|
|
 |
|
ORIGINAL ARTICLE |
|
Year : 2020 | Volume
: 10
| Issue : 7 | Page : 333-340 |
|
Anti-proliferative potential of sodium thiosulfate against HT 29 human colon cancer cells with augmented effect in the presence of mitochondrial electron transport chain inhibitors
Bhavana Sivakumar, Sri Rahavi Boovarahan, Gino A Kurian
Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
Date of Submission | 24-Dec-2019 |
Date of Decision | 14-Jan-2020 |
Date of Acceptance | 26-Feb-2020 |
Date of Web Publication | 03-Jun-2020 |
Correspondence Address: Gino A Kurian Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur India
 Source of Support: None, Conflict of Interest: None  | 3 |
DOI: 10.4103/2221-1691.284948
Objective: To compare the anti-proliferative effect of sodium thiosulfate on human colorectal cancer cells (HT-29) and normal small intestine cells (IEC6). Methods: Cells (HT-29 and IEC6) were treated with different concentrations of sodium thiosulfate ranging from 0.5 mM to 80 mM for 24 h. Cell viability was measured via crystal violet and MTT assays. HT-29 cells were further treated in the presence and absence of mitochondrial electron transport chain (ETC) inhibitors, KATP channel opener and closer and H2S inhibitors for 24 h followed by sodium thiosulfate in order to study their respective roles in the anti-proliferative activity of sodium thiosulfate. Results: The IC50 values of sodium thiosulfate on HT-29 cells were 40.93 mM and 42.45 mM by crystal violet and MTT assay whereas, in the case of IEC6 cells, the values were 45.17 mM and 47.22 mM. The inhibition of endogenous H2S enzymes and KATP channel induced no change in the anti-proliferative capacity of sodium thiosulfate. However, the anti-proliferative activity of sodium thiosulfate was enhanced in the presence of mitochondrial ETC inhibitors. Conclusions: HT-29 cell growth is effectively attenuated by sodium thiosulfate and the anti-proliferative activity of sodium thiosulfate is enhanced in the presence of mitochondrial ETC inhibitors.
Keywords: Colorectal cancer; Sodium thiosulfate; HT-29; IEC6; Mitochondria; Rotenone; Clonogenic assay
How to cite this article: Sivakumar B, Boovarahan SR, Kurian GA. Anti-proliferative potential of sodium thiosulfate against HT 29 human colon cancer cells with augmented effect in the presence of mitochondrial electron transport chain inhibitors. Asian Pac J Trop Biomed 2020;10:333-40 |
How to cite this URL: Sivakumar B, Boovarahan SR, Kurian GA. Anti-proliferative potential of sodium thiosulfate against HT 29 human colon cancer cells with augmented effect in the presence of mitochondrial electron transport chain inhibitors. Asian Pac J Trop Biomed [serial online] 2020 [cited 2023 Mar 24];10:333-40. Available from: https://www.apjtb.org/text.asp?2020/10/7/333/284948 |
Bhavana Sivakumar, Sri Rahavi Boovarahan, Equal contribution.
1. Introduction | |  |
Colorectal cancer is becoming one of the emergent cancers and according to the Global Cancer Observatory, the number of new cases in 2018 was around 18 495 118[1]. The incidence rate of colorectal cancer is linked to people’s nutritional status and its transition is followed by rapid change in the economy and society. The proportion of colorectal cancer cases in the world has increased in individuals younger than 50 years of age, primarily associated with dietary habits and sedentary lifestyle (which generally induces inflammatory response) along with smoking and alcohol consumption[2]. Under chronic inflammatory conditions, the colon polyps develop as the aberrant crypt foci which leads to malignant tumors[3]. Chemotherapeutic agents play a key role in the management of colorectal cancer and are becoming an indispensable part of the treatment in these days[4].
Many sulfur-based drugs like thiophene derivatives which include sulfonamide, isoxazole, benzothiazole, quinoline and anthracene are very promising agents in the treatment of colon cancer[5]. In addition, sulfur-based naturally occurring products are widely used in the traditional system of medicines in India (Ayurveda) and China. The sulfur-containing compound in garlic namely, allicin is found to be effective in the management of gastric cancer because of its pro-apoptotic, anti-proliferative, and anti-helicobacter activities[6]. However, sulfur-based compound efficiency may be linked to its bioavailability as the half-life of many of these compounds are low and the possible cure depends on its concentration in the cancer environment. Documents from the literature have shown that hydrogen sulfide (H2S), a sulfur-based endogenous molecule, possesses both pro and anti-proliferative effects on colon cancer cells[7], thereby putting a hold on the bioavailability issue. By using the slow-releasing H2S donor, GYY4137, investigators have shown that H2S is specifically targeted to induce cancer cell death via activating caspase systems[8], but without making an apprehension on its cellular toxicity on normal cells.
Unlike H2S, sodium thiosulfate (a metabolite of H2S), a sulfur- based compound has high tissue tolerance even up to 250 mg/mL (according to Food and Drug Administration reports). A number of studies have shown that thiosulfate possesses antioxidant potential, calcium chelation effect and can also modulate mitochondrial activity[9] and thereby regulate different signaling pathways that involve NFkB and PI3K[10]. However, the anti-proliferative effect of thiosulfate has not been widely recognized except for a few investigators who have shown the detoxifying effect of platinum-based chemotherapeutic drugs[11]. Also, recent research developments have shown that one of the major etiology of cancer lies within mitochondrial DNA mutations and many investigators consider cancer as a type of mitochondrial metabolic disease[12],[13] The treatment with sodium thiosulfate leads to mitochondrial functional modulation due to its interference with NF-κB and PI3K signaling pathways that converge into the mitochondria, an effective target in the treatment of cancer. Hence, more studies are required to explore its potential to act as an anti-proliferative agent, which is addressed in this study.
2. Materials and Methods | |  |
2.1. Chemicals and reagents
All the chemicals and reagents used in this study were purchased from Sigma-Aldrich (St. Louis, USA).
2.2. Cell culture
The human colon cancer cell line (HT-29) and intestinal epithelial cells (IEC6) were purchased from National Centre for Cell Science, Pune. The cells were maintained in Dulbecco’s Modified Eagle Medium (DMEM, Invitrogen, USA) and 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin were used as supplements. 5% CO2 humidified incubator at 37 °C was used for the incubation of the cells which were later passaged after it reached its 70% confluence.
2.3. Cell viability tests
Cells (HT-29 and IEC6) were plated in a 96-well plate of 1×105 cell density and treated with different concentrations of sodium thiosulfate (0.5 mM, 1 mM,10 mM, 20 mM, 40 mM, 60 mM, 80 mM) for 24 h. The cell viability was measured via crystal violet and MTT assays.
2.3.1.Crystal violet assay
Cell viability was assessed using the crystal violet assay according to the protocol described[14]. Paraformaldehyde at 4% was used to fix HT-29 and IEC6 cells and was then incubated for 30 min at room temperature. The plate was washed twice with water in order to remove the fixative. The cells were stained with 0.5% crystal violet and incubated for 20 min. To dissolve the dye, 200 μL methanol was added and gently shook for 20 min. The absorbance was measured at 570 nm.
2.3.2. 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay
The MTT assay was done to assess the cell viability and the ability of mitochondrial malate dehydrogenase enzyme in viable cells to reduce yellow-colored MTT to form purple-colored formazan, which was measured[15]. A total of 10 μL MTT solution was added to each well containing IEC6 and HT-29 cells. The cells were incubated at 37 °C for 4 h. Then 150 μL of 0.1% acidic isopropanol was used to dissolve the dye. The absorbance was measured using a multimode reader at 540 nm.
2.3.3. Clonogenic assay
Cells (HT-29 and IEC6) were plated in a 35 mm culture dish at 0.3 ×106 cell density and treated with different concentrations of sodium thiosulfate (0.5 mM, 1 mM, 10 mM, 20 mM, 40 mM, 60 mM) for 15 d.
The clonogenic assay or colony formation assay determines the ability of a cell to grow in a colony that determines its proliferation capacity. IEC6 and HT-29 cells were plated at different concentrations (0.5 mM, 1 mM, 10 mM, 20 mM, 40 mM, 60 mM) in order to form colonies in 1-3 weeks[16]. The colonies were fixed with glutaraldehyde (6.0% v/v), further stained with crystal violet (0.5% w/v) and counted. Cell viability was measured by crystal violet assay.
2.4. Inhibitor studies
HT-29 was plated in a 96-well plate of 1×105 cell density and treated with 1 μM sodium azide and 1 μM rotenone [mitochondrial electron transport chain (ETC) inhibitors], 300 μM diazoxide and 25 μM glibenclamide (KATP channel opener and closer), H2S inhibitors 0.2 mM DL-propargyl glycine (PAG) and 0.2 mM aminooxy acetic acid (AOA) for 24 h followed by a combination with sodium thiosulfate at different concentrations (0.1 mM, 0.5 mM, 1 mM, 25 mM).
2.4.1. Effect of sodium thiosulfate in the presence of mitochondrial ETC inhibitors
To evaluate whether sodium thiosulfate modulates its cytotoxic action in HT-29 through mitochondria, we used ETC inhibitors in the presence and absence of sodium thiosulfate. Rotenone is a naturally occurring mitochondrial complex I inhibitor that affects two reversible conformational states of complex I, which leads to the activation and inactivation of the complex. Similarly, selective reduction of complex IV activity was mediated by administrating the cells with sodium azide. Therefore, the HT-29 cells were first treated with 1 μM sodium azide (mitochondrial complex IV inhibitor) for 24 h and different concentrations of sodium thiosulfate (0.1 mM, 0.5 mM, 1 mM, 25 mM). Similarly, the cells were treated with 1 μM rotenone (mitochondrial complex I inhibitor) followed by different concentrations of sodium thiosulfate (0.1 mM, 0.5 mM, 1 mM, 25 mM) in order to understand the cytoprotective action which was measured by crystal violet assay
.
2.4.2. Effect of sodium thiosulfate in the presence of KATP channel modulators
KATP channel of HT-29 cells was modulated by using diazoxide (300 μM acts as KATP channel opener) and glibenclamide (25 μM acts as KATP channel closer). Prior to sodium thiosulfate treatment, HT-29 cells were incubated with KATP channel modulators for 24 h. At the end of the experiment, cytotoxicity was determined by crystal violet assay.
2.4.3. Effect of sodium thiosulfate in the presence of endogenous H2S inhibitors
The influential role of endogenous H2S enzyme in the anti- proliferative effect of sodium thiosulfate was evaluated by using H2S biosynthetic enzyme inhibitors like DL-PAG and AOA. Prior to sodium thiosulfate administration, the cells were treated with PAG and AOA for 24 h and the cytotoxicity effect was evaluated by crystal violet assay.
2.5. Statistical analysis
All values were expressed as mean ± SD. The data were subjected to ANOVA for comparison of differences between groups using GraphPad Prism 7 software. P <0.05 was considered statistically significant.
3. Results | |  |
3.1. Sodium thiosulfate induces dose-dependent decline in survival of HT-29 colon cancer cells
HT-29 and IEC6 cells were cultured in the presence or absence of sodium thiosulfate at various concentrations (0.5 mM, 1 mM, 10 mM, 20 mM, 40 mM, 60 mM, and 80 mM) for 24 h and cell viability was measured [Figure 1]. Exposure to 20 mM sodium thiosulfate was found to be toxic to HT-29, but IEC6 cells could withstand the 20 mM sodium thiosulfate and sodium thiosulfate was toxic from 40 mM concentration. The IC50 value of HT-29 was found to be 40.93 mM and 42.45 mM when evaluated by crystal violet and MTT assays, respectively. IEC6 cells showed IC50 values of 45.17 mM and 47.22 mM. | Figure 1: Cytotoxicity of sodium thiosulfate (STS) in HT-29 cells and IEC6 cells. HT-29 and IEC6 cells were cultured in the presence or absence of sodium thiosulfate at various concentrations. A: HT-29 cells measured via MTT; B: IEC6 cells measured via MTT; C: HT-29 cells measured via crystal violet assays; D: IEC6 cells measured via crystal violet assays. * indicates P<0.05 vs. control
Click here to view |
In order to determine the anti-proliferative activity of sodium thiosulfate, the clonogenic assay was performed and the results are shown in [Figure 2]. HT-29 cells treated with 40 mM of sodium thiosulfate showed only 46% of live cells in 15 d whereas IEC6 cells showed 68% of live cells at the same concentration of sodium thiosulfate and same time period. | Figure 2: Clonogenic assay. The anti-proliferative capacity of sodium thiosulfate (STS) reconfirmed by clonogenic assay done on (A) HT-29 cells and (B) IEC6 cells. * indicates P<0.05 vs. control.
Click here to view |
3.2. Sodium thiosulfate augments the cytotoxicity effect in the presence of ETC inhibitors in HT-29 colon cancer cells
Treatment with complex] IV inhibitor sodium azide resulted in (28±7)% and (30±6)% cell death, estimated via MTT and crystal violet assays, respectively [Figure 3]. However, a combination of sodium azide with different concentrations of sodium thiosulfate (0.1 mM, 0.5 mM, 1 mM, 25 mM) led to approximately (60±4)%, (65±4)%, (67±3)%, and (69±3)% cell death, respectively in MTT assay. In the case of crystal violet assay, cell death of (57±4)%, (61±4)%, (63±3)%, and (65±3)% were found at 0.1 mM, 0.5 mM, 1 mM, and 25 mM, respectively. These results indicate a significant (P <0.05) cell death in both cases. | Figure 3: Effect of electron transport chain (complex -IV) inhibitor, 1 μM sodium azide on the anti-proliferative effect of sodium thiosulfate (STS) in HT-29 cell line evaluated by (A) MTT assay and (B) crystal violet assay. * indicates P<0.05 vs. control
Click here to view |
Similarly, 1 μM rotenone treatment of HT-29 cells for 24 h showed (57±4)% cell death in MTT and (55±4)% death in crystal violet assay. Conversely, the combined treatment of rotenone with different sodium thiosulfate concentrations as mentioned above led to (72±4)%, (75±4)%, (78±3)%, and (81±3)% cell death in MTT assay and (70±4)%, (72±4)%, (75±3)%, and (77±3)% in crystal violet assay, indicating a significant (P <0.05) cytotoxicity of sodium thiosulfate in the presence of rotenone [Figure 4]. Hence it can be concluded that neither sodium azide nor rotenone, when given individually, can impart prominent cytotoxicity to the HT-29 cells. On the other hand, the presence of ETC inhibitors enhanced the cytotoxic effect of sodium thiosulfate on HT-29 cells. | Figure 4: Effect of electron transport chain (complex -I) inhibitor, 1 μM rotenone on the anti-proliferative effect of sodium thiosulfate (STS) in HT-29 cell line evaluated by (A) MTT assay and (B) crystal violet assay. * indicates P<0.05 vs. control.
Click here to view |
3.3. Sodium thiosulfate mediated cytotoxicity in HT-29 colon cancer cells is independent of mito-KATP channel
Since ETC enzymes need intact KATP channel for its functional activity, we further assessed the effect of sodium thiosulfate on HT-29 in the presence of KATP channel modulators. According to [Figure 5] and [Figure 6], the cytotoxic effect of sodium thiosulfate was not significantly changed in the presence of 300 μM diazoxide (KATP channel opener) and 25 μM glibenclamide (KATP channel closer), when compared with the sodium thiosulfate group without the above-mentioned inhibitors. | Figure 5: Effect of sodium thiosulfate (STS) on HT-29 in the presence of KATP channel modulator-diazoxide (300 μM) evaluated by (A) MTT assay and (B) crystal violet assay. No significant changes were observed.
Click here to view |
 | Figure 6: Effect of sodium thiosulfate (STS) on HT-29 in the presence of KATP channel modulator-glibenclamide (25 μM), evaluated by (A) MTT assay and (B) crystal violet assay. No significant changes were observed.
Click here to view |
3.4. Anti-proliferative effect of sodium thiosulfate was intact in HT-29 cells in the presence of DL-PAG or AOA
Next, we evaluated whether the anti-proliferative effect of sodium thiosulfate depends on endogenous H2S, where the latter has already been proven to be a potent anticancer agent on HT-29 cells. We found that the cytotoxic effect of sodium thiosulfate on HT-29 was unaltered in the presence of DL-PAG or AOA, indicating the sodium thiosulfate mediated cytotoxicity is H2S independent [Figure 7] & [Figure 8]. | Figure 7: Cytotoxicity of sodium thiosulfate (STS) in the presence of endogenous H2S inhibitor- propargyl glycine (PAG) in the HT-29 cell line evaluated by (A) MTT assay and (B) crystal violet assay. No significant changes were observed.
Click here to view |
 | Figure 8: Cytotoxicity of sodium thiosulfate (STS) in the presence of endogenous H2S inhibitor-aminooxy acetic acid (AOA) in HT-29 cell lines evaluated by (A) MTT assay and (B) crystal violet assay. No significant changes were observed.
Click here to view |
4. Discussion | |  |
The ambiguous mechanisms in cancer pathology make cancer therapy difficult and thus the research focusing on its eradication is still attracting greater attention of both clinicians and scientific investigators. In the present study, the anti-proliferative capacity of sodium thiosulfate was evaluated on HT-29 colorectal cancer cells and the effects were compared with IEC6 normal small intestine cells. Based on the clonogenic and cytotoxicity assays, we found that sodium thiosulfate can inhibit the growth of HT-29 cells. The IC50 values of HT-29 cells were 40.93 mM and 42.45 mM by crystal violet and MTT assay whereas in IEC6 cells, the values were 45.17 mM and 47.22 mM, respectively. Moreover, we also found that the anti-proliferative effect of sodium thiosulfate was augmented in the presence of rotenone (complex I) and sodium azide (complex IV) inhibitors.
Among the different anticancer drugs approved by the Food and Drug Administration from 2010 to 2015, two-thirds of them are heterocyclic compounds[17]. Sulfur-based heterocyclic compounds have exhibited potent anticancer property primarily because of its role in co-enzyme chemistry and its ability to interact with different regulatory molecules[18]. A recent clinical trial by Hauschild and his co-workers showed that dabrafenib, a well-known sulfur-based heterocyclic compound, is known to inhibit mutated BRAF and thereby improve the survival rates in BRAF mutated colorectal cancer patients[19]. Similarly, few investigators have shown that thiazole derivatives are effective against colon cancer cell lines[20]. Even though sulfur-based heterocyclic compounds provide many promises in the management of colorectal cancer, they have their own limitations. Sulfur-based heterocyclic compounds work in tight regulatory control of cells, making it difficult to distinguish the delicate balance of cell metabolism in normal and cancer cells. Hence it has become an utmost necessity to identify a potential sulfur-based molecule that is endogenous in nature. This shortcoming can be addressed to a certain extent by recent findings that prove the anti-proliferative action of endogenous sulfur compounds like hydrogen sulfide[18]. Accordingly, literature against cancer suggested that H2S can inhibit the growth of cancer cells[21.22]. On the contrary, there are evidences which suggest the tumor-promoting activity of H2S as well[23]. The controversial findings of H2S (pro and anti- cancer effects) will bring doubt in the use of H2S as a therapeutic agent in the management of colon cancer. So this calls for a new therapeutic endogenous sulfur-based drug which is safer than H2S.
Thiosulfate, one of the main metabolites of hydrogen sulfide, is reported to interact with the mitochondrial electron transport system[24], apoptosis and is believed to be a potent candidate in the management of cancer. In the present study, we found that exogenous administration of thiosulfate attenuated the proliferation capacity of HT-29 cells. Exposure to 20 mM sodium thiosulfate inhibited the growth of colon cancer epithelial cells (HT-29) while the normal intestinal epithelial cell growth (IEC6) was found to be inhibited from 40 mM sodium thiosulfate.
In agreement with our findings, Freyer and his group have shown that sodium thiosulfate can reduce hearing loss due to cisplatin in cancer patients[25]. Similarly, Ramasamy and his workers demonstrated that thiosulfate sulfur transferase can act as a tumor marker for colorectal cancer as its mRNA expression level was found to be markedly declined in cancer colonocytes. Thiosulfate sulfur transferase and mercapto pyruvate sulfur transferase are the isoforms of rhodanese enzymes that catabolize intracellular hydrogen sulfide to thiosulfate[26].
Since the anti-proliferative property of sodium thiosulfate was confirmed in HT-29 cells, we evaluated whether the inhibition of HT-29 cell growth was mediated via thiosulfate metabolites (H2S, sulphate). We used H2S metabolizing enzyme inhibitors PAG and AOA before the administration of sodium thiosulfate. These results indicate the cytotoxic effect of sodium thiosulfate on HT-29 cells was unaltered even in the presence of PAG or AOA, suggesting sodium thiosulfate mediated cytotoxicity is H2S independent.
A recent study by Brown and his coworkers showed that caspase-3 knockout in colorectal cancer cells leads to the sensitization of colon cancer cells towards chemotherapeutic agents by promoting receptor-interacting serine/threonine-protein kinase 1, procaspase 8 and ROS dependent necrosis[27]. Similarly, an in silico study predicts the interaction of sodium thiosulfate with the cascade of the caspase system via binding to caspase 3, which is one of the major executors of apoptosis[28]. Evidence from literature signifies the influence of dysregulated apoptosis (downregulation of caspase 9&3) system in cancer pathology[29].
Thus, the anti-proliferative activity of sodium thiosulfate may be attributed to the activation of caspase 3 (based on our previous in silico result[28]. Since caspase-3 is widely present in mitochondria[30] and its site of action is linked with complex 1 enzyme activity, we further investigated the role of mitochondria in the anti-proliferative property of sodium thiosulfate. Accordingly, we found that the underlying mode of action of sodium thiosulfate was not mediated through the mitochondrial ETC enzyme activity. In fact, in the presence of ETC enzyme inhibitors, the anti-proliferative action of sodium thiosulfate was enhanced, thus suggesting a synergetic effect. In addition, we used mitochondrial KATP channel inhibitors along with sodium thiosulfate to confirm its interactive effect with mitochondria. The result suggests that sodium thiosulfate action was intact even in the presence of KATP channel inhibitors, confirming the absence of mitochondrial linked sodium thiosulfate anti-proliferative effect. Similarly, H2S mediated sodium thiosulfate anti-proliferative action was ruled out by using inhibitors of endogenous H2S metabolizing enzymes, thereby asserting the anti-proliferative effect attributed to sodium thiosulfate rather than its metabolite.
Based on the above findings, we conclude that sodium thiosulfate possesses the potential to curb HT-29 growth. Moreover, the anti- proliferative activity of sodium thiosulfate can be enhanced in the presence of mitochondrial ETC inhibitors.
Conflict of interest statement
We declare that there is no conflict of interest.
Authors’ contributions
GAK desiged the work. SRB and BS analysed data, wrote and revised the article The final approval of the version to be published was done by GAK.
References | |  |
1. | Ferlay J, Colombet M, Soeijomataram I, Mathers C, Parkin DM, Piñeros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019; 144(8): 19411953. |
2. | Weinberg BA, Marshall JL. Colon cancer in young adults: Trends and their implications. Curr Oncol Rep 2019; 21: 3. |
3. | Simon K. Colorectal cancer development and advances in screening. Clin Interv Aging 2016; 11: 967-976. |
4. | Lansdorp-Vogelaar I, Van Ballegooijen M, Zauber AG, Habbema JD, Kuipers EJ. Effect of rising chemotherapy costs on the cost savings of colorectal cancer screening. J Natl Cancer Inst 2009; 101: 1412-1422. |
5. | Ghorab MM, Bashandy MS, Alsaid MS. Novel thiophene derivatives with sulfonamide, isoxazole, benzothiazole, quinoline and anthracene moieties as potential anticancer agents. Acta Pharm 2014; 64: 419-431. |
6. | Omar SH, Al-Wabel NA. Organosulfur compounds and possible mechanism of garlic in cancer. Saudi Pharm J 2010; 18: 51-58. |
7. | Wu YC, Wang XJ, Yu L, Chan FKL, Cheng ASL, Yu J, et al. Hydrogen sulfide lowers proliferation and induces protective autophagy in colon epithelial cells. PLoS One 2012; 7: e37572. |
8. | Lee ZW, Zhou J, Chen CS, Zhao Y, Tan CH, Li L, et al. The slow- releasing hydrogen sulfide donor, GYY4137, exhibits novel anti-cancer effects in vitro and in vivo. PLoS One 2011; 6: e21077. |
9. | Ravindran S, Boovarahan SR, Shanmugam K, Vedarathinam RC, Kurian GA. Sodium thiosulfate preconditioning ameliorates ischemia/reperfusion injury in rat hearts via reduction of oxidative stress and apoptosis. Cardiovasc Drugs Ther 2017; 31(5-6): 511-524. |
10. | Hayden MR, Goldsmith DJ. Sodium thiosulfate: New hope for the treatment of calciphylaxis. Semin Dial 2010; 23: 258-262. |
11. | Yan F, Liu JJ, Ip V, Jamieson SM, McKeage MJ. Role of platinum DNA damage-induced transcriptional inhibition in chemotherapy-induced neuronal atrophy and peripheral neurotoxicity. J Neurochem 2015; 135: 1099-1112. |
12. | Kalyanaraman B, Cheng G, Hardy M, Ouari O, Lopez M, Joseph J, et al. A review of the basics of mitochondrial bioenergetics, metabolism, and related signaling pathways in cancer cells: Therapeutic targeting of tumor mitochondria with lipophilic cationic compounds. Redox Biol 2018; 14: 316-327. |
13. | Yang Y, Karakhanova S, Hartwig W, D’Haese JG, Philippov PP, Werner J, et al. Mitochondria and mitochondrial ROS in cancer: Novel targets for anticancer therapy. Cell Physiol 2016; 231: 2570-2581. |
14. | Feoktistova M, Geserick P, Leverkus M. Crystal violet assay for determining viability of cultured cells. Cold Spring Harb Prot 2016; 2016. Doi: 10.1101/pdb.prot087379. |
15. | van Meerloo J, Kaspers GJL, Cloos J. Cell sensitivity assays: The MTT assay. Methods Mol Biol 2011; 731: 237-245. |
16. | Franken NAP, Rodermond HM, Stap J, Haveman J, Bree CV. Clonogenic assay of cells in vitro. Nat Prot 2006; 1: 2315-2319. |
17. | Tandon R, Singh I, Luxami V, Tansdon N, Paul K. Recent advances and developments of in vitro evaluation of heterocyclic moieties on cancer cell lines. Chem Rec 2019; 19(2-3): 362-393. |
18. | Martins P, Jesus J, Santos S, Raposo LR, Roma-Rodrigues C, Baptista PV, et al. Heterocyclic anticancer compounds: Recent advances and the paradigm shift towards the use of nanomedicine’s tool box. Molecules 2015; 20: 16852-16891. |
19. | Hauschild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, Millward M, et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012; 380(9839): 358-365. |
20. | Mohareb RM, Abdallah AEM, Ahmed EA. Synthesis and cytotoxicity evaluation of thiazole derivatives obtained from 2-amino-4,5,6,7- tetrahydrobenzobthiophene- 3-carbonitrile. Acta Pharm 2017; 67(4): 495510. |
21. | Fiorucci S, Santucci L. Hydrogen sulfide-based therapies: Focus on H 2S releasing NSAIDs. Inflamm Allergy Drug Targets 2011; 10(2): 133-140. |
22. | Lee ZW, Teo XY, Tay EYW, Tan CH, Hagen T, Moore PK, et al. Utilizing hydrogen sulfide as a novel anti-cancer agent by targeting cancer glycolysis and pH imbalance. Br J Pharmacol 2014; 171(18): 4322-4336. |
23. | Kumarasamy A, Kurian GA. Hydrogen sulfide promotes proliferation of HT-29 colon cancer cells in a mitochondria-independent pathway. Indian J Pharm Sci 2019; 81(3): 456-463. |
24. | Ravindran S, Kurian GA. Effect of sodium thiosulfate postconditioning on ischemia-reperfusion injury induced mitochondrial dysfunction in rat heart. J Cardiovasc Transl Res 2018; 11(3): 246-258. |
25. | Freyer DR, Chen L, Krailo MD, Knight K, Luna DV, Bliss B, et al. Effects of sodium thiosulfate versus observation on development of cisplatin-induced hearing loss in children with cancer. Lancet Oncol 2017; 18(1): 63-74. |
26. | Ramasamy S, Singh S, Taniere P, Langman MJ, Eggo MC. Sulfide- detoxifying enzymes in the human colon are decreased in cancer and upregulated in differentiation. Am J Physiol Gastrointest Liver Physiol 2006; 291(2): G288-G296. |
27. | Brown MF, Leibowitz BJ, Chen D, He K, Zou F, Sobol RW, et al. Loss of caspase-3 sensitizes colon cancer cells to genotoxic stress via RIP1- dependent necrosis. Cell Death Dis 2015; 6: e1729. |
28. | Ravindran S, Boovarahan SR, Shanmugam K, Vedarathinam RC, Kurian GA. Sodium thiosulfate preconditioning ameliorates ischemia/ reperfusion injury in rat hearts via reduction of oxidative stress and apoptosis. Cardiovasc Drugs Ther 2017; 31(5-6): 511-524. |
29. | Asadi M, Shanehbandi D, Kermani TA, Sanaat Z, Zafari V, et al. Expression level of caspase genes in colorectal cancer. Asian Pac J Cancer Prev 2018; 19(5): 1277-1280. |
30. | Mancini M, Nicholson DW, Roy S, Thornberry NA, Peterson EP, Casciola-rosen LA, et al. The caspase-3 precursor has a cytosolic and mitochondrial distribution: Implications for apoptotic signaling. J Cell Biol 1998; 140(6): 1485-1495. |
[Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5], [Figure 6], [Figure 7], [Figure 8]
This article has been cited by | 1 |
Effects of a dietary anionic alkali mineral complex in juvenile olive flounder (Paralichthys olivaceus) during low water temperature season |
|
| Buddhi E. Gunathilaka, Nalin Medagoda, Ji-Hoon Cha, Byung-Woo Yoo, Soo-Il Choi, Chang-Hoon Shin, Kyeong-Jun Lee | | Aquaculture Reports. 2022; 27: 101420 | | [Pubmed] | [DOI] | | 2 |
Diesel particulate matter exposure deteriorates cardiovascular health and increases the sensitivity of rat heart towards ischemia reperfusion injury via suppressing mitochondrial bioenergetics function |
|
| Bhavana Sivakumar, Gino A. Kurian | | Chemico-Biological Interactions. 2021; : 109769 | | [Pubmed] | [DOI] | | 3 |
Fisetin ameliorates ischemia re-oxygenation injury in H9c2 cardiomyocytes via targeting the PI3K signalling pathway |
|
| Bhavana Sivakumar,Sri Rahavi Boovarahan,Priyanka N Prem,Gino A Kurian | | Phytomedicine Plus. 2021; 1(3): 100094 | | [Pubmed] | [DOI] | |
|
 |
 |
|