Illuminating Neuropeptide Y Y4 Receptor Binding: Fluorescent Cyclic Peptides with Subnanomolar Binding Affinity as Novel Molecular ToolsClick to copy article linkArticle link copied!
- Jakob GleixnerJakob GleixnerInstitute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Jakob Gleixner
- Sergei KopanchukSergei KopanchukInstitute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, EstoniaMore by Sergei Kopanchuk
- Lukas GrätzLukas GrätzInstitute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Lukas Grätz
- Maris-Johanna TahkMaris-Johanna TahkInstitute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, EstoniaMore by Maris-Johanna Tahk
- Tõnis LaasfeldTõnis LaasfeldInstitute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, EstoniaMore by Tõnis Laasfeld
- Santa VeikšinaSanta VeikšinaInstitute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, EstoniaMore by Santa Veikšina
- Carina HöringCarina HöringInstitute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Carina Höring
- Albert O. GattorAlbert O. GattorInstitute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Albert O. Gattor
- Laura J. HumphrysLaura J. HumphrysInstitute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Laura J. Humphrys
- Christoph MüllerChristoph MüllerInstitute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Christoph Müller
- Nataliya ArchipowaNataliya ArchipowaInstitute of Biophysics and Physical Biochemistry, Faculty of Biology and Preclinical Medicine, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Nataliya Archipowa
- Johannes KöckenbergerJohannes KöckenbergerDepartment of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, GermanyMore by Johannes Köckenberger
- Markus R. HeinrichMarkus R. HeinrichDepartment of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, GermanyMore by Markus R. Heinrich
- Roger Jan KuttaRoger Jan KuttaInstitute of Physical and Theoretical Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, GermanyMore by Roger Jan Kutta
- Ago Rinken*Ago Rinken*Email: [email protected]. Tel: (+372) 7375-249.Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, EstoniaMore by Ago Rinken
- Max Keller*Max Keller*Email: [email protected]. Tel: (+49) 941-9433329.Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93040 Regensburg, GermanyMore by Max Keller
Abstract
The neuropeptide Y (NPY) Y4 receptor (Y4R), a member of the family of NPY receptors, is physiologically activated by the linear 36-amino acid peptide pancreatic polypeptide (PP). The Y4R is involved in the regulation of various biological processes, most importantly pancreatic secretion, gastrointestinal motility, and regulation of food intake. So far, Y4R binding affinities have been mostly studied in radiochemical binding assays. Except for a few fluorescently labeled PP derivatives, fluorescence-tagged Y4R ligands with high affinity have not been reported. Here, we introduce differently fluorescence-labeled (Sulfo-Cy5, Cy3B, Py-1, Py-5) Y4R ligands derived from recently reported cyclic hexapeptides showing picomolar Y4R binding affinity. With pKi values of 9.22–9.71 (radioligand competition binding assay), all fluorescent ligands (16–19) showed excellent Y4R affinity. Y4R saturation binding, binding kinetics, and competition binding with reference ligands were studied using different fluorescence-based methods: flow cytometry (Sulfo-Cy5, Cy3B, and Py-1 label), fluorescence anisotropy (Cy3B label), and NanoBRET (Cy3B label) binding assays. These experiments confirmed the high binding affinity to Y4R (equilibrium pKd: 9.02–9.9) and proved the applicability of the probes for fluorescence-based Y4R competition binding studies and imaging techniques such as single-receptor molecule tracking.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Figure 1
Figure 1. (A) Structures and Y4R affinities of reported Y4R fluorescent ligands. 1 (26) and 2 (27) represent derivatives of the endogenous Y4R ligand hPP labeled with sulfo-Cy5 (S0223) and 5-TAMRA, respectively. Compound 3 (29) represents a derivative of the hexapeptide UR-KK236 (28) labeled with Sulfo-Cy5.5 (lumiprobe, ref no. 7330). (B) Structures of the reported cyclic Y4R ligands 4–7 showing high binding affinity to Y4R (pKi > 10). (30,31)
Figure 2
Results and Discussion
Synthesis and Chemical Stability
Figure 3
Scheme 1
aReagents and conditions: (a) Fmoc amino acid/HOBt/HBTU/DIPEA (5/5/4.9/10 equiv), DMF/NMP 8:2 v/v, 38 °C, 2 × 45 min (“double” coupling); Fmoc deprotection (following amino acid coupling): 20% piperidine in DMF/NMP 8:2 v/v, rt, 2 × 10 min; (b) building block 8 or 9/HOBt/HBTU/DIPEA (3/3/2.9/6 equiv), DMF/NMP 8:2 v/v, 38 °C, 16 h (single coupling); Fmoc deprotection (following amino acid coupling): as under (a); (c) succinic anhydride/DIPEA (10/10 equiv), DMF/NMP 8:2 v/v, 38 °C, 45 min; (d) (1) TFA/CH2Cl2 1:3 v/v, rt, 2 × 20 min; (2) TFA/H2O 95:5 v/v, rt, 5 h; overall yield: 37%; (e) peptide cyclization: HOBt/PyBOP/DIPEA (3/2.9/6 equiv), DMF/NMP 8:2 v/v, rt, 16 h, 21%.
Scheme 2
aReagents and conditions: (a) DIPEA, DMF, rt, 2 h, 34–69%; (b) CuSO4, sodium ascorbate, H2O/NMP 1:1 v/v, rt, 2 h, 27%.
Investigation of YR Binding in Radiochemical Binding Assays
pKi ± SEM/Ki [nM] | ||||
---|---|---|---|---|
compd. | hY1Ra | hY2Rb | hY4Rc | hY5Rd |
hPP | 6.4/440e | <5.5/>3000e | 10.02 ± 0.06/0.10f | 7.8/17e |
4 | <5.5/>3000g | <5/>10000g | 10.36 ± 0.11/0.048g | <5.5/>3000g |
5 | <5/>10000g | <5/>10000g | 10.48 ± 0.04/0.033g | <5.5/>3000g |
6 | <6/>1000h | 6.04 ± 0.07/950h | 10.04 ± 0.04/0.093h | <6/>1000h |
11 | <5/>10000 | <5/>10000 | 9.38 ± 0.04/0.42 | <5.5/>3000 |
16 | 6.91 ± 0.01/120 | <6/>1000 | 9.71 ± 0.09/0.21 | 6.32 ± 0.04/480 |
17 | <6/>1000 | <6/>1000 | 9.48 ± 0.09/0.35 | <6/>1000 |
18 | <6/>1000 | <6/>1000 | 9.22 ± 0.03/0.61 | 6.08 ± 0.05/840 |
19 | <6/>1000 | <6/>1000 | 9.22 ± 0.06/0.62 | <6/>1000 |
Determined by competition binding at SK-N-MC neuroblastoma cells using [3H]UR-MK299 (Kd = 0.054 nM, c = 0.15 nM) as radioligand.
Determined by competition binding with [3H]propionyl-pNPY (Kd = 0.14 nM, (34) c = 0.5 nM) at CHO-hY2R cells.
Determined by competition binding at CHO-hY4R-Gqi5-mtAEQ cells using [3H]UR-KK200 (Kd = 0.67 nM, (35) c = 1 nM) as radioligand.
Determined by competition binding at HEC-1B-hY5R cells using [3H]propionyl-pNPY (Kd = 11 nM, (26) c = 5 nM) as radioligand.
Berlicki et al. (reported Ki values were converted to pKi values). (36)
Wirth et al. (37)
Konieczny et al. (30)
Gleixner et al. (31) Data represent mean values ± SEM (pKi) or mean values (Ki) from 3 to 4 independent experiments performed in triplicate.
Functional Studies at Y4R
Ca2+-aequorin | miniGsi recruitment | CAMYEN cAMP | ||||
---|---|---|---|---|---|---|
cmpd. | pEC50 ± SEM/EC50 [nM] | Emax ± SEM/% | pEC50 ± SEM/EC50 [nM] | Emax ± SEM/% | pEC50 ± SEM/EC50 [nM] | Emax ± SEM/% |
hPP | 7.90 ± 0.2/17b | 100b | 8.94 ± 0.01/1.1b | 100b | 9.85 ± 0.09/0.15b | 100b |
4 | 8.57 ± 0.03/2.7c | 81 ± 1c | n.d. | n.d. | 9.79 ± 0.1/0.17b | 113 ± 9b |
5 | 9.00 ± 0.07/0.99c | 84 ± 2c | 8.74 ± 0.04/1.8 | 61 ± 2 | n.d. | n.d. |
6 | 7.76 ± 0.05/18b | 69 ± 3b | 8.60 ± 0.03/2.5b | 62 ± 2b | n.d. | n.d. |
11 | 7.01 ± 0.01/98 | 56 ± 3 | 7.2 ± 0.1/73 | 62 ± 1 | n.d. | n.d. |
16 | 7.8 ± 0.1/19 | 66 ± 2 | 7.4 ± 0.1/41 | 72 ± 9 | 9.58 ± 0.06/0.26 | 93 ± 5 |
17 | 7.41 ± 0.05/40 | 71 ± 3 | 7.07 ± 0.09/93 | 70 ± 9 | 9.65 ± 0.05/0.22 | 90 ± 3 |
18 | 7.3 ± 0.1/65 | 61 ± 5 | 8.15 ± 0.01/7.1 | 44 ± 2 | n.d. | n.d. |
19 | 7.25 ± 0.07/58 | 64 ± 2 | 7.38 ± 0.02/42 | 43 ± 1 | n.d. | n.d. |
Agonistic potencies (pEC50, EC50) and efficacies Emax (relative to the maximum effect elicited by 1 μM hPP, Emax = 100%) determined in a Ca2+-aequorin assay (CHO-hY4R-Gqi5-mtAEQ cells), a miniGsi recruitment assay (HEK293T-NlucN-mGsi/Y4R-NlucC cells) and in a CAMYEN cAMP assay (HEK293T-CAMYEN-hY4R cells).
Gleixner et al. (31)
Konieczny et al. (30) Data represent mean values ± SEM (pEC50, Emax) or mean values (EC50) from three or four independent experiments performed in triplicate. n.d. = not determined.
Fluorescence Characterization
λex [nm]/λem [nm]/Δ [eV] | Φ [%] | ||||
---|---|---|---|---|---|
compd. | dye | PBS | PBS with 1% BSA | PBS | PBS with 1% BSA |
16 | Cy3B | 571/584/0.048 | 572/586/0.052 | 69 | 67 |
17 | Sulfo-Cy5 | 648/665/0.049 | 657/670/0.037 | 26 | 33 |
18 | py-1 | 517/633/0.439 | 506/595/0.367 | 2 | 36 |
19 | py-5 | 496/692/0.708 | 515/644/0.482 | 4 | 24 |
Flow Cytometric Y4R Binding Studies
Figure 4
Figure 4. Flow cytometric saturation binding of 16 (A), 17 (B), and 18 (C) at whole CHO-hY4R-Gqi5-mtAEQ cells at 22 ± 2 °C. (A) Representative saturation isotherms (red circle) of 16 from experiments performed in sodium-free buffer (buffer I) and sodium-containing buffer (DPBS, 137 mM Na+). (B, C) Representative saturation isotherms (red circle) of 17 (B) and 18 (C) from experiments performed in sodium-containing buffer (DPBS). Unspecific binding (blue squares) was determined in the presence of 1 μM hPP (A–C). Total and unspecific binding data represent mean values ± SEM. Specific binding, representing calculated values ± propagated error, were fitted according to an equation describing a hyperbolic isotherm (binding-saturation: one site-specific binding, GraphPad Prism 5).
saturation binding | binding kinetics | |||||||
---|---|---|---|---|---|---|---|---|
cmpd. | buffer | pKd/Kd [nM]a | kobs(mono) [min–1]b | kobs(bi,fast) [min–1]ckobs(bi,slow) [min–1]c | koff [min–1]d | kon(mono) [nM–1 min–1]e | kon(bi,fast) [nM–1 min–1]e kon(bi,slow) [nM–1 min–1]e | Kd(kin) [nM]f |
16 | buffer I (Na+-free) | 9.56 ± 0.09/0.30 | 0.13 ± 0.02 | n.a. | 0.0069 ± 0.0006 | 0.41 ± 0.07 | n.a. | 0.017 ± 0.004 |
16 | DPBS (137 mM Na+) | 9.16 ± 0.05/0.70 | 0.18 ± 0.04 | 1.0 ± 0.2 | 0.012 ± 0.001 | 0.24 ± 0.05 | 1.3 ± 0.3 | with kon(mono): 0.05 ± 0.02 |
0.048 ± 0.003 | 0.050 ± 0.006 | with kon(bi,fast): 0.009 ± 0.002 | ||||||
with kon(bi,slow): 0.25 ± 0.05 | ||||||||
17 | DPBS (137 mM Na+) | 9.03 ± 0.02/0.93 | 0.15 ± 0.01 | 1.89 ± 0.05 | 0.026 ± 0.002 | 0.12 ± 0.01 | 1.87 ± 0.05 | with kon(mono): 0.21 ± 0.03 |
0.071 ± 0.008 | 0.045 ± 0.009 | with kon(bi,fast): 0.007 ± 0.001 | ||||||
with kon(bi,slow): 0.29 ± 0.07 | ||||||||
18 | DPBS (137 mM Na+) | 9.3 ± 0.1/0.51 | 0.10 ± 0.02 | 0.7 ± 0.1 | 0.010 ± 0.001 | 0.18 ± 0.04 | 1.3 ± 0.2 | with kon(mono): 0.06 ± 0.02 |
0.034 ± 0.001 | 0.048 ± 0.004 | with kon(bi,fast): 0.008 ± 0.002 | ||||||
with kon(bi,slow): 0.20 ± 0.05 |
Equilibrium dissociation constant expressed as pKd (mean values ± SEM) and Kd (mean values) obtained from at least three independent experiments (performed in triplicate).
Observed association rate constant obtained by monophasic fitting (exponential rise to a maximum); mean values ± SEM from three independent experiments (performed in duplicate).
Observed association rate constant obtained by biphasic fitting; mean values ± SEM from three or four independent experiments (performed in duplicate).
Dissociation rate constant obtained from three-parameter monophasic fits (exponential decline); mean values ± SEM from three or four independent experiments (performed in duplicate).
Association rate constant ± propagated error calculated from kobs(mono), kobs(bi,fast), or kobs(bi,slow) values, the respective koff value, and the ligand concentration used for the association experiments.
Kinetically derived dissociation constant ± propagated error calculated from koff and kon values. n.a.: not applicable
Figure 5
Figure 5. Binding kinetics of 16–18 determined by flow cytometry at whole CHO-hY4R-Gqi5-mtAEQ cells at 22 ± 2 °C. (A) Association of 16 to the hY4R under sodium-free conditions (buffer I) and in sodium-containing buffer (DPBS, 137 mM Na+). Concentration of 16: 0.3 and 0.7 nM, respectively. Proportion of the initial fast association (two-phase association fit, GraphPad Prism 5): 38 ± 4% (mean ± SEM). (B) Dissociation of 16 from the hY4R determined in buffer I and DPBS. The dissociation was initiated after 1.5 h of preincubation with 16 (c = 1.5 nM (Na+-free) and 3.5 nM (137 mM Na+)) by the addition of an excess of hPP (1000-fold) and 5 (100-fold). Plateau values of the three-parameter fits (monophasic exponential decline): 13% (Na+-free), 22% (137 mM Na+). (C) Association of 17 (c = 1 nM) and 18 (c = 0.5 nM) to the hY4R determined in DPBS (137 mM Na+). Proportion of the initial fast association (two-phase association fit, GraphPad Prism 5): 74 ± 1% (17), 29 ± 3% (18) (mean values ± SEM). (D) Dissociation of 17 and 18 from the hY4R in DPBS. The dissociation was initiated after 1.5 h of preincubation with 17 (c = 5 nM) or 18 (c = 2.5 nM) by the addition of an excess of hPP (1000-fold) and 5 (100-fold). Plateau values of the three-parameter fits (monophasic exponential decline): 13% (17), 22% (18). Data (A–D) represent mean values ± SEM from three independent experiments performed in duplicate.
Fluorescence Anisotropy-Based Y4R Binding Studies
Figure 6
Figure 6. Binding of 16 to hY4R displaying BBVs studied by FA measurement at 27 °C. (A) Binding isotherms of 16 obtained from experiments using fixed concentrations of 16 (0.5 or 2 nM) and increasing amounts of Y4R. Total binding is represented by filled symbols and unspecific binding (determined in the presence of 1 μM hPP) is represented by open symbols. Depicted data (mean values ± SEM from a representative experiment performed in duplicate) represent snapshots at 90 min incubation. Y4R concentrations displayed on the abscissa were calculated after global analysis of the data from three or four individual experiments by a modified version of a model described by Veiksina et al., (19) affording the estimated binding site (Y4R) concentration of the applied BBV stock which amounted to 6 ± 1 nM (Y4Rnonglyco, mean value ± SEM, n = 3) or 2.1 ± 0.1 nM (Y4RSwBac, mean value ± SEM, n = 4). (B) Association and dissociation of 16 (0.5 nM) determined in real time for three different Y4R concentrations (green, blue, and red symbols). Total binding is represented by filled symbols and unspecific binding (determined in the presence of 1 μM hPP) is represented by open symbols. Data represent mean values ± SEM from a representative experiment performed in duplicate.
saturation binding | binding kinetics | |||
---|---|---|---|---|
BBV | pKd/Kd [nM]a | kon [nM–1 min–1]b | koff [min–1]c | Kd(kin) [nM]d |
Y4Rnonglyco | 9.2 ± 0.1/0.60 | 0.015 ± 0.004 | 0.0012 ± 0.0001 | 0.11 ± 0.03 |
Y4RSwBac | 9.9 ± 0.1/0.14 | 0.021 ± 0.002 | 0.00098 ± 0.00003 | 0.052 ± 0.006 |
Equilibrium dissociation constant expressed as pKd (mean values ± SEM) and Kd (mean value) obtained from three independent experiments (performed in duplicate).
Association rate constant ± SEM obtained from global analysis (19) of data from three individual experiments (performed in duplicate) each involving two different concentrations of 16 (0.5 and 7.0, or 0.5 and 2.0 nM).
Dissociation rate constants obtained from three-parameter monophasic fits (exponential decline). Mean values ± SEM from three independent experiments (performed in triplicate).
Kinetically derived dissociation constants ± SEM calculated from the mean koff value and individual kon values.
Figure 7
Figure 7. Characterization of Y4R binding of fluorescent ligand 16 in a NanoBRET-based binding assay at 25 °C using intact HEK293T-hY4R-NLuc(intraECL2) cells. (A) Representative saturation isotherm (specific binding) from saturation binding experiments. Unspecific binding was determined in the presence of 1 μM 5. Total and unspecific binding data represent mean values ± SEM. Specific binding data represent calculated values ± propagated error. (B) Association of 16 (c = 1 nM) to Y4R. Mean values ± SEM from three independent experiments performed in triplicate. (C) Dissociation of 16 from Y4R. The dissociation was initiated after 1.5 h of preincubation with 16 (c = 3.5 nM) by the addition of a 1000-fold excess of 5. Mean values ± SEM from three independent experiments performed in duplicate. Plateau value of the three-parameter fit describing a monophasic exponential decline: 6% (note: for the biphasic fit the plateau value was not different from zero, see discussion).
NanoBRET Y4R Binding Studies
Figure 8
Figure 8. Determination of Y4R affinities of Y4R reference ligands (hPP, 5, 7, UR-MK188, UR-MEK388, UR-KK200) in different fluorescence-based assays by competition binding with 16. (A) Displacement curves based on data from flow cytometric competition binding experiments performed with intact CHO-hY4R-Gqi5-mtAEQ cells in sodium-free buffer (buffer I) and sodium-containing buffer (DPBS, 137 mM Na+). (B) Displacement curves from fluorescence anisotropy-based competition binding experiments performed with Y4RSwBac-displaying BBVs in DPBS. (C) Displacement curves from NanoBRET-based competition binding experiments performed with intact HEK293T-hY4R-NLuc(intraECL2) cells in L15-HEPES (140 mM Na+). Data (A–C), representing mean values ± SEM from three to five independent experiments performed in duplicate (B) or triplicate (A, C), were fitted according to a four-parameter logistic model.
saturation binding | binding kinetics | |||
---|---|---|---|---|
pKd/Kd [nM]a | kobs [min–1]b | koff [min–1]c | kon [nM–1min–1]d | Kd(kin) [nM]e |
9.02 ± 0.04/0.94 | mono: 0.18 ± 0.04 | mono: 0.06 ± 0.01 | mono: 0.18 ± 0.07 | mono: 0.3 ± 0.2 |
bi, fast: 0.58 ± 0.02 | bi, fast: 0.34 ± 0.03 | bi, fast: 0.24 ± 0.05 | bi, fast: 1.4 ± 0.4 | |
bi, slow: 0.095 ± 0.005 | bi, slow: 0.016 ± 0.002 | bi, slow: 0.078 ± 0.008 | bi, slow: 0.21 ± 0.05 |
Equilibrium dissociation constant expressed as pKd (mean values ± SEM) and Kd (mean value) obtained from three independent experiments (performed in triplicate).
Observed association rate constants obtained by monophasic and biphasic fitting. Mean values ± SEM from four independent experiments (performed in triplicate).
Dissociation rate constants obtained from three-parameter monophasic (Y0 constrained to 100%) and five-parameter biphasic (Y0 constrained to 100%, plateau constrained to zero) fits (exponential decline). Mean values ± SEM from three independent experiments (performed in triplicate).
Association rate constant ± propagated error calculated from kobs, koff, and the ligand concentration used for the association studies.
Kinetically derived dissociation constants ± propagated error calculated from various koff and kon values.
Fluorescence-Based Y4R Competition Binding
flow cytometrya | FAb | nanoBRETc | literature | |||
---|---|---|---|---|---|---|
Na+ free | 137 mM Na+ | 137 mM Na+ | 140 mM Na+ | Na+ free | 137 mM Na+ | |
cmpd. | pKi | |||||
hPP | 8.5 ± 0.1 | 8.9 ± 0.1 | 8.0 ± 0.2 | 7.7 ± 0.1 | 10.02d | 10.1e |
5 | 9.49 ± 0.07 | 9.15 ± 0.08 | 9.26 ± 0.04 | 8.89 ± 0.06 | 10.48f | n.a. |
7 | 9.61 ± 0.06 | 8.8 ± 0.1 | 9.4 ± 0.1 | 9.07 ± 0.07 | 10.5e | 9.79e |
UR-KK200 | n.d. | n.d. | n.d. | 7.8 ± 0.1 | 8.92g | 7.99e |
UR-MEK388 | 6.29 ± 0.08 | 6.68 ± 0.09 | n.d. | 6.85 ± 0.05 | 6.58h | n.a. |
UR-MK188 | n.d. | n.d. | 7.7 ± 0.1 | n.d. | 6.88h | 6.82g |
6.18g |
Determined at intact CHO-hY4R-Gqi5-mtAEQ cells; mean values ± SEM from three or four independent experiments performed in triplicate.
Determined at Y4RSwBac displaying BBVs; mean values ± SEM from three independent experiments performed in duplicate.
Determined at intact HEK293T-hY4R-NLuc(intraECL2) cells; mean values ± SEM from three or four independent experiments performed in triplicate.
Wirth et al. (37)
Gleixner et al. (31)
Konieczny et al. (30)
Kuhn et al. (reported Ki values were converted to pKi). (35)
Microscopy Studies
Figure 9
Figure 9. Visualization of fluorescent ligand (16, 17) binding to CHO-hY4R-Gqi5-mtAEQ cells by confocal microscopy. Shown are representative images acquired after incubation of the cells with 16 or 17 (each 20 nM) at 22 °C for 10 and 30 min. Unspecific binding was determined in the presence of 1 μM hPP. Nuclei were stained with H33342 (2 μM). Fluorescence of 16 and 17 is shown in green and red, respectively. Fluorescence of H33342 is shown in blue. Scale bar: 10 μm.
Figure 10
Figure 10. Visualization of binding of 16 to the hY4R transiently expressed by SK-OV-3 cells using wide-field and TIRF microscopy. (A) Wide-field fluorescence images acquired after incubation of the cells with 16 at 37 °C for 30 min. The two-color composite of individual focal planes after Z-stack deconvolution is shown with the green pseudocolor for 16 (561 nm excitation) and the blue pseudocolor for the nuclear stain channel (Hoechst 34580, 405 nm excitation). (B) Wide-field fluorescence and TIRF images of the same cells obtained after incubation of the cells with 16 (1 nM) at 37 °C for 30 min. Wide-field images were processed as under (A). In TIRF images, fluorescence of 16 is shown in white pseudocolor. Scale bar: 10 μm.
Conclusions
Experimental Section
Materials
NMR Spectroscopy
Mass Spectrometry
Preparative HPLC
Analytical HPLC
Compound Characterization
Synthesis of Peptide 11 and the Fluorescent Ligands 16–19
Nα-Succinyl-Nω[hex-5-ynylaminocarbonyl]-Arg-Tyr-Nω-[(aminobutyl)aminocarbonyl] Arg-Leu-Arg-Tyr-Amide Tetrakis(hydrotrifluoroacetate) (10)
(9S,12S,15S,E)-4-Amino-N-{(S)-1-[((S)-1-{[(S)-1-amino-3-(4-hydroxyphenyl)-1-oxopropan-2-yl]amino}-5-guanidino-1-oxopentan-2-yl)amino]-4-methyl-1-oxopentan-2-yl}-15-{3-[(E)-2-(hex-5-yn-1-ylcarbamoyl)guanidino]propyl}-12-(4-hydroxybenzyl)-2,11,14,17,20-pentaoxo-1,3,5,10,13,16,21-heptaazacyclopentacos-3-ene-9-carboxamide Tris(hydrotrifluoroacetate) (11)
24-{5-[(4-{[(9S,12S,15S,19R,E)-4-Amino-9-({(S)-1-[((S)-1-{[(S)-1-amino-3-(4-hydroxyphenyl)-1-oxopropan-2-yl]amino}-5-guanidino-1-oxopentan-2-yl)amino]-4-methyl-1-oxopentan-2-yl}carbamoyl)-15-(3-guanidinopropyl)-12-(4-hydroxybenzyl)-2,11,14,17,20-pentaoxo-1,3,5,10,13,16,21-heptaazacyclopentacos-3-en-19-yl]amino}-4-oxobutyl)amino]-5-oxopent-1-yn-1-yl}-5,5,27,27-tetramethyl-16-oxa-20-aza-12-azoniaheptacyclo[15.11.0.03,15.04,12.06,11.020,28.021,26]octacosa-1(28),2,4(12),6(11),7,9,21(26),22,24-nonaene-8-sulfonate Bis(hydrotrifluoroacetate) (16)
4-(2-{(1E,3E)-5-[(E)-1-(6-{[4-({(9S,12S,15S,19R,E)-4-Amino-9-[((S)-1-{[(S)-1-({(S)-1-amino-3-(4-hydroxyphenyl)-1-oxopropan-2-yl}amino)-5-guanidino-1-oxopentan-2-yl]amino}-4-methyl-1-oxopentan-2-yl)carbamoyl]-15-(3-guanidinopropyl)-12-(4-hydroxybenzyl)-2,11,14,17,20-pentaoxo-1,3,5,10,13,16,21-heptaazacyclopentacos-3-en-19-yl}amino)-4-oxobutyl]amino}-6-oxohexyl)-3,3-dimethyl-5-sulfoindolin-2-ylidene]penta-1,3-dien-1-yl}-3,3-dimethyl-3H-indol-1-ium-1-yl)butane-1-sulfonate Bis(hydrotrifluoroacetate) (17)
1-{4-[((9S,12S,15S,19R,E)-4-Amino-9-{[(S)-1-({(S)-1-[((S)-1-amino-3-(4-hydroxyphenyl)-1-oxopropan-2-yl)amino]-5-guanidino-1-oxopentan-2-yl}amino)-4-methyl-1-oxopentan-2-yl]carbamoyl}-15-(3-guanidinopropyl)-12-(4-hydroxybenzyl)-2,11,14,17,20-pentaoxo-1,3,5,10,13,16,21-heptaazacyclopentacos-3-en-19-yl)amino]-4-oxobutyl}-2,6-dimethyl-4-[(E)-2-(2,3,6,7-tetrahydro-1H,5H-pyrido[3,2,1-ij]quinolin-9-yl)vinyl]pyridin-1-ium Bis(hydrotrifluoroacetate) Trifluoroacetate (18)
1-{3-[4-(4-{3-[(E)-Amino({3-[(9S,12S,15S,E)-4-amino-9-({(S)-1-[((S)-1-{[(S)-1-amino-3-(4-hydroxyphenyl)-1-oxopropan-2-yl]amino}-5-guanidino-1-oxopentan-2-yl)amino]-4-methyl-1-oxopentan-2-yl}carbamoyl)-12-(4-hydroxybenzyl)-2,11,14,17,20-pentaoxo-1,3,5,10,13,16,21-heptaazacyclopentacos-3-en-15-yl]propyl}amino)methylene]ureido}butyl)-1H-1,2,3-triazol-1-yl]propyl}-4-{(1E,3E)-4-[4-(dimethylamino)phenyl]buta-1,3-dien-1-yl}-2,6-dimethylpyridin-1-ium Bis(hydrotrifluoroacetate) Trifluoroacetate (19)
Chemical Stability
Fluorescence Excitation and Emission Spectra
Determination of Fluorescence Quantum Yields
Cell Culture
Molecular Cloning
Generation of the Stable HEK293T-Nluc-hY4R and HEK293T-hY4R-Nluc(intraECL2) Cell Lines
Buffers and Media Used for Binding and Functional Assays
Radioligand Binding Assays
Y1R Binding
Y2R Binding
Y4R Binding
Y5R Binding
Functional Y4R Assays
Y4R Ca2+ Aequorin Assay
Y4R miniGsi Recruitment Assay
Y4R cAMP CAMYEN Assay
Flow Cytometric Y4R Binding Assays
Fluorescence Anisotropy Y4R Binding Assays
NanoBRET Y4R Binding Assay
Confocal Microscopy
Wide-Field Epifluorescence and TIRF Microscopy
Statistical Significance
Calculation of Propagated Errors
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsptsci.4c00013.
Preparation of the azido-functionalized Py-5 derivative 15; chromatograms of the investigation of the chemical stability of 16–18 in PBS pH 7.4 (Figure S1); radioligand displacement curves from competition binding experiments with [3H]UR-KK200 (Figure S2); concentration–response curves of hPP, 11 and 16–19 obtained from a hY4R Ca2+ aequorin assay (Figure S3); concentration–response curves of hPP, 5, 11, and 16–19 obtained from a hY4R mini-Gsi protein recruitment assay (Figure S4); concentration–response curves of hPP, 16 and 17 obtained from a hY4R CAMYEN cAMP assay (Figure S5); study of dummy fluorescent ligands in functional assays (Figure S6); excitation and emission spectra (Figure S7); viability of CHO-hY4R-Gqi5-mtAEQ cells (Figure S8); analyses of flow cytometric saturation binding data of 16 based on nonviable and viable cell populations (Figure S9); syntax of the equation used to fit FA equilibrium binding data (GraphPad Prism 5); RP-HPLC chromatograms of compounds 11 and 16–19; and 1H NMR spectra of compound 11 (PDF)
TIRF video sequence (recorded and shown at a 30 Hz resolution, shown at double speed) showing the interactions of 16 with the basal plasma membrane of adherent SK-OV-3-Y4R cells including single-particle tracking (magnified region) (AVI)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
The authors thank Susanne Bollwein, Maria Beer-Krön, Brigitte Wenzl, Carmen Piirsalu, and Merve Gökalp for excellent technical assistance, and Sabrina Diwisch, Luise Liebig, and Tobias Spitzl for support with the flow cytometric assays. This work was funded by the Deutsche Forschungsgemeinschaft (DFG) (research grant KE 1857/1-3) and the Estonian Research Council grant PSG230 and was additionally supported by the Graduate Training Program GRK 1910 of the DFG.
Arg(carb) | Nω-carbamoylated arginine containing a tetramethylene spacer or a hexyne structure with a terminal alkyne group in the carbamoyl substituent (cf. Figure 3) |
BSA | bovine serum albumin |
CAMYEN | cAMP sensor using YFP-Epac-nanoluciferase |
CHO | Chinese hamster ovary |
Cy3B-SE | Cy3B succinimidyl ester |
DIPEA | N,N-diisopropylethylamine |
Emax | efficacy (maximum effect) of a receptor agonist determined in a functional assay |
Epac | exchange protein activated by cAMP |
EtOH | ethanol |
FA | fluorescence anisotropy |
FBS | fetal bovine serum |
FL | fluorescent ligand |
Fmoc amino acid | Nα-Fmoc-protected and side-chain-protected (if required) amino acid |
HBTU | 3-[bis(dimethylamino)methyliumyl]-3H-benzotriazol-1-oxide hexafluorophosphate |
HEC | human endometrial cancer |
HEPES | 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid |
HOBt | hydroxybenzotriazole |
hPP | human pancreatic polypeptide |
hY4R | human Y4 receptor |
k | retention (or capacity) factor (HPLC) |
MeOH | methanol |
Nluc | nanoluciferase |
Pbf | 2,2,4,6,7-pentamethyl-dihydrobenzofuran-5-sulfonyl |
PBS | phosphate-buffered saline |
pEC50 | negative decadic logarithm of the half-maximal effective concentration (functional assays) |
pKi | negative decadic logarithm of the dissociation constant Ki (in M) obtained from a competition binding experiment |
pNPY | porcine neuropeptide Y |
PS | polystyrene |
PyBOP | benzotriazol-1-yl-oxytripyrrolidinophosphonium-hexafluorophosphate |
PYY | peptide YY |
RP | reversed phase |
SEM | standard error of the mean |
SPPS | solid-phase peptide synthesis |
TFA | trifluoroacetic acid |
TIRF | total internal reflection fluorescence |
tR | retention time |
YFP | yellow fluorescent protein |
YR | NPY receptor |
References
This article references 68 other publications.
- 1Michel, M. C.; Beck-Sickinger, A.; Cox, H.; Doods, H. N.; Herzog, H.; Larhammar, D.; Quirion, R.; Schwartz, T.; Westfall, T. XVI. International Union of Pharmacology recommendations for the nomenclature of neuropeptide Y, peptide YY, and pancreatic polypeptide receptors. Pharmacol. Rev. 1998, 50, 143– 150Google Scholar1XVI. International union of pharmacology recommendations for the nomenclature of neuropeptide Y, peptide YY, and pancreatic polypeptide receptorsMichel, Martin C.; Beck-Sickinger, Annette; Cox, Helen; Doods, Henri N.; Herzog, Herbert; Larhammar, Dan; Quirion, Remi; Schwartz, Thue; Westfall, ThomasPharmacological Reviews (1998), 50 (1), 143-150CODEN: PAREAQ; ISSN:0031-6997. (Williams & Wilkins)A review with ∼80 refs.
- 2Pedragosa-Badia, X.; Stichel, J.; Beck-Sickinger, A. G. Neuropeptide Y receptors: how to get subtype selectivity. Front. Endocrinol. 2013, 4, 5 DOI: 10.3389/fendo.2013.00005Google Scholar2Neuropeptide Y receptors: how to get subtype selectivityPedragosa-Badia Xavier; Stichel Jan; Beck-Sickinger Annette GFrontiers in endocrinology (2013), 4 (), 5 ISSN:1664-2392.The neuropeptide Y (NPY) system is a multireceptor/multiligand system consisting of four receptors in humans (hY(1), hY(2), hY(4), hY(5)) and three agonists (NPY, PYY, PP) that activate these receptors with different potency. The relevance of this system in diseases like obesity or cancer, and the different role that each receptor plays influencing different biological processes makes this system suitable for the design of subtype selectivity studies. In this review we focus on the latest findings within the NPY system, we summarize recent mutagenesis studies, structure activity relationship studies, receptor chimera, and selective ligands focusing also on the binding mode of the native agonists.
- 3Ueno, N.; Inui, A.; Iwamoto, M.; Kaga, T.; Asakawa, A.; Okita, M.; Fujimiya, M.; Nakajima, Y.; Ohmoto, Y.; Ohnaka, M.; Nakaya, Y.; Miyazaki, J.-I.; Kasuga, M. Decreased food intake and body weight in pancreatic polypeptide-overexpressing mice. Gastroenterology 1999, 117, 1427– 1432, DOI: 10.1016/S0016-5085(99)70293-3Google Scholar3Decreased food intake and body weight in pancreatic polypeptide-overexpressing miceUeno N; Inui A; Iwamoto M; Kaga T; Asakawa A; Okita M; Fujimiya M; Nakajima Y; Ohmoto Y; Ohnaka M; Nakaya Y; Miyazaki J I; Kasuga MGastroenterology (1999), 117 (6), 1427-32 ISSN:0016-5085.BACKGROUND & AIMS: Pancreatic polypeptide (PP) is a 36-amino acid hormone produced by F cells within the pancreatic islets and the exocrine pancreas. The definitive function of PP in mammalian physiology remains to be determined. This study examined the effects of chronic overexpression of PP through the development of PP transgenic mice. METHODS: PP transgenic mice were created by using mouse PP complementary DNA under the control of the cytomegalovirus immediate early enhancer-chicken beta-actin hybrid promoter (pCAGGS expression vector). RESULTS: A unique line of transgenic mice was created that overexpresses PP in the pancreatic islets with low levels of expression in other tissues including the brain. Plasma PP concentrations were more than 20 times higher than those of control littermates. However, PP overproduction led to postnatal lethality in half of the pups because of markedly decreased milk intake. The remaining PP transgenic mice gained less weight with specifically reduced food intake and fat mass compared with controls, a result that was more evident in male than in female mice. The transgenic mice exhibited a reduced rate of gastric emptying of a solid meal but had normal oxygen consumption and fasting leptin levels. Immunoneutralization with anti-PP antiserum reversed the phenotypic changes of transgenic animals. CONCLUSIONS: PP could be involved in feeding and body weight regulation partly through regulation of gastric emptying.
- 4Batterham, R. L.; Le Roux, C. W.; Cohen, M. A.; Park, A. J.; Ellis, S. M.; Patterson, M.; Frost, G. S.; Ghatei, M. A.; Bloom, S. R. Pancreatic polypeptide reduces appetite and food intake in humans. J. Clin. Endocrinol. Metab. 2003, 88, 3989– 3992, DOI: 10.1210/jc.2003-030630Google Scholar4Pancreatic polypeptide reduces appetite and food intake in humansBatterham, R. L.; Le Roux, C. W.; Cohen, M. A.; Park, A. J.; Ellis, S. M.; Patterson, M.; Frost, G. S.; Ghatei, M. A.; Bloom, S. R.Journal of Clinical Endocrinology and Metabolism (2003), 88 (8), 3989-3992CODEN: JCEMAZ; ISSN:0021-972X. (Endocrine Society)Pancreatic polypeptide (PP) is a gut hormone released from the pancreas in response to ingestion of food. Plasma PP has been shown to be reduced in conditions assocd. with increased food intake and elevated in anorexia nervosa. In addn. peripheral administration of PP has been shown to decrease food intake in rodents. These findings suggest that PP may act as a circulating factor that regulates food intake. Therefore we investigated the effect of i.v. infusion of PP (10 pmol/kg/min) on appetite and food intake in a randomized double-blind placebo-controlled crossover study in ten healthy volunteers. Infusion of PP reduced appetite and decreased the energy intake at a buffet lunch two hours post-infusion by 21.8±5.7% (P < 0.01). More importantly the inhibition of food intake was sustained, such that energy intake, as assessed by food diaries, was significantly reduced both the evening of the study and the following morning. Overall PP infusion reduced cumulative 24-h energy intake by 25.3±5.8%. In conclusion our data demonstrates that PP causes a sustained decrease in both appetite and food intake.
- 5Schmidt, P. T.; Näslund, E.; Grybäck, P.; Jacobsson, H.; Holst, J. J.; Hilsted, L.; Hellström, P. M. A role for pancreatic polypeptide in the regulation of gastric emptying and short-term metabolic control. J. Clin. Endocrinol. Metab. 2005, 90, 5241– 5246, DOI: 10.1210/jc.2004-2089Google Scholar5A role for pancreatic polypeptide in the regulation of gastric emptying and short-term metabolic controlSchmidt, P. T.; Naeslund, E.; Grybaeck, P.; Jacobsson, H.; Holst, J. J.; Hilsted, L.; Hellstroem, P. M.Journal of Clinical Endocrinology and Metabolism (2005), 90 (9), 5241-5246CODEN: JCEMAZ; ISSN:0021-972X. (Endocrine Society)Context: Previous studies using pancreatic polypeptide (PP) infusions in humans have failed to show an effect on gastric emptying, glucose metab., and insulin secretion. This might be due to the use of nonhuman sequences of the peptide. Objective: The objective of this study was to use synthetic human PP to study gastric emptying rates of a solid meal and postprandial hormone secretion and glucose disposal as well as the gastric emptying rate of water. Design: This was a single-blind study. Setting: The study was performed at a university hospital. Participants: Fourteen healthy adult subjects were studied. Interventions: Infusion of saline or PP at 0.75 or 2.25 pmol/kg·min was given to eight subjects (gastric emptying of solid food), and infusion of saline or PP at 2.25 pmol/kg·min was given to six subjects (gastric emptying of water). Main Outcome Measures: The main outcome measures were gastric emptying of solids (scintigraphy), hunger ratings (visual analog scale), and plasma concns. of PP, insulin, glucagon, somatostatin, glucagon-like peptide 1, glucose, and gastric emptying of plain water (scintigraphy). Results: PP prolonged the lag phase and the half-time of emptying of the solid meal. The change in hunger rating, satiety, desire to eat after the meal, or prospective consumption was not affected. The postprandial rise in plasma glucose was prolonged by PP. The postprandial rise in insulin was also delayed by PP. PP had no significant effect on the emptying of water. Conclusions: PP inhibits gastric emptying of solid food and delays the postprandial rise in plasma glucose and insulin. PP is suggested to have a physiol. role in the pancreatic postprandial counter-regulation of gastric emptying and insulin secretion.
- 6Painsipp, E.; Wultsch, T.; Edelsbrunner, M. E.; Tasan, R. O.; Singewald, N.; Herzog, H.; Holzer, P. Reduced anxiety-like and depression-related behavior in neuropeptide Y Y4 receptor knockout mice. Genes, Brain Behav. 2008, 7, 532– 542, DOI: 10.1111/j.1601-183X.2008.00389.xGoogle Scholar6Reduced anxiety-like and depression-related behavior in neuropeptide Y Y4 receptor knockout micePainsipp E; Wultsch T; Edelsbrunner M E; Tasan R O; Singewald N; Herzog H; Holzer PGenes, brain, and behavior (2008), 7 (5), 532-42 ISSN:.Neuropeptide Y (NPY) acting through Y1 receptors reduces anxiety- and depression-like behavior in rodents, whereas Y2 receptor stimulation has the opposite effect. This study addressed the implication of Y4 receptors in emotional behavior by comparing female germ line Y4 knockout (Y4-/-) mice with control and germ line Y2-/- animals. Anxiety- and depression-like behavior was assessed with the open field (OF), elevated plus maze (EPM), stress-induced hyperthermia (SIH) and tail suspension tests (TST), respectively. Learning and memory were evaluated with the object recognition test (ORT). In the OF and EPM, both Y4-/- and Y2-/- mice exhibited reduced anxiety-related behavior and enhanced locomotor activity relative to control animals. Locomotor activity in a familiar environment was unchanged in Y4-/- but reduced in Y2-/- mice. The basal rectal temperature exhibited diurnal and genotype-related alterations. Control mice had temperature minima at noon and midnight, whereas Y4-/- and Y2-/- mice displayed only one temperature minimum at noon. The magnitude of SIH was related to time of the day and genotype in a complex manner. In the TST, the duration of immobility was significantly shorter in Y4-/- and Y2-/- mice than in controls. Object memory 6 h after initial exposure to the ORT was impaired in Y2-/- but not in Y4-/- mice, relative to control mice. These results show that genetic deletion of Y4 receptors, like that of Y2 receptors, reduces anxiety-like and depression-related behavior. Unlike Y2 receptor knockout, Y4 receptor knockout does not impair object memory. We propose that Y4 receptors play an important role in the regulation of behavioral homeostasis.
- 7Lin, S.; Shi, Y.-C.; Yulyaningsih, E.; Aljanova, A.; Zhang, L.; Macia, L.; Nguyen, A. D.; Lin, E.-J. D.; During, M. J.; Herzog, H.; Sainsbury, A. Critical role of arcuate Y4 receptors and the melanocortin system in pancreatic polypeptide-induced reduction in food intake in mice. PLoS One 2009, 4, e8488 DOI: 10.1371/journal.pone.0008488Google ScholarThere is no corresponding record for this reference.
- 8Tasan, R. O.; Lin, S.; Hetzenauer, A.; Singewald, N.; Herzog, H.; Sperk, G. Increased novelty-induced motor activity and reduced depression-like behavior in neuropeptide Y (NPY)-Y4 receptor knockout mice. Neuroscience 2009, 158, 1717– 1730, DOI: 10.1016/j.neuroscience.2008.11.048Google ScholarThere is no corresponding record for this reference.
- 9Sainsbury, A.; Shi, Y. C.; Zhang, L.; Aljanova, A.; Lin, Z.; Nguyen, A. D.; Herzog, H.; Lin, S. Y4 receptors and pancreatic polypeptide regulate food intake via hypothalamic orexin and brain-derived neurotropic factor dependent pathways. Neuropeptides 2010, 44, 261– 268, DOI: 10.1016/j.npep.2010.01.001Google Scholar9Y4 receptors and pancreatic polypeptide regulate food intake via hypothalamic orexin and brain-derived neurotropic factor dependent pathwaysSainsbury, Amanda; Shi, Yan-Chuan; Zhang, Lei; Aljanova, Aygul; Lin, Zhou; Nguyen, Amy D.; Herzog, Herbert; Lin, ShuNeuropeptides (Oxford, United Kingdom) (2010), 44 (3), 261-268CODEN: NRPPDD; ISSN:0143-4179. (Elsevier Ltd.)Gut-derived peptides are known to regulate food intake by activating specific receptors in the brain, but the target nuclei and neurons influenced are largely unknown. Here we show that peripherally administered pancreatic polypeptide (PP) stimulates neurons in key nuclei of the hypothalamus crit. for appetite and satiety regulation. In the lateral hypothalamic area (LHA), also known as the feeding center, neurons expressing the orexigenic neuropeptide orexin colocalize with the early neuronal activation marker c-Fos upon i.p. injection of PP into mice. In the ventromedial hypothalamus (VMH), also known as the satiety center, neurons activated by PP, as indicated by induction of c-Fos immunoreactivity, express the anorexigenic brain-derived neurotrophic factor (BDNF). Activation of neurons in the LHA and VMH in response to PP occurs via a Y4 receptor-dependent process as it is not seen in Y4 receptor knockout mice. We further demonstrate that in response to i.p. PP, orexin mRNA expression in the LHA is down-regulated, with Y4 receptors being crit. for this effect as it is not seen in Y4 receptor knockout mice, whereas BDNF mRNA expression is up-regulated in the VMH in response to i.p. PP in the fasted, but not in the non-fasted state. Taken together these data suggest that PP can regulate food intake by suppressing orexigenic pathways by down-regulation of orexin and simultaneously increasing anorexigenic pathways by up-regulating BDNF.
- 10Li, J. B.; Asakawa, A.; Terashi, M.; Cheng, K.; Chaolu, H.; Zoshiki, T.; Ushikai, M.; Sheriff, S.; Balasubramaniam, A.; Inui, A. Regulatory effects of Y4 receptor agonist (BVD-74D) on food intake. Peptides 2010, 31, 1706– 1710, DOI: 10.1016/j.peptides.2010.06.011Google Scholar10Regulatory effects of Y4 receptor agonist (BVD-74D) on food intakeLi, Jiang-Bo; Asakawa, Akihiro; Terashi, Mutsumi; Cheng, Kai-Chun; Chaolu, Huhe; Zoshiki, Takahiro; Ushikai, Miharu; Sheriff, Sulaiman; Balasubramaniam, Ambikaipakan; Inui, AkioPeptides (New York, NY, United States) (2010), 31 (9), 1706-1710CODEN: PPTDD5; ISSN:0196-9781. (Elsevier)The objective of this study was to clarify the role of a novel agonist with high selectivity and affinity for Y4 receptors in the regulation of food intake. The Y4 receptor agonist BVD-74D was administered to C57BL/6J mice that were fed with a normal or high-fat diet, and to fatty liver Shionogi (FLS)-ob/ob mice; the food intake, water intake, and body wt. gain were measured in these mice. In the mice fed with a normal diet, the cumulative food intake significantly decreased at 20 min and 1 h after the administration of 1 mg/kg of BVD-74D and at 1, 2, 4, 8, and 24 h after the administration of 10 mg/kg of BVD-74D. Moreover, the cumulative water intake and body wt. gain significantly decreased in these mice. Among the mice that were fed with a high-fat diet, the cumulative food intake and water intake significantly decreased 1, 2, and 4 h after BVD-74D (10 mg/kg) administration. Furthermore, the cumulative food intake significantly decreased 2 and 4 h after BVD-74D (10 mg/kg) administration in the FLS-ob/ob mice. Thus, we propose that the novel Y4 receptor agonist BVD-74D has suppressive effects on food intake, water intake, and wt. gain in normal mice fed with normal diets and on food intake in normal mice fed with high-fat diets and in FLS-ob/ob mice. These findings indicate that the Y4 receptor and its agonist would be promising targets for obesity.
- 11Moriya, R.; Fujikawa, T.; Ito, J.; Shirakura, T.; Hirose, H.; Suzuki, J.; Fukuroda, T.; MacNeil, D. J.; Kanatani, A. Pancreatic polypeptide enhances colonic muscle contraction and fecal output through neuropeptide Y Y4 receptor in mice. Eur. J. Pharmacol. 2010, 627, 258– 264, DOI: 10.1016/j.ejphar.2009.09.057Google ScholarThere is no corresponding record for this reference.
- 12Brothers, S. P.; Wahlestedt, C. Therapeutic potential of neuropeptide Y (NPY) receptor ligands. EMBO Mol. Med. 2010, 2, 429– 439, DOI: 10.1002/emmm.201000100Google Scholar12Therapeutic potential of neuropeptide Y (NPY) receptor ligandsBrothers, Shaun P.; Wahlestedt, ClaesEMBO Molecular Medicine (2010), 2 (11), 429-439CODEN: EMMMAM; ISSN:1757-4684. (Wiley-Blackwell)A review. Neuropeptide Y (NPY) is widely distributed in the human body and contributes to a vast no. of physiol. processes. Since its discovery, NPY has been implicated in metabolic regulation and, although interest in its role in central mechanisms related to food intake and obesity has somewhat diminished, the topic remains a strong focus of research concerning NPY signalling. In addn., a no. of other uses for modulators of NPY receptors have been implied in a range of diseases, although the development of NPY receptor ligands has been slow, with no clin. approved receptor therapeutics currently available. Nevertheless, several interesting small mol. compds., notably Y2 receptor antagonists, have been published recently, fueling optimism in the field. Herein we review the role of NPY in the pathophysiol. of a no. of diseases and highlight instances where NPY receptor signalling systems are attractive therapeutic targets.
- 13Verma, D.; Hörmer, B.; Bellmann-Sickert, K.; Thieme, V.; Beck-Sickinger, A. G.; Herzog, H.; Sperk, G.; Tasan, R. O. Pancreatic polypeptide and its central Y4 receptors are essential for cued fear extinction and permanent suppression of fear. Br. J. Pharmacol. 2016, 173, 1925– 1938, DOI: 10.1111/bph.13456Google Scholar13Pancreatic polypeptide and its central Y4 receptors are essential for cued fear extinction and permanent suppression of fearVerma, D.; Hoermer, B.; Bellmann-Sickert, K.; Thieme, V.; Beck-Sickinger, A. G.; Herzog, H.; Sperk, G.; Tasan, R. O.British Journal of Pharmacology (2016), 173 (12), 1925-1938CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)Background and purpose : Avoiding danger and finding food are closely related behaviors that are essential for surviving in a natural environment. Growing evidence supports an important role of gut-brain peptides in modulating energy homeostasis and emotional-affective behavior. For instance, postprandial release of pancreatic polypeptide (PP) reduced food intake and altered stress-induced motor activity and anxiety by activating central Y4 receptors. Exptl. approach : We characterized [K30(PEG2)]hPP2-36 as long-acting Y4 receptor agonist and injected it peripherally into wildtype and Y4 receptor knockout (Y4KO) C57Bl/6NCrl mice to investigate the role of Y4 receptors in fear conditioning. Extinction and relapse after extinction was measured by spontaneous recovery and renewal. Key results : The Y4KO mice showed impaired cued and context fear extinction without affecting acquisition, consolidation or recall of fear. Correspondingly, peripheral injection of [K30(PEG2)]hPP2-36 facilitated extinction learning upon fasting, an effect that was long-lasting and generalized. Furthermore, peripherally applied [K30(PEG2)]hPP2-36 before extinction inhibited the activation of orexin-expressing neurons in the lateral hypothalamus in WT, but not in Y4KO mice. Conclusions and implications : Our findings suggests suppression of excessive arousal as a possible mechanism for the extinction-promoting effect of central Y4 receptors and provide strong evidence that fear extinction requires integration of vegetative stimuli with cortical and subcortical information, a process crucially depending on Y4 receptors. Importantly, in the lateral hypothalamus two peptide systems, PP and orexin, interact to generate an emotional response adapted to the current homeostatic state. Detailed investigations of feeding-relevant genes may thus deliver multiple intervention points for treating anxiety-related disorders.
- 14Zhang, L.; Bijker, M. S.; Herzog, H. The neuropeptide Y system: Pathophysiological and therapeutic implications in obesity and cancer. Pharmacol. Ther. 2011, 131, 91– 113, DOI: 10.1016/j.pharmthera.2011.03.011Google Scholar14The neuropeptide Y system: Pathophysiological and therapeutic implications in obesity and cancerZhang, Lei; Bijker, Martijn S.; Herzog, HerbertPharmacology & Therapeutics (2011), 131 (1), 91-113CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)A review. The neuropeptide Y (NPY) system - comprising of neuropeptide Y, peptide YY, pancreatic polypeptide and the corresponding Y receptors through which they act (Y1, Y2, Y4, Y5 and y6) - is well known for its role in the regulation of energy homeostasis and assocd. processes. Dysfunctions of the system have been implicated in human diseases such as obesity and cancer, raising the possibility that correction of the system may provide therapeutic benefits for these diseases. In addn. to the regulation of appetite and satiety that has attracted most attention during the past years, insight has also been gained into the crit. role of NPY in the control of energy expenditure, oxidative fuel selection and bone metab. Studies using conditional knockout models further shed light on the central vs. peripheral, and hypothalamic vs. extra-hypothalamic mechanisms of these regulatory effects of NPY. Moreover, a role of NPY family peptides and Y receptors in modulating the growth of tumors has emerged. These findings provide the basis for novel NPY system-targeted strategies to treat obesity as well as cancer. Such strategies include modifying both sides of the energy balance equation - energy intake vs. energy expenditure - to achieve a greater wt./fat loss by particularly modulating peripheral Y receptor(s) to ameliorate metabolic conditions without interfering with central functions of Y receptors. In addn., targeting multiple Y receptors and/or multiple systems involved in the regulation of energy balance will have greater beneficial effects. However, long-term interference with the NPY system to target obesity or cancer related aspects needs to consider potential side effects on bone health.
- 15Yi, M.; Li, H.; Wu, Z.; Yan, J.; Liu, Q.; Ou, C.; Chen, M. A promising therapeutic target for metabolic diseases: Neuropeptide Y receptors in humans. Cell. Physiol. Biochem. 2018, 45, 88– 107, DOI: 10.1159/000486225Google Scholar15A Promising Therapeutic Target for Metabolic Diseases: Neuropeptide Y Receptors in HumansYi, Min; Li, Hekai; Wu, Zhiye; Yan, Jianyun; Liu, Qicai; Ou, Caiwen; Chen, MinshengCellular Physiology and Biochemistry (2018), 45 (1), 88-107CODEN: CEPBEW; ISSN:1015-8987. (S. Karger AG)Human neuropeptide Y (hNPY) is one of the most widely expressed neurotransmitters in the human central and peripheral nervous systems. It consists of 36 highly conserved amino acid residues, and was first isolated from the porcine hypothalamus in 1982. While it is the most recently discovered member of the pancreatic polypeptide family (which includes neuropeptide Y, gut-derived hormone peptide YY, and pancreatic polypeptide), NPY is the most abundant peptide found in the mammalian brain. In order to exert particular functions, NPY needs to bind to the NPY receptor to activate specific signaling pathways. NPY receptors belong to the class A or rhodopsin-like G-protein coupled receptor (GPCR) family and signal via cell-surface receptors. By binding to GPCRs, NPY plays a crucial role in various biol. processes, including cortical excitability, stress response, food intake, circadian rhythms, and cardiovascular function. Abnormal regulation of NPY is involved in the development of a wide range of diseases, including obesity, hypertension, atherosclerosis, epilepsy, metabolic disorders, and many cancers. Thus far, five receptors have been cloned from mammals (Y1, Y2, Y4, Y5, and y6), but only four of these (hY1, hY2, hY4, and hY5) are functional in humans. In this review, we summarize the structural characteristics of human NPY receptors and their role in metabolic diseases.
- 16Allen, M.; Reeves, J.; Mellor, G. High throughput fluorescence polarization: a homogeneous alternative to radioligand binding for cell surface receptors. J. Biomol. Screening 2000, 5, 63– 69, DOI: 10.1177/108705710000500202Google Scholar16High throughput fluorescence polarization: a homogeneous alternative to radioligand binding for cell surface receptorsAllen, Michael; Reeves, Julian; Mellor, GeoffreyJournal of Biomolecular Screening (2000), 5 (2), 63-69CODEN: JBISF3; ISSN:1087-0571. (Mary Ann Liebert, Inc.)High throughput fluorescence polarization (FP) assays are described that offer a nonradioactive, homogeneous, and low-cost alternative to radioligand binding assays for cell surface receptors (G protein-coupled receptors and ligand-gated ion channels). FP assays were shown to work across a range of both peptide (vasopressin V1a and δ-opioid) and nonpeptide (β1-adrenoceptor, 5-hydroxytryptamine3) receptors. Structure-activity relationships were investigated at β1-receptors and were found to be consistent with radioligand binding assays. FP was shown to tolerate up to 5% DMSO with no loss in sensitivity or signal window. From a random set of 1,280 compds., 1.9% were found to significantly interfere with FP measurement. If fluorescent or quenching compds. were eliminated (3% of all compds.), less than 0.4% of compds. were found to interfere with FP measurement. Assays could be run in 384-well plates with little loss of signal window or sensitivity compared to 96-well plate assays. New advances in FP measurement have therefore enabled FP to offer a high throughput alternative to radioligand binding for cell surface receptors.
- 17Leyris, J.-P.; Roux, T.; Trinquet, E.; Verdié, P.; Fehrentz, J.-A.; Oueslati, N.; Douzon, S.; Bourrier, E.; Lamarque, L.; Gagne, D. Homogeneous time-resolved fluorescence-based assay to screen for ligands targeting the growth hormone secretagogue receptor type 1a. Anal. Biochem. 2011, 408, 253– 262, DOI: 10.1016/j.ab.2010.09.030Google Scholar17Homogeneous time-resolved fluorescence-based assay to screen for ligands targeting the growth hormone secretagogue receptor type 1aLeyris, Jean-Philippe; Roux, Thomas; Trinquet, Eric; Verdie, Pascal; Fehrentz, Jean-Alain; Oueslati, Nadia; Douzon, Stephanie; Bourrier, Emmanuel; Lamarque, Laurent; Gagne, Didier; Galleyrand, Jean-Claude; M'kadmi, Celine; Martinez, Jean; Mary, Sophie; Baneres, Jean-Louis; Marie, JackyAnalytical Biochemistry (2011), 408 (2), 253-262CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)The growth hormone secretagogue receptor type 1a (GHS-R1a) belongs to class A G-protein-coupled receptors (GPCR). This receptor mediates pleiotropic effects of ghrelin and represents a promising target for dysfunctions of growth hormone secretion and energy homeostasis including obesity. Identification of new compds. which bind GHS-R1a is traditionally achieved using radioactive binding assays. Here we propose a new fluorescence-based assay, called Tag-lite binding assay, based on a fluorescence resonance energy transfer (FRET) process between a terbium cryptate covalently attached to a SNAP-tag fused GHS-R1a (SNAP-GHS-R1a) and a high-affinity red fluorescent ghrelin ligand. The long fluorescence lifetime of the terbium cryptate allows a time-resolved detection of the FRET signal. The assay was made compatible with high-throughput screening by using prelabeled cells in suspension under a 384-well plate format. Ki values for a panel of 14 compds. displaying agonist, antagonist, or inverse agonist properties were detd. using both the radioactive and the Tag-lite binding assays performed on the same batches of GHS-R1a-expressing cells. Compd. potencies obtained in the two assays were nicely correlated. This study is the first description of a sensitive and reliable nonradioactive binding assay for GHS-R1a in a format amenable to high-throughput screening.
- 18Emami-Nemini, A.; Roux, T.; Leblay, M.; Bourrier, E.; Lamarque, L.; Trinquet, E.; Lohse, M. J. Time-resolved fluorescence ligand binding for G protein–coupled receptors. Nat. Protoc. 2013, 8, 1307– 1320, DOI: 10.1038/nprot.2013.073Google Scholar18Time-resolved fluorescence ligand binding for G protein-coupled receptorsEmami-Nemini, Alexander; Roux, Thomas; Leblay, Marion; Bourrier, Emmanuel; Lamarque, Laurent; Trinquet, Eric; Lohse, Martin J.Nature Protocols (2013), 8 (7), 1307-1320, 14 pp.CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)G protein-coupled receptors (GPCRs) and their ligands are traditionally characterized by radioligand-binding expts. These expts. yield excellent quant. data, but have low temporal and spatial resoln. In addn., the use of radioligands presents safety concerns. Here we provide a general procedure for an alternative approach with high temporal and spatial resoln., based on Tb+-labeled fluorescent receptor ligands and time-resolved fluorescence resonance energy transfer (TR-FRET). This protocol and its design are detailed here for the parathyroid hormone receptor, a class B GPCR, and its fluorescently labeled 34-amino acid peptide ligand, but it can be easily modified for other receptors and their appropriately labeled ligands. We discuss three protocol options that use Tb+-labeled fluorescent ligands: a time-resolved fluorescence sepn. option that works on native receptors but requires sepn. of bound and unbound ligand; a TR-FRET option using SNAP-tag-labeled receptors for high-throughput screening; and a TR-FRET option that uses fluorescently labeled antibodies directed against an epitope engineered into the Flag-labeled receptors' N terminus. These protocol options can be used as std. procedures with very high signal-to-background ratios in order to characterize ligands and their receptors in living cells and in cell membranes via straightforward plate-reader measurements.
- 19Veiksina, S.; Kopanchuk, S.; Rinken, A. Budded baculoviruses as a tool for a homogeneous fluorescence anisotropy-based assay of ligand binding to G protein-coupled receptors: The case of melanocortin 4 receptors. Biochim. Biophys. Acta, Biomembr. 2014, 1838, 372– 381, DOI: 10.1016/j.bbamem.2013.09.015Google Scholar19Budded baculoviruses as a tool for a homogeneous fluorescence anisotropy-based assay of ligand binding to G protein-coupled receptors: The case of melanocortin 4 receptorsVeiksina, Santa; Kopanchuk, Sergei; Rinken, AgoBiochimica et Biophysica Acta, Biomembranes (2014), 1838 (1PB), 372-381CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)We present here the implementation of budded baculoviruses that display G protein-coupled receptors on their surfaces for the investigation of ligand-receptor interactions using fluorescence anisotropy (FA). Melanocortin 4 (MC4) receptors and the fluorescent ligand Cy3B-NDP-α-MSH were used as the model system. The real-time monitoring of reactions and the high assay quality allow the application of global data anal. with kinetic mechanistic models that take into account the effect of nonspecific interactions and the depletion of the fluorescent ligand during the reaction. The receptor concn., affinity and kinetic parameters of fluorescent ligand binding as well as state anisotropies for different fluorescent ligand populations were detd. At low Cy3B-NDP-α-MSH concns., a one-site receptor-ligand binding model described the processes, whereas divergence from this model was obsd. at higher ligand concns., which indicated a more complex mechanism of interactions similar to those mechanisms that have been found in expts. with radioactive ligands. The information obtained from our kinetic expts. and the inherent flexibility of FA assays also allowed the estn. of binding parameters for several MC4 receptor-specific unlabeled compds. In summary, the FA assay that was developed with budded baculoviruses led the exptl. data to a level that would solve complex models of receptor-ligand interactions also for other receptor systems and would become as a valuable tool for the screening of pharmacol. active compds.
- 20Schiele, F.; Ayaz, P.; Fernández-Montalván, A. A universal homogeneous assay for high-throughput determination of binding kinetics. Anal. Biochem. 2015, 468, 42– 49, DOI: 10.1016/j.ab.2014.09.007Google Scholar20A universal homogeneous assay for high-throughput determination of binding kineticsSchiele, Felix; Ayaz, Pelin; Fernandez-Montalvan, AmauryAnalytical Biochemistry (2015), 468 (), 42-49CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)There is an increasing demand for assay technologies that enable accurate, cost-effective, and high-throughput measurements of drug-target assocn. and dissocn. rates. Here the authors introduce a universal homogeneous kinetic probe competition assay (kPCA) that meets these requirements. The time-resolved fluorescence energy transfer (TR-FRET) procedure combines the versatility of radioligand binding assays with the advantages of homogeneous nonradioactive techniques while approaching the time resoln. of surface plasmon resonance (SPR) and related biosensors. The authors show application of kPCA for three important target classes: enzymes, protein-protein interactions, and G protein-coupled receptors (GPCRs). This method is capable of supporting early stages of drug discovery with large amts. of kinetic information.
- 21Antoine, T.; Ott, D.; Ebell, K.; Hansen, K.; Henry, L.; Becker, F.; Hannus, S. Homogeneous time-resolved G protein-coupled receptor-ligand binding assay based on fluorescence cross-correlation spectroscopy. Anal. Biochem. 2016, 502, 24– 35, DOI: 10.1016/j.ab.2016.02.017Google Scholar21Homogeneous time-resolved G protein-coupled receptor-ligand binding assay based on fluorescence cross-correlation spectroscopyAntoine, Thomas; Ott, David; Ebell, Katharina; Hansen, Kerrin; Henry, Luc; Becker, Frank; Hannus, StefanAnalytical Biochemistry (2016), 502 (), 24-35CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)G protein-coupled receptors (GPCRs) mediate many important physiol. functions and are considered as one of the most successful therapeutic target classes for a wide spectrum of diseases. Drug discovery projects generally benefit from a broad range of exptl. approaches for screening compd. libraries and for the characterization of binding modes of drug candidates. Owing to the difficulties in solubilizing and purifying GPCRs, assay formats have been so far mainly limited to cell-based functional assays and radioligand binding assays. In this study, we used fluorescence cross-correlation spectroscopy (FCCS) to analyze the interaction of detergent-solubilized receptors to various types of GPCR ligands: endogenous peptides, small mols., and a large surrogate antagonist represented by a blocking monoclonal antibody. Our work demonstrates the suitability of the homogeneous and time-resolved FCCS assay format for a robust, high-throughput detn. of receptor-ligand binding affinities and kinetic rate consts. for various therapeutically relevant GPCRs.
- 22Stoddart, L. A.; White, C. W.; Nguyen, K.; Hill, S. J.; Pfleger, K. D. Fluorescence- and bioluminescence-based approaches to study GPCR ligand binding. Br. J. Pharmacol. 2016, 173, 3028– 3037, DOI: 10.1111/bph.13316Google Scholar22Fluorescence- and bioluminescence-based approaches to study GPCR ligand bindingStoddart, Leigh A.; White, Carl W.; Nguyen, Kim; Hill, Stephen J.; Pfleger, Kevin D. G.British Journal of Pharmacology (2016), 173 (20), 3028-3037CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)Ligand binding is a vital component of any pharmacologist's toolbox and allows the detailed investigation of how a mol. binds to its receptor. These studies enable the exptl. detn. of binding affinity of labeled and unlabeled compds. through kinetic, satn. (Kd) and competition (Ki) binding assays. Traditionally, these studies have used mols. labeled with radioisotopes; however, more recently, fluorescent ligands have been developed for this purpose. This review will briefly cover receptor ligand binding theory and then discuss the use of fluorescent ligands with some of the different technologies currently employed to examine ligand binding. Fluorescent ligands can be used for direct measurement of receptor-assocd. fluorescence using confocal microscopy and flow cytometry as well as in assays such as fluorescence polarization, where ligand binding is monitored by changes in the free rotation when a fluorescent ligand is bound to a receptor. Addnl., fluorescent ligands can act as donors or acceptors for fluorescence resonance energy transfer (FRET) with the development of assays based on FRET and time-resolved FRET (TR-FRET). Finally, we have recently developed a novel bioluminescence resonance energy transfer (BRET) ligand binding assay utilizing a small (19 kDa), super-bright luciferase subunit (NanoLuc) from a deep sea shrimp. In combination with fluorescent ligands, measurement of RET now provides an array of methodologies to study ligand binding. While each method has its own advantages and drawbacks, binding studies using fluorescent ligands are now a viable alternative to the use of radioligands.
- 23Stoddart, L. A.; Kilpatrick, L. E.; Hill, S. J. NanoBRET approaches to study ligand binding to GPCRs and RTKs. Trends Pharmacol. Sci. 2018, 39, 136– 147, DOI: 10.1016/j.tips.2017.10.006Google Scholar23NanoBRET Approaches to Study Ligand Binding to GPCRs and RTKsStoddart, Leigh A.; Kilpatrick, Laura E.; Hill, Stephen J.Trends in Pharmacological Sciences (2018), 39 (2), 136-147CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)Recent advances in the development of fluorescent ligands for G-protein-coupled receptors (GPCRs) and receptor tyrosine kinase receptors (RTKs) have facilitated the study of these receptors in living cells. A limitation of these ligands is potential uptake into cells and increased nonspecific binding. However, this can largely be overcome by using proximity approaches, such as bioluminescence resonance energy transfer (BRET), which localize the signal (within 10 nm) to the specific receptor target. The recent engineering of NanoLuc has resulted in a luciferase variant that is smaller and significantly brighter (up to tenfold) than existing variants. Here, we review the use of BRET from N-terminal NanoLuc-tagged GPCRs or a RTK to a receptor-bound fluorescent ligand to provide quant. pharmacol. of ligand-receptor interactions in living cells in real time.
- 24Iliopoulos-Tsoutsouvas, C.; Kulkarni, R. N.; Makriyannis, A.; Nikas, S. P. Fluorescent probes for G-protein-coupled receptor drug discovery. Expert Opin. Drug Discovery 2018, 13, 933– 947, DOI: 10.1080/17460441.2018.1518975Google Scholar24Fluorescent probes for G-protein-coupled receptor drug discoveryIliopoulos-Tsoutsouvas, Christos; Kulkarni, Rohit N.; Makriyannis, Alexandros; Nikas, Spyros P.Expert Opinion on Drug Discovery (2018), 13 (10), 933-947CODEN: EODDBX; ISSN:1746-0441. (Taylor & Francis Ltd.)A review. G-protein-coupled receptors (GPCRs) mediate the effects of approx. 33% of all marketed drugs. The development of tools to study GPCR pharmacol. is urgently needed as it can lead to the discovery of safer and more effective medications. Fluorescent GPCR ligands represent highly sensitive and safe small-mol. tools for real-time exploration of the life of the receptor, cellular signaling, and ligand-/receptor-receptor interactions in cellulo and/or in vivo.: This review summarizes relevant information from published literature and provides crit. insights into the design of successful small-mol. fluorescent probes for Class A GPCRs as potential major targets for drug development.: Considering the rapid progress of fluorescence technologies, effective small-mol. fluorescent probes represent valuable pharmacol. tools for studying GPCRs. However, the design and development of such probes are challenging, largely due to the low affinity/specificity of the probe for its target, inadequate photophys. properties, extensive non-specific binding, and/or low signal-to-noise ratio. Generally speaking, fluorescent and luminescent small-mol. probes, receptors, and G proteins in combination with FRET and BRET technologies hold great promise for studying kinetic profiles of GPCR signaling.
- 25Ziemek, R.; Schneider, E.; Kraus, A.; Cabrele, C.; Beck-Sickinger, A. G.; Bernhardt, G.; Buschauer, A. Determination of affinity and activity of ligands at the human neuropeptide Y Y4 receptor by flow cytometry and aequorin luminescence. J. Recept. Signal Transduction 2007, 27, 217– 233, DOI: 10.1080/10799890701505206Google Scholar25Determination of Affinity and Activity of Ligands at the Human Neuropeptide Y Y4 Receptor by Flow Cytometry and Aequorin LuminescenceZiemek, Ralf; Schneider, Erich; Kraus, Anja; Cabrele, Chiara; Beck-Sickinger, Annette G.; Bernhardt, Guenther; Buschauer, ArminJournal of Receptors and Signal Transduction (2007), 27 (4), 217-233CODEN: JRSTCT ISSN:. (Informa Healthcare)Fluorescence-labeled neuropeptide Y (NPY) has been used in flow cytometric binding assays for the detn. of affinity consts. of NPY Y1, Y2, and Y5 receptor ligands. Because the binding of fluorescent NPY is insufficient for competition studies at the human Y4 receptor (hY4R), the authors replaced Glu 4 in hPP with Lys for the derivatization with cyanine-5. Because cy5-[K4]hPP has high affinity (Kd 5.6 nM) to the hY4R, it was used as a probe in a flow cytometric binding assay. Specific binding of cy5-[K4]hPP to hY4R was visualized by confocal microscopy. The hY4R, the chimeric G protein Gqi5 and mitochondrially targeted apoaequorin were stably coexpressed in CHO cells. Aequorin luminescence was quantified in a microplate reader and by a CCD camera. By application of these methods 3-cyclohexyl-N-[(3-1H-imidazol-4-ylpropylamino)(imino)methyl]propanamide (UR-AK49)was discovered as the first nonpeptidic Y4R antagonist (pKi 4.17), a lead to be optimized in terms of potency and selectivity.
- 26Dukorn, S.; Littmann, T.; Keller, M.; Kuhn, K.; Cabrele, C.; Baumeister, P.; Bernhardt, G.; Buschauer, A. Fluorescence- and radiolabeling of [Lys4,Nle17,30]hPP yields molecular tools for the NPY Y4 receptor. Bioconjugate Chem. 2017, 28, 1291– 1304, DOI: 10.1021/acs.bioconjchem.7b00103Google Scholar26Fluorescence- and Radiolabeling of [Lys4,Nle17,30]hPP Yields Molecular Tools for the NPY Y4 ReceptorDukorn, Stefanie; Littmann, Timo; Keller, Max; Kuhn, Kilian; Cabrele, Chiara; Baumeister, Paul; Bernhardt, Guenther; Buschauer, ArminBioconjugate Chemistry (2017), 28 (4), 1291-1304CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The neuropeptide Y (NPY) Y4 receptor (Y4R) is involved in energy homeostasis and considered a potential drug target for the treatment of obesity. Only a few mol. tools, i.e., radiolabeled and fluorescent ligands, for the investigation of the Y4R were reported. Previously, [Lys4]hPP proved to be an appropriate full-length PP analog to prep. a fluorescent ligand by derivatization at the ε-amino group. To preclude oxidn. upon long-term storage, the authors replaced the two methionine residues in [Lys4]hPP by norleucine and prepd. the corresponding [3H]propionylated ([3H]12) and cyanine labeled (13) peptides, which were characterized and compared with a set of ref. compds. in binding (Y1, Y2, Y4, and Y5 receptors) and functional (luciferase gene reporter, beta-arrestin-1,2) Y4R assays. Both mol. probes proved to be useful in radiochem. and flow cytometric satn. and competition Y4R binding expts. Most strikingly, there was a different influence of the compn. of buffer on equil. binding and kinetics: [3H]12 affinity (Kd in Na+-free buffer: 1.1 nM) clearly decreased with increasing sodium ion concn., whereas dissocn. and Y4R-mediated internalization of 13 (Kd in Na+-free buffer: 10.8 nM) were strongly affected by the osmolarity of the buffer as demonstrated by confocal microscopy. Displacement of [3H]12 and 13 revealed a tendency to higher apparent affinities for a set of ref. peptides in hypotonic (Na+-free) compared to isotonic buffers. The differences were negligible in the case of hPP but up to 270-fold in the case of GW1229 (GR231118). By contrast, no relevant influence of Na+ on Y5R affinity became obvious, when the radioligands [H]12 and [3H]propionyl-pNPY were investigated in satn. binding and competition binding.
- 27Tang, T.; Tan, Q.; Han, S.; Diemar, A.; Löbner, K.; Wang, H.; Schüß, C.; Behr, V.; Mörl, K.; Wang, M.; Chu, X.; Yi, C.; Keller, M.; Kofoed, J.; Reedtz-Runge, S.; Kaiser, A.; Beck-Sickinger, A. G.; Zhao, Q.; Wu, B. Receptor-specific recognition of NPY peptides revealed by structures of NPY receptors. Sci. Adv. 2022, 8, eabm1232 DOI: 10.1126/sciadv.abm1232Google ScholarThere is no corresponding record for this reference.
- 28Kuhn, K. K.; Littmann, T.; Dukorn, S.; Tanaka, M.; Keller, M.; Ozawa, T.; Bernhardt, G.; Buschauer, A. In search of NPY Y4R antagonists: Incorporation of carbamoylated arginine, aza-amino acids, or D-amino acids into oligopeptides derived from the C-termini of the endogenous agonists. ACS Omega 2017, 2, 3616– 3631, DOI: 10.1021/acsomega.7b00451Google Scholar28In Search of NPY Y4R Antagonists: Incorporation of Carbamoylated Arginine, Aza-Amino Acids, or D-Amino Acids into Oligopeptides Derived from the C-Termini of the Endogenous AgonistsKuhn, Kilian K.; Littmann, Timo; Dukorn, Stefanie; Tanaka, Miho; Keller, Max; Ozawa, Takeaki; Bernhardt, Guenther; Buschauer, ArminACS Omega (2017), 2 (7), 3616-3631CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)The crosslinked pentapeptides (2R,7R)-diaminooctanedioyl-bis(Tyr-Arg-Leu-Arg-Tyr-amide) ((2R,7R)-BVD-74D, (2R,7R)-1) and octanedioyl-bis(Tyr-Arg-Leu-Arg-Tyr-amide) (2) as well as the pentapeptide Ac-Tyr-Arg-Leu-Arg-Tyr-amide (3) were previously described as neuropeptide Y Y4 receptor (Y4R) partial agonists. Here the authors report on a series of analogs of (2R,7R)-1 and 2 in which Arg2, Leu3 or Arg4 were replaced by the resp. aza-amino acids. The replacement of Arg2 in 3 with a carbamoylated arginine building block and the extension of the N-terminus by an addnl. arginine led to the high-affinity hexapeptide Ac-Arg-Tyr-Nω-[(4-aminobutyl)aminocarbonyl]Arg-Leu-Arg-Tyr-amide that was used as a precursor for a D-amino acid scan. The target compds. were investigated for Y4R functional activity in assays with complementary readouts: aequorin Ca2+ and β-arrestin 1 or β-arrestin 2 assays. In contrast to the parent compds., which are Y4R agonists, several ligands were able to suppress the effect elicited by the endogenous ligand pancreatic polypeptide and therefore represent a novel class of peptide Y4R antagonists.
- 29Spinnler, K.; von Krüchten, L.; Konieczny, A.; Schindler, L.; Bernhardt, G.; Keller, M. An alkyne-functionalized arginine for solid-phase synthesis enabling “bioorthogonal” peptide conjugation. ACS Med. Chem. Lett. 2020, 11, 334– 339, DOI: 10.1021/acsmedchemlett.9b00388Google Scholar29An alkyne-functionalized arginine for solid-phase synthesis enabling "bioorthogonal" peptide conjugationSpinnler, Katrin; von Kruechten, Lara; Konieczny, Adam; Schindler, Lisa; Bernhardt, Guenther; Keller, MaxACS Medicinal Chemistry Letters (2020), 11 (3), 334-339CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Lately, amino-functionalized Nω-carbamoylated arginines were introduced as arginine surrogates enabling peptide labeling. However, this approach is hardly compatible with peptides also contg. lysine or cysteine. Here, we present the synthesis of an alkyne-functionalized, Nω-carbamoylated arginine building block, which is compatible with Fmoc-strategy solid-phase peptide synthesis. The alkynylated arginine was incorporated into three biol. active linear hexapeptides and into a cyclic pentapeptide. Peptide conjugation to an azido-functionalized fluorescent dye via "click" chem. was successfully demonstrated. In the case of a peptide also contg. lysine besides the alkyne-functionalized arginine, this was feasible in a "bioorthogonal" manner.
- 30Konieczny, A.; Conrad, M.; Ertl, F. J.; Gleixner, J.; Gattor, A. O.; Grätz, L.; Schmidt, M. F.; Neu, E.; Horn, A. H. C.; Wifling, D.; Gmeiner, P.; Clark, T.; Sticht, H.; Keller, M. N-terminus to arginine side-chain cyclization of linear peptidic neuropeptide Y Y4 receptor ligands results in picomolar binding constants. J. Med. Chem. 2021, 64, 16746– 16769, DOI: 10.1021/acs.jmedchem.1c01574Google Scholar30N-Terminus to arginine side-chain cyclization of linear peptidic neuropeptide Y Y4 receptor ligands results in picomolar binding constantsKonieczny, Adam; Conrad, Marcus; Ertl, Fabian J.; Gleixner, Jakob; Gattor, Albert O.; Graetz, Lukas; Schmidt, Maximilian F.; Neu, Eduard; Horn, Anselm H. C.; Wifling, David; Gmeiner, Peter; Clark, Timothy; Sticht, Heinrich; Keller, MaxJournal of Medicinal Chemistry (2021), 64 (22), 16746-16769CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The family of neuropeptide Y (NPY) receptors comprises four subtypes (Y1R, Y2R, Y4R, Y5R), which are addressed by at least three endogenous peptides, i.e., NPY, peptide YY, and pancreatic polypeptide (PP), the latter showing a preference for Y4R. A series of cyclic oligopeptidic Y4R ligands were prepd. by applying a novel approach, i.e., N-terminus to arginine side-chain cyclization. Most peptides acted as Y4R partial agonists, showing up to 60-fold higher Y4R affinity compared to the linear precursor peptides. Two cyclic hexapeptides (I) [cyclo[succinyl-Arg-Tyr-Arg(CO-NH-(CH2)4-NH)]-Leu-Arg-Tyr-NH2] and (II) [cyclo[succinyl-Arg-Trp-Arg(CO-NH-(CH2)4-NH)]-Leu-Arg-Tyr-NH2] showed higher Y4R potency (Ca2+ aequorin assay) and, with pKi values >10, also higher Y4R affinity compared to human pancreatic polypeptide (hPP). Compds. such as I and II exhibiting considerably lower mol. wt. and considerably more pronounced Y4R selectivity than PP and previously described dimeric peptidic ligands with high Y4R affinity, represent promising leads for the prepn. of labeled tool compds. and might support the development of drug-like Y4R ligands.
- 31Gleixner, J.; Gattor, A. O.; Humphrys, L. J.; Brunner, T.; Keller, M. 3H]UR-JG102 - a radiolabeled cyclic peptide with high affinity and excellent selectivity for the neuropeptide Y Y4 receptor. J. Med. Chem. 2023, 66, 13788– 13808, DOI: 10.1021/acs.jmedchem.3c01224Google ScholarThere is no corresponding record for this reference.
- 32Keller, M.; Kuhn, K. K.; Einsiedel, J.; Hübner, H.; Biselli, S.; Mollereau, C.; Wifling, D.; Svobodová, J.; Bernhardt, G.; Cabrele, C.; Vanderheyden, P. M. L.; Gmeiner, P.; Buschauer, A. Mimicking of arginine by functionalized Nω-carbamoylated arginine as a new broadly applicable approach to labeled bioactive peptides: high affinity angiotensin, neuropeptide Y, neuropeptide FF, and neurotensin receptor ligands as examples. J. Med. Chem. 2016, 59, 1925– 1945, DOI: 10.1021/acs.jmedchem.5b01495Google Scholar32Mimicking of Arginine by Functionalized Nω-Carbamoylated Arginine As a New Broadly Applicable Approach to Labeled Bioactive Peptides: High Affinity Angiotensin, Neuropeptide Y, Neuropeptide FF, and Neurotensin Receptor Ligands As ExamplesKeller, Max; Kuhn, Kilian K.; Einsiedel, Juergen; Huebner, Harald; Biselli, Sabrina; Mollereau, Catherine; Wifling, David; Svobodova, Jaroslava; Bernhardt, Guenther; Cabrele, Chiara; Vanderheyden, Patrick M. L.; Gmeiner, Peter; Buschauer, ArminJournal of Medicinal Chemistry (2016), 59 (5), 1925-1945CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Derivatization of biol. active peptides by conjugation with fluorophores or radionuclide-bearing moieties is an effective and commonly used approach to prep. mol. tools and diagnostic agents. Whereas lysine, cysteine, and N-terminal amino acids have been mostly used for peptide conjugation, we describe a new, widely applicable approach to peptide conjugation based on the nonclassical bioisosteric replacement of the guanidine group in arginine by a functionalized carbamoylguanidine moiety. Four arginine-contg. peptide receptor ligands (angiotensin II, neurotensin(8-13), an analog of the C-terminal pentapeptide of neuropeptide Y, and a neuropeptide FF analog) were subject of this proof-of-concept study. The Nω-carbamoylated arginines, bearing spacers with a terminal amino group, were incorporated into the peptides by std. Fmoc solid phase peptide synthesis. The synthesized chem. stable peptide derivs. showed high receptor affinities with Ki values in the low nanomolar range, even when bulky fluorophores had been attached. Two new tritiated tracers for angiotensin and neurotensin receptors are described.
- 33Keller, M.; Weiss, S.; Hutzler, C.; Kuhn, K. K.; M?llereau, C.; Dukorn, S.; Schindler, L.; Bernhardt, G.; König, B.; Buschauer, A. Nω-carbamoylation of the argininamide moiety: An avenue to insurmountable NPY Y1 receptor antagonists and a radiolabeled selective high-affinity molecular tool ([3H]UR-MK299) with extended residence time. J. Med. Chem. 2015, 58, 8834– 8849, DOI: 10.1021/acs.jmedchem.5b00925Google Scholar33Nω-Carbamoylation of the Argininamide Moiety: An Avenue to Insurmountable NPY Y1 Receptor Antagonists and a Radiolabeled Selective High-Affinity Molecular Tool ([3H]UR-MK299) with Extended Residence TimeKeller, Max; Weiss, Stefan; Hutzler, Christoph; Kuhn, Kilian K.; Mollereau, Catherine; Dukorn, Stefanie; Schindler, Lisa; Bernhardt, Guenther; Koenig, Burkhard; Buschauer, ArminJournal of Medicinal Chemistry (2015), 58 (22), 8834-8849CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Analogs of the argininamide-type NPY Y1 receptor (Y1R) antagonist BIBP3226, bearing carbamoyl moieties at the guanidine group, revealed subnanomolar Ki values and caused depression of the maximal response to NPY (calcium assay) by up to 90% in a concn.- and time-dependent manner, suggesting insurmountable antagonism. To gain insight into the mechanism of binding of the synthesized compds., a tritiated antagonist, (R)-Nα-diphenylacetyl-Nω-[2-([2,3-3H]propionylamino)ethyl]aminocarbonyl-(4-hydroxybenzyl)arginin-amide ([3H]UR-MK299, [3H]38), was prepd. [3H]38 revealed a dissocn. const. in the picomolar range (Kd 0.044 nM, SK-N-MC cells) and very high Y1R selectivity. Apart from superior affinity, a considerably lower target off-rate (t1/2 95 min) was characteristic of [3H]38 compared to that of the higher homolog contg. a tetramethylene instead of an ethylene spacer (t1/2 3 min, Kd 2.0 nM). Y1R binding of [3H]38 was fully reversible and fully displaceable by nonpeptide antagonists and the agonist pNPY. Therefore, the insurmountable antagonism obsd. in the functional assay has to be attributed to the extended target-residence time, a phenomenon of relevance in drug research beyond the NPY receptor field.
- 34Konieczny, A.; Braun, D.; Wifling, D.; Bernhardt, G.; Keller, M. Oligopeptides as neuropeptide Y Y4 receptor ligands: identification of a high-affinity tetrapeptide agonist and a hexapeptide antagonist. J. Med. Chem. 2020, 63, 8198– 8215, DOI: 10.1021/acs.jmedchem.0c00426Google Scholar34Oligopeptides as Neuropeptide Y Y4 Receptor Ligands: Identification of a High-Affinity Tetrapeptide Agonist and a Hexapeptide AntagonistKonieczny, Adam; Braun, Diana; Wifling, David; Bernhardt, Guenther; Keller, MaxJournal of Medicinal Chemistry (2020), 63 (15), 8198-8215CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Within the family of neuropeptide Y (NPY) receptors, the Y4 receptor (Y4R) is unique as it prefers pancreatic polypeptide (PP) over NPY and peptide YY (PYY). Today, low mol. wt. Y4R ligands are lacking, in particular antagonists. We synthesized a series of peptidic NPY Y4R ligands, derived from the hexapeptide acetyl-Arg-Tyr-Arg-Leu-Arg-Tyr-NH2 (1), reported to be a Y4R partial agonist with high affinity (pKi Y4R: 8.43). Peptide 1 was N-terminally extended as well as truncated, subjected to a D-amino acid scan, and Leu was replaced by different amino acids. Compds. were characterized by radioligand competition binding and functional studies (Cai2+ mobilization and β-arrestin 1/2 recruitment). N-terminal truncation of 1 resulted in a tetrapeptide (Arg-Leu-Arg-Tyr-NH2) being a Y4R partial agonist with retained Y4R affinity (pKi: 8.47). Remarkably, replacement of Leu in 1 and in derivs. of 1 by Trp turned Y4R agonism to antagonism, giving Y4R antagonists with pKi values ≤ 7.57.
- 35Kuhn, K. K.; Ertl, T.; Dukorn, S.; Keller, M.; Bernhardt, G.; Reiser, O.; Buschauer, A. High affinity agonists of the neuropeptide Y (NPY) Y4 receptor derived from the C-terminal pentapeptide of human pancreatic polypeptide (hPP): Synthesis, stereochemical discrimination, and radiolabeling. J. Med. Chem. 2016, 59, 6045– 6058, DOI: 10.1021/acs.jmedchem.6b00309Google Scholar35High Affinity Agonists of the Neuropeptide Y (NPY) Y4 Receptor Derived from the C-Terminal Pentapeptide of Human Pancreatic Polypeptide (hPP): Synthesis, Stereochemical Discrimination, and RadiolabelingKuhn, Kilian K.; Ertl, Thomas; Dukorn, Stefanie; Keller, Max; Bernhardt, Guenther; Reiser, Oliver; Buschauer, ArminJournal of Medicinal Chemistry (2016), 59 (13), 6045-6058CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The diastereomeric mixt. of D/L-2,7-diaminooctanedioyl-bis(YRLRY-NH2) (BVD-74D, 2) was described in the literature as a high affinity Y4 receptor agonist. Here we report on the synthesis and pharmacol. characterization of the pure diastereomers (2R,7R)- and (2S,7S)-2 and a series of homo- and heterodimeric analogs in which octanedioic acid was used as an achiral linker. To investigate the role of the Arg residues, one or two arginines were replaced by Ala. Moreover, Nω-(6-aminohexylaminocarbonyl)Arg was introduced as an arginine replacement (17). (2R,7R)-2 was superior to (2S,7S)-2 in binding and functional cellular assays and equipotent with 17. [3H]Propionylation of one amino group in the linker of (2R,7R)-2 or at the primary amino group in 17 resulted in high affinity Y4R radioligands ([3H]-(2R,7R)-10, [3H]18) with subnanomolar Kd values.
- 36Berlicki, Ł.; Kaske, M.; Gutiérrez-Abad, R.; Bernhardt, G.; Illa, O.; Ortuño, R. M.; Cabrele, C.; Buschauer, A.; Reiser, O. Replacement of Thr32 and Gln34 in the C-terminal neuropeptide Y fragment 25–36 by cis-cyclobutane and cis-cyclopentane β-amino acids shifts selectivity toward the Y4 receptor. J. Med. Chem. 2013, 56, 8422– 8431, DOI: 10.1021/jm4008505Google Scholar36Replacement of Thr32 and Gln34 in the C-Terminal Neuropeptide Y Fragment 25-36 by cis-Cyclobutane and cis-Cyclopentane β Amino Acids Shifts Selectivity toward the Y4 ReceptorBerlicki, Lukasz; Kaske, Melanie; Gutierrez-Abad, Raquel; Bernhardt, Guenther; Illa, Ona; Ortuno, Rosa M.; Cabrele, Chiara; Buschauer, Armin; Reiser, OliverJournal of Medicinal Chemistry (2013), 56 (21), 8422-8431CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Neuropeptide Y (NPY) and pancreatic polypeptide (PUP) control central and peripheral processes by activating the G protein coupled receptors YxR (x = 1, 2, 4, 5). We present analogs of the C-terminal fragments 25-36 and 32-36 of NPY and PP contg. (1R,2S)-cyclobutane (βCbu) or (1R,2S)-cyclopentane (βCpe) β-amino acids, which display exclusively Y4R affinity. In particular, [βCpe34]-NPY-(25-36) is a Y4R selective partial agonist (EC50 41 nM, Emax 71%) that binds Y4R with a Ki of 10 nM and a selectivity >100-fold relative to Y1R and Y2R and >50-fold relative to Y5R. Comparably, [Y32, βCpe34]-NPY-(PP)-(32-36) selectively binds and activates Y4R (EC50 94 nM, Emax 73%). The NMR structure of [βCpe34]-NPY-(25-36) in dodecylphosphatidylcholine micelles shows a short helix at residues 27-32, while the C-terminal segment R33βCpe34R35Y36 is extended. The biol. properties of the βCbu- or βCpe-contg. NPY and PP C-terminal fragments encourage the future application of these β-amino acids in the synthesis of selective Y4R ligands.
- 37Wirth, U.; Erl, J.; Azzam, S.; Höring, C.; Skiba, M.; Singh, R.; Hochmuth, K.; Keller, M.; Wegener, J.; König, B. Monitoring the reversibility of GPCR signaling by combining photochromic ligands with label-free impedance analysis. Angew. Chem., Int. Ed. 2023, 62, e202215547 DOI: 10.1002/anie.202215547Google Scholar37Monitoring the Reversibility of GPCR Signaling by Combining Photochromic Ligands with Label-free Impedance AnalysisWirth, Ulrike; Erl, Julia; Azzam, Saphia; Horing, Carina; Skiba, Michael; Singh, Ritu; Hochmuth, Kathrin; Keller, Max; Wegener, Joachim; Konig, BurkhardAngewandte Chemie, International Edition (2023), 62 (21), e202215547CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)G protein-coupled cell surface receptors (GPCR) trigger complex intracellular signaling cascades upon agonist binding. Classic pharmacol. assays provide information about binding affinities, activation or blockade at different stages of the signaling cascade, but real time dynamics and reversibility of these processes remain often disguised. We show that combining photochromic NPY receptor ligands, which can be toggled in their receptor activation ability by irradn. with light of different wavelengths, with whole cell label-free impedance assays allows observing the cell response to receptor activation and its reversibility over time. The concept demonstrated on NPY receptors may be well applicable to many other GPCRs providing a deeper insight into the time course of intracellular signaling processes.
- 38Archipowa, N.; Wittmann, L.; Köckenberger, J.; Ertl, F. J.; Gleixner, J.; Keller, M.; Heinrich, M. R.; Kutta, R. J. Characterization of fluorescent dyes frequently used for bioimaging: Photophysics and photocatalytical reactions with proteins. J. Phys. Chem. B 2023, 127, 9532– 9542, DOI: 10.1021/acs.jpcb.3c04484Google ScholarThere is no corresponding record for this reference.
- 39Keller, M.; Mahuroof, S. A.; Yee, V. H.; Carpenter, J.; Schindler, L.; Littmann, T.; Pegoli, A.; Hübner, H.; Bernhardt, G.; Gmeiner, P.; Holliday, N. D. Fluorescence labeling of neurotensin(8–13) via arginine residues gives molecular tools with high receptor affinity. ACS Med. Chem. Lett. 2020, 11, 16– 22, DOI: 10.1021/acsmedchemlett.9b00462Google Scholar39Fluorescence labeling of neurotensin(8-13) via arginine residues gives molecular tools with high receptor affinityKeller, Max; Mahuroof, Shahani A.; Hong Yee, Vivyanne; Carpenter, Jessica; Schindler, Lisa; Littmann, Timo; Pegoli, Andrea; Huebner, Harald; Bernhardt, Guenther; Gmeiner, Peter; Holliday, Nicholas D.ACS Medicinal Chemistry Letters (2020), 11 (1), 16-22CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Fluorescence-labeled receptor ligands have emerged as valuable mol. tools, being indispensable for studying receptor-ligand interactions by fluorescence-based techniques such as high-content imaging, fluorescence microscopy, and fluorescence polarization. Through application of a new labeling strategy for peptides, a series of fluorescent neurotensin(8-13) derivs. was synthesized by attaching red-emitting fluorophores (indolinium- and pyridinium-type cyanine dyes) to carbamoylated arginine residues in neurotensin(8-13) analogs, yielding fluorescent probes with high NTS1R affinity (pKi values: 8.15-9.12) and potency (pEC50 values (Ca2+ mobilization): 8.23-9.43). Selected fluorescent ligands were investigated by flow cytometry and high-content imaging (satn. binding, kinetic studies, and competition binding) as well as by confocal microscopy using intact CHO-hNTS1R cells. The study demonstrates the applicability of the fluorescent probes as mol. tools to obtain, for example, information about the localization of receptors in cells and to det. binding affinities of nonlabeled ligands.
- 40Müller, C.; Gleixner, J.; Tahk, M.-J.; Kopanchuk, S.; Laasfeld, T.; Weinhart, M.; Schollmeyer, D.; Betschart, M. U.; Lüdeke, S.; Koch, P.; Rinken, A.; Keller, M. Structure-based design of high-affinity fluorescent probes for the neuropeptide Y Y1 receptor. J. Med. Chem. 2022, 65, 4832– 4853, DOI: 10.1021/acs.jmedchem.1c02033Google Scholar40Structure-based design of high-affinity fluorescent probes for the neuropeptide Y Y1 receptorMueller, Christoph; Gleixner, Jakob; Tahk, Maris-Johanna; Kopanchuk, Sergei; Laasfeld, Tonis; Weinhart, Michael; Schollmeyer, Dieter; Betschart, Martin U.; Luedeke, Steffen; Koch, Pierre; Rinken, Ago; Keller, MaxJournal of Medicinal Chemistry (2022), 65 (6), 4832-4853CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent crystn. of the neuropeptide Y Y1 receptor (Y1R) in complex with the argininamide-type Y1R selective antagonist UR-MK299 (2) opened up a new approach toward structure-based design of nonpeptidic Y1R ligands. We designed novel fluorescent probes showing excellent Y1R selectivity and, in contrast to previously described fluorescent Y1R ligands, considerably higher (~ 100-fold) binding affinity. This was achieved through the attachment of different fluorescent dyes to the diphenylacetyl moiety in 2 via an amine-functionalized linker. The fluorescent ligands exhibited picomolar Y1R binding affinities (pKi values of 9.36-9.95) and proved to be Y1R antagonists, as validated in a Fura-2 calcium assay. The versatile applicability of the probes as tool compds. was demonstrated by flow cytometry- and fluorescence anisotropy-based Y1R binding studies (satn. and competition binding and assocn. and dissocn. kinetics) as well as by widefield and total internal reflection fluorescence (TIRF) microscopy of live tumor cells, revealing that fluorescence was mainly localized at the plasma membrane.
- 41Keller, M.; Erdmann, D.; Pop, N.; Pluym, N.; Teng, S.; Bernhardt, G.; Buschauer, A. Red-fluorescent argininamide-type NPY Y1 receptor antagonists as pharmacological tools. Biorg. Med. Chem. 2011, 19, 2859– 2878, DOI: 10.1016/j.bmc.2011.03.045Google Scholar41Red-fluorescent argininamide-type NPY Y1 receptor antagonists as pharmacological toolsKeller, Max; Erdmann, Daniela; Pop, Nathalie; Pluym, Nikola; Teng, Shangjun; Bernhardt, Guenther; Buschauer, ArminBioorganic & Medicinal Chemistry (2011), 19 (9), 2859-2878CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Fluorescently labeled NPY Y1 receptor (Y1R) ligands were synthesized by connecting pyrylium and cyanine dyes with the argininamide-type Y1R antagonist core structure by linkers, covering a wide variety in length and chem. nature, attached to the guanidine group. The most promising fluorescent probes had Y1R affinities (radioligand binding) and antagonistic activities (calcium assay) in the one- to two-digit nanomolar range. These compds. turned out to be stable under assay conditions and to be appropriate for the detection of Y1Rs by confocal microscopy in live cells. To improve the signal-to-noise ratio by shifting the emission into the near IR, a new benzothiazolium-type fluorescent cyanine dye (UR-DE99) was synthesized and attached to the parent antagonist via a carbamoyl linker yielding UR-MK131, a highly potent fluorescent Y1R probe, which was also successfully applied in flow cytometry.
- 42She, X.; Pegoli, A.; Gruber, C. G.; Wifling, D.; Carpenter, J.; Hübner, H.; Chen, M.; Wan, J.; Bernhardt, G.; Gmeiner, P.; Holliday, N. D.; Keller, M. Red-emitting dibenzodiazepinone derivatives as fluorescent dualsteric probes for the muscarinic acetylcholine M2 receptor. J. Med. Chem. 2020, 63, 4133– 4154, DOI: 10.1021/acs.jmedchem.9b02172Google Scholar42Red-Emitting Dibenzodiazepinone Derivatives as Fluorescent Dualsteric Probes for the Muscarinic Acetylcholine M2 ReceptorShe, Xueke; Pegoli, Andrea; Gruber, Corinna G.; Wifling, David; Carpenter, Jessica; Huebner, Harald; Chen, Mengya; Wan, Jianfei; Bernhardt, Guenther; Gmeiner, Peter; Holliday, Nicholas D.; Keller, MaxJournal of Medicinal Chemistry (2020), 63 (8), 4133-4154CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fluorescently labeled dibenzodiazepinone-type muscarinic acetylcholine receptor (MR) antagonists, including dimeric ligands, were prepd. using red-emitting cyanine dyes. Probes contg. a fluorophore with neg. charge showed high M2R affinities (pKi (radioligand competition binding): 9.10-9.59). Binding studies at M1 and M3-M5 receptors indicated a M2R preference. Flow cytometric and high-content imaging satn. and competition binding (M1R, M2R, and M4R) confirmed occupation of the orthosteric site. Confocal microscopy revealed that fluorescence was located mainly at the cell membrane (CHO-hM2R cells). Results from dissocn. and satn. binding expts. (M2R) in the presence of allosteric M2R modulators (dissocn.: W84, LY2119620, and alcuronium; satn. binding: W84) were consistent with a competitive mode of action between the fluorescent probes and the allosteric ligands. Taken together, these lines of evidence indicate that these ligands are useful fluorescent mol. tools to label the M2R in imaging and binding studies and suggest that they have a dualsteric mode of action.
- 43Katritch, V.; Fenalti, G.; Abola, E. E.; Roth, B. L.; Cherezov, V.; Stevens, R. C. Allosteric sodium in class A GPCR signaling. Trends Biochem. Sci. 2014, 39, 233– 244, DOI: 10.1016/j.tibs.2014.03.002Google Scholar43Allosteric sodium in class A GPCR signalingKatritch, Vsevolod; Fenalti, Gustavo; Abola, Enrique E.; Roth, Bryan L.; Cherezov, Vadim; Stevens, Raymond C.Trends in Biochemical Sciences (2014), 39 (5), 233-244CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. Despite their functional and structural diversity, G-protein-coupled receptors (GPCRs) share a common mechanism of signal transduction via conformational changes in the seven-transmembrane (7TM) helical domain. New major insights into this mechanism come from the recent crystallog. discoveries of a partially hydrated sodium ion that is specifically bound in the middle of the 7TM bundle of multiple class A GPCRs. This review discusses the remarkable structural conservation and distinct features of the Na+ pocket in this most populous GPCR class, as well as the conformational collapse of the pocket upon receptor activation. New insights help to explain allosteric effects of sodium on GPCR agonist binding and activation, and sodium's role as a potential co-factor in class A GPCR function.
- 44White, K. L.; Eddy, M. T.; Gao, Z. G.; Han, G. W.; Lian, T.; Deary, A.; Patel, N.; Jacobson, K. A.; Katritch, V.; Stevens, R. C. Structural connection between activation microswitch and allosteric sodium site in GPCR Signaling. Structure 2018, 26, 259– 269, DOI: 10.1016/j.str.2017.12.013Google Scholar44Structural Connection between Activation Microswitch and Allosteric Sodium Site in GPCR SignalingWhite, Kate L.; Eddy, Matthew T.; Gao, Zhan-Guo; Han, Gye Won; Lian, Tiffany; Deary, Alexander; Patel, Nilkanth; Jacobson, Kenneth A.; Katritch, Vsevolod; Stevens, Raymond C.Structure (Oxford, United Kingdom) (2018), 26 (2), 259-269.e5CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)Sodium ions are endogenous allosteric modulators of many G-protein-coupled receptors (GPCRs). Mutation of key residues in the sodium binding motif causes a striking effect on G-protein signaling. We report the crystal structures of agonist complexes for two variants in the first sodium coordination shell of the human A2A adenosine receptor, D522.50N and S913.39A. Both structures present an overall active-like conformation; however, the variants show key changes in the activation motif NPxxY. Changes in the hydrogen bonding network in this microswitch suggest a possible mechanism for modified G-protein signaling and enhanced thermal stability. These structures, signaling data, and thermal stability anal. with a panel of pharmacol. ligands provide a basis for understanding the role of the sodium-coordinating residues on stability and G-protein signaling. Utilizing the D2.50N variant is a promising method for stabilizing class A GPCRs to accelerate structural efforts and drug discovery.
- 45DeVree, B. T.; Mahoney, J. P.; Velez-Ruiz, G. A.; Rasmussen, S. G.; Kuszak, A. J.; Edwald, E.; Fung, J. J.; Manglik, A.; Masureel, M.; Du, Y.; Matt, R. A.; Pardon, E.; Steyaert, J.; Kobilka, B. K.; Sunahara, R. K. Allosteric coupling from G protein to the agonist-binding pocket in GPCRs. Nature 2016, 535, 182– 186, DOI: 10.1038/nature18324Google Scholar45Allosteric coupling from G protein to the agonist-binding pocket in GPCRsDeVree, Brian T.; Mahoney, Jacob P.; Velez-Ruiz, Gisselle A.; Rasmussen, Soren G. F.; Kuszak, Adam J.; Edwald, Elin; Fung, Juan-Jose; Manglik, Aashish; Masureel, Matthieu; Du, Yang; Matt, Rachel A.; Pardon, Els; Steyaert, Jan; Kobilka, Brian K.; Sunahara, Roger K.Nature (London, United Kingdom) (2016), 535 (7610), 182-186CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)G protein-coupled receptors (GPCRs) remain the primary conduit by which cells detect environmental stimuli and communicate with each other. Upon activation by extracellular agonists, these seven-transmembrane-domain-contg. receptors interact with heterotrimeric G proteins to regulate downstream second messenger and/or protein kinase cascades. Crystallog. evidence from a prototypic GPCR, the β2-adrenergic receptor (β2AR), in complex with its cognate G protein, Gs, has provided a model for how agonist binding promotes conformational changes that propagate through the GPCR and into the nucleotide-binding pocket of the G protein α-subunit to catalyze GDP release, the key step required for GTP binding and activation of G proteins. The structure also offers hints about how G protein binding may, in turn, allosterically influence ligand binding. Here we provide functional evidence that G protein coupling to the β2AR stabilizes a 'closed' receptor conformation characterized by restricted access to and egress from the hormone-binding site. Surprisingly, the effects of G protein on the hormone-binding site can be obsd. in the absence of a bound agonist, where G protein coupling driven by basal receptor activity impedes the assocn. of agonists, partial agonists, antagonists and inverse agonists. The ability of bound ligands to dissoc. from the receptor is also hindered, providing a structural explanation for the G protein-mediated enhancement of agonist affinity, which has been obsd. for many GPCR-G protein pairs. Our data also indicate that in contrast to agonist binding alone, coupling of a G protein in the absence of an agonist stabilizes large structural changes in a GPCR. The effects of nucleotide-free G protein on ligand-binding kinetics are shared by other members of the superfamily of GPCRs, suggesting that a common mechanism may underlie G protein-mediated enhancement of agonist affinity.
- 46Wingler, L. M.; Lefkowitz, R. J. Conformational basis of G protein-coupled receptor signaling versatility. Trends Cell Biol. 2020, 30, 736– 747, DOI: 10.1016/j.tcb.2020.06.002Google Scholar46Conformational Basis of G Protein-Coupled Receptor Signaling VersatilityWingler, Laura M.; Lefkowitz, Robert J.Trends in Cell Biology (2020), 30 (9), 736-747CODEN: TCBIEK; ISSN:0962-8924. (Elsevier Ltd.)A review. G protein-coupled receptors (GPCRs) are privileged structural scaffolds in biol. that have the versatility to regulate diverse physiol. processes. Interestingly, many GPCR ligands exhibit significant 'bias' - the ability to preferentially activate subsets of the many cellular pathways downstream of these receptors. Recently, complementary information from structural and spectroscopic approaches has made significant inroads into understanding the mechanisms of these biased ligands. The consistently emerging theme is that GPCRs are highly dynamic proteins, and ligands with varying pharmacol. properties differentially modulate the equil. among multiple conformations. Biased signaling and other recently appreciated complexities of GPCR signaling thus appear to be a natural consequence of the conformational heterogeneity of GPCRs and GPCR-transducer complexes.
- 47Copeland, R. A. Conformational adaptation in drug–target interactions and residence time. Future Med. Chem. 2011, 3, 1491– 1501, DOI: 10.4155/fmc.11.112Google Scholar47Conformational adaptation in drug-target interactions and residence timeCopeland, Robert A.Future Medicinal Chemistry (2011), 3 (12), 1491-1501CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. Although drug-target interactions are commonly illustrated in terms of structurally static binding and dissocn. events, such descriptions are inadequate to explain the impact of conformational dynamics on these processes. For high-affinity interactions, both the assocn. and dissocn. of drug mols. to and from their targets are often controlled by conformational changes of the target. Conformational adaptation can greatly influence the residence time of a drug on its target (i.e., the lifetime of the binary drug-target complex); long residence time can lead to sustained pharmacol. and may also mitigate off-target toxicity. In this perspective, the kinetics of drug-target assocn. and dissocn. reactions are explored, with particular emphasis on the impact of conformational adaptation on drug-target residence time.
- 48Vauquelin, G. Simplified models for heterobivalent ligand binding: when are they applicable and which are the factors that affect their target residence time. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2013, 386, 949– 962, DOI: 10.1007/s00210-013-0881-0Google Scholar48Simplified models for heterobivalent ligand binding: when are they applicable and which are the factors that affect their target residence timeVauquelin, GeorgesNaunyn-Schmiedeberg's Archives of Pharmacology (2013), 386 (11), 949-962CODEN: NSAPCC; ISSN:0028-1298. (Springer)Bivalent ligands often display high affinity/avidity for and long residence time at their target. Thereto responsible is the synergy that emanates from the simultaneous binding of their two pharmacophores to their resp. target sites. Thermodn. cycle models permit the most complete description of the binding process, and thereto, corresponding differential equation-based simulations link the "microscopic" rate consts. that govern the individual binding steps to the "macroscopic" bivalent ligand's binding properties. Present simulations of heterobivalent ligand binding led to an appreciably simpler description thereof. The thermodn. cycle model can be split into two pathways/lanes that the bivalent ligand can solicit to reach fully bound state. Since the first binding event prompts the still free pharmacophore to stay into "forced proximity" of its target site, such lanes can be looked into by the equations that also apply to induced fit binding mechanisms. Interestingly, the simplest equations apply when bivalency goes along with a large gain in avidity. The overall bivalent ligand assocn. and dissocn. will be swifter than via each lane apart, but it is the lane that allows the fastest bidirectional "transit" between the free and the fully bound target that is chiefly solicited. The bivalent ligand's residence time is governed not only by the stability of the fully bound complex but also by the ability of freshly dissocd. pharmacophores to successfully rebind. Hence, the presence of a slow-assocg. pharmacophore could be counterproductive. Yet, a long residence time is unfortunately also responsible for the slow attainment of binding equil.
- 49Palmberger, D.; Wilson, I. B.; Berger, I.; Grabherr, R.; Rendic, D. SweetBac: a new approach for the production of mammalianised glycoproteins in insect cells. PLoS One 2012, 7, e34226 DOI: 10.1371/journal.pone.0034226Google Scholar49SweetBac: a new approach for the production of mammalianised glycoproteins in insect cellsPalmberger, Dieter; Wilson, Iain B. H.; Berger, Imre; Grabherr, Reingard; Rendic, DubravkoPLoS One (2012), 7 (4), e34226CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Recombinant prodn. of therapeutically active proteins has become a central focus of contemporary life science research. These proteins are often produced in mammalian cells, in order to obtain products with post-translational modifications similar to their natural counterparts. However, in cases where a fast and flexible system for recombinant prodn. of proteins is needed, the use of mammalian cells is limited. The baculoviral insect cell system has proven to be a powerful alternative for the expression of a wide range of recombinant proteins in short time frames. The major drawback of baculoviral systems lies in the inability to perform mammalian-like glycosylation required for the prodn. of therapeutic glycoproteins. In this study we integrated sequences encoding Caenorhabditis elegans N-acetylglucosaminyl-transferase II and bovine β1,4-galactosyltransferase I into the backbone of a baculovirus genome. The thereby generated SweetBac virus was subsequently used for the prodn. of the human HIV anti-gp41 antibody 3D6 by integrating heavy and light chain open reading frames into the SweetBac genome. The parallel expression of target genes and glycosyltransferases reduced the yield of secreted antibody. However, the overall expression rate, esp. in the recently established Tnao38 cell line, was comparable to that of transient expression in mammalian cells. In order to evaluate the ability of SweetBac to generate mammalian-like N-glycan structures on 3D6 antibody, we performed SDS-PAGE and tested for the presence of terminal galactose using Riccinus communis agglutinin I. The mammalianized variants of 3D6 showed highly specific binding to the lectin, indicating proper functionality. To confirm these results, PNGase A released N-glycans were analyzed by MALDI-TOF-MS and shown to contain structures with mainly one or two terminal galactose residues. Since the presence of specific N-glycans has an impact on antibodies' ability to exert different effector functions, we tested the binding to human Fc gamma receptor I present on U937 cells.
- 50Schneider, E. H.; Seifert, R. Sf9 cells: A versatile model system to investigate the pharmacological properties of G protein-coupled receptors. Pharmacol. Ther. 2010, 128, 387– 418, DOI: 10.1016/j.pharmthera.2010.07.005Google Scholar50Sf9 cells: A versatile model system to investigate the pharmacological properties of G protein-coupled receptorsSchneider, Erich H.; Seifert, RolandPharmacology & Therapeutics (2010), 128 (3), 387-418CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)A review. The Sf9 cell/baculovirus expression system is widely used for high-level protein expression, often with the purpose of purifn. However, proteins may also be functionally expressed in the defined Sf9 cell environment. According to the literature, the pharmacol. of G-protein-coupled receptors (GPCRs) functionally reconstituted in Sf9 cells is similar to the receptor properties in mammalian cells. Sf9 cells express both recombinant GPCRs and G-proteins at much higher levels than mammalian cells. Sf9 cells can be grown in suspension culture, providing an inexpensive way of obtaining large protein amts. Co-infection with various baculoviruses allows free combination of GPCRs with different G-proteins. The absence of constitutively active receptors in Sf9 cells provides an excellent signal-to background ratio in functional assays, allowing the detection of agonist-independent receptor activity and of small ligand-induced signals including partial agonistic and inverse agonistic effects. Insect cell Gαi-like proteins mostly do not couple productively to mammalian GPCRs. Thus, unlike in mammalian cells, Sf9 cells do not require pertussis toxin treatment to obtain a Gαi-free environment. Co-expression of GPCRs with Gαi1, Gαi2, Gαi3 or Gαo in Sf9 cells allows the generation of a selectivity profile for these Gαi/o-isoforms. Addnl., GPCR-G-protein combinations can be compared with defined 1:1 stoichiometry by expressing GPCR-Gα fusion proteins. Sf9 cells can also be employed for ligand screening in medicinal chem. programs, using radioligand binding assays or functional assays, like the steady-state GTPase- or [35S]GTPγS binding assay. This review shows that Sf9 cells are a versatile model system to investigate the pharmacol. properties of GPCRs.
- 51Grätz, L.; Müller, C.; Pegoli, A.; Schindler, L.; Bernhardt, G.; Littmann, T. Insertion of Nanoluc into the extracellular loops as a complementary method to establish BRET-based binding assays for GPCRs. ACS Pharmacol. Transl. Sci. 2022, 5, 1142– 1155, DOI: 10.1021/acsptsci.2c00162Google ScholarThere is no corresponding record for this reference.
- 52Calderón, J. C.; Plut, E.; Keller, M.; Cabrele, C.; Reiser, O.; Gervasio, F. L.; Clark, T. Extended metadynamics protocol for binding/unbinding free energies of peptide ligands to class A G-protein-coupled receptors. J. Chem. Inf. Model. 2024, 64, 205– 218, DOI: 10.1021/acs.jcim.3c01574Google ScholarThere is no corresponding record for this reference.
- 53Hall, M. P.; Unch, J.; Binkowski, B. F.; Valley, M. P.; Butler, B. L.; Wood, M. G.; Otto, P.; Zimmerman, K.; Vidugiris, G.; Machleidt, T.; Robers, M. B.; Benink, H. A.; Eggers, C. T.; Slater, M. R.; Meisenheimer, P. L.; Klaubert, D. H.; Fan, F.; Encell, L. P.; Wood, K. V. Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone substrate. ACS Chem. Biol. 2012, 7, 1848– 1857, DOI: 10.1021/cb3002478Google Scholar53Engineered Luciferase Reporter from a Deep Sea Shrimp Utilizing a Novel Imidazopyrazinone SubstrateHall, Mary P.; Unch, James; Binkowski, Brock F.; Valley, Michael P.; Butler, Braeden L.; Wood, Monika G.; Otto, Paul; Zimmerman, Kristopher; Vidugiris, Gediminas; Machleidt, Thomas; Robers, Matthew B.; Benink, Helene A.; Eggers, Christopher T.; Slater, Michael R.; Meisenheimer, Poncho L.; Klaubert, Dieter H.; Fan, Frank; Encell, Lance P.; Wood, Keith V.ACS Chemical Biology (2012), 7 (11), 1848-1857CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Bioluminescence methodologies have been extraordinarily useful due to their high sensitivity, broad dynamic range, and operational simplicity. These capabilities have been realized largely through incremental adaptations of native enzymes and substrates, originating from luminous organisms of diverse evolutionary lineages. We engineered both an enzyme and substrate in combination to create a novel bioluminescence system capable of more efficient light emission with superior biochem. and phys. characteristics. Using a small luciferase subunit (19 kDa) from the deep sea shrimp Oplophorus gracilirostris, we have improved luminescence expression in mammalian cells ∼2.5 million-fold by merging optimization of protein structure with development of a novel imidazopyrazinone substrate (furimazine). The new luciferase, NanoLuc, produces glow-type luminescence (signal half-life >2 h) with a specific activity ∼150-fold greater than that of either firefly (Photinus pyralis) or Renilla luciferases similarly configured for glow-type assays. In mammalian cells, NanoLuc shows no evidence of post-translational modifications or subcellular partitioning. The enzyme exhibits high phys. stability, retaining activity with incubation up to 55 °C or in culture medium for >15 h at 37 °C. As a genetic reporter, NanoLuc may be configured for high sensitivity or for response dynamics by appending a degrdn. sequence to reduce intracellular accumulation. Appending a signal sequence allows NanoLuc to be exported to the culture medium, where reporter expression can be measured without cell lysis. Fusion onto other proteins allows luminescent assays of their metab. or localization within cells. Reporter quantitation is achievable even at very low expression levels to facilitate more reliable coupling with endogenous cellular processes.
- 54Keller, M.; Kaske, M.; Holzammer, T.; Bernhardt, G.; Buschauer, A. Dimeric argininamide-type neuropeptide Y receptor antagonists: Chiral discrimination between Y1 and Y4 receptors. Biorg. Med. Chem. 2013, 21, 6303– 6322, DOI: 10.1016/j.bmc.2013.08.065Google ScholarThere is no corresponding record for this reference.
- 55Pegoli, A.; She, X.; Wifling, D.; Hubner, H.; Bernhardt, G.; Gmeiner, P.; Keller, M. Radiolabeled dibenzodiazepinone-type antagonists give evidence of dualsteric binding at the M(2) muscarinic acetylcholine receptor. J. Med. Chem. 2017, 60, 3314– 3334, DOI: 10.1021/acs.jmedchem.6b01892Google Scholar55Radiolabeled Dibenzodiazepinone-Type Antagonists Give Evidence of Dualsteric Binding at the M2 Muscarinic Acetylcholine ReceptorPegoli Andrea; She Xueke; Wifling David; Bernhardt Gunther; Keller Max; Hubner Harald; Gmeiner PeterJournal of medicinal chemistry (2017), 60 (8), 3314-3334 ISSN:.The dualsteric ligand approach, aiming at ligands with improved subtype selectivity, has been increasingly applied to muscarinic receptors (MRs). In this article, we present the synthesis and characterization of a M2R subtype-preferring radiolabeled dibenzodiazepinone-type antagonist ([(3)H]UNSW-MK259, [(3)H]19) and its homodimeric analogue [(3)H]UR-AP060 ([(3)H]33). Saturation binding studies at the M2R, using the orthosteric antagonist atropine to determine unspecific binding, proved that the monomeric and the dimeric compound bind to the orthosteric binding site (apparent Kd: 0.87 and 0.31 nM, respectively). Various binding studies with [(3)H]19 and [(3)H]33 at the M2R, for instance, saturation binding experiments in the presence of the allosteric MR modulators W84 (8) or LY2119620 (9) (Schild-like analysis) suggested a competitive mechanism between the allosteric modulator and the dibenzodiazepinone derivatives, and thus a dualsteric binding mode of both 19 and 33. This was consistent with the results of M2R MD simulations (≥2 μs) performed with 19 and 33.
- 56Schindler, L.; Moosbauer, J.; Schmidt, D.; Spruss, T.; Grätz, L.; Lüdeke, S.; Hofheinz, F.; Meister, S.; Echtenacher, B.; Bernhardt, G.; Pietzsch, J.; Hellwig, D.; Keller, M. Development of a neurotensin-derived 68Ga-labeled PET ligand with high in vivo stability for imaging of NTS1 receptor-expressing tumors. Cancers 2022, 14, 4922 DOI: 10.3390/cancers14194922Google ScholarThere is no corresponding record for this reference.
- 57Schindler, L.; Wohlfahrt, K.; von Krüchten, L. G.; Prante, O.; Keller, M.; Maschauer, S. Neurotensin analogs by fluoroglycosylation at Nω-carbamoylated arginines for PET imaging of NTS1-positive tumors. Sci. Rep. 2022, 12, 15028 DOI: 10.1038/s41598-022-19296-0Google ScholarThere is no corresponding record for this reference.
- 58Höfelschweiger, B. K. The pyrylium dyes: A new class of biolabels. Synthesis, spectroscopy, and application as labels and in general protein assay. Doctoral thesis, University of Regensburg, Regensburg, 2005.Google ScholarThere is no corresponding record for this reference.
- 59Moser, C.; Bernhardt, G.; Michel, J.; Schwarz, H.; Buschauer, A. Cloning and functional expression of the hNPY Y5 receptor in human endometrial cancer (HEC-1B) cells. Can. J. Physiol. Pharmacol. 2000, 78, 134– 142, DOI: 10.1139/y99-125Google Scholar59Cloning and functional expression of the hNPY Y5 receptor in human endometrial cancer (HEC-1B) cellsMoser, C.; Bernhardt, G.; Michel, J.; Schwarz, H.; Buschauer, A.Canadian Journal of Physiology and Pharmacology (2000), 78 (2), 134-142CODEN: CJPPA3; ISSN:0008-4212. (National Research Council of Canada)Aiming to develop a functional assay for the human NPY Y5 receptor based on adenylyl cyclase activity, HEC-1B cells, in which cAMP synthesis can be efficiently stimulated with forskolin, were selected for the transfection with the pcDNA3-Y5-FLAG and the pcDEF3-Y5 vectors. After optimization of the transfection procedure, the binding of [3H]propionyl-NPY to transiently and stably expressed Y5 receptors was detd. The affinities of NPY, NPY derivs., and rPP (pNPY ≥ p(Leu31Pro34)NPY = p(2-36)NPY ≥ p(D-Trp32)NPY > p(13-36)NPY > rPP) were in accordance with the NPY Y5 receptor subtype. For [3H]propionyl-pNPY approx. 1.7×105 and 1×106 binding sites per transiently and stably transfected cell, resp., were detd. The KD values were 2.4±0.4 and 1.7±0.2 nM, resp. Due to the high expression of the receptor protein, both stably and transiently transfected cells can be conveniently used in routine radioligand binding studies. By contrast, functional assays were only feasible with HEC-1B cells stably expressing the Y5 receptor. In these cells, 10 nM pNPY inhibited the forskolin-stimulated cAMP synthesis by 75%. This effect was partially antagonized by the Y5 antagonist N-{trans-[4-(2-naphthylmethylamino)-methyl]cyclohexylmethyl}naphthalene-2-sulfonamide. Although the genetic variability of cancer cells is in principle incompatible with a stable phenotype, both ligand binding characteristics and functionality of the Y5 receptor remained unchanged for more than 30 passages.
- 60Bartole, E.; Grätz, L.; Littmann, T.; Wifling, D.; Seibel, U.; Buschauer, A.; Bernhardt, G. UR-DEBa242: A Py-5-labeled fluorescent multipurpose probe for investigations on the histamine H3 and H4 receptors. J. Med. Chem. 2020, 63, 5297– 5311, DOI: 10.1021/acs.jmedchem.0c00160Google Scholar60UR-DEBa242: A Py-5-Labeled Fluorescent Multipurpose Probe for Investigations on the Histamine H3 and H4 ReceptorsBartole, Edith; Graetz, Lukas; Littmann, Timo; Wifling, David; Seibel, Ulla; Buschauer, Armin; Bernhardt, GuentherJournal of Medicinal Chemistry (2020), 63 (10), 5297-5311CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Comprehensively characterized fluorescent probes for the histamine H3 receptor (H3R) and esp. for the H4R orthologs [e.g., human (h) and mouse (m)] are highly needed as versatile complementary tools to radioligands. In view of fluorescent probes for BRET-based binding studies and for localizing the H4R in live cells, we synthesized and biol. characterized Py-5-labeled histamine derivs. The most notable compd. was UR-DEBa242 (26, 1-[4-(1H-Imidazol-4-yl)butyl]-4-{(1E,3E)-4-[4-(dimethylamino)phenyl]buta-1,3-dienyl}-2, 6-dimethylpyridinium hydrotrifluoroacetate trifluoroacetate), acting as a partial agonist at the hH3R [pEC50 (reporter gene) 8.77] and as an inverse agonist/antagonist at the h/mH4Rs [pIC50(reporter gene) 8.76/7.08; pIC50/pKb (β-arrestin2) 7.81/7.30]. In confocal microscopy, 26 proved suitable for hH4R localization and trafficking studies in live cells. BRET-based binding at the NLuc-hH3,4Rs/mH4R [pKd 8.78/7.75/7.18, comparable to binding consts. from radioligand binding/flow cytometry; fast assocn./dissocn. (~ 2 min)] revealed 26 as a useful mol. tool to det. hH3,4Rs/mH4R binding affinities of ligands binding to these receptors.
- 61Mansouri, M.; Bellon-Echeverria, I.; Rizk, A.; Ehsaei, Z.; Cosentino, C. C.; Silva, C. S.; Xie, Y.; Boyce, F. M.; Davis, M. W.; Neuhauss, S. C. F.; Taylor, V.; Ballmer-Hofer, K.; Berger, I.; Berger, P. Highly efficient baculovirus-mediated multigene delivery in primary cells. Nat. Commun. 2016, 7, 11529 DOI: 10.1038/ncomms11529Google Scholar61Highly efficient baculovirus-mediated multigene delivery in primary cellsMansouri, Maysam; Bellon-Echeverria, Itxaso; Rizk, Aurelien; Ehsaei, Zahra; Cianciolo Cosentino, Chiara; Silva, Catarina S.; Xie, Ye; Boyce, Frederick M.; Davis, M. Wayne; Neuhauss, Stephan C. F.; Taylor, Verdon; Ballmer-Hofer, Kurt; Berger, Imre; Berger, PhilippNature Communications (2016), 7 (), 11529CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Multigene delivery and subsequent cellular expression is emerging as a key technol. required in diverse research fields including, synthetic and structural biol., cellular reprogramming and functional pharmaceutical screening. Current viral delivery systems such as retro- and adenoviruses suffer from limited DNA cargo capacity, thus impeding unrestricted multigene expression. We developed MultiPrime, a modular, non-cytotoxic, non-integrating, baculovirus-based vector system expediting highly efficient transient multigene expression from a variety of promoters. MultiPrime viruses efficiently transduce a wide range of cell types, including non-dividing primary neurons and induced-pluripotent stem cells (iPS). We show that MultiPrime can be used for reprogramming, and for genome editing and engineering by CRISPR/Cas9. Moreover, we implemented dual-host-specific cassettes enabling multiprotein expression in insect and mammalian cells using a single reagent. Our expts. establish MultiPrime as a powerful and highly efficient tool, to deliver multiple genes for a wide range of applications in primary and established mammalian cells.
- 62Nie, Y.; Chaillet, M.; Becke, C.; Haffke, M.; Pelosse, M.; Fitzgerald, D.; Collinson, I.; Schaffitzel, C.; Berger, I. ACEMBL tool-kits for high-throughput multigene delivery and expression in prokaryotic and eukaryotic hosts. Adv. Exp. Med. Biol. 2016, 896, 27– 42, DOI: 10.1007/978-3-319-27216-0_3Google ScholarThere is no corresponding record for this reference.
- 63Cheng, Y.-C.; Prusoff, W. H. Relationship between the inhibition constant (KI) and the concentration of inhibitor which causes 50% inhibition (IC50) of an enzymatic reaction. Biochem. Pharmacol. 1973, 22, 3099– 3108, DOI: 10.1016/0006-2952(73)90196-2Google Scholar63Relation between the inhibition constant K1) and the concentration of inhibitor which causes fifty per cent inhibition (I50) of an enzymic reactionCheng, Yung-Chi; Prusoff, William H.Biochemical Pharmacology (1973), 22 (23), 3099-108CODEN: BCPCA6; ISSN:0006-2952.The Ki and I50 values are equal where the kinetics are non- or uncompetitive, but not where they are competitive. The relation between the 2 values was analyzed for non- and uncompetitive bi- and monosubstrate reactions and for competitive bisubstrate reactions.
- 64Laasfeld, T.; Kopanchuk, S.; Rinken, A. Image-based cell-size estimation for baculovirus quantification. BioTechniques 2017, 63, 161– 168, DOI: 10.2144/000114595Google Scholar64Image-based cell-size estimation for baculovirus quantificationLaasfeld, Tanis; Kopanchuk, Sergei; Rinken, AgoBioTechniques (2017), 63 (4), 161-168CODEN: BTNQDO; ISSN:0736-6205. (Informa Healthcare)Measurement of virus concn. is essential for effective virus-based transfection technologies. Here, we describe a user-friendly, image-based cell-size estn. (ICSE) assay for bacufovirus quantification that relies on automated detn. of celf diams. from bright-field microscopy images. In the !CSC assay, microplate-based imaging systems and our custom ICSE-Tools software enable measurement of cell morphol. parameters over time. Results from the ICSE assay were in agreement with virus concn. measurements obtained using the traditional plaque assay as well as the Coulter principle- based cell-size measurement assay. ICSE-Tools is designed for data organization and image anal. from microplate-based imaging systems, and is freely available at www.gper.ut.eelsoftware.html.
- 65Veiksina, S.; Tahk, M.-J.; Laasfeld, T.; Link, R.; Kopanchuk, S.; Rinken, A. Fluorescence Anisotropy-Based Assay for Characterization of Ligand Binding Dynamics to GPCRs: The Case of Cy3B-Labeled Ligands Binding to MC4 Receptors in Budded Baculoviruses. In G Protein-Coupled Receptor Screening Assays: Methods and Protocols; Martins, S. A. M.; Prazeres, D. M. F., Eds.; Springer: US, New York, NY, 2021; pp 119– 136.Google ScholarThere is no corresponding record for this reference.
- 66Kopanchuk, S.; Vavers, E.; Veiksina, S.; Ligi, K.; Zvejniece, L.; Dambrova, M.; Rinken, A. Intracellular dynamics of the Sigma-1 receptor observed with super-resolution imaging microscopy. PLoS One 2022, 17, e0268563 DOI: 10.1371/journal.pone.0268563Google ScholarThere is no corresponding record for this reference.
- 67Soulez, F. In A “Learn 2D, Apply 3D” Method for 3D Deconvolution Microscopy, 2014 IEEE 11th International Symposium on Biomedical Imaging (ISBI); IEEE, 2014; pp 1075– 1078.Google ScholarThere is no corresponding record for this reference.
- 68Chenouard, N.; Bloch, I.; Olivo-Marin, J. C. Multiple hypothesis tracking for cluttered biological image sequences. IEEE Trans. Pattern Anal. Mach. Intell. 2013, 35, 2736– 3750, DOI: 10.1109/TPAMI.2013.97Google Scholar68Multiple hypothesis tracking for cluttered biological image sequencesChenouard Nicolas; Bloch Isabelle; Olivo-Marin Jean-ChristopheIEEE transactions on pattern analysis and machine intelligence (2013), 35 (11), 2736-3750 ISSN:.In this paper, we present a method for simultaneously tracking thousands of targets in biological image sequences, which is of major importance in modern biology. The complexity and inherent randomness of the problem lead us to propose a unified probabilistic framework for tracking biological particles in microscope images. The framework includes realistic models of particle motion and existence and of fluorescence image features. For the track extraction process per se, the very cluttered conditions motivate the adoption of a multiframe approach that enforces tracking decision robustness to poor imaging conditions and to random target movements. We tackle the large-scale nature of the problem by adapting the multiple hypothesis tracking algorithm to the proposed framework, resulting in a method with a favorable tradeoff between the model complexity and the computational cost of the tracking procedure. When compared to the state-of-the-art tracking techniques for bioimaging, the proposed algorithm is shown to be the only method providing high-quality results despite the critically poor imaging conditions and the dense target presence. We thus demonstrate the benefits of advanced Bayesian tracking techniques for the accurate computational modeling of dynamical biological processes, which is promising for further developments in this domain.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 3 publications.
- Fabian J. Ertl, Sergei Kopanchuk, Nicola C. Dijon, Santa Veikšina, Maris-Johanna Tahk, Tõnis Laasfeld, Franziska Schettler, Albert O. Gattor, Harald Hübner, Nataliya Archipowa, Johannes Köckenberger, Markus R. Heinrich, Peter Gmeiner, Roger J. Kutta, Nicholas D. Holliday, Ago Rinken, Max Keller. Dually Labeled Neurotensin NTS1R Ligands for Probing Radiochemical and Fluorescence-Based Binding Assays. Journal of Medicinal Chemistry 2024, 67
(18)
, 16664-16691. https://doi.org/10.1021/acs.jmedchem.4c01470
- Renáta Szabó, Ágnes Hornyánszky, Dóra Judit Kiss, György Miklós Keserű. Fluorescent tools for imaging class A G-protein coupled receptors. European Journal of Pharmaceutical Sciences 2025, 209 , 107074. https://doi.org/10.1016/j.ejps.2025.107074
- Max E. Huber, Silas L. Wurnig, Aurélien F. A. Moumbock, Lara Toy, Evi Kostenis, Ana Alonso Bartolomé, Martyna Szpakowska, Andy Chevigné, Stefan Günther, Finn K. Hansen, Matthias Schiedel. Development of a NanoBRET Assay Platform to Detect Intracellular Ligands for the Chemokine Receptors CCR6 and CXCR1. ChemMedChem 2024, 19
(20)
https://doi.org/10.1002/cmdc.202400284
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. (A) Structures and Y4R affinities of reported Y4R fluorescent ligands. 1 (26) and 2 (27) represent derivatives of the endogenous Y4R ligand hPP labeled with sulfo-Cy5 (S0223) and 5-TAMRA, respectively. Compound 3 (29) represents a derivative of the hexapeptide UR-KK236 (28) labeled with Sulfo-Cy5.5 (lumiprobe, ref no. 7330). (B) Structures of the reported cyclic Y4R ligands 4–7 showing high binding affinity to Y4R (pKi > 10). (30,31)
Figure 2
Figure 3
Scheme 1
Scheme 1. Synthesis of the Cyclic Peptide 11 via SPPSaaReagents and conditions: (a) Fmoc amino acid/HOBt/HBTU/DIPEA (5/5/4.9/10 equiv), DMF/NMP 8:2 v/v, 38 °C, 2 × 45 min (“double” coupling); Fmoc deprotection (following amino acid coupling): 20% piperidine in DMF/NMP 8:2 v/v, rt, 2 × 10 min; (b) building block 8 or 9/HOBt/HBTU/DIPEA (3/3/2.9/6 equiv), DMF/NMP 8:2 v/v, 38 °C, 16 h (single coupling); Fmoc deprotection (following amino acid coupling): as under (a); (c) succinic anhydride/DIPEA (10/10 equiv), DMF/NMP 8:2 v/v, 38 °C, 45 min; (d) (1) TFA/CH2Cl2 1:3 v/v, rt, 2 × 20 min; (2) TFA/H2O 95:5 v/v, rt, 5 h; overall yield: 37%; (e) peptide cyclization: HOBt/PyBOP/DIPEA (3/2.9/6 equiv), DMF/NMP 8:2 v/v, rt, 16 h, 21%.
Scheme 2
Scheme 2. Synthesis of the Fluorescent Y4R Ligands 16–19aaReagents and conditions: (a) DIPEA, DMF, rt, 2 h, 34–69%; (b) CuSO4, sodium ascorbate, H2O/NMP 1:1 v/v, rt, 2 h, 27%.
Figure 4
Figure 4. Flow cytometric saturation binding of 16 (A), 17 (B), and 18 (C) at whole CHO-hY4R-Gqi5-mtAEQ cells at 22 ± 2 °C. (A) Representative saturation isotherms (red circle) of 16 from experiments performed in sodium-free buffer (buffer I) and sodium-containing buffer (DPBS, 137 mM Na+). (B, C) Representative saturation isotherms (red circle) of 17 (B) and 18 (C) from experiments performed in sodium-containing buffer (DPBS). Unspecific binding (blue squares) was determined in the presence of 1 μM hPP (A–C). Total and unspecific binding data represent mean values ± SEM. Specific binding, representing calculated values ± propagated error, were fitted according to an equation describing a hyperbolic isotherm (binding-saturation: one site-specific binding, GraphPad Prism 5).
Figure 5
Figure 5. Binding kinetics of 16–18 determined by flow cytometry at whole CHO-hY4R-Gqi5-mtAEQ cells at 22 ± 2 °C. (A) Association of 16 to the hY4R under sodium-free conditions (buffer I) and in sodium-containing buffer (DPBS, 137 mM Na+). Concentration of 16: 0.3 and 0.7 nM, respectively. Proportion of the initial fast association (two-phase association fit, GraphPad Prism 5): 38 ± 4% (mean ± SEM). (B) Dissociation of 16 from the hY4R determined in buffer I and DPBS. The dissociation was initiated after 1.5 h of preincubation with 16 (c = 1.5 nM (Na+-free) and 3.5 nM (137 mM Na+)) by the addition of an excess of hPP (1000-fold) and 5 (100-fold). Plateau values of the three-parameter fits (monophasic exponential decline): 13% (Na+-free), 22% (137 mM Na+). (C) Association of 17 (c = 1 nM) and 18 (c = 0.5 nM) to the hY4R determined in DPBS (137 mM Na+). Proportion of the initial fast association (two-phase association fit, GraphPad Prism 5): 74 ± 1% (17), 29 ± 3% (18) (mean values ± SEM). (D) Dissociation of 17 and 18 from the hY4R in DPBS. The dissociation was initiated after 1.5 h of preincubation with 17 (c = 5 nM) or 18 (c = 2.5 nM) by the addition of an excess of hPP (1000-fold) and 5 (100-fold). Plateau values of the three-parameter fits (monophasic exponential decline): 13% (17), 22% (18). Data (A–D) represent mean values ± SEM from three independent experiments performed in duplicate.
Figure 6
Figure 6. Binding of 16 to hY4R displaying BBVs studied by FA measurement at 27 °C. (A) Binding isotherms of 16 obtained from experiments using fixed concentrations of 16 (0.5 or 2 nM) and increasing amounts of Y4R. Total binding is represented by filled symbols and unspecific binding (determined in the presence of 1 μM hPP) is represented by open symbols. Depicted data (mean values ± SEM from a representative experiment performed in duplicate) represent snapshots at 90 min incubation. Y4R concentrations displayed on the abscissa were calculated after global analysis of the data from three or four individual experiments by a modified version of a model described by Veiksina et al., (19) affording the estimated binding site (Y4R) concentration of the applied BBV stock which amounted to 6 ± 1 nM (Y4Rnonglyco, mean value ± SEM, n = 3) or 2.1 ± 0.1 nM (Y4RSwBac, mean value ± SEM, n = 4). (B) Association and dissociation of 16 (0.5 nM) determined in real time for three different Y4R concentrations (green, blue, and red symbols). Total binding is represented by filled symbols and unspecific binding (determined in the presence of 1 μM hPP) is represented by open symbols. Data represent mean values ± SEM from a representative experiment performed in duplicate.
Figure 7
Figure 7. Characterization of Y4R binding of fluorescent ligand 16 in a NanoBRET-based binding assay at 25 °C using intact HEK293T-hY4R-NLuc(intraECL2) cells. (A) Representative saturation isotherm (specific binding) from saturation binding experiments. Unspecific binding was determined in the presence of 1 μM 5. Total and unspecific binding data represent mean values ± SEM. Specific binding data represent calculated values ± propagated error. (B) Association of 16 (c = 1 nM) to Y4R. Mean values ± SEM from three independent experiments performed in triplicate. (C) Dissociation of 16 from Y4R. The dissociation was initiated after 1.5 h of preincubation with 16 (c = 3.5 nM) by the addition of a 1000-fold excess of 5. Mean values ± SEM from three independent experiments performed in duplicate. Plateau value of the three-parameter fit describing a monophasic exponential decline: 6% (note: for the biphasic fit the plateau value was not different from zero, see discussion).
Figure 8
Figure 8. Determination of Y4R affinities of Y4R reference ligands (hPP, 5, 7, UR-MK188, UR-MEK388, UR-KK200) in different fluorescence-based assays by competition binding with 16. (A) Displacement curves based on data from flow cytometric competition binding experiments performed with intact CHO-hY4R-Gqi5-mtAEQ cells in sodium-free buffer (buffer I) and sodium-containing buffer (DPBS, 137 mM Na+). (B) Displacement curves from fluorescence anisotropy-based competition binding experiments performed with Y4RSwBac-displaying BBVs in DPBS. (C) Displacement curves from NanoBRET-based competition binding experiments performed with intact HEK293T-hY4R-NLuc(intraECL2) cells in L15-HEPES (140 mM Na+). Data (A–C), representing mean values ± SEM from three to five independent experiments performed in duplicate (B) or triplicate (A, C), were fitted according to a four-parameter logistic model.
Figure 9
Figure 9. Visualization of fluorescent ligand (16, 17) binding to CHO-hY4R-Gqi5-mtAEQ cells by confocal microscopy. Shown are representative images acquired after incubation of the cells with 16 or 17 (each 20 nM) at 22 °C for 10 and 30 min. Unspecific binding was determined in the presence of 1 μM hPP. Nuclei were stained with H33342 (2 μM). Fluorescence of 16 and 17 is shown in green and red, respectively. Fluorescence of H33342 is shown in blue. Scale bar: 10 μm.
Figure 10
Figure 10. Visualization of binding of 16 to the hY4R transiently expressed by SK-OV-3 cells using wide-field and TIRF microscopy. (A) Wide-field fluorescence images acquired after incubation of the cells with 16 at 37 °C for 30 min. The two-color composite of individual focal planes after Z-stack deconvolution is shown with the green pseudocolor for 16 (561 nm excitation) and the blue pseudocolor for the nuclear stain channel (Hoechst 34580, 405 nm excitation). (B) Wide-field fluorescence and TIRF images of the same cells obtained after incubation of the cells with 16 (1 nM) at 37 °C for 30 min. Wide-field images were processed as under (A). In TIRF images, fluorescence of 16 is shown in white pseudocolor. Scale bar: 10 μm.
References
This article references 68 other publications.
- 1Michel, M. C.; Beck-Sickinger, A.; Cox, H.; Doods, H. N.; Herzog, H.; Larhammar, D.; Quirion, R.; Schwartz, T.; Westfall, T. XVI. International Union of Pharmacology recommendations for the nomenclature of neuropeptide Y, peptide YY, and pancreatic polypeptide receptors. Pharmacol. Rev. 1998, 50, 143– 1501XVI. International union of pharmacology recommendations for the nomenclature of neuropeptide Y, peptide YY, and pancreatic polypeptide receptorsMichel, Martin C.; Beck-Sickinger, Annette; Cox, Helen; Doods, Henri N.; Herzog, Herbert; Larhammar, Dan; Quirion, Remi; Schwartz, Thue; Westfall, ThomasPharmacological Reviews (1998), 50 (1), 143-150CODEN: PAREAQ; ISSN:0031-6997. (Williams & Wilkins)A review with ∼80 refs.
- 2Pedragosa-Badia, X.; Stichel, J.; Beck-Sickinger, A. G. Neuropeptide Y receptors: how to get subtype selectivity. Front. Endocrinol. 2013, 4, 5 DOI: 10.3389/fendo.2013.000052Neuropeptide Y receptors: how to get subtype selectivityPedragosa-Badia Xavier; Stichel Jan; Beck-Sickinger Annette GFrontiers in endocrinology (2013), 4 (), 5 ISSN:1664-2392.The neuropeptide Y (NPY) system is a multireceptor/multiligand system consisting of four receptors in humans (hY(1), hY(2), hY(4), hY(5)) and three agonists (NPY, PYY, PP) that activate these receptors with different potency. The relevance of this system in diseases like obesity or cancer, and the different role that each receptor plays influencing different biological processes makes this system suitable for the design of subtype selectivity studies. In this review we focus on the latest findings within the NPY system, we summarize recent mutagenesis studies, structure activity relationship studies, receptor chimera, and selective ligands focusing also on the binding mode of the native agonists.
- 3Ueno, N.; Inui, A.; Iwamoto, M.; Kaga, T.; Asakawa, A.; Okita, M.; Fujimiya, M.; Nakajima, Y.; Ohmoto, Y.; Ohnaka, M.; Nakaya, Y.; Miyazaki, J.-I.; Kasuga, M. Decreased food intake and body weight in pancreatic polypeptide-overexpressing mice. Gastroenterology 1999, 117, 1427– 1432, DOI: 10.1016/S0016-5085(99)70293-33Decreased food intake and body weight in pancreatic polypeptide-overexpressing miceUeno N; Inui A; Iwamoto M; Kaga T; Asakawa A; Okita M; Fujimiya M; Nakajima Y; Ohmoto Y; Ohnaka M; Nakaya Y; Miyazaki J I; Kasuga MGastroenterology (1999), 117 (6), 1427-32 ISSN:0016-5085.BACKGROUND & AIMS: Pancreatic polypeptide (PP) is a 36-amino acid hormone produced by F cells within the pancreatic islets and the exocrine pancreas. The definitive function of PP in mammalian physiology remains to be determined. This study examined the effects of chronic overexpression of PP through the development of PP transgenic mice. METHODS: PP transgenic mice were created by using mouse PP complementary DNA under the control of the cytomegalovirus immediate early enhancer-chicken beta-actin hybrid promoter (pCAGGS expression vector). RESULTS: A unique line of transgenic mice was created that overexpresses PP in the pancreatic islets with low levels of expression in other tissues including the brain. Plasma PP concentrations were more than 20 times higher than those of control littermates. However, PP overproduction led to postnatal lethality in half of the pups because of markedly decreased milk intake. The remaining PP transgenic mice gained less weight with specifically reduced food intake and fat mass compared with controls, a result that was more evident in male than in female mice. The transgenic mice exhibited a reduced rate of gastric emptying of a solid meal but had normal oxygen consumption and fasting leptin levels. Immunoneutralization with anti-PP antiserum reversed the phenotypic changes of transgenic animals. CONCLUSIONS: PP could be involved in feeding and body weight regulation partly through regulation of gastric emptying.
- 4Batterham, R. L.; Le Roux, C. W.; Cohen, M. A.; Park, A. J.; Ellis, S. M.; Patterson, M.; Frost, G. S.; Ghatei, M. A.; Bloom, S. R. Pancreatic polypeptide reduces appetite and food intake in humans. J. Clin. Endocrinol. Metab. 2003, 88, 3989– 3992, DOI: 10.1210/jc.2003-0306304Pancreatic polypeptide reduces appetite and food intake in humansBatterham, R. L.; Le Roux, C. W.; Cohen, M. A.; Park, A. J.; Ellis, S. M.; Patterson, M.; Frost, G. S.; Ghatei, M. A.; Bloom, S. R.Journal of Clinical Endocrinology and Metabolism (2003), 88 (8), 3989-3992CODEN: JCEMAZ; ISSN:0021-972X. (Endocrine Society)Pancreatic polypeptide (PP) is a gut hormone released from the pancreas in response to ingestion of food. Plasma PP has been shown to be reduced in conditions assocd. with increased food intake and elevated in anorexia nervosa. In addn. peripheral administration of PP has been shown to decrease food intake in rodents. These findings suggest that PP may act as a circulating factor that regulates food intake. Therefore we investigated the effect of i.v. infusion of PP (10 pmol/kg/min) on appetite and food intake in a randomized double-blind placebo-controlled crossover study in ten healthy volunteers. Infusion of PP reduced appetite and decreased the energy intake at a buffet lunch two hours post-infusion by 21.8±5.7% (P < 0.01). More importantly the inhibition of food intake was sustained, such that energy intake, as assessed by food diaries, was significantly reduced both the evening of the study and the following morning. Overall PP infusion reduced cumulative 24-h energy intake by 25.3±5.8%. In conclusion our data demonstrates that PP causes a sustained decrease in both appetite and food intake.
- 5Schmidt, P. T.; Näslund, E.; Grybäck, P.; Jacobsson, H.; Holst, J. J.; Hilsted, L.; Hellström, P. M. A role for pancreatic polypeptide in the regulation of gastric emptying and short-term metabolic control. J. Clin. Endocrinol. Metab. 2005, 90, 5241– 5246, DOI: 10.1210/jc.2004-20895A role for pancreatic polypeptide in the regulation of gastric emptying and short-term metabolic controlSchmidt, P. T.; Naeslund, E.; Grybaeck, P.; Jacobsson, H.; Holst, J. J.; Hilsted, L.; Hellstroem, P. M.Journal of Clinical Endocrinology and Metabolism (2005), 90 (9), 5241-5246CODEN: JCEMAZ; ISSN:0021-972X. (Endocrine Society)Context: Previous studies using pancreatic polypeptide (PP) infusions in humans have failed to show an effect on gastric emptying, glucose metab., and insulin secretion. This might be due to the use of nonhuman sequences of the peptide. Objective: The objective of this study was to use synthetic human PP to study gastric emptying rates of a solid meal and postprandial hormone secretion and glucose disposal as well as the gastric emptying rate of water. Design: This was a single-blind study. Setting: The study was performed at a university hospital. Participants: Fourteen healthy adult subjects were studied. Interventions: Infusion of saline or PP at 0.75 or 2.25 pmol/kg·min was given to eight subjects (gastric emptying of solid food), and infusion of saline or PP at 2.25 pmol/kg·min was given to six subjects (gastric emptying of water). Main Outcome Measures: The main outcome measures were gastric emptying of solids (scintigraphy), hunger ratings (visual analog scale), and plasma concns. of PP, insulin, glucagon, somatostatin, glucagon-like peptide 1, glucose, and gastric emptying of plain water (scintigraphy). Results: PP prolonged the lag phase and the half-time of emptying of the solid meal. The change in hunger rating, satiety, desire to eat after the meal, or prospective consumption was not affected. The postprandial rise in plasma glucose was prolonged by PP. The postprandial rise in insulin was also delayed by PP. PP had no significant effect on the emptying of water. Conclusions: PP inhibits gastric emptying of solid food and delays the postprandial rise in plasma glucose and insulin. PP is suggested to have a physiol. role in the pancreatic postprandial counter-regulation of gastric emptying and insulin secretion.
- 6Painsipp, E.; Wultsch, T.; Edelsbrunner, M. E.; Tasan, R. O.; Singewald, N.; Herzog, H.; Holzer, P. Reduced anxiety-like and depression-related behavior in neuropeptide Y Y4 receptor knockout mice. Genes, Brain Behav. 2008, 7, 532– 542, DOI: 10.1111/j.1601-183X.2008.00389.x6Reduced anxiety-like and depression-related behavior in neuropeptide Y Y4 receptor knockout micePainsipp E; Wultsch T; Edelsbrunner M E; Tasan R O; Singewald N; Herzog H; Holzer PGenes, brain, and behavior (2008), 7 (5), 532-42 ISSN:.Neuropeptide Y (NPY) acting through Y1 receptors reduces anxiety- and depression-like behavior in rodents, whereas Y2 receptor stimulation has the opposite effect. This study addressed the implication of Y4 receptors in emotional behavior by comparing female germ line Y4 knockout (Y4-/-) mice with control and germ line Y2-/- animals. Anxiety- and depression-like behavior was assessed with the open field (OF), elevated plus maze (EPM), stress-induced hyperthermia (SIH) and tail suspension tests (TST), respectively. Learning and memory were evaluated with the object recognition test (ORT). In the OF and EPM, both Y4-/- and Y2-/- mice exhibited reduced anxiety-related behavior and enhanced locomotor activity relative to control animals. Locomotor activity in a familiar environment was unchanged in Y4-/- but reduced in Y2-/- mice. The basal rectal temperature exhibited diurnal and genotype-related alterations. Control mice had temperature minima at noon and midnight, whereas Y4-/- and Y2-/- mice displayed only one temperature minimum at noon. The magnitude of SIH was related to time of the day and genotype in a complex manner. In the TST, the duration of immobility was significantly shorter in Y4-/- and Y2-/- mice than in controls. Object memory 6 h after initial exposure to the ORT was impaired in Y2-/- but not in Y4-/- mice, relative to control mice. These results show that genetic deletion of Y4 receptors, like that of Y2 receptors, reduces anxiety-like and depression-related behavior. Unlike Y2 receptor knockout, Y4 receptor knockout does not impair object memory. We propose that Y4 receptors play an important role in the regulation of behavioral homeostasis.
- 7Lin, S.; Shi, Y.-C.; Yulyaningsih, E.; Aljanova, A.; Zhang, L.; Macia, L.; Nguyen, A. D.; Lin, E.-J. D.; During, M. J.; Herzog, H.; Sainsbury, A. Critical role of arcuate Y4 receptors and the melanocortin system in pancreatic polypeptide-induced reduction in food intake in mice. PLoS One 2009, 4, e8488 DOI: 10.1371/journal.pone.0008488There is no corresponding record for this reference.
- 8Tasan, R. O.; Lin, S.; Hetzenauer, A.; Singewald, N.; Herzog, H.; Sperk, G. Increased novelty-induced motor activity and reduced depression-like behavior in neuropeptide Y (NPY)-Y4 receptor knockout mice. Neuroscience 2009, 158, 1717– 1730, DOI: 10.1016/j.neuroscience.2008.11.048There is no corresponding record for this reference.
- 9Sainsbury, A.; Shi, Y. C.; Zhang, L.; Aljanova, A.; Lin, Z.; Nguyen, A. D.; Herzog, H.; Lin, S. Y4 receptors and pancreatic polypeptide regulate food intake via hypothalamic orexin and brain-derived neurotropic factor dependent pathways. Neuropeptides 2010, 44, 261– 268, DOI: 10.1016/j.npep.2010.01.0019Y4 receptors and pancreatic polypeptide regulate food intake via hypothalamic orexin and brain-derived neurotropic factor dependent pathwaysSainsbury, Amanda; Shi, Yan-Chuan; Zhang, Lei; Aljanova, Aygul; Lin, Zhou; Nguyen, Amy D.; Herzog, Herbert; Lin, ShuNeuropeptides (Oxford, United Kingdom) (2010), 44 (3), 261-268CODEN: NRPPDD; ISSN:0143-4179. (Elsevier Ltd.)Gut-derived peptides are known to regulate food intake by activating specific receptors in the brain, but the target nuclei and neurons influenced are largely unknown. Here we show that peripherally administered pancreatic polypeptide (PP) stimulates neurons in key nuclei of the hypothalamus crit. for appetite and satiety regulation. In the lateral hypothalamic area (LHA), also known as the feeding center, neurons expressing the orexigenic neuropeptide orexin colocalize with the early neuronal activation marker c-Fos upon i.p. injection of PP into mice. In the ventromedial hypothalamus (VMH), also known as the satiety center, neurons activated by PP, as indicated by induction of c-Fos immunoreactivity, express the anorexigenic brain-derived neurotrophic factor (BDNF). Activation of neurons in the LHA and VMH in response to PP occurs via a Y4 receptor-dependent process as it is not seen in Y4 receptor knockout mice. We further demonstrate that in response to i.p. PP, orexin mRNA expression in the LHA is down-regulated, with Y4 receptors being crit. for this effect as it is not seen in Y4 receptor knockout mice, whereas BDNF mRNA expression is up-regulated in the VMH in response to i.p. PP in the fasted, but not in the non-fasted state. Taken together these data suggest that PP can regulate food intake by suppressing orexigenic pathways by down-regulation of orexin and simultaneously increasing anorexigenic pathways by up-regulating BDNF.
- 10Li, J. B.; Asakawa, A.; Terashi, M.; Cheng, K.; Chaolu, H.; Zoshiki, T.; Ushikai, M.; Sheriff, S.; Balasubramaniam, A.; Inui, A. Regulatory effects of Y4 receptor agonist (BVD-74D) on food intake. Peptides 2010, 31, 1706– 1710, DOI: 10.1016/j.peptides.2010.06.01110Regulatory effects of Y4 receptor agonist (BVD-74D) on food intakeLi, Jiang-Bo; Asakawa, Akihiro; Terashi, Mutsumi; Cheng, Kai-Chun; Chaolu, Huhe; Zoshiki, Takahiro; Ushikai, Miharu; Sheriff, Sulaiman; Balasubramaniam, Ambikaipakan; Inui, AkioPeptides (New York, NY, United States) (2010), 31 (9), 1706-1710CODEN: PPTDD5; ISSN:0196-9781. (Elsevier)The objective of this study was to clarify the role of a novel agonist with high selectivity and affinity for Y4 receptors in the regulation of food intake. The Y4 receptor agonist BVD-74D was administered to C57BL/6J mice that were fed with a normal or high-fat diet, and to fatty liver Shionogi (FLS)-ob/ob mice; the food intake, water intake, and body wt. gain were measured in these mice. In the mice fed with a normal diet, the cumulative food intake significantly decreased at 20 min and 1 h after the administration of 1 mg/kg of BVD-74D and at 1, 2, 4, 8, and 24 h after the administration of 10 mg/kg of BVD-74D. Moreover, the cumulative water intake and body wt. gain significantly decreased in these mice. Among the mice that were fed with a high-fat diet, the cumulative food intake and water intake significantly decreased 1, 2, and 4 h after BVD-74D (10 mg/kg) administration. Furthermore, the cumulative food intake significantly decreased 2 and 4 h after BVD-74D (10 mg/kg) administration in the FLS-ob/ob mice. Thus, we propose that the novel Y4 receptor agonist BVD-74D has suppressive effects on food intake, water intake, and wt. gain in normal mice fed with normal diets and on food intake in normal mice fed with high-fat diets and in FLS-ob/ob mice. These findings indicate that the Y4 receptor and its agonist would be promising targets for obesity.
- 11Moriya, R.; Fujikawa, T.; Ito, J.; Shirakura, T.; Hirose, H.; Suzuki, J.; Fukuroda, T.; MacNeil, D. J.; Kanatani, A. Pancreatic polypeptide enhances colonic muscle contraction and fecal output through neuropeptide Y Y4 receptor in mice. Eur. J. Pharmacol. 2010, 627, 258– 264, DOI: 10.1016/j.ejphar.2009.09.057There is no corresponding record for this reference.
- 12Brothers, S. P.; Wahlestedt, C. Therapeutic potential of neuropeptide Y (NPY) receptor ligands. EMBO Mol. Med. 2010, 2, 429– 439, DOI: 10.1002/emmm.20100010012Therapeutic potential of neuropeptide Y (NPY) receptor ligandsBrothers, Shaun P.; Wahlestedt, ClaesEMBO Molecular Medicine (2010), 2 (11), 429-439CODEN: EMMMAM; ISSN:1757-4684. (Wiley-Blackwell)A review. Neuropeptide Y (NPY) is widely distributed in the human body and contributes to a vast no. of physiol. processes. Since its discovery, NPY has been implicated in metabolic regulation and, although interest in its role in central mechanisms related to food intake and obesity has somewhat diminished, the topic remains a strong focus of research concerning NPY signalling. In addn., a no. of other uses for modulators of NPY receptors have been implied in a range of diseases, although the development of NPY receptor ligands has been slow, with no clin. approved receptor therapeutics currently available. Nevertheless, several interesting small mol. compds., notably Y2 receptor antagonists, have been published recently, fueling optimism in the field. Herein we review the role of NPY in the pathophysiol. of a no. of diseases and highlight instances where NPY receptor signalling systems are attractive therapeutic targets.
- 13Verma, D.; Hörmer, B.; Bellmann-Sickert, K.; Thieme, V.; Beck-Sickinger, A. G.; Herzog, H.; Sperk, G.; Tasan, R. O. Pancreatic polypeptide and its central Y4 receptors are essential for cued fear extinction and permanent suppression of fear. Br. J. Pharmacol. 2016, 173, 1925– 1938, DOI: 10.1111/bph.1345613Pancreatic polypeptide and its central Y4 receptors are essential for cued fear extinction and permanent suppression of fearVerma, D.; Hoermer, B.; Bellmann-Sickert, K.; Thieme, V.; Beck-Sickinger, A. G.; Herzog, H.; Sperk, G.; Tasan, R. O.British Journal of Pharmacology (2016), 173 (12), 1925-1938CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)Background and purpose : Avoiding danger and finding food are closely related behaviors that are essential for surviving in a natural environment. Growing evidence supports an important role of gut-brain peptides in modulating energy homeostasis and emotional-affective behavior. For instance, postprandial release of pancreatic polypeptide (PP) reduced food intake and altered stress-induced motor activity and anxiety by activating central Y4 receptors. Exptl. approach : We characterized [K30(PEG2)]hPP2-36 as long-acting Y4 receptor agonist and injected it peripherally into wildtype and Y4 receptor knockout (Y4KO) C57Bl/6NCrl mice to investigate the role of Y4 receptors in fear conditioning. Extinction and relapse after extinction was measured by spontaneous recovery and renewal. Key results : The Y4KO mice showed impaired cued and context fear extinction without affecting acquisition, consolidation or recall of fear. Correspondingly, peripheral injection of [K30(PEG2)]hPP2-36 facilitated extinction learning upon fasting, an effect that was long-lasting and generalized. Furthermore, peripherally applied [K30(PEG2)]hPP2-36 before extinction inhibited the activation of orexin-expressing neurons in the lateral hypothalamus in WT, but not in Y4KO mice. Conclusions and implications : Our findings suggests suppression of excessive arousal as a possible mechanism for the extinction-promoting effect of central Y4 receptors and provide strong evidence that fear extinction requires integration of vegetative stimuli with cortical and subcortical information, a process crucially depending on Y4 receptors. Importantly, in the lateral hypothalamus two peptide systems, PP and orexin, interact to generate an emotional response adapted to the current homeostatic state. Detailed investigations of feeding-relevant genes may thus deliver multiple intervention points for treating anxiety-related disorders.
- 14Zhang, L.; Bijker, M. S.; Herzog, H. The neuropeptide Y system: Pathophysiological and therapeutic implications in obesity and cancer. Pharmacol. Ther. 2011, 131, 91– 113, DOI: 10.1016/j.pharmthera.2011.03.01114The neuropeptide Y system: Pathophysiological and therapeutic implications in obesity and cancerZhang, Lei; Bijker, Martijn S.; Herzog, HerbertPharmacology & Therapeutics (2011), 131 (1), 91-113CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)A review. The neuropeptide Y (NPY) system - comprising of neuropeptide Y, peptide YY, pancreatic polypeptide and the corresponding Y receptors through which they act (Y1, Y2, Y4, Y5 and y6) - is well known for its role in the regulation of energy homeostasis and assocd. processes. Dysfunctions of the system have been implicated in human diseases such as obesity and cancer, raising the possibility that correction of the system may provide therapeutic benefits for these diseases. In addn. to the regulation of appetite and satiety that has attracted most attention during the past years, insight has also been gained into the crit. role of NPY in the control of energy expenditure, oxidative fuel selection and bone metab. Studies using conditional knockout models further shed light on the central vs. peripheral, and hypothalamic vs. extra-hypothalamic mechanisms of these regulatory effects of NPY. Moreover, a role of NPY family peptides and Y receptors in modulating the growth of tumors has emerged. These findings provide the basis for novel NPY system-targeted strategies to treat obesity as well as cancer. Such strategies include modifying both sides of the energy balance equation - energy intake vs. energy expenditure - to achieve a greater wt./fat loss by particularly modulating peripheral Y receptor(s) to ameliorate metabolic conditions without interfering with central functions of Y receptors. In addn., targeting multiple Y receptors and/or multiple systems involved in the regulation of energy balance will have greater beneficial effects. However, long-term interference with the NPY system to target obesity or cancer related aspects needs to consider potential side effects on bone health.
- 15Yi, M.; Li, H.; Wu, Z.; Yan, J.; Liu, Q.; Ou, C.; Chen, M. A promising therapeutic target for metabolic diseases: Neuropeptide Y receptors in humans. Cell. Physiol. Biochem. 2018, 45, 88– 107, DOI: 10.1159/00048622515A Promising Therapeutic Target for Metabolic Diseases: Neuropeptide Y Receptors in HumansYi, Min; Li, Hekai; Wu, Zhiye; Yan, Jianyun; Liu, Qicai; Ou, Caiwen; Chen, MinshengCellular Physiology and Biochemistry (2018), 45 (1), 88-107CODEN: CEPBEW; ISSN:1015-8987. (S. Karger AG)Human neuropeptide Y (hNPY) is one of the most widely expressed neurotransmitters in the human central and peripheral nervous systems. It consists of 36 highly conserved amino acid residues, and was first isolated from the porcine hypothalamus in 1982. While it is the most recently discovered member of the pancreatic polypeptide family (which includes neuropeptide Y, gut-derived hormone peptide YY, and pancreatic polypeptide), NPY is the most abundant peptide found in the mammalian brain. In order to exert particular functions, NPY needs to bind to the NPY receptor to activate specific signaling pathways. NPY receptors belong to the class A or rhodopsin-like G-protein coupled receptor (GPCR) family and signal via cell-surface receptors. By binding to GPCRs, NPY plays a crucial role in various biol. processes, including cortical excitability, stress response, food intake, circadian rhythms, and cardiovascular function. Abnormal regulation of NPY is involved in the development of a wide range of diseases, including obesity, hypertension, atherosclerosis, epilepsy, metabolic disorders, and many cancers. Thus far, five receptors have been cloned from mammals (Y1, Y2, Y4, Y5, and y6), but only four of these (hY1, hY2, hY4, and hY5) are functional in humans. In this review, we summarize the structural characteristics of human NPY receptors and their role in metabolic diseases.
- 16Allen, M.; Reeves, J.; Mellor, G. High throughput fluorescence polarization: a homogeneous alternative to radioligand binding for cell surface receptors. J. Biomol. Screening 2000, 5, 63– 69, DOI: 10.1177/10870571000050020216High throughput fluorescence polarization: a homogeneous alternative to radioligand binding for cell surface receptorsAllen, Michael; Reeves, Julian; Mellor, GeoffreyJournal of Biomolecular Screening (2000), 5 (2), 63-69CODEN: JBISF3; ISSN:1087-0571. (Mary Ann Liebert, Inc.)High throughput fluorescence polarization (FP) assays are described that offer a nonradioactive, homogeneous, and low-cost alternative to radioligand binding assays for cell surface receptors (G protein-coupled receptors and ligand-gated ion channels). FP assays were shown to work across a range of both peptide (vasopressin V1a and δ-opioid) and nonpeptide (β1-adrenoceptor, 5-hydroxytryptamine3) receptors. Structure-activity relationships were investigated at β1-receptors and were found to be consistent with radioligand binding assays. FP was shown to tolerate up to 5% DMSO with no loss in sensitivity or signal window. From a random set of 1,280 compds., 1.9% were found to significantly interfere with FP measurement. If fluorescent or quenching compds. were eliminated (3% of all compds.), less than 0.4% of compds. were found to interfere with FP measurement. Assays could be run in 384-well plates with little loss of signal window or sensitivity compared to 96-well plate assays. New advances in FP measurement have therefore enabled FP to offer a high throughput alternative to radioligand binding for cell surface receptors.
- 17Leyris, J.-P.; Roux, T.; Trinquet, E.; Verdié, P.; Fehrentz, J.-A.; Oueslati, N.; Douzon, S.; Bourrier, E.; Lamarque, L.; Gagne, D. Homogeneous time-resolved fluorescence-based assay to screen for ligands targeting the growth hormone secretagogue receptor type 1a. Anal. Biochem. 2011, 408, 253– 262, DOI: 10.1016/j.ab.2010.09.03017Homogeneous time-resolved fluorescence-based assay to screen for ligands targeting the growth hormone secretagogue receptor type 1aLeyris, Jean-Philippe; Roux, Thomas; Trinquet, Eric; Verdie, Pascal; Fehrentz, Jean-Alain; Oueslati, Nadia; Douzon, Stephanie; Bourrier, Emmanuel; Lamarque, Laurent; Gagne, Didier; Galleyrand, Jean-Claude; M'kadmi, Celine; Martinez, Jean; Mary, Sophie; Baneres, Jean-Louis; Marie, JackyAnalytical Biochemistry (2011), 408 (2), 253-262CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)The growth hormone secretagogue receptor type 1a (GHS-R1a) belongs to class A G-protein-coupled receptors (GPCR). This receptor mediates pleiotropic effects of ghrelin and represents a promising target for dysfunctions of growth hormone secretion and energy homeostasis including obesity. Identification of new compds. which bind GHS-R1a is traditionally achieved using radioactive binding assays. Here we propose a new fluorescence-based assay, called Tag-lite binding assay, based on a fluorescence resonance energy transfer (FRET) process between a terbium cryptate covalently attached to a SNAP-tag fused GHS-R1a (SNAP-GHS-R1a) and a high-affinity red fluorescent ghrelin ligand. The long fluorescence lifetime of the terbium cryptate allows a time-resolved detection of the FRET signal. The assay was made compatible with high-throughput screening by using prelabeled cells in suspension under a 384-well plate format. Ki values for a panel of 14 compds. displaying agonist, antagonist, or inverse agonist properties were detd. using both the radioactive and the Tag-lite binding assays performed on the same batches of GHS-R1a-expressing cells. Compd. potencies obtained in the two assays were nicely correlated. This study is the first description of a sensitive and reliable nonradioactive binding assay for GHS-R1a in a format amenable to high-throughput screening.
- 18Emami-Nemini, A.; Roux, T.; Leblay, M.; Bourrier, E.; Lamarque, L.; Trinquet, E.; Lohse, M. J. Time-resolved fluorescence ligand binding for G protein–coupled receptors. Nat. Protoc. 2013, 8, 1307– 1320, DOI: 10.1038/nprot.2013.07318Time-resolved fluorescence ligand binding for G protein-coupled receptorsEmami-Nemini, Alexander; Roux, Thomas; Leblay, Marion; Bourrier, Emmanuel; Lamarque, Laurent; Trinquet, Eric; Lohse, Martin J.Nature Protocols (2013), 8 (7), 1307-1320, 14 pp.CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)G protein-coupled receptors (GPCRs) and their ligands are traditionally characterized by radioligand-binding expts. These expts. yield excellent quant. data, but have low temporal and spatial resoln. In addn., the use of radioligands presents safety concerns. Here we provide a general procedure for an alternative approach with high temporal and spatial resoln., based on Tb+-labeled fluorescent receptor ligands and time-resolved fluorescence resonance energy transfer (TR-FRET). This protocol and its design are detailed here for the parathyroid hormone receptor, a class B GPCR, and its fluorescently labeled 34-amino acid peptide ligand, but it can be easily modified for other receptors and their appropriately labeled ligands. We discuss three protocol options that use Tb+-labeled fluorescent ligands: a time-resolved fluorescence sepn. option that works on native receptors but requires sepn. of bound and unbound ligand; a TR-FRET option using SNAP-tag-labeled receptors for high-throughput screening; and a TR-FRET option that uses fluorescently labeled antibodies directed against an epitope engineered into the Flag-labeled receptors' N terminus. These protocol options can be used as std. procedures with very high signal-to-background ratios in order to characterize ligands and their receptors in living cells and in cell membranes via straightforward plate-reader measurements.
- 19Veiksina, S.; Kopanchuk, S.; Rinken, A. Budded baculoviruses as a tool for a homogeneous fluorescence anisotropy-based assay of ligand binding to G protein-coupled receptors: The case of melanocortin 4 receptors. Biochim. Biophys. Acta, Biomembr. 2014, 1838, 372– 381, DOI: 10.1016/j.bbamem.2013.09.01519Budded baculoviruses as a tool for a homogeneous fluorescence anisotropy-based assay of ligand binding to G protein-coupled receptors: The case of melanocortin 4 receptorsVeiksina, Santa; Kopanchuk, Sergei; Rinken, AgoBiochimica et Biophysica Acta, Biomembranes (2014), 1838 (1PB), 372-381CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)We present here the implementation of budded baculoviruses that display G protein-coupled receptors on their surfaces for the investigation of ligand-receptor interactions using fluorescence anisotropy (FA). Melanocortin 4 (MC4) receptors and the fluorescent ligand Cy3B-NDP-α-MSH were used as the model system. The real-time monitoring of reactions and the high assay quality allow the application of global data anal. with kinetic mechanistic models that take into account the effect of nonspecific interactions and the depletion of the fluorescent ligand during the reaction. The receptor concn., affinity and kinetic parameters of fluorescent ligand binding as well as state anisotropies for different fluorescent ligand populations were detd. At low Cy3B-NDP-α-MSH concns., a one-site receptor-ligand binding model described the processes, whereas divergence from this model was obsd. at higher ligand concns., which indicated a more complex mechanism of interactions similar to those mechanisms that have been found in expts. with radioactive ligands. The information obtained from our kinetic expts. and the inherent flexibility of FA assays also allowed the estn. of binding parameters for several MC4 receptor-specific unlabeled compds. In summary, the FA assay that was developed with budded baculoviruses led the exptl. data to a level that would solve complex models of receptor-ligand interactions also for other receptor systems and would become as a valuable tool for the screening of pharmacol. active compds.
- 20Schiele, F.; Ayaz, P.; Fernández-Montalván, A. A universal homogeneous assay for high-throughput determination of binding kinetics. Anal. Biochem. 2015, 468, 42– 49, DOI: 10.1016/j.ab.2014.09.00720A universal homogeneous assay for high-throughput determination of binding kineticsSchiele, Felix; Ayaz, Pelin; Fernandez-Montalvan, AmauryAnalytical Biochemistry (2015), 468 (), 42-49CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)There is an increasing demand for assay technologies that enable accurate, cost-effective, and high-throughput measurements of drug-target assocn. and dissocn. rates. Here the authors introduce a universal homogeneous kinetic probe competition assay (kPCA) that meets these requirements. The time-resolved fluorescence energy transfer (TR-FRET) procedure combines the versatility of radioligand binding assays with the advantages of homogeneous nonradioactive techniques while approaching the time resoln. of surface plasmon resonance (SPR) and related biosensors. The authors show application of kPCA for three important target classes: enzymes, protein-protein interactions, and G protein-coupled receptors (GPCRs). This method is capable of supporting early stages of drug discovery with large amts. of kinetic information.
- 21Antoine, T.; Ott, D.; Ebell, K.; Hansen, K.; Henry, L.; Becker, F.; Hannus, S. Homogeneous time-resolved G protein-coupled receptor-ligand binding assay based on fluorescence cross-correlation spectroscopy. Anal. Biochem. 2016, 502, 24– 35, DOI: 10.1016/j.ab.2016.02.01721Homogeneous time-resolved G protein-coupled receptor-ligand binding assay based on fluorescence cross-correlation spectroscopyAntoine, Thomas; Ott, David; Ebell, Katharina; Hansen, Kerrin; Henry, Luc; Becker, Frank; Hannus, StefanAnalytical Biochemistry (2016), 502 (), 24-35CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)G protein-coupled receptors (GPCRs) mediate many important physiol. functions and are considered as one of the most successful therapeutic target classes for a wide spectrum of diseases. Drug discovery projects generally benefit from a broad range of exptl. approaches for screening compd. libraries and for the characterization of binding modes of drug candidates. Owing to the difficulties in solubilizing and purifying GPCRs, assay formats have been so far mainly limited to cell-based functional assays and radioligand binding assays. In this study, we used fluorescence cross-correlation spectroscopy (FCCS) to analyze the interaction of detergent-solubilized receptors to various types of GPCR ligands: endogenous peptides, small mols., and a large surrogate antagonist represented by a blocking monoclonal antibody. Our work demonstrates the suitability of the homogeneous and time-resolved FCCS assay format for a robust, high-throughput detn. of receptor-ligand binding affinities and kinetic rate consts. for various therapeutically relevant GPCRs.
- 22Stoddart, L. A.; White, C. W.; Nguyen, K.; Hill, S. J.; Pfleger, K. D. Fluorescence- and bioluminescence-based approaches to study GPCR ligand binding. Br. J. Pharmacol. 2016, 173, 3028– 3037, DOI: 10.1111/bph.1331622Fluorescence- and bioluminescence-based approaches to study GPCR ligand bindingStoddart, Leigh A.; White, Carl W.; Nguyen, Kim; Hill, Stephen J.; Pfleger, Kevin D. G.British Journal of Pharmacology (2016), 173 (20), 3028-3037CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)Ligand binding is a vital component of any pharmacologist's toolbox and allows the detailed investigation of how a mol. binds to its receptor. These studies enable the exptl. detn. of binding affinity of labeled and unlabeled compds. through kinetic, satn. (Kd) and competition (Ki) binding assays. Traditionally, these studies have used mols. labeled with radioisotopes; however, more recently, fluorescent ligands have been developed for this purpose. This review will briefly cover receptor ligand binding theory and then discuss the use of fluorescent ligands with some of the different technologies currently employed to examine ligand binding. Fluorescent ligands can be used for direct measurement of receptor-assocd. fluorescence using confocal microscopy and flow cytometry as well as in assays such as fluorescence polarization, where ligand binding is monitored by changes in the free rotation when a fluorescent ligand is bound to a receptor. Addnl., fluorescent ligands can act as donors or acceptors for fluorescence resonance energy transfer (FRET) with the development of assays based on FRET and time-resolved FRET (TR-FRET). Finally, we have recently developed a novel bioluminescence resonance energy transfer (BRET) ligand binding assay utilizing a small (19 kDa), super-bright luciferase subunit (NanoLuc) from a deep sea shrimp. In combination with fluorescent ligands, measurement of RET now provides an array of methodologies to study ligand binding. While each method has its own advantages and drawbacks, binding studies using fluorescent ligands are now a viable alternative to the use of radioligands.
- 23Stoddart, L. A.; Kilpatrick, L. E.; Hill, S. J. NanoBRET approaches to study ligand binding to GPCRs and RTKs. Trends Pharmacol. Sci. 2018, 39, 136– 147, DOI: 10.1016/j.tips.2017.10.00623NanoBRET Approaches to Study Ligand Binding to GPCRs and RTKsStoddart, Leigh A.; Kilpatrick, Laura E.; Hill, Stephen J.Trends in Pharmacological Sciences (2018), 39 (2), 136-147CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)Recent advances in the development of fluorescent ligands for G-protein-coupled receptors (GPCRs) and receptor tyrosine kinase receptors (RTKs) have facilitated the study of these receptors in living cells. A limitation of these ligands is potential uptake into cells and increased nonspecific binding. However, this can largely be overcome by using proximity approaches, such as bioluminescence resonance energy transfer (BRET), which localize the signal (within 10 nm) to the specific receptor target. The recent engineering of NanoLuc has resulted in a luciferase variant that is smaller and significantly brighter (up to tenfold) than existing variants. Here, we review the use of BRET from N-terminal NanoLuc-tagged GPCRs or a RTK to a receptor-bound fluorescent ligand to provide quant. pharmacol. of ligand-receptor interactions in living cells in real time.
- 24Iliopoulos-Tsoutsouvas, C.; Kulkarni, R. N.; Makriyannis, A.; Nikas, S. P. Fluorescent probes for G-protein-coupled receptor drug discovery. Expert Opin. Drug Discovery 2018, 13, 933– 947, DOI: 10.1080/17460441.2018.151897524Fluorescent probes for G-protein-coupled receptor drug discoveryIliopoulos-Tsoutsouvas, Christos; Kulkarni, Rohit N.; Makriyannis, Alexandros; Nikas, Spyros P.Expert Opinion on Drug Discovery (2018), 13 (10), 933-947CODEN: EODDBX; ISSN:1746-0441. (Taylor & Francis Ltd.)A review. G-protein-coupled receptors (GPCRs) mediate the effects of approx. 33% of all marketed drugs. The development of tools to study GPCR pharmacol. is urgently needed as it can lead to the discovery of safer and more effective medications. Fluorescent GPCR ligands represent highly sensitive and safe small-mol. tools for real-time exploration of the life of the receptor, cellular signaling, and ligand-/receptor-receptor interactions in cellulo and/or in vivo.: This review summarizes relevant information from published literature and provides crit. insights into the design of successful small-mol. fluorescent probes for Class A GPCRs as potential major targets for drug development.: Considering the rapid progress of fluorescence technologies, effective small-mol. fluorescent probes represent valuable pharmacol. tools for studying GPCRs. However, the design and development of such probes are challenging, largely due to the low affinity/specificity of the probe for its target, inadequate photophys. properties, extensive non-specific binding, and/or low signal-to-noise ratio. Generally speaking, fluorescent and luminescent small-mol. probes, receptors, and G proteins in combination with FRET and BRET technologies hold great promise for studying kinetic profiles of GPCR signaling.
- 25Ziemek, R.; Schneider, E.; Kraus, A.; Cabrele, C.; Beck-Sickinger, A. G.; Bernhardt, G.; Buschauer, A. Determination of affinity and activity of ligands at the human neuropeptide Y Y4 receptor by flow cytometry and aequorin luminescence. J. Recept. Signal Transduction 2007, 27, 217– 233, DOI: 10.1080/1079989070150520625Determination of Affinity and Activity of Ligands at the Human Neuropeptide Y Y4 Receptor by Flow Cytometry and Aequorin LuminescenceZiemek, Ralf; Schneider, Erich; Kraus, Anja; Cabrele, Chiara; Beck-Sickinger, Annette G.; Bernhardt, Guenther; Buschauer, ArminJournal of Receptors and Signal Transduction (2007), 27 (4), 217-233CODEN: JRSTCT ISSN:. (Informa Healthcare)Fluorescence-labeled neuropeptide Y (NPY) has been used in flow cytometric binding assays for the detn. of affinity consts. of NPY Y1, Y2, and Y5 receptor ligands. Because the binding of fluorescent NPY is insufficient for competition studies at the human Y4 receptor (hY4R), the authors replaced Glu 4 in hPP with Lys for the derivatization with cyanine-5. Because cy5-[K4]hPP has high affinity (Kd 5.6 nM) to the hY4R, it was used as a probe in a flow cytometric binding assay. Specific binding of cy5-[K4]hPP to hY4R was visualized by confocal microscopy. The hY4R, the chimeric G protein Gqi5 and mitochondrially targeted apoaequorin were stably coexpressed in CHO cells. Aequorin luminescence was quantified in a microplate reader and by a CCD camera. By application of these methods 3-cyclohexyl-N-[(3-1H-imidazol-4-ylpropylamino)(imino)methyl]propanamide (UR-AK49)was discovered as the first nonpeptidic Y4R antagonist (pKi 4.17), a lead to be optimized in terms of potency and selectivity.
- 26Dukorn, S.; Littmann, T.; Keller, M.; Kuhn, K.; Cabrele, C.; Baumeister, P.; Bernhardt, G.; Buschauer, A. Fluorescence- and radiolabeling of [Lys4,Nle17,30]hPP yields molecular tools for the NPY Y4 receptor. Bioconjugate Chem. 2017, 28, 1291– 1304, DOI: 10.1021/acs.bioconjchem.7b0010326Fluorescence- and Radiolabeling of [Lys4,Nle17,30]hPP Yields Molecular Tools for the NPY Y4 ReceptorDukorn, Stefanie; Littmann, Timo; Keller, Max; Kuhn, Kilian; Cabrele, Chiara; Baumeister, Paul; Bernhardt, Guenther; Buschauer, ArminBioconjugate Chemistry (2017), 28 (4), 1291-1304CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The neuropeptide Y (NPY) Y4 receptor (Y4R) is involved in energy homeostasis and considered a potential drug target for the treatment of obesity. Only a few mol. tools, i.e., radiolabeled and fluorescent ligands, for the investigation of the Y4R were reported. Previously, [Lys4]hPP proved to be an appropriate full-length PP analog to prep. a fluorescent ligand by derivatization at the ε-amino group. To preclude oxidn. upon long-term storage, the authors replaced the two methionine residues in [Lys4]hPP by norleucine and prepd. the corresponding [3H]propionylated ([3H]12) and cyanine labeled (13) peptides, which were characterized and compared with a set of ref. compds. in binding (Y1, Y2, Y4, and Y5 receptors) and functional (luciferase gene reporter, beta-arrestin-1,2) Y4R assays. Both mol. probes proved to be useful in radiochem. and flow cytometric satn. and competition Y4R binding expts. Most strikingly, there was a different influence of the compn. of buffer on equil. binding and kinetics: [3H]12 affinity (Kd in Na+-free buffer: 1.1 nM) clearly decreased with increasing sodium ion concn., whereas dissocn. and Y4R-mediated internalization of 13 (Kd in Na+-free buffer: 10.8 nM) were strongly affected by the osmolarity of the buffer as demonstrated by confocal microscopy. Displacement of [3H]12 and 13 revealed a tendency to higher apparent affinities for a set of ref. peptides in hypotonic (Na+-free) compared to isotonic buffers. The differences were negligible in the case of hPP but up to 270-fold in the case of GW1229 (GR231118). By contrast, no relevant influence of Na+ on Y5R affinity became obvious, when the radioligands [H]12 and [3H]propionyl-pNPY were investigated in satn. binding and competition binding.
- 27Tang, T.; Tan, Q.; Han, S.; Diemar, A.; Löbner, K.; Wang, H.; Schüß, C.; Behr, V.; Mörl, K.; Wang, M.; Chu, X.; Yi, C.; Keller, M.; Kofoed, J.; Reedtz-Runge, S.; Kaiser, A.; Beck-Sickinger, A. G.; Zhao, Q.; Wu, B. Receptor-specific recognition of NPY peptides revealed by structures of NPY receptors. Sci. Adv. 2022, 8, eabm1232 DOI: 10.1126/sciadv.abm1232There is no corresponding record for this reference.
- 28Kuhn, K. K.; Littmann, T.; Dukorn, S.; Tanaka, M.; Keller, M.; Ozawa, T.; Bernhardt, G.; Buschauer, A. In search of NPY Y4R antagonists: Incorporation of carbamoylated arginine, aza-amino acids, or D-amino acids into oligopeptides derived from the C-termini of the endogenous agonists. ACS Omega 2017, 2, 3616– 3631, DOI: 10.1021/acsomega.7b0045128In Search of NPY Y4R Antagonists: Incorporation of Carbamoylated Arginine, Aza-Amino Acids, or D-Amino Acids into Oligopeptides Derived from the C-Termini of the Endogenous AgonistsKuhn, Kilian K.; Littmann, Timo; Dukorn, Stefanie; Tanaka, Miho; Keller, Max; Ozawa, Takeaki; Bernhardt, Guenther; Buschauer, ArminACS Omega (2017), 2 (7), 3616-3631CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)The crosslinked pentapeptides (2R,7R)-diaminooctanedioyl-bis(Tyr-Arg-Leu-Arg-Tyr-amide) ((2R,7R)-BVD-74D, (2R,7R)-1) and octanedioyl-bis(Tyr-Arg-Leu-Arg-Tyr-amide) (2) as well as the pentapeptide Ac-Tyr-Arg-Leu-Arg-Tyr-amide (3) were previously described as neuropeptide Y Y4 receptor (Y4R) partial agonists. Here the authors report on a series of analogs of (2R,7R)-1 and 2 in which Arg2, Leu3 or Arg4 were replaced by the resp. aza-amino acids. The replacement of Arg2 in 3 with a carbamoylated arginine building block and the extension of the N-terminus by an addnl. arginine led to the high-affinity hexapeptide Ac-Arg-Tyr-Nω-[(4-aminobutyl)aminocarbonyl]Arg-Leu-Arg-Tyr-amide that was used as a precursor for a D-amino acid scan. The target compds. were investigated for Y4R functional activity in assays with complementary readouts: aequorin Ca2+ and β-arrestin 1 or β-arrestin 2 assays. In contrast to the parent compds., which are Y4R agonists, several ligands were able to suppress the effect elicited by the endogenous ligand pancreatic polypeptide and therefore represent a novel class of peptide Y4R antagonists.
- 29Spinnler, K.; von Krüchten, L.; Konieczny, A.; Schindler, L.; Bernhardt, G.; Keller, M. An alkyne-functionalized arginine for solid-phase synthesis enabling “bioorthogonal” peptide conjugation. ACS Med. Chem. Lett. 2020, 11, 334– 339, DOI: 10.1021/acsmedchemlett.9b0038829An alkyne-functionalized arginine for solid-phase synthesis enabling "bioorthogonal" peptide conjugationSpinnler, Katrin; von Kruechten, Lara; Konieczny, Adam; Schindler, Lisa; Bernhardt, Guenther; Keller, MaxACS Medicinal Chemistry Letters (2020), 11 (3), 334-339CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Lately, amino-functionalized Nω-carbamoylated arginines were introduced as arginine surrogates enabling peptide labeling. However, this approach is hardly compatible with peptides also contg. lysine or cysteine. Here, we present the synthesis of an alkyne-functionalized, Nω-carbamoylated arginine building block, which is compatible with Fmoc-strategy solid-phase peptide synthesis. The alkynylated arginine was incorporated into three biol. active linear hexapeptides and into a cyclic pentapeptide. Peptide conjugation to an azido-functionalized fluorescent dye via "click" chem. was successfully demonstrated. In the case of a peptide also contg. lysine besides the alkyne-functionalized arginine, this was feasible in a "bioorthogonal" manner.
- 30Konieczny, A.; Conrad, M.; Ertl, F. J.; Gleixner, J.; Gattor, A. O.; Grätz, L.; Schmidt, M. F.; Neu, E.; Horn, A. H. C.; Wifling, D.; Gmeiner, P.; Clark, T.; Sticht, H.; Keller, M. N-terminus to arginine side-chain cyclization of linear peptidic neuropeptide Y Y4 receptor ligands results in picomolar binding constants. J. Med. Chem. 2021, 64, 16746– 16769, DOI: 10.1021/acs.jmedchem.1c0157430N-Terminus to arginine side-chain cyclization of linear peptidic neuropeptide Y Y4 receptor ligands results in picomolar binding constantsKonieczny, Adam; Conrad, Marcus; Ertl, Fabian J.; Gleixner, Jakob; Gattor, Albert O.; Graetz, Lukas; Schmidt, Maximilian F.; Neu, Eduard; Horn, Anselm H. C.; Wifling, David; Gmeiner, Peter; Clark, Timothy; Sticht, Heinrich; Keller, MaxJournal of Medicinal Chemistry (2021), 64 (22), 16746-16769CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The family of neuropeptide Y (NPY) receptors comprises four subtypes (Y1R, Y2R, Y4R, Y5R), which are addressed by at least three endogenous peptides, i.e., NPY, peptide YY, and pancreatic polypeptide (PP), the latter showing a preference for Y4R. A series of cyclic oligopeptidic Y4R ligands were prepd. by applying a novel approach, i.e., N-terminus to arginine side-chain cyclization. Most peptides acted as Y4R partial agonists, showing up to 60-fold higher Y4R affinity compared to the linear precursor peptides. Two cyclic hexapeptides (I) [cyclo[succinyl-Arg-Tyr-Arg(CO-NH-(CH2)4-NH)]-Leu-Arg-Tyr-NH2] and (II) [cyclo[succinyl-Arg-Trp-Arg(CO-NH-(CH2)4-NH)]-Leu-Arg-Tyr-NH2] showed higher Y4R potency (Ca2+ aequorin assay) and, with pKi values >10, also higher Y4R affinity compared to human pancreatic polypeptide (hPP). Compds. such as I and II exhibiting considerably lower mol. wt. and considerably more pronounced Y4R selectivity than PP and previously described dimeric peptidic ligands with high Y4R affinity, represent promising leads for the prepn. of labeled tool compds. and might support the development of drug-like Y4R ligands.
- 31Gleixner, J.; Gattor, A. O.; Humphrys, L. J.; Brunner, T.; Keller, M. 3H]UR-JG102 - a radiolabeled cyclic peptide with high affinity and excellent selectivity for the neuropeptide Y Y4 receptor. J. Med. Chem. 2023, 66, 13788– 13808, DOI: 10.1021/acs.jmedchem.3c01224There is no corresponding record for this reference.
- 32Keller, M.; Kuhn, K. K.; Einsiedel, J.; Hübner, H.; Biselli, S.; Mollereau, C.; Wifling, D.; Svobodová, J.; Bernhardt, G.; Cabrele, C.; Vanderheyden, P. M. L.; Gmeiner, P.; Buschauer, A. Mimicking of arginine by functionalized Nω-carbamoylated arginine as a new broadly applicable approach to labeled bioactive peptides: high affinity angiotensin, neuropeptide Y, neuropeptide FF, and neurotensin receptor ligands as examples. J. Med. Chem. 2016, 59, 1925– 1945, DOI: 10.1021/acs.jmedchem.5b0149532Mimicking of Arginine by Functionalized Nω-Carbamoylated Arginine As a New Broadly Applicable Approach to Labeled Bioactive Peptides: High Affinity Angiotensin, Neuropeptide Y, Neuropeptide FF, and Neurotensin Receptor Ligands As ExamplesKeller, Max; Kuhn, Kilian K.; Einsiedel, Juergen; Huebner, Harald; Biselli, Sabrina; Mollereau, Catherine; Wifling, David; Svobodova, Jaroslava; Bernhardt, Guenther; Cabrele, Chiara; Vanderheyden, Patrick M. L.; Gmeiner, Peter; Buschauer, ArminJournal of Medicinal Chemistry (2016), 59 (5), 1925-1945CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Derivatization of biol. active peptides by conjugation with fluorophores or radionuclide-bearing moieties is an effective and commonly used approach to prep. mol. tools and diagnostic agents. Whereas lysine, cysteine, and N-terminal amino acids have been mostly used for peptide conjugation, we describe a new, widely applicable approach to peptide conjugation based on the nonclassical bioisosteric replacement of the guanidine group in arginine by a functionalized carbamoylguanidine moiety. Four arginine-contg. peptide receptor ligands (angiotensin II, neurotensin(8-13), an analog of the C-terminal pentapeptide of neuropeptide Y, and a neuropeptide FF analog) were subject of this proof-of-concept study. The Nω-carbamoylated arginines, bearing spacers with a terminal amino group, were incorporated into the peptides by std. Fmoc solid phase peptide synthesis. The synthesized chem. stable peptide derivs. showed high receptor affinities with Ki values in the low nanomolar range, even when bulky fluorophores had been attached. Two new tritiated tracers for angiotensin and neurotensin receptors are described.
- 33Keller, M.; Weiss, S.; Hutzler, C.; Kuhn, K. K.; M?llereau, C.; Dukorn, S.; Schindler, L.; Bernhardt, G.; König, B.; Buschauer, A. Nω-carbamoylation of the argininamide moiety: An avenue to insurmountable NPY Y1 receptor antagonists and a radiolabeled selective high-affinity molecular tool ([3H]UR-MK299) with extended residence time. J. Med. Chem. 2015, 58, 8834– 8849, DOI: 10.1021/acs.jmedchem.5b0092533Nω-Carbamoylation of the Argininamide Moiety: An Avenue to Insurmountable NPY Y1 Receptor Antagonists and a Radiolabeled Selective High-Affinity Molecular Tool ([3H]UR-MK299) with Extended Residence TimeKeller, Max; Weiss, Stefan; Hutzler, Christoph; Kuhn, Kilian K.; Mollereau, Catherine; Dukorn, Stefanie; Schindler, Lisa; Bernhardt, Guenther; Koenig, Burkhard; Buschauer, ArminJournal of Medicinal Chemistry (2015), 58 (22), 8834-8849CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Analogs of the argininamide-type NPY Y1 receptor (Y1R) antagonist BIBP3226, bearing carbamoyl moieties at the guanidine group, revealed subnanomolar Ki values and caused depression of the maximal response to NPY (calcium assay) by up to 90% in a concn.- and time-dependent manner, suggesting insurmountable antagonism. To gain insight into the mechanism of binding of the synthesized compds., a tritiated antagonist, (R)-Nα-diphenylacetyl-Nω-[2-([2,3-3H]propionylamino)ethyl]aminocarbonyl-(4-hydroxybenzyl)arginin-amide ([3H]UR-MK299, [3H]38), was prepd. [3H]38 revealed a dissocn. const. in the picomolar range (Kd 0.044 nM, SK-N-MC cells) and very high Y1R selectivity. Apart from superior affinity, a considerably lower target off-rate (t1/2 95 min) was characteristic of [3H]38 compared to that of the higher homolog contg. a tetramethylene instead of an ethylene spacer (t1/2 3 min, Kd 2.0 nM). Y1R binding of [3H]38 was fully reversible and fully displaceable by nonpeptide antagonists and the agonist pNPY. Therefore, the insurmountable antagonism obsd. in the functional assay has to be attributed to the extended target-residence time, a phenomenon of relevance in drug research beyond the NPY receptor field.
- 34Konieczny, A.; Braun, D.; Wifling, D.; Bernhardt, G.; Keller, M. Oligopeptides as neuropeptide Y Y4 receptor ligands: identification of a high-affinity tetrapeptide agonist and a hexapeptide antagonist. J. Med. Chem. 2020, 63, 8198– 8215, DOI: 10.1021/acs.jmedchem.0c0042634Oligopeptides as Neuropeptide Y Y4 Receptor Ligands: Identification of a High-Affinity Tetrapeptide Agonist and a Hexapeptide AntagonistKonieczny, Adam; Braun, Diana; Wifling, David; Bernhardt, Guenther; Keller, MaxJournal of Medicinal Chemistry (2020), 63 (15), 8198-8215CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Within the family of neuropeptide Y (NPY) receptors, the Y4 receptor (Y4R) is unique as it prefers pancreatic polypeptide (PP) over NPY and peptide YY (PYY). Today, low mol. wt. Y4R ligands are lacking, in particular antagonists. We synthesized a series of peptidic NPY Y4R ligands, derived from the hexapeptide acetyl-Arg-Tyr-Arg-Leu-Arg-Tyr-NH2 (1), reported to be a Y4R partial agonist with high affinity (pKi Y4R: 8.43). Peptide 1 was N-terminally extended as well as truncated, subjected to a D-amino acid scan, and Leu was replaced by different amino acids. Compds. were characterized by radioligand competition binding and functional studies (Cai2+ mobilization and β-arrestin 1/2 recruitment). N-terminal truncation of 1 resulted in a tetrapeptide (Arg-Leu-Arg-Tyr-NH2) being a Y4R partial agonist with retained Y4R affinity (pKi: 8.47). Remarkably, replacement of Leu in 1 and in derivs. of 1 by Trp turned Y4R agonism to antagonism, giving Y4R antagonists with pKi values ≤ 7.57.
- 35Kuhn, K. K.; Ertl, T.; Dukorn, S.; Keller, M.; Bernhardt, G.; Reiser, O.; Buschauer, A. High affinity agonists of the neuropeptide Y (NPY) Y4 receptor derived from the C-terminal pentapeptide of human pancreatic polypeptide (hPP): Synthesis, stereochemical discrimination, and radiolabeling. J. Med. Chem. 2016, 59, 6045– 6058, DOI: 10.1021/acs.jmedchem.6b0030935High Affinity Agonists of the Neuropeptide Y (NPY) Y4 Receptor Derived from the C-Terminal Pentapeptide of Human Pancreatic Polypeptide (hPP): Synthesis, Stereochemical Discrimination, and RadiolabelingKuhn, Kilian K.; Ertl, Thomas; Dukorn, Stefanie; Keller, Max; Bernhardt, Guenther; Reiser, Oliver; Buschauer, ArminJournal of Medicinal Chemistry (2016), 59 (13), 6045-6058CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The diastereomeric mixt. of D/L-2,7-diaminooctanedioyl-bis(YRLRY-NH2) (BVD-74D, 2) was described in the literature as a high affinity Y4 receptor agonist. Here we report on the synthesis and pharmacol. characterization of the pure diastereomers (2R,7R)- and (2S,7S)-2 and a series of homo- and heterodimeric analogs in which octanedioic acid was used as an achiral linker. To investigate the role of the Arg residues, one or two arginines were replaced by Ala. Moreover, Nω-(6-aminohexylaminocarbonyl)Arg was introduced as an arginine replacement (17). (2R,7R)-2 was superior to (2S,7S)-2 in binding and functional cellular assays and equipotent with 17. [3H]Propionylation of one amino group in the linker of (2R,7R)-2 or at the primary amino group in 17 resulted in high affinity Y4R radioligands ([3H]-(2R,7R)-10, [3H]18) with subnanomolar Kd values.
- 36Berlicki, Ł.; Kaske, M.; Gutiérrez-Abad, R.; Bernhardt, G.; Illa, O.; Ortuño, R. M.; Cabrele, C.; Buschauer, A.; Reiser, O. Replacement of Thr32 and Gln34 in the C-terminal neuropeptide Y fragment 25–36 by cis-cyclobutane and cis-cyclopentane β-amino acids shifts selectivity toward the Y4 receptor. J. Med. Chem. 2013, 56, 8422– 8431, DOI: 10.1021/jm400850536Replacement of Thr32 and Gln34 in the C-Terminal Neuropeptide Y Fragment 25-36 by cis-Cyclobutane and cis-Cyclopentane β Amino Acids Shifts Selectivity toward the Y4 ReceptorBerlicki, Lukasz; Kaske, Melanie; Gutierrez-Abad, Raquel; Bernhardt, Guenther; Illa, Ona; Ortuno, Rosa M.; Cabrele, Chiara; Buschauer, Armin; Reiser, OliverJournal of Medicinal Chemistry (2013), 56 (21), 8422-8431CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Neuropeptide Y (NPY) and pancreatic polypeptide (PUP) control central and peripheral processes by activating the G protein coupled receptors YxR (x = 1, 2, 4, 5). We present analogs of the C-terminal fragments 25-36 and 32-36 of NPY and PP contg. (1R,2S)-cyclobutane (βCbu) or (1R,2S)-cyclopentane (βCpe) β-amino acids, which display exclusively Y4R affinity. In particular, [βCpe34]-NPY-(25-36) is a Y4R selective partial agonist (EC50 41 nM, Emax 71%) that binds Y4R with a Ki of 10 nM and a selectivity >100-fold relative to Y1R and Y2R and >50-fold relative to Y5R. Comparably, [Y32, βCpe34]-NPY-(PP)-(32-36) selectively binds and activates Y4R (EC50 94 nM, Emax 73%). The NMR structure of [βCpe34]-NPY-(25-36) in dodecylphosphatidylcholine micelles shows a short helix at residues 27-32, while the C-terminal segment R33βCpe34R35Y36 is extended. The biol. properties of the βCbu- or βCpe-contg. NPY and PP C-terminal fragments encourage the future application of these β-amino acids in the synthesis of selective Y4R ligands.
- 37Wirth, U.; Erl, J.; Azzam, S.; Höring, C.; Skiba, M.; Singh, R.; Hochmuth, K.; Keller, M.; Wegener, J.; König, B. Monitoring the reversibility of GPCR signaling by combining photochromic ligands with label-free impedance analysis. Angew. Chem., Int. Ed. 2023, 62, e202215547 DOI: 10.1002/anie.20221554737Monitoring the Reversibility of GPCR Signaling by Combining Photochromic Ligands with Label-free Impedance AnalysisWirth, Ulrike; Erl, Julia; Azzam, Saphia; Horing, Carina; Skiba, Michael; Singh, Ritu; Hochmuth, Kathrin; Keller, Max; Wegener, Joachim; Konig, BurkhardAngewandte Chemie, International Edition (2023), 62 (21), e202215547CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)G protein-coupled cell surface receptors (GPCR) trigger complex intracellular signaling cascades upon agonist binding. Classic pharmacol. assays provide information about binding affinities, activation or blockade at different stages of the signaling cascade, but real time dynamics and reversibility of these processes remain often disguised. We show that combining photochromic NPY receptor ligands, which can be toggled in their receptor activation ability by irradn. with light of different wavelengths, with whole cell label-free impedance assays allows observing the cell response to receptor activation and its reversibility over time. The concept demonstrated on NPY receptors may be well applicable to many other GPCRs providing a deeper insight into the time course of intracellular signaling processes.
- 38Archipowa, N.; Wittmann, L.; Köckenberger, J.; Ertl, F. J.; Gleixner, J.; Keller, M.; Heinrich, M. R.; Kutta, R. J. Characterization of fluorescent dyes frequently used for bioimaging: Photophysics and photocatalytical reactions with proteins. J. Phys. Chem. B 2023, 127, 9532– 9542, DOI: 10.1021/acs.jpcb.3c04484There is no corresponding record for this reference.
- 39Keller, M.; Mahuroof, S. A.; Yee, V. H.; Carpenter, J.; Schindler, L.; Littmann, T.; Pegoli, A.; Hübner, H.; Bernhardt, G.; Gmeiner, P.; Holliday, N. D. Fluorescence labeling of neurotensin(8–13) via arginine residues gives molecular tools with high receptor affinity. ACS Med. Chem. Lett. 2020, 11, 16– 22, DOI: 10.1021/acsmedchemlett.9b0046239Fluorescence labeling of neurotensin(8-13) via arginine residues gives molecular tools with high receptor affinityKeller, Max; Mahuroof, Shahani A.; Hong Yee, Vivyanne; Carpenter, Jessica; Schindler, Lisa; Littmann, Timo; Pegoli, Andrea; Huebner, Harald; Bernhardt, Guenther; Gmeiner, Peter; Holliday, Nicholas D.ACS Medicinal Chemistry Letters (2020), 11 (1), 16-22CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Fluorescence-labeled receptor ligands have emerged as valuable mol. tools, being indispensable for studying receptor-ligand interactions by fluorescence-based techniques such as high-content imaging, fluorescence microscopy, and fluorescence polarization. Through application of a new labeling strategy for peptides, a series of fluorescent neurotensin(8-13) derivs. was synthesized by attaching red-emitting fluorophores (indolinium- and pyridinium-type cyanine dyes) to carbamoylated arginine residues in neurotensin(8-13) analogs, yielding fluorescent probes with high NTS1R affinity (pKi values: 8.15-9.12) and potency (pEC50 values (Ca2+ mobilization): 8.23-9.43). Selected fluorescent ligands were investigated by flow cytometry and high-content imaging (satn. binding, kinetic studies, and competition binding) as well as by confocal microscopy using intact CHO-hNTS1R cells. The study demonstrates the applicability of the fluorescent probes as mol. tools to obtain, for example, information about the localization of receptors in cells and to det. binding affinities of nonlabeled ligands.
- 40Müller, C.; Gleixner, J.; Tahk, M.-J.; Kopanchuk, S.; Laasfeld, T.; Weinhart, M.; Schollmeyer, D.; Betschart, M. U.; Lüdeke, S.; Koch, P.; Rinken, A.; Keller, M. Structure-based design of high-affinity fluorescent probes for the neuropeptide Y Y1 receptor. J. Med. Chem. 2022, 65, 4832– 4853, DOI: 10.1021/acs.jmedchem.1c0203340Structure-based design of high-affinity fluorescent probes for the neuropeptide Y Y1 receptorMueller, Christoph; Gleixner, Jakob; Tahk, Maris-Johanna; Kopanchuk, Sergei; Laasfeld, Tonis; Weinhart, Michael; Schollmeyer, Dieter; Betschart, Martin U.; Luedeke, Steffen; Koch, Pierre; Rinken, Ago; Keller, MaxJournal of Medicinal Chemistry (2022), 65 (6), 4832-4853CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent crystn. of the neuropeptide Y Y1 receptor (Y1R) in complex with the argininamide-type Y1R selective antagonist UR-MK299 (2) opened up a new approach toward structure-based design of nonpeptidic Y1R ligands. We designed novel fluorescent probes showing excellent Y1R selectivity and, in contrast to previously described fluorescent Y1R ligands, considerably higher (~ 100-fold) binding affinity. This was achieved through the attachment of different fluorescent dyes to the diphenylacetyl moiety in 2 via an amine-functionalized linker. The fluorescent ligands exhibited picomolar Y1R binding affinities (pKi values of 9.36-9.95) and proved to be Y1R antagonists, as validated in a Fura-2 calcium assay. The versatile applicability of the probes as tool compds. was demonstrated by flow cytometry- and fluorescence anisotropy-based Y1R binding studies (satn. and competition binding and assocn. and dissocn. kinetics) as well as by widefield and total internal reflection fluorescence (TIRF) microscopy of live tumor cells, revealing that fluorescence was mainly localized at the plasma membrane.
- 41Keller, M.; Erdmann, D.; Pop, N.; Pluym, N.; Teng, S.; Bernhardt, G.; Buschauer, A. Red-fluorescent argininamide-type NPY Y1 receptor antagonists as pharmacological tools. Biorg. Med. Chem. 2011, 19, 2859– 2878, DOI: 10.1016/j.bmc.2011.03.04541Red-fluorescent argininamide-type NPY Y1 receptor antagonists as pharmacological toolsKeller, Max; Erdmann, Daniela; Pop, Nathalie; Pluym, Nikola; Teng, Shangjun; Bernhardt, Guenther; Buschauer, ArminBioorganic & Medicinal Chemistry (2011), 19 (9), 2859-2878CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Fluorescently labeled NPY Y1 receptor (Y1R) ligands were synthesized by connecting pyrylium and cyanine dyes with the argininamide-type Y1R antagonist core structure by linkers, covering a wide variety in length and chem. nature, attached to the guanidine group. The most promising fluorescent probes had Y1R affinities (radioligand binding) and antagonistic activities (calcium assay) in the one- to two-digit nanomolar range. These compds. turned out to be stable under assay conditions and to be appropriate for the detection of Y1Rs by confocal microscopy in live cells. To improve the signal-to-noise ratio by shifting the emission into the near IR, a new benzothiazolium-type fluorescent cyanine dye (UR-DE99) was synthesized and attached to the parent antagonist via a carbamoyl linker yielding UR-MK131, a highly potent fluorescent Y1R probe, which was also successfully applied in flow cytometry.
- 42She, X.; Pegoli, A.; Gruber, C. G.; Wifling, D.; Carpenter, J.; Hübner, H.; Chen, M.; Wan, J.; Bernhardt, G.; Gmeiner, P.; Holliday, N. D.; Keller, M. Red-emitting dibenzodiazepinone derivatives as fluorescent dualsteric probes for the muscarinic acetylcholine M2 receptor. J. Med. Chem. 2020, 63, 4133– 4154, DOI: 10.1021/acs.jmedchem.9b0217242Red-Emitting Dibenzodiazepinone Derivatives as Fluorescent Dualsteric Probes for the Muscarinic Acetylcholine M2 ReceptorShe, Xueke; Pegoli, Andrea; Gruber, Corinna G.; Wifling, David; Carpenter, Jessica; Huebner, Harald; Chen, Mengya; Wan, Jianfei; Bernhardt, Guenther; Gmeiner, Peter; Holliday, Nicholas D.; Keller, MaxJournal of Medicinal Chemistry (2020), 63 (8), 4133-4154CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fluorescently labeled dibenzodiazepinone-type muscarinic acetylcholine receptor (MR) antagonists, including dimeric ligands, were prepd. using red-emitting cyanine dyes. Probes contg. a fluorophore with neg. charge showed high M2R affinities (pKi (radioligand competition binding): 9.10-9.59). Binding studies at M1 and M3-M5 receptors indicated a M2R preference. Flow cytometric and high-content imaging satn. and competition binding (M1R, M2R, and M4R) confirmed occupation of the orthosteric site. Confocal microscopy revealed that fluorescence was located mainly at the cell membrane (CHO-hM2R cells). Results from dissocn. and satn. binding expts. (M2R) in the presence of allosteric M2R modulators (dissocn.: W84, LY2119620, and alcuronium; satn. binding: W84) were consistent with a competitive mode of action between the fluorescent probes and the allosteric ligands. Taken together, these lines of evidence indicate that these ligands are useful fluorescent mol. tools to label the M2R in imaging and binding studies and suggest that they have a dualsteric mode of action.
- 43Katritch, V.; Fenalti, G.; Abola, E. E.; Roth, B. L.; Cherezov, V.; Stevens, R. C. Allosteric sodium in class A GPCR signaling. Trends Biochem. Sci. 2014, 39, 233– 244, DOI: 10.1016/j.tibs.2014.03.00243Allosteric sodium in class A GPCR signalingKatritch, Vsevolod; Fenalti, Gustavo; Abola, Enrique E.; Roth, Bryan L.; Cherezov, Vadim; Stevens, Raymond C.Trends in Biochemical Sciences (2014), 39 (5), 233-244CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. Despite their functional and structural diversity, G-protein-coupled receptors (GPCRs) share a common mechanism of signal transduction via conformational changes in the seven-transmembrane (7TM) helical domain. New major insights into this mechanism come from the recent crystallog. discoveries of a partially hydrated sodium ion that is specifically bound in the middle of the 7TM bundle of multiple class A GPCRs. This review discusses the remarkable structural conservation and distinct features of the Na+ pocket in this most populous GPCR class, as well as the conformational collapse of the pocket upon receptor activation. New insights help to explain allosteric effects of sodium on GPCR agonist binding and activation, and sodium's role as a potential co-factor in class A GPCR function.
- 44White, K. L.; Eddy, M. T.; Gao, Z. G.; Han, G. W.; Lian, T.; Deary, A.; Patel, N.; Jacobson, K. A.; Katritch, V.; Stevens, R. C. Structural connection between activation microswitch and allosteric sodium site in GPCR Signaling. Structure 2018, 26, 259– 269, DOI: 10.1016/j.str.2017.12.01344Structural Connection between Activation Microswitch and Allosteric Sodium Site in GPCR SignalingWhite, Kate L.; Eddy, Matthew T.; Gao, Zhan-Guo; Han, Gye Won; Lian, Tiffany; Deary, Alexander; Patel, Nilkanth; Jacobson, Kenneth A.; Katritch, Vsevolod; Stevens, Raymond C.Structure (Oxford, United Kingdom) (2018), 26 (2), 259-269.e5CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)Sodium ions are endogenous allosteric modulators of many G-protein-coupled receptors (GPCRs). Mutation of key residues in the sodium binding motif causes a striking effect on G-protein signaling. We report the crystal structures of agonist complexes for two variants in the first sodium coordination shell of the human A2A adenosine receptor, D522.50N and S913.39A. Both structures present an overall active-like conformation; however, the variants show key changes in the activation motif NPxxY. Changes in the hydrogen bonding network in this microswitch suggest a possible mechanism for modified G-protein signaling and enhanced thermal stability. These structures, signaling data, and thermal stability anal. with a panel of pharmacol. ligands provide a basis for understanding the role of the sodium-coordinating residues on stability and G-protein signaling. Utilizing the D2.50N variant is a promising method for stabilizing class A GPCRs to accelerate structural efforts and drug discovery.
- 45DeVree, B. T.; Mahoney, J. P.; Velez-Ruiz, G. A.; Rasmussen, S. G.; Kuszak, A. J.; Edwald, E.; Fung, J. J.; Manglik, A.; Masureel, M.; Du, Y.; Matt, R. A.; Pardon, E.; Steyaert, J.; Kobilka, B. K.; Sunahara, R. K. Allosteric coupling from G protein to the agonist-binding pocket in GPCRs. Nature 2016, 535, 182– 186, DOI: 10.1038/nature1832445Allosteric coupling from G protein to the agonist-binding pocket in GPCRsDeVree, Brian T.; Mahoney, Jacob P.; Velez-Ruiz, Gisselle A.; Rasmussen, Soren G. F.; Kuszak, Adam J.; Edwald, Elin; Fung, Juan-Jose; Manglik, Aashish; Masureel, Matthieu; Du, Yang; Matt, Rachel A.; Pardon, Els; Steyaert, Jan; Kobilka, Brian K.; Sunahara, Roger K.Nature (London, United Kingdom) (2016), 535 (7610), 182-186CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)G protein-coupled receptors (GPCRs) remain the primary conduit by which cells detect environmental stimuli and communicate with each other. Upon activation by extracellular agonists, these seven-transmembrane-domain-contg. receptors interact with heterotrimeric G proteins to regulate downstream second messenger and/or protein kinase cascades. Crystallog. evidence from a prototypic GPCR, the β2-adrenergic receptor (β2AR), in complex with its cognate G protein, Gs, has provided a model for how agonist binding promotes conformational changes that propagate through the GPCR and into the nucleotide-binding pocket of the G protein α-subunit to catalyze GDP release, the key step required for GTP binding and activation of G proteins. The structure also offers hints about how G protein binding may, in turn, allosterically influence ligand binding. Here we provide functional evidence that G protein coupling to the β2AR stabilizes a 'closed' receptor conformation characterized by restricted access to and egress from the hormone-binding site. Surprisingly, the effects of G protein on the hormone-binding site can be obsd. in the absence of a bound agonist, where G protein coupling driven by basal receptor activity impedes the assocn. of agonists, partial agonists, antagonists and inverse agonists. The ability of bound ligands to dissoc. from the receptor is also hindered, providing a structural explanation for the G protein-mediated enhancement of agonist affinity, which has been obsd. for many GPCR-G protein pairs. Our data also indicate that in contrast to agonist binding alone, coupling of a G protein in the absence of an agonist stabilizes large structural changes in a GPCR. The effects of nucleotide-free G protein on ligand-binding kinetics are shared by other members of the superfamily of GPCRs, suggesting that a common mechanism may underlie G protein-mediated enhancement of agonist affinity.
- 46Wingler, L. M.; Lefkowitz, R. J. Conformational basis of G protein-coupled receptor signaling versatility. Trends Cell Biol. 2020, 30, 736– 747, DOI: 10.1016/j.tcb.2020.06.00246Conformational Basis of G Protein-Coupled Receptor Signaling VersatilityWingler, Laura M.; Lefkowitz, Robert J.Trends in Cell Biology (2020), 30 (9), 736-747CODEN: TCBIEK; ISSN:0962-8924. (Elsevier Ltd.)A review. G protein-coupled receptors (GPCRs) are privileged structural scaffolds in biol. that have the versatility to regulate diverse physiol. processes. Interestingly, many GPCR ligands exhibit significant 'bias' - the ability to preferentially activate subsets of the many cellular pathways downstream of these receptors. Recently, complementary information from structural and spectroscopic approaches has made significant inroads into understanding the mechanisms of these biased ligands. The consistently emerging theme is that GPCRs are highly dynamic proteins, and ligands with varying pharmacol. properties differentially modulate the equil. among multiple conformations. Biased signaling and other recently appreciated complexities of GPCR signaling thus appear to be a natural consequence of the conformational heterogeneity of GPCRs and GPCR-transducer complexes.
- 47Copeland, R. A. Conformational adaptation in drug–target interactions and residence time. Future Med. Chem. 2011, 3, 1491– 1501, DOI: 10.4155/fmc.11.11247Conformational adaptation in drug-target interactions and residence timeCopeland, Robert A.Future Medicinal Chemistry (2011), 3 (12), 1491-1501CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. Although drug-target interactions are commonly illustrated in terms of structurally static binding and dissocn. events, such descriptions are inadequate to explain the impact of conformational dynamics on these processes. For high-affinity interactions, both the assocn. and dissocn. of drug mols. to and from their targets are often controlled by conformational changes of the target. Conformational adaptation can greatly influence the residence time of a drug on its target (i.e., the lifetime of the binary drug-target complex); long residence time can lead to sustained pharmacol. and may also mitigate off-target toxicity. In this perspective, the kinetics of drug-target assocn. and dissocn. reactions are explored, with particular emphasis on the impact of conformational adaptation on drug-target residence time.
- 48Vauquelin, G. Simplified models for heterobivalent ligand binding: when are they applicable and which are the factors that affect their target residence time. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2013, 386, 949– 962, DOI: 10.1007/s00210-013-0881-048Simplified models for heterobivalent ligand binding: when are they applicable and which are the factors that affect their target residence timeVauquelin, GeorgesNaunyn-Schmiedeberg's Archives of Pharmacology (2013), 386 (11), 949-962CODEN: NSAPCC; ISSN:0028-1298. (Springer)Bivalent ligands often display high affinity/avidity for and long residence time at their target. Thereto responsible is the synergy that emanates from the simultaneous binding of their two pharmacophores to their resp. target sites. Thermodn. cycle models permit the most complete description of the binding process, and thereto, corresponding differential equation-based simulations link the "microscopic" rate consts. that govern the individual binding steps to the "macroscopic" bivalent ligand's binding properties. Present simulations of heterobivalent ligand binding led to an appreciably simpler description thereof. The thermodn. cycle model can be split into two pathways/lanes that the bivalent ligand can solicit to reach fully bound state. Since the first binding event prompts the still free pharmacophore to stay into "forced proximity" of its target site, such lanes can be looked into by the equations that also apply to induced fit binding mechanisms. Interestingly, the simplest equations apply when bivalency goes along with a large gain in avidity. The overall bivalent ligand assocn. and dissocn. will be swifter than via each lane apart, but it is the lane that allows the fastest bidirectional "transit" between the free and the fully bound target that is chiefly solicited. The bivalent ligand's residence time is governed not only by the stability of the fully bound complex but also by the ability of freshly dissocd. pharmacophores to successfully rebind. Hence, the presence of a slow-assocg. pharmacophore could be counterproductive. Yet, a long residence time is unfortunately also responsible for the slow attainment of binding equil.
- 49Palmberger, D.; Wilson, I. B.; Berger, I.; Grabherr, R.; Rendic, D. SweetBac: a new approach for the production of mammalianised glycoproteins in insect cells. PLoS One 2012, 7, e34226 DOI: 10.1371/journal.pone.003422649SweetBac: a new approach for the production of mammalianised glycoproteins in insect cellsPalmberger, Dieter; Wilson, Iain B. H.; Berger, Imre; Grabherr, Reingard; Rendic, DubravkoPLoS One (2012), 7 (4), e34226CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Recombinant prodn. of therapeutically active proteins has become a central focus of contemporary life science research. These proteins are often produced in mammalian cells, in order to obtain products with post-translational modifications similar to their natural counterparts. However, in cases where a fast and flexible system for recombinant prodn. of proteins is needed, the use of mammalian cells is limited. The baculoviral insect cell system has proven to be a powerful alternative for the expression of a wide range of recombinant proteins in short time frames. The major drawback of baculoviral systems lies in the inability to perform mammalian-like glycosylation required for the prodn. of therapeutic glycoproteins. In this study we integrated sequences encoding Caenorhabditis elegans N-acetylglucosaminyl-transferase II and bovine β1,4-galactosyltransferase I into the backbone of a baculovirus genome. The thereby generated SweetBac virus was subsequently used for the prodn. of the human HIV anti-gp41 antibody 3D6 by integrating heavy and light chain open reading frames into the SweetBac genome. The parallel expression of target genes and glycosyltransferases reduced the yield of secreted antibody. However, the overall expression rate, esp. in the recently established Tnao38 cell line, was comparable to that of transient expression in mammalian cells. In order to evaluate the ability of SweetBac to generate mammalian-like N-glycan structures on 3D6 antibody, we performed SDS-PAGE and tested for the presence of terminal galactose using Riccinus communis agglutinin I. The mammalianized variants of 3D6 showed highly specific binding to the lectin, indicating proper functionality. To confirm these results, PNGase A released N-glycans were analyzed by MALDI-TOF-MS and shown to contain structures with mainly one or two terminal galactose residues. Since the presence of specific N-glycans has an impact on antibodies' ability to exert different effector functions, we tested the binding to human Fc gamma receptor I present on U937 cells.
- 50Schneider, E. H.; Seifert, R. Sf9 cells: A versatile model system to investigate the pharmacological properties of G protein-coupled receptors. Pharmacol. Ther. 2010, 128, 387– 418, DOI: 10.1016/j.pharmthera.2010.07.00550Sf9 cells: A versatile model system to investigate the pharmacological properties of G protein-coupled receptorsSchneider, Erich H.; Seifert, RolandPharmacology & Therapeutics (2010), 128 (3), 387-418CODEN: PHTHDT; ISSN:0163-7258. (Elsevier)A review. The Sf9 cell/baculovirus expression system is widely used for high-level protein expression, often with the purpose of purifn. However, proteins may also be functionally expressed in the defined Sf9 cell environment. According to the literature, the pharmacol. of G-protein-coupled receptors (GPCRs) functionally reconstituted in Sf9 cells is similar to the receptor properties in mammalian cells. Sf9 cells express both recombinant GPCRs and G-proteins at much higher levels than mammalian cells. Sf9 cells can be grown in suspension culture, providing an inexpensive way of obtaining large protein amts. Co-infection with various baculoviruses allows free combination of GPCRs with different G-proteins. The absence of constitutively active receptors in Sf9 cells provides an excellent signal-to background ratio in functional assays, allowing the detection of agonist-independent receptor activity and of small ligand-induced signals including partial agonistic and inverse agonistic effects. Insect cell Gαi-like proteins mostly do not couple productively to mammalian GPCRs. Thus, unlike in mammalian cells, Sf9 cells do not require pertussis toxin treatment to obtain a Gαi-free environment. Co-expression of GPCRs with Gαi1, Gαi2, Gαi3 or Gαo in Sf9 cells allows the generation of a selectivity profile for these Gαi/o-isoforms. Addnl., GPCR-G-protein combinations can be compared with defined 1:1 stoichiometry by expressing GPCR-Gα fusion proteins. Sf9 cells can also be employed for ligand screening in medicinal chem. programs, using radioligand binding assays or functional assays, like the steady-state GTPase- or [35S]GTPγS binding assay. This review shows that Sf9 cells are a versatile model system to investigate the pharmacol. properties of GPCRs.
- 51Grätz, L.; Müller, C.; Pegoli, A.; Schindler, L.; Bernhardt, G.; Littmann, T. Insertion of Nanoluc into the extracellular loops as a complementary method to establish BRET-based binding assays for GPCRs. ACS Pharmacol. Transl. Sci. 2022, 5, 1142– 1155, DOI: 10.1021/acsptsci.2c00162There is no corresponding record for this reference.
- 52Calderón, J. C.; Plut, E.; Keller, M.; Cabrele, C.; Reiser, O.; Gervasio, F. L.; Clark, T. Extended metadynamics protocol for binding/unbinding free energies of peptide ligands to class A G-protein-coupled receptors. J. Chem. Inf. Model. 2024, 64, 205– 218, DOI: 10.1021/acs.jcim.3c01574There is no corresponding record for this reference.
- 53Hall, M. P.; Unch, J.; Binkowski, B. F.; Valley, M. P.; Butler, B. L.; Wood, M. G.; Otto, P.; Zimmerman, K.; Vidugiris, G.; Machleidt, T.; Robers, M. B.; Benink, H. A.; Eggers, C. T.; Slater, M. R.; Meisenheimer, P. L.; Klaubert, D. H.; Fan, F.; Encell, L. P.; Wood, K. V. Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone substrate. ACS Chem. Biol. 2012, 7, 1848– 1857, DOI: 10.1021/cb300247853Engineered Luciferase Reporter from a Deep Sea Shrimp Utilizing a Novel Imidazopyrazinone SubstrateHall, Mary P.; Unch, James; Binkowski, Brock F.; Valley, Michael P.; Butler, Braeden L.; Wood, Monika G.; Otto, Paul; Zimmerman, Kristopher; Vidugiris, Gediminas; Machleidt, Thomas; Robers, Matthew B.; Benink, Helene A.; Eggers, Christopher T.; Slater, Michael R.; Meisenheimer, Poncho L.; Klaubert, Dieter H.; Fan, Frank; Encell, Lance P.; Wood, Keith V.ACS Chemical Biology (2012), 7 (11), 1848-1857CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Bioluminescence methodologies have been extraordinarily useful due to their high sensitivity, broad dynamic range, and operational simplicity. These capabilities have been realized largely through incremental adaptations of native enzymes and substrates, originating from luminous organisms of diverse evolutionary lineages. We engineered both an enzyme and substrate in combination to create a novel bioluminescence system capable of more efficient light emission with superior biochem. and phys. characteristics. Using a small luciferase subunit (19 kDa) from the deep sea shrimp Oplophorus gracilirostris, we have improved luminescence expression in mammalian cells ∼2.5 million-fold by merging optimization of protein structure with development of a novel imidazopyrazinone substrate (furimazine). The new luciferase, NanoLuc, produces glow-type luminescence (signal half-life >2 h) with a specific activity ∼150-fold greater than that of either firefly (Photinus pyralis) or Renilla luciferases similarly configured for glow-type assays. In mammalian cells, NanoLuc shows no evidence of post-translational modifications or subcellular partitioning. The enzyme exhibits high phys. stability, retaining activity with incubation up to 55 °C or in culture medium for >15 h at 37 °C. As a genetic reporter, NanoLuc may be configured for high sensitivity or for response dynamics by appending a degrdn. sequence to reduce intracellular accumulation. Appending a signal sequence allows NanoLuc to be exported to the culture medium, where reporter expression can be measured without cell lysis. Fusion onto other proteins allows luminescent assays of their metab. or localization within cells. Reporter quantitation is achievable even at very low expression levels to facilitate more reliable coupling with endogenous cellular processes.
- 54Keller, M.; Kaske, M.; Holzammer, T.; Bernhardt, G.; Buschauer, A. Dimeric argininamide-type neuropeptide Y receptor antagonists: Chiral discrimination between Y1 and Y4 receptors. Biorg. Med. Chem. 2013, 21, 6303– 6322, DOI: 10.1016/j.bmc.2013.08.065There is no corresponding record for this reference.
- 55Pegoli, A.; She, X.; Wifling, D.; Hubner, H.; Bernhardt, G.; Gmeiner, P.; Keller, M. Radiolabeled dibenzodiazepinone-type antagonists give evidence of dualsteric binding at the M(2) muscarinic acetylcholine receptor. J. Med. Chem. 2017, 60, 3314– 3334, DOI: 10.1021/acs.jmedchem.6b0189255Radiolabeled Dibenzodiazepinone-Type Antagonists Give Evidence of Dualsteric Binding at the M2 Muscarinic Acetylcholine ReceptorPegoli Andrea; She Xueke; Wifling David; Bernhardt Gunther; Keller Max; Hubner Harald; Gmeiner PeterJournal of medicinal chemistry (2017), 60 (8), 3314-3334 ISSN:.The dualsteric ligand approach, aiming at ligands with improved subtype selectivity, has been increasingly applied to muscarinic receptors (MRs). In this article, we present the synthesis and characterization of a M2R subtype-preferring radiolabeled dibenzodiazepinone-type antagonist ([(3)H]UNSW-MK259, [(3)H]19) and its homodimeric analogue [(3)H]UR-AP060 ([(3)H]33). Saturation binding studies at the M2R, using the orthosteric antagonist atropine to determine unspecific binding, proved that the monomeric and the dimeric compound bind to the orthosteric binding site (apparent Kd: 0.87 and 0.31 nM, respectively). Various binding studies with [(3)H]19 and [(3)H]33 at the M2R, for instance, saturation binding experiments in the presence of the allosteric MR modulators W84 (8) or LY2119620 (9) (Schild-like analysis) suggested a competitive mechanism between the allosteric modulator and the dibenzodiazepinone derivatives, and thus a dualsteric binding mode of both 19 and 33. This was consistent with the results of M2R MD simulations (≥2 μs) performed with 19 and 33.
- 56Schindler, L.; Moosbauer, J.; Schmidt, D.; Spruss, T.; Grätz, L.; Lüdeke, S.; Hofheinz, F.; Meister, S.; Echtenacher, B.; Bernhardt, G.; Pietzsch, J.; Hellwig, D.; Keller, M. Development of a neurotensin-derived 68Ga-labeled PET ligand with high in vivo stability for imaging of NTS1 receptor-expressing tumors. Cancers 2022, 14, 4922 DOI: 10.3390/cancers14194922There is no corresponding record for this reference.
- 57Schindler, L.; Wohlfahrt, K.; von Krüchten, L. G.; Prante, O.; Keller, M.; Maschauer, S. Neurotensin analogs by fluoroglycosylation at Nω-carbamoylated arginines for PET imaging of NTS1-positive tumors. Sci. Rep. 2022, 12, 15028 DOI: 10.1038/s41598-022-19296-0There is no corresponding record for this reference.
- 58Höfelschweiger, B. K. The pyrylium dyes: A new class of biolabels. Synthesis, spectroscopy, and application as labels and in general protein assay. Doctoral thesis, University of Regensburg, Regensburg, 2005.There is no corresponding record for this reference.
- 59Moser, C.; Bernhardt, G.; Michel, J.; Schwarz, H.; Buschauer, A. Cloning and functional expression of the hNPY Y5 receptor in human endometrial cancer (HEC-1B) cells. Can. J. Physiol. Pharmacol. 2000, 78, 134– 142, DOI: 10.1139/y99-12559Cloning and functional expression of the hNPY Y5 receptor in human endometrial cancer (HEC-1B) cellsMoser, C.; Bernhardt, G.; Michel, J.; Schwarz, H.; Buschauer, A.Canadian Journal of Physiology and Pharmacology (2000), 78 (2), 134-142CODEN: CJPPA3; ISSN:0008-4212. (National Research Council of Canada)Aiming to develop a functional assay for the human NPY Y5 receptor based on adenylyl cyclase activity, HEC-1B cells, in which cAMP synthesis can be efficiently stimulated with forskolin, were selected for the transfection with the pcDNA3-Y5-FLAG and the pcDEF3-Y5 vectors. After optimization of the transfection procedure, the binding of [3H]propionyl-NPY to transiently and stably expressed Y5 receptors was detd. The affinities of NPY, NPY derivs., and rPP (pNPY ≥ p(Leu31Pro34)NPY = p(2-36)NPY ≥ p(D-Trp32)NPY > p(13-36)NPY > rPP) were in accordance with the NPY Y5 receptor subtype. For [3H]propionyl-pNPY approx. 1.7×105 and 1×106 binding sites per transiently and stably transfected cell, resp., were detd. The KD values were 2.4±0.4 and 1.7±0.2 nM, resp. Due to the high expression of the receptor protein, both stably and transiently transfected cells can be conveniently used in routine radioligand binding studies. By contrast, functional assays were only feasible with HEC-1B cells stably expressing the Y5 receptor. In these cells, 10 nM pNPY inhibited the forskolin-stimulated cAMP synthesis by 75%. This effect was partially antagonized by the Y5 antagonist N-{trans-[4-(2-naphthylmethylamino)-methyl]cyclohexylmethyl}naphthalene-2-sulfonamide. Although the genetic variability of cancer cells is in principle incompatible with a stable phenotype, both ligand binding characteristics and functionality of the Y5 receptor remained unchanged for more than 30 passages.
- 60Bartole, E.; Grätz, L.; Littmann, T.; Wifling, D.; Seibel, U.; Buschauer, A.; Bernhardt, G. UR-DEBa242: A Py-5-labeled fluorescent multipurpose probe for investigations on the histamine H3 and H4 receptors. J. Med. Chem. 2020, 63, 5297– 5311, DOI: 10.1021/acs.jmedchem.0c0016060UR-DEBa242: A Py-5-Labeled Fluorescent Multipurpose Probe for Investigations on the Histamine H3 and H4 ReceptorsBartole, Edith; Graetz, Lukas; Littmann, Timo; Wifling, David; Seibel, Ulla; Buschauer, Armin; Bernhardt, GuentherJournal of Medicinal Chemistry (2020), 63 (10), 5297-5311CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Comprehensively characterized fluorescent probes for the histamine H3 receptor (H3R) and esp. for the H4R orthologs [e.g., human (h) and mouse (m)] are highly needed as versatile complementary tools to radioligands. In view of fluorescent probes for BRET-based binding studies and for localizing the H4R in live cells, we synthesized and biol. characterized Py-5-labeled histamine derivs. The most notable compd. was UR-DEBa242 (26, 1-[4-(1H-Imidazol-4-yl)butyl]-4-{(1E,3E)-4-[4-(dimethylamino)phenyl]buta-1,3-dienyl}-2, 6-dimethylpyridinium hydrotrifluoroacetate trifluoroacetate), acting as a partial agonist at the hH3R [pEC50 (reporter gene) 8.77] and as an inverse agonist/antagonist at the h/mH4Rs [pIC50(reporter gene) 8.76/7.08; pIC50/pKb (β-arrestin2) 7.81/7.30]. In confocal microscopy, 26 proved suitable for hH4R localization and trafficking studies in live cells. BRET-based binding at the NLuc-hH3,4Rs/mH4R [pKd 8.78/7.75/7.18, comparable to binding consts. from radioligand binding/flow cytometry; fast assocn./dissocn. (~ 2 min)] revealed 26 as a useful mol. tool to det. hH3,4Rs/mH4R binding affinities of ligands binding to these receptors.
- 61Mansouri, M.; Bellon-Echeverria, I.; Rizk, A.; Ehsaei, Z.; Cosentino, C. C.; Silva, C. S.; Xie, Y.; Boyce, F. M.; Davis, M. W.; Neuhauss, S. C. F.; Taylor, V.; Ballmer-Hofer, K.; Berger, I.; Berger, P. Highly efficient baculovirus-mediated multigene delivery in primary cells. Nat. Commun. 2016, 7, 11529 DOI: 10.1038/ncomms1152961Highly efficient baculovirus-mediated multigene delivery in primary cellsMansouri, Maysam; Bellon-Echeverria, Itxaso; Rizk, Aurelien; Ehsaei, Zahra; Cianciolo Cosentino, Chiara; Silva, Catarina S.; Xie, Ye; Boyce, Frederick M.; Davis, M. Wayne; Neuhauss, Stephan C. F.; Taylor, Verdon; Ballmer-Hofer, Kurt; Berger, Imre; Berger, PhilippNature Communications (2016), 7 (), 11529CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Multigene delivery and subsequent cellular expression is emerging as a key technol. required in diverse research fields including, synthetic and structural biol., cellular reprogramming and functional pharmaceutical screening. Current viral delivery systems such as retro- and adenoviruses suffer from limited DNA cargo capacity, thus impeding unrestricted multigene expression. We developed MultiPrime, a modular, non-cytotoxic, non-integrating, baculovirus-based vector system expediting highly efficient transient multigene expression from a variety of promoters. MultiPrime viruses efficiently transduce a wide range of cell types, including non-dividing primary neurons and induced-pluripotent stem cells (iPS). We show that MultiPrime can be used for reprogramming, and for genome editing and engineering by CRISPR/Cas9. Moreover, we implemented dual-host-specific cassettes enabling multiprotein expression in insect and mammalian cells using a single reagent. Our expts. establish MultiPrime as a powerful and highly efficient tool, to deliver multiple genes for a wide range of applications in primary and established mammalian cells.
- 62Nie, Y.; Chaillet, M.; Becke, C.; Haffke, M.; Pelosse, M.; Fitzgerald, D.; Collinson, I.; Schaffitzel, C.; Berger, I. ACEMBL tool-kits for high-throughput multigene delivery and expression in prokaryotic and eukaryotic hosts. Adv. Exp. Med. Biol. 2016, 896, 27– 42, DOI: 10.1007/978-3-319-27216-0_3There is no corresponding record for this reference.
- 63Cheng, Y.-C.; Prusoff, W. H. Relationship between the inhibition constant (KI) and the concentration of inhibitor which causes 50% inhibition (IC50) of an enzymatic reaction. Biochem. Pharmacol. 1973, 22, 3099– 3108, DOI: 10.1016/0006-2952(73)90196-263Relation between the inhibition constant K1) and the concentration of inhibitor which causes fifty per cent inhibition (I50) of an enzymic reactionCheng, Yung-Chi; Prusoff, William H.Biochemical Pharmacology (1973), 22 (23), 3099-108CODEN: BCPCA6; ISSN:0006-2952.The Ki and I50 values are equal where the kinetics are non- or uncompetitive, but not where they are competitive. The relation between the 2 values was analyzed for non- and uncompetitive bi- and monosubstrate reactions and for competitive bisubstrate reactions.
- 64Laasfeld, T.; Kopanchuk, S.; Rinken, A. Image-based cell-size estimation for baculovirus quantification. BioTechniques 2017, 63, 161– 168, DOI: 10.2144/00011459564Image-based cell-size estimation for baculovirus quantificationLaasfeld, Tanis; Kopanchuk, Sergei; Rinken, AgoBioTechniques (2017), 63 (4), 161-168CODEN: BTNQDO; ISSN:0736-6205. (Informa Healthcare)Measurement of virus concn. is essential for effective virus-based transfection technologies. Here, we describe a user-friendly, image-based cell-size estn. (ICSE) assay for bacufovirus quantification that relies on automated detn. of celf diams. from bright-field microscopy images. In the !CSC assay, microplate-based imaging systems and our custom ICSE-Tools software enable measurement of cell morphol. parameters over time. Results from the ICSE assay were in agreement with virus concn. measurements obtained using the traditional plaque assay as well as the Coulter principle- based cell-size measurement assay. ICSE-Tools is designed for data organization and image anal. from microplate-based imaging systems, and is freely available at www.gper.ut.eelsoftware.html.
- 65Veiksina, S.; Tahk, M.-J.; Laasfeld, T.; Link, R.; Kopanchuk, S.; Rinken, A. Fluorescence Anisotropy-Based Assay for Characterization of Ligand Binding Dynamics to GPCRs: The Case of Cy3B-Labeled Ligands Binding to MC4 Receptors in Budded Baculoviruses. In G Protein-Coupled Receptor Screening Assays: Methods and Protocols; Martins, S. A. M.; Prazeres, D. M. F., Eds.; Springer: US, New York, NY, 2021; pp 119– 136.There is no corresponding record for this reference.
- 66Kopanchuk, S.; Vavers, E.; Veiksina, S.; Ligi, K.; Zvejniece, L.; Dambrova, M.; Rinken, A. Intracellular dynamics of the Sigma-1 receptor observed with super-resolution imaging microscopy. PLoS One 2022, 17, e0268563 DOI: 10.1371/journal.pone.0268563There is no corresponding record for this reference.
- 67Soulez, F. In A “Learn 2D, Apply 3D” Method for 3D Deconvolution Microscopy, 2014 IEEE 11th International Symposium on Biomedical Imaging (ISBI); IEEE, 2014; pp 1075– 1078.There is no corresponding record for this reference.
- 68Chenouard, N.; Bloch, I.; Olivo-Marin, J. C. Multiple hypothesis tracking for cluttered biological image sequences. IEEE Trans. Pattern Anal. Mach. Intell. 2013, 35, 2736– 3750, DOI: 10.1109/TPAMI.2013.9768Multiple hypothesis tracking for cluttered biological image sequencesChenouard Nicolas; Bloch Isabelle; Olivo-Marin Jean-ChristopheIEEE transactions on pattern analysis and machine intelligence (2013), 35 (11), 2736-3750 ISSN:.In this paper, we present a method for simultaneously tracking thousands of targets in biological image sequences, which is of major importance in modern biology. The complexity and inherent randomness of the problem lead us to propose a unified probabilistic framework for tracking biological particles in microscope images. The framework includes realistic models of particle motion and existence and of fluorescence image features. For the track extraction process per se, the very cluttered conditions motivate the adoption of a multiframe approach that enforces tracking decision robustness to poor imaging conditions and to random target movements. We tackle the large-scale nature of the problem by adapting the multiple hypothesis tracking algorithm to the proposed framework, resulting in a method with a favorable tradeoff between the model complexity and the computational cost of the tracking procedure. When compared to the state-of-the-art tracking techniques for bioimaging, the proposed algorithm is shown to be the only method providing high-quality results despite the critically poor imaging conditions and the dense target presence. We thus demonstrate the benefits of advanced Bayesian tracking techniques for the accurate computational modeling of dynamical biological processes, which is promising for further developments in this domain.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsptsci.4c00013.
Preparation of the azido-functionalized Py-5 derivative 15; chromatograms of the investigation of the chemical stability of 16–18 in PBS pH 7.4 (Figure S1); radioligand displacement curves from competition binding experiments with [3H]UR-KK200 (Figure S2); concentration–response curves of hPP, 11 and 16–19 obtained from a hY4R Ca2+ aequorin assay (Figure S3); concentration–response curves of hPP, 5, 11, and 16–19 obtained from a hY4R mini-Gsi protein recruitment assay (Figure S4); concentration–response curves of hPP, 16 and 17 obtained from a hY4R CAMYEN cAMP assay (Figure S5); study of dummy fluorescent ligands in functional assays (Figure S6); excitation and emission spectra (Figure S7); viability of CHO-hY4R-Gqi5-mtAEQ cells (Figure S8); analyses of flow cytometric saturation binding data of 16 based on nonviable and viable cell populations (Figure S9); syntax of the equation used to fit FA equilibrium binding data (GraphPad Prism 5); RP-HPLC chromatograms of compounds 11 and 16–19; and 1H NMR spectra of compound 11 (PDF)
TIRF video sequence (recorded and shown at a 30 Hz resolution, shown at double speed) showing the interactions of 16 with the basal plasma membrane of adherent SK-OV-3-Y4R cells including single-particle tracking (magnified region) (AVI)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.