In Vivo Photocontrol of Microtubule Dynamics and Integrity, Migration and Mitosis, by the Potent GFP-Imaging-Compatible Photoswitchable Reagents SBTubA4P and SBTub2MClick to copy article linkArticle link copied!
- Li GaoLi GaoDepartment of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, GermanyMore by Li Gao
- Joyce C. M. MeiringJoyce C. M. MeiringCell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht CH 3584, NetherlandsMore by Joyce C. M. Meiring
- Adam VaradyAdam VaradySt. Anna Children’s Cancer Research Institute (CCRI), Vienna 1090, AustriaMore by Adam Varady
- Iris E. RuiderIris E. RuiderPhysics Department and Center for Protein Assemblies CPA, Technical University of Munich, Garching 85747, GermanyMore by Iris E. Ruider
- Constanze HeiseConstanze HeiseDepartment of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, GermanyMore by Constanze Heise
- Maximilian WranikMaximilian WranikLaboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, SwitzerlandMore by Maximilian Wranik
- Cecilia D. VelascoCecilia D. VelascoLaboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Barcelona 08907, SpainBellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona 08907, SpainMore by Cecilia D. Velasco
- Jennifer A. TaylorJennifer A. TaylorDepartment of Biology, University of Washington, Seattle, Washington 98195, United StatesMore by Jennifer A. Taylor
- Beatrice TerniBeatrice TerniLaboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Barcelona 08907, SpainBellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona 08907, SpainMore by Beatrice Terni
- Tobias WeinertTobias WeinertLaboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, SwitzerlandMore by Tobias Weinert
- Jörg StandfussJörg StandfussLaboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, SwitzerlandMore by Jörg Standfuss
- Clemens C. CabernardClemens C. CabernardDepartment of Biology, University of Washington, Seattle, Washington 98195, United StatesMore by Clemens C. Cabernard
- Artur LlobetArtur LlobetLaboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Barcelona 08907, SpainBellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona 08907, SpainMore by Artur Llobet
- Michel O. SteinmetzMichel O. SteinmetzLaboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, SwitzerlandBiozentrum, University of Basel, Basel 4056, SwitzerlandMore by Michel O. Steinmetz
- Andreas R. BauschAndreas R. BauschPhysics Department and Center for Protein Assemblies CPA, Technical University of Munich, Garching 85747, GermanyMore by Andreas R. Bausch
- Martin DistelMartin DistelSt. Anna Children’s Cancer Research Institute (CCRI), Vienna 1090, AustriaZebrafish Platform Austria for Preclinical Drug Screening (ZANDR), Vienna 1090, AustriaMore by Martin Distel
- Julia Thorn-SesholdJulia Thorn-SesholdDepartment of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, GermanyMore by Julia Thorn-Seshold
- Anna AkhmanovaAnna AkhmanovaCell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht CH 3584, NetherlandsMore by Anna Akhmanova
- Oliver Thorn-Seshold*Oliver Thorn-Seshold*Email: [email protected]Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, GermanyMore by Oliver Thorn-Seshold
Abstract
Photoswitchable reagents are powerful tools for high-precision studies in cell biology. When these reagents are globally administered yet locally photoactivated in two-dimensional (2D) cell cultures, they can exert micron- and millisecond-scale biological control. This gives them great potential for use in biologically more relevant three-dimensional (3D) models and in vivo, particularly for studying systems with inherent spatiotemporal complexity, such as the cytoskeleton. However, due to a combination of photoswitch isomerization under typical imaging conditions, metabolic liabilities, and insufficient water solubility at effective concentrations, the in vivo potential of photoswitchable reagents addressing cytosolic protein targets remains largely unrealized. Here, we optimized the potency and solubility of metabolically stable, druglike colchicinoid microtubule inhibitors based on the styrylbenzothiazole (SBT) scaffold that are nonresponsive to typical fluorescent protein imaging wavelengths and so enable multichannel imaging studies. We applied these reagents both to 3D organoids and tissue explants and to classic model organisms (zebrafish, clawed frog) in one- and two-protein imaging experiments, in which spatiotemporally localized illuminations allowed them to photocontrol microtubule dynamics, network architecture, and microtubule-dependent processes in vivo with cellular precision and second-level resolution. These nanomolar, in vivo capable photoswitchable reagents should open up new dimensions for high-precision cytoskeleton research in cargo transport, cell motility, cell division, and development. More broadly, their design can also inspire similarly capable optical reagents for a range of cytosolic protein targets, thus bringing in vivo photopharmacology one step closer to general realization.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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.
1. Introduction
Figure 1
Figure 1. Design and synthesis. (a) The colchicinoid pharmacophore (gray shaded trimethoxyphenyl south ring and isovanillyl north ring) can be applied to various scaffolds, giving photoswitchable azobenzene-based PST and SBT-based SBTub antimitotics. Previously published SBTub2/3 lacked key interaction residues (red shaded sites). (b) Z-SBTub3 inhibits tubulin polymerization and MT-dependent processes. (c) X-ray structure of tubulin:Z-SBTub3 complex (carbons as purple spheres). The south ring is buried in β-tubulin (green); only the north ring interacts with α-tubulin at the α-T5 loop (cyan). (d) Evolved SBTub compound library used in this paper. (e) Typical synthesis of SBTubs proceeds by acetanilide sulfurization, Jacobson cyclization, and basic condensation. Phosphate prodrug SBTubA4P is further accessed by phosphoester formation and deprotection.
2. Results
2.1. Cellular Structure–Activity Optimization of SBTubs
EC50 (μM) | EC50 ratio | [I]WC | ||
---|---|---|---|---|
compound | hν | dark | dark/lit | (μM) |
SBTub2 | 1.4 | >25 | >18 | 6.0 |
SBTub3 | 1.2 | >25 | >21 | 3.2 |
SBTubA4 | 0.12 | 3.8 | 31 | 0.21 |
2 | 0.52 | >100 | >190 | 1.6 |
3 | 0.20 | 15 | 75 | 0.65 |
4 | 0.67 | 9.3 | 14 | 1.5 |
5 | 1.4 | 18 | 13 | 3.2 |
6 | 23 | 27 | na | na |
7 | 2.6 | >35 | >13 | na |
8 | 1.2 | >100 | >83 | 3.0 |
9 | 1.0 | 48 | 48 | 2.6 |
10 | 3.0 | >100 | >33 | 10 |
11 | 1.7 | >100 | >58 | 4.3 |
12 | 0.77 | 64 | 83 | 1.6 |
13 | 0.42 | 8.8 | 21 | 1.8 |
14 | 1.0 | >15 | >14 | 2.7 |
SBTub2M | 0.035 | 7.0 | 200 | 0.06 |
16 | 0.52 | 48 | 92 | 1.1 |
17 | 0.23 | 40 | 174 | 0.65 |
18 | 3.0 | >15 | >5 | 10 |
SBTub3P | 3.4 | >20 | >5 | 7.5 |
SBTubA4P | 0.052 | 0.45 | 8.7 | 0.10 |
Figure 2
Figure 2. (a) Leads SBTub2M and SBTubA4 have highly nonlinear dose–response profiles, high lit/dark ratio of bioactivity, and mid-nanomolar [I]WC values. (b, c) Photocharacterization: (b) SBTub2M is not isomerized from its all-E dark state by 488 nm illumination, but is photoswitched to majority-Z lit states by UV/violet light (78% Z at 405 nm by NMR; 1:1 phosphate-buffered saline/dimethylsulfoxide, PBS:DMSO). (c) Comparison of absorbance spectra of SBTubA4 and azobenzene PST-1 illustrates the SBT’s ideal match to 405 nm photoactivation, combining stronger 405 nm absorption, with sharper absorption cutoff above 405 nm, which makes it orthogonal to GFP (488 nm), YFP (514 nm), and RFP (561 nm) imaging.
2.2. SBTub Photoswitch Performance Studies
2.3. SBTubs Isomer-Dependently Target Tubulin in Cells
Figure 3
Figure 3. Tubulin-specific cellular mechanism. (a) SBTubs light-dependently inhibit tubulin polymerization (turbidimetric cell-free assay; absorbance mirrors the extent of polymerization; lit indicates majority-Z-SBTub (20 μM) preisomerized to PSS at 405 nm; nocodazole control at 10 μM). (b) Cell cycle analysis of Jurkat cells treated with SBTub2M/SBTubA4P matches photoswitchable reference PST-1: with significant G2/M arrest under 405 nm pulsing (lit), but without cell cycle effects in the dark (matching cosolvent controls). (c) Immunofluorescence imaging of cells treated with SBTubA4P under pulsed 405 nm illuminations (lit, mostly-Z) and in the dark (all-E), compared to cosolvent control (HeLa cells, 20 h incubation; α-tubulin in green, DNA (stained with 4′,6-diamidino-2-phenylindole, DAPI) in blue). (d) Close-up views at the colchicine-binding site of X-ray co-crystal structures (PDB 7Z01, 7Z02) of Z-SBTubA4 (green) and E-SBTub2M (orange) bound to the Darpin D1:tubulin complex (dark gray α-tubulin, light gray β-tubulin in cartoon representation; Z-SBTubA4 and Z-SBTub2M in stick representation, oxygens red, nitrogen blue, and sulfur yellow). (e) Superimposition of tubulin:CA4 (white carbons; PDB 5LYJ) and TD1:Z-SBTubA4 (PDB 7Z01) shows that lead SBTubs share the same binding site as the parent natural product CA4 (see also Figure S11).
2.4. SBTub Photocontrol Enables Cell-Precise, Temporally Reversible MT Inhibition in 2D Cell Culture
Figure 4
Figure 4. Spatiotemporal control over MT dynamics in 2D-cultured HeLa cells. (a, b) MT inhibition in SBTubA4P-treated cells is initiated only upon 405 nm illumination pulses and only in ROI-targeted cells (data related to Movie S1; live-cell EB3-tdTomato comets quantify polymerizing MTs). (a) Comet count statistics are similar to cosolvent-only baseline in both ROI-pulsed-cosolvent and non-ROI-SBTubA4P conditions; ROI-SBTubA4P statistics show inhibition spikes. (b) Stills from Movie S1 at the times indicated in (a), initially during the untreated timecourse, then during the SBTubA4P-treated timecourse on the same cells. Purple arrowhead indicates the ROI cell; purple dotted circle indicates where the 405 nm ROI is applied at times 26, 88, and 148 s; and white arrowhead indicates the non-ROI cell quantified as the internal control (scale bar 15 μm). (c) EB3 comet counts of cells imaged at 561 nm only (dark, gray), with 47 frames at 487 nm applied to full field of view during the time span indicated with dashed lines (“487”, cyan) and SBTubA4P (6 μM), or with single-frame 405 nm pulses, SBTubA4P (0.6 μM), applied to full field of view at times indicated with dashed lines (“405”, violet) (n = 3 cells). Temporally precise onset and full-field diffusional reversibility are shown (data related to Movies S2 and S3). [(a, c) Mean ± standard error of the mean (SEM) EB3 comet counts as normalized to the means of the first five time points; 405 nm ROIs applied at indicated times; for further details, see the Supporting Information].
2.5. From Cell Culture to MT Photocontrol in 3D Models, Tissue Explants, and Animals
2.6. SBTub Photocontrol in 3D Organoids Enables Spatially Targeted Blockade of Migration and Mitosis in the Long Term
Figure 5
Figure 5. Spatiotemporal control over MT architecture, migration, and mitosis in 3D culture and tissue explant. (a) 3D human mammary gland organoids embedded in collagen gels only have inhibited branch outgrowth when treated with both SBTubA4P and UV pulses. (b) Local applications of UV light to ROItarg regions of SBTubA4P-treated organoids (blue box, one ca. 450 ms pulse per 7 min per z-stack) stops branch proliferation and outgrowth (red outline), while branches in untargeted ROIctrl regions develop dramatically (start: solid green line, final: dotted green line) (related to Movie S4). (c) Radial progress of branch tip fronts (directed and collective behavior) in ROItarg and ROIctrl regions. (d) Still image timecourse, zoomed on a branch tip in the ROIctrl (blue box) region, showing cell proliferation (yellow arrowhead) and matrix invasion (one representative of the migrating cells is tracked over time with green arrows), while branch tip of ROItarg region has static non-proliferating cells and even slight branch retraction (red arrows) (data related to Movie S5). (e) Branch progression and proliferation are unimpeded and continuous in ROIctrl regions, while ROItarg regions are static, and branches growing into the ROItarg stop their growth (color code as in (e), data related to Movie S6). [(a–e) Cell location in organoids tracked with nuclear stain SiR-DNA imaged at 647 nm]. (f) Whole-field-of-view 405 nm photoactivation of SBTub2M-treated intact 3D brain explants of larval Drosophila melanogaster (bottom row) causes neuroblast centrosomes (red arrows) to rapidly shrink in size and signal intensity (45 s and 3 min) and prevents the cell from progressing through division (13 min). Some MT signal accumulates at mid-cell at later time points (purple arrow) (data related to Movie S8). In DMSO-only controls (top row), centrosome integrity (white arrows, 45 s and 3 min) and progression through the cell cycle (13 min) are unaffected, indicated by myosin accumulation at the cleavage furrow (cyan arrows) (data related to Movie S9). [MTs in white (Jupiter::mCherry imaged at 561 nm), myosin in green (Squash::GFP imaged at 488 nm)]. (g) Relative mCherry fluorescence intensity of centrosomal ROIs in SBTub2M-treated prophase neuroblasts (red) after activation at 405 nm drops notably during the approximately 45 s activation period (blue box) compared to the DMSO control prophase neuroblasts (black). Signal intensities are shown as the proportion of the per-cell maximum preactivation signal intensity (shading indicates ±1 standard deviation, 1–2 centrosomes quantified from a total of five neuroblasts from three different animals). For details, see the Supporting Information.
2.7. SBTub Photocontrol in Intact 3D Tissue Explants Allows Temporally Precise MT Depolymerization and Mitotic Control in the Short Term
2.8. SBTub Photocontrol in Live Animals Enables Targeted Blockade of Embryonic Development
Figure 6
Figure 6. Photoinhibition of X. tropicalis development, and in vivo photocontrol of MT dynamics in Danio rerio. (a, b) Xenopus embryos incubated with compounds for 1 h at the two-cell stage, before medium exchange optionally with 410 nm photoactivation. Embryos show irreversible development inhibition by in situ formed Z-SBTubA4 in lit conditions but had no effects in the dark or with a low concentration of SBTubA4P [(a) SBTubA4P at 5 μM; (b) development quantified by the ratio of major to minor embryo axis lengths, six embryos per condition, mean ± SEM]. (c) Development of D. rerio treated at the indicated stages for 24 h with SBTubA4P or control compounds under dark or pulsed lit (1 s/5 min) conditions. SBTubA4P (1 or 25 μM) causes morphological abnormalities only in the lit state, showing that it remains effective in vivo. (d, e) Reversible modulation of MT dynamics in 48 hpf zebrafish embryo (25 μM). (EB3-GFP in green, histone H2B in red). (data related to Movies S12–S16; see the Supporting Information).
2.9. SBTub Photocontrol in Live Animals Enables Cell-Precise Temporally Reversible Inhibition of MT Dynamics
3. Conclusions
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/jacs.2c01020.
Temporally precise and cell-precise inhibitions of cellular MT polymerization dynamics by photoactivations of E-SBTubA4P at 405 nm (Movie S1) (MP4)
Temporally precise full-field-of-view inhibitions of cellular MT polymerization dynamics by photoactivations of E-SBTubA4P at 405 nm (Movie S2) (MP4)
No inhibition of cellular MT polymerization dynamics by illumination of E-SBTubA4P at 487 nm (Movie S3) (MP4)
SBTubA4P blocks branch development in primary human mammary gland organoids, light-dependently and with spatiotemporal precision (Movies S4−S6) (MP4, MP4, MP4)
No-compound control shows no photoinhibition of organoid branch development in both ROItarg and ROIctrl (Movie S7) (MP4)
Temporally precise depolymerization of the mitotic spindle in a prophase Drosophila neuroblast by photoactivation of E-SBTub2M (30 μM) at 405 nm (Movie S8) (MP4)
DMSO-only control to Movie S8 shows prophase Drosophila neuroblast undergoing normal mitosis after 405 nm illumination (Movie S9) (MP4)
Temporally precise depolymerization of the mitotic spindle in a prophase Drosophila neuroblast by photoactivation of E-SBTub2M (30 μM) at 405 nm (Movie S10) (MP4)
DMSO-only control to Movie S10 shows prophase Drosophila neuroblast undergoing normal mitosis after 405 nm illumination (Movie S11) (MP4)
Reaction of hatchling frog X. tropicalis embryos to mechanical stimulus depends on the temporally precise application of Z-SBTubA4 during prior development (Movie S12) (MP4)
Photoactivation of SBTubA4P (25 μM) in a live zebrafish embryo shows temporally reversible inhibitions of EB3 dynamics over several cycles (Movies S13 and S14) (MP4, MP4)
Inhibition of MT polymerization dynamics in zebrafish embryo when SBTubA4P (25 μM) is photoactivated (Movie S15) (MP4)
Photoactivation of SBTubA4P (25 μM) stops both EB3 dynamics and cell division in the developing zebrafish embryo (Movie S16) (MP4)
Chemical synthesis, photocharacterization, biological data, protein crystallization, and NMR spectra (PDF)
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
We are grateful to Henrietta Lacks, now deceased, and to her surviving family members for their contributions to biomedical research. We thank Monique Preusse for early cell viability testing, and Rebekkah Hammar for performing the tubulin polymerization assay. We thank Christian Gabka from the Nymphenburg Clinic for Plastic and Aesthetic Surgery, Munich 80637, Germany, for providing primary human mammary gland tissue.
References
This article references 59 other publications.
- 1Stepanova, T.; Slemmer, J.; Hoogenraad, C. C.; Lansbergen, G.; Dortland, B.; De Zeeuw, C. I.; Grosveld, F.; van Cappellen, G.; Akhmanova, A.; Galjart, N. Visualization of Microtubule Growth in Cultured Neurons via the Use of EB3-GFP (End-Binding Protein 3-Green Fluorescent Protein). J. Neurosci. 2003, 23, 2655– 2664, DOI: 10.1523/JNEUROSCI.23-07-02655.2003Google Scholar1Visualization of microtubule growth in cultured neurons via the use of EB3-GFP (end-binding protein 3-green fluorescent protein)Stepanova, Tatiana; Slemmer, Jenny; Hoogenraad, Casper C.; Lansbergen, Gideon; Dortland, Bjorn; De Zeeuw, Chris I.; Grosveld, Frank; van Cappellen, Gert; Akhmanova, Anna; Galjart, NielsJournal of Neuroscience (2003), 23 (7), 2655-2664CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Several microtubule binding proteins, including CLIP-170 (cytoplasmic linker protein-170), CLIP-115, and EB1 (end-binding protein 1), have been shown to assoc. specifically with the ends of growing microtubules in non-neuronal cells, thereby regulating microtubule dynamics and the binding of microtubules to protein complexes, organelles, and membranes. When fused to GFP (green fluorescent protein), these proteins, which collectively are called +TIPs (plus end tracking proteins), also serve as powerful markers for visualizing microtubule growth events. Here we demonstrate that endogenous +TIPs are present at distal ends of microtubules in fixed neurons. Using EB3-GFP as a marker of microtubule growth in live cells, we subsequently analyze microtubule dynamics in neurons. Our results indicate that microtubules grow slower in neurons than in glia and COS-1 cells. The av. speed and length of EB3-GFP movements are comparable in cell bodies, dendrites, axons, and growth cones. In the proximal region of differentiated dendrites ∼65% of EB3-GFP movements are directed toward the distal end, whereas 35% are directed toward the cell body. In more distal dendritic regions and in axons most EB3-GFP dots move toward the growth cone. This difference in directionality of EB3-GFP movements in dendrites and axons reflects the highly specific microtubule organization in neurons. Together, these results suggest that local microtubule polymn. contributes to the formation of the microtubule network in all neuronal compartments. We propose that similar mechanisms underlie the specific assocn. of CLIPs and EB1-related proteins with the ends of growing microtubules in non-neuronal and neuronal cells.
- 2Kleele, T.; Marinković, P.; Williams, P. R.; Stern, S.; Weigand, E. E.; Engerer, P.; Naumann, R.; Hartmann, J.; Karl, R. M.; Bradke, F.; Bishop, D.; Herms, J.; Konnerth, A.; Kerschensteiner, M.; Godinho, L.; Misgeld, T. An Assay to Image Neuronal Microtubule Dynamics in Mice. Nat. Commun. 2014, 5, 4827 DOI: 10.1038/ncomms5827Google Scholar2An assay to image neuronal microtubule dynamics in miceKleele, Tatjana; Marinkovic, Petar; Williams, Philip R.; Stern, Sina; Weigand, Emily E.; Engerer, Peter; Naumann, Ronald; Hartmann, Jana; Karl, Rosa M.; Bradke, Frank; Bishop, Derron; Herms, Jochen; Konnerth, Arthur; Kerschensteiner, Martin; Godinho, Leanne; Misgeld, ThomasNature Communications (2014), 5 (), 4827CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Microtubule dynamics in neurons play crit. roles in physiol., injury and disease and det. microtubule orientation, the cell biol. correlate of neurite polarization. Several microtubule binding proteins, including end-binding protein 3 (EB3), specifically bind to the growing plus tip of microtubules. In the past, fluorescently tagged end-binding proteins have revealed microtubule dynamics in vitro and in non-mammalian model organisms. Here, we devise an imaging assay based on transgenic mice expressing yellow fluorescent protein-tagged EB3 to study microtubules in intact mammalian neurites. Our approach allows measurement of microtubule dynamics in vivo and ex vivo in peripheral nervous system and central nervous system neurites under physiol. conditions and after exposure to microtubule-modifying drugs. We find an increase in dynamic microtubules after injury and in neurodegenerative disease states, before axons show morphol. indications of degeneration or regrowth. Thus increased microtubule dynamics might serve as a general indicator of neurite remodelling in health and disease.
- 3Goglia, A. G.; Toettcher, J. E. A Bright Future: Optogenetics to Dissect the Spatiotemporal Control of Cell Behavior. Curr. Opin. Chem. Biol. 2019, 48, 106– 113, DOI: 10.1016/j.cbpa.2018.11.010Google Scholar3A bright future: optogenetics to dissect the spatiotemporal control of cell behaviorGoglia, Alexander G.; Toettcher, Jared E.Current Opinion in Chemical Biology (2019), 48 (), 106-113CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)Cells sense, process, and respond to extracellular information using signaling networks: collections of proteins that act as precise biochem. sensors. These protein networks are characterized by both complex temporal organization, such as pulses of signaling activity, and by complex spatial organization, where proteins assemble structures at particular locations and times within the cell. Yet despite their ubiquity, studying these spatial and temporal properties has remained challenging because they emerge from the entire protein network rather than a single node, and cannot be easily tuned by drugs or mutations. These challenges are being met by a new generation of optogenetic tools capable of directly controlling the activity of individual signaling nodes over time and the assembly of protein complexes in space. Here, we outline how these recent innovations are being used in conjunction with engineering-influenced exptl. design to address longstanding questions in signaling biol.
- 4Hoorens, M. W. H.; Szymanski, W. Reversible, Spatial and Temporal Control over Protein Activity Using Light. Trends Biochem. Sci. 2018, 43, 567– 575, DOI: 10.1016/j.tibs.2018.05.004Google Scholar4Reversible, Spatial and Temporal Control over Protein Activity Using LightHoorens, Mark W. H.; Szymanski, WiktorTrends in Biochemical Sciences (2018), 43 (8), 567-575CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. In biomedical sciences, the function of a protein of interest is investigated by altering its net activity and assessing the consequences for the cell or organism. To change the activity of a protein, a wide variety of chem. and genetic tools have been developed. The drawback of most of these tools is that they do not allow for reversible, spatial and temporal control. Here, we describe selected developments in photopharmacol. that aim at establishing such control over protein activity through bioactive mols. with photo-controlled potency. We also discuss why such control is desired and what challenges still need to be overcome for photopharmacol. to reach its maturity as a chem. biol. research tool.
- 5Borowiak, M.; Nahaboo, W.; Reynders, M.; Nekolla, K.; Jalinot, P.; Hasserodt, J.; Rehberg, M.; Delattre, M.; Zahler, S.; Vollmar, A.; Trauner, D.; Thorn-Seshold, O. Photoswitchable Inhibitors of Microtubule Dynamics Optically Control Mitosis and Cell Death. Cell 2015, 162, 403– 411, DOI: 10.1016/j.cell.2015.06.049Google Scholar5Photoswitchable Inhibitors of Microtubule Dynamics Optically Control Mitosis and Cell DeathBorowiak, Malgorzata; Nahaboo, Wallis; Reynders, Martin; Nekolla, Katharina; Jalinot, Pierre; Hasserodt, Jens; Rehberg, Markus; Delattre, Marie; Zahler, Stefan; Vollmar, Angelika; Trauner, Dirk; Thorn-Seshold, OliverCell (Cambridge, MA, United States) (2015), 162 (2), 403-411CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Small mols. that interfere with microtubule dynamics, such as Taxol and the Vinca alkaloids, are widely used in cell biol. research and as clin. anticancer drugs. However, their activity cannot be restricted to specific target cells, which also causes severe side effects in chemotherapy. Here, the authors introduce the photostatins, inhibitors that can be switched on and off in vivo by visible light, to optically control microtubule dynamics. Photostatins modulate microtubule dynamics with a subsecond response time and control mitosis in living organisms with single-cell spatial precision. In longer-term applications in cell culture, photostatins are up to 250 times more cytotoxic when switched on with blue light than when kept in the dark. Therefore, photostatins are both valuable tools for cell biol., and are promising as a new class of precision chemotherapeutics whose toxicity may be spatiotemporally constrained using light.
- 6Borowiak, M.; Küllmer, F.; Gegenfurtner, F.; Peil, S.; Nasufovic, V.; Zahler, S.; Thorn-Seshold, O.; Trauner, D.; Arndt, H.-D. Optical Manipulation of F-Actin with Photoswitchable Small Molecules. J. Am. Chem. Soc. 2020, 142, 9240– 9249, DOI: 10.1021/jacs.9b12898Google Scholar6Optical Manipulation of F-Actin with Photoswitchable Small MoleculesBorowiak, Malgorzata; Kuellmer, Florian; Gegenfurtner, Florian; Peil, Sebastian; Nasufovic, Veselin; Zahler, Stefan; Thorn-Seshold, Oliver; Trauner, Dirk; Arndt, Hans-DieterJournal of the American Chemical Society (2020), 142 (20), 9240-9249CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Cell-permeable photoswitchable small mols., termed optojasps, are introduced to optically control the dynamics of the actin cytoskeleton and cellular functions that depend on it. These light-dependent effectors were designed from the F-actin-stabilizing marine depsipeptide jasplakinolide by functionalizing them with azobenzene photoswitches. As demonstrated, optojasps can be employed to control cell viability, cell motility, and cytoskeletal signaling with the high spatial and temporal resoln. that light affords. Optojasps can be expected to find applications in diverse areas of cell biol. research. They may also provide a template for photopharmacol. targeting the ubiquitous actin cytoskeleton with precision control in the micrometer range.
- 7Glotzer, M. The 3Ms of Central Spindle Assembly: Microtubules, Motors and MAPs. Nat. Rev. Mol. Cell Biol. 2009, 10, 9– 20, DOI: 10.1038/nrm2609Google Scholar7The 3Ms of central spindle assembly: microtubules, motors and MAPsGlotzer, MichaelNature Reviews Molecular Cell Biology (2009), 10 (1), 9-20CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. During metaphase, sister chromatids are positioned at the midpoint of the microtubule-based mitotic spindle in prepn. for their segregation. The onset of anaphase triggers inactivation of the key mitotic kinase, cyclin-dependent kinase 1 (CDK1), and the poleward movement of sister chromatids. During anaphase, the mitotic spindle reorganizes in prepn. for cytokinesis. Kinesin motor proteins and microtubule-assocd. proteins bundle the plus ends of interpolar microtubules and generate the central spindle, which regulates cleavage furrow initiation and the completion of cytokinesis. Complementary approaches, including cell biol., genetics, and computational modeling, have provided new insights into the mechanism and regulation of central spindle assembly.
- 8Kapitein, L. C.; Hoogenraad, C. C. Building the Neuronal Microtubule Cytoskeleton. Neuron 2015, 87, 492– 506, DOI: 10.1016/j.neuron.2015.05.046Google Scholar8Building the Neuronal Microtubule CytoskeletonKapitein, Lukas C.; Hoogenraad, Casper C.Neuron (2015), 87 (3), 492-506CODEN: NERNET; ISSN:0896-6273. (Cell Press)Microtubules are one of the major cytoskeletal components of neurons, essential for many fundamental cellular and developmental processes, such as neuronal migration, polarity, and differentiation. Microtubules have been regarded as crit. structures for stable neuronal morphol. because they serve as tracks for long-distance transport, provide dynamic and mech. functions, and control local signaling events. Establishment and maintenance of the neuronal microtubule architecture requires tight control over different dynamic parameters, such as microtubule no., length, distribution, orientations, and bundling. Recent genetic studies have identified mutations in a wide variety of tubulin isotypes and microtubule-related proteins in many of the major neurodevelopmental and neurodegenerative diseases. Here, we highlight the functions of the neuronal microtubule cytoskeleton, its architecture, and the way its organization and dynamics are shaped by microtubule-related proteins.
- 9van Haren, J.; Charafeddine, R. A.; Ettinger, A.; Wang, H.; Hahn, K. M.; Wittmann, T. Local Control of Intracellular Microtubule Dynamics by EB1 Photodissociation. Nat. Cell Biol. 2018, 20, 252– 261, DOI: 10.1038/s41556-017-0028-5Google Scholar9Local control of intracellular microtubule dynamics by EB1 photodissociationvan Haren, Jeffrey; Charafeddine, Rabab A.; Ettinger, Andreas; Wang, Hui; Hahn, Klaus M.; Wittmann, TorstenNature Cell Biology (2018), 20 (3), 252-261CODEN: NCBIFN; ISSN:1465-7392. (Nature Research)End-binding proteins (EBs) are adaptors that recruit functionally diverse microtubule plus-end-tracking proteins (+TIPs) to growing microtubule plus ends. To test with high spatial and temporal accuracy how, when and where +TIP complexes contribute to dynamic cell biol., it developed a photo-inactivated EB1 variant (.product. -EB1) by inserting a blue-light-sensitive protein-protein interaction module between the microtubule-binding and +TIP-binding domains of EB1. .product. -EB1 replaces endogenous EB1 function in the absence of blue light. By contrast, blue-light-mediated .product. -EB1 photodissocn. results in rapid +TIP complex disassembly, and acutely and reversibly attenuates microtubule growth independent of microtubule end assocn. of the microtubule polymerase CKAP5 (also known as ch-TOG and XMAP215). Local .product. -EB1 photodissocn. allows subcellular control of microtubule dynamics at the second and micrometer scale, and elicits aversive turning of migrating cancer cells. Importantly, light-mediated domain splitting can serve as a template to optically control other intracellular protein activities.
- 10Adikes, R. C.; Hallett, R. A.; Saway, B. F.; Kuhlman, B.; Slep, K. C. Control of Microtubule Dynamics Using an Optogenetic Microtubule plus End–F-Actin Cross-Linker. J. Cell Biol. 2018, 217, 779– 793, DOI: 10.1083/jcb.201705190Google Scholar10Control of microtubule dynamics using an optogenetic microtubule plus end-F-actin cross-linkerAdikes, Rebecca C.; Hallett, Ryan A.; Saway, Brian F.; Kuhlman, Brian; Slep, Kevin C.Journal of Cell Biology (2018), 217 (2), 779-793CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)We developed a novel optogenetic tool, SxIP-improved light-inducible dimer (iLID), to facilitate the reversible recruitment of factors to microtubule (MT) plus ends in an end-binding protein-dependent manner using blue light. We show that SxIP-iLID can track MT plus ends and recruit tgRFP-SspB upon blue light activation. We used this system to investigate the effects of crosslinking MT plus ends and F-actin in Drosophila melanogaster S2 cells to gain insight into spectraplakin function and mechanism. We show that SxIP-iLID can be used to temporally recruit an F-actin binding domain to MT plus ends and cross-link the MT and F-actin networks. Crosslinking decreases MT growth velocities and generates a peripheral MT exclusion zone. SxIP-iLID facilitates the general recruitment of specific factors to MT plus ends with temporal control enabling researchers to systematically regulate MT plus end dynamics and probe MT plus end function in many biol. processes.
- 11Meiring, J. C. M.; Grigoriev, I.; Nijenhuis, W.; Kapitein, L. C.; Akhmanova, A. Opto-Katanin: An Optogenetic Tool for Localized Microtubule Disassembly. bioRxiv 2021, 22, 473806 DOI: 10.1101/2021.12.22.473806Google ScholarThere is no corresponding record for this reference.
- 12Liu, G. Y.; Chen, S.-C.; Shaiv, K.; Hong, S.-R.; Yang, W.-T.; Huang, S.-H.; Chang, Y.-C.; Cheng, H.; Lin, Y.-C. Precise Control of Microtubule Disassembly in Living Cells. bioRxiv 2021, 31, 463668 DOI: 10.1101/2021.10.08.463668Google ScholarThere is no corresponding record for this reference.
- 13Wühr, M.; Tan, E. S.; Parker, S. K.; Detrich, H. W.; Mitchison, T. J. A Model for Cleavage Plane Determination in Early Amphibian and Fish Embryos. Curr. Biol. 2010, 20, 2040– 2045, DOI: 10.1016/j.cub.2010.10.024Google Scholar13A Model for Cleavage Plane Determination in Early Amphibian and Fish EmbryosWuehr, Martin; Tan, Edwin S.; Parker, Sandra K.; Detrich, H. William, III; Mitchison, Timothy J.Current Biology (2010), 20 (22), 2040-2045CODEN: CUBLE2; ISSN:0960-9822. (Cell Press)Summary: Current models for cleavage plane detn. propose that metaphase spindles are positioned and oriented by interactions of their astral microtubules with the cellular cortex, followed by cleavage in the plane of the metaphase plate []. We show that in early frog and fish embryos, where cells are unusually large, astral microtubules in metaphase are too short to position and orient the spindle. Rather, the preceding interphase aster centers and orients a pair of centrosomes prior to nuclear envelope breakdown, and the spindle assembles between these prepositioned centrosomes. Interphase asters center and orient centrosomes with dynein-mediated pulling forces. These forces act before astral microtubules contact the cortex; thus, dynein must pull from sites in the cytoplasm, not the cell cortex as is usually proposed for smaller cells. Aster shape is detd. by interactions of the expanding periphery with the cell cortex or with an interaction zone that forms between sister-asters in telophase. We propose a model to explain cleavage plane geometry in which the length of astral microtubules is limited by interaction with these boundaries, causing length asymmetries. Dynein anchored in the cytoplasm then generates length-dependent pulling forces, which move and orient centrosomes.
- 14Josa-Culleré, L.; Llebaria, A. In the Search for Photocages Cleavable with Visible Light: An Overview of Recent Advances and Chemical Strategies. ChemPhotoChem 2021, 5, 296– 314, DOI: 10.1002/cptc.202000253Google ScholarThere is no corresponding record for this reference.
- 15Müller-Deku, A.; Meiring, J. C. M.; Loy, K.; Kraus, Y.; Heise, C.; Bingham, R.; Jansen, K. I.; Qu, X.; Bartolini, F.; Kapitein, L. C.; Akhmanova, A.; Ahlfeld, J.; Trauner, D.; Thorn-Seshold, O. Photoswitchable Paclitaxel-Based Microtubule Stabilisers Allow Optical Control over the Microtubule Cytoskeleton. Nat. Commun. 2020, 11, 4640 DOI: 10.1038/s41467-020-18389-6Google Scholar15Photoswitchable paclitaxel-based microtubule stabilisers allow optical control over the microtubule cytoskeletonMueller-Deku, Adrian; Meiring, Joyce C. M.; Loy, Kristina; Kraus, Yvonne; Heise, Constanze; Bingham, Rebekkah; Jansen, Klara I.; Qu, Xiaoyi; Bartolini, Francesca; Kapitein, Lukas C.; Akhmanova, Anna; Ahlfeld, Julia; Trauner, Dirk; Thorn-Seshold, OliverNature Communications (2020), 11 (1), 4640CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Small mol. inhibitors are prime reagents for studies in microtubule cytoskeleton research, being applicable across a range of biol. models and not requiring genetic engineering. However, traditional chem. inhibitors cannot be exptl. applied with spatiotemporal precision suiting the length and time scales inherent to microtubule-dependent cellular processes. We have synthesized photoswitchable paclitaxel-based microtubule stabilizers, whose binding is induced by photoisomerisation to their metastable state. Photoisomerising these reagents in living cells allows optical control over microtubule network integrity and dynamics, cell division and survival, with biol. response on the timescale of seconds and spatial precision to the level of individual cells within a population. In primary neurons, they enable regulation of microtubule dynamics resolved to subcellular regions within individual neurites. These azobenzene-based microtubule stabilizers thus enable non-invasive, spatiotemporally precise modulation of the microtubule cytoskeleton in living cells, and promise new possibilities for studying intracellular transport, cell motility, and neuronal physiol.
- 16Gao, L.; Meiring, J. C. M.; Heise, C.; Rai, A.; Müller-Deku, A.; Akhmanova, A.; Thorn-Seshold, J.; Thorn-Seshold, O. Photoswitchable Epothilone-Based Microtubule Stabilisers Allow GFP-Imaging-Compatible, Optical Control over the Microtubule Cytoskeleton. Angew. Chem., Int. Ed. 2021, 48, e202114614 DOI: 10.1002/anie.202114614Google ScholarThere is no corresponding record for this reference.
- 17Rastogi, S. K.; Zhao, Z.; Gildner, M. B.; Shoulders, B. A.; Velasquez, T. L.; Blumenthal, M. O.; Wang, L.; Li, X.; Hudnall, T. W.; Betancourt, T.; Du, L.; Brittain, W. J. Synthesis, Optical Properties and in Vitro Cell Viability of Novel Spiropyrans and Their Photostationary States. Tetrahedron 2021, 80, 131854 DOI: 10.1016/j.tet.2020.131854Google Scholar17Synthesis, optical properties and in vitro cell viability of novel spiropyrans and their photostationary statesRastogi, Shiva K.; Zhao, Zhenze; Gildner, M. Brenton; Shoulders, Ben A.; Velasquez, Tara L.; Blumenthal, Madeleine O.; Wang, Lei; Li, Xiaopeng; Hudnall, Todd W.; Betancourt, Tania; Du, Liqin; Brittain, William J.Tetrahedron (2021), 80 (), 131854CODEN: TETRAB; ISSN:0040-4020. (Elsevier Ltd.)A novel class of spiropyrans I (R = H, OEt, OMe; R1 = R2 = R3 = H, OMe; R4 = H, NO2) was synthesized using a multistep process that involves three key intermediates: (a) diazonium-tetrafluoroborate, (b) hydrazine and (c) indolium iodide. The SP analogs I were confirmed as being able to isomerize and attain a photostationary state (PSS) upon irradn. with UV light (UV, 365 nm) in an aq. environment. UV-visible absorption spectra were recorded to confirm the isomerization properties. The ability of the synthesized compds. I to induce growth inhibition of HeLa cervical cancer cells was assessed via the MTT assay after incubation with either the SP or their PSS. The IC50 values of two PSS II (R = H; R1 = R2 = R3 = OMe; R4 = nitro, R = R1 = R2 = R3 = OMe; R4 = H), were obsd. to be around 14 ± 4 fold lower (26 ± 3μM) than their corresponding SPs. The most cytotoxic compds. I [R = R1 = R2 = R3 = R4 = H (III)] and II [R = R1 = R2 = R3 = R4 = H (IV)] showed the lowest IC50 values (12μM). An in vitro tubulin polymn. assay showed that III and IV exhibited the greatest difference in tubulin inhibition relative to unirradiated I (R = R1 = R2 = R3 = H, R4 = NO2; R = R1 = R2 = R3 = OMe, R4 = H).
- 18Gao, L.; Meiring, J. C. M.; Kraus, Y.; Wranik, M.; Weinert, T.; Pritzl, S. D.; Bingham, R.; Ntouliou, E.; Jansen, K. I.; Olieric, N.; Standfuss, J.; Kapitein, L. C.; Lohmüller, T.; Ahlfeld, J.; Akhmanova, A.; Steinmetz, M. O.; Thorn-Seshold, O. A Robust, GFP-Orthogonal Photoswitchable Inhibitor Scaffold Extends Optical Control over the Microtubule Cytoskeleton. Cell Chem. Biol. 2021, 28, 228– 241, DOI: 10.1016/j.chembiol.2020.11.007Google Scholar18A Robust, GFP-Orthogonal Photoswitchable Inhibitor Scaffold Extends Optical Control over the Microtubule CytoskeletonGao, Li; Meiring, Joyce C. M.; Kraus, Yvonne; Wranik, Maximilian; Weinert, Tobias; Pritzl, Stefanie D.; Bingham, Rebekkah; Ntouliou, Evangelia; Jansen, Klara I.; Olieric, Natacha; Standfuss, Joerg; Kapitein, Lukas C.; Lohmueller, Theobald; Ahlfeld, Julia; Akhmanova, Anna; Steinmetz, Michel O.; Thorn-Seshold, OliverCell Chemical Biology (2021), 28 (2), 228-241.e6CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Optically controlled chem. reagents, termed photopharmaceuticals, are powerful tools for precise spatiotemporal control of proteins particularly when genetic methods, such as knockouts or optogenetics are not viable options. However, current photopharmaceutical scaffolds, such as azobenzenes are intolerant of GFP/YFP imaging and are metabolically labile, posing severe limitations for biol. use. We rationally designed a photoswitchable SBT scaffold to overcome these problems, then derivatized it to create exceptionally metabolically robust and fully GFP/YFP-orthogonal SBTub photopharmaceutical tubulin inhibitors. Lead compd. SBTub3 allows temporally reversible, cell-precise, and even subcellularly precise photomodulation of microtubule dynamics, organization, and microtubule-dependent processes. By overcoming the previous limitations of microtubule photopharmaceuticals, SBTubs offer powerful applications in cell biol., and their robustness and druglikeness are favorable for intracellular biol. control in in vivo applications. We furthermore expect that the robustness and imaging orthogonality of the SBT scaffold will inspire other derivatizations directed at extending the photocontrol of a range of other biol. targets.
- 19Sailer, A.; Meiring, J. C. M.; Heise, C.; Pettersson, L. N.; Akhmanova, A.; Thorn-Seshold, J.; Thorn-Seshold, O. Pyrrole Hemithioindigo Antimitotics with Near-Quantitative Bidirectional Photoswitching That Photocontrol Cellular Microtubule Dynamics with Single-Cell Precision. Angew. Chem., Int. Ed. 2021, 60, 23695– 23704, DOI: 10.1002/anie.202104794Google Scholar19Pyrrole Hemithioindigo Antimitotics with Near-Quantitative Bidirectional Photoswitching that Photocontrol Cellular Microtubule Dynamics with Single-Cell PrecisionSailer, Alexander; Meiring, Joyce C. M.; Heise, Constanze; Pettersson, Linda N.; Akhmanova, Anna; Thorn-Seshold, Julia; Thorn-Seshold, OliverAngewandte Chemie, International Edition (2021), 60 (44), 23695-23704CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We report the first cellular application of the emerging near-quant. photoswitch pyrrole hemithioindigo, by rationally designing photopharmaceutical PHTub inhibitors of the cytoskeletal protein tubulin. PHTubs allow simultaneous visible-light imaging and photoswitching in live cells, delivering cell-precise photomodulation of microtubule dynamics, and photocontrol over cell cycle progression and cell death. This is the first acute use of a hemithioindigo photopharmaceutical for high-spatiotemporal-resoln. biol. control in live cells. It addnl. demonstrates the utility of near-quant. photoswitches, by enabling a dark-active design to overcome residual background activity during cellular photopatterning. This work opens up new horizons for high-precision microtubule research using PHTubs and shows the cellular applicability of pyrrole hemithioindigo as a valuable scaffold for photocontrol of a range of other biol. targets.
- 20Sailer, A.; Ermer, F.; Kraus, Y.; Lutter, F. H.; Donau, C.; Bremerich, M.; Ahlfeld, J.; Thorn-Seshold, O. Hemithioindigos for Cellular Photopharmacology: Desymmetrised Molecular Switch Scaffolds Enabling Design Control over the Isomer-Dependency of Potent Antimitotic Bioactivity. ChemBioChem 2019, 20, 1305– 1314, DOI: 10.1002/cbic.201800752Google Scholar20Hemithioindigos for Cellular Photopharmacology: Desymmetrised Molecular Switch Scaffolds Enabling Design Control over the Isomer-Dependency of Potent Antimitotic BioactivitySailer, Alexander; Ermer, Franziska; Kraus, Yvonne; Lutter, Ferdinand H.; Donau, Carsten; Bremerich, Maximilian; Ahlfeld, Julia; Thorn-Seshold, OliverChemBioChem (2019), 20 (10), 1305-1314CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Druglike small mols. with photoswitchable bioactivity-photopharmaceuticals-allow biologists to perform studies with exquisitely precise and reversible, spatial and temporal control over crit. biol. systems inaccessible to genetic manipulation. The photoresponsive pharmacophores disclosed have been almost exclusively azobenzenes, which has limited the structural and substituent scope of photopharmacol. More detrimentally, for azobenzene reagents, it is not researchers' needs for adapted exptl. tools, but rather protein binding site sterics, that typically force whether the trans (dark) or cis (lit) isomer is the more bioactive. We now present the rational design of HOTubs, the first hemithioindigo-based pharmacophores enabling photoswitchable control over endogenous biol. activity in cellulo. HOTubs optically control microtubule depolymn. and cell death in unmodified mammalian cells. Notably, we show how the asymmetry of hemithioindigos allows a priori design of either Z- or E- (dark- or lit)-toxic antimitotics, whereas the corresponding azobenzenes are exclusively lit-toxic. We thus demonstrate that hemithioindigos enable an important expansion of the substituent and design scope of photopharmacol. interventions for biol. systems.
- 21Sailer, A.; Ermer, F.; Kraus, Y.; Bingham, R.; Lutter, F. H.; Ahlfeld, J.; Thorn-Seshold, O. Potent Hemithioindigo-Based Antimitotics Photocontrol the Microtubule Cytoskeleton in Cellulo. Beilstein J. Org. Chem. 2020, 16, 125– 134, DOI: 10.3762/bjoc.16.14Google Scholar21Potent hemithioindigo-based antimitotics photocontrol the microtubule cytoskeleton in celluloSailer, Alexander; Ermer, Franziska; Kraus, Yvonne; Bingham, Rebekkah; Lutter, Ferdinand H.; Ahlfeld, Julia; Thorn-Seshold, OliverBeilstein Journal of Organic Chemistry (2020), 16 (), 125-134CODEN: BJOCBH; ISSN:1860-5397. (Beilstein-Institut zur Foerderung der Chemischen Wissenschaften)Uniquely, in contrast to other photoswitches that have been applied to biol., the pseudosym. hemithioindigo scaffold has allowed the creation of both dark-active and lit-active photopharmaceuticals for the same binding site by a priori design. However, the potency of previous hemithioindigo photopharmaceuticals has not been optimal for their translation to other biol. models. Inspired by the structure of tubulin-inhibiting indanones, we created hemithioindigo-based indanone-like tubulin inhibitors (HITubs) and optimized their cellular potency as antimitotic photopharmaceuticals. The use of the hemithioindigo scaffold also permitted us to employ a parahydroxyhemistilbene motif, a structural feature which is denied to most azobenzenes due to the negligibly short lifetimes of their metastable Z-isomers, which proved crucial to enhancing the potency and photoswitchability. The HITubs were ten times more potent than previously reported hemithioindigo photopharmaceutical antimitotics in a series of cell-free and cellular assays, and allowed robust photocontrol over tubulin polymn., microtubule (MT) network structure, cell cycle, and cell survival. Addnl., as the hemithioindigo scaffold allows photoswitchable bioactivity for substituent patterns inaccessible to the majority of current photopharmaceuticals, wider adoption of the hemithioindigo scaffold may significantly expand the scope of cellular and in vivo targets addressable by photopharmacol.
- 22Engdahl, A. J.; Torres, E. A.; Lock, S. E.; Engdahl, T. B.; Mertz, P. S.; Streu, C. N. Synthesis, Characterization, and Bioactivity of the Photoisomerizable Tubulin Polymerization Inhibitor Azo-Combretastatin A4. Org. Lett. 2015, 17, 4546– 4549, DOI: 10.1021/acs.orglett.5b02262Google Scholar22Synthesis, Characterization, and Bioactivity of the Photoisomerizable Tubulin Polymerization Inhibitor azo-Combretastatin A4Engdahl, Ashton J.; Torres, Edith A.; Lock, Sarah E.; Engdahl, Taylor B.; Mertz, Pamela S.; Streu, Craig N.Organic Letters (2015), 17 (18), 4546-4549CODEN: ORLEF7; ISSN:1523-7052. (American Chemical Society)Combretastatin A4 is a stilbenoid tubulin binding mitotic inhibitor whose conformation greatly influences its potency, making it an excellent candidate for adaptation as a photoactivatable tool. Herein, the authors report a novel synthesis, the facile isomerization with com. grade equipment, and biol. activity of azo-combretastatin A4 in vitro and in human cancer cells. Photoisomerized azo-combretastatin A4 is at least 200-fold more potent in cellular culture, making it a promising phototherapeutic and biomedical research tool.
- 23Zenker, J.; White, M. D.; Gasnier, M.; Alvarez, Y. D.; Lim, H. Y. G.; Bissiere, S.; Biro, M.; Plachta, N. Expanding Actin Rings Zipper the Mouse Embryo for Blastocyst Formation. Cell 2018, 173, 776– 791, DOI: 10.1016/j.cell.2018.02.035Google Scholar23Expanding Actin Rings Zipper the Mouse Embryo for Blastocyst FormationZenker, Jennifer; White, Melanie D.; Gasnier, Maxime; Alvarez, Yanina D.; Lim, Hui Yi Grace; Bissiere, Stephanie; Biro, Mate; Plachta, NicolasCell (Cambridge, MA, United States) (2018), 173 (3), 776-791.e17CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Transformation from morula to blastocyst is a defining event of preimplantation embryo development. During this transition, the embryo must establish a paracellular permeability barrier to enable expansion of the blastocyst cavity. Here, using live imaging of mouse embryos, we reveal an actin-zippering mechanism driving this embryo sealing. Preceding blastocyst stage, a cortical F-actin ring assembles at the apical pole of the embryo's outer cells. The ring structure forms when cortical actin flows encounter a network of polar microtubules that exclude F-actin. Unlike stereotypical actin rings, the actin rings of the mouse embryo are not contractile, but instead, they expand to the cell-cell junctions. Here, they couple to the junctions by recruiting and stabilizing adherens and tight junction components. Coupling of the actin rings triggers localized myosin II accumulation, and it initiates a tension-dependent zippering mechanism along the junctions that is required to seal the embryo for blastocyst formation.
- 24Theisen, U.; Ernst, A. U.; Heyne, R. L. S.; Ring, T. P.; Thorn-Seshold, O.; Köster, R. W. Microtubules and Motor Proteins Support Zebrafish Neuronal Migration by Directing Cargo. J. Cell Biol. 2020, 219, e201908040 DOI: 10.1083/jcb.201908040Google Scholar24Microtubules and motor proteins support zebrafish neuronal migration by directing cargoTheisen, Ulrike; Ernst, Alexander U.; Heyne, Ronja L. S.; Ring, Tobias P.; Thorn-Seshold, Oliver; Koester, Reinhard W.Journal of Cell Biology (2020), 219 (10), e201908040CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Neuronal migration during development is necessary to form an ordered and functional brain. Postmitotic neurons require microtubules and dynein to move, but the mechanisms by which they contribute to migration are not fully characterized. Using tegmental hindbrain nuclei neurons in zebrafish embryos together with subcellular imaging, optogenetics, and photopharmacol., we show that, in vivo, the centrosome's position relative to the nucleus is not linked to greatest motility in this cell type. Nevertheless, microtubules, dynein, and kinesin-1 are essential for migration, and we find that interference with endosome formation or the Golgi app. impairs migration to a similar extent as disrupting microtubules. In addn., an imbalance in the traffic of the model cargo Cadherin-2 also reduces neuronal migration. These results lead us to propose that microtubules act as cargo carriers to control spatiotemporal protein distribution, which in turn controls motility. This adds crucial insights into the variety of ways that microtubules can support successful neuronal migration in vivo.
- 25Gavin, J.; Ruiz, J. F. M.; Kedziora, K.; Windle, H.; Kelleher, D. P.; Gilmer, J. F. Structure Requirements for Anaerobe Processing of Azo Compounds: Implications for Prodrug Design. Bioorg. Med. Chem. Lett. 2012, 22, 7647– 7652, DOI: 10.1016/j.bmcl.2012.10.014Google Scholar25Structure requirements for anaerobe processing of azo compounds: Implications for prodrug designGavin, Jason; Ruiz, Juan F. Marquez; Kedziora, Kinga; Windle, Henry; Kelleher, Dermot P.; Gilmer, John F.Bioorganic & Medicinal Chemistry Letters (2012), 22 (24), 7647-7652CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)This Letter generalizes the metab. of the azo class of compds. by Clostridium perfringens, an anaerobe found in the human colon. A recently reported 5-aminosalicylic acid-based prednisolone prodrug was shown to release the drug when incubated with the bacteria, while the para-aminobenzoic acid (PABA) based analog did not. Instead, it showed a new HPLC peak with a relatively close retention time to the parent which was identified by LCMS as the partially reduced hydrazine product. This Letter investigates azoredn. across a panel of substrates with varying degrees of electronic and steric similarity to the PABA-based compd. Azo compds. with an electron donating group on the azo-contg. arom. ring showed immediate disproportionation to their parent amines without any detection of hydrazine intermediates by HPLC. Compds. contg. only electron withdrawing groups are partially and reversibly reduced to produce a stable detectable hydrazine. They do not disproportionate to their parent amines, but regenerate the parent azo compd. This incomplete redn. is relevant to the design of azo-based prodrugs and the toxicol. of azo-based dyes.
- 26Sheldon, J. E.; Dcona, M. M.; Lyons, C. E.; Hackett, J. C.; Hartman, M. C. T. Photoswitchable Anticancer Activity via Trans-Cis Isomerization of a Combretastatin A-4 Analog. Org. Biomol. Chem. 2016, 14, 40– 49, DOI: 10.1039/c5ob02005kGoogle Scholar26Photoswitchable anticancer activity via trans-cis isomerization of a combretastatin A-4 analogSheldon, Jonathon E.; Dcona, M. Michael; Lyons, Charles E.; Hackett, John C.; Hartman, Matthew C. T.Organic & Biomolecular Chemistry (2016), 14 (1), 40-49CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)Combretastatin A-4 (CA4) is highly potent anticancer drug that acts as an inhibitor of tubulin polymn. The core of the CA4 structure contains a cis-stilbene, and it is known that the trans isomer is significantly less potent. The authors prepd. an azobenzene analog of CA4 (Azo-CA4) that shows 13-35-fold enhancement in potency upon illumination. EC50 values in the light were in the mid nM range. Due to its ability to thermally revert to less toxic trans form, Azo-CA4 also has the ability to automatically turn its activity off with time. Azo-CA4 is less potent than CA-4 because it degrades in the presence of glutathione as evidenced by UV-Vis spectroscopy and ESI-MS. Nevertheless, Azo-CA4 represents a promising strategy for switchable potency for treatment of cancer.
- 27An, Y.; Chen, C.; Zhu, J.; Dwivedi, P.; Zhao, Y.; Wang, Z. Hypoxia-Induced Activity Loss of a Photo-Responsive Microtubule Inhibitor Azobenzene Combretastatin A4. Front. Chem. Sci. Eng. 2020, 14, 880– 888, DOI: 10.1007/s11705-019-1864-6Google Scholar27Hypoxia-induced activity loss of a photo-responsive microtubule inhibitor azobenzene combretastatin A4An, Yang; Chen, Chao; Zhu, Jundong; Dwivedi, Pankaj; Zhao, Yanjun; Wang, ZhengFrontiers of Chemical Science and Engineering (2020), 14 (5), 880-888CODEN: FCSEA3; ISSN:2095-0187. (Springer)The conformation-dependent activity of azobenzene combretastatin A4 (Azo-CA4) provides a unique approach to reduce the side-effects of chemotherapy, due to the light-triggered conformation transition of its azobenzene moiety. Under hypoxic tumor microenvironment, however, the high expression of azoreductase can reduce azobenzene to aniline. It was postulated that the Azo-CA4 might be degraded under hypoxia, resulting in the decrease of its anti-tumor activity. The aim of this study was to verify such hypothesis in HeLa cells in vitro. The quant. drug concn. anal. shows the ratio-metric formation of degrdn. end-products, confirming the bioredn. of Azo-CA4. The tubulin staining study indicates that Azo-CA4 loses the potency of switching off microtubule dynamics under hypoxia. Furthermore, the cell cycle anal. shows that the ability of Azo-CA4 to induce mitotic arrest is lost at low oxygen content. Therefore, the cytotoxicity of Azo-CA4 is compromised under hypoxia. In contrast, combretastatin A4 as a pos. control maintains the potency to inhibit tubulin polymn. and break down the nuclei irresp. of light irradn. and oxygen level. This work highlights the influence of hypoxic tumor microenvironment on the anti-tumor potency of Azo-CA4, which should be considered during the early stage of designing translational Azo-CA4 delivery systems.
- 28Hüll, K.; Morstein, J.; Trauner, D. In Vivo Photopharmacology. Chem. Rev. 2018, 118, 10710– 10747, DOI: 10.1021/acs.chemrev.8b00037Google Scholar28In Vivo PhotopharmacologyHull, Katharina; Morstein, Johannes; Trauner, DirkChemical Reviews (Washington, DC, United States) (2018), 118 (21), 10710-10747CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Synthetic photoswitches have been known for many years, but their usefulness in biol., pharmacol., and medicine has only recently been systematically explored. Over the past decade photopharmacol. has grown into a vibrant field. As the photophys., pharmacodynamic, and pharmacokinetic properties of photoswitches, such as azobenzenes, have become established, they have been applied to a wide range of biol. targets. These include transmembrane proteins (ion channels, transporters, G protein-coupled receptors, receptor-linked enzymes), sol. proteins (kinases, proteases, factors involved in epigenetic regulation), lipid membranes, and nucleic acids. In this review, the authors provide an overview of photopharmacol. using synthetic switches that have been applied in vivo, i.e., in living cells and organisms. The authors discuss the scope and limitations of this approach to study biol. function and the challenges it faces in translational medicine. The relationships between synthetic photoswitches, natural chromophores used in optogenetics, and caged ligands are addressed.
- 29Lachmann, D.; Lahmy, R.; König, B. Fulgimides as Light-Activated Tools in Biological Investigations: Fulgimides as Light-Activated Tools in Biological Investigations. Eur. J. Org. Chem. 2019, 2019, 5018– 5024, DOI: 10.1002/ejoc.201900219Google Scholar29Fulgimides as Light-Activated Tools in Biological InvestigationsLachmann, D.; Lahmy, R.; Koenig, B.European Journal of Organic Chemistry (2019), 2019 (31-32), 5018-5024CODEN: EJOCFK; ISSN:1099-0690. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. With high spatiotemporal control, the conformation, rigidity and electronics of photoresponsive bioactive mols. can be altered. This, in turn, allows for control over the biol. properties of these mols. Incorporation of a photoswitchable moiety into a no. of reported inhibitors, ligands and modulators has demonstrated the ability to modulate enzyme, receptor and ion channel responses using light. To date, the major classes of photoswitches explored in biol. applications have been the azobenzenes and diarylethenes. Even though the use of these photoswitches has established the value of photoresponsive mols. as biol. tools, several limitations have become apparent. Fulgimides represent a promising class of photoswitches that are not widely used for such biol. purposes. Their properties are similar to that of diarylethenes, as their photochromism is based on a 6π-electrocyclic rearrangement, however, fulgimides have the added advantage of thermal stability for both isomers. Fulgimides exhibit high photostationary states and fatigue resistance, with the ability to switch in aq. buffer solns. In this minireview, these advantageous photophys. properties will be discussed, as well as the use of fulgimides in biol. investigations.
- 30Fuchter, M. J. On the Promise of Photopharmacology Using Photoswitches: A Medicinal Chemist’s Perspective. J. Med. Chem. 2020, 63, 11436– 11447, DOI: 10.1021/acs.jmedchem.0c00629Google Scholar30On the Promise of Photopharmacology Using Photoswitches: A Medicinal Chemist's PerspectiveFuchter, Matthew J.Journal of Medicinal Chemistry (2020), 63 (20), 11436-11447CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Photopharmacol. is a growing area of endeavor that employs photoswitchable ligands to allow for light-dependent pharmacol. activity. By coupling light to therapeutic action, improved spatial and temporal selectivity can be achieved and subsequently harnessed for new concepts in therapy. Tremendous progress has already been made, with photopharmacol. agents now reported against a wide array of target classes and light-dependent results demonstrated in a range of live cell and animal models. Several challenges remain, however, esp. in order for photopharmacol. to truly impact the clin. management of disease. This Perspective aims to summarize these challenges, particularly with attention to the medicinal chem. that will be unavoidably required for the further translation of these agents/approaches. By clearly defining challenges for drug hunters, it is hoped that further research into the medicinal chem. of photopharmacol. agents will be stimulated, ultimately enabling full realization of the huge potential for this exciting field.
- 31Welleman, I. M.; Hoorens, M. W. H.; Feringa, B. L.; Boersma, H. H.; Szymański, W. Photoresponsive Molecular Tools for Emerging Applications of Light in Medicine. Chem. Sci. 2020, 11, 11672– 11691, DOI: 10.1039/D0SC04187DGoogle Scholar31Photoresponsive molecular tools for emerging applications of light in medicineWelleman, Ilse M.; Hoorens, Mark W. H.; Feringa, Ben L.; Boersma, Hendrikus H.; Szymanski, WiktorChemical Science (2020), 11 (43), 11672-11691CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A review. Light-based therapeutic and imaging modalities, which emerge in clin. applications, rely on mol. tools, such as photocleavable protecting groups and photoswitches that respond to photonic stimulus and translate it into a biol. effect. However, optimization of their key parameters (activation wavelength, band sepn., fatigue resistance and half-life) is necessary to enable application in the medical field. In this perspective, we describe the applications scenarios that can be envisioned in clin. practice and then we use those scenarios to explain the necessary properties that the photoresponsive tools used to control biol. function should possess, highlighted by examples from medical imaging, drug delivery and photopharmacol. We then present how the (photo)chem. parameters are currently being optimized and an outlook is given on pharmacol. aspects (toxicity, soly., and stability) of light-responsive mols. With these interdisciplinary insights, we aim to inspire the future directions for the development of photocontrolled tools that will empower clin. applications of light.
- 32Gaspari, R.; Prota, A. E.; Bargsten, K.; Cavalli, A.; Steinmetz, M. O. Structural Basis of Cis- and Trans-Combretastatin Binding to Tubulin. Chem 2017, 2, 102– 113, DOI: 10.1016/j.chempr.2016.12.005Google Scholar32Structural Basis of cis- and trans-Combretastatin Binding to TubulinGaspari, Roberto; Prota, Andrea E.; Bargsten, Katja; Cavalli, Andrea; Steinmetz, Michel O.Chem (2017), 2 (1), 102-113CODEN: CHEMVE; ISSN:2451-9294. (Cell Press)Combretastatin A4 (CA-4) derivs. are microtubule-destabilizing agents, some of which are in advanced clin. trials for cancer therapy. The active cis conformation of CA-4 can readily isomerize into a thermodynamically more stable but significantly less active trans form. Here, we solved the high-resoln. crystal structure of cis-CA-4 in complex with tubulin. The compd. binds to the colchicine site of tubulin and displays both common and distinct interaction points with colchicine. Using metadynamics simulations, we generated the trans form of the ligand within its binding site and computed the relative binding free energy of the cis-CA-4 and trans-CA-4 isomers via a thermodn. cycle. The calcns. suggest structural distortions of the bound trans-CA-4 mol. as the likely cause of its reduced activity in comparison with that of its cis isomer. Our findings could open up unique possibilities for structure-guided drug engineering with the aim of discovering combretastatin variants with improved chem. properties and pharmacol. profiles.
- 33Tarade, D.; Ma, D.; Pignanelli, C.; Mansour, F.; Simard, D.; Berg, S.; van den Gauld, J.; McNulty, J.; Pandey, S. Structurally Simplified Biphenyl Combretastatin A4 Derivatives Retain in Vitro Anti-Cancer Activity Dependent on Mitotic Arrest. PLoS One 2017, 12, e0171806 DOI: 10.1371/journal.pone.0171806Google ScholarThere is no corresponding record for this reference.
- 34Jacobson, P. Ueber Bildung von Anhydroverbindungen Des Orthoamidophenylmercaptans Aus Thioaniliden. Ber. Dtsch. Chem. Ges. 1886, 19, 1067– 1077, DOI: 10.1002/cber.188601901239Google ScholarThere is no corresponding record for this reference.
- 35Ma, D.; Xie, S.; Xue, P.; Zhang, X.; Dong, J.; Jiang, Y. Efficient and Economical Access to Substituted Benzothiazoles: Copper-Catalyzed Coupling of 2-Haloanilides with Metal Sulfides and Subsequent Condensation. Angew. Chem., Int. Ed. 2009, 48, 4222– 4225, DOI: 10.1002/anie.200900486Google Scholar35Efficient and economical access to substituted benzothiazoles: copper-catalyzed coupling of 2-haloanilides with metal sulfides and subsequent condensationMa, Dawei; Xie, Siwei; Xue, Peng; Zhang, Xiaojing; Dong, Jinhua; Jiang, YongwenAngewandte Chemie, International Edition (2009), 48 (23), 4222-4225, S4222/1-S4222/18CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The first metal-catalyzed direct coupling of metal sulfides with aryl halides and subsequent intramol. condensation provided substituted benzothiazoles. A wide range of functional groups are tolerated under the reaction conditions.
- 36Tron, G. C.; Pirali, T.; Sorba, G.; Pagliai, F.; Busacca, S.; Genazzani, A. A. Medicinal Chemistry of Combretastatin A4: Present and Future Directions. J. Med. Chem. 2006, 49, 3033– 3044, DOI: 10.1021/jm0512903Google Scholar36Medicinal chemistry of combretastatin A4: Present and future directionsTron, Gian Cesare; Pirali, Tracey; Sorba, Giovanni; Pagliai, Francesca; Busacca, Sara; Genazzani, Armando A.Journal of Medicinal Chemistry (2006), 49 (11), 3033-3044CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review with refs. A review on the medicinal chem. of combretastatin A4 (CA-4), a compd. with antitumor properties. The mechanism of action of CA-4, including its ability to inhibit tubulin polymn., is discussed, along with selected analogs of CA-4 modified on the arom. rings A and B, substituted Ph rings, heterocycling rings, non-substituted arom. rings, modifications on the double bond, and analogs where the olefinic group is replaced by a ring. Some issues related to the pharmacol. of CA-4 and future directions in research are considered.
- 37Kraus, Y.; Glas, C.; Melzer, B.; Gao, L.; Heise, C.; Preuße, M.; Ahlfeld, J.; Bracher, F.; Thorn-Seshold, O. Isoquinoline-Based Biaryls as a Robust Scaffold for Microtubule Inhibitors. Eur. J. Med. Chem. 2020, 186, 111865 DOI: 10.1016/j.ejmech.2019.111865Google Scholar37Isoquinoline-based biaryls as a robust scaffold for microtubule inhibitorsKraus, Yvonne; Glas, Carina; Melzer, Benedikt; Gao, Li; Heise, Constanze; Preusse, Monique; Ahlfeld, Julia; Bracher, Franz; Thorn-Seshold, OliverEuropean Journal of Medicinal Chemistry (2020), 186 (), 111865CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)We here report the discovery of isoquinoline-based biaryls as a new scaffold for colchicine domain tubulin inhibitors. Colchicinoid inhibitors offer highly desirable cytotoxic and vascular disrupting bioactivities, but their further development requires improving in vivo robustness and tolerability: properties that both depend on the scaffold structure employed. We have developed isoquinoline-based biaryls as a novel scaffold for high-potency tubulin inhibitors, with excellent robustness, druglikeness, and facile late-stage structural diversification, accessible through a tolerant synthetic route. We confirmed their bioactivity mechanism in vitro, developed sol. prodrugs, and established safe in vivo dosing in mice. By addressing several problems facing the current families of inhibitors, we expect that this new scaffold will find a range of in vivo applications towards translational use in cancer therapy.
- 38Thorn-Seshold, O.; Meiring, J. C. M. Photocontrolling Microtubule Dynamics with Photoswitchable Chemical Reagents. In Microtubules─Methods and Protocols; Springer International Publishing, 2022; Chapter 26.Google ScholarThere is no corresponding record for this reference.
- 39Florian, S.; Mitchison, T. J. Anti-Microtubule Drugs. In The Mitotic Spindle: Methods and Protocols; Chang, P.; Ohi, R., Eds.; Springer: New York, NY, 2016; Vol. 1413, pp 403– 421.Google ScholarThere is no corresponding record for this reference.
- 40Roostalu, J.; Thomas, C.; Cade, N. I.; Kunzelmann, S.; Taylor, I. A.; Surrey, T. The Speed of GTP Hydrolysis Determines GTP Cap Size and Controls Microtubule Stability. eLife 2020, 9, e51992 DOI: 10.7554/eLife.51992Google Scholar40The speed of GTP hydrolysis determines GTP cap size and controls microtubule stabilityRoostalu, Johanna; Thomas, Claire; Cade, Nicholas Ian; Kunzelmann, Simone; Taylor, Ian A.; Surrey, ThomaseLife (2020), 9 (), e51992CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Microtubules are cytoskeletal polymers whose function depends on their property to switch between states of growth and shrinkage. Growing microtubules are thought to be stabilized by a GTP cap at their ends. The nature of this cap, however, is still poorly understood. End Binding proteins (EBs) recruit a diverse range of regulators of microtubule function to growing microtubule ends. Whether the EB binding region is identical to the GTP cap is unclear. Using mutated human tubulin with blocked GTP hydrolysis, we demonstrate that EBs bind with high affinity to the GTP conformation of microtubules. Slowing-down GTP hydrolysis leads to extended GTP caps. We find that cap length dets. microtubule stability and that the microtubule conformation changes gradually in the cap as GTP is hydrolyzed. These results demonstrate the crit. importance of the kinetics of GTP hydrolysis for microtubule stability and establish that the GTP cap coincides with the EB-binding region.
- 41Linnemann, J. R.; Miura, H.; Meixner, L. K.; Irmler, M.; Kloos, U. J.; Hirschi, B.; Bartsch, H. S.; Sass, S.; Beckers, J.; Theis, F. J.; Gabka, C.; Sotlar, K.; Scheel, C. H. Quantification of Regenerative Potential in Primary Human Mammary Epithelial Cells. Development 2015, 31, 3239– 3251, DOI: 10.1242/dev.123554Google ScholarThere is no corresponding record for this reference.
- 42Buchmann, B.; Engelbrecht, L. K.; Fernandez, P.; Hutterer, F. P.; Raich, M. K.; Scheel, C. H.; Bausch, A. R. Mechanical Plasticity of Collagen Directs Branch Elongation in Human Mammary Gland Organoids. Nat. Commun. 2021, 12, 2759 DOI: 10.1038/s41467-021-22988-2Google Scholar42Mechanical plasticity of collagen directs branch elongation in human mammary gland organoidsBuchmann, B.; Engelbrecht, L. K.; Fernandez, P.; Hutterer, F. P.; Raich, M. K.; Scheel, C. H.; Bausch, A. R.Nature Communications (2021), 12 (1), 2759CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Epithelial branch elongation is a central developmental process during branching morphogenesis in diverse organs. This fundamental growth process into large arborized epithelial networks is accompanied by structural reorganization of the surrounding extracellular matrix (ECM), well beyond its mech. linear response regime. Here, we report that epithelial ductal elongation within human mammary organoid branches relies on the non-linear and plastic mech. response of the surrounding collagen. Specifically, we demonstrate that collective back-and-forth motion of cells within the branches generates tension that is strong enough to induce a plastic reorganization of the surrounding collagen network which results in the formation of mech. stable collagen cages. Such matrix encasing in turn directs further tension generation, branch outgrowth and plastic deformation of the matrix. The identified mech. tension equil. sets a framework to understand how mech. cues can direct ductal branch elongation.
- 43Hofer, M.; Lutolf, M. P. Engineering Organoids. Nat. Rev. Mater. 2021, 6, 402– 420, DOI: 10.1038/s41578-021-00279-yGoogle Scholar43Engineering organoidsHofer, Moritz; Lutolf, Matthias P.Nature Reviews Materials (2021), 6 (5), 402-420CODEN: NRMADL; ISSN:2058-8437. (Nature Portfolio)A review. Abstr.: Organoids are in vitro miniaturized and simplified model systems of organs that have gained enormous interest for modeling tissue development and disease, and for personalized medicine, drug screening and cell therapy. Despite considerable success in culturing physiol. relevant organoids, challenges remain to achieve real-life applications. In particular, the high variability of self-organizing growth and restricted exptl. and anal. access hamper the translatability of organoid systems. In this Review, we argue that many limitations of traditional organoid culture can be addressed by engineering approaches at all levels of organoid systems. We investigate cell surface and genetic engineering approaches, and discuss stem cell niche engineering based on the design of matrixes that allow spatiotemporal control of organoid growth and shape-guided morphogenesis. We examine how microfluidic approaches and lessons learnt from organs-on-a-chip enable the integration of mechano-physiol. parameters and increase accessibility of organoids to improve functional readouts. Applying engineering principles to organoids increases reproducibility and provides exptl. control, which will, ultimately, be required to enable clin. translation.
- 44Kopf, A.; Renkawitz, J.; Hauschild, R.; Girkontaite, I.; Tedford, K.; Merrin, J.; Thorn-Seshold, O.; Trauner, D.; Häcker, H.; Fischer, K.-D.; Kiermaier, E.; Sixt, M. Microtubules Control Cellular Shape and Coherence in Amoeboid Migrating Cells. J. Cell Biol. 2020, 219, e201907154 DOI: 10.1083/jcb.201907154Google Scholar44Microtubules control cellular shape and coherence in amoeboid migrating cellsKopf, Aglaja; Renkawitz, Joerg; Hauschild, Robert; Girkontaite, Irute; Tedford, Kerry; Merrin, Jack; Thorn-Seshold, Oliver; Trauner, Dirk; Haecker, Hans; Fischer, Klaus-Dieter; Kiermaier, Eva; Sixt, MichaelJournal of Cell Biology (2020), 219 (6), e201907154/1-e201907154/24CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Cells navigating through complex tissues face a fundamental challenge: while multiple protrusions explore different paths, the cell needs to avoid entanglement. How a cell surveys and then corrects its own shape is poorly understood. Here, we demonstrate that spatially distinct microtubule dynamics regulate amoeboid cell migration by locally promoting the retraction of protrusions. In migrating dendritic cells, local microtubule depolymn. within protrusions remote from the microtubule organizing center triggers actomyosin contractility controlled by RhoA and its exchange factor Lfc. Depletion of Lfc leads to aberrant myosin localization, thereby causing two effects that rate-limit locomotion: (1) impaired cell edge coordination during path finding and (2) defective adhesion resoln. Compromised shape control is particularly hindering in geometrically complex microenvironments, where it leads to entanglement and ultimately fragmentation of the cell body. We thus demonstrate that microtubules can act as a proprioceptive device: they sense cell shape and control actomyosin retraction to sustain cellular coherence.
- 45Vandestadt, C.; Vanwalleghem, G. C.; Castillo, H. A.; Li, M.; Schulze, K.; Khabooshan, M.; Don, E.; Anko, M.-L.; Scott, E. K.; Kaslin, J. Early Migration of Precursor Neurons Initiates Cellular and Functional Regeneration after Spinal Cord Injury in Zebrafish. bioRxiv 2019, 539940 DOI: 10.1101/539940Google ScholarThere is no corresponding record for this reference.
- 46Cabernard, C.; Doe, C. Q. Apical/Basal Spindle Orientation Is Required for Neuroblast Homeostasis and Neuronal Differentiation in Drosophila. Dev. Cell 2009, 17, 134– 141, DOI: 10.1016/j.devcel.2009.06.009Google Scholar46Apical/basal spindle orientation is required for neuroblast homeostasis and neuronal differentiation in DrosophilaCabernard, Clemens; Doe, Chris Q.Developmental Cell (2009), 17 (1), 134-141CODEN: DCEEBE; ISSN:1534-5807. (Cell Press)Precise regulation of stem cell self-renewal/differentiation is essential for embryogenesis and tumor suppression. Drosophila neural progenitors (neuroblasts) align their spindle along an apical/basal polarity axis to generate a self-renewed apical neuroblast and a differentiating basal cell. Here, the authors genetically disrupt spindle orientation without altering cell polarity to test the role of spindle orientation in self-renewal/differentiation. The authors perform correlative live imaging of polarity markers and spindle orientation over multiple divisions within intact brains, followed by mol. marker anal. of cell fate. The authors find that spindle alignment orthogonal to apical/basal polarity always segregates apical determinants into both siblings, which invariably assume a neuroblast identity. Basal determinants can all be localized into one sibling without inducing neuronal differentiation, but overexpression of the basal determinant Prospero can deplete neuroblasts. The authors conclude that the ratio of apical/basal determinants specifies neuroblast/GMC identity, and that apical/basal spindle orientation is required for neuroblast homeostasis and neuronal differentiation.
- 47Karpova, N.; Bobinnec, Y.; Fouix, S.; Huitorel, P.; Debec, A. Jupiter, a New Drosophila Protein Associated with Microtubules. Cell Motil. 2006, 63, 301– 312, DOI: 10.1002/cm.20124Google Scholar47Jupiter, a new Drosophila protein associated with microtubulesKarpova, Nina; Bobinnec, Yves; Fouix, Sylvaine; Huitorel, Philippe; Debec, AlainCell Motility and the Cytoskeleton (2006), 63 (5), 301-312CODEN: CMCYEO; ISSN:0886-1544. (Wiley-Liss, Inc.)We describe a novel Drosophila protein Jupiter, which shares properties with several structural microtubule-assocd. proteins (MAPs) including TAU, MAP2, MAP4. Jupiter is a sol. unfolded mol. with the high net pos. charge, rich in glycine. It possesses 2 degenerated repeats around the sequence PPGG, sepd. by a serine-rich region. Jupiter assocs. with microtubules in vitro and, fused with the green fluorescent protein (GFP), is an excellent marker to follow microtubule dynamics in vivo. In a jupiter transgenic Drosophila strain generated by the protein-trap technique, Jupiter:GFP fusion protein localizes to the microtubule network through the cell cycle at the different stages of development. We found particularly high Jupiter:GFP concns. in the young embryo, larval nervous system, precursors of eye photoreceptors and adult ovary. Moreover, from jupiter:gfp embryos we have established 2 permanent cell lines presenting strongly fluorescent microtubules during the whole cell cycle. In these cells, the distribution of the Jupiter:GFP fusion protein reproduces microtubule behavior upon treatment by the drugs colchicine and taxol. The Jupiter cell lines and fly strain should be of wide interest for biologists interested in in vivo anal. of microtubule dynamics.
- 48Royou, A.; Sullivan, W.; Karess, R. Cortical Recruitment of Nonmuscle Myosin II in Early Syncytial Drosophila Embryos: Its Role in Nuclear Axial Expansion and Its Regulation by Cdc2 Activity. J. Cell Biol. 2002, 158, 127– 137, DOI: 10.1083/jcb.200203148Google Scholar48Cortical recruitment of nonmuscle myosin II in early syncytial Drosophila embryos: its role in nuclear axial expansion and its regulation by Cdc2 activityRoyou, Anne; Sullivan, William; Karess, RogerJournal of Cell Biology (2002), 158 (1), 127-137CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)The nuclei of early syncytial Drosophila embryos migrate dramatically toward the poles. The cellular mechanisms driving this process, called axial expansion, are unclear, but myosin II activity is required. By following regulatory myosin light chain (RLC)-green fluorescent protein dynamics in living embryos, we obsd. cycles of myosin recruitment to the cortex synchronized with mitotic cycles. Cortical myosin is first seen in a patch at the anterocentral part of the embryo at cycle 4. With each succeeding cycle, the patch expands poleward, dispersing at the beginning of each mitosis and reassembling at the end of telophase. Each cycle of actin and myosin recruitment is accompanied by a cortical contraction. The cortical myosin cycle does not require microtubules but correlates inversely with Cdc2/cyclin B (mitosis-promoting factor) activity. A mutant RLC lacking inhibitory phosphorylation sites was fully functional with no effect on the cortical myosin cycle, indicating that Cdc2 must be modulating myosin activity by some other mechanism. An inhibitor of Rho kinase blocks the cortical myosin recruitment cycles and provokes a concomitant failure of axial expansion. These studies suggest a model in which cycles of myosin-mediated contraction and relaxation, tightly linked to Cdc2 and Rho kinase activity, are directly responsible for the axial expansion of the syncytial nuclei.
- 49Rusan, N. M.; Peifer, M. A Role for a Novel Centrosome Cycle in Asymmetric Cell Division. J. Cell Biol. 2007, 177, 13– 20, DOI: 10.1083/jcb.200612140Google Scholar49A role for a novel centrosome cycle in asymmetric cell divisionRusan, Nasser M.; Peifer, MarkJournal of Cell Biology (2007), 177 (1), 13-20CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)Drosophila melanogaster central brain neuroblasts are excellent models for stem cell asym. division. Earlier work showed that their mitotic spindle orientation is established before spindle formation. We investigated the mechanism by which this occurs, revealing a novel centrosome cycle. In interphase, the 2 centrioles sep., but only 1 is active, retaining pericentriolar material and forming a dominant centrosome. This centrosome acts as a microtubule organizing center (MTOC) and remains stationary, forming 1 pole of the future spindle. The 2nd centriole is inactive and moves to the opposite side of the cell before being activated as a centrosome/MTOC. This is accompanied by asym. localization of Polo kinase, a key centrosome regulator. Disruption of centrosomes disrupts the high fidelity of asym. division. We propose a 2-step mechanism to ensure faithful spindle positioning: the novel centrosome cycle produces a single interphase MTOC, coarsely aligning the spindle, and spindle-cortex interactions refine this alignment.
- 50Cabernard, C.; Prehoda, K. E.; Doe, C. Q. A Spindle-Independent Cleavage Furrow Positioning Pathway. Nature 2010, 467, 91– 94, DOI: 10.1038/nature09334Google Scholar50A spindle-independent cleavage furrow positioning pathwayCabernard, Clemens; Prehoda, Kenneth E.; Doe, Chris Q.Nature (London, United Kingdom) (2010), 467 (7311), 91-94CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The mitotic spindle dets. the cleavage furrow site during metazoan cell division, but whether other mechanisms exist remains unknown. Here we identify a spindle-independent mechanism for cleavage furrow positioning in Drosophila neuroblasts. We show that early and late furrow proteins (Pavarotti, Anillin, and Myosin) are localized to the neuroblast basal cortex at anaphase onset by a Pins cortical polarity pathway, and can induce a basally displaced furrow even in the complete absence of a mitotic spindle. Rotation or displacement of the spindle results in 2 furrows: an early polarity-induced basal furrow and a later spindle-induced furrow. This spindle-independent cleavage furrow mechanism may be relevant to other highly polarized mitotic cells, such as mammalian neural progenitors.
- 51Connell, M.; Cabernard, C.; Ricketson, D.; Doe, C. Q.; Prehoda, K. E. Asymmetric Cortical Extension Shifts Cleavage Furrow Position in Drosophila Neuroblasts. Mol. Biol. Cell 2011, 22, 4220– 4226, DOI: 10.1091/mbc.e11-02-0173Google Scholar51Asymmetric cortical extension shifts cleavage furrow position in Drosophila neuroblastsConnell, Marisa; Cabernard, Clemens; Ricketson, Derek; Doe, Chris Q.; Prehoda, Kenneth E.Molecular Biology of the Cell (2011), 22 (22), 4220-4226CODEN: MBCEEV; ISSN:1939-4586. (American Society for Cell Biology)The cytokinetic cleavage furrow is typically positioned sym. relative to the cortical cell boundaries, but it can also be asym. The mechanisms that control furrow site specification have been intensively studied, but how polar cortex movements influence ultimate furrow position remains poorly understood. We measured the position of the apical and the basal cortex in asym. dividing Drosophila neuroblasts and obsd. preferential displacement of the apical cortex that becomes the larger daughter cell during anaphase, effectively shifting the cleavage furrow toward the smaller daughter cell. Asym. cortical extension is correlated with the presence of cortical myosin II, which is polarized in neuroblasts. Loss of myosin II asymmetry by perturbing heterotrimeric G-protein signaling results in sym. extension and equal-sized daughter cells. We propose a model in which contraction-driven asym. polar extension of the neuroblast cortex during anaphase contributes to asym. furrow position and daughter cell size.
- 52Roberts, A.; Borisyuk, R.; Buhl, E.; Ferrario, A.; Koutsikou, S.; Li, W.-C.; Soffe, S. R. The Decision to Move: Response Times, Neuronal Circuits and Sensory Memory in a Simple Vertebrate. Proc. R. Soc. B 2019, 286, 20190297 DOI: 10.1098/rspb.2019.0297Google ScholarThere is no corresponding record for this reference.
- 53Distel, M.; Hocking, J. C.; Volkmann, K.; Köster, R. W. The Centrosome Neither Persistently Leads Migration nor Determines the Site of Axonogenesis in Migrating Neurons in Vivo. J. Cell Biol. 2010, 191, 875– 890, DOI: 10.1083/jcb.201004154Google Scholar53The centrosome neither persistently leads migration nor determines the site of axonogenesis in migrating neurons in vivoDistel, Martin; Hocking, Jennifer C.; Volkmann, Katrin; Koester, Reinhard W.Journal of Cell Biology (2010), 191 (4), 875-890CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)The position of the centrosome ahead of the nucleus has been considered crucial for coordinating neuronal migration in most developmental situations. The proximity of the centrosome has also been correlated with the site of axonogenesis in certain differentiating neurons. Despite these pos. correlations, accumulating exptl. findings appear to negate a universal role of the centrosome in detg. where an axon forms, or in leading the migration of neurons. To further examine this controversy in an in vivo setting, we have generated cell type-specific multi-cistronic gene expression to monitor subcellular dynamics in the developing zebrafish cerebellum. We show that migration of rhombic lip-derived neurons is characterized by a centrosome that does not persistently lead the nucleus, but which is instead regularly overtaken by the nucleus. In addn., axonogenesis is initiated during the onset of neuronal migration and occurs independently of centrosome proximity. These in vivo data reveal a new temporal orchestration of organelle dynamics and provide important insights into the variation in intracellular processes during vertebrate brain differentiation.
- 54Distel, M.; Wullimann, M. F.; Köster, R. W. Optimized Gal4 Genetics for Permanent Gene Expression Mapping in Zebrafish. Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 13365– 13370, DOI: 10.1073/pnas.0903060106Google Scholar54Optimized Gal4 genetics for permanent gene expression mapping in zebrafishDistel, Martin; Wullimann, Mario F.; Koster, Reinhard W.Proceedings of the National Academy of Sciences of the United States of America (2009), 106 (32), 13365-13370, S13365/1-S13365/9CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Combinatorial genetics for conditional transgene activation allows studying gene function with temporal and tissue specific control like the Gal4-UAS system, which has enabled sophisticated genetic studies in Drosophila. Recently this system was adapted for zebrafish and promising applications have been introduced. Here, we report a systematic optimization of zebrafish Gal4-UAS genetics by establishing an optimized Gal4-activator (KalTA4). We provide quant. data for KalTA4-mediated transgene activation in dependence of UAS copy nos. to allow for studying dosage effects of transgene expression. Employing a Tol2 transposon-mediated KalTA4 enhancer trap screen biased for central nervous system expression, we present a collection of self-reporting red fluorescent KalTA4 activator strains. These strains reliably transactivate UAS-dependent transgenes and can be rendered homozygous. Furthermore, we have characterized the transactivation kinetics of tissue-specific KalTA4 activation, which led to the development of a self-maintaining effector strain "Kaloop.". This strain relates transient KalTA4 expression during embryogenesis via a KalTA4-mediated autoregulatory mechanism to live adult structures. We demonstrate its use by showing that the secondary octaval nucleus in the adult hindbrain is likely derived from egr2b-expressing cells in rhombomere 5 during stages of early embryogenesis. These data demonstrate prolonged and maintained expression by Kalooping, a technique that can be used for permanent spatiotemporal genetic fate mapping and targeted transgene expression in zebrafish.
- 55Fortin, D. L.; Banghart, M. R.; Dunn, T. W.; Borges, K.; Wagenaar, D. A.; Gaudry, Q.; Karakossian, M. H.; Otis, T. S.; Kristan, W. B.; Trauner, D.; Kramer, R. H. Photochemical Control of Endogenous Ion Channels and Cellular Excitability. Nat. Methods 2008, 5, 331– 338, DOI: 10.1038/nmeth.1187Google Scholar55Photochemical control of endogenous ion channels and cellular excitabilityFortin, Doris L.; Banghart, Matthew R.; Dunn, Timothy W.; Borges, Katharine; Wagenaar, Daniel A.; Gaudry, Quentin; Karakossian, Movses H.; Otis, Thomas S.; Kristan, William B.; Trauner, Dirk; Kramer, Richard H.Nature Methods (2008), 5 (4), 331-338CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Light-activated ion channels provide a precise and noninvasive optical means for controlling action potential firing, but the genes encoding these channels must first be delivered and expressed in target cells. Here we describe a method for bestowing light sensitivity onto endogenous ion channels that does not rely on exogenous gene expression. The method uses a synthetic photoisomerizable small mol., or photoswitchable affinity label (PAL), that specifically targets K+ channels. PALs contain a reactive electrophile, enabling covalent attachment of the photoswitch to naturally occurring nucleophiles in K+ channels. Ion flow through PAL-modified channels is turned on or off by photoisomerizing PAL with different wavelengths of light. We showed that PAL treatment confers light sensitivity onto endogenous K+ channels in isolated rat neurons and in intact neural structures from rat and leech, allowing rapid optical regulation of excitability without genetic modification.
- 56Laprell, L.; Tochitsky, I.; Kaur, K.; Manookin, M. B.; Stein, M.; Barber, D. M.; Schön, C.; Michalakis, S.; Biel, M.; Kramer, R. H.; Sumser, M. P.; Trauner, D.; Gelder, R. N. V. Photopharmacological Control of Bipolar Cells Restores Visual Function in Blind Mice. J. Clin. Invest. 2017, 127, 2598– 2611, DOI: 10.1172/JCI92156Google Scholar56Photopharmacological control of bipolar cells restores visual function in blind miceLaprell Laura; Stein Marco; Barber David M; Sumser Martin P; Trauner Dirk; Laprell Laura; Kaur Kuldeep; Manookin Michael B; Van Gelder Russell N; Tochitsky Ivan; Kramer Richard H; Schon Christian; Michalakis Stylianos; Biel Martin; Van Gelder Russell NThe Journal of clinical investigation (2017), 127 (7), 2598-2611 ISSN:.Photopharmacological control of neuronal activity using synthetic photochromic ligands, or photoswitches, is a promising approach for restoring visual function in patients suffering from degenerative retinal diseases. Azobenzene photoswitches, such as AAQ and DENAQ, have been shown to restore the responses of retinal ganglion cells to light in mouse models of retinal degeneration but do not recapitulate native retinal signal processing. Here, we describe diethylamino-azo-diethylamino (DAD), a third-generation photoswitch that is capable of restoring retinal ganglion cell light responses to blue or white light. In acute brain slices of murine layer 2/3 cortical neurons, we determined that the photoswitch quickly relaxes to its inactive form in the dark. DAD is not permanently charged, and the uncharged form enables the photoswitch to rapidly and effectively cross biological barriers and thereby access and photosensitize retinal neurons. Intravitreal injection of DAD restored retinal light responses and light-driven behavior to blind mice. Unlike DENAQ, DAD acts upstream of retinal ganglion cells, primarily conferring light sensitivity to bipolar cells. Moreover, DAD was capable of generating ON and OFF visual responses in the blind retina by utilizing intrinsic retinal circuitry, which may be advantageous for restoring visual function.
- 57Matera, C.; Gomila, A. M. J.; Camarero, N.; Libergoli, M.; Soler, C.; Gorostiza, P. Photoswitchable Antimetabolite for Targeted Photoactivated Chemotherapy. J. Am. Chem. Soc. 2018, 140, 15764– 15773, DOI: 10.1021/jacs.8b08249Google Scholar57Photoswitchable Antimetabolite for Targeted Photoactivated ChemotherapyMatera, Carlo; Gomila, Alexandre M. J.; Camarero, Nuria; Libergoli, Michela; Soler, Concepcio; Gorostiza, PauJournal of the American Chemical Society (2018), 140 (46), 15764-15773CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The efficacy and tolerability of systemically administered anticancer agents are limited by their off-target effects. Precise spatiotemporal control over their cytotoxic activity would allow improving chemotherapy treatments, and light-regulated drugs are well suited to this purpose. We have developed phototrexate, the first photoswitchable inhibitor of the human dihydrofolate reductase (DHFR), as a photochromic analog of methotrexate, a widely prescribed chemotherapeutic drug to treat cancer and psoriasis. Quantification of the light-regulated DHFR enzymic activity, cell proliferation, and in vivo effects in zebrafish show that phototrexate behaves as a potent antifolate in its photoactivated cis configuration and that it is nearly inactive in its dark-relaxed trans form. Thus, phototrexate constitutes a proof-of-concept to design light-regulated cytotoxic small mols. and a step forward to develop targeted anticancer photochemotherapies with localized efficacy and reduced adverse effects.
- 58Babii, O.; Afonin, S.; Schober, T.; Garmanchuk, L. V.; Ostapchenko, L. I.; Yurchenko, V.; Zozulya, S.; Tarasov, O.; Pishel, I.; Ulrich, A. S.; Komarov, I. V. Peptide Drugs for Photopharmacology: How Much of a Safety Advantage Can Be Gained by Photocontrol?. Future Drug Discovery 2020, 2, FDD28 DOI: 10.4155/fdd-2019-0033Google ScholarThere is no corresponding record for this reference.
- 59Afonin, S.; Babii, O.; Reuter, A.; Middel, V.; Takamiya, M.; Strähle, U.; Komarov, I. V.; Ulrich, A. S. Light-Controllable Dithienylethene-Modified Cyclic Peptides: Photoswitching the in Vivo Toxicity in Zebrafish Embryos. Beilstein J. Org. Chem. 2020, 16, 39– 49, DOI: 10.3762/bjoc.16.6Google Scholar59Light-controllable dithienylethene-modified cyclic peptides: photoswitching the in vivo toxicity in zebrafish embryosAfonin, Sergii; Babii, Oleg; Reuter, Aline; Middel, Volker; Takamiya, Masanari; Straehle, Uwe; Komarov, Igor V.; Ulrich, Anne S.Beilstein Journal of Organic Chemistry (2020), 16 (), 39-49CODEN: BJOCBH; ISSN:1860-5397. (Beilstein-Institut zur Foerderung der Chemischen Wissenschaften)This study evaluates the embryotoxicity of dithienylethene-modified peptides upon photoswitching, using 19 analogs based on the β-hairpin scaffold of the natural membranolytic peptide gramicidin S. We established an in vivo assay in two variations (with ex vivo and in situ photoisomerization), using larvae of the model organism Danio rerio, and detd. the toxicities of the peptides in terms of 50% LDs (LD50). This study allowed us to: (i) demonstrate the feasibility of evaluating peptide toxicity with D. rerio larvae at 3-4 days post fertilization, (ii) det. the phototherapeutic safety windows for all peptides, (iii) demonstrate photoswitching of the whole-body toxicity for the dithienylethene-modified peptides in vivo, (iv) re-analyze previous structure-toxicity relationship data, and (v) select promising candidates for potential clin. development.
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 38 publications.
- Sudeep Koppayithodi, Palash Jana, Subhajit Bandyopadhyay. Heteroarylstilbenes: Visible-Light-Tunable Photochromic Systems in Water. The Journal of Organic Chemistry 2025, 90
(13)
, 4518-4524. https://doi.org/10.1021/acs.joc.4c02844
- Atin Chatterjee, Sandip Sarkar, Sangheeta Bhattacharjee, Arpan Bhattacharyya, Surajit Barman, Uttam Pal, Raviranjan Pandey, Anitha Ethirajan, Batakrishna Jana, Benu Brata Das, Amitava Das. Microtubule-Targeting NAP Peptide-Ru(II)-polypyridyl Conjugate As a Bimodal Therapeutic Agent for Triple Negative Breast Carcinoma. Journal of the American Chemical Society 2025, 147
(1)
, 532-547. https://doi.org/10.1021/jacs.4c11820
- Anaëlle Dumazer, Xavier Gómez-Santacana, Fanny Malhaire, Chris Jopling, Damien Maurel, Guillaume Lebon, Amadeu Llebaria, Cyril Goudet. Optical Control of Adenosine A2A Receptor Using Istradefylline Photosensitivity. ACS Chemical Neuroscience 2024, 15
(3)
, 645-655. https://doi.org/10.1021/acschemneuro.3c00721
- Adrian Müller-Deku, Oliver Thorn-Seshold. Exhaustive Catalytic ortho-Alkoxylation of Azobenzenes: Flexible Access to Functionally Diverse Yellow-Light-Responsive Photoswitches. The Journal of Organic Chemistry 2022, 87
(24)
, 16526-16531. https://doi.org/10.1021/acs.joc.2c02214
- James L. Rushworth, Aditya R. Thawani, Elena Fajardo-Ruiz, Joyce C. M. Meiring, Constanze Heise, Andrew J. P. White, Anna Akhmanova, Jochen R. Brandt, Oliver Thorn-Seshold, Matthew J. Fuchter. [5]-Helistatins: Tubulin-Binding Helicenes with Antimitotic Activity. JACS Au 2022, 2
(11)
, 2561-2570. https://doi.org/10.1021/jacsau.2c00435
- Saugata Sahu, Ammathnadu S. Amrutha, Nobuyuki Tamaoki. Controlling Protein Functionalities With Temporal and Cellular/Subcellular Dimensions of Spatial Resolution With Molecular Photoswitches. Medicinal Research Reviews 2025, 52 https://doi.org/10.1002/med.22106
- Adam Varady, Sarah Grissenberger, Andrea Wenninger-Weinzierl, Hugo Poplimont, Caterina Sturtzel, Nicole Schmitner, Li Gao, Robin A. Kimmel, Martin Distel. Precise photopharmacological eradication of metastatic tumor cells. Disease Models & Mechanisms 2025, 18
(2)
https://doi.org/10.1242/dmm.052016
- Anh-Khoa Nguyen, Sompop Khotwong, Sumrit Wacharasindhu, Preecha Phuwapraisirisan. Three new α-glucosidase inhibitors from aqueous extract of
Cannabis sativa
leaves: isolation, characterisation, and kinetic study. Natural Product Research 2025, 5 , 1-10. https://doi.org/10.1080/14786419.2024.2448841
- Martin Reynders, Sabine Willems, Julian A. Marschner, Thomas Wein, Daniel Merk, Oliver Thorn‐Seshold. Eine hochwertige photoschaltbare Sonde, die selektiv und wirksam den Transkriptionsfaktor RORγ moduliert. Angewandte Chemie 2024, 136
(49)
https://doi.org/10.1002/ange.202410139
- Martin Reynders, Sabine Willems, Julian A. Marschner, Thomas Wein, Daniel Merk, Oliver Thorn‐Seshold. A High‐Quality Photoswitchable Probe that Selectively and Potently Regulates the Transcription Factor RORγ. Angewandte Chemie International Edition 2024, 63
(49)
https://doi.org/10.1002/anie.202410139
- Carina Schmitt, Philipp Mauker, Nynke A. Vepřek, Carolin Gierse, Joyce C. M. Meiring, Jürgen Kuch, Anna Akhmanova, Leif Dehmelt, Oliver Thorn‐Seshold. A Photocaged Microtubule‐Stabilising Epothilone Allows Spatiotemporal Control of Cytoskeletal Dynamics. Angewandte Chemie 2024, 136
(43)
https://doi.org/10.1002/ange.202410169
- Carina Schmitt, Philipp Mauker, Nynke A. Vepřek, Carolin Gierse, Joyce C. M. Meiring, Jürgen Kuch, Anna Akhmanova, Leif Dehmelt, Oliver Thorn‐Seshold. A Photocaged Microtubule‐Stabilising Epothilone Allows Spatiotemporal Control of Cytoskeletal Dynamics. Angewandte Chemie International Edition 2024, 219 https://doi.org/10.1002/anie.202410169
- Gulia Bikbaeva, Anna Pilip, Anastasiya Egorova, Vasiliy Medvedev, Daria Mamonova, Dmitrii Pankin, Alexey Kalinichev, Natalya Mayachkina, Lyudmila Bakina, Ilya Kolesnikov, Gerd Leuchs, Alina Manshina. Smart photopharmacological agents: LaVO
4
:Eu
3+
@vinyl phosphonate combining luminescence imaging and photoswitchable butyrylcholinesterase inhibition. Nanoscale Advances 2024, 6
(17)
, 4417-4425. https://doi.org/10.1039/D4NA00389F
- Martin Reynders, Małgorzata Garścia, Adrian Müller-Deku, Maximilian Wranik, Kristina Krauskopf, Luis de la Osa de la Rosa, Konstantin Schaffer, Anna Jötten, Alexander Rode, Valentin Stierle, Yvonne Kraus, Benedikt Baumgartner, Ahmed Ali, Andrei Bubeneck, Trina Seal, Michel O. Steinmetz, Philipp Paulitschke, Oliver Thorn-Seshold. A photo-SAR study of photoswitchable azobenzene tubulin-inhibiting antimitotics identifying a general method for near-quantitative photocontrol. Chemical Science 2024, 15
(31)
, 12301-12309. https://doi.org/10.1039/D4SC03072A
- Akshay Saroha, Monica Swetha Bosco, Sneha Menon, Pratibha Kumari, Tanmoy Maity, Subinoy Rana, Sachin Kotak, Jagannath Mondal, Sarit S. Agasti. Regulation of microtubule dynamics and function in living cells
via
cucurbit[7]uril host–guest assembly. Chemical Science 2024, 15
(30)
, 11981-11994. https://doi.org/10.1039/D4SC00204K
- Charlotte N. Stindt, Stefano Crespi, Ben L. Feringa. Synthesis of Styrylbenzazole Photoswitches and Evaluation of their Photochemical Properties. Chemistry – A European Journal 2024, 30
(39)
https://doi.org/10.1002/chem.202401409
- Yen-Ling Lian, Yu-Chun Lin. The emerging tools for precisely manipulating microtubules. Current Opinion in Cell Biology 2024, 88 , 102360. https://doi.org/10.1016/j.ceb.2024.102360
- Angelika Seliwjorstow, Masanari Takamiya, Sepand Rastegar, Zbigniew Pianowski. Reversible Influence of Hemipiperazine Photochromism on the Early Development of Zebrafish Embryo. ChemBioChem 2024, 25
(8)
https://doi.org/10.1002/cbic.202400143
- Yu Zhao, Qingqing Huang, Qiushi Li, Zihan Chen, Yang Liu. Bidirectional Regulation of Intracellular Enzyme Activity Using Light‐Driven Nano‐Inhibitors. Angewandte Chemie 2024, 136
(8)
https://doi.org/10.1002/ange.202318533
- Yu Zhao, Qingqing Huang, Qiushi Li, Zihan Chen, Yang Liu. Bidirectional Regulation of Intracellular Enzyme Activity Using Light‐Driven Nano‐Inhibitors. Angewandte Chemie International Edition 2024, 63
(8)
https://doi.org/10.1002/anie.202318533
- Hiroshi Inaba, Kazunori Matsuura. Functionalization of microtubules by Tau-derived peptides: encapsulation, cell manipulation, and construction of superstructures. 2024, 27-44. https://doi.org/10.1039/BK9781839169328-00027
- Bo-Wen Wu, Wen-Jing Huang, Yun-He Liu, Qiu-Ge Liu, Jian Song, Tao Hu, Ping Chen, Sai-Yang Zhang. Design, synthesis and biological evaluation of 1,2,3-triazole benzothiazole derivatives as tubulin polymerization inhibitors with potent anti-esophageal cancer activities. European Journal of Medicinal Chemistry 2024, 265 , 116118. https://doi.org/10.1016/j.ejmech.2023.116118
- Hiroshi Inaba. Construction of functional microtubules and artificial motile systems based on peptide design. Polymer Journal 2023, 55
(12)
, 1261-1274. https://doi.org/10.1038/s41428-023-00838-w
- Maximilian Wranik, Tobias Weinert, Chavdar Slavov, Tiziana Masini, Antonia Furrer, Natacha Gaillard, Dario Gioia, Marco Ferrarotti, Daniel James, Hannah Glover, Melissa Carrillo, Demet Kekilli, Robin Stipp, Petr Skopintsev, Steffen Brünle, Tobias Mühlethaler, John Beale, Dardan Gashi, Karol Nass, Dmitry Ozerov, Philip J. M. Johnson, Claudio Cirelli, Camila Bacellar, Markus Braun, Meitian Wang, Florian Dworkowski, Chris Milne, Andrea Cavalli, Josef Wachtveitl, Michel O. Steinmetz, Jörg Standfuss. Watching the release of a photopharmacological drug from tubulin using time-resolved serial crystallography. Nature Communications 2023, 14
(1)
https://doi.org/10.1038/s41467-023-36481-5
- Maximilian Wranik, Michal W. Kepa, Emma V. Beale, Daniel James, Quentin Bertrand, Tobias Weinert, Antonia Furrer, Hannah Glover, Dardan Gashi, Melissa Carrillo, Yasushi Kondo, Robin T. Stipp, Georgii Khusainov, Karol Nass, Dmitry Ozerov, Claudio Cirelli, Philip J. M. Johnson, Florian Dworkowski, John H. Beale, Scott Stubbs, Thierry Zamofing, Marco Schneider, Kristina Krauskopf, Li Gao, Oliver Thorn-Seshold, Christoph Bostedt, Camila Bacellar, Michel O. Steinmetz, Christopher Milne, Jörg Standfuss. A multi-reservoir extruder for time-resolved serial protein crystallography and compound screening at X-ray free-electron lasers. Nature Communications 2023, 14
(1)
https://doi.org/10.1038/s41467-023-43523-5
- Kazunori Matsuura, Hiroshi Inaba. Photoresponsive peptide materials: Spatiotemporal control of self-assembly and biological functions. Biophysics Reviews 2023, 4
(4)
https://doi.org/10.1063/5.0179171
- Sergio Ramos, Alba Vicente-Blázquez, Marta López-Rubio, Laura Gallego-Yerga, Raquel Álvarez, Rafael Peláez. Frentizole, a Nontoxic Immunosuppressive Drug, and Its Analogs Display Antitumor Activity via Tubulin Inhibition. International Journal of Molecular Sciences 2023, 24
(24)
, 17474. https://doi.org/10.3390/ijms242417474
- Matthias Gehringer, Felix Pape, María Méndez, Philipp Barbie, Andrea Unzue Lopez, Julien Lefranc, Franca‐Maria Klingler, Gerhard Hessler, Thierry Langer, Eleonora Diamanti, Matthias Schiedel. Back in Person: Frontiers in Medicinal Chemistry 2023. ChemMedChem 2023, 18
(19)
https://doi.org/10.1002/cmdc.202300344
- Xingyu Wang, Benoît Gigant, Xi Zheng, Qiang Chen. Microtubule‐targeting agents for cancer treatment: Seven binding sites and three strategies. MedComm – Oncology 2023, 2
(3)
https://doi.org/10.1002/mog2.46
- Ana Patricia Ramos, Alicja Szalapak, Lucrezia Camilla Ferme, Carl D. Modes. From cells to form: A roadmap to study shape emergence in vivo. Biophysical Journal 2023, 122
(18)
, 3587-3599. https://doi.org/10.1016/j.bpj.2023.05.015
- Filip Borys, Piotr Tobiasz, Hanna Fabczak, Ewa Joachimiak, Hanna Krawczyk. First-in-Class Colchicine-Based Visible Light Photoswitchable Microtubule Dynamics Disrupting Agent. Cells 2023, 12
(14)
, 1866. https://doi.org/10.3390/cells12141866
- Hiroshi Inaba, Minamo Sakaguchi, Soei Watari, Shigesaburo Ogawa, Arif Md. Rashedul Kabir, Akira Kakugo, Kazuki Sada, Kazunori Matsuura. Reversible Photocontrol of Microtubule Stability by Spiropyran‐Conjugated Tau‐Derived Peptides. ChemBioChem 2023, 24
(8)
https://doi.org/10.1002/cbic.202200782
- Susanne Kirchner, Anna-Lena Leistner, Peter Gödtel, Angelika Seliwjorstow, Sven Weber, Johannes Karcher, Martin Nieger, Zbigniew Pianowski. Hemipiperazines as peptide-derived molecular photoswitches with low-nanomolar cytotoxicity. Nature Communications 2022, 13
(1)
https://doi.org/10.1038/s41467-022-33750-7
- Eleonora Diamanti, María Méndez, Tatjana Ross, Christian A. Kuttruff, Julien Lefranc, Franca‐Maria Klingler, Franz von Nussbaum, Manfred Jung, Matthias Gehringer. Frontiers in Medicinal Chemistry 2022 Goes Virtual. ChemMedChem 2022, 17
(21)
https://doi.org/10.1002/cmdc.202200419
- Yan Zhang, Shouguo Peng, Songwen Lin, Ming Ji, Tingting Du, Xiaoguang Chen, Heng Xu. Discovery of a novel photoswitchable PI3K inhibitor toward optically-controlled anticancer activity. Bioorganic & Medicinal Chemistry 2022, 72 , 116975. https://doi.org/10.1016/j.bmc.2022.116975
- Filip Borys, Piotr Tobiasz, Jakub Sobel, Hanna Krawczyk. Synthesis and Study of Dibenzo[b, f]oxepine Combined with Fluoroazobenzenes—New Photoswitches for Application in Biological Systems. Molecules 2022, 27
(18)
, 5836. https://doi.org/10.3390/molecules27185836
- Soei Watari, Hiroshi Inaba, Tomonori Tamura, Arif Md. Rashedul Kabir, Akira Kakugo, Kazuki Sada, Itaru Hamachi, Kazunori Matsuura. Light-induced stabilization of microtubules by photo-crosslinking of a Tau-derived peptide. Chemical Communications 2022, 58
(66)
, 9190-9193. https://doi.org/10.1039/D2CC01890J
- Susanne Kirchner, Zbigniew Pianowski. Photopharmacology of Antimitotic Agents. International Journal of Molecular Sciences 2022, 23
(10)
, 5657. https://doi.org/10.3390/ijms23105657
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. Design and synthesis. (a) The colchicinoid pharmacophore (gray shaded trimethoxyphenyl south ring and isovanillyl north ring) can be applied to various scaffolds, giving photoswitchable azobenzene-based PST and SBT-based SBTub antimitotics. Previously published SBTub2/3 lacked key interaction residues (red shaded sites). (b) Z-SBTub3 inhibits tubulin polymerization and MT-dependent processes. (c) X-ray structure of tubulin:Z-SBTub3 complex (carbons as purple spheres). The south ring is buried in β-tubulin (green); only the north ring interacts with α-tubulin at the α-T5 loop (cyan). (d) Evolved SBTub compound library used in this paper. (e) Typical synthesis of SBTubs proceeds by acetanilide sulfurization, Jacobson cyclization, and basic condensation. Phosphate prodrug SBTubA4P is further accessed by phosphoester formation and deprotection.
Figure 2
Figure 2. (a) Leads SBTub2M and SBTubA4 have highly nonlinear dose–response profiles, high lit/dark ratio of bioactivity, and mid-nanomolar [I]WC values. (b, c) Photocharacterization: (b) SBTub2M is not isomerized from its all-E dark state by 488 nm illumination, but is photoswitched to majority-Z lit states by UV/violet light (78% Z at 405 nm by NMR; 1:1 phosphate-buffered saline/dimethylsulfoxide, PBS:DMSO). (c) Comparison of absorbance spectra of SBTubA4 and azobenzene PST-1 illustrates the SBT’s ideal match to 405 nm photoactivation, combining stronger 405 nm absorption, with sharper absorption cutoff above 405 nm, which makes it orthogonal to GFP (488 nm), YFP (514 nm), and RFP (561 nm) imaging.
Figure 3
Figure 3. Tubulin-specific cellular mechanism. (a) SBTubs light-dependently inhibit tubulin polymerization (turbidimetric cell-free assay; absorbance mirrors the extent of polymerization; lit indicates majority-Z-SBTub (20 μM) preisomerized to PSS at 405 nm; nocodazole control at 10 μM). (b) Cell cycle analysis of Jurkat cells treated with SBTub2M/SBTubA4P matches photoswitchable reference PST-1: with significant G2/M arrest under 405 nm pulsing (lit), but without cell cycle effects in the dark (matching cosolvent controls). (c) Immunofluorescence imaging of cells treated with SBTubA4P under pulsed 405 nm illuminations (lit, mostly-Z) and in the dark (all-E), compared to cosolvent control (HeLa cells, 20 h incubation; α-tubulin in green, DNA (stained with 4′,6-diamidino-2-phenylindole, DAPI) in blue). (d) Close-up views at the colchicine-binding site of X-ray co-crystal structures (PDB 7Z01, 7Z02) of Z-SBTubA4 (green) and E-SBTub2M (orange) bound to the Darpin D1:tubulin complex (dark gray α-tubulin, light gray β-tubulin in cartoon representation; Z-SBTubA4 and Z-SBTub2M in stick representation, oxygens red, nitrogen blue, and sulfur yellow). (e) Superimposition of tubulin:CA4 (white carbons; PDB 5LYJ) and TD1:Z-SBTubA4 (PDB 7Z01) shows that lead SBTubs share the same binding site as the parent natural product CA4 (see also Figure S11).
Figure 4
Figure 4. Spatiotemporal control over MT dynamics in 2D-cultured HeLa cells. (a, b) MT inhibition in SBTubA4P-treated cells is initiated only upon 405 nm illumination pulses and only in ROI-targeted cells (data related to Movie S1; live-cell EB3-tdTomato comets quantify polymerizing MTs). (a) Comet count statistics are similar to cosolvent-only baseline in both ROI-pulsed-cosolvent and non-ROI-SBTubA4P conditions; ROI-SBTubA4P statistics show inhibition spikes. (b) Stills from Movie S1 at the times indicated in (a), initially during the untreated timecourse, then during the SBTubA4P-treated timecourse on the same cells. Purple arrowhead indicates the ROI cell; purple dotted circle indicates where the 405 nm ROI is applied at times 26, 88, and 148 s; and white arrowhead indicates the non-ROI cell quantified as the internal control (scale bar 15 μm). (c) EB3 comet counts of cells imaged at 561 nm only (dark, gray), with 47 frames at 487 nm applied to full field of view during the time span indicated with dashed lines (“487”, cyan) and SBTubA4P (6 μM), or with single-frame 405 nm pulses, SBTubA4P (0.6 μM), applied to full field of view at times indicated with dashed lines (“405”, violet) (n = 3 cells). Temporally precise onset and full-field diffusional reversibility are shown (data related to Movies S2 and S3). [(a, c) Mean ± standard error of the mean (SEM) EB3 comet counts as normalized to the means of the first five time points; 405 nm ROIs applied at indicated times; for further details, see the Supporting Information].
Figure 5
Figure 5. Spatiotemporal control over MT architecture, migration, and mitosis in 3D culture and tissue explant. (a) 3D human mammary gland organoids embedded in collagen gels only have inhibited branch outgrowth when treated with both SBTubA4P and UV pulses. (b) Local applications of UV light to ROItarg regions of SBTubA4P-treated organoids (blue box, one ca. 450 ms pulse per 7 min per z-stack) stops branch proliferation and outgrowth (red outline), while branches in untargeted ROIctrl regions develop dramatically (start: solid green line, final: dotted green line) (related to Movie S4). (c) Radial progress of branch tip fronts (directed and collective behavior) in ROItarg and ROIctrl regions. (d) Still image timecourse, zoomed on a branch tip in the ROIctrl (blue box) region, showing cell proliferation (yellow arrowhead) and matrix invasion (one representative of the migrating cells is tracked over time with green arrows), while branch tip of ROItarg region has static non-proliferating cells and even slight branch retraction (red arrows) (data related to Movie S5). (e) Branch progression and proliferation are unimpeded and continuous in ROIctrl regions, while ROItarg regions are static, and branches growing into the ROItarg stop their growth (color code as in (e), data related to Movie S6). [(a–e) Cell location in organoids tracked with nuclear stain SiR-DNA imaged at 647 nm]. (f) Whole-field-of-view 405 nm photoactivation of SBTub2M-treated intact 3D brain explants of larval Drosophila melanogaster (bottom row) causes neuroblast centrosomes (red arrows) to rapidly shrink in size and signal intensity (45 s and 3 min) and prevents the cell from progressing through division (13 min). Some MT signal accumulates at mid-cell at later time points (purple arrow) (data related to Movie S8). In DMSO-only controls (top row), centrosome integrity (white arrows, 45 s and 3 min) and progression through the cell cycle (13 min) are unaffected, indicated by myosin accumulation at the cleavage furrow (cyan arrows) (data related to Movie S9). [MTs in white (Jupiter::mCherry imaged at 561 nm), myosin in green (Squash::GFP imaged at 488 nm)]. (g) Relative mCherry fluorescence intensity of centrosomal ROIs in SBTub2M-treated prophase neuroblasts (red) after activation at 405 nm drops notably during the approximately 45 s activation period (blue box) compared to the DMSO control prophase neuroblasts (black). Signal intensities are shown as the proportion of the per-cell maximum preactivation signal intensity (shading indicates ±1 standard deviation, 1–2 centrosomes quantified from a total of five neuroblasts from three different animals). For details, see the Supporting Information.
Figure 6
Figure 6. Photoinhibition of X. tropicalis development, and in vivo photocontrol of MT dynamics in Danio rerio. (a, b) Xenopus embryos incubated with compounds for 1 h at the two-cell stage, before medium exchange optionally with 410 nm photoactivation. Embryos show irreversible development inhibition by in situ formed Z-SBTubA4 in lit conditions but had no effects in the dark or with a low concentration of SBTubA4P [(a) SBTubA4P at 5 μM; (b) development quantified by the ratio of major to minor embryo axis lengths, six embryos per condition, mean ± SEM]. (c) Development of D. rerio treated at the indicated stages for 24 h with SBTubA4P or control compounds under dark or pulsed lit (1 s/5 min) conditions. SBTubA4P (1 or 25 μM) causes morphological abnormalities only in the lit state, showing that it remains effective in vivo. (d, e) Reversible modulation of MT dynamics in 48 hpf zebrafish embryo (25 μM). (EB3-GFP in green, histone H2B in red). (data related to Movies S12–S16; see the Supporting Information).
References
This article references 59 other publications.
- 1Stepanova, T.; Slemmer, J.; Hoogenraad, C. C.; Lansbergen, G.; Dortland, B.; De Zeeuw, C. I.; Grosveld, F.; van Cappellen, G.; Akhmanova, A.; Galjart, N. Visualization of Microtubule Growth in Cultured Neurons via the Use of EB3-GFP (End-Binding Protein 3-Green Fluorescent Protein). J. Neurosci. 2003, 23, 2655– 2664, DOI: 10.1523/JNEUROSCI.23-07-02655.20031Visualization of microtubule growth in cultured neurons via the use of EB3-GFP (end-binding protein 3-green fluorescent protein)Stepanova, Tatiana; Slemmer, Jenny; Hoogenraad, Casper C.; Lansbergen, Gideon; Dortland, Bjorn; De Zeeuw, Chris I.; Grosveld, Frank; van Cappellen, Gert; Akhmanova, Anna; Galjart, NielsJournal of Neuroscience (2003), 23 (7), 2655-2664CODEN: JNRSDS; ISSN:0270-6474. (Society for Neuroscience)Several microtubule binding proteins, including CLIP-170 (cytoplasmic linker protein-170), CLIP-115, and EB1 (end-binding protein 1), have been shown to assoc. specifically with the ends of growing microtubules in non-neuronal cells, thereby regulating microtubule dynamics and the binding of microtubules to protein complexes, organelles, and membranes. When fused to GFP (green fluorescent protein), these proteins, which collectively are called +TIPs (plus end tracking proteins), also serve as powerful markers for visualizing microtubule growth events. Here we demonstrate that endogenous +TIPs are present at distal ends of microtubules in fixed neurons. Using EB3-GFP as a marker of microtubule growth in live cells, we subsequently analyze microtubule dynamics in neurons. Our results indicate that microtubules grow slower in neurons than in glia and COS-1 cells. The av. speed and length of EB3-GFP movements are comparable in cell bodies, dendrites, axons, and growth cones. In the proximal region of differentiated dendrites ∼65% of EB3-GFP movements are directed toward the distal end, whereas 35% are directed toward the cell body. In more distal dendritic regions and in axons most EB3-GFP dots move toward the growth cone. This difference in directionality of EB3-GFP movements in dendrites and axons reflects the highly specific microtubule organization in neurons. Together, these results suggest that local microtubule polymn. contributes to the formation of the microtubule network in all neuronal compartments. We propose that similar mechanisms underlie the specific assocn. of CLIPs and EB1-related proteins with the ends of growing microtubules in non-neuronal and neuronal cells.
- 2Kleele, T.; Marinković, P.; Williams, P. R.; Stern, S.; Weigand, E. E.; Engerer, P.; Naumann, R.; Hartmann, J.; Karl, R. M.; Bradke, F.; Bishop, D.; Herms, J.; Konnerth, A.; Kerschensteiner, M.; Godinho, L.; Misgeld, T. An Assay to Image Neuronal Microtubule Dynamics in Mice. Nat. Commun. 2014, 5, 4827 DOI: 10.1038/ncomms58272An assay to image neuronal microtubule dynamics in miceKleele, Tatjana; Marinkovic, Petar; Williams, Philip R.; Stern, Sina; Weigand, Emily E.; Engerer, Peter; Naumann, Ronald; Hartmann, Jana; Karl, Rosa M.; Bradke, Frank; Bishop, Derron; Herms, Jochen; Konnerth, Arthur; Kerschensteiner, Martin; Godinho, Leanne; Misgeld, ThomasNature Communications (2014), 5 (), 4827CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Microtubule dynamics in neurons play crit. roles in physiol., injury and disease and det. microtubule orientation, the cell biol. correlate of neurite polarization. Several microtubule binding proteins, including end-binding protein 3 (EB3), specifically bind to the growing plus tip of microtubules. In the past, fluorescently tagged end-binding proteins have revealed microtubule dynamics in vitro and in non-mammalian model organisms. Here, we devise an imaging assay based on transgenic mice expressing yellow fluorescent protein-tagged EB3 to study microtubules in intact mammalian neurites. Our approach allows measurement of microtubule dynamics in vivo and ex vivo in peripheral nervous system and central nervous system neurites under physiol. conditions and after exposure to microtubule-modifying drugs. We find an increase in dynamic microtubules after injury and in neurodegenerative disease states, before axons show morphol. indications of degeneration or regrowth. Thus increased microtubule dynamics might serve as a general indicator of neurite remodelling in health and disease.
- 3Goglia, A. G.; Toettcher, J. E. A Bright Future: Optogenetics to Dissect the Spatiotemporal Control of Cell Behavior. Curr. Opin. Chem. Biol. 2019, 48, 106– 113, DOI: 10.1016/j.cbpa.2018.11.0103A bright future: optogenetics to dissect the spatiotemporal control of cell behaviorGoglia, Alexander G.; Toettcher, Jared E.Current Opinion in Chemical Biology (2019), 48 (), 106-113CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)Cells sense, process, and respond to extracellular information using signaling networks: collections of proteins that act as precise biochem. sensors. These protein networks are characterized by both complex temporal organization, such as pulses of signaling activity, and by complex spatial organization, where proteins assemble structures at particular locations and times within the cell. Yet despite their ubiquity, studying these spatial and temporal properties has remained challenging because they emerge from the entire protein network rather than a single node, and cannot be easily tuned by drugs or mutations. These challenges are being met by a new generation of optogenetic tools capable of directly controlling the activity of individual signaling nodes over time and the assembly of protein complexes in space. Here, we outline how these recent innovations are being used in conjunction with engineering-influenced exptl. design to address longstanding questions in signaling biol.
- 4Hoorens, M. W. H.; Szymanski, W. Reversible, Spatial and Temporal Control over Protein Activity Using Light. Trends Biochem. Sci. 2018, 43, 567– 575, DOI: 10.1016/j.tibs.2018.05.0044Reversible, Spatial and Temporal Control over Protein Activity Using LightHoorens, Mark W. H.; Szymanski, WiktorTrends in Biochemical Sciences (2018), 43 (8), 567-575CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. In biomedical sciences, the function of a protein of interest is investigated by altering its net activity and assessing the consequences for the cell or organism. To change the activity of a protein, a wide variety of chem. and genetic tools have been developed. The drawback of most of these tools is that they do not allow for reversible, spatial and temporal control. Here, we describe selected developments in photopharmacol. that aim at establishing such control over protein activity through bioactive mols. with photo-controlled potency. We also discuss why such control is desired and what challenges still need to be overcome for photopharmacol. to reach its maturity as a chem. biol. research tool.
- 5Borowiak, M.; Nahaboo, W.; Reynders, M.; Nekolla, K.; Jalinot, P.; Hasserodt, J.; Rehberg, M.; Delattre, M.; Zahler, S.; Vollmar, A.; Trauner, D.; Thorn-Seshold, O. Photoswitchable Inhibitors of Microtubule Dynamics Optically Control Mitosis and Cell Death. Cell 2015, 162, 403– 411, DOI: 10.1016/j.cell.2015.06.0495Photoswitchable Inhibitors of Microtubule Dynamics Optically Control Mitosis and Cell DeathBorowiak, Malgorzata; Nahaboo, Wallis; Reynders, Martin; Nekolla, Katharina; Jalinot, Pierre; Hasserodt, Jens; Rehberg, Markus; Delattre, Marie; Zahler, Stefan; Vollmar, Angelika; Trauner, Dirk; Thorn-Seshold, OliverCell (Cambridge, MA, United States) (2015), 162 (2), 403-411CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Small mols. that interfere with microtubule dynamics, such as Taxol and the Vinca alkaloids, are widely used in cell biol. research and as clin. anticancer drugs. However, their activity cannot be restricted to specific target cells, which also causes severe side effects in chemotherapy. Here, the authors introduce the photostatins, inhibitors that can be switched on and off in vivo by visible light, to optically control microtubule dynamics. Photostatins modulate microtubule dynamics with a subsecond response time and control mitosis in living organisms with single-cell spatial precision. In longer-term applications in cell culture, photostatins are up to 250 times more cytotoxic when switched on with blue light than when kept in the dark. Therefore, photostatins are both valuable tools for cell biol., and are promising as a new class of precision chemotherapeutics whose toxicity may be spatiotemporally constrained using light.
- 6Borowiak, M.; Küllmer, F.; Gegenfurtner, F.; Peil, S.; Nasufovic, V.; Zahler, S.; Thorn-Seshold, O.; Trauner, D.; Arndt, H.-D. Optical Manipulation of F-Actin with Photoswitchable Small Molecules. J. Am. Chem. Soc. 2020, 142, 9240– 9249, DOI: 10.1021/jacs.9b128986Optical Manipulation of F-Actin with Photoswitchable Small MoleculesBorowiak, Malgorzata; Kuellmer, Florian; Gegenfurtner, Florian; Peil, Sebastian; Nasufovic, Veselin; Zahler, Stefan; Thorn-Seshold, Oliver; Trauner, Dirk; Arndt, Hans-DieterJournal of the American Chemical Society (2020), 142 (20), 9240-9249CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Cell-permeable photoswitchable small mols., termed optojasps, are introduced to optically control the dynamics of the actin cytoskeleton and cellular functions that depend on it. These light-dependent effectors were designed from the F-actin-stabilizing marine depsipeptide jasplakinolide by functionalizing them with azobenzene photoswitches. As demonstrated, optojasps can be employed to control cell viability, cell motility, and cytoskeletal signaling with the high spatial and temporal resoln. that light affords. Optojasps can be expected to find applications in diverse areas of cell biol. research. They may also provide a template for photopharmacol. targeting the ubiquitous actin cytoskeleton with precision control in the micrometer range.
- 7Glotzer, M. The 3Ms of Central Spindle Assembly: Microtubules, Motors and MAPs. Nat. Rev. Mol. Cell Biol. 2009, 10, 9– 20, DOI: 10.1038/nrm26097The 3Ms of central spindle assembly: microtubules, motors and MAPsGlotzer, MichaelNature Reviews Molecular Cell Biology (2009), 10 (1), 9-20CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. During metaphase, sister chromatids are positioned at the midpoint of the microtubule-based mitotic spindle in prepn. for their segregation. The onset of anaphase triggers inactivation of the key mitotic kinase, cyclin-dependent kinase 1 (CDK1), and the poleward movement of sister chromatids. During anaphase, the mitotic spindle reorganizes in prepn. for cytokinesis. Kinesin motor proteins and microtubule-assocd. proteins bundle the plus ends of interpolar microtubules and generate the central spindle, which regulates cleavage furrow initiation and the completion of cytokinesis. Complementary approaches, including cell biol., genetics, and computational modeling, have provided new insights into the mechanism and regulation of central spindle assembly.
- 8Kapitein, L. C.; Hoogenraad, C. C. Building the Neuronal Microtubule Cytoskeleton. Neuron 2015, 87, 492– 506, DOI: 10.1016/j.neuron.2015.05.0468Building the Neuronal Microtubule CytoskeletonKapitein, Lukas C.; Hoogenraad, Casper C.Neuron (2015), 87 (3), 492-506CODEN: NERNET; ISSN:0896-6273. (Cell Press)Microtubules are one of the major cytoskeletal components of neurons, essential for many fundamental cellular and developmental processes, such as neuronal migration, polarity, and differentiation. Microtubules have been regarded as crit. structures for stable neuronal morphol. because they serve as tracks for long-distance transport, provide dynamic and mech. functions, and control local signaling events. Establishment and maintenance of the neuronal microtubule architecture requires tight control over different dynamic parameters, such as microtubule no., length, distribution, orientations, and bundling. Recent genetic studies have identified mutations in a wide variety of tubulin isotypes and microtubule-related proteins in many of the major neurodevelopmental and neurodegenerative diseases. Here, we highlight the functions of the neuronal microtubule cytoskeleton, its architecture, and the way its organization and dynamics are shaped by microtubule-related proteins.
- 9van Haren, J.; Charafeddine, R. A.; Ettinger, A.; Wang, H.; Hahn, K. M.; Wittmann, T. Local Control of Intracellular Microtubule Dynamics by EB1 Photodissociation. Nat. Cell Biol. 2018, 20, 252– 261, DOI: 10.1038/s41556-017-0028-59Local control of intracellular microtubule dynamics by EB1 photodissociationvan Haren, Jeffrey; Charafeddine, Rabab A.; Ettinger, Andreas; Wang, Hui; Hahn, Klaus M.; Wittmann, TorstenNature Cell Biology (2018), 20 (3), 252-261CODEN: NCBIFN; ISSN:1465-7392. (Nature Research)End-binding proteins (EBs) are adaptors that recruit functionally diverse microtubule plus-end-tracking proteins (+TIPs) to growing microtubule plus ends. To test with high spatial and temporal accuracy how, when and where +TIP complexes contribute to dynamic cell biol., it developed a photo-inactivated EB1 variant (.product. -EB1) by inserting a blue-light-sensitive protein-protein interaction module between the microtubule-binding and +TIP-binding domains of EB1. .product. -EB1 replaces endogenous EB1 function in the absence of blue light. By contrast, blue-light-mediated .product. -EB1 photodissocn. results in rapid +TIP complex disassembly, and acutely and reversibly attenuates microtubule growth independent of microtubule end assocn. of the microtubule polymerase CKAP5 (also known as ch-TOG and XMAP215). Local .product. -EB1 photodissocn. allows subcellular control of microtubule dynamics at the second and micrometer scale, and elicits aversive turning of migrating cancer cells. Importantly, light-mediated domain splitting can serve as a template to optically control other intracellular protein activities.
- 10Adikes, R. C.; Hallett, R. A.; Saway, B. F.; Kuhlman, B.; Slep, K. C. Control of Microtubule Dynamics Using an Optogenetic Microtubule plus End–F-Actin Cross-Linker. J. Cell Biol. 2018, 217, 779– 793, DOI: 10.1083/jcb.20170519010Control of microtubule dynamics using an optogenetic microtubule plus end-F-actin cross-linkerAdikes, Rebecca C.; Hallett, Ryan A.; Saway, Brian F.; Kuhlman, Brian; Slep, Kevin C.Journal of Cell Biology (2018), 217 (2), 779-793CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)We developed a novel optogenetic tool, SxIP-improved light-inducible dimer (iLID), to facilitate the reversible recruitment of factors to microtubule (MT) plus ends in an end-binding protein-dependent manner using blue light. We show that SxIP-iLID can track MT plus ends and recruit tgRFP-SspB upon blue light activation. We used this system to investigate the effects of crosslinking MT plus ends and F-actin in Drosophila melanogaster S2 cells to gain insight into spectraplakin function and mechanism. We show that SxIP-iLID can be used to temporally recruit an F-actin binding domain to MT plus ends and cross-link the MT and F-actin networks. Crosslinking decreases MT growth velocities and generates a peripheral MT exclusion zone. SxIP-iLID facilitates the general recruitment of specific factors to MT plus ends with temporal control enabling researchers to systematically regulate MT plus end dynamics and probe MT plus end function in many biol. processes.
- 11Meiring, J. C. M.; Grigoriev, I.; Nijenhuis, W.; Kapitein, L. C.; Akhmanova, A. Opto-Katanin: An Optogenetic Tool for Localized Microtubule Disassembly. bioRxiv 2021, 22, 473806 DOI: 10.1101/2021.12.22.473806There is no corresponding record for this reference.
- 12Liu, G. Y.; Chen, S.-C.; Shaiv, K.; Hong, S.-R.; Yang, W.-T.; Huang, S.-H.; Chang, Y.-C.; Cheng, H.; Lin, Y.-C. Precise Control of Microtubule Disassembly in Living Cells. bioRxiv 2021, 31, 463668 DOI: 10.1101/2021.10.08.463668There is no corresponding record for this reference.
- 13Wühr, M.; Tan, E. S.; Parker, S. K.; Detrich, H. W.; Mitchison, T. J. A Model for Cleavage Plane Determination in Early Amphibian and Fish Embryos. Curr. Biol. 2010, 20, 2040– 2045, DOI: 10.1016/j.cub.2010.10.02413A Model for Cleavage Plane Determination in Early Amphibian and Fish EmbryosWuehr, Martin; Tan, Edwin S.; Parker, Sandra K.; Detrich, H. William, III; Mitchison, Timothy J.Current Biology (2010), 20 (22), 2040-2045CODEN: CUBLE2; ISSN:0960-9822. (Cell Press)Summary: Current models for cleavage plane detn. propose that metaphase spindles are positioned and oriented by interactions of their astral microtubules with the cellular cortex, followed by cleavage in the plane of the metaphase plate []. We show that in early frog and fish embryos, where cells are unusually large, astral microtubules in metaphase are too short to position and orient the spindle. Rather, the preceding interphase aster centers and orients a pair of centrosomes prior to nuclear envelope breakdown, and the spindle assembles between these prepositioned centrosomes. Interphase asters center and orient centrosomes with dynein-mediated pulling forces. These forces act before astral microtubules contact the cortex; thus, dynein must pull from sites in the cytoplasm, not the cell cortex as is usually proposed for smaller cells. Aster shape is detd. by interactions of the expanding periphery with the cell cortex or with an interaction zone that forms between sister-asters in telophase. We propose a model to explain cleavage plane geometry in which the length of astral microtubules is limited by interaction with these boundaries, causing length asymmetries. Dynein anchored in the cytoplasm then generates length-dependent pulling forces, which move and orient centrosomes.
- 14Josa-Culleré, L.; Llebaria, A. In the Search for Photocages Cleavable with Visible Light: An Overview of Recent Advances and Chemical Strategies. ChemPhotoChem 2021, 5, 296– 314, DOI: 10.1002/cptc.202000253There is no corresponding record for this reference.
- 15Müller-Deku, A.; Meiring, J. C. M.; Loy, K.; Kraus, Y.; Heise, C.; Bingham, R.; Jansen, K. I.; Qu, X.; Bartolini, F.; Kapitein, L. C.; Akhmanova, A.; Ahlfeld, J.; Trauner, D.; Thorn-Seshold, O. Photoswitchable Paclitaxel-Based Microtubule Stabilisers Allow Optical Control over the Microtubule Cytoskeleton. Nat. Commun. 2020, 11, 4640 DOI: 10.1038/s41467-020-18389-615Photoswitchable paclitaxel-based microtubule stabilisers allow optical control over the microtubule cytoskeletonMueller-Deku, Adrian; Meiring, Joyce C. M.; Loy, Kristina; Kraus, Yvonne; Heise, Constanze; Bingham, Rebekkah; Jansen, Klara I.; Qu, Xiaoyi; Bartolini, Francesca; Kapitein, Lukas C.; Akhmanova, Anna; Ahlfeld, Julia; Trauner, Dirk; Thorn-Seshold, OliverNature Communications (2020), 11 (1), 4640CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Small mol. inhibitors are prime reagents for studies in microtubule cytoskeleton research, being applicable across a range of biol. models and not requiring genetic engineering. However, traditional chem. inhibitors cannot be exptl. applied with spatiotemporal precision suiting the length and time scales inherent to microtubule-dependent cellular processes. We have synthesized photoswitchable paclitaxel-based microtubule stabilizers, whose binding is induced by photoisomerisation to their metastable state. Photoisomerising these reagents in living cells allows optical control over microtubule network integrity and dynamics, cell division and survival, with biol. response on the timescale of seconds and spatial precision to the level of individual cells within a population. In primary neurons, they enable regulation of microtubule dynamics resolved to subcellular regions within individual neurites. These azobenzene-based microtubule stabilizers thus enable non-invasive, spatiotemporally precise modulation of the microtubule cytoskeleton in living cells, and promise new possibilities for studying intracellular transport, cell motility, and neuronal physiol.
- 16Gao, L.; Meiring, J. C. M.; Heise, C.; Rai, A.; Müller-Deku, A.; Akhmanova, A.; Thorn-Seshold, J.; Thorn-Seshold, O. Photoswitchable Epothilone-Based Microtubule Stabilisers Allow GFP-Imaging-Compatible, Optical Control over the Microtubule Cytoskeleton. Angew. Chem., Int. Ed. 2021, 48, e202114614 DOI: 10.1002/anie.202114614There is no corresponding record for this reference.
- 17Rastogi, S. K.; Zhao, Z.; Gildner, M. B.; Shoulders, B. A.; Velasquez, T. L.; Blumenthal, M. O.; Wang, L.; Li, X.; Hudnall, T. W.; Betancourt, T.; Du, L.; Brittain, W. J. Synthesis, Optical Properties and in Vitro Cell Viability of Novel Spiropyrans and Their Photostationary States. Tetrahedron 2021, 80, 131854 DOI: 10.1016/j.tet.2020.13185417Synthesis, optical properties and in vitro cell viability of novel spiropyrans and their photostationary statesRastogi, Shiva K.; Zhao, Zhenze; Gildner, M. Brenton; Shoulders, Ben A.; Velasquez, Tara L.; Blumenthal, Madeleine O.; Wang, Lei; Li, Xiaopeng; Hudnall, Todd W.; Betancourt, Tania; Du, Liqin; Brittain, William J.Tetrahedron (2021), 80 (), 131854CODEN: TETRAB; ISSN:0040-4020. (Elsevier Ltd.)A novel class of spiropyrans I (R = H, OEt, OMe; R1 = R2 = R3 = H, OMe; R4 = H, NO2) was synthesized using a multistep process that involves three key intermediates: (a) diazonium-tetrafluoroborate, (b) hydrazine and (c) indolium iodide. The SP analogs I were confirmed as being able to isomerize and attain a photostationary state (PSS) upon irradn. with UV light (UV, 365 nm) in an aq. environment. UV-visible absorption spectra were recorded to confirm the isomerization properties. The ability of the synthesized compds. I to induce growth inhibition of HeLa cervical cancer cells was assessed via the MTT assay after incubation with either the SP or their PSS. The IC50 values of two PSS II (R = H; R1 = R2 = R3 = OMe; R4 = nitro, R = R1 = R2 = R3 = OMe; R4 = H), were obsd. to be around 14 ± 4 fold lower (26 ± 3μM) than their corresponding SPs. The most cytotoxic compds. I [R = R1 = R2 = R3 = R4 = H (III)] and II [R = R1 = R2 = R3 = R4 = H (IV)] showed the lowest IC50 values (12μM). An in vitro tubulin polymn. assay showed that III and IV exhibited the greatest difference in tubulin inhibition relative to unirradiated I (R = R1 = R2 = R3 = H, R4 = NO2; R = R1 = R2 = R3 = OMe, R4 = H).
- 18Gao, L.; Meiring, J. C. M.; Kraus, Y.; Wranik, M.; Weinert, T.; Pritzl, S. D.; Bingham, R.; Ntouliou, E.; Jansen, K. I.; Olieric, N.; Standfuss, J.; Kapitein, L. C.; Lohmüller, T.; Ahlfeld, J.; Akhmanova, A.; Steinmetz, M. O.; Thorn-Seshold, O. A Robust, GFP-Orthogonal Photoswitchable Inhibitor Scaffold Extends Optical Control over the Microtubule Cytoskeleton. Cell Chem. Biol. 2021, 28, 228– 241, DOI: 10.1016/j.chembiol.2020.11.00718A Robust, GFP-Orthogonal Photoswitchable Inhibitor Scaffold Extends Optical Control over the Microtubule CytoskeletonGao, Li; Meiring, Joyce C. M.; Kraus, Yvonne; Wranik, Maximilian; Weinert, Tobias; Pritzl, Stefanie D.; Bingham, Rebekkah; Ntouliou, Evangelia; Jansen, Klara I.; Olieric, Natacha; Standfuss, Joerg; Kapitein, Lukas C.; Lohmueller, Theobald; Ahlfeld, Julia; Akhmanova, Anna; Steinmetz, Michel O.; Thorn-Seshold, OliverCell Chemical Biology (2021), 28 (2), 228-241.e6CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Optically controlled chem. reagents, termed photopharmaceuticals, are powerful tools for precise spatiotemporal control of proteins particularly when genetic methods, such as knockouts or optogenetics are not viable options. However, current photopharmaceutical scaffolds, such as azobenzenes are intolerant of GFP/YFP imaging and are metabolically labile, posing severe limitations for biol. use. We rationally designed a photoswitchable SBT scaffold to overcome these problems, then derivatized it to create exceptionally metabolically robust and fully GFP/YFP-orthogonal SBTub photopharmaceutical tubulin inhibitors. Lead compd. SBTub3 allows temporally reversible, cell-precise, and even subcellularly precise photomodulation of microtubule dynamics, organization, and microtubule-dependent processes. By overcoming the previous limitations of microtubule photopharmaceuticals, SBTubs offer powerful applications in cell biol., and their robustness and druglikeness are favorable for intracellular biol. control in in vivo applications. We furthermore expect that the robustness and imaging orthogonality of the SBT scaffold will inspire other derivatizations directed at extending the photocontrol of a range of other biol. targets.
- 19Sailer, A.; Meiring, J. C. M.; Heise, C.; Pettersson, L. N.; Akhmanova, A.; Thorn-Seshold, J.; Thorn-Seshold, O. Pyrrole Hemithioindigo Antimitotics with Near-Quantitative Bidirectional Photoswitching That Photocontrol Cellular Microtubule Dynamics with Single-Cell Precision. Angew. Chem., Int. Ed. 2021, 60, 23695– 23704, DOI: 10.1002/anie.20210479419Pyrrole Hemithioindigo Antimitotics with Near-Quantitative Bidirectional Photoswitching that Photocontrol Cellular Microtubule Dynamics with Single-Cell PrecisionSailer, Alexander; Meiring, Joyce C. M.; Heise, Constanze; Pettersson, Linda N.; Akhmanova, Anna; Thorn-Seshold, Julia; Thorn-Seshold, OliverAngewandte Chemie, International Edition (2021), 60 (44), 23695-23704CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We report the first cellular application of the emerging near-quant. photoswitch pyrrole hemithioindigo, by rationally designing photopharmaceutical PHTub inhibitors of the cytoskeletal protein tubulin. PHTubs allow simultaneous visible-light imaging and photoswitching in live cells, delivering cell-precise photomodulation of microtubule dynamics, and photocontrol over cell cycle progression and cell death. This is the first acute use of a hemithioindigo photopharmaceutical for high-spatiotemporal-resoln. biol. control in live cells. It addnl. demonstrates the utility of near-quant. photoswitches, by enabling a dark-active design to overcome residual background activity during cellular photopatterning. This work opens up new horizons for high-precision microtubule research using PHTubs and shows the cellular applicability of pyrrole hemithioindigo as a valuable scaffold for photocontrol of a range of other biol. targets.
- 20Sailer, A.; Ermer, F.; Kraus, Y.; Lutter, F. H.; Donau, C.; Bremerich, M.; Ahlfeld, J.; Thorn-Seshold, O. Hemithioindigos for Cellular Photopharmacology: Desymmetrised Molecular Switch Scaffolds Enabling Design Control over the Isomer-Dependency of Potent Antimitotic Bioactivity. ChemBioChem 2019, 20, 1305– 1314, DOI: 10.1002/cbic.20180075220Hemithioindigos for Cellular Photopharmacology: Desymmetrised Molecular Switch Scaffolds Enabling Design Control over the Isomer-Dependency of Potent Antimitotic BioactivitySailer, Alexander; Ermer, Franziska; Kraus, Yvonne; Lutter, Ferdinand H.; Donau, Carsten; Bremerich, Maximilian; Ahlfeld, Julia; Thorn-Seshold, OliverChemBioChem (2019), 20 (10), 1305-1314CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Druglike small mols. with photoswitchable bioactivity-photopharmaceuticals-allow biologists to perform studies with exquisitely precise and reversible, spatial and temporal control over crit. biol. systems inaccessible to genetic manipulation. The photoresponsive pharmacophores disclosed have been almost exclusively azobenzenes, which has limited the structural and substituent scope of photopharmacol. More detrimentally, for azobenzene reagents, it is not researchers' needs for adapted exptl. tools, but rather protein binding site sterics, that typically force whether the trans (dark) or cis (lit) isomer is the more bioactive. We now present the rational design of HOTubs, the first hemithioindigo-based pharmacophores enabling photoswitchable control over endogenous biol. activity in cellulo. HOTubs optically control microtubule depolymn. and cell death in unmodified mammalian cells. Notably, we show how the asymmetry of hemithioindigos allows a priori design of either Z- or E- (dark- or lit)-toxic antimitotics, whereas the corresponding azobenzenes are exclusively lit-toxic. We thus demonstrate that hemithioindigos enable an important expansion of the substituent and design scope of photopharmacol. interventions for biol. systems.
- 21Sailer, A.; Ermer, F.; Kraus, Y.; Bingham, R.; Lutter, F. H.; Ahlfeld, J.; Thorn-Seshold, O. Potent Hemithioindigo-Based Antimitotics Photocontrol the Microtubule Cytoskeleton in Cellulo. Beilstein J. Org. Chem. 2020, 16, 125– 134, DOI: 10.3762/bjoc.16.1421Potent hemithioindigo-based antimitotics photocontrol the microtubule cytoskeleton in celluloSailer, Alexander; Ermer, Franziska; Kraus, Yvonne; Bingham, Rebekkah; Lutter, Ferdinand H.; Ahlfeld, Julia; Thorn-Seshold, OliverBeilstein Journal of Organic Chemistry (2020), 16 (), 125-134CODEN: BJOCBH; ISSN:1860-5397. (Beilstein-Institut zur Foerderung der Chemischen Wissenschaften)Uniquely, in contrast to other photoswitches that have been applied to biol., the pseudosym. hemithioindigo scaffold has allowed the creation of both dark-active and lit-active photopharmaceuticals for the same binding site by a priori design. However, the potency of previous hemithioindigo photopharmaceuticals has not been optimal for their translation to other biol. models. Inspired by the structure of tubulin-inhibiting indanones, we created hemithioindigo-based indanone-like tubulin inhibitors (HITubs) and optimized their cellular potency as antimitotic photopharmaceuticals. The use of the hemithioindigo scaffold also permitted us to employ a parahydroxyhemistilbene motif, a structural feature which is denied to most azobenzenes due to the negligibly short lifetimes of their metastable Z-isomers, which proved crucial to enhancing the potency and photoswitchability. The HITubs were ten times more potent than previously reported hemithioindigo photopharmaceutical antimitotics in a series of cell-free and cellular assays, and allowed robust photocontrol over tubulin polymn., microtubule (MT) network structure, cell cycle, and cell survival. Addnl., as the hemithioindigo scaffold allows photoswitchable bioactivity for substituent patterns inaccessible to the majority of current photopharmaceuticals, wider adoption of the hemithioindigo scaffold may significantly expand the scope of cellular and in vivo targets addressable by photopharmacol.
- 22Engdahl, A. J.; Torres, E. A.; Lock, S. E.; Engdahl, T. B.; Mertz, P. S.; Streu, C. N. Synthesis, Characterization, and Bioactivity of the Photoisomerizable Tubulin Polymerization Inhibitor Azo-Combretastatin A4. Org. Lett. 2015, 17, 4546– 4549, DOI: 10.1021/acs.orglett.5b0226222Synthesis, Characterization, and Bioactivity of the Photoisomerizable Tubulin Polymerization Inhibitor azo-Combretastatin A4Engdahl, Ashton J.; Torres, Edith A.; Lock, Sarah E.; Engdahl, Taylor B.; Mertz, Pamela S.; Streu, Craig N.Organic Letters (2015), 17 (18), 4546-4549CODEN: ORLEF7; ISSN:1523-7052. (American Chemical Society)Combretastatin A4 is a stilbenoid tubulin binding mitotic inhibitor whose conformation greatly influences its potency, making it an excellent candidate for adaptation as a photoactivatable tool. Herein, the authors report a novel synthesis, the facile isomerization with com. grade equipment, and biol. activity of azo-combretastatin A4 in vitro and in human cancer cells. Photoisomerized azo-combretastatin A4 is at least 200-fold more potent in cellular culture, making it a promising phototherapeutic and biomedical research tool.
- 23Zenker, J.; White, M. D.; Gasnier, M.; Alvarez, Y. D.; Lim, H. Y. G.; Bissiere, S.; Biro, M.; Plachta, N. Expanding Actin Rings Zipper the Mouse Embryo for Blastocyst Formation. Cell 2018, 173, 776– 791, DOI: 10.1016/j.cell.2018.02.03523Expanding Actin Rings Zipper the Mouse Embryo for Blastocyst FormationZenker, Jennifer; White, Melanie D.; Gasnier, Maxime; Alvarez, Yanina D.; Lim, Hui Yi Grace; Bissiere, Stephanie; Biro, Mate; Plachta, NicolasCell (Cambridge, MA, United States) (2018), 173 (3), 776-791.e17CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Transformation from morula to blastocyst is a defining event of preimplantation embryo development. During this transition, the embryo must establish a paracellular permeability barrier to enable expansion of the blastocyst cavity. Here, using live imaging of mouse embryos, we reveal an actin-zippering mechanism driving this embryo sealing. Preceding blastocyst stage, a cortical F-actin ring assembles at the apical pole of the embryo's outer cells. The ring structure forms when cortical actin flows encounter a network of polar microtubules that exclude F-actin. Unlike stereotypical actin rings, the actin rings of the mouse embryo are not contractile, but instead, they expand to the cell-cell junctions. Here, they couple to the junctions by recruiting and stabilizing adherens and tight junction components. Coupling of the actin rings triggers localized myosin II accumulation, and it initiates a tension-dependent zippering mechanism along the junctions that is required to seal the embryo for blastocyst formation.
- 24Theisen, U.; Ernst, A. U.; Heyne, R. L. S.; Ring, T. P.; Thorn-Seshold, O.; Köster, R. W. Microtubules and Motor Proteins Support Zebrafish Neuronal Migration by Directing Cargo. J. Cell Biol. 2020, 219, e201908040 DOI: 10.1083/jcb.20190804024Microtubules and motor proteins support zebrafish neuronal migration by directing cargoTheisen, Ulrike; Ernst, Alexander U.; Heyne, Ronja L. S.; Ring, Tobias P.; Thorn-Seshold, Oliver; Koester, Reinhard W.Journal of Cell Biology (2020), 219 (10), e201908040CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Neuronal migration during development is necessary to form an ordered and functional brain. Postmitotic neurons require microtubules and dynein to move, but the mechanisms by which they contribute to migration are not fully characterized. Using tegmental hindbrain nuclei neurons in zebrafish embryos together with subcellular imaging, optogenetics, and photopharmacol., we show that, in vivo, the centrosome's position relative to the nucleus is not linked to greatest motility in this cell type. Nevertheless, microtubules, dynein, and kinesin-1 are essential for migration, and we find that interference with endosome formation or the Golgi app. impairs migration to a similar extent as disrupting microtubules. In addn., an imbalance in the traffic of the model cargo Cadherin-2 also reduces neuronal migration. These results lead us to propose that microtubules act as cargo carriers to control spatiotemporal protein distribution, which in turn controls motility. This adds crucial insights into the variety of ways that microtubules can support successful neuronal migration in vivo.
- 25Gavin, J.; Ruiz, J. F. M.; Kedziora, K.; Windle, H.; Kelleher, D. P.; Gilmer, J. F. Structure Requirements for Anaerobe Processing of Azo Compounds: Implications for Prodrug Design. Bioorg. Med. Chem. Lett. 2012, 22, 7647– 7652, DOI: 10.1016/j.bmcl.2012.10.01425Structure requirements for anaerobe processing of azo compounds: Implications for prodrug designGavin, Jason; Ruiz, Juan F. Marquez; Kedziora, Kinga; Windle, Henry; Kelleher, Dermot P.; Gilmer, John F.Bioorganic & Medicinal Chemistry Letters (2012), 22 (24), 7647-7652CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)This Letter generalizes the metab. of the azo class of compds. by Clostridium perfringens, an anaerobe found in the human colon. A recently reported 5-aminosalicylic acid-based prednisolone prodrug was shown to release the drug when incubated with the bacteria, while the para-aminobenzoic acid (PABA) based analog did not. Instead, it showed a new HPLC peak with a relatively close retention time to the parent which was identified by LCMS as the partially reduced hydrazine product. This Letter investigates azoredn. across a panel of substrates with varying degrees of electronic and steric similarity to the PABA-based compd. Azo compds. with an electron donating group on the azo-contg. arom. ring showed immediate disproportionation to their parent amines without any detection of hydrazine intermediates by HPLC. Compds. contg. only electron withdrawing groups are partially and reversibly reduced to produce a stable detectable hydrazine. They do not disproportionate to their parent amines, but regenerate the parent azo compd. This incomplete redn. is relevant to the design of azo-based prodrugs and the toxicol. of azo-based dyes.
- 26Sheldon, J. E.; Dcona, M. M.; Lyons, C. E.; Hackett, J. C.; Hartman, M. C. T. Photoswitchable Anticancer Activity via Trans-Cis Isomerization of a Combretastatin A-4 Analog. Org. Biomol. Chem. 2016, 14, 40– 49, DOI: 10.1039/c5ob02005k26Photoswitchable anticancer activity via trans-cis isomerization of a combretastatin A-4 analogSheldon, Jonathon E.; Dcona, M. Michael; Lyons, Charles E.; Hackett, John C.; Hartman, Matthew C. T.Organic & Biomolecular Chemistry (2016), 14 (1), 40-49CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)Combretastatin A-4 (CA4) is highly potent anticancer drug that acts as an inhibitor of tubulin polymn. The core of the CA4 structure contains a cis-stilbene, and it is known that the trans isomer is significantly less potent. The authors prepd. an azobenzene analog of CA4 (Azo-CA4) that shows 13-35-fold enhancement in potency upon illumination. EC50 values in the light were in the mid nM range. Due to its ability to thermally revert to less toxic trans form, Azo-CA4 also has the ability to automatically turn its activity off with time. Azo-CA4 is less potent than CA-4 because it degrades in the presence of glutathione as evidenced by UV-Vis spectroscopy and ESI-MS. Nevertheless, Azo-CA4 represents a promising strategy for switchable potency for treatment of cancer.
- 27An, Y.; Chen, C.; Zhu, J.; Dwivedi, P.; Zhao, Y.; Wang, Z. Hypoxia-Induced Activity Loss of a Photo-Responsive Microtubule Inhibitor Azobenzene Combretastatin A4. Front. Chem. Sci. Eng. 2020, 14, 880– 888, DOI: 10.1007/s11705-019-1864-627Hypoxia-induced activity loss of a photo-responsive microtubule inhibitor azobenzene combretastatin A4An, Yang; Chen, Chao; Zhu, Jundong; Dwivedi, Pankaj; Zhao, Yanjun; Wang, ZhengFrontiers of Chemical Science and Engineering (2020), 14 (5), 880-888CODEN: FCSEA3; ISSN:2095-0187. (Springer)The conformation-dependent activity of azobenzene combretastatin A4 (Azo-CA4) provides a unique approach to reduce the side-effects of chemotherapy, due to the light-triggered conformation transition of its azobenzene moiety. Under hypoxic tumor microenvironment, however, the high expression of azoreductase can reduce azobenzene to aniline. It was postulated that the Azo-CA4 might be degraded under hypoxia, resulting in the decrease of its anti-tumor activity. The aim of this study was to verify such hypothesis in HeLa cells in vitro. The quant. drug concn. anal. shows the ratio-metric formation of degrdn. end-products, confirming the bioredn. of Azo-CA4. The tubulin staining study indicates that Azo-CA4 loses the potency of switching off microtubule dynamics under hypoxia. Furthermore, the cell cycle anal. shows that the ability of Azo-CA4 to induce mitotic arrest is lost at low oxygen content. Therefore, the cytotoxicity of Azo-CA4 is compromised under hypoxia. In contrast, combretastatin A4 as a pos. control maintains the potency to inhibit tubulin polymn. and break down the nuclei irresp. of light irradn. and oxygen level. This work highlights the influence of hypoxic tumor microenvironment on the anti-tumor potency of Azo-CA4, which should be considered during the early stage of designing translational Azo-CA4 delivery systems.
- 28Hüll, K.; Morstein, J.; Trauner, D. In Vivo Photopharmacology. Chem. Rev. 2018, 118, 10710– 10747, DOI: 10.1021/acs.chemrev.8b0003728In Vivo PhotopharmacologyHull, Katharina; Morstein, Johannes; Trauner, DirkChemical Reviews (Washington, DC, United States) (2018), 118 (21), 10710-10747CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Synthetic photoswitches have been known for many years, but their usefulness in biol., pharmacol., and medicine has only recently been systematically explored. Over the past decade photopharmacol. has grown into a vibrant field. As the photophys., pharmacodynamic, and pharmacokinetic properties of photoswitches, such as azobenzenes, have become established, they have been applied to a wide range of biol. targets. These include transmembrane proteins (ion channels, transporters, G protein-coupled receptors, receptor-linked enzymes), sol. proteins (kinases, proteases, factors involved in epigenetic regulation), lipid membranes, and nucleic acids. In this review, the authors provide an overview of photopharmacol. using synthetic switches that have been applied in vivo, i.e., in living cells and organisms. The authors discuss the scope and limitations of this approach to study biol. function and the challenges it faces in translational medicine. The relationships between synthetic photoswitches, natural chromophores used in optogenetics, and caged ligands are addressed.
- 29Lachmann, D.; Lahmy, R.; König, B. Fulgimides as Light-Activated Tools in Biological Investigations: Fulgimides as Light-Activated Tools in Biological Investigations. Eur. J. Org. Chem. 2019, 2019, 5018– 5024, DOI: 10.1002/ejoc.20190021929Fulgimides as Light-Activated Tools in Biological InvestigationsLachmann, D.; Lahmy, R.; Koenig, B.European Journal of Organic Chemistry (2019), 2019 (31-32), 5018-5024CODEN: EJOCFK; ISSN:1099-0690. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. With high spatiotemporal control, the conformation, rigidity and electronics of photoresponsive bioactive mols. can be altered. This, in turn, allows for control over the biol. properties of these mols. Incorporation of a photoswitchable moiety into a no. of reported inhibitors, ligands and modulators has demonstrated the ability to modulate enzyme, receptor and ion channel responses using light. To date, the major classes of photoswitches explored in biol. applications have been the azobenzenes and diarylethenes. Even though the use of these photoswitches has established the value of photoresponsive mols. as biol. tools, several limitations have become apparent. Fulgimides represent a promising class of photoswitches that are not widely used for such biol. purposes. Their properties are similar to that of diarylethenes, as their photochromism is based on a 6π-electrocyclic rearrangement, however, fulgimides have the added advantage of thermal stability for both isomers. Fulgimides exhibit high photostationary states and fatigue resistance, with the ability to switch in aq. buffer solns. In this minireview, these advantageous photophys. properties will be discussed, as well as the use of fulgimides in biol. investigations.
- 30Fuchter, M. J. On the Promise of Photopharmacology Using Photoswitches: A Medicinal Chemist’s Perspective. J. Med. Chem. 2020, 63, 11436– 11447, DOI: 10.1021/acs.jmedchem.0c0062930On the Promise of Photopharmacology Using Photoswitches: A Medicinal Chemist's PerspectiveFuchter, Matthew J.Journal of Medicinal Chemistry (2020), 63 (20), 11436-11447CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Photopharmacol. is a growing area of endeavor that employs photoswitchable ligands to allow for light-dependent pharmacol. activity. By coupling light to therapeutic action, improved spatial and temporal selectivity can be achieved and subsequently harnessed for new concepts in therapy. Tremendous progress has already been made, with photopharmacol. agents now reported against a wide array of target classes and light-dependent results demonstrated in a range of live cell and animal models. Several challenges remain, however, esp. in order for photopharmacol. to truly impact the clin. management of disease. This Perspective aims to summarize these challenges, particularly with attention to the medicinal chem. that will be unavoidably required for the further translation of these agents/approaches. By clearly defining challenges for drug hunters, it is hoped that further research into the medicinal chem. of photopharmacol. agents will be stimulated, ultimately enabling full realization of the huge potential for this exciting field.
- 31Welleman, I. M.; Hoorens, M. W. H.; Feringa, B. L.; Boersma, H. H.; Szymański, W. Photoresponsive Molecular Tools for Emerging Applications of Light in Medicine. Chem. Sci. 2020, 11, 11672– 11691, DOI: 10.1039/D0SC04187D31Photoresponsive molecular tools for emerging applications of light in medicineWelleman, Ilse M.; Hoorens, Mark W. H.; Feringa, Ben L.; Boersma, Hendrikus H.; Szymanski, WiktorChemical Science (2020), 11 (43), 11672-11691CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A review. Light-based therapeutic and imaging modalities, which emerge in clin. applications, rely on mol. tools, such as photocleavable protecting groups and photoswitches that respond to photonic stimulus and translate it into a biol. effect. However, optimization of their key parameters (activation wavelength, band sepn., fatigue resistance and half-life) is necessary to enable application in the medical field. In this perspective, we describe the applications scenarios that can be envisioned in clin. practice and then we use those scenarios to explain the necessary properties that the photoresponsive tools used to control biol. function should possess, highlighted by examples from medical imaging, drug delivery and photopharmacol. We then present how the (photo)chem. parameters are currently being optimized and an outlook is given on pharmacol. aspects (toxicity, soly., and stability) of light-responsive mols. With these interdisciplinary insights, we aim to inspire the future directions for the development of photocontrolled tools that will empower clin. applications of light.
- 32Gaspari, R.; Prota, A. E.; Bargsten, K.; Cavalli, A.; Steinmetz, M. O. Structural Basis of Cis- and Trans-Combretastatin Binding to Tubulin. Chem 2017, 2, 102– 113, DOI: 10.1016/j.chempr.2016.12.00532Structural Basis of cis- and trans-Combretastatin Binding to TubulinGaspari, Roberto; Prota, Andrea E.; Bargsten, Katja; Cavalli, Andrea; Steinmetz, Michel O.Chem (2017), 2 (1), 102-113CODEN: CHEMVE; ISSN:2451-9294. (Cell Press)Combretastatin A4 (CA-4) derivs. are microtubule-destabilizing agents, some of which are in advanced clin. trials for cancer therapy. The active cis conformation of CA-4 can readily isomerize into a thermodynamically more stable but significantly less active trans form. Here, we solved the high-resoln. crystal structure of cis-CA-4 in complex with tubulin. The compd. binds to the colchicine site of tubulin and displays both common and distinct interaction points with colchicine. Using metadynamics simulations, we generated the trans form of the ligand within its binding site and computed the relative binding free energy of the cis-CA-4 and trans-CA-4 isomers via a thermodn. cycle. The calcns. suggest structural distortions of the bound trans-CA-4 mol. as the likely cause of its reduced activity in comparison with that of its cis isomer. Our findings could open up unique possibilities for structure-guided drug engineering with the aim of discovering combretastatin variants with improved chem. properties and pharmacol. profiles.
- 33Tarade, D.; Ma, D.; Pignanelli, C.; Mansour, F.; Simard, D.; Berg, S.; van den Gauld, J.; McNulty, J.; Pandey, S. Structurally Simplified Biphenyl Combretastatin A4 Derivatives Retain in Vitro Anti-Cancer Activity Dependent on Mitotic Arrest. PLoS One 2017, 12, e0171806 DOI: 10.1371/journal.pone.0171806There is no corresponding record for this reference.
- 34Jacobson, P. Ueber Bildung von Anhydroverbindungen Des Orthoamidophenylmercaptans Aus Thioaniliden. Ber. Dtsch. Chem. Ges. 1886, 19, 1067– 1077, DOI: 10.1002/cber.188601901239There is no corresponding record for this reference.
- 35Ma, D.; Xie, S.; Xue, P.; Zhang, X.; Dong, J.; Jiang, Y. Efficient and Economical Access to Substituted Benzothiazoles: Copper-Catalyzed Coupling of 2-Haloanilides with Metal Sulfides and Subsequent Condensation. Angew. Chem., Int. Ed. 2009, 48, 4222– 4225, DOI: 10.1002/anie.20090048635Efficient and economical access to substituted benzothiazoles: copper-catalyzed coupling of 2-haloanilides with metal sulfides and subsequent condensationMa, Dawei; Xie, Siwei; Xue, Peng; Zhang, Xiaojing; Dong, Jinhua; Jiang, YongwenAngewandte Chemie, International Edition (2009), 48 (23), 4222-4225, S4222/1-S4222/18CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The first metal-catalyzed direct coupling of metal sulfides with aryl halides and subsequent intramol. condensation provided substituted benzothiazoles. A wide range of functional groups are tolerated under the reaction conditions.
- 36Tron, G. C.; Pirali, T.; Sorba, G.; Pagliai, F.; Busacca, S.; Genazzani, A. A. Medicinal Chemistry of Combretastatin A4: Present and Future Directions. J. Med. Chem. 2006, 49, 3033– 3044, DOI: 10.1021/jm051290336Medicinal chemistry of combretastatin A4: Present and future directionsTron, Gian Cesare; Pirali, Tracey; Sorba, Giovanni; Pagliai, Francesca; Busacca, Sara; Genazzani, Armando A.Journal of Medicinal Chemistry (2006), 49 (11), 3033-3044CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review with refs. A review on the medicinal chem. of combretastatin A4 (CA-4), a compd. with antitumor properties. The mechanism of action of CA-4, including its ability to inhibit tubulin polymn., is discussed, along with selected analogs of CA-4 modified on the arom. rings A and B, substituted Ph rings, heterocycling rings, non-substituted arom. rings, modifications on the double bond, and analogs where the olefinic group is replaced by a ring. Some issues related to the pharmacol. of CA-4 and future directions in research are considered.
- 37Kraus, Y.; Glas, C.; Melzer, B.; Gao, L.; Heise, C.; Preuße, M.; Ahlfeld, J.; Bracher, F.; Thorn-Seshold, O. Isoquinoline-Based Biaryls as a Robust Scaffold for Microtubule Inhibitors. Eur. J. Med. Chem. 2020, 186, 111865 DOI: 10.1016/j.ejmech.2019.11186537Isoquinoline-based biaryls as a robust scaffold for microtubule inhibitorsKraus, Yvonne; Glas, Carina; Melzer, Benedikt; Gao, Li; Heise, Constanze; Preusse, Monique; Ahlfeld, Julia; Bracher, Franz; Thorn-Seshold, OliverEuropean Journal of Medicinal Chemistry (2020), 186 (), 111865CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)We here report the discovery of isoquinoline-based biaryls as a new scaffold for colchicine domain tubulin inhibitors. Colchicinoid inhibitors offer highly desirable cytotoxic and vascular disrupting bioactivities, but their further development requires improving in vivo robustness and tolerability: properties that both depend on the scaffold structure employed. We have developed isoquinoline-based biaryls as a novel scaffold for high-potency tubulin inhibitors, with excellent robustness, druglikeness, and facile late-stage structural diversification, accessible through a tolerant synthetic route. We confirmed their bioactivity mechanism in vitro, developed sol. prodrugs, and established safe in vivo dosing in mice. By addressing several problems facing the current families of inhibitors, we expect that this new scaffold will find a range of in vivo applications towards translational use in cancer therapy.
- 38Thorn-Seshold, O.; Meiring, J. C. M. Photocontrolling Microtubule Dynamics with Photoswitchable Chemical Reagents. In Microtubules─Methods and Protocols; Springer International Publishing, 2022; Chapter 26.There is no corresponding record for this reference.
- 39Florian, S.; Mitchison, T. J. Anti-Microtubule Drugs. In The Mitotic Spindle: Methods and Protocols; Chang, P.; Ohi, R., Eds.; Springer: New York, NY, 2016; Vol. 1413, pp 403– 421.There is no corresponding record for this reference.
- 40Roostalu, J.; Thomas, C.; Cade, N. I.; Kunzelmann, S.; Taylor, I. A.; Surrey, T. The Speed of GTP Hydrolysis Determines GTP Cap Size and Controls Microtubule Stability. eLife 2020, 9, e51992 DOI: 10.7554/eLife.5199240The speed of GTP hydrolysis determines GTP cap size and controls microtubule stabilityRoostalu, Johanna; Thomas, Claire; Cade, Nicholas Ian; Kunzelmann, Simone; Taylor, Ian A.; Surrey, ThomaseLife (2020), 9 (), e51992CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Microtubules are cytoskeletal polymers whose function depends on their property to switch between states of growth and shrinkage. Growing microtubules are thought to be stabilized by a GTP cap at their ends. The nature of this cap, however, is still poorly understood. End Binding proteins (EBs) recruit a diverse range of regulators of microtubule function to growing microtubule ends. Whether the EB binding region is identical to the GTP cap is unclear. Using mutated human tubulin with blocked GTP hydrolysis, we demonstrate that EBs bind with high affinity to the GTP conformation of microtubules. Slowing-down GTP hydrolysis leads to extended GTP caps. We find that cap length dets. microtubule stability and that the microtubule conformation changes gradually in the cap as GTP is hydrolyzed. These results demonstrate the crit. importance of the kinetics of GTP hydrolysis for microtubule stability and establish that the GTP cap coincides with the EB-binding region.
- 41Linnemann, J. R.; Miura, H.; Meixner, L. K.; Irmler, M.; Kloos, U. J.; Hirschi, B.; Bartsch, H. S.; Sass, S.; Beckers, J.; Theis, F. J.; Gabka, C.; Sotlar, K.; Scheel, C. H. Quantification of Regenerative Potential in Primary Human Mammary Epithelial Cells. Development 2015, 31, 3239– 3251, DOI: 10.1242/dev.123554There is no corresponding record for this reference.
- 42Buchmann, B.; Engelbrecht, L. K.; Fernandez, P.; Hutterer, F. P.; Raich, M. K.; Scheel, C. H.; Bausch, A. R. Mechanical Plasticity of Collagen Directs Branch Elongation in Human Mammary Gland Organoids. Nat. Commun. 2021, 12, 2759 DOI: 10.1038/s41467-021-22988-242Mechanical plasticity of collagen directs branch elongation in human mammary gland organoidsBuchmann, B.; Engelbrecht, L. K.; Fernandez, P.; Hutterer, F. P.; Raich, M. K.; Scheel, C. H.; Bausch, A. R.Nature Communications (2021), 12 (1), 2759CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Epithelial branch elongation is a central developmental process during branching morphogenesis in diverse organs. This fundamental growth process into large arborized epithelial networks is accompanied by structural reorganization of the surrounding extracellular matrix (ECM), well beyond its mech. linear response regime. Here, we report that epithelial ductal elongation within human mammary organoid branches relies on the non-linear and plastic mech. response of the surrounding collagen. Specifically, we demonstrate that collective back-and-forth motion of cells within the branches generates tension that is strong enough to induce a plastic reorganization of the surrounding collagen network which results in the formation of mech. stable collagen cages. Such matrix encasing in turn directs further tension generation, branch outgrowth and plastic deformation of the matrix. The identified mech. tension equil. sets a framework to understand how mech. cues can direct ductal branch elongation.
- 43Hofer, M.; Lutolf, M. P. Engineering Organoids. Nat. Rev. Mater. 2021, 6, 402– 420, DOI: 10.1038/s41578-021-00279-y43Engineering organoidsHofer, Moritz; Lutolf, Matthias P.Nature Reviews Materials (2021), 6 (5), 402-420CODEN: NRMADL; ISSN:2058-8437. (Nature Portfolio)A review. Abstr.: Organoids are in vitro miniaturized and simplified model systems of organs that have gained enormous interest for modeling tissue development and disease, and for personalized medicine, drug screening and cell therapy. Despite considerable success in culturing physiol. relevant organoids, challenges remain to achieve real-life applications. In particular, the high variability of self-organizing growth and restricted exptl. and anal. access hamper the translatability of organoid systems. In this Review, we argue that many limitations of traditional organoid culture can be addressed by engineering approaches at all levels of organoid systems. We investigate cell surface and genetic engineering approaches, and discuss stem cell niche engineering based on the design of matrixes that allow spatiotemporal control of organoid growth and shape-guided morphogenesis. We examine how microfluidic approaches and lessons learnt from organs-on-a-chip enable the integration of mechano-physiol. parameters and increase accessibility of organoids to improve functional readouts. Applying engineering principles to organoids increases reproducibility and provides exptl. control, which will, ultimately, be required to enable clin. translation.
- 44Kopf, A.; Renkawitz, J.; Hauschild, R.; Girkontaite, I.; Tedford, K.; Merrin, J.; Thorn-Seshold, O.; Trauner, D.; Häcker, H.; Fischer, K.-D.; Kiermaier, E.; Sixt, M. Microtubules Control Cellular Shape and Coherence in Amoeboid Migrating Cells. J. Cell Biol. 2020, 219, e201907154 DOI: 10.1083/jcb.20190715444Microtubules control cellular shape and coherence in amoeboid migrating cellsKopf, Aglaja; Renkawitz, Joerg; Hauschild, Robert; Girkontaite, Irute; Tedford, Kerry; Merrin, Jack; Thorn-Seshold, Oliver; Trauner, Dirk; Haecker, Hans; Fischer, Klaus-Dieter; Kiermaier, Eva; Sixt, MichaelJournal of Cell Biology (2020), 219 (6), e201907154/1-e201907154/24CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Cells navigating through complex tissues face a fundamental challenge: while multiple protrusions explore different paths, the cell needs to avoid entanglement. How a cell surveys and then corrects its own shape is poorly understood. Here, we demonstrate that spatially distinct microtubule dynamics regulate amoeboid cell migration by locally promoting the retraction of protrusions. In migrating dendritic cells, local microtubule depolymn. within protrusions remote from the microtubule organizing center triggers actomyosin contractility controlled by RhoA and its exchange factor Lfc. Depletion of Lfc leads to aberrant myosin localization, thereby causing two effects that rate-limit locomotion: (1) impaired cell edge coordination during path finding and (2) defective adhesion resoln. Compromised shape control is particularly hindering in geometrically complex microenvironments, where it leads to entanglement and ultimately fragmentation of the cell body. We thus demonstrate that microtubules can act as a proprioceptive device: they sense cell shape and control actomyosin retraction to sustain cellular coherence.
- 45Vandestadt, C.; Vanwalleghem, G. C.; Castillo, H. A.; Li, M.; Schulze, K.; Khabooshan, M.; Don, E.; Anko, M.-L.; Scott, E. K.; Kaslin, J. Early Migration of Precursor Neurons Initiates Cellular and Functional Regeneration after Spinal Cord Injury in Zebrafish. bioRxiv 2019, 539940 DOI: 10.1101/539940There is no corresponding record for this reference.
- 46Cabernard, C.; Doe, C. Q. Apical/Basal Spindle Orientation Is Required for Neuroblast Homeostasis and Neuronal Differentiation in Drosophila. Dev. Cell 2009, 17, 134– 141, DOI: 10.1016/j.devcel.2009.06.00946Apical/basal spindle orientation is required for neuroblast homeostasis and neuronal differentiation in DrosophilaCabernard, Clemens; Doe, Chris Q.Developmental Cell (2009), 17 (1), 134-141CODEN: DCEEBE; ISSN:1534-5807. (Cell Press)Precise regulation of stem cell self-renewal/differentiation is essential for embryogenesis and tumor suppression. Drosophila neural progenitors (neuroblasts) align their spindle along an apical/basal polarity axis to generate a self-renewed apical neuroblast and a differentiating basal cell. Here, the authors genetically disrupt spindle orientation without altering cell polarity to test the role of spindle orientation in self-renewal/differentiation. The authors perform correlative live imaging of polarity markers and spindle orientation over multiple divisions within intact brains, followed by mol. marker anal. of cell fate. The authors find that spindle alignment orthogonal to apical/basal polarity always segregates apical determinants into both siblings, which invariably assume a neuroblast identity. Basal determinants can all be localized into one sibling without inducing neuronal differentiation, but overexpression of the basal determinant Prospero can deplete neuroblasts. The authors conclude that the ratio of apical/basal determinants specifies neuroblast/GMC identity, and that apical/basal spindle orientation is required for neuroblast homeostasis and neuronal differentiation.
- 47Karpova, N.; Bobinnec, Y.; Fouix, S.; Huitorel, P.; Debec, A. Jupiter, a New Drosophila Protein Associated with Microtubules. Cell Motil. 2006, 63, 301– 312, DOI: 10.1002/cm.2012447Jupiter, a new Drosophila protein associated with microtubulesKarpova, Nina; Bobinnec, Yves; Fouix, Sylvaine; Huitorel, Philippe; Debec, AlainCell Motility and the Cytoskeleton (2006), 63 (5), 301-312CODEN: CMCYEO; ISSN:0886-1544. (Wiley-Liss, Inc.)We describe a novel Drosophila protein Jupiter, which shares properties with several structural microtubule-assocd. proteins (MAPs) including TAU, MAP2, MAP4. Jupiter is a sol. unfolded mol. with the high net pos. charge, rich in glycine. It possesses 2 degenerated repeats around the sequence PPGG, sepd. by a serine-rich region. Jupiter assocs. with microtubules in vitro and, fused with the green fluorescent protein (GFP), is an excellent marker to follow microtubule dynamics in vivo. In a jupiter transgenic Drosophila strain generated by the protein-trap technique, Jupiter:GFP fusion protein localizes to the microtubule network through the cell cycle at the different stages of development. We found particularly high Jupiter:GFP concns. in the young embryo, larval nervous system, precursors of eye photoreceptors and adult ovary. Moreover, from jupiter:gfp embryos we have established 2 permanent cell lines presenting strongly fluorescent microtubules during the whole cell cycle. In these cells, the distribution of the Jupiter:GFP fusion protein reproduces microtubule behavior upon treatment by the drugs colchicine and taxol. The Jupiter cell lines and fly strain should be of wide interest for biologists interested in in vivo anal. of microtubule dynamics.
- 48Royou, A.; Sullivan, W.; Karess, R. Cortical Recruitment of Nonmuscle Myosin II in Early Syncytial Drosophila Embryos: Its Role in Nuclear Axial Expansion and Its Regulation by Cdc2 Activity. J. Cell Biol. 2002, 158, 127– 137, DOI: 10.1083/jcb.20020314848Cortical recruitment of nonmuscle myosin II in early syncytial Drosophila embryos: its role in nuclear axial expansion and its regulation by Cdc2 activityRoyou, Anne; Sullivan, William; Karess, RogerJournal of Cell Biology (2002), 158 (1), 127-137CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)The nuclei of early syncytial Drosophila embryos migrate dramatically toward the poles. The cellular mechanisms driving this process, called axial expansion, are unclear, but myosin II activity is required. By following regulatory myosin light chain (RLC)-green fluorescent protein dynamics in living embryos, we obsd. cycles of myosin recruitment to the cortex synchronized with mitotic cycles. Cortical myosin is first seen in a patch at the anterocentral part of the embryo at cycle 4. With each succeeding cycle, the patch expands poleward, dispersing at the beginning of each mitosis and reassembling at the end of telophase. Each cycle of actin and myosin recruitment is accompanied by a cortical contraction. The cortical myosin cycle does not require microtubules but correlates inversely with Cdc2/cyclin B (mitosis-promoting factor) activity. A mutant RLC lacking inhibitory phosphorylation sites was fully functional with no effect on the cortical myosin cycle, indicating that Cdc2 must be modulating myosin activity by some other mechanism. An inhibitor of Rho kinase blocks the cortical myosin recruitment cycles and provokes a concomitant failure of axial expansion. These studies suggest a model in which cycles of myosin-mediated contraction and relaxation, tightly linked to Cdc2 and Rho kinase activity, are directly responsible for the axial expansion of the syncytial nuclei.
- 49Rusan, N. M.; Peifer, M. A Role for a Novel Centrosome Cycle in Asymmetric Cell Division. J. Cell Biol. 2007, 177, 13– 20, DOI: 10.1083/jcb.20061214049A role for a novel centrosome cycle in asymmetric cell divisionRusan, Nasser M.; Peifer, MarkJournal of Cell Biology (2007), 177 (1), 13-20CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)Drosophila melanogaster central brain neuroblasts are excellent models for stem cell asym. division. Earlier work showed that their mitotic spindle orientation is established before spindle formation. We investigated the mechanism by which this occurs, revealing a novel centrosome cycle. In interphase, the 2 centrioles sep., but only 1 is active, retaining pericentriolar material and forming a dominant centrosome. This centrosome acts as a microtubule organizing center (MTOC) and remains stationary, forming 1 pole of the future spindle. The 2nd centriole is inactive and moves to the opposite side of the cell before being activated as a centrosome/MTOC. This is accompanied by asym. localization of Polo kinase, a key centrosome regulator. Disruption of centrosomes disrupts the high fidelity of asym. division. We propose a 2-step mechanism to ensure faithful spindle positioning: the novel centrosome cycle produces a single interphase MTOC, coarsely aligning the spindle, and spindle-cortex interactions refine this alignment.
- 50Cabernard, C.; Prehoda, K. E.; Doe, C. Q. A Spindle-Independent Cleavage Furrow Positioning Pathway. Nature 2010, 467, 91– 94, DOI: 10.1038/nature0933450A spindle-independent cleavage furrow positioning pathwayCabernard, Clemens; Prehoda, Kenneth E.; Doe, Chris Q.Nature (London, United Kingdom) (2010), 467 (7311), 91-94CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The mitotic spindle dets. the cleavage furrow site during metazoan cell division, but whether other mechanisms exist remains unknown. Here we identify a spindle-independent mechanism for cleavage furrow positioning in Drosophila neuroblasts. We show that early and late furrow proteins (Pavarotti, Anillin, and Myosin) are localized to the neuroblast basal cortex at anaphase onset by a Pins cortical polarity pathway, and can induce a basally displaced furrow even in the complete absence of a mitotic spindle. Rotation or displacement of the spindle results in 2 furrows: an early polarity-induced basal furrow and a later spindle-induced furrow. This spindle-independent cleavage furrow mechanism may be relevant to other highly polarized mitotic cells, such as mammalian neural progenitors.
- 51Connell, M.; Cabernard, C.; Ricketson, D.; Doe, C. Q.; Prehoda, K. E. Asymmetric Cortical Extension Shifts Cleavage Furrow Position in Drosophila Neuroblasts. Mol. Biol. Cell 2011, 22, 4220– 4226, DOI: 10.1091/mbc.e11-02-017351Asymmetric cortical extension shifts cleavage furrow position in Drosophila neuroblastsConnell, Marisa; Cabernard, Clemens; Ricketson, Derek; Doe, Chris Q.; Prehoda, Kenneth E.Molecular Biology of the Cell (2011), 22 (22), 4220-4226CODEN: MBCEEV; ISSN:1939-4586. (American Society for Cell Biology)The cytokinetic cleavage furrow is typically positioned sym. relative to the cortical cell boundaries, but it can also be asym. The mechanisms that control furrow site specification have been intensively studied, but how polar cortex movements influence ultimate furrow position remains poorly understood. We measured the position of the apical and the basal cortex in asym. dividing Drosophila neuroblasts and obsd. preferential displacement of the apical cortex that becomes the larger daughter cell during anaphase, effectively shifting the cleavage furrow toward the smaller daughter cell. Asym. cortical extension is correlated with the presence of cortical myosin II, which is polarized in neuroblasts. Loss of myosin II asymmetry by perturbing heterotrimeric G-protein signaling results in sym. extension and equal-sized daughter cells. We propose a model in which contraction-driven asym. polar extension of the neuroblast cortex during anaphase contributes to asym. furrow position and daughter cell size.
- 52Roberts, A.; Borisyuk, R.; Buhl, E.; Ferrario, A.; Koutsikou, S.; Li, W.-C.; Soffe, S. R. The Decision to Move: Response Times, Neuronal Circuits and Sensory Memory in a Simple Vertebrate. Proc. R. Soc. B 2019, 286, 20190297 DOI: 10.1098/rspb.2019.0297There is no corresponding record for this reference.
- 53Distel, M.; Hocking, J. C.; Volkmann, K.; Köster, R. W. The Centrosome Neither Persistently Leads Migration nor Determines the Site of Axonogenesis in Migrating Neurons in Vivo. J. Cell Biol. 2010, 191, 875– 890, DOI: 10.1083/jcb.20100415453The centrosome neither persistently leads migration nor determines the site of axonogenesis in migrating neurons in vivoDistel, Martin; Hocking, Jennifer C.; Volkmann, Katrin; Koester, Reinhard W.Journal of Cell Biology (2010), 191 (4), 875-890CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)The position of the centrosome ahead of the nucleus has been considered crucial for coordinating neuronal migration in most developmental situations. The proximity of the centrosome has also been correlated with the site of axonogenesis in certain differentiating neurons. Despite these pos. correlations, accumulating exptl. findings appear to negate a universal role of the centrosome in detg. where an axon forms, or in leading the migration of neurons. To further examine this controversy in an in vivo setting, we have generated cell type-specific multi-cistronic gene expression to monitor subcellular dynamics in the developing zebrafish cerebellum. We show that migration of rhombic lip-derived neurons is characterized by a centrosome that does not persistently lead the nucleus, but which is instead regularly overtaken by the nucleus. In addn., axonogenesis is initiated during the onset of neuronal migration and occurs independently of centrosome proximity. These in vivo data reveal a new temporal orchestration of organelle dynamics and provide important insights into the variation in intracellular processes during vertebrate brain differentiation.
- 54Distel, M.; Wullimann, M. F.; Köster, R. W. Optimized Gal4 Genetics for Permanent Gene Expression Mapping in Zebrafish. Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 13365– 13370, DOI: 10.1073/pnas.090306010654Optimized Gal4 genetics for permanent gene expression mapping in zebrafishDistel, Martin; Wullimann, Mario F.; Koster, Reinhard W.Proceedings of the National Academy of Sciences of the United States of America (2009), 106 (32), 13365-13370, S13365/1-S13365/9CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Combinatorial genetics for conditional transgene activation allows studying gene function with temporal and tissue specific control like the Gal4-UAS system, which has enabled sophisticated genetic studies in Drosophila. Recently this system was adapted for zebrafish and promising applications have been introduced. Here, we report a systematic optimization of zebrafish Gal4-UAS genetics by establishing an optimized Gal4-activator (KalTA4). We provide quant. data for KalTA4-mediated transgene activation in dependence of UAS copy nos. to allow for studying dosage effects of transgene expression. Employing a Tol2 transposon-mediated KalTA4 enhancer trap screen biased for central nervous system expression, we present a collection of self-reporting red fluorescent KalTA4 activator strains. These strains reliably transactivate UAS-dependent transgenes and can be rendered homozygous. Furthermore, we have characterized the transactivation kinetics of tissue-specific KalTA4 activation, which led to the development of a self-maintaining effector strain "Kaloop.". This strain relates transient KalTA4 expression during embryogenesis via a KalTA4-mediated autoregulatory mechanism to live adult structures. We demonstrate its use by showing that the secondary octaval nucleus in the adult hindbrain is likely derived from egr2b-expressing cells in rhombomere 5 during stages of early embryogenesis. These data demonstrate prolonged and maintained expression by Kalooping, a technique that can be used for permanent spatiotemporal genetic fate mapping and targeted transgene expression in zebrafish.
- 55Fortin, D. L.; Banghart, M. R.; Dunn, T. W.; Borges, K.; Wagenaar, D. A.; Gaudry, Q.; Karakossian, M. H.; Otis, T. S.; Kristan, W. B.; Trauner, D.; Kramer, R. H. Photochemical Control of Endogenous Ion Channels and Cellular Excitability. Nat. Methods 2008, 5, 331– 338, DOI: 10.1038/nmeth.118755Photochemical control of endogenous ion channels and cellular excitabilityFortin, Doris L.; Banghart, Matthew R.; Dunn, Timothy W.; Borges, Katharine; Wagenaar, Daniel A.; Gaudry, Quentin; Karakossian, Movses H.; Otis, Thomas S.; Kristan, William B.; Trauner, Dirk; Kramer, Richard H.Nature Methods (2008), 5 (4), 331-338CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Light-activated ion channels provide a precise and noninvasive optical means for controlling action potential firing, but the genes encoding these channels must first be delivered and expressed in target cells. Here we describe a method for bestowing light sensitivity onto endogenous ion channels that does not rely on exogenous gene expression. The method uses a synthetic photoisomerizable small mol., or photoswitchable affinity label (PAL), that specifically targets K+ channels. PALs contain a reactive electrophile, enabling covalent attachment of the photoswitch to naturally occurring nucleophiles in K+ channels. Ion flow through PAL-modified channels is turned on or off by photoisomerizing PAL with different wavelengths of light. We showed that PAL treatment confers light sensitivity onto endogenous K+ channels in isolated rat neurons and in intact neural structures from rat and leech, allowing rapid optical regulation of excitability without genetic modification.
- 56Laprell, L.; Tochitsky, I.; Kaur, K.; Manookin, M. B.; Stein, M.; Barber, D. M.; Schön, C.; Michalakis, S.; Biel, M.; Kramer, R. H.; Sumser, M. P.; Trauner, D.; Gelder, R. N. V. Photopharmacological Control of Bipolar Cells Restores Visual Function in Blind Mice. J. Clin. Invest. 2017, 127, 2598– 2611, DOI: 10.1172/JCI9215656Photopharmacological control of bipolar cells restores visual function in blind miceLaprell Laura; Stein Marco; Barber David M; Sumser Martin P; Trauner Dirk; Laprell Laura; Kaur Kuldeep; Manookin Michael B; Van Gelder Russell N; Tochitsky Ivan; Kramer Richard H; Schon Christian; Michalakis Stylianos; Biel Martin; Van Gelder Russell NThe Journal of clinical investigation (2017), 127 (7), 2598-2611 ISSN:.Photopharmacological control of neuronal activity using synthetic photochromic ligands, or photoswitches, is a promising approach for restoring visual function in patients suffering from degenerative retinal diseases. Azobenzene photoswitches, such as AAQ and DENAQ, have been shown to restore the responses of retinal ganglion cells to light in mouse models of retinal degeneration but do not recapitulate native retinal signal processing. Here, we describe diethylamino-azo-diethylamino (DAD), a third-generation photoswitch that is capable of restoring retinal ganglion cell light responses to blue or white light. In acute brain slices of murine layer 2/3 cortical neurons, we determined that the photoswitch quickly relaxes to its inactive form in the dark. DAD is not permanently charged, and the uncharged form enables the photoswitch to rapidly and effectively cross biological barriers and thereby access and photosensitize retinal neurons. Intravitreal injection of DAD restored retinal light responses and light-driven behavior to blind mice. Unlike DENAQ, DAD acts upstream of retinal ganglion cells, primarily conferring light sensitivity to bipolar cells. Moreover, DAD was capable of generating ON and OFF visual responses in the blind retina by utilizing intrinsic retinal circuitry, which may be advantageous for restoring visual function.
- 57Matera, C.; Gomila, A. M. J.; Camarero, N.; Libergoli, M.; Soler, C.; Gorostiza, P. Photoswitchable Antimetabolite for Targeted Photoactivated Chemotherapy. J. Am. Chem. Soc. 2018, 140, 15764– 15773, DOI: 10.1021/jacs.8b0824957Photoswitchable Antimetabolite for Targeted Photoactivated ChemotherapyMatera, Carlo; Gomila, Alexandre M. J.; Camarero, Nuria; Libergoli, Michela; Soler, Concepcio; Gorostiza, PauJournal of the American Chemical Society (2018), 140 (46), 15764-15773CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The efficacy and tolerability of systemically administered anticancer agents are limited by their off-target effects. Precise spatiotemporal control over their cytotoxic activity would allow improving chemotherapy treatments, and light-regulated drugs are well suited to this purpose. We have developed phototrexate, the first photoswitchable inhibitor of the human dihydrofolate reductase (DHFR), as a photochromic analog of methotrexate, a widely prescribed chemotherapeutic drug to treat cancer and psoriasis. Quantification of the light-regulated DHFR enzymic activity, cell proliferation, and in vivo effects in zebrafish show that phototrexate behaves as a potent antifolate in its photoactivated cis configuration and that it is nearly inactive in its dark-relaxed trans form. Thus, phototrexate constitutes a proof-of-concept to design light-regulated cytotoxic small mols. and a step forward to develop targeted anticancer photochemotherapies with localized efficacy and reduced adverse effects.
- 58Babii, O.; Afonin, S.; Schober, T.; Garmanchuk, L. V.; Ostapchenko, L. I.; Yurchenko, V.; Zozulya, S.; Tarasov, O.; Pishel, I.; Ulrich, A. S.; Komarov, I. V. Peptide Drugs for Photopharmacology: How Much of a Safety Advantage Can Be Gained by Photocontrol?. Future Drug Discovery 2020, 2, FDD28 DOI: 10.4155/fdd-2019-0033There is no corresponding record for this reference.
- 59Afonin, S.; Babii, O.; Reuter, A.; Middel, V.; Takamiya, M.; Strähle, U.; Komarov, I. V.; Ulrich, A. S. Light-Controllable Dithienylethene-Modified Cyclic Peptides: Photoswitching the in Vivo Toxicity in Zebrafish Embryos. Beilstein J. Org. Chem. 2020, 16, 39– 49, DOI: 10.3762/bjoc.16.659Light-controllable dithienylethene-modified cyclic peptides: photoswitching the in vivo toxicity in zebrafish embryosAfonin, Sergii; Babii, Oleg; Reuter, Aline; Middel, Volker; Takamiya, Masanari; Straehle, Uwe; Komarov, Igor V.; Ulrich, Anne S.Beilstein Journal of Organic Chemistry (2020), 16 (), 39-49CODEN: BJOCBH; ISSN:1860-5397. (Beilstein-Institut zur Foerderung der Chemischen Wissenschaften)This study evaluates the embryotoxicity of dithienylethene-modified peptides upon photoswitching, using 19 analogs based on the β-hairpin scaffold of the natural membranolytic peptide gramicidin S. We established an in vivo assay in two variations (with ex vivo and in situ photoisomerization), using larvae of the model organism Danio rerio, and detd. the toxicities of the peptides in terms of 50% LDs (LD50). This study allowed us to: (i) demonstrate the feasibility of evaluating peptide toxicity with D. rerio larvae at 3-4 days post fertilization, (ii) det. the phototherapeutic safety windows for all peptides, (iii) demonstrate photoswitching of the whole-body toxicity for the dithienylethene-modified peptides in vivo, (iv) re-analyze previous structure-toxicity relationship data, and (v) select promising candidates for potential clin. development.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/jacs.2c01020.
Temporally precise and cell-precise inhibitions of cellular MT polymerization dynamics by photoactivations of E-SBTubA4P at 405 nm (Movie S1) (MP4)
Temporally precise full-field-of-view inhibitions of cellular MT polymerization dynamics by photoactivations of E-SBTubA4P at 405 nm (Movie S2) (MP4)
No inhibition of cellular MT polymerization dynamics by illumination of E-SBTubA4P at 487 nm (Movie S3) (MP4)
SBTubA4P blocks branch development in primary human mammary gland organoids, light-dependently and with spatiotemporal precision (Movies S4−S6) (MP4, MP4, MP4)
No-compound control shows no photoinhibition of organoid branch development in both ROItarg and ROIctrl (Movie S7) (MP4)
Temporally precise depolymerization of the mitotic spindle in a prophase Drosophila neuroblast by photoactivation of E-SBTub2M (30 μM) at 405 nm (Movie S8) (MP4)
DMSO-only control to Movie S8 shows prophase Drosophila neuroblast undergoing normal mitosis after 405 nm illumination (Movie S9) (MP4)
Temporally precise depolymerization of the mitotic spindle in a prophase Drosophila neuroblast by photoactivation of E-SBTub2M (30 μM) at 405 nm (Movie S10) (MP4)
DMSO-only control to Movie S10 shows prophase Drosophila neuroblast undergoing normal mitosis after 405 nm illumination (Movie S11) (MP4)
Reaction of hatchling frog X. tropicalis embryos to mechanical stimulus depends on the temporally precise application of Z-SBTubA4 during prior development (Movie S12) (MP4)
Photoactivation of SBTubA4P (25 μM) in a live zebrafish embryo shows temporally reversible inhibitions of EB3 dynamics over several cycles (Movies S13 and S14) (MP4, MP4)
Inhibition of MT polymerization dynamics in zebrafish embryo when SBTubA4P (25 μM) is photoactivated (Movie S15) (MP4)
Photoactivation of SBTubA4P (25 μM) stops both EB3 dynamics and cell division in the developing zebrafish embryo (Movie S16) (MP4)
Chemical synthesis, photocharacterization, biological data, protein crystallization, and NMR spectra (PDF)
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.