ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img
RETURN TO ISSUEPREVBiological and Medic...Biological and Medical Applications of Materials and InterfacesNEXT

The Atomically Precise Gold/Captopril Nanocluster Au25(Capt)18 Gains Anticancer Activity by Inhibiting Mitochondrial Oxidative Phosphorylation

  • Sarita Roy Bhattacharya
    Sarita Roy Bhattacharya
    Department of Physical Chemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva, Switzerland
  • Kaushik Bhattacharya
    Kaushik Bhattacharya
    Department of Molecular and Cellular Biology, University of Geneva, Sciences III, Geneva 1205, Switzerland
  • Vanessa Joanne Xavier
    Vanessa Joanne Xavier
    Department of Molecular and Cellular Biology, University of Geneva, Sciences III, Geneva 1205, Switzerland
  • Abolfazl Ziarati
    Abolfazl Ziarati
    Department of Physical Chemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva, Switzerland
  • Didier Picard*
    Didier Picard
    Department of Molecular and Cellular Biology, University of Geneva, Sciences III, Geneva 1205, Switzerland
    *Email: [email protected]
  • , and 
  • Thomas Bürgi*
    Thomas Bürgi
    Department of Physical Chemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva, Switzerland
    *Email: [email protected]
Cite this: ACS Appl. Mater. Interfaces 2022, 14, 26, 29521–29536
Publication Date (Web):June 21, 2022
https://doi.org/10.1021/acsami.2c05054

Copyright © 2022 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0.
  • Open Access

Article Views

1627

Altmetric

-

Citations

LEARN ABOUT THESE METRICS
PDF (7 MB)
Supporting Info (1)»

Abstract

Atomically precise gold nanoclusters (AuNCs) are an emerging class of quantum-sized nanomaterials with well-defined molecular structures and unique biophysical properties, rendering them highly attractive for biological applications. We set out to study the impact of different ligand shells of atomically similar nanoclusters on cellular recognition and response. To understand the effects of atomically precise nanoclusters with identical composition on cells, we selected two different water-soluble gold nanoclusters protected with captopril (Capt) and glutathione (GSH): Au25(Capt)18 (CNC) and Au25(GSH)18 (GNC), respectively. We demonstrated that a change of the ligand of the cluster completely changes its biological functions. Whereas both nanoclusters are capable of internalization, only CNC exhibits remarkable cytotoxicity, more specifically on cancer cells. CNC shows enhanced cytotoxicity by inhibiting the OXPHOS of mitochondria, possibly by inhibiting the ATP synthase complex of the electron transport chain (ETC), and by initiating the leakage of electrons into the mitochondrial lumen. The resulting increase in both mitochondrial and total cellular ROS triggers cell death indicated by the appearance of cellular markers of apoptosis. Remarkably, this effect of nanoclusters is independent of any external light source excitation. Our findings point to the prevailing importance of the ligand shell for applications of atomically precise nanoclusters in biology and medicine.

This publication is licensed under

CC-BY-NC-ND 4.0.
  • cc licence
  • by licence
  • nc licence
  • nd licence

Introduction

ARTICLE SECTIONS
Jump To

The field of nanotechnology has always intrigued researchers to find and develop novel nanomaterials for applications in our life. Nanoparticles have some disadvantages regarding the precise control of their size and the number of ligands on their surface. In contrast, ligand-protected metal nanoclusters are atomically precise and exhibit enhanced photoluminescence and other appealing properties for applications such as intrinsic chirality and magnetism. (1,2) Precise control of the size and structure provides a handle to tune the electronic properties of these nanoscale objects, which makes them attractive for applications in various domains like sensing, optics, enantio-separation, photocatalysis, and the biomedical field. (3−5)
The nontoxic nature of gold, the size-dependent fluorescence, and other molecule-like features make AuNCs appealing candidates for biomedical applications. (4) Furthermore, the ligand shell of AuNCs can easily be modified to impart proper functionality and recognition properties. For example, the detection of hydrogen sulfide (H2S) and sensing of l-dopamine in cerebrospinal fluid are the examples of the ligand shell modification. (6,7) The fluorescence of AuNCs ranges from near infra-red (NIR) over the visible range to ultraviolet (UV), and this can be achieved by tuning the size of nanoclusters to have a high quantum yield and large Stokes shift. (8) One of the early applications of water-soluble AuNCs in imaging relied on d-penicillamine-capped AuNCs. (9−11) The internalization of these clusters on Hela cells was studied by confocal microscopy with two-photon excitation for bio-imaging. (12)
Another emerging application field for nanoclusters is nanomedicine with the use of hydrophilic ligands. 6-Mercaptohexanoic acid (MHA)-protected AuNCs were successfully shown to impart antimicrobial activities toward gram-positive and gram-negative bacteria. (13) 6-MHA is also utilized for the synthesis of Au25(MHA)18 as a delivery vehicle for melittin. (14) In addition to the Au25(SR)18 nanocluster system, Au18(MPA)14 (MPA, mercaptopropionic acid) is also internalized by both gram-positive and gram-negative bacteria. The Au18(MPA)14 nanocluster by itself has some bactericidal effect, but a nanocomposite of Au18(MPA)14 with a photoporphyrin (PpIX) within a chitosan polymer matrix shows an enhanced bactericidal effect toward both gram-positive and gram-negative bacteria upon white light irradiation. (15) Similarly, in combination with photothermal and photodynamic therapy, AuNCs were shown to possess enhanced anticancer properties. AuNCs are promising candidates for photodynamic therapy due to singlet oxygen generation upon two-photon excitation. One can also increase the local bioavailability of singlet oxygen by embedding the nanocluster into hydrogel nanoparticles, thus enhancing the photoexcitation properties of AuNCs. (16)
Most of the earlier applications of water-soluble nanoclusters use intrinsic fluorescence properties of nanoclusters for bio-imaging or use the nanoclusters in combination with single-photon or two-photon excitation. In the latter applications, the cells were killed only upon laser exposure. In the field of monolayer-protected metal clusters, it has been shown that minor changes in the composition, such as the number of metal atoms and/or the change of ligand, can have a drastic effect on the properties of the sample (every atom count). It is likely that in a highly complex environment such as in cells, only minor changes of the nanoclusters can have drastic consequences. Therefore, it is essential to use atomically precise clusters rather than a mixture of different clusters in such studies because, in the latter case, only one cluster in the mixture may be responsible for an observed effect.
Not much attention was paid to the atomic precision of AuNCs when evaluating their interaction with cells. None of the reported studies revealed the influence of the ligand shell on the surface of atomically precise nanoclusters for cellular recognition and responses. Therefore, a thorough study of the impact of atomically precise nanoclusters on cells is warranted. Here, we explored the impacts of different capping ligands of an atomically precise Au25(SR)18 (-SR; thiolate ligand) nanocluster on cellular physiology and metabolism. We have selected and synthesized two well-defined water-soluble AuNCs, CNC and GNC, where the ligands are Capt and GSH, respectively. The choice of these two ligands is typically based on their nontoxicity to cells and their ability to give rise to identical atomically precise (Au25(-SR)18) AuNCs. (17,18) Additionally, it does not involve complicated synthesis methods for both AuNCs. Our study on the cell–nanocluster interactions reveals that the physiological impacts of an atomically precise nanocluster remarkably depend on the capping ligands. We discover that the formation of a type of nanocluster with a specific ligand leads to a completely different chemical entity with a ″gain of function″ property compared to the bare ligand by itself since, in comparison to Capt, CNC emerges as cancer-cell-specific cytotoxic agent.

Experimental Section

ARTICLE SECTIONS
Jump To

Reagents and Resources

Details of all the reagents and resources are provided in Table S1 of the supplementary file.

Synthesis of Captopril (Capt) and Glutathione (GSH) Stabilized Nanoclusters

Synthesis of CNC and GNC was done according to previously reported methods. (19,20) For CNC synthesis, 0.2 mmol HAuCl4·3H2O (78.7 mg) was mixed with 0.23 mmol TOABr (126.8 mg) in 10 mL of methanol at room temperature for ∼25 min followed by the rapid addition of 1 mmol captopril (217.2 mg) in 5 mL of methanol. After ∼30 min, 2 mmol NaBH4 dissolved in ice-cold Milli Q water was injected rapidly at once. The reaction was continued for a minimum of 8 h until a reddish-brown solution was formed, indicative of Au25 nanocluster formation. Later, the solution was centrifuged to eliminate unreacted and insoluble gold-thiolate polymers. The supernatant collected was concentrated by rotatory evaporation followed by an ethanol precipitation step, and the pure CNC was extracted by dissolving the residue in a minimum volume of methanol. This purification step was repeated at least two more times to obtain pure CNC. Finally, the cluster was dried under a vacuum (Telstar Cryodo) to obtain a dry powder of pure CNC and stored at 4 °C for further experiments.
For GNC synthesis, in brief, an equal volume of 0.013 mmol HAuCl4·3H2O (5 mg) in Milli-Q water was mixed with a 0.052 mmol l-glutathione reduced (16 mg) solution in Milli-Q water. After the above mixture turned cloudy white, a solution of 0.132 mmol NaBH4 (5 mg) in 10 mL of ice-cold Milli-Q water was rapidly injected. Instantaneously, the color of the solution turned brown, indicative of Au25 nanocluster. The reaction mixture was kept in an oil bath at 60 °C for 2 h to complete the GNC synthesis. The solvent was then evaporated at low pressure and 50 °C by a rotatory evaporator to reduce the volume of the solution followed by the addition of methanol and centrifugation at high speed to precipitate the nanocluster. The reddish-brown precipitate of the nanocluster was dissolved in fresh Milli-Q water. The purification step was repeated once more to obtain the pure form of the GNC. The purified GNC solution was dried under a vacuum, and the nanocluster powder collected was preserved at 4 °C for further experiments.

Characterization of CNC and GNC

UV–vis absorption spectra were recorded on a Varian Cary 50 spectrophotometer using a quartz cuvette of 1 cm path length. All the spectra were recorded after dissolving the nanocluster solution in Milli-Q water. The spectra were normalized at 400 nm when required.
Mass spectra were recorded in positive mode using a Bruker Autoflex mass spectrometer equipped with a nitrogen laser. α-Cyano-4-hydroxycinnamic acid (α-CHCA) was used for CNC as the matrix with a 1:10, 1:100, and 1:1000 analyte/matrix ratio. α-CHCA (25 mM) was prepared in 50% acetonitrile/H2O with 0.1% trifluoroacetic acid. A volume of 2 μL of the analyte/matrix mixture was applied to the target and air-dried for the measurement. Electrospray high-resolution mass spectra (ESI HRMS) were measured on a QSTAR pulsar i (AB Sciex) using quadrupole and time-of-flight analyzers. The spectra were measured in the negative ion mode.
The fluorescence spectra were recorded at room temperature on a Fluorolog 3 (Horiba) spectrometer using a 10 × 2 mm quartz cuvette. The excitation wavelength was fixed at 514 nm (from a 450 W Xe lamp) in emission measurements. The emission was measured between 550 and 900 nm with identical excitation and emission slit widths of 5.0 nm.
Infrared spectra were measured on a Bruker Vertex 80V FTIR instrument. Briefly, the ligand (Capt/GSH) or cluster solution (CNC/GNC) was drop cast on CaF2 window and allowed to dry in air. IR spectra were recorded with a spectral resolution of 4 cm–1 in the range of 800–4000 cm–1.
A few drops of the samples (CNC and GNC) were placed on the carbon-coated copper grids (200 mesh), and the solvent was allowed to evaporate. The grids were then analyzed with a transmission electron microscope (TEM) at 120 or 160 kV (Tecnai G2, FEI, Eindhoven, Netherlands). Images were further analyzed with the ImageJ software to measure the diameter distribution and average values.
The hydrodynamic diameter and zeta potential of CNC and GNC were measured at room temperature using the dynamic light scattering (DLS) technique (Nano-ZS; Malvern Instruments, Malvern, United Kingdom). (20)

Cell Lines and Cell Culture

The following cells were used: HEK293T, human embryonic kidney cells; MDA-MB-231, triple-negative human breast carcinoma cells; HCT116, human colorectal carcinoma cells; A549, human lung epithelial carcinoma cells; HeLa, human cervical carcinoma cells; MCF7, estrogen receptor-positive human breast carcinoma cells; MAFs, mouse fibroblast derived from adult mouse ear tissue; HFs, human fibroblast cells derived from normal foreskin; HFs-hTERT, HFs immortalized by exogenous hTERT expression; HFs-hTERT + SV40 LT + Ras G12V, HFs-hTERT cells transformed into cancerous cells by overexpression of SV40 large T-antigen and mutant (G12V) H-Ras; (21) and RPE-1, retinal epithelial cells immortalized by hTERT expression. All these cells were maintained in Dulbecco’s modified Eagle medium (DMEM) supplemented with GlutaMAX, 10% fetal bovine serum (FBS), and penicillin/streptomycin (100 μg/mL) with 5% CO2 in a 37 °C humidified incubator. MCF10A (noncancerous epithelial) cells were grown in similar conditions in DMEM/F-12 supplemented with 5% heat-inactivated horse serum containing 10 ng/mL EGF, 5 μg/mL insulin, 1 μM dexamethasone, and penicillin/streptomycin (100 μg/mL).

Stability of Nanocluster in the Cell Culture Medium

CNC or GNC (300 μg/mL) was dissolved in pure Milli Q water or the cell culture medium, and UV–vis spectra were measured at different time (0–72 h) intervals.

Cell Viability Assay

The cell viability assay was performed by a CellTiter-Glo (CTG) luminescent assay (Promega) to directly quantify ATP generated by the cells, an indicator of metabolically active cells, as per the manufacturer’s instructions. Cells were seeded at a density of 10,000 cells/well (unless mentioned otherwise) at a volume of 100 μL/well in a complete medium in a 96-well plate, and after ∼12 h of seeding, the cells were treated with varied concentrations of CNC or GNC (0, 50, 100, 200, 300, 400, and 500 μg/mL) and their corresponding free ligands, i.e., Capt (0, 22.4, 44.4, 88.8, 133.2, 177.6, and 222 μg/mL) or GSH (0, 26.5, 53, 106, 159, 212, and 265 μg/mL) for 72 to 96 h (unless mentioned otherwise). Then 30 μL of the CellTiter-Glo reagent was added. After 15 min of incubation at room temperature (RT) in the dark, the luminescence was measured with a Cytation 3 (BioTek) cell imaging multimode microplate reader. The luminescence from the vehicle-treated cells was set to 100% viability. (22)

Flow Cytometry

For the flow-cytometric-based assay, a minimum of 10,000 cells was analyzed for each sample. We used FACS Gallios (Beckman Coulter) for acquiring the data, and the acquired data were analyzed with the FlowJo software package.

Measurement of Cell Death

Another way of measuring cytotoxicity is calculating the propidium iodide (PI) positive cells as membrane-altered dead cells by flow cytometry. For this, 4.0 × 105 cells/2 mL of medium in six-well plates were treated with CNC or GNC (0, 300, and 500 μg/mL) and their corresponding ligand: Capt (0, 133.2, and 222 μg/mL) or GSH (0, 159, and 265 μg/mL). After 72 h, cells were harvested by trypsinization, washed in phosphate-buffered saline (PBS), resuspended in 100 μL of PBS containing PI (2.5 μg/mL), and incubated for 15–20 min at 4 °C before flow cytometric acquisition was performed. (23)

Cell Cycle Analyses

Cells were harvested (as before) and fixed in 70% ice-cold ethanol, washed, incubated with 50 μg/mL PI for 15–20 min after treating with 100 μg/mL RNase A at RT for 5 min, and analyzed by flow cytometry. In some experiments, Hela cells (2 × 105) were pretreated with either a clathrin-mediated endocytosis (CME) inhibitor, Pitstop (40 μM), or a ROS scavenger, vitamin C (75 μM), for 1 h before the addition of CNC (0, 300, and 500 μg/mL), and the cell cycle assay was performed as described. Sub G0 cells (cells with nuclei containing less than a 2n DNA content) were used for the quantification of apoptotic cells. In experiments where G2/M phase arrests were measured, only diploid cell populations (nuclei containing 2n to 4n DNA content) were analyzed during data processing. (23)

Cell Counting by Trypan Blue Exclusion Assay

For this experiment, after treatments, viable cells were counted by trypan blue exclusion assay using a hemocytometer under the light microscope.

Scratch Assay

Highly invasive MDA-MB-231 cells were grown in complete DMEM as described before. After reaching 90% confluency, the cells were serum-starved for 24 h. Physical scratches were made with 10 μL pipette tips through the center of the wells, and cells were incubated with 222 μg/mL Capt and 500 μg/mL CNC. Images were captured by phase-contrast microscopy (described below) at different time intervals (0–24 h). (24)

Phase-Contrast Microscopy

Cellular morphology was analyzed using an inverted light microscope (Olympus CK2), and the images were captured with a Dino-lite camera using the DinoXcope software.

Confocal Microscopy

For confocal imaging experiments, HeLa or HEK293T cells (3 × 105) were seeded on a glass coverslip and transfected with plasmid pEGFP-C1 (for exogenous expression of intracellular enhanced green fluorescent protein (EGFP)). After ∼24 h post-transfection, cells were treated with CNC or GNC (500 μg/mL) and incubated for another 8 h in the incubator. Later, coverslips containing cells were washed with PBS and subsequently fixed with 4% formaldehyde for 20 min. After washing, the coverslips were mounted on a microscopic grade glass slide with Mowiol, and images were taken with Leica TCS SP5 confocal microscope with a 63× oil immersion objective. Cells labeled with a nanocluster (either CNC or GNC) were excited at 514 nm, and fluorescence was detected in the range of 640–788 nm. A sequential scanning setup was used to record the fluorescence from cells expressing EGFP proteins to avoid bleed-over of green fluorescence of EGFP into the red fluorescence (AuNC fluorescence region) channel. The excitation wavelength for measuring the fluorescence from EGFP was set at 488 nm, and the emission range at which fluorescence was measured was 501–540 nm. Digital images were acquired with 2.0–3.0× digital magnifications. Images were analyzed using the Image J-Fiji software. (25)
The study of endocytosis of CNC or GNC was carried out as described earlier. (26) HeLa cells were grown on coverslips for confocal imaging as described above at a low temperature (4 °C) in a complete medium containing 10 mM HEPES buffer for 1 h before the addition of CNC (300 μg/mL) and then incubated for another 8 h at 4 °C before slides were prepared for confocal imaging. The same set of experiments was performed at 37 °C, which served as a control.
For the endocytosis inhibition experiment by chemical inhibitors, slides were prepared for confocal imaging as described above. Briefly, HeLa cells were pretreated with Pitstop 2 (20 μM), Dynasore (80 μM), and cytochalasin D (5 μM) for 1 h, and subsequently, cells were treated with CNC (300 μg/mL) for an additional 8 h. Cells were processed as described above, and images were analyzed using the Image J-Fiji software. To quantitatively evaluate the uptake of nanoclusters into the cells, the imaging parameters were kept identical throughout the experiments. The relative amount of intracellular CNCs was estimated by measuring the intensity inside individual cells and is expressed as the mean fluorescence intensity calculated by selecting the area covered by cells in the collected images. This analysis was done using the Image J-Fiji software. A minimum of 50 cells was considered for calculating the mean fluorescence intensity. The software ImarisX64 9.5.1 was used for calculating Pearson’s correlation coefficient values.
For localization of CNC and GNC in mitochondria, we performed a confocal imaging assay. However, instead of the intracellular expression of EGFP, HeLa cells were transfected with mitochondrially targeted EGFP (mitoEGFP) and then treated with either CNC or GNC (300 μg/mL). (27) After 8 h of treatment, slides for confocal imaging were prepared, and data were analyzed with the Image J-Fiji software as described above.

Energy Metabolism Assays

The mitochondrial oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) measurements were monitored with the Seahorse Bioscience Extracellular Flux Analyzer (XFe24, Seahorse Agilent) in real time, and cells were pretreated with the nanoclusters and ligands. (27,28) In brief, Hela cells (6 × 104) were cultured overnight in custom XF24 microplates in the DMEM GlutaMax medium. Before measurements, the cells were washed with unbuffered DMEM and incubated without CO2 for 1 h to acclimatize them to the assay medium.
For real-time treatment of nanoclusters (both CNC and GNC, 500 μg/mL) and corresponding ligands (222 μg/mL Capt and 265 μg/mL GSH), basal OCR and ECAR were determined for the first few minutes (∼20 min). Then CNC/GNC or ligands were added to record the mitochondrial stress test profile. After ∼90 min, sequential injections of oligomycin (5 μM), carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP; 1 μM), and a mixture of rotenone and antimycin (Rot: 1 μM, AA: 1 μM) were performed, and data were recorded for ∼24 min after each injection.
In pretreatment experiments, Hela cells were pretreated with nanoclusters, i.e., CNC/GNC (500 μg/mL), for different times (0–4 h) depending on the experiment, and 1 h before measurements, the cells were incubated in an unbuffered medium as described before. Following incubation in an unbuffered medium, the sequential addition of different inhibitors of mitochondrial ETC [(oligomycin, FCCP, and AA + Rot) or (AA + Rot, FCCP, and oligomycin)] was performed, and the metabolic profiles were measured as described above.
For HFs-hTERT cells and their transformed variant, 1 × 104 cells/well were seeded into XF24 cell culture microplates and processed as described above to compare their OCR profiles without any pretreatments with nanoclusters.

Measurements of Intracellular ROS

Cells (2 × 105) were treated with CNC/GNC (500 μg/mL), Capt (222 μg/mL), and GSH (265 μg/mL) for different time intervals (0–4 h), and the intracellular H2O2 (ROS) was assessed flow cytometrically by incubating with H2DCFDA (20 μM) for 30 min at 37 °C. For some experiments, HeLa cells (2 × 105) were pretreated with the inhibitors of CME (Pitstop 2, 40 μM and Dynasore, 80 μM) for 1 h before the treatment with CNC (500 μg/mL), and ROS was measured as described. (29)

Measurements of Mitochondrial ROS

Cells (2 × 105) were treated with CNC (500 μg/mL) for different times, and intramitochondrial ROS was measured by incubating the CNC/GNC- and ligands-treated cells with 5 μM Mitosox red for 15 min at 37 °C. Before acquiring the samples with a flow cytometer, stained cells were fixed with 4% formaldehyde. (29)

Mitochondrial Membrane Potential Assay

HeLa cells (2 × 105) were treated with CNC (500 μg/mL) at different time intervals (0–36 h), and after harvesting, the cells were incubated with MitoTracker Deep Red (500 nM) by incubating for 15 min at 37 °C, and the corresponding fluorescence was measured and analyzed by FACS. (29)

Immunoblot Assay

HeLa cells (1.5 × 106) were treated with CNC and GNC (0, 200, 300, 400, and 500 μg/mL) for 48 h. Proteins were extracted from the cells in the lysis buffer (20 mM Tris–HCl pH 7.4, 2 mM EDTA, 150 mM NaCl, 1.2% sodium deoxycholate, 1.2% TritonX-100, protease inhibitor cocktail (PIC)) and quantified by the Bradford method. An equivalent amount of proteins (75 μg) were resolved by 15% SDS-PAGE and transferred onto a nitrocellulose membrane (GVS Life Science) with a wet blot transfer system (VWR). The membranes were blocked with 2–5% nonfat milk in TBS-Tween 20 (0.2%) and incubated with primary antibodies (see Table S1 for reagent details) with the following dilutions: anti-Caspase 9 (1:1000) and anti-α-tubulin (1:5000). Lysates (50 μg) of HFs-hTERT cells and their transformed variant were resolved by 10% SDS-PAGE, processed as described above, and anti-H-Ras (1:1000) and anti-SV40 LT (1:750) antibodies were used to detect the corresponding proteins. Then membranes were washed with TBS-Tween 20 (0.2%) and incubated with the appropriate HRP-conjugated secondary antibodies (1:10,000 dilutions). Immunoblots were developed using the WesternBright chemiluminescent substrate (Advansta). Images were recorded with a LI-COR Odyssey image recorder.

RNA Extraction and Quantitative RT-PCR (qRT-PCR)

The TRIzol reagent (Thermo Fisher Scientific) was added directly to the cell monolayer for total RNA extraction, and RNA extraction was performed according to the manufacturer’s instructions. RNA (1 μg) from each sample was digested with RNase-free DNase (Promega), and the digested RNA was then reverse-transcribed to cDNA using random primers and the GoScript reverse transcription kit (Promega). qRT-PCR was performed using the GoTaq master mix kit (Promega) with CXF96 or Connect thermocyclers (BioRad). GAPDH was used as a reference gene for these experiments.

General Data Analyses

Data processing and analyses were performed using GraphPad Prism (version 8).

Results

ARTICLE SECTIONS
Jump To

Characterization of Au25(Capt)18 and Au25(GSH)18 Nanoclusters

We selected two well-characterized AuNCs pairs, CNC and GNC, and their corresponding free ligands (Capt and GSH) to evaluate their biological effects on cellular functions (Figure 1a). The reported UV–vis absorption spectrum of Au25(SR)18 (R; Capt or GSH) has a prominent peak at 670 nm with two additional peaks at 450 and 400 nm, which is in agreement with previous findings and confirms the presence of CNC and GNC (Figure 1b). (19,20) The higher peak at 450 nm compared to the one at 400 nm in CNC and GNC confirms the anionic charge state for both nanoclusters. (19,20,30,31)

Figure 1

Figure 1. Characterization of CNC and GNC. (a) The top image shows the structure of the Au25(SR)18 nanocluster (color code: Au atoms, dark yellow; S atoms, green); the ligands (Capt or GSH) are omitted for clarity. The bottom image shows the structure of captopril (Capt) and glutathione (GSH) ligands used for the synthesis of CNC (Au25(Capt)18) and GNC (Au25(GSH)18). (b) UV–vis spectra of the synthesized CNC (red dotted line) and GNC (green squared line). The inset of the figure represents the color of the synthesized CNC and GNC in an aqueous solution. (c) The MALDI mass spectrum of CNC (Au25(Capt)18) recorded in positive mode with α-CHCA as the matrix. The peak marked with an asterisk (*) corresponds to the most important fragment Au21(Capt)14. (d) The ESI spectrum of GNC (Au25(GSH)18) gives characteristic peaks at m/z 809 and 1005 due to [Au(SG)2-H]−1 and [Au2(SG)2-H]−1, respectively. (e) The upper and lower panels, respectively, represent the morphology and size distributions of CNC and GNC obtained by TEM.

We further confirmed the synthesis of CNC by performing matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) using α-CHCA as a matrix. We found the molecular ion peak for CNC (Figure 1c, left panel) to be at ∼8822 m/z (theoretical: ∼8818 m/z) with the predominant fragment at ∼7166 m/z (for Au21(Capt)14, theoretical: ∼7165 m/z) corresponding to a loss of ∼1656 m/z, i.e., Au4(Capt)4 unit (theoretical: ∼1653 m/z). The detailed analysis of the fragment peaks was described in the literature. (20) In ESI HRMS, since the spray is orthogonal to ion extraction in the instrument, only certain fragments such as [Au(SG)2-H]−1 (m/z) 808 (experimentally obtained value m/z, 809) and [Au2(SG)2-H]−1 (m/z) 1005 are detectable, which are in good agreement with the earlier reported value. (32) The spectra showed only the features due to the monolayers and smaller Au clusters in all the samples. Both CNC and GNC possess a well-characterized fluorescence emission peak in the range of 550–900 nm when excited at 514 nm wavelength (Figure S1a). We exploited these fluorescence properties of CNC/GNC to monitor their cellular uptake by confocal microscopy as described below.
The infrared spectra of Capt/CNC (Figure S1b, left panel) and GSH/GNC (Figure S1b, right panel) show the absence or reduction of the S–H stretching vibrational mode (2550–2600 cm–1) in CNC and GNC, which suggests the cleavage of S–H bond and binding of Capt or GSH molecules onto the surfaces of gold nanocluster (CNC/GNC) through Au–S bonding. From Figure S1b, it is also evident that the stretching mode ν(C═O) of free carboxylic acid group (∼1735 cm–1) of the ligand (Capt/GSH) is diminished or disappeared due to the formation of nanoclusters (CNC/GNC). (20)
The TEM images show well-dispersed spherical CNC (Figure 1e, upper panel) and GNC (Figure 1e, lower panel) with comparable average sizes (diameters) of 1.71 ± 0.02 and 1.6 ± 0.02 nm, respectively. The mean hydrodynamic sizes of the CNC and GNC were also measured by the dynamic light scattering technique (Figure S1c). The mean diameters of CNC (1.656 ± 0.26 nm) and GNC (2.116 ± 0.31 nm) are well within the range of previously reported values and agree with TEM analyses. (20,33) The average zeta potentials of CNC and GNC in the aqueous solution were 27.1 ± 3.23 and −33.1 ± 2.62, respectively.
Next, we monitored the stability of the nanoclusters in the cell culture medium by a UV–vis spectral analysis and found that both nanoclusters were stable even after incubation of 72 h (Figure S1d). The two major peaks at 670 and 450 nm, characteristic of Au25(SR)18 nanoclusters, remain unchanged, suggesting the structural integrity of the nanoclusters even in the nutrient-rich medium. However, a time-dependent increase in a peak at ∼550 nm was observed for CNC and GNC (Figure S1d). We speculate that the increase in the peak height at 550 nm could be indicative of the formation of a protein-nanocluster corona. The formation of such protein corona has been reported, and it is proposed that this phenomenon helps the cellular entry of nanoparticles. (4,34) We speculate that also nanoclusters are prone to form such protein corona.

Differential Cellular Response to Au25(Capt)18 and Au25(GSH)18 Nanoclusters

To evaluate the impacts of AuNCs on cellular viability, HEK293T cells were treated with various concentrations of CNC, GNC, and the corresponding free ligands. To our surprise, we observed that only CNC is cytotoxic to HEK293T cells (Figure 2a and Figure S2a). In contrast, GNC and the free ligands Capt and GSH are completely nontoxic within the used concentration range, as determined by cellular viability and morphological analyses (Figure 2a and Figure S2a).

Figure 2

Figure 2. Cytotoxicity of nanoclusters and ligands on cancer cells. (a) Effects of CNC and GNC (0–500 μg/mL) and the corresponding ligands (Capt and GSH, respectively) on cell viability measured by the CTG assay in HEK293T cells after 72 h treatment (n = 3 biologically independent samples). (b) Only CNC induces a higher percentage of cell death in HEK293T cells as determined by the PI staining assay measured by flow cytometry, whereas GNC and the corresponding ligands (Capt and GSH) are neutral (n = 4 biologically independent samples). Membrane ruptured cells can only be visualized by PI staining and designated as the ″dead cell″ population. (c) The left panel represents the apoptotic cell percentage calculated by the flow-cytometry-based cell cycle assay. Only CNC induces significant apoptosis. The right panel shows that CNC arrests HEK293T cells in the G2/M phase of the cell cycle (n = 3 biologically independent samples). For the bar graphs, the data are represented as mean values ± SEM (standard error of the mean).

To further validate our results, we performed a flow-cytometry-based propidium iodide (PI) uptake assay to evaluate the cytotoxic effects of CNC. HEK293T cells treated with CNC showed PI-stained dead cells (Figure 2b). However, GNC, Capt, or GSH treatment showed only a marginal increase of dead cells compared to the control (Figure 2b). Next, we wanted to evaluate the impacts of CNC, GNC, and the free ligands on the cell cycle and apoptosis. HEK293T cells were treated with 300 and 500 μg/mL of CNC and GNC and the corresponding concentrations of the free ligands. As expected, only the CNC induced apoptosis as determined by the evaluation of the SubG0 (fragmented and <2n nucleus) cell population in the cell cycle analyses (Figure 2c, left panel). We also determined that CNC strikingly blocks the cell cycle progression of HEK293T cells in the G2/M phase (Figure 2c, right panel). However, GNC and the free ligands are totally nontoxic. These results were intriguing and showed that, specifically, CNC triggers apoptosis and a G2/M phase cell cycle arrest between two atomically precise identical AuNCs with different primarily nontoxic ligand shells.
All the above-mentioned cell-based cytotoxicity assays indicate a surprising and striking ″gain of function″ of CNC but not GNC. We speculate that when Capt ligands are clustered around an atomically precise gold core (Au25(SR)18 system), Capt changes its properties and becomes a new chemical species with additional functions. This phenomenon is ligand-dependent as the effect of GSH regarding cellular toxicity remains the same even in the form of a nanocluster.
To broaden the cytotoxicity spectrum of CNC, we performed cell viability assays and morphological assessments on different cancer cell lines upon treatment with CNC and Capt. CNC showed a dose-dependent inhibition of cell viability and cytotoxicity on MDA-MB-231 (triple-negative breast cancer), A549 (lung adenocarcinoma), HCT116 (colon carcinoma), and HeLa (cervical carcinoma) cells (Figure S2b,c).
The results shown above focus exclusively on cancer cells, and we wondered whether the cytotoxicity of CNC is limited to those. Therefore, we performed a similar cell viability assay on several noncancerous normal cell lines. Intriguingly, CNC is not as cytotoxic on normal cells as on cancer cells (Figure S2d). This result suggested that the cytotoxic ″gain of function″ of CNC is primarily specific to cancer cells.
We also checked the impact of CNC and its ligand on the migration of the highly invasive breast cancer cell line MDA-MB-231. CNC significantly reduces the migration of MDA-MB-231 cells into the scratched area, as evidenced by the scratch assay after 24 h of treatment (Figure S3). Free ligands behave like an inert component toward the cellular migratory property (Figure S3). Hence, CNC is not only cytotoxic but also inhibits cell migration and cell cycle progression.

Both CNC and GNC Internalize into Cells, and CNC Specifically Internalizes via Clathrin-Mediated Endocytosis (CME)

Next, we checked the intracellular uptake of these two AuNCs to understand the CNC-specific cytotoxicity. To study the intracellular uptake of CNC and GNC, Hela cells were treated with nanoclusters, and exploiting the red (pseudocolor) fluorescence (RF) properties of nanoclusters in confocal microscopy, comparative internalization was monitored qualitatively. (20,35) Colocalization of the RF and the green fluorescence (GF) from the exogenously expressed intracellular enhanced green fluorescence protein (EGFP; expressed from the pEGFP-C1 plasmid) served as an indicator of the internalization and intracellular localization of the nanoclusters. To confirm that the RF and the GF signals (specific to the nanocluster and EGFP, respectively) are only detected in the respective fluorescence channels, control sets with unstained cells, cells with only pEGFP-C1 transfection, and cells only treated with nanoclusters were analyzed (Figure 3a; first three rows of micrographs) and ruled out the possibility of ″bleed-over″ between the RF and GF signals. It must be noted that these transfection assays with pEGFP-C1 were transient transfections, and hence, it is expected that only a subset of the total cell population is transfected and ends up expressing EGFP.

Figure 3

Figure 3. Energy-dependent CME drives CNC internalization. (a) Dual-color confocal images of HeLa cells labeled with CNC (red color) and exogenously expressed intracellular EGFP (green color). The first row represents HeLa cells without transfection with the EGFP expression plasmid pEGFP-C1. The second and third rows represent cells stained with either CNC or cells transiently transfected with pEGFP-C1. The last row represents the transfected Hela cells with pEGFP-C1 and treated with CNC. The merging of the DIC (differential interference contrast) channel and the other two channels (red: CNC and green: EGFP channel) indicates the colocalization of CNC with EGFP as evident by the yellow color (n = at least 3 independent experiments). It must be noted that these transfection assays with pEGFP-C1 are transient transfections. Scale bar represents 10 μm. (b) The effect of temperature on CNC uptake by HeLa cells. Confocal microscopy of HeLa cells treated with CNC grown in two different temperatures: 4 and 37 °C (n = 2 independent biological samples). Scale bar represents 10 μm. (c) Bar graph representing the mean fluorescence intensity in arbitrary unit (a.u.) showing a comparison of CNC uptake at two different temperatures compared to control (n = a minimum of 50 cells was considered for calculating the mean fluorescence intensity, MFI). (d) Confocal images of HeLa cells pretreated with different endocytosis inhibitors and further incubated with CNC for 8 h to evaluate the uptake efficiency (n = 2 independent biological samples). Scale bar represents 10 μm. (e) Bar graph of mean fluorescence intensity (a.u.) relative to the control sample showing the effect of different inhibitors of the endocytic pathway on the uptake of CNC (n = a minimum of 50 cells was taken into account for calculating the MFI of a particular set). The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests.

AuNC treatment on EGFP-expressing cells revealed the colocalization of these two fluorescence signals in a subset of cells as indicated by the yellow color in the merged images (Figure 3a and Figures S4 and S5). We calculated the Pearson’s correlation coefficients (PCC) to confirm the intracellular localization of nanoclusters by correlating the fluorescence derived from nanoclusters and intracellular EGFP. The PCC values for both CNC (0.269 ± 0.11) and GNC (0.343 ± 0.14) for Hela cells (Figure 3a and Figure S5, respectively), and CNC (0.628 ± 0.14) and GNC (0.728 ± 0.13) for HEK293T cells (Figure S4) indicated that, irrespective of the ligand, both the nanoclusters are equally capable of internalizing into the cells. Therefore, we ruled out the possibility that a differential ability of internalization into the cells is the primary reason for the differential cytotoxic behaviors of CNC and GNC.
To further explore how these nanoclusters might enter into cells, we decided to focus on the cytotoxic nanocluster, CNC. To determine whether the energy-dependent endocytosis mechanism drives the internalization of CNC into cells, HeLa cells were treated with CNC at a low temperature (4 °C). In agreement with the literature, (36,37) we also found reduced intracellular localization of CNC at 4 °C compared to the normal growth conditions (37 °C, Figure 3b,c). This result indicated that an energy-dependent endocytic mechanism is responsible for the internalization of CNC into cells.
One of the most well-studied mechanisms of internalizing foreign particles into cells is clathrin-mediated endocytosis (CME). To evaluate whether the intracellular uptake of CNC is CME-driven, (38,39) HeLa cells were pretreated with the known CME inhibitors Pitstop 2 and Dynasore before incubating with CNC. Both CME inhibitors markedly blocked the internalization of CNC into cells (Figure 3d,e). These findings were further confirmed by the treatment with cytochalasin D (CytoD, Figure 3d,e), which inhibits CME by blocking actin polymerization. (40) Together, these results confirmed that the internalization of CNC is CME-driven.

CNC Inhibits Mitochondrial OXPHOS

We further investigated the impact of these nanoclusters on the major cellular metabolic processes oxidative phosphorylation (OXPHOS) and glycolysis. OXPHOS coupled with the electron transport chain (ETC) and aerobic glycolysis are the sources of cellular energy production in the form of adenosine triphosphate (ATP). (41,42) The ETC is a series of large protein complexes (complex I, II, III, IV, and V) and is responsible for the sequential electron transfer from the most electropositive end (complex I) to the most electronegative end (molecular O2) via electron carriers including ubiquinone and cytochrome c. Complex V, the ATP synthase, generates ATP by utilizing the energy from the proton gradient generated across the mitochondrial inner membrane during the sequential electron transfer process. (43,44)
Impacts of nanoclusters on OXPHOS and glycolysis were monitored by measuring the oxygen consumption rates (OCRs) and the extracellular acidification rates (ECARs), respectively, with a Seahorse XF analyzer. (27,28,41,45) Real-time monitoring of the OCR after injection of the nanoclusters on the cells revealed that CNC, but not GNC, affects the OCR with a sudden increase followed by a gradual decrease over time (Figure 4a). The sudden increase in OCR after CNC injection was intriguing (Figure 4a). We speculate that this may be a cellular feedback mechanism to overcome the initial inhibition of OCR after exposure to CNC. To evaluate the activity profile of the ETC complexes, we sequentially injected oligomycin A (complex V/ATP synthase inhibitor), carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), and a mixture of antimycin A and rotenone (complex III and I inhibitors, respectively) on the cells. The addition of oligomycin A reduced the OCR as expected in the control and GNC treatment (Figure 4a). However, CNC-treated cells were unresponsive to oligomycin A, and the OCR followed the same slope as before (Figure 4a). These results indicated that CNC might be a complex V inhibitor, and since complex V is possibly inhibited in the CNC-treated cells, oligomycin A cannot further inhibit the same substrate. The subsequent addition of FCCP increased the OCR, a measure of the respiratory reserve capacity, in control and the GNC-treated sets as expected (Figure 4a). On the apparent contrary, the addition of FCCP did not positively impact the OCR of CNC-treated cells (Figure 4a), which seemed to have lost their respiratory reserve capacity. The subsequent addition of antimycin A and rotenone entirely killed the respiratory capacity in all the experimental sets (Figure 4a). These results suggested that although complex V is potentially blocked by CNC treatment, complexes I and III are still active and, therefore, can be inhibited by their respective inhibitors.

Figure 4

Figure 4. CNC inhibits complex V of mitochondrial ETC. (a) OCR and (b) ECAR of HeLa cells were monitored using the Seahorse Bioscience Extra Cellular Flux Analyzer in real time. The first few minutes (∼20 min) represent the basal OCR or ECAR of HeLa cells followed by the injection of nanoclusters and then the sequential addition of the ATP synthase inhibitor (oligomycin 5 μM), FCCP (1 μM), and a mixture of rotenone (1 μM) and antimycin (1 μM) was performed. OCR is an indicator of mitochondrial respiration, and ECAR is predominately a measure of glycolytic flux. (c) OCR and (d) ECAR of HeLa cells pretreated with either CNC or GNC for different times (1, 2, and 4 h) and effect of mitochondrial inhibitors in the same order (oligomycin, FCCP, and rotenone + antimycin). (e) OCR and (f) ECAR of Hela cells pretreated with either CNC or GNC for different times (1, 2, and 4 h). Note that the order of addition of the inhibitors was changed. Here, pretreated cells were first treated with the antimycin A and rotenone mixture and subsequently with FCCP and oligomycin A. All data are reported as means ± SD (n = 3 experimental sets). (g) ATP production rates are calculated from the data presented in panel c for HeLa cells pretreated with CNC and GNC. The ATP production rate is calculated as (last OCR measurement before oligomycin injection) – (OCR rate measured after oligomycin injection). All data are presented as means ± SEM (n = minimum of 3 independent biological samples). The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests.

Any inhibition of the OCR can induce the glycolytic flux, which can be monitored by the ECAR. (46) We found that the inhibition of the OCR by the CNC treatment is complemented by an enhanced glycolytic flux (Figure 4b). To exclude the possibility that the addition of CNC can lower the pH of the solution by itself, the pH of the unbuffered medium was measured before and after the addition of CNC and GNC. None of these two nanoclusters affect the pH of the medium (data not shown). Therefore, the CNC-induced enhancement of ECAR is due to the inhibition of the OCR rather than the direct influence of the nanoclusters on pH. Additionally, in the identical experimental setups, the free ligands did not significantly impact the OCR and ECAR (Figure S6).
Metabolic flux measurements with CNC- and GNC-pretreated HeLa cells were performed to validate these findings further. Reminiscent of the real-time metabolic flux assays, we found that only the CNC-pretreated cells were nonresponsive to oligomycin A, which further confirmed the inhibition of complex V upon CNC treatment (Figure 4c). The enhanced basal ECAR of CNC-pretreated cells also supported the inhibition of OXPHOS (Figure 4d, first 20 min). Additionally, as expected, CNC-pretreated cells totally lost their respiratory reserve capacity (Figure 4c).
To find the impact of CNC on the activities of complexes I and III, CNC- and GNC-pretreated cells were first injected with the mixture of antimycin A and rotenone and, subsequently, with FCCP and oligomycin A. Irrespective of the pretreatment, the mixture of antimycin A and rotenone almost completely killed the OCR in all the experimental sets (Figure 4e). Interestingly, unlike CNC-pretreated cells, the mixture of antimycin A and rotenone augmented the ECAR of control and GNC-pretreated cells (Figure 4f). This result further confirmed that although complexes I and III remain active after the CNC pretreatment, glycolytic flux is already saturated due to the CNC-mediated inhibition of complex V.
The inhibition of complex V should reduce the production of ATP linked with mitochondria. Therefore, the rate of mitochondria-linked ATP production was measured. (27) As expected, only CNC but not GNC blocks the mitochondria-linked ATP production (Figure 4g). Together, these results demonstrated the following: (1) CNC blocks mitochondrial respiration, (2) CNC may be a novel inhibitor of mitochondrial ATP synthase (complex V), and (3) the ″gain of function″ of CNC compared to free captopril is due to a novel ″mito-poison″ activity. (29,47)

CNC Induces the Generation of Both Total Cellular and Mitochondrial ROS

Specific blockage in the ETC can initiate premature leakage of electrons from the mitochondrial respiratory complexes to the mitochondrial lumen, which induces the partial reduction of molecular oxygen to superoxide radical and further conversion into hydrogen peroxide. Hence, mitochondrial and overall intracellular ROS levels are augmented. (48,49) Since CNC inhibits complex V, we hypothesize that it should also increase intracellular ROS levels. The total intracellular ROS levels in HeLa cells were measured by 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA) staining upon treatment with both nanoclusters and their corresponding free ligands. In agreement with our metabolic efficiency analyses (Figure 4a–g), we found that only CNC induces the generation of ROS, supporting the idea of the ″gain of function″ upon nanocluster formation with Capt (Figure 5a). The augmentation of CNC-driven mitochondrial ROS was measured with MitoSox Red, a mitochondrial superoxide reactive dye (Figure 5b). Moreover, this augmentation of mitochondrial ROS was perfectly correlated with the depolarization of the mitochondrial membrane potential upon CNC treatment as determined by the staining of MitoTracker Deep Red, a mitochondrial-membrane-potential-dependent dye (Figure 5c).

Figure 5

Figure 5. CNC-induced mitochondrial respiratory dysfunction triggers intracellular ROS and apoptosis. (a) Intracellular ROS levels were measured by flow cytometry using the H2DCFDA staining assay in HeLa cells treated with CNC, GNC, and their corresponding ligands. For the line graphs, the data are represented as mean values ± SEM (n = 4 biologically independent samples). (b) Mitochondrial ROS levels were measured in HeLa cells after treatment with CNC (500 μg/mL) using MitoSox staining (n = 4 biologically independent samples). (c) Bar graph representing the percentage of mitochondrial membrane depolarized cells as a function of time (0, 3, 6, 12, 24, and 36 h) upon CNC (500 μg/mL) treatment (n = 4 biologically independent samples). (d) Confocal images of HeLa cells transiently transfected with mitochondrially targeted EGFP (mitoEGFP) and later treated with CNC to study the colocalization of CNC into mitochondria. The first row represents HeLa cells without transfection with mitoEGFP. The second and third rows represent cells either stained with CNC or transiently transfected with mitoEGFP. The last row represents HeLa cells transiently transfected with mitoEGFP and then treated with CNC to study the colocalization of CNC into mitochondria. The yellow color in the merged image confirms the localization of CNC in mitochondria. It has to be noted that these transfection assays with pEGFP-C1 are transient transfections. (e) Western blot analysis of Hela cells after treating them with different concentrations of CNC and GNC (200, 300, 400, and 500 μg/mL). The analysis indicates that CNC strikingly reduces the pro-form of caspase 9. α-Tubulin served as a loading control.

Both Nanoclusters Localize in Mitochondria, but Only CNC Triggers Caspase-Induced Apoptosis

Since CNC treatment leads to the inhibition of complex V, we speculated that CNC might also localize into mitochondria. To test this hypothesis, we expressed a mitochondrially targeted EGFP (mitoEGFP) in HeLa cells (27) and evaluated the colocalization of nanoclusters and mitoEGFP by confocal microscopy. Nevertheless, we confirmed no ″bleed-over″ between RF and GF signals derived from CNC/GNC and mitoEGFP, respectively (Figure 5d and Figure S7).
The dual-color confocal microscopy confirmed the localization of both CNC (Figure 5d, bottom row, and Figure S7a) and GNC in mitochondria (Figure S7b). It is important to note that due to the toxic nature of CNC, we reduced the incubation time with the nanoclusters to only 8 h for these colocalization studies. That might explain why there are relatively few and only weakly stained CNC- or GNC-positive cells in these confocal microscopy images. Additionally, the reduced fluorescence of nanoclusters in some cells may also be explained by the phenomenon of quenching of fluorescence of metal nanoclusters in the presence of various cellular substances. (6,50,51)
An increase in ROS leads to apoptosis via DNA damage, lipid peroxidation, mitochondrial dysfunction, and protein structure/function alterations. (52,53) Since CNC is cytotoxic and is also shown to inhibit the mitochondrial ETC, perhaps through complex V, we decided to examine the molecular signatures of apoptosis in CNC-treated cells.
We checked one of the major caspase proteins, caspase 9, associated with mitochondrial-dysfunction-mediated apoptosis. (54) We found that the expression level of pro-caspase 9 is strikingly reduced only in CNC treatment (Figure 5e). The downregulation of the pro-form of caspases in the process of cell death is a direct measure of their cleavage and formation of the enzymatically active form. (55) This result suggested that the mitochondrial-dysfunction-mediated induction of caspase 9 conceivably induces the caspase cascade activation only in the CNC treatment. Note that, although we observed the downregulation or reduction of the levels of pro-caspase tested here, we did not observe any strong signal for the activated caspase form. Since we performed this experiment after 48 h of treatment, we speculate that the labile nature of the active forms of caspases is the potential reason behind this observation. (56) Together, we concluded that CME-mediated internalized CNC localizes to mitochondria, potentially inhibits complex V of the mitochondrial ETC, triggers ROS generation, and subsequently initiates the mitochondria-mediated apoptotic response.

Inhibition of Internalization of CNC Protects Cells from Death

To test the hypothesis that impairing the internalization of CNC might result in reduced ROS generation, HeLa cells were pretreated with CME inhibitors and subsequently treated with CNC to check the total ROS generation by H2DCFDA staining. Both CME inhibitors, Pitstop 2 and Dynasore, reduce the CNC-induced ROS generation in HeLa cells (Figure 6a).

Figure 6

Figure 6. Restriction in the internalization of CNC protects cells from cell cycle arrest and reduced viability. (a) A comparison of intracellular ROS levels measured in Hela cells treated with CNC (500 μg/mL, red line) and HeLa cells pretreated with endocytic inhibitors Pitstop 2 (40 μM, blue line) and Dynasore (80 μM) to block the uptake of CNC inside the cells. (b) Cell cycle analysis revealed that pretreatment of Pitstop 2 (40 μM) on Hela cells partially protects from CNC (500 μg/mL)-induced G2/M phase cell cycle arrest. (c) A significantly higher number of viable cells were found in the case of HeLa cells pretreated with Pitstop 2 (40 μM) followed by CNC treatment compared to cells treated with only CNC. (d) Cell cycle analysis revealed that vitamin C (75 μM) pretreatment on Hela cells partially protects them from CNC-induced G2/M phase cell cycle arrest. (e) A significantly higher number of viable cells were found in the case of HeLa cells pretreated with vitamin C (75 μM) followed by CNC treatment compared to cells treated with only CNC. The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests.

Since CME inhibitors reduce the CNC-mediated ROS generation, we hypothesized that they might also protect cells from CNC-induced death. Hence, HeLa cells were treated with the CME-inhibitor Pitstop 2 and CNC, alone and in combination. Remarkably, Pitstop 2 inhibits the cytotoxic effect of CNC, as demonstrated by a reduced G2/M phase cell cycle arrest and by a relatively higher number of viable cells (Figure 6b,c). Additionally, vitamin C was used as a ROS-scavenging agent to check whether CNC-induced ROS promotes cytotoxicity. Remarkably, vitamin C significantly protects cells from CNC-mediated cytotoxicity (Figure 6d,e). Therefore, intracellular uptake and subsequent enhanced ROS generation are the molecular basis of the ″gain-of-function″ cytotoxicity of CNC.

CNC Targets Metabolically Active Cancer Cells

Although OXPHOS is required for both normal and cancer cells, we wondered how specifically CNC targets cancer cells with remarkably lower IC50 and higher therapeutic index (Figure 2a and Figure S2b,d). To elucidate this apparent difference between normal and cancer cells in response to CNC treatment, we generated oncogenically transformed cells by overexpressing the SV40 large T-antigen and mutant (G12V) H-Ras in the normal immortalized human fibroblasts (HFs-hTERT) (Figure S8a,b). We observed a negligible effect of hTERT-driven immortalization on OXPHOS and mitochondria-linked ATP production (Figure 7a,b).

Figure 7

Figure 7. Oncogenic transformation renders cells vulnerable to CNC-induced death. (a) Comparative OCR profiles of HFs (normal parental), HFs-hTERT (normal immortalized), and HFs-hTERT + SV40 + Ras G12V (oncogenically transformed) and effects of mitochondrial inhibitors (oligomycin, FCCP, and rotenone + antimycin) on them (n = 5 biologically independent samples). (b) Bar graphs represent the comparative basal OCR (left; first ∼20 min of respiration), rate of ATP production [middle; ATP production is calculated as (last OCR measurement before oligomycin injection) – (OCR rate measured after oligomycin injection)], and maximal respiratory rate [right; maximal respiratory rate is calculated as (last OCR measurement after oligomycin injection) – (OCR rate measured after FCCP injection)] of HFs (normal parental), HFs-hTERT (normal immortalized), and HFs-hTERT + SV40 + Ras G12V (cancer) cells. All these calculations were done on the experiment presented in panel a. (c) Measurements of total cellular ROS by H2DCFDA staining in the indicated cells in the presence and absence of CNC (n = 3 biologically independent samples). (d) Comparison of cell viability of the indicated cells after 5 days of treatment with CNC (n = 2 biologically independent samples). Note that, in this experiment, we seeded 6 × 103 cells. (e) Measurements of apoptotic cells (<2n nucleus in cell cycle analysis) after 48 h of treatment with CNC of the indicated cells (n = 2 biologically independent samples). The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests. *p < 0.05, **p < 0.01.

The oncogenic transformation of normal human fibroblasts remarkably increases OXPHOS by augmenting the basal OCR, maximal respiratory capacity, and mitochondria-linked ATP production (Figure 7a,b). These results are reminiscent of our earlier findings with a mouse fibroblast cell line showing that transformation by the H-Ras oncogene augments OXPHOS. (57) The CNC-driven inhibition of this hyperactivated OXPHOS specifically generates enhanced ROS in cancer cells compared to the immortalized normal cells (Figure 7c). This finding is further supported by the fact that CNC also generates enhanced ROS in breast cancer cells compared to normal breast epithelial cells (Figure S8c). These cancer cells are remarkably vulnerable to CNC-mediated cytotoxicity and apoptosis (Figure 7d,e), consistent with enhanced CNC-driven ROS generation in these cancer cells. We speculate that cancer cells with an ″OXPHOS-centric″ elevated energy metabolism (57,58) are selectively sensitized to CNC-mediated cytotoxicity.

Discussion

ARTICLE SECTIONS
Jump To

One of the significant advances emerging from this study is establishing the cellular internalization mechanism of gold-nanocluster systems. Like most cytotoxic nanoparticles described before, our nanoclusters are also internalized into cells by CME. (38,39,59−61) Nevertheless, we did examine the other pathway involved in the uptake of foreign particles by using genistein, an inhibitor of caveolin-mediated endocytosis, and found no disruption of the internalization of CNCs (data not shown). It should be noted that cultured cell lines are a heterogeneous population of cells with respect to cell cycle phases, and it is well-known that cell cycle phases strongly influence endocytic processes. (62,63) Therefore, it can be speculated that those cells with stronger RF from nanoclusters might have internalized more because they happened to be in a cell cycle phase more favorable for endocytosis. This further explains the differential intensities in the staining of cells by nanoclusters in the confocal analyses.
In this study, we also proved that nanoclusters by themselves can induce ROS via blocking OXPHOS but without being excited by an external energy source. Significantly, this ability depends on the ligand used to synthesize the nanocluster system. (64)
CME is an energy-dependent process that requires a continuous supply of ATP to uptake substances into cells. Any alteration in mitochondrial ETC and excessive leakage of protons lead to cytoplasmic acidification. (65) In our case, CNC inhibits the mitochondrial ETC, probably via blocking complex V, resulting in a drop in ATP production and further acidifying the cytoplasm of the cells (Figure 4). Thus, this could be a plausible reason for not showing enhanced differences in the cytotoxicity imparted by CNC beyond a concentration of 200 μg/mL in cancer cells (Figure 2 and Figure S2). We speculate that after the initial uptake of CNC by CME and inhibition of OXPHOS, further internalization of CNC into the cells is impaired due to this negative feedback mechanism.
Our finding that the cytotoxicity of CNC is selective for cancer cells relative to noncancerous cells correlates with the elevated ROS generation (Figures S8c and S2b,d). Normal cells are metabolically less active than cancer cells, and cancer cells have a higher requirement for receptor-ligand recycling via endocytosis. (58) We propose that CNC hijacks this mechanism in cancer cells and it preferentially accumulates in cancer cells, leading to the generation of elevated ROS levels, and shows more pronounced cytotoxicity. (66)
The production of cellular ATP is an essential parameter for maintaining healthy cellular physiology. (41,67) Most of the cellular ATP is produced by mitochondrial OXPHOS. (67) Minimal alterations or disruptions of OXPHOS significantly affect aging and are correlated with various disease conditions, including cancer and neurodegenerative disorders. (68,69) CNC but not GNC interferes explicitly with the mitochondrial ETC, leading to electron leakage and elevated ROS levels. Excessive generation of ROS can chemically damage cellular macromolecules, including DNA, proteins, and lipids, and induce cells to undergo apoptosis. (70,71)
We propose that CNC could be used as an anticancer therapeutic agent based on our discoveries. A key feature of CNC is that it shows selective cytotoxicity toward cancer cells, with an impressive therapeutic window, as evident from our cell-based assays (Figure 2 and Figure S2). However, for future applications, a systematic evaluation is warranted regarding the anticancer activity of CNC, specifically in cancer organoid systems and with cancer models in animals.
The cytotoxicity of water-soluble AuNCs is not only restricted to the Au25(SR)18 nanocluster system since Au18(MPA)14 nanoclusters have also been shown to exhibit some cytotoxic effect toward gram-positive and gram-negative bacteria and since this effect is enhanced in a nanocomposite system with photoporphyrin. (15) In the future, it will indeed be interesting to study the cytotoxic behavior of different magic-numbered AuNCs protected with different water-soluble ligands such as Capt or GSH. It will also be interesting to characterize the conformational states of the ligand Capt in the AuNC system, which may account for the altered topology or physical–chemical properties of Capt in the ligand shell of Au25(Capt)18 compared to free Capt. It has been shown before that similar thiolate ligands such as N-acetyl-l-cysteine and d-penicillamine adsorbed on gold interact in multiple ways with metal surfaces and clusters. (72,73) These studies indicate that, in addition to the thiol group, the carboxylic acid group can also interact with the metal.

Conclusions

ARTICLE SECTIONS
Jump To

The current study highlights the physicochemical interaction between atomically precise nanoclusters and cells. We demonstrate that different ligands of an atomically precise nanocluster with otherwise identical compositions might trigger different cellular responses. In our experimental model, the atomically precise Au25(SR)18 nanocluster gains cytotoxic property only in the presence of the Capt ligand via inhibition of mitochondrial respiration, whereas the GSH-protected nanocluster does not show such effects.
Our study suggests a plausible mechanism underlying the CNC-induced apoptosis. CNC inhibits the ATP synthase complex (complex V) of the mitochondrial ETC. Mitochondrial respiration inhibitors are toxic to cells by nature, and therefore, CNC might have the potential to be a new member of the broad family of ″mito-poisons″. However, its selectivity toward cancer cells prompts the idea that a specific respiratory or metabolic inhibitor for cancer treatment may be synthesized based on nanoclusters. An exciting outcome of the study is the complete change of target specificity and the biological effect of Capt upon nanocluster formation. Whereas the ligand Capt is used against hypertension, it becomes an anticancer molecule in the form of CNC based on our in vitro cell-based assays. This change of biological function may be related to the altered topology or physical–chemical properties of Capt in the ligand shell of Au25(Capt)18 compared to free Capt. Moreover, in the growing applied field of nanoclusters in biological systems, we need to know whether one can generalize the use of nanoclusters based on their physical properties or whether each nanocluster must be differentiated based on their specific impacts on biological systems. Our study indicates that nanoclusters need to be studied extensively, both physicochemically and biologically, for their compatibility before applying them for any physiological applications.

Supporting Information

ARTICLE SECTIONS
Jump To

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsami.2c05054.

  • Supporting figures including fluorescence emission spectra, FTIR spectra, size of the nanocluster, and stability in the cellular medium for CNC/GNC; cytotoxicity of nanocluster toward different cancer and normal cells; wound healing assay in the presence of CNC and Capt; internalization of CNC and GNC into cell and mitochondria; Seahorse assay with free ligand Capt and GSH; and measurement of intracellular ROS on normal and cancer cell lines (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.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Authors
  • Authors
    • Sarita Roy Bhattacharya - Department of Physical Chemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva, Switzerland
    • Kaushik Bhattacharya - Department of Molecular and Cellular Biology, University of Geneva, Sciences III, Geneva 1205, Switzerland
    • Vanessa Joanne Xavier - Department of Molecular and Cellular Biology, University of Geneva, Sciences III, Geneva 1205, Switzerland
    • Abolfazl Ziarati - Department of Physical Chemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva, SwitzerlandOrcidhttps://orcid.org/0000-0001-6686-4758
  • Author Contributions

    S.R.B. and K.B. contributed equally to this work. They conceived the study, designed and performed experiments, analyzed the data, prepared figures, and wrote the manuscript. V.J.X. generated immortalized and cancer-transformed HFs cells and performed the basic characterization and metabolic profiling assays with them. A.Z. contributed to the experiments with TEM. D.P. and T.B. conceived and supervised the study, contributed to designing experiments and analyzing the data, and wrote and edited the manuscript.

  • Funding

    T.B. and D.P. thank the Swiss National Science Foundation for generous support (grants 200020_172511 and 200020_192232 to T.B. and 31003A_172789/1 to D.P.). D.P. also acknowledges the funding from Canton de Genève and Fondation Medic.

  • Notes
    The authors declare no competing financial interest.

    Original data can be found at: https://doi.org/10.5281/zenodo.6594191.

Acknowledgments

ARTICLE SECTIONS
Jump To

We acknowledge the confocal facility of the Bioimaging Center and Jérôme Bosset for confocal measurements and the mass spectrometry core facility (MZ 2.0) of the University of Geneva. Lilia Bernasconi and Dina Hany are also kindly acknowledged for technical help in several experiments. We also thank Xianwei Wang for help with the IR studies. We thank Prof. Jean-Claude Martinou for mentoring VJX.

References

ARTICLE SECTIONS
Jump To

This article references 73 other publications.

  1. 1
    Knoppe, S.; Bürgi, T. Chirality in Thiolate-Protected Gold Clusters. Acc. Chem. Res. 2014, 47, 13181326,  DOI: 10.1021/ar400295d
  2. 2
    Jin, R. Atomically Precise Metal Nanoclusters: Stable Sizes and Optical Properties. Nanoscale 2015, 7, 15491565,  DOI: 10.1039/C4NR05794E
  3. 3
    Aires, A.; Lopez-Martinez, E.; Cortajarena, A. L. Sensors Based on Metal Nanoclusters Stabilized on Designed Proteins. Biosensors 2018, 8, 110,  DOI: 10.3390/bios8040110
  4. 4
    Porret, E.; Le Guével, X.; Coll, J.-L. Gold Nanoclusters for Biomedical Applications: Toward In Vivo Studies. J. Mater. Chem. B 2020, 8, 22162232,  DOI: 10.1039/C9TB02767J
  5. 5
    Du, Y.; Sheng, H.; Astruc, D.; Zhu, M. Atomically Precise Noble Metal Nanoclusters as Efficient Catalysts: A Bridge between Structure and Properties. Chem. Rev. 2020, 120, 526622,  DOI: 10.1021/acs.chemrev.8b00726
  6. 6
    Govindaraju, S.; Ankireddy, S. R.; Viswanath, B.; Kim, J.; Yun, K. Fluorescent Gold Nanoclusters for Selective Detection of Dopamine in Cerebrospinal fluid. Sci. Rep. 2017, 7, 4029840298,  DOI: 10.1038/srep40298
  7. 7
    Yu, Q.; Gao, P.; Zhang, K. Y.; Tong, X.; Yang, H.; Liu, S.; Du, J.; Zhao, Q.; Huang, W. Luminescent Gold Nanocluster-Based Sensing Platform for Accurate H2S Detection In Vitro and In Vivo with Improved Anti-Interference. Light: Sci. Appl. 2017, e17107  DOI: 10.1038/lsa.2017.107
  8. 8
    Jin, R.; Zeng, C.; Zhou, M.; Chen, Y. Atomically Precise Colloidal Metal Nanoclusters and Nanoparticles: Fundamentals and Opportunities. Chem. Rev. 2016, 116, 1034610413,  DOI: 10.1021/acs.chemrev.5b00703
  9. 9
    Yang, J.; Wang, F.; Yuan, H.; Zhang, L.; Jiang, Y.; Zhang, X.; Liu, C.; Chai, L.; Li, H.; Stenzel, M. Recent advances in ultra-small fluorescent Au nanoclusters toward oncological research. Nanoscale 2019, 11, 1796717980,  DOI: 10.1039/C9NR04301B
  10. 10
    Zheng, Y.; Lai, L.; Liu, W.; Jiang, H.; Wang, X. Recent advances in biomedical applications of fluorescent gold nanoclusters. Adv. Colloid Interface Sci. 2017, 242, 116,  DOI: 10.1016/j.cis.2017.02.005
  11. 11
    Zheng, Y.; Wu, J.; Jiang, H.; Wang, X. Gold nanoclusters for theranostic applications. Coord. Chem. Rev. 2021, 431, 213689  DOI: 10.1016/j.ccr.2020.213689
  12. 12
    Shang, L.; Dörlich, R. M.; Brandholt, S.; Schneider, R.; Trouillet, V.; Bruns, M.; Gerthsen, D.; Nienhaus, G. U. Facile Preparation of Water-Soluble Fluorescent Gold Nanoclusters for Cellular Imaging Applications. Nanoscale 2011, 3, 20092014,  DOI: 10.1039/c0nr00947d
  13. 13
    Zheng, K.; Setyawati, M. I.; Leong, D. T.; Xie, J. Antimicrobial Gold Nanoclusters. ACS Nano 2017, 11, 69046910,  DOI: 10.1021/acsnano.7b02035
  14. 14
    Qi, J.; Liu, Y.; Xu, H.; Xue, T.; Su, Y.; Lin, Z. Anti-Cancer Effect of Melittin-Au25(MHA)18 Complexes on Human Cervical Cancer HeLa Cells. J. Drug Delivery Sci. Technol. 2022, 68, 103078  DOI: 10.1016/j.jddst.2021.103078
  15. 15
    Srinivasulu, Y. G.; Mozhi, A.; Goswami, N.; Yao, Q.; Xie, J. Gold Nanocluster Based Nanocomposites for Combinatorial Antibacterial Therapy for Eradicating Biofilm Forming Pathogens. Mater. Chem. Front. 2022, 6, 689706,  DOI: 10.1039/D1QM00936B
  16. 16
    McLean, A.; Wang, R.; Huo, Y.; Cooke, A.; Hopkins, T.; Potter, N.; Li, Q.; Isaac, J.; Haidar, J.; Jin, R.; Kopelman, R. Synthesis and Optical Properties of Two-Photon-Absorbing Au25(Captopril)18-Embedded Polyacrylamide Nanoparticles for Cancer Therapy. ACS Appl. Nano Mater. 2020, 3, 14201430,  DOI: 10.1021/acsanm.9b02272
  17. 17
    Carlos-Escalante, J. A.; de Jesús-Sánchez, M.; Rivas-Castro, A.; Pichardo-Rojas, P. S.; Arce, C.; Wegman-Ostrosky, T. The Use of Antihypertensive Drugs as Coadjuvant Therapy in Cancer. Front. Oncol. 2021, 11, 660943  DOI: 10.3389/fonc.2021.660943
  18. 18
    Kennedy, L.; Sandhu, J. K.; Harper, M.-E.; Cuperlovic-Culf, M. Role of Glutathione in Cancer: From Mechanisms to Therapies. Biomolecules 2020, 10, 1429,  DOI: 10.3390/biom10101429
  19. 19
    Katla, S. K.; Zhang, J.; Castro, E.; Bernal, R. A.; Li, X. Atomically Precise Au25(SG)18 Nanoclusters: Rapid Single-Step Synthesis and Application in Photothermal Therapy. ACS Appl. Mater. Interfaces 2018, 10, 7582,  DOI: 10.1021/acsami.7b12614
  20. 20
    Roy Bhattacharya, S.; Bürgi, T. Amplified Vibrational Circular Dichroism as a Manifestation of the Interaction Between a Water Soluble Gold Nanocluster and Cobalt Salt. Nanoscale 2019, 11, 2322623233,  DOI: 10.1039/C9NR07534H
  21. 21
    Hahn, W. C.; Counter, C. M.; Lundberg, A. S.; Beijersbergen, R. L.; Brooks, M. W.; Weinberg, R. A. Creation of Human Tumour Cells with Defined Genetic Elements. Nature 1999, 400, 464468,  DOI: 10.1038/22780
  22. 22
    Jung, K.-J.; Dasgupta, A.; Huang, K.; Jeong, S.-J.; Pise-Masison, C.; Gurova, K. V.; Brady, J. N. Small-Molecule Inhibitor Which Reactivates p53 in Human T-Cell Leukemia Virus Type 1-Transformed Cells. J. Virol. 2008, 82, 85378547,  DOI: 10.1128/JVI.00690-08
  23. 23
    Bhattacharya, K.; Weidenauer, L.; Luengo, T. M.; Pieters, E. C.; Echeverría, P. C.; Bernasconi, L.; Wider, D.; Sadian, Y.; Koopman, M. B.; Villemin, M.; Bauer, C.; Rüdiger, S. G. D.; Quadroni, M.; Picard, D. The Hsp70-Hsp90 Co-Chaperone Hop/Stip1 Shifts the Proteostatic Balance from Folding Towards Degradation. Nat. Commun. 2020, 11, 5975,  DOI: 10.1038/s41467-020-19783-w
  24. 24
    Mondal, S.; Bhattacharya, K.; Mandal, C. Nutritional Stress Reprograms Dedifferention in Glioblastoma Multiforme Driven by PTEN/Wnt/Hedgehog Axis: a Stochastic Model of Cancer Stem Cells. Cell Death Discovery 2018, 4, 110,  DOI: 10.1038/s41420-018-0126-6
  25. 25
    Schindelin, J.; Arganda-Carreras, I.; Frise, E.; Kaynig, V.; Longair, M.; Pietzsch, T.; Preibisch, S.; Rueden, C.; Saalfeld, S.; Schmid, B.; Tinevez, J.-Y.; White, D. J.; Hartenstein, V.; Eliceiri, K.; Tomancak, P.; Cardona, A. Fiji: an Open-Source Platform for Biological-Image Analysis. Nat. Methods 2012, 9, 676682,  DOI: 10.1038/nmeth.2019
  26. 26
    Xie, J.; Pan, X.; Wang, M.; Ma, J.; Fei, Y.; Wang, P.-N.; Mi, L. The Role of Surface Modification for TiO2 Nanoparticles in Cancer Cells. Colloids Surf., B 2016, 143, 148155,  DOI: 10.1016/j.colsurfb.2016.03.029
  27. 27
    Joshi, A.; Dai, L.; Liu, Y.; Lee, J.; Ghahhari, N. M.; Segala, G.; Beebe, K.; Jenkins, L. M.; Lyons, G. C.; Bernasconi, L.; Tsai, F. T. F.; Agard, D. A.; Neckers, L.; Picard, D. The Mitochondrial HSP90 Paralog TRAP1 Forms an OXPHOS-Regulated Tetramer and is Involved in Mitochondrial Metabolic Homeostasis. BMC Biol. 2020, 18, 10,  DOI: 10.1186/s12915-020-0740-7
  28. 28
    Yoshida, S.; Tsutsumi, S.; Muhlebach, G.; Sourbier, C.; Lee, M.-J.; Lee, S.; Vartholomaiou, E.; Tatokoro, M.; Beebe, K.; Miyajima, N.; Mohney, R. P.; Chen, Y.; Hasumi, H.; Xu, W.; Fukushima, H.; Nakamura, K.; Koga, F.; Kihara, K.; Trepel, J.; Picard, D.; Neckers, L. Molecular Chaperone TRAP1 Regulates a Metabolic Switch Between Mitochondrial Respiration and Aerobic Glycolysis. Proc. Natl. Acad. Sci. U. S. A. 2013, 110, E1604E1612,  DOI: 10.1073/pnas.1220659110
  29. 29
    Bhattacharya, K.; Bag, A. K.; Tripathi, R.; Samanta, S. K.; Pal, B. C.; Shaha, C.; Mandal, C. Mahanine, a Novel Mitochondrial Complex-III Inhibitor Induces G0/G1 Arrest Through Redox Alteration-Mediated DNA Damage Response and Regresses Glioblastoma Multiforme. Am. J. Cancer Res. 2014, 4, 629647
  30. 30
    Tofanelli, M. A.; Ackerson, C. J. Superatom Electron Configuration Predicts Thermal Stability of Au25(SR)18 Nanoclusters. J. Am. Chem. Soc. 2012, 134, 1693716940,  DOI: 10.1021/ja3072644
  31. 31
    Tofanelli, M. A.; Salorinne, K.; Ni, T. W.; Malola, S.; Newell, B.; Phillips, B.; Häkkinen, H.; Ackerson, C. J. Jahn–Teller Effects in Au25(SR)18. Chem. Sci. 2016, 7, 18821890,  DOI: 10.1039/C5SC02134K
  32. 32
    Shibu, E. S.; Muhammed, M. A. H.; Tsukuda, T.; Pradeep, T. J. Phys. Chem. C 2008, 112, 1216812176,  DOI: 10.1021/jp800508d
  33. 33
    Tay, C. Y.; Yu, Y.; Setyawati, M. I.; Xie, J.; Leong, D. T. Presentation Matters: Identity of Gold Nanocluster Capping Agent Governs Intracellular Uptake and Cell Metabolism. Nano Res. 2014, 7, 805815,  DOI: 10.1007/s12274-014-0441-z
  34. 34
    Sabuncu, A. C.; Grubbs, J.; Qian, S.; Abdel-Fattah, T. M.; Stacey, M. W.; Beskok, A. Probing Nanoparticle Interactions in Cell Culture Media. Colloids Surf., B 2012, 95, 96102,  DOI: 10.1016/j.colsurfb.2012.02.022
  35. 35
    Wang, M.; Wu, Z.; Yang, J.; Wang, G.; Wang, H.; Cai, W. Au25(SG)18 as a Fluorescent Iodide Sensor. Nanoscale 2012, 4, 40874090,  DOI: 10.1039/c2nr30169e
  36. 36
    Dalal, C.; Saha, A.; Jana, N. R. Nanoparticle Multivalency Directed Shifting of Cellular Uptake Mechanism. J. Phys. Chem. C 2016, 120, 67786786,  DOI: 10.1021/acs.jpcc.5b11059
  37. 37
    Bera, K.; Maiti, S.; Maity, M.; Mandal, C.; Maiti, N. C. Porphyrin–Gold Nanomaterial for Efficient Drug Delivery to Cancerous Cells. ACS Omega 2018, 3, 46024619,  DOI: 10.1021/acsomega.8b00419
  38. 38
    Von Kleist, L.; Stahlschmidt, W.; Bulut, H.; Gromova, K.; Puchkov, D.; Robertson, M. J.; MacGregor, K. A.; Tomilin, N.; Pechstein, A.; Chau, N.; Chircop, M.; Sakoff, J.; Peter von Kries, J.; Saenger, W.; Kräusslich, H.-G.; Shupliakov, O.; Robinson, P. J.; McCluskey, A.; Haucke, V. Role of the Clathrin Terminal Domain in Regulating Coated Pit Dynamics Revealed by Small Molecule Inhibition. Cell 2011, 146, 841,  DOI: 10.1016/j.cell.2011.08.014
  39. 39
    Preta, G.; Cronin, J. G.; Sheldon, I. M. Dynasore - Not Just a Dynamin Inhibitor. Cell Commun. Signaling 2015, 13, 24,  DOI: 10.1186/s12964-015-0102-1
  40. 40
    Kuhn, D. A.; Vanhecke, D.; Michen, B.; Blank, F.; Gehr, P.; Petri-Fink, A.; Rothen-Rutishauser, B. Different Endocytotic Uptake Mechanisms for Nanoparticles in Epithelial Cells and Macrophages. Beilstein J. Nanotechnol. 2014, 5, 16251636,  DOI: 10.3762/bjnano.5.174
  41. 41
    Nolfi-Donegan, D.; Braganza, A.; Shiva, S. Mitochondrial Electron Transport Chain: Oxidative Phosphorylation, Oxidant Production, and Methods of Measurement. Redox Biol. 2020, 37, 101674  DOI: 10.1016/j.redox.2020.101674
  42. 42
    Zhao, Y.; Butler, E. B.; Tan, M. Targeting Cellular Metabolism to Improve Cancer Therapeutics. Cell Death Discovery 2013, 4, e532  DOI: 10.1038/cddis.2013.60
  43. 43
    Guo, R.; Gu, J.; Zong, S.; Wu, M.; Yang, M. Structure and Mechanism of Mitochondrial Electron Transport Chain. Biomed. J. 2018, 41, 920,  DOI: 10.1016/j.bj.2017.12.001
  44. 44
    Zhao, R. Z.; Jiang, S.; Zhang, L.; Yu, Z.-B. Mitochondrial Electron Transport Chain, ROS Generation and Uncoupling (Review). Int. J. Mol. Med. 2019, 44, 315,  DOI: 10.3892/ijmm.2019.4188
  45. 45
    Yépez, V. A.; Kremer, L. S.; Iuso, A.; Gusic, M.; Kopajtich, R.; Koňaříková, E.; Nadel, A.; Wachutka, L.; Prokisch, H.; Gagneur, J. OCR-Stats: Robust Estimation and Statistical Testing of Mitochondrial Respiration Activities Using Seahorse XF Analyzer. PLoS One 2018, 13, e0199938  DOI: 10.1371/journal.pone.0199938
  46. 46
    Pike Winer, L. S.; Wu, M. Rapid Analysis of Glycolytic and Oxidative Substrate Flux of Cancer Cells in a Microplate. PLoS One 2014, 9, e109916  DOI: 10.1371/journal.pone.0109916
  47. 47
    Pelicano, H.; Feng, L.; Zhou, Y.; Carew, J. S.; Hileman, E. O.; Plunkett, W.; Keating, M. J.; Huang, P. Inhibition of Mitochondrial Respiration: a Novel Strategy to Enhance Drug-Induced Apoptosis in Human Leukemia Cells by a Reactive Oxygen Species-Mediated Mechanism. J. Biol. Chem. 2003, 278, 3783237839,  DOI: 10.1074/jbc.M301546200
  48. 48
    Kausar, S.; Wang, F.; Cui, H. The Role of Mitochondria in Reactive Oxygen Species Generation and Its Implications for Neurodegenerative Diseases. Cell 2018, 7, 274,  DOI: 10.3390/cells7120274
  49. 49
    Zorov, D. B.; Juhaszova, M.; Sollott, S. J. Mitochondrial Reactive Oxygen Species (ROS) and ROS-Induced ROS Release. Physiol. Rev. 2014, 94, 909950,  DOI: 10.1152/physrev.00026.2013
  50. 50
    Hu, L.; Han, S.; Parveen, S.; Yuan, Y.; Zhang, L.; Xu, G. Highly Sensitive Fluorescent Detection of Trypsin Based on BSA-Stabilized Gold Nanoclusters. Biosens. Bioelectron. 2012, 32, 297299,  DOI: 10.1016/j.bios.2011.12.007
  51. 51
    McDonagh, B. H.; Singh, G.; Bandyopadhyay, S.; Lystvet, S. M.; Ryan, J. A.; Volden, S.; Kim, E.; Sandvig, I.; Sandvig, A.; Glomm, W. R. Controlling the Self-Assembly and Optical Properties of Gold Nanoclusters and Gold Nanoparticles Biomineralized with Bovine Serum Albumin. RSC Adv. 2015, 5, 101101101109,  DOI: 10.1039/C5RA23423A
  52. 52
    Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in Cancer Therapy: The Bright Side of the Moon. Exp. Mol. Med. 2020, 52, 192203,  DOI: 10.1038/s12276-020-0384-2
  53. 53
    Yang, H.; Villani, R. M.; Wang, H.; Simpson, M. J.; Roberts, M. S.; Tang, M.; Liang, X. The Role of Cellular Reactive Oxygen Species in Cancer Chemotherapy. J. Exp. Clin. Cancer Res. 2018, 37, 266,  DOI: 10.1186/s13046-018-0909-x
  54. 54
    McIlwain, D. R.; Berger, T.; Mak, T. W. Caspase Functions in Cell Death and Disease. Cold Spring Harbor Perspect. Biol. 2013, 5, a008656  DOI: 10.1101/cshperspect.a008656
  55. 55
    Roy, S.; Bhattacharya, K.; Mandal, C.; Dasgupta, A. K. Cellular Response to Chirality and Amplified Chirality. J. Mater. Chem. B 2013, 1, 66346643,  DOI: 10.1039/c3tb21322f
  56. 56
    Bhattacharya, K.; Samanta, S. K.; Tripathi, R.; Mallick, A.; Chandra, S.; Pal, B. C.; Shaha, C.; Mandal, C. Apoptotic Effects of Mahanine on Human Leukemic Cells are Mediated Through Crosstalk Between Apo-1/Fas Signaling and the Bid Protein and via Mitochondrial Pathways. Biochem. Pharmacol. 2010, 79, 361372,  DOI: 10.1016/j.bcp.2009.09.007
  57. 57
    Echeverria, P. C.; Bhattacharya, K.; Joshi, A.; Wang, T.; Picard, D. The Sensitivity to Hsp90 Inhibitors of Both Normal and Oncogenically Transformed Cells is Determined by the Equilibrium Between Cellular Quiescence and Activity. PLoS One 2019, 14, e0208287  DOI: 10.1371/journal.pone.0208287
  58. 58
    Lanzetti, L.; Di Fiore, P. P. Endocytosis and Cancer: an ‘Insider’ Network with Dangerous Liaisons. Traffic 2008, 9, 20112021,  DOI: 10.1111/j.1600-0854.2008.00816.x
  59. 59
    Faklaris, O.; Joshi, V.; Irinopoulou, T.; Tauc, P.; Sennour, M.; Girard, H.; Gesset, C.; Arnault, J.-C.; Thorel, A.; Boudou, J.-P.; Curmi, P. A.; Treussart, F. Photoluminescent Diamond Nanoparticles for Cell Labeling: Study of the Uptake Mechanism in Mammalian Cells. ACS Nano 2009, 3, 39553962,  DOI: 10.1021/nn901014j
  60. 60
    Jiang, X.; Röcker, C.; Hafner, M.; Brandholt, S.; Dörlich, R. M.; Nienhaus, G. U. Endo- and Exocytosis of Zwitterionic Quantum Dot Nanoparticles by Live HeLa Cells. ACS Nano 2010, 4, 67876797,  DOI: 10.1021/nn101277w
  61. 61
    Zhao, J.; Stenzel, M. H. Entry of Nanoparticles into Cells: The Importance of Nanoparticle Properties. Polym. Chem. 2018, 9, 259272,  DOI: 10.1039/C7PY01603D
  62. 62
    Fielding, A. B.; Willox, A. K.; Okeke, E.; Royle, S. J. Clathrin-Mediated Endocytosis is Inhibited During Mitosis. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 6572,  DOI: 10.1073/pnas.1117401109
  63. 63
    Schweitzer, J. K.; Burke, E. E.; Goodson, H. V.; D’Souza-Schorey, C. Endocytosis Resumes during Late Mitosis and Is Required for Cytokinesis. J. Biol. Chem. 2005, 280, 4162841635,  DOI: 10.1074/jbc.M504497200
  64. 64
    Zheng, K.; Xie, J. Engineering Ultrasmall Metal Nanoclusters as Promising Theranostic Agents. Trends Chem. 2020, 2, 665679,  DOI: 10.1016/j.trechm.2020.04.011
  65. 65
    Dejonghe, W.; Kuenen, S.; Mylle, E.; Vasileva, M.; Keech, O.; Viotti, C.; Swerts, J.; Fendrych, M.; Ortiz-Morea, F. A.; Mishev, K.; Delang, S.; Scholl, S.; Zarza, X.; Heilmann, M.; Kourelis, J.; Kasprowicz, J.; Nguyen, L. S. L.; Drozdzecki, A.; Van Houtte, I.; Szatmári, A.-M.; Majda, M.; Baisa, G.; Bednarek, S. Y.; Robert, S.; Audenaert, D.; Testerink, C.; Munnik, T.; Van Damme, D.; Heilmann, I.; Schumacher, K.; Winne, J.; Friml, J.; Verstreken, P.; Russinova, E. Mitochondrial Uncouplers Inhibit Clathrin-Mediated Endocytosis Largely Through Cytoplasmic Acidification. Nat. Commun. 2016, 7, 11710,  DOI: 10.1038/ncomms11710
  66. 66
    Engelberg, S.; Modrejewski, J.; Walter, J. G.; Livney, Y. D.; Assaraf, Y. G. Cancer Cell-Selective, Clathrin-Mediated Endocytosis of Aptamer Decorated Nanoparticles. Oncotarget 2018, 9, 2099321006,  DOI: 10.18632/oncotarget.24772
  67. 67
    Bonora, M.; Patergnani, S.; Rimessi, A.; De Marchi, E.; Suski, J. M.; Bononi, A.; Giorgi, C.; Marchi, S.; Missiroli, S.; Poletti, F.; Wieckowski, M. R.; Pinton, P. ATP Synthesis and Storage. Purinergic Signalling 2012, 8, 343357,  DOI: 10.1007/s11302-012-9305-8
  68. 68
    Moro, L. Mitochondrial Dysfunction in Aging and Cancer. J. Clin. Med. 2019, 8, 1983,  DOI: 10.3390/jcm8111983
  69. 69
    Garrido-Pérez, N.; Vela-Sebastián, A.; López-Gallardo, E.; Emperador, S.; Iglesias, E.; Meade, P.; Jiménez-Mallebrera, C.; Montoya, J.; Bayona-Bafaluy, M. P.; Ruiz-Pesini, E. Oxidative Phosphorylation Dysfunction Modifies the Cell Secretome. Int. J. Mol. Sci. 2020, 21, 3374,  DOI: 10.3390/ijms21093374
  70. 70
    Fiers, W.; Beyaert, R.; Declercq, W.; Vandenabeele, P. More Than One Way to Die: Apoptosis, Necrosis and Reactive Oxygen Damage. Oncogene 1999, 18, 77197730,  DOI: 10.1038/sj.onc.1203249
  71. 71
    Schieber, M.; Chandel, N. S. ROS Function in Redox Signaling and Oxidative Stress. Curr. Biol. 2014, 24, R453R462,  DOI: 10.1016/j.cub.2014.03.034
  72. 72
    Bieri, M.; Bürgi, T. d-Penicillamine Adsorption on Gold: An in Situ ATR-IR Spectroscopic and QCM Study. Langmuir 2006, 22, 83798386,  DOI: 10.1021/la061454y
  73. 73
    Gautier, C.; Bürgi, T. Vibrational Circular Dichroism of N-Acetyl-l-Cysteine Protected Gold Nanoparticles. Chem. Commun. 2005, 43, 53935395

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 5 publications.

  1. Vikas Tiwari, Tarak Karmakar. Understanding Molecular Aggregation of Ligand-Protected Atomically-Precise Metal Nanoclusters. The Journal of Physical Chemistry Letters 2023, 14 (29) , 6686-6694. https://doi.org/10.1021/acs.jpclett.3c01770
  2. Vikas Tiwari, Sonali Garg, Tarak Karmakar. Insights into the Interactions of Peptides with Monolayer-Protected Metal Nanoclusters. ACS Applied Bio Materials 2023, Article ASAP.
  3. Dina Hany, Vasiliki Vafeiadou, Didier Picard. CRISPR-Cas9 screen reveals a role of purine synthesis for estrogen receptor α activity and tamoxifen resistance of breast cancer cells. Science Advances 2023, 9 (19) https://doi.org/10.1126/sciadv.add3685
  4. Ling-Qi Du, Tian-Yu Zhang, Xiao-Mei Huang, Yue Xu, Ming-Xiong Tan, Yan Huang, Yuan Chen, Qi-Pin Qin. Synthesis and anticancer mechanisms of zinc( ii )-8-hydroxyquinoline complexes with 1,10-phenanthroline ancillary ligands. Dalton Transactions 2023, 52 (15) , 4737-4751. https://doi.org/10.1039/D3DT00150D
  5. Youkun Zheng, Yuxin Zhu, Jianghong Dai, Jiaojiao Lei, Jingcan You, Ni Chen, Liqun Wang, Mao Luo, Jianbo Wu. Atomically precise Au nanocluster-embedded carrageenan for single near-infrared light-triggered photothermal and photodynamic antibacterial therapy. International Journal of Biological Macromolecules 2023, 230 , 123452. https://doi.org/10.1016/j.ijbiomac.2023.123452
  • Abstract

    Figure 1

    Figure 1. Characterization of CNC and GNC. (a) The top image shows the structure of the Au25(SR)18 nanocluster (color code: Au atoms, dark yellow; S atoms, green); the ligands (Capt or GSH) are omitted for clarity. The bottom image shows the structure of captopril (Capt) and glutathione (GSH) ligands used for the synthesis of CNC (Au25(Capt)18) and GNC (Au25(GSH)18). (b) UV–vis spectra of the synthesized CNC (red dotted line) and GNC (green squared line). The inset of the figure represents the color of the synthesized CNC and GNC in an aqueous solution. (c) The MALDI mass spectrum of CNC (Au25(Capt)18) recorded in positive mode with α-CHCA as the matrix. The peak marked with an asterisk (*) corresponds to the most important fragment Au21(Capt)14. (d) The ESI spectrum of GNC (Au25(GSH)18) gives characteristic peaks at m/z 809 and 1005 due to [Au(SG)2-H]−1 and [Au2(SG)2-H]−1, respectively. (e) The upper and lower panels, respectively, represent the morphology and size distributions of CNC and GNC obtained by TEM.

    Figure 2

    Figure 2. Cytotoxicity of nanoclusters and ligands on cancer cells. (a) Effects of CNC and GNC (0–500 μg/mL) and the corresponding ligands (Capt and GSH, respectively) on cell viability measured by the CTG assay in HEK293T cells after 72 h treatment (n = 3 biologically independent samples). (b) Only CNC induces a higher percentage of cell death in HEK293T cells as determined by the PI staining assay measured by flow cytometry, whereas GNC and the corresponding ligands (Capt and GSH) are neutral (n = 4 biologically independent samples). Membrane ruptured cells can only be visualized by PI staining and designated as the ″dead cell″ population. (c) The left panel represents the apoptotic cell percentage calculated by the flow-cytometry-based cell cycle assay. Only CNC induces significant apoptosis. The right panel shows that CNC arrests HEK293T cells in the G2/M phase of the cell cycle (n = 3 biologically independent samples). For the bar graphs, the data are represented as mean values ± SEM (standard error of the mean).

    Figure 3

    Figure 3. Energy-dependent CME drives CNC internalization. (a) Dual-color confocal images of HeLa cells labeled with CNC (red color) and exogenously expressed intracellular EGFP (green color). The first row represents HeLa cells without transfection with the EGFP expression plasmid pEGFP-C1. The second and third rows represent cells stained with either CNC or cells transiently transfected with pEGFP-C1. The last row represents the transfected Hela cells with pEGFP-C1 and treated with CNC. The merging of the DIC (differential interference contrast) channel and the other two channels (red: CNC and green: EGFP channel) indicates the colocalization of CNC with EGFP as evident by the yellow color (n = at least 3 independent experiments). It must be noted that these transfection assays with pEGFP-C1 are transient transfections. Scale bar represents 10 μm. (b) The effect of temperature on CNC uptake by HeLa cells. Confocal microscopy of HeLa cells treated with CNC grown in two different temperatures: 4 and 37 °C (n = 2 independent biological samples). Scale bar represents 10 μm. (c) Bar graph representing the mean fluorescence intensity in arbitrary unit (a.u.) showing a comparison of CNC uptake at two different temperatures compared to control (n = a minimum of 50 cells was considered for calculating the mean fluorescence intensity, MFI). (d) Confocal images of HeLa cells pretreated with different endocytosis inhibitors and further incubated with CNC for 8 h to evaluate the uptake efficiency (n = 2 independent biological samples). Scale bar represents 10 μm. (e) Bar graph of mean fluorescence intensity (a.u.) relative to the control sample showing the effect of different inhibitors of the endocytic pathway on the uptake of CNC (n = a minimum of 50 cells was taken into account for calculating the MFI of a particular set). The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests.

    Figure 4

    Figure 4. CNC inhibits complex V of mitochondrial ETC. (a) OCR and (b) ECAR of HeLa cells were monitored using the Seahorse Bioscience Extra Cellular Flux Analyzer in real time. The first few minutes (∼20 min) represent the basal OCR or ECAR of HeLa cells followed by the injection of nanoclusters and then the sequential addition of the ATP synthase inhibitor (oligomycin 5 μM), FCCP (1 μM), and a mixture of rotenone (1 μM) and antimycin (1 μM) was performed. OCR is an indicator of mitochondrial respiration, and ECAR is predominately a measure of glycolytic flux. (c) OCR and (d) ECAR of HeLa cells pretreated with either CNC or GNC for different times (1, 2, and 4 h) and effect of mitochondrial inhibitors in the same order (oligomycin, FCCP, and rotenone + antimycin). (e) OCR and (f) ECAR of Hela cells pretreated with either CNC or GNC for different times (1, 2, and 4 h). Note that the order of addition of the inhibitors was changed. Here, pretreated cells were first treated with the antimycin A and rotenone mixture and subsequently with FCCP and oligomycin A. All data are reported as means ± SD (n = 3 experimental sets). (g) ATP production rates are calculated from the data presented in panel c for HeLa cells pretreated with CNC and GNC. The ATP production rate is calculated as (last OCR measurement before oligomycin injection) – (OCR rate measured after oligomycin injection). All data are presented as means ± SEM (n = minimum of 3 independent biological samples). The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests.

    Figure 5

    Figure 5. CNC-induced mitochondrial respiratory dysfunction triggers intracellular ROS and apoptosis. (a) Intracellular ROS levels were measured by flow cytometry using the H2DCFDA staining assay in HeLa cells treated with CNC, GNC, and their corresponding ligands. For the line graphs, the data are represented as mean values ± SEM (n = 4 biologically independent samples). (b) Mitochondrial ROS levels were measured in HeLa cells after treatment with CNC (500 μg/mL) using MitoSox staining (n = 4 biologically independent samples). (c) Bar graph representing the percentage of mitochondrial membrane depolarized cells as a function of time (0, 3, 6, 12, 24, and 36 h) upon CNC (500 μg/mL) treatment (n = 4 biologically independent samples). (d) Confocal images of HeLa cells transiently transfected with mitochondrially targeted EGFP (mitoEGFP) and later treated with CNC to study the colocalization of CNC into mitochondria. The first row represents HeLa cells without transfection with mitoEGFP. The second and third rows represent cells either stained with CNC or transiently transfected with mitoEGFP. The last row represents HeLa cells transiently transfected with mitoEGFP and then treated with CNC to study the colocalization of CNC into mitochondria. The yellow color in the merged image confirms the localization of CNC in mitochondria. It has to be noted that these transfection assays with pEGFP-C1 are transient transfections. (e) Western blot analysis of Hela cells after treating them with different concentrations of CNC and GNC (200, 300, 400, and 500 μg/mL). The analysis indicates that CNC strikingly reduces the pro-form of caspase 9. α-Tubulin served as a loading control.

    Figure 6

    Figure 6. Restriction in the internalization of CNC protects cells from cell cycle arrest and reduced viability. (a) A comparison of intracellular ROS levels measured in Hela cells treated with CNC (500 μg/mL, red line) and HeLa cells pretreated with endocytic inhibitors Pitstop 2 (40 μM, blue line) and Dynasore (80 μM) to block the uptake of CNC inside the cells. (b) Cell cycle analysis revealed that pretreatment of Pitstop 2 (40 μM) on Hela cells partially protects from CNC (500 μg/mL)-induced G2/M phase cell cycle arrest. (c) A significantly higher number of viable cells were found in the case of HeLa cells pretreated with Pitstop 2 (40 μM) followed by CNC treatment compared to cells treated with only CNC. (d) Cell cycle analysis revealed that vitamin C (75 μM) pretreatment on Hela cells partially protects them from CNC-induced G2/M phase cell cycle arrest. (e) A significantly higher number of viable cells were found in the case of HeLa cells pretreated with vitamin C (75 μM) followed by CNC treatment compared to cells treated with only CNC. The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests.

    Figure 7

    Figure 7. Oncogenic transformation renders cells vulnerable to CNC-induced death. (a) Comparative OCR profiles of HFs (normal parental), HFs-hTERT (normal immortalized), and HFs-hTERT + SV40 + Ras G12V (oncogenically transformed) and effects of mitochondrial inhibitors (oligomycin, FCCP, and rotenone + antimycin) on them (n = 5 biologically independent samples). (b) Bar graphs represent the comparative basal OCR (left; first ∼20 min of respiration), rate of ATP production [middle; ATP production is calculated as (last OCR measurement before oligomycin injection) – (OCR rate measured after oligomycin injection)], and maximal respiratory rate [right; maximal respiratory rate is calculated as (last OCR measurement after oligomycin injection) – (OCR rate measured after FCCP injection)] of HFs (normal parental), HFs-hTERT (normal immortalized), and HFs-hTERT + SV40 + Ras G12V (cancer) cells. All these calculations were done on the experiment presented in panel a. (c) Measurements of total cellular ROS by H2DCFDA staining in the indicated cells in the presence and absence of CNC (n = 3 biologically independent samples). (d) Comparison of cell viability of the indicated cells after 5 days of treatment with CNC (n = 2 biologically independent samples). Note that, in this experiment, we seeded 6 × 103 cells. (e) Measurements of apoptotic cells (<2n nucleus in cell cycle analysis) after 48 h of treatment with CNC of the indicated cells (n = 2 biologically independent samples). The statistical significance between the groups was analyzed by two-tailed unpaired Student’s t tests. *p < 0.05, **p < 0.01.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 73 other publications.

    1. 1
      Knoppe, S.; Bürgi, T. Chirality in Thiolate-Protected Gold Clusters. Acc. Chem. Res. 2014, 47, 13181326,  DOI: 10.1021/ar400295d
    2. 2
      Jin, R. Atomically Precise Metal Nanoclusters: Stable Sizes and Optical Properties. Nanoscale 2015, 7, 15491565,  DOI: 10.1039/C4NR05794E
    3. 3
      Aires, A.; Lopez-Martinez, E.; Cortajarena, A. L. Sensors Based on Metal Nanoclusters Stabilized on Designed Proteins. Biosensors 2018, 8, 110,  DOI: 10.3390/bios8040110
    4. 4
      Porret, E.; Le Guével, X.; Coll, J.-L. Gold Nanoclusters for Biomedical Applications: Toward In Vivo Studies. J. Mater. Chem. B 2020, 8, 22162232,  DOI: 10.1039/C9TB02767J
    5. 5
      Du, Y.; Sheng, H.; Astruc, D.; Zhu, M. Atomically Precise Noble Metal Nanoclusters as Efficient Catalysts: A Bridge between Structure and Properties. Chem. Rev. 2020, 120, 526622,  DOI: 10.1021/acs.chemrev.8b00726
    6. 6
      Govindaraju, S.; Ankireddy, S. R.; Viswanath, B.; Kim, J.; Yun, K. Fluorescent Gold Nanoclusters for Selective Detection of Dopamine in Cerebrospinal fluid. Sci. Rep. 2017, 7, 4029840298,  DOI: 10.1038/srep40298
    7. 7
      Yu, Q.; Gao, P.; Zhang, K. Y.; Tong, X.; Yang, H.; Liu, S.; Du, J.; Zhao, Q.; Huang, W. Luminescent Gold Nanocluster-Based Sensing Platform for Accurate H2S Detection In Vitro and In Vivo with Improved Anti-Interference. Light: Sci. Appl. 2017, e17107  DOI: 10.1038/lsa.2017.107
    8. 8
      Jin, R.; Zeng, C.; Zhou, M.; Chen, Y. Atomically Precise Colloidal Metal Nanoclusters and Nanoparticles: Fundamentals and Opportunities. Chem. Rev. 2016, 116, 1034610413,  DOI: 10.1021/acs.chemrev.5b00703
    9. 9
      Yang, J.; Wang, F.; Yuan, H.; Zhang, L.; Jiang, Y.; Zhang, X.; Liu, C.; Chai, L.; Li, H.; Stenzel, M. Recent advances in ultra-small fluorescent Au nanoclusters toward oncological research. Nanoscale 2019, 11, 1796717980,  DOI: 10.1039/C9NR04301B
    10. 10
      Zheng, Y.; Lai, L.; Liu, W.; Jiang, H.; Wang, X. Recent advances in biomedical applications of fluorescent gold nanoclusters. Adv. Colloid Interface Sci. 2017, 242, 116,  DOI: 10.1016/j.cis.2017.02.005
    11. 11
      Zheng, Y.; Wu, J.; Jiang, H.; Wang, X. Gold nanoclusters for theranostic applications. Coord. Chem. Rev. 2021, 431, 213689  DOI: 10.1016/j.ccr.2020.213689
    12. 12
      Shang, L.; Dörlich, R. M.; Brandholt, S.; Schneider, R.; Trouillet, V.; Bruns, M.; Gerthsen, D.; Nienhaus, G. U. Facile Preparation of Water-Soluble Fluorescent Gold Nanoclusters for Cellular Imaging Applications. Nanoscale 2011, 3, 20092014,  DOI: 10.1039/c0nr00947d
    13. 13
      Zheng, K.; Setyawati, M. I.; Leong, D. T.; Xie, J. Antimicrobial Gold Nanoclusters. ACS Nano 2017, 11, 69046910,  DOI: 10.1021/acsnano.7b02035
    14. 14
      Qi, J.; Liu, Y.; Xu, H.; Xue, T.; Su, Y.; Lin, Z. Anti-Cancer Effect of Melittin-Au25(MHA)18 Complexes on Human Cervical Cancer HeLa Cells. J. Drug Delivery Sci. Technol. 2022, 68, 103078  DOI: 10.1016/j.jddst.2021.103078
    15. 15
      Srinivasulu, Y. G.; Mozhi, A.; Goswami, N.; Yao, Q.; Xie, J. Gold Nanocluster Based Nanocomposites for Combinatorial Antibacterial Therapy for Eradicating Biofilm Forming Pathogens. Mater. Chem. Front. 2022, 6, 689706,  DOI: 10.1039/D1QM00936B
    16. 16
      McLean, A.; Wang, R.; Huo, Y.; Cooke, A.; Hopkins, T.; Potter, N.; Li, Q.; Isaac, J.; Haidar, J.; Jin, R.; Kopelman, R. Synthesis and Optical Properties of Two-Photon-Absorbing Au25(Captopril)18-Embedded Polyacrylamide Nanoparticles for Cancer Therapy. ACS Appl. Nano Mater. 2020, 3, 14201430,  DOI: 10.1021/acsanm.9b02272
    17. 17
      Carlos-Escalante, J. A.; de Jesús-Sánchez, M.; Rivas-Castro, A.; Pichardo-Rojas, P. S.; Arce, C.; Wegman-Ostrosky, T. The Use of Antihypertensive Drugs as Coadjuvant Therapy in Cancer. Front. Oncol. 2021, 11, 660943  DOI: 10.3389/fonc.2021.660943
    18. 18
      Kennedy, L.; Sandhu, J. K.; Harper, M.-E.; Cuperlovic-Culf, M. Role of Glutathione in Cancer: From Mechanisms to Therapies. Biomolecules 2020, 10, 1429,  DOI: 10.3390/biom10101429
    19. 19
      Katla, S. K.; Zhang, J.; Castro, E.; Bernal, R. A.; Li, X. Atomically Precise Au25(SG)18 Nanoclusters: Rapid Single-Step Synthesis and Application in Photothermal Therapy. ACS Appl. Mater. Interfaces 2018, 10, 7582,  DOI: 10.1021/acsami.7b12614
    20. 20
      Roy Bhattacharya, S.; Bürgi, T. Amplified Vibrational Circular Dichroism as a Manifestation of the Interaction Between a Water Soluble Gold Nanocluster and Cobalt Salt. Nanoscale 2019, 11, 2322623233,  DOI: 10.1039/C9NR07534H
    21. 21
      Hahn, W. C.; Counter, C. M.; Lundberg, A. S.; Beijersbergen, R. L.; Brooks, M. W.; Weinberg, R. A. Creation of Human Tumour Cells with Defined Genetic Elements. Nature 1999, 400, 464468,  DOI: 10.1038/22780
    22. 22
      Jung, K.-J.; Dasgupta, A.; Huang, K.; Jeong, S.-J.; Pise-Masison, C.; Gurova, K. V.; Brady, J. N. Small-Molecule Inhibitor Which Reactivates p53 in Human T-Cell Leukemia Virus Type 1-Transformed Cells. J. Virol. 2008, 82, 85378547,  DOI: 10.1128/JVI.00690-08
    23. 23
      Bhattacharya, K.; Weidenauer, L.; Luengo, T. M.; Pieters, E. C.; Echeverría, P. C.; Bernasconi, L.; Wider, D.; Sadian, Y.; Koopman, M. B.; Villemin, M.; Bauer, C.; Rüdiger, S. G. D.; Quadroni, M.; Picard, D. The Hsp70-Hsp90 Co-Chaperone Hop/Stip1 Shifts the Proteostatic Balance from Folding Towards Degradation. Nat. Commun. 2020, 11, 5975,  DOI: 10.1038/s41467-020-19783-w
    24. 24
      Mondal, S.; Bhattacharya, K.; Mandal, C. Nutritional Stress Reprograms Dedifferention in Glioblastoma Multiforme Driven by PTEN/Wnt/Hedgehog Axis: a Stochastic Model of Cancer Stem Cells. Cell Death Discovery 2018, 4, 110,  DOI: 10.1038/s41420-018-0126-6
    25. 25
      Schindelin, J.; Arganda-Carreras, I.; Frise, E.; Kaynig, V.; Longair, M.; Pietzsch, T.; Preibisch, S.; Rueden, C.; Saalfeld, S.; Schmid, B.; Tinevez, J.-Y.; White, D. J.; Hartenstein, V.; Eliceiri, K.; Tomancak, P.; Cardona, A. Fiji: an Open-Source Platform for Biological-Image Analysis. Nat. Methods 2012, 9, 676682,  DOI: 10.1038/nmeth.2019
    26. 26
      Xie, J.; Pan, X.; Wang, M.; Ma, J.; Fei, Y.; Wang, P.-N.; Mi, L. The Role of Surface Modification for TiO2 Nanoparticles in Cancer Cells. Colloids Surf., B 2016, 143, 148155,  DOI: 10.1016/j.colsurfb.2016.03.029
    27. 27
      Joshi, A.; Dai, L.; Liu, Y.; Lee, J.; Ghahhari, N. M.; Segala, G.; Beebe, K.; Jenkins, L. M.; Lyons, G. C.; Bernasconi, L.; Tsai, F. T. F.; Agard, D. A.; Neckers, L.; Picard, D. The Mitochondrial HSP90 Paralog TRAP1 Forms an OXPHOS-Regulated Tetramer and is Involved in Mitochondrial Metabolic Homeostasis. BMC Biol. 2020, 18, 10,  DOI: 10.1186/s12915-020-0740-7
    28. 28
      Yoshida, S.; Tsutsumi, S.; Muhlebach, G.; Sourbier, C.; Lee, M.-J.; Lee, S.; Vartholomaiou, E.; Tatokoro, M.; Beebe, K.; Miyajima, N.; Mohney, R. P.; Chen, Y.; Hasumi, H.; Xu, W.; Fukushima, H.; Nakamura, K.; Koga, F.; Kihara, K.; Trepel, J.; Picard, D.; Neckers, L. Molecular Chaperone TRAP1 Regulates a Metabolic Switch Between Mitochondrial Respiration and Aerobic Glycolysis. Proc. Natl. Acad. Sci. U. S. A. 2013, 110, E1604E1612,  DOI: 10.1073/pnas.1220659110
    29. 29
      Bhattacharya, K.; Bag, A. K.; Tripathi, R.; Samanta, S. K.; Pal, B. C.; Shaha, C.; Mandal, C. Mahanine, a Novel Mitochondrial Complex-III Inhibitor Induces G0/G1 Arrest Through Redox Alteration-Mediated DNA Damage Response and Regresses Glioblastoma Multiforme. Am. J. Cancer Res. 2014, 4, 629647
    30. 30
      Tofanelli, M. A.; Ackerson, C. J. Superatom Electron Configuration Predicts Thermal Stability of Au25(SR)18 Nanoclusters. J. Am. Chem. Soc. 2012, 134, 1693716940,  DOI: 10.1021/ja3072644
    31. 31
      Tofanelli, M. A.; Salorinne, K.; Ni, T. W.; Malola, S.; Newell, B.; Phillips, B.; Häkkinen, H.; Ackerson, C. J. Jahn–Teller Effects in Au25(SR)18. Chem. Sci. 2016, 7, 18821890,  DOI: 10.1039/C5SC02134K
    32. 32
      Shibu, E. S.; Muhammed, M. A. H.; Tsukuda, T.; Pradeep, T. J. Phys. Chem. C 2008, 112, 1216812176,  DOI: 10.1021/jp800508d
    33. 33
      Tay, C. Y.; Yu, Y.; Setyawati, M. I.; Xie, J.; Leong, D. T. Presentation Matters: Identity of Gold Nanocluster Capping Agent Governs Intracellular Uptake and Cell Metabolism. Nano Res. 2014, 7, 805815,  DOI: 10.1007/s12274-014-0441-z
    34. 34
      Sabuncu, A. C.; Grubbs, J.; Qian, S.; Abdel-Fattah, T. M.; Stacey, M. W.; Beskok, A. Probing Nanoparticle Interactions in Cell Culture Media. Colloids Surf., B 2012, 95, 96102,  DOI: 10.1016/j.colsurfb.2012.02.022
    35. 35
      Wang, M.; Wu, Z.; Yang, J.; Wang, G.; Wang, H.; Cai, W. Au25(SG)18 as a Fluorescent Iodide Sensor. Nanoscale 2012, 4, 40874090,  DOI: 10.1039/c2nr30169e
    36. 36
      Dalal, C.; Saha, A.; Jana, N. R. Nanoparticle Multivalency Directed Shifting of Cellular Uptake Mechanism. J. Phys. Chem. C 2016, 120, 67786786,  DOI: 10.1021/acs.jpcc.5b11059
    37. 37
      Bera, K.; Maiti, S.; Maity, M.; Mandal, C.; Maiti, N. C. Porphyrin–Gold Nanomaterial for Efficient Drug Delivery to Cancerous Cells. ACS Omega 2018, 3, 46024619,  DOI: 10.1021/acsomega.8b00419
    38. 38
      Von Kleist, L.; Stahlschmidt, W.; Bulut, H.; Gromova, K.; Puchkov, D.; Robertson, M. J.; MacGregor, K. A.; Tomilin, N.; Pechstein, A.; Chau, N.; Chircop, M.; Sakoff, J.; Peter von Kries, J.; Saenger, W.; Kräusslich, H.-G.; Shupliakov, O.; Robinson, P. J.; McCluskey, A.; Haucke, V. Role of the Clathrin Terminal Domain in Regulating Coated Pit Dynamics Revealed by Small Molecule Inhibition. Cell 2011, 146, 841,  DOI: 10.1016/j.cell.2011.08.014
    39. 39
      Preta, G.; Cronin, J. G.; Sheldon, I. M. Dynasore - Not Just a Dynamin Inhibitor. Cell Commun. Signaling 2015, 13, 24,  DOI: 10.1186/s12964-015-0102-1
    40. 40
      Kuhn, D. A.; Vanhecke, D.; Michen, B.; Blank, F.; Gehr, P.; Petri-Fink, A.; Rothen-Rutishauser, B. Different Endocytotic Uptake Mechanisms for Nanoparticles in Epithelial Cells and Macrophages. Beilstein J. Nanotechnol. 2014, 5, 16251636,  DOI: 10.3762/bjnano.5.174
    41. 41
      Nolfi-Donegan, D.; Braganza, A.; Shiva, S. Mitochondrial Electron Transport Chain: Oxidative Phosphorylation, Oxidant Production, and Methods of Measurement. Redox Biol. 2020, 37, 101674  DOI: 10.1016/j.redox.2020.101674
    42. 42
      Zhao, Y.; Butler, E. B.; Tan, M. Targeting Cellular Metabolism to Improve Cancer Therapeutics. Cell Death Discovery 2013, 4, e532  DOI: 10.1038/cddis.2013.60
    43. 43
      Guo, R.; Gu, J.; Zong, S.; Wu, M.; Yang, M. Structure and Mechanism of Mitochondrial Electron Transport Chain. Biomed. J. 2018, 41, 920,  DOI: 10.1016/j.bj.2017.12.001
    44. 44
      Zhao, R. Z.; Jiang, S.; Zhang, L.; Yu, Z.-B. Mitochondrial Electron Transport Chain, ROS Generation and Uncoupling (Review). Int. J. Mol. Med. 2019, 44, 315,  DOI: 10.3892/ijmm.2019.4188
    45. 45
      Yépez, V. A.; Kremer, L. S.; Iuso, A.; Gusic, M.; Kopajtich, R.; Koňaříková, E.; Nadel, A.; Wachutka, L.; Prokisch, H.; Gagneur, J. OCR-Stats: Robust Estimation and Statistical Testing of Mitochondrial Respiration Activities Using Seahorse XF Analyzer. PLoS One 2018, 13, e0199938  DOI: 10.1371/journal.pone.0199938
    46. 46
      Pike Winer, L. S.; Wu, M. Rapid Analysis of Glycolytic and Oxidative Substrate Flux of Cancer Cells in a Microplate. PLoS One 2014, 9, e109916  DOI: 10.1371/journal.pone.0109916
    47. 47
      Pelicano, H.; Feng, L.; Zhou, Y.; Carew, J. S.; Hileman, E. O.; Plunkett, W.; Keating, M. J.; Huang, P. Inhibition of Mitochondrial Respiration: a Novel Strategy to Enhance Drug-Induced Apoptosis in Human Leukemia Cells by a Reactive Oxygen Species-Mediated Mechanism. J. Biol. Chem. 2003, 278, 3783237839,  DOI: 10.1074/jbc.M301546200
    48. 48
      Kausar, S.; Wang, F.; Cui, H. The Role of Mitochondria in Reactive Oxygen Species Generation and Its Implications for Neurodegenerative Diseases. Cell 2018, 7, 274,  DOI: 10.3390/cells7120274
    49. 49
      Zorov, D. B.; Juhaszova, M.; Sollott, S. J. Mitochondrial Reactive Oxygen Species (ROS) and ROS-Induced ROS Release. Physiol. Rev. 2014, 94, 909950,  DOI: 10.1152/physrev.00026.2013
    50. 50
      Hu, L.; Han, S.; Parveen, S.; Yuan, Y.; Zhang, L.; Xu, G. Highly Sensitive Fluorescent Detection of Trypsin Based on BSA-Stabilized Gold Nanoclusters. Biosens. Bioelectron. 2012, 32, 297299,  DOI: 10.1016/j.bios.2011.12.007
    51. 51
      McDonagh, B. H.; Singh, G.; Bandyopadhyay, S.; Lystvet, S. M.; Ryan, J. A.; Volden, S.; Kim, E.; Sandvig, I.; Sandvig, A.; Glomm, W. R. Controlling the Self-Assembly and Optical Properties of Gold Nanoclusters and Gold Nanoparticles Biomineralized with Bovine Serum Albumin. RSC Adv. 2015, 5, 101101101109,  DOI: 10.1039/C5RA23423A
    52. 52
      Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in Cancer Therapy: The Bright Side of the Moon. Exp. Mol. Med. 2020, 52, 192203,  DOI: 10.1038/s12276-020-0384-2
    53. 53
      Yang, H.; Villani, R. M.; Wang, H.; Simpson, M. J.; Roberts, M. S.; Tang, M.; Liang, X. The Role of Cellular Reactive Oxygen Species in Cancer Chemotherapy. J. Exp. Clin. Cancer Res. 2018, 37, 266,  DOI: 10.1186/s13046-018-0909-x
    54. 54
      McIlwain, D. R.; Berger, T.; Mak, T. W. Caspase Functions in Cell Death and Disease. Cold Spring Harbor Perspect. Biol. 2013, 5, a008656  DOI: 10.1101/cshperspect.a008656
    55. 55
      Roy, S.; Bhattacharya, K.; Mandal, C.; Dasgupta, A. K. Cellular Response to Chirality and Amplified Chirality. J. Mater. Chem. B 2013, 1, 66346643,  DOI: 10.1039/c3tb21322f
    56. 56
      Bhattacharya, K.; Samanta, S. K.; Tripathi, R.; Mallick, A.; Chandra, S.; Pal, B. C.; Shaha, C.; Mandal, C. Apoptotic Effects of Mahanine on Human Leukemic Cells are Mediated Through Crosstalk Between Apo-1/Fas Signaling and the Bid Protein and via Mitochondrial Pathways. Biochem. Pharmacol. 2010, 79, 361372,  DOI: 10.1016/j.bcp.2009.09.007
    57. 57
      Echeverria, P. C.; Bhattacharya, K.; Joshi, A.; Wang, T.; Picard, D. The Sensitivity to Hsp90 Inhibitors of Both Normal and Oncogenically Transformed Cells is Determined by the Equilibrium Between Cellular Quiescence and Activity. PLoS One 2019, 14, e0208287  DOI: 10.1371/journal.pone.0208287
    58. 58
      Lanzetti, L.; Di Fiore, P. P. Endocytosis and Cancer: an ‘Insider’ Network with Dangerous Liaisons. Traffic 2008, 9, 20112021,  DOI: 10.1111/j.1600-0854.2008.00816.x
    59. 59
      Faklaris, O.; Joshi, V.; Irinopoulou, T.; Tauc, P.; Sennour, M.; Girard, H.; Gesset, C.; Arnault, J.-C.; Thorel, A.; Boudou, J.-P.; Curmi, P. A.; Treussart, F. Photoluminescent Diamond Nanoparticles for Cell Labeling: Study of the Uptake Mechanism in Mammalian Cells. ACS Nano 2009, 3, 39553962,  DOI: 10.1021/nn901014j
    60. 60
      Jiang, X.; Röcker, C.; Hafner, M.; Brandholt, S.; Dörlich, R. M.; Nienhaus, G. U. Endo- and Exocytosis of Zwitterionic Quantum Dot Nanoparticles by Live HeLa Cells. ACS Nano 2010, 4, 67876797,  DOI: 10.1021/nn101277w
    61. 61
      Zhao, J.; Stenzel, M. H. Entry of Nanoparticles into Cells: The Importance of Nanoparticle Properties. Polym. Chem. 2018, 9, 259272,  DOI: 10.1039/C7PY01603D
    62. 62
      Fielding, A. B.; Willox, A. K.; Okeke, E.; Royle, S. J. Clathrin-Mediated Endocytosis is Inhibited During Mitosis. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 6572,  DOI: 10.1073/pnas.1117401109
    63. 63
      Schweitzer, J. K.; Burke, E. E.; Goodson, H. V.; D’Souza-Schorey, C. Endocytosis Resumes during Late Mitosis and Is Required for Cytokinesis. J. Biol. Chem. 2005, 280, 4162841635,  DOI: 10.1074/jbc.M504497200
    64. 64
      Zheng, K.; Xie, J. Engineering Ultrasmall Metal Nanoclusters as Promising Theranostic Agents. Trends Chem. 2020, 2, 665679,  DOI: 10.1016/j.trechm.2020.04.011
    65. 65
      Dejonghe, W.; Kuenen, S.; Mylle, E.; Vasileva, M.; Keech, O.; Viotti, C.; Swerts, J.; Fendrych, M.; Ortiz-Morea, F. A.; Mishev, K.; Delang, S.; Scholl, S.; Zarza, X.; Heilmann, M.; Kourelis, J.; Kasprowicz, J.; Nguyen, L. S. L.; Drozdzecki, A.; Van Houtte, I.; Szatmári, A.-M.; Majda, M.; Baisa, G.; Bednarek, S. Y.; Robert, S.; Audenaert, D.; Testerink, C.; Munnik, T.; Van Damme, D.; Heilmann, I.; Schumacher, K.; Winne, J.; Friml, J.; Verstreken, P.; Russinova, E. Mitochondrial Uncouplers Inhibit Clathrin-Mediated Endocytosis Largely Through Cytoplasmic Acidification. Nat. Commun. 2016, 7, 11710,  DOI: 10.1038/ncomms11710
    66. 66
      Engelberg, S.; Modrejewski, J.; Walter, J. G.; Livney, Y. D.; Assaraf, Y. G. Cancer Cell-Selective, Clathrin-Mediated Endocytosis of Aptamer Decorated Nanoparticles. Oncotarget 2018, 9, 2099321006,  DOI: 10.18632/oncotarget.24772
    67. 67
      Bonora, M.; Patergnani, S.; Rimessi, A.; De Marchi, E.; Suski, J. M.; Bononi, A.; Giorgi, C.; Marchi, S.; Missiroli, S.; Poletti, F.; Wieckowski, M. R.; Pinton, P. ATP Synthesis and Storage. Purinergic Signalling 2012, 8, 343357,  DOI: 10.1007/s11302-012-9305-8
    68. 68
      Moro, L. Mitochondrial Dysfunction in Aging and Cancer. J. Clin. Med. 2019, 8, 1983,  DOI: 10.3390/jcm8111983
    69. 69
      Garrido-Pérez, N.; Vela-Sebastián, A.; López-Gallardo, E.; Emperador, S.; Iglesias, E.; Meade, P.; Jiménez-Mallebrera, C.; Montoya, J.; Bayona-Bafaluy, M. P.; Ruiz-Pesini, E. Oxidative Phosphorylation Dysfunction Modifies the Cell Secretome. Int. J. Mol. Sci. 2020, 21, 3374,  DOI: 10.3390/ijms21093374
    70. 70
      Fiers, W.; Beyaert, R.; Declercq, W.; Vandenabeele, P. More Than One Way to Die: Apoptosis, Necrosis and Reactive Oxygen Damage. Oncogene 1999, 18, 77197730,  DOI: 10.1038/sj.onc.1203249
    71. 71
      Schieber, M.; Chandel, N. S. ROS Function in Redox Signaling and Oxidative Stress. Curr. Biol. 2014, 24, R453R462,  DOI: 10.1016/j.cub.2014.03.034
    72. 72
      Bieri, M.; Bürgi, T. d-Penicillamine Adsorption on Gold: An in Situ ATR-IR Spectroscopic and QCM Study. Langmuir 2006, 22, 83798386,  DOI: 10.1021/la061454y
    73. 73
      Gautier, C.; Bürgi, T. Vibrational Circular Dichroism of N-Acetyl-l-Cysteine Protected Gold Nanoparticles. Chem. Commun. 2005, 43, 53935395
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsami.2c05054.

    • Supporting figures including fluorescence emission spectra, FTIR spectra, size of the nanocluster, and stability in the cellular medium for CNC/GNC; cytotoxicity of nanocluster toward different cancer and normal cells; wound healing assay in the presence of CNC and Capt; internalization of CNC and GNC into cell and mitochondria; Seahorse assay with free ligand Capt and GSH; and measurement of intracellular ROS on normal and cancer cell lines (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.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect