1
|
Poulios E, Roupaka V, Giaginis C, Galaris D, Spyrou G. Implication of Thioredoxin 1 and Glutaredoxin 1 in H 2O 2-induced Phosphorylation of JNK and p38 MAP Kinases. Curr Mol Med 2025; 25:305-319. [PMID: 38243922 DOI: 10.2174/0115665240271103231127072635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Aerobic organisms continuously generate small amounts of Reactive Oxygen Species (ROS), which are involved in the oxidation of sensitive cysteine residues in proteins, leading to the formation of disulfide bonds. Thioredoxin (Trx1) and Glutaredoxin (Grx1) represent key antioxidant enzymes reducing disulfide bonds. OBJECTIVE In this work, we have focused on the possible protective effect of Trx1 and Grx1 against oxidative stress-induced DNA damage and apoptosis-signaling, by studying the phosphorylation of MAP kinases. METHODS Trx1 and Grx1 were overexpressed or silenced in cultured H1299 non-small cell lung cancer epithelial cells. We examined cell growth, DNA damage, and the phosphorylation status of MAP kinases following treatment with H2O2. RESULTS Overexpression of both Trx1 and Grx1 had a significant impact on the growth of H1299 cells and provided protection against H2O2-induced toxicity, as well as acute DNA single-strand breaks. Conversely, silencing of these proteins exacerbated DNA damage. Furthermore, overexpression of Trx1 and Grx1 inhibited the rapid phosphorylation of JNK (especially at 360 min of treatment, ****p=0.004 and **p=0.0033 respectively) and p38 MAP kinases (especially at 360 min of treatment, ****p<0.0001 and ***p=0.0008 respectively) during H2O2 exposure, while their silencing had the opposite effect (especially at 360 min of treatment, ****p<0.0001). CONCLUSION These results suggest that both Trx1 and Grx1 have protective roles against H2O2 induced toxicity, emphasizing their significance in mitigating oxidative stress-related cellular damage.
Collapse
Affiliation(s)
- Efthymios Poulios
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Metropolite Ioakeim 2, 81400 Myrina, Lemnos, Greece
| | - Vasiliki Roupaka
- Laboratory of Biological Chemistry, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Metropolite Ioakeim 2, 81400 Myrina, Lemnos, Greece
| | - Dimitrios Galaris
- Laboratory of Biological Chemistry, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Giannis Spyrou
- Department of Biomedical and Clinical Sciences, Division of Clinical Chemistry, Medical Faculty, S-581 85 Linköping, Sweden
| |
Collapse
|
2
|
Einstein SA, Steyn LV, Weegman BP, Suszynski TM, Sambanis A, O'Brien TD, Avgoustiniatos ES, Firpo MT, Graham ML, Janecek J, Eberly LE, Garwood M, Putnam CW, Papas KK. Hypoxia within subcutaneously implanted macroencapsulation devices limits the viability and functionality of densely loaded islets. FRONTIERS IN TRANSPLANTATION 2023; 2:1257029. [PMID: 38993891 PMCID: PMC11235299 DOI: 10.3389/frtra.2023.1257029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/20/2023] [Indexed: 07/13/2024]
Abstract
Introduction Subcutaneous macroencapsulation devices circumvent disadvantages of intraportal islet therapy. However, a curative dose of islets within reasonably sized devices requires dense cell packing. We measured internal PO2 of implanted devices, mathematically modeled oxygen availability within devices and tested the predictions with implanted devices containing densely packed human islets. Methods Partial pressure of oxygen (PO2) within implanted empty devices was measured by noninvasive 19F-MRS. A mathematical model was constructed, predicting internal PO2, viability and functionality of densely packed islets as a function of external PO2. Finally, viability was measured by oxygen consumption rate (OCR) in day 7 explants loaded at various islet densities. Results In empty devices, PO2 was 12 mmHg or lower, despite successful external vascularization. Devices loaded with human islets implanted for 7 days, then explanted and assessed by OCR confirmed trends proffered by the model but viability was substantially lower than predicted. Co-localization of insulin and caspase-3 immunostaining suggested that apoptosis contributed to loss of beta cells. Discussion Measured PO2 within empty devices declined during the first few days post-transplant then modestly increased with neovascularization around the device. Viability of islets is inversely related to islet density within devices.
Collapse
Affiliation(s)
- Samuel A Einstein
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
- Department of Radiology, The Pennsylvania State University, Hershey, PA, United States
| | - Leah V Steyn
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Bradley P Weegman
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
- Sylvatica Biotech Inc., North Charleston, SC, United States
| | - Thomas M Suszynski
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Athanassios Sambanis
- Department of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Timothy D O'Brien
- Veterinary Population Medicine Department, University of Minnesota, Saint Paul, MN, United States
- Department of Medicine, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | | | - Meri T Firpo
- Department of Medicine, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Melanie L Graham
- Veterinary Population Medicine Department, University of Minnesota, Saint Paul, MN, United States
- Department of Surgery, Preclinical Research Center, University of Minnesota, Saint Paul, MN, United States
| | - Jody Janecek
- Department of Surgery, Preclinical Research Center, University of Minnesota, Saint Paul, MN, United States
| | - Lynn E Eberly
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, United States
| | - Michael Garwood
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| | - Charles W Putnam
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Klearchos K Papas
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
3
|
Mansor NI, Ling KH, Rosli R, Hassan Z, Adenan MI, Nordin N. Centella asiatica (L.) Urban. Attenuates Cell Damage in Hydrogen Peroxide-Induced Oxidative Stress in Transgenic Murine Embryonic Stem Cell Line-Derived Neural-Like Cells: A Preliminary Study for Potential Treatment of Alzheimer's Disease. J Alzheimers Dis 2023; 94:S21-S44. [PMID: 37334592 PMCID: PMC10473099 DOI: 10.3233/jad-221233] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Centella asiatica (L.) (C. asiatica) is commonly known in South East and South East Asia communities for its nutritional and medicinal benefits. Besides being traditionally used to enhance memory and accelerate wound healing, its phytochemicals have been extensively documented for their neuroprotective, neuroregenerative, and antioxidant properties. OBJECTIVE The present study aims to investigate the effects of a standardized raw extract of C. asiatica (RECA) on hydrogen peroxide (H2O2)-induced oxidative stress and apoptotic death in neural-like cells derived from mouse embryonic stem (ES) cell line. METHODS A transgenic mouse ES cell (46C) was differentiated into neural-like cells using 4-/4+ protocol with addition of all-trans retinoic acid. These cells were then exposed to H2O2 for 24 h. The effects of RECA on H2O2-induced neural-like cells were assessed through cell viability, apoptosis, and reactive oxygen species (ROS) assays, as well as neurite length measurement. The gene expression levels of neuronal-specific and antioxidant markers were assessed by RT-qPCR analysis. RESULTS Pre-treatment with H2O2 for 24 hours, in a dose-dependent manner, damaged neural-like cells as marked by a decrease in cell viability, substantial increase in intracellular ROS accumulation, and increase in apoptotic rate compared to untreated cells. These cells were used to treat with RECA. Treatment with RECA for 48 h remarkably restored cell survival and promoted neurite outgrowth in the H2O2- damaged neurons by increasing cell viability and decreasing ROS activity. RT-qPCR analysis revealed that RECA upregulated the level of antioxidant genes such as thioredoxin-1 (Trx-1) and heme oxygenase-1 (HO-1) of treated cells, as well as the expression level of neuronal-specific markers such as Tuj1 and MAP2 genes, suggesting their contribution in neuritogenic effect. CONCLUSION Our findings indicate that RECA promotes neuroregenerative effects and exhibits antioxidant properties, suggesting a valuable synergistic activity of its phytochemical constituents, thus, making the extract a promising candidate in preventing or treating oxidative stress-associated Alzheimer's disease.
Collapse
Affiliation(s)
- Nur Izzati Mansor
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras Kuala Lumpur, Malaysia
| | - King-Hwa Ling
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeing™), Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Rozita Rosli
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Gelugor, Penang, Malaysia
| | - Mohd Ilham Adenan
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA, Puncak Alam Campus, Bandar PuncakAlam, Selangor Darul Ehsan, Malaysia
| | - Norshariza Nordin
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeing™), Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
4
|
Kowluru A. Roles of GTP and Rho GTPases in pancreatic islet beta cell function and dysfunction. Small GTPases 2020; 12:323-335. [PMID: 32867592 DOI: 10.1080/21541248.2020.1815508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A growing body of evidence implicates requisite roles for GTP and its binding proteins (Rho GTPases) in the cascade of events leading to physiological insulin secretion from the islet beta cell. Interestingly, chronic exposure of these cells to hyperglycaemic conditions appears to result in sustained activation of specific Rho GTPases (e.g. Rac1) leading to significant alterations in cellular functions including defects in mitochondrial function and nuclear collapse culminating in beta cell demise. One of the objectives of this review is to highlight our current understanding of the regulatory roles of GTP and Rho GTPases in normal islet function (e.g. proliferation and insulin secretion) as well potential defects in these signalling molecules and metabolic pathways that could contribute islet beta cell dysfunction and loss of functional beta cell mass leading to the onset of diabetes. Potential knowledge gaps in this field and possible avenues for future research are also highlighted. ABBREVIATIONS ARNO: ADP-ribosylation factor nucleotide binding site opener; CML: carboxyl methylation; Epac: exchange protein directly activated by cAMP; ER stress: endoplasmic reticulum stress; FTase: farnesyltransferase; GAP: GTPase activating protein; GDI: GDP dissociation inhibitor; GEF: guanine nucleotide exchange factor; GGTase: geranylgeranyltransferase; GGpp: geranylgeranylpyrophosphate; GGPPS: geranylgeranyl pyrophosphate synthase; GSIS: glucose-stimulated insulin secretion; HGPRTase: hypoxanthine-guanine phosphoribosyltransferase; IMPDH: inosine monophosphate dehydrogenase; α-KIC: α-ketoisocaproic acid; MPA: mycophenolic acid; MVA: mevalonic acid; NDPK: nucleoside diphosphate kinase; NMPK: nucleoside monophosphate kinase; Nox2: phagocyte-like NADPH oxidase; PAK-I: p21-activated kinase-I; β-PIX: β-Pak-interacting exchange factor; PRMT: protein arginine methyltransferase; Rac1: ras-related C3 botulinum toxin substrate 1; Tiam1: T-cell lymphoma invasion and metastasis-inducing protein 1; Trx-1: thioredoxin-1; Vav2: vav guanine nucleotide exchange factor 2.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Biomedical Research Service, John D. Dingell VA Medical Center and Department of Pharmaceutical Sciences and Medicine, Wayne State University, Detroit, MI, USA
| |
Collapse
|
5
|
Wang W, Wu Y, Chen S, Liu X, He J, Wang S, Lu W, Tang Y, Huang J. Shikonin is a novel and selective IMPDH2 inhibitor that target triple-negative breast cancer. Phytother Res 2020; 35:463-476. [PMID: 32779300 DOI: 10.1002/ptr.6825] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/17/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022]
Abstract
Triple-negative breast cancer (TNBC) is heterogeneous disease with a poor prognosis. It is therefore important to explore novel therapeutic agents to improve the clinical efficacy for TNBC. The inosine 5'-monophosphate dehydrogenase 2 (IMPDH2) is a rate-limiting enzyme in the de novo synthesis of guanine nucleotides. It is always overexpressed in many types of tumors, including TNBC and regarded as a potential target for cancer therapy. Through screening a library of natural products, we identified shikonin, a natural bioactive component of Lithospermum erythrorhizon, is a novel and selective IMPDH2 inhibitor. Enzymatic analysis using Lineweaver-Burk plot indicates that shikonin is a competitive inhibitor of IMPDH2. The interaction between shikonin and IMDPH2 was further investigated by thermal shift assay, fluorescence quenching, and molecular docking simulation. Shikonin treatment effectively inhibits the growth of human TNBC cell line MDA-MB-231, and murine TNBC cell line, 4T1 in a dose-dependent manner, which is impaired by exogenous supplementation of guanosine, a salvage pathway of purine nucleotides. Most importantly, IMPDH2 knockdown significantly reduced cell proliferation and conferred resistance to shikonin in TNBC. Collectively, our findings showed the natural product shikonin as a selective inhibitor of IMPDH2 with anti-TNBC activity, impelling its further study in clinical trials.
Collapse
Affiliation(s)
- Wanyan Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yu Wu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Si Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xi Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiacheng He
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Shuyi Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yong Tang
- Department of Urology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jin Huang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
6
|
Hsu YJ, Lin CW, Cho SL, Yang WS, Yang CM, Yang CH. Protective Effect of Fenofibrate on Oxidative Stress-Induced Apoptosis in Retinal-Choroidal Vascular Endothelial Cells: Implication for Diabetic Retinopathy Treatment. Antioxidants (Basel) 2020; 9:antiox9080712. [PMID: 32764528 PMCID: PMC7464418 DOI: 10.3390/antiox9080712] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/28/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022] Open
Abstract
Diabetic retinopathy (DR) is an important microvascular complication of diabetes and one of the leading causes of blindness in developed countries. Two large clinical studies showed that fenofibrate, a peroxisome proliferator-activated receptor type α (PPAR-α) agonist, reduces DR progression. We evaluated the protective effects of fenofibrate on retinal/choroidal vascular endothelial cells under oxidative stress and investigated the underlying mechanisms using RF/6A cells as the model system and paraquat (PQ) to induce oxidative stress. Pretreatment with fenofibrate suppressed reactive oxygen species (ROS) production, decreased cellular apoptosis, diminished the changes in the mitochondrial membrane potential, increased the mRNA levels of peroxiredoxin (Prx), thioredoxins (Trxs), B-cell lymphoma 2 (Bcl-2), and Bcl-xl, and reduced the level of B-cell lymphoma 2-associated X protein (Bax) in PQ-stimulated RF/6A cells. Western blot analysis revealed that fenofibrate repressed apoptosis through cytosolic and mitochondrial apoptosis signal-regulated kinase-1 (Ask)-Trx-related signaling pathways, including c-Jun amino-terminal kinase (JNK) phosphorylation, cytochrome c release, caspase 3 activation, and poly (ADP-ribose) polymerase-1 (PARP-1) cleavage. These protective effects of fenofibrate on RF/6A cells may be attributable to its anti-oxidative ability. Our research suggests that fenofibrate could serve as an effective adjunct therapy for ocular oxidative stress-related disorders, such as DR.
Collapse
Affiliation(s)
- Ying-Jung Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 1, Jen Ai Road Section 1, Taipei 100, Taiwan; (Y.-J.H.); (C.-W.L.); (W.-S.Y.)
| | - Chao-Wen Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 1, Jen Ai Road Section 1, Taipei 100, Taiwan; (Y.-J.H.); (C.-W.L.); (W.-S.Y.)
- Department of Ophthalmology, National Taiwan University Hospital, No. 7, Zhongshan South Road, Taipei 100, Taiwan;
| | - Sheng-Li Cho
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Zhongshan South Road, Taipei 100, Taiwan;
| | - Wei-Shiung Yang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 1, Jen Ai Road Section 1, Taipei 100, Taiwan; (Y.-J.H.); (C.-W.L.); (W.-S.Y.)
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Zhongshan South Road, Taipei 100, Taiwan;
| | - Chung-May Yang
- Department of Ophthalmology, National Taiwan University Hospital, No. 7, Zhongshan South Road, Taipei 100, Taiwan;
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, No. 7, Zhongshan South Road, Taipei 100, Taiwan;
- Correspondence: ; Tel.: +886-2-23123456 (ext. 63193)
| |
Collapse
|
7
|
Wang W, Fang D, Zhang H, Xue J, Wangchuk D, Du J, Jiang L. Sodium Butyrate Selectively Kills Cancer Cells and Inhibits Migration in Colorectal Cancer by Targeting Thioredoxin-1. Onco Targets Ther 2020; 13:4691-4704. [PMID: 32547098 PMCID: PMC7263851 DOI: 10.2147/ott.s235575] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/07/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Sodium butyrate (NaB) is a short-chain fatty acid which is produced by bacterial fermentation of nondigestible dietary fiber and has been reported to exert anti-tumor effects in many tumors including colorectal cancer (CRC). However, the role of thioredoxin-1 (Trx-1) in NaB-induced anti-tumor effect has not been completely clarified. MATERIALS AND METHODS Effects of NaB on the growth of CRC cell lines HT29 and SW480 were detected by the Cell Counting Kit-8 (CCK-8) and colony formation assays. The apoptotic cells were determined by flow cytometry, and cell migration was assessed by a Transwell assay. Western blot analysis was used to test the Trx-1 and epithelial-to-mesenchymal transition (EMT)-related proteins level. Reactive oxygen species (ROS) level was determined and N-acetylcysteine (NAC) recovery experiment was performed in CRC cells. In addition, mice xenograft model was established to test the effect of NaB on CRC growth in vivo. Further, the effects of NaB on CRC cells with overexpression or knockdown were tested by the CCK-8 and Transwell assays. RESULTS NaB treatment significantly inhibited cell growth and decreased Trx-1 protein expression in CRC cells but not in normal colon epithelial cells. NaB also induced apoptosis, inhibited colony formation, migration and EMT in CRC cells. Besides, NaB increased ROS level in CRC cells and NAC reversed NaB-induced inhibition of cell proliferation. Moreover, downregulation of Trx-1 significantly enhanced NaB-induced inhibitory effects on cell growth and migration, whereas overexpression of Trx-1 attenuated NaB-induced inhibitory effects on growth and migration in CRC cells. CONCLUSION These findings indicate that the NaB-mediated anti-tumor effects on CRC cells are related to downregulation of Trx-1.
Collapse
Affiliation(s)
- Wenqi Wang
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou325000, People’s Republic of China
- Department of Laboratory Medicine, Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, Shanghai 201318, People’s Republic of China
| | - Daoquan Fang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou325000, People’s Republic of China
| | - Hao Zhang
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou325000, People’s Republic of China
| | - Jiao Xue
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou325000, People’s Republic of China
| | - Drugyel Wangchuk
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou325000, People’s Republic of China
| | - Jimei Du
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou325000, People’s Republic of China
| | - Lei Jiang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou325000, People’s Republic of China
| |
Collapse
|
8
|
Liu C, Cao B, Zhang Q, Zhang Y, Chen X, Kong X, Dong Y. Inhibition of thioredoxin 2 by intracellular methylglyoxal accumulation leads to mitochondrial dysfunction and apoptosis in INS-1 cells. Endocrine 2020; 68:103-115. [PMID: 31939094 DOI: 10.1007/s12020-020-02191-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/05/2020] [Indexed: 01/17/2023]
Abstract
PURPOSE To investigate the role of thioredoxin 2 (Trx2) inhibition induced by intracellular methylglyoxal (MGO) in pancreatic beta-cell mitochondrial dysfunction and apoptosis. METHODS Rat pancreatic beta-cell line INS-1 cells were treated with Glo1 siRNAs or exogenous MGO to increase intracellular MGO. AGEs formation was detected by ELISA and mitochondrial ROS was detected by probe MitoSOX. Transmission electron microscopy (TEM) analysis and ATP content were measured to evaluate mitochondrial function. Trx2 expression was manipulated by overexpression with recombinant Trx2 lentivirus or knockdown with Trx2 siRNAs, and effects on apoptosis and insulin secretion were measured by flow cytometry and ELISA, respectively. RESULTS The increase of intracellular MGO by Glo1 blockage or MGO treatment led to advanced glycation end products (AGEs) overproduction, mitochondrial ROS increase, and insulin secretion paralysis. These were probably due to MGO-induced inhibition of mitochondrial Trx2. Trx2 inhibition by blockage of either Glo1 or Trx2 impaired mitochondrial integrity, inhibited cytochrome C oxidases subunit 1 and 4 (Cox1 and Cox4) expression and further reduced ATP generation, and all of these might lead to insulin paralysis; whereas Trx2 overexpression partially reversed MGO-induced oxidative stress, attenuated insulin secretion by preventing mitochondrial damage. Trx2 overexpression also retarded MGO-induced apoptosis of INS-1 cell through inhibiting ASK1 activation and downregulation of the ASK1-p38 MAPK pathway. CONCLUSIONS Our results reveal a possible mechanism for beta-cell oxidative damage upon intracellular MGO-induced Trx2 inactivation and mitochondrial dysfunction and apoptosis.
Collapse
Affiliation(s)
- Chongxiao Liu
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Department of Endocrinology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Baige Cao
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Qianren Zhang
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yifan Zhang
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xueru Chen
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiang Kong
- Department of Endocrinology, Yijishan Hospital Affiliated Wannan Medical College, Anhui, 241000, China
| | - Yan Dong
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Shanghai Institute for Pediatric Research, Shanghai, 200092, China.
| |
Collapse
|
9
|
Yang L, Guo Y, Huang M, Wu X, Li X, Chen G, Li Y, Bai J. Thioredoxin-1 Protects Spinal Cord from Demyelination Induced by Methamphetamine through Suppressing Endoplasmic Reticulum Stress and Inflammation. Front Neurol 2018; 9:49. [PMID: 29467717 PMCID: PMC5808126 DOI: 10.3389/fneur.2018.00049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/18/2018] [Indexed: 01/19/2023] Open
Abstract
Methamphetamine (METH) is a psychostimulant abused around the world. Emerging evidence indicates that METH causes brain damage. However, there are very few reports on METH-induced demyelination. Thioredoxin-1 (Trx-1) is a redox regulating protein and plays the roles in protecting neurons from various stresses. However, whether Trx-1 resists demyelination induced by METH has not been reported. In this study, we found that METH-induced thin myelin sheaths in spinal cord, whereas Trx-1 overexpression transgenic (TG) mice restored the myelin sheaths thickness. The expressions of myelin-associated glycoprotein, myelin basic protein, and cyclin-dependent kinase 5 were decreased by METH, whereas these alterations were blocked in Trx-1 TG mice. The expressions of procaspase-12 and procaspase-3 were decreased by METH, the expression of calpain1 was increased by METH, whereas the alterations were suppressed in Trx-1 TG mice. As same as, the expressions of the extracellular signal-regulated kinase, nuclear factor κB, tumor necrosis factor-alpha, and interleukin-1beta were induced by METH, which were suppressed in Trx-1 TG mice. These data suggest that Trx-1 may play a critical role in resisting the METH-mediated demyelination in spinal cord through regulating endoplasmic reticulum stress and inflammation pathways.
Collapse
Affiliation(s)
- Lihua Yang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China.,Medical School, Kunming University of Science and Technology, Kunming, China.,Narcotics Control School, Yunnan Police College, Kunming, China
| | - Yinli Guo
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Mengbin Huang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiaoli Wu
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiang Li
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Guobing Chen
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Ye Li
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
10
|
Bianchi-Smiraglia A, Rana MS, Foley CE, Paul LM, Lipchick BC, Moparthy S, Moparthy K, Fink EE, Bagati A, Hurley E, Affronti HC, Bakin AV, Kandel ES, Smiraglia DJ, Feltri ML, Sousa R, Nikiforov MA. Internally ratiometric fluorescent sensors for evaluation of intracellular GTP levels and distribution. Nat Methods 2017; 14:1003-1009. [PMID: 28869758 PMCID: PMC5636219 DOI: 10.1038/nmeth.4404] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023]
Abstract
GTP is a major regulator of multiple cellular processes, but tools for quantitative evaluation of GTP levels in live cells have not been available. We report the development and characterization of genetically encoded GTP sensors, which we constructed by inserting a circularly permuted yellow fluorescent protein (cpYFP) into a region of the bacterial G protein FeoB that undergoes a GTP-driven conformational change. GTP binding to these sensors results in a ratiometric change in their fluorescence, thereby providing an internally normalized response to changes in GTP levels while minimally perturbing those levels. Mutations introduced into FeoB to alter its affinity for GTP created a series of sensors with a wide dynamic range. Critically, in mammalian cells the sensors showed consistent changes in ratiometric signal upon depletion or restoration of GTP pools. We show that these GTP evaluators (GEVALs) are suitable for detection of spatiotemporal changes in GTP levels in living cells and for high-throughput screening of molecules that modulate GTP levels.
Collapse
Affiliation(s)
- Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Mitra S. Rana
- Department of Biochemistry and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Colleen E. Foley
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Leslie M. Paul
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Brittany C. Lipchick
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Sudha Moparthy
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Kalyana Moparthy
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Emily E. Fink
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Archis Bagati
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Edward Hurley
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Hayley C. Affronti
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Andrei V. Bakin
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Eugene S. Kandel
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Dominic J. Smiraglia
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Rui Sousa
- Department of Biochemistry and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Mikhail A. Nikiforov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
11
|
Ren X, Lu H, Wang N, Zhang C, Ji Y, Cui S, Dong Y, Yang K, Du M, Diao F, Kong L. Thioredoxin is implicated in the anti‑apoptotic effects of grape seed proanthocyanidin extract during hyperglycemia. Mol Med Rep 2017; 16:7731-7737. [PMID: 28944891 DOI: 10.3892/mmr.2017.7508] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 07/11/2017] [Indexed: 11/05/2022] Open
Abstract
Diabetic retinopathy has long been recognized as a microvascular disease, however, recent research has indicated that diabetic retinopathy may also be considered a neurodegenerative disease. The elucidation of the molecular mechanisms underlying the development of diabetic retinopathy is imperative for the development of preventive and treatment strategies for patients with diabetes. In the present study, grape seed proanthocyanidin extract (GSPE) was used to upregulate the expression of thioredoxin (Trx), in order to evaluate its potential as a novel agent for the prevention and treatment of neurodegenerative diseases, including diabetic retinopathy. Hematoxylin and eosin staining was performed to observe the morphology of retinal neurons, whereas flow cytometry and terminal deoxynucleotidyl transferase 2'‑deoxyuridine, 5'‑triphosphate nick‑end labeling were employed to investigate cellular apoptosis. Reverse transcription‑quantitative polymerase chain reaction and western blot analysis were performed to assess the mRNA and protein expression of target proteins in order to investigate the underlying molecular mechanisms. In vivo, it was found that the photoreceptor cell was damaged in diabetic mice but following GSPE treatment, the process could be inhibited. In vitro, the results of the current study demonstrated that, under hyperglycemic culture conditions, the expression of 78 kDa glucose‑regulated protein, which is an endoplasmic reticulum stress marker, was upregulated. In addition, the expression of Trx was downregulated and cell apoptosis was enhanced. Notably, treatment with GSPE was revealed to inhibit the neurodegenerative process induced by hyperglycemia. However, treatment with the Trx inhibitor PX12 in combination with GSPE was demonstrated to potentiate apoptosis compared with GSPE treatment alone under hyperglycemic conditions. Furthermore, the protein expression of apoptosis signal‑regulating kinase (ASK) 1 and Trx‑interacting protein (Txnip) was also upregulated by hyperglycemia, whereas GSPE was revealed to counteract this upregulation. In conclusion, the results of the present study indicate that Trx may be implicated in the mechanisms underlying the protective effects of GSPE against hyperglycemia‑induced cell degeneration and apoptosis. The molecular mechanisms may also involve inhibition of the activation of the Trx/ASK1/Txnip signaling pathway.
Collapse
Affiliation(s)
- Xiang Ren
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Heyuan Lu
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Nina Wang
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Chenghong Zhang
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yunpeng Ji
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Shiqi Cui
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yichen Dong
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Kaiyuan Yang
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Mengyi Du
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Fengsheng Diao
- Department of Traditional Chinese Medicine, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Li Kong
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
12
|
Chau GC, Im DU, Kang TM, Bae JM, Kim W, Pyo S, Moon EY, Um SH. mTOR controls ChREBP transcriptional activity and pancreatic β cell survival under diabetic stress. J Cell Biol 2017; 216:2091-2105. [PMID: 28606928 PMCID: PMC5496625 DOI: 10.1083/jcb.201701085] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/05/2017] [Accepted: 05/01/2017] [Indexed: 02/06/2023] Open
Abstract
Through in vivo analyses of mTOR deficiency and in vitro studies of human and mouse pancreatic islets, Chau et al. show that mTOR plays a critical role in β cell survival in diabetes. mTOR associates with and inhibits the transcriptional ChREBP–Mlx complex, suppressing TXNIP expression and β cell death. Impaired nutrient sensing and dysregulated glucose homeostasis are common in diabetes. However, how nutrient-sensitive signaling components control glucose homeostasis and β cell survival under diabetic stress is not well understood. Here, we show that mice lacking the core nutrient-sensitive signaling component mammalian target of rapamycin (mTOR) in β cells exhibit reduced β cell mass and smaller islets. mTOR deficiency leads to a severe reduction in β cell survival and increased mitochondrial oxidative stress in chemical-induced diabetes. Mechanistically, we find that mTOR associates with the carbohydrate-response element–binding protein (ChREBP)–Max-like protein complex and inhibits its transcriptional activity, leading to decreased expression of thioredoxin-interacting protein (TXNIP), a potent inducer of β cell death and oxidative stress. Consistent with this, the levels of TXNIP and ChREBP were highly elevated in human diabetic islets and mTOR-deficient mouse islets. Thus, our results suggest that a nutrient-sensitive mTOR-regulated transcriptional network could be a novel target to improve β cell survival and glucose homeostasis in diabetes.
Collapse
Affiliation(s)
- Gia Cac Chau
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, Korea
| | - Dong Uk Im
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| | - Tong Mook Kang
- Department of Physiology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, Korea .,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
13
|
Bianchi-Smiraglia A, Wawrzyniak JA, Bagati A, Marvin EK, Ackroyd J, Moparthy S, Bshara W, Fink EE, Foley CE, Morozevich GE, Berman AE, Shewach DS, Nikiforov MA. Pharmacological targeting of guanosine monophosphate synthase suppresses melanoma cell invasion and tumorigenicity. Cell Death Differ 2015; 22:1858-64. [PMID: 25909885 PMCID: PMC4648332 DOI: 10.1038/cdd.2015.47] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/12/2015] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
Malignant melanoma possesses one of the highest metastatic potentials among human cancers. Acquisition of invasive phenotypes is a prerequisite for melanoma metastases. Elucidation of the molecular mechanisms underlying melanoma invasion will greatly enhance the design of novel agents for melanoma therapeutic intervention. Here, we report that guanosine monophosphate synthase (GMPS), an enzyme required for the de novo biosynthesis of GMP, has a major role in invasion and tumorigenicity of cells derived from either BRAF(V600E) or NRAS(Q61R) human metastatic melanomas. Moreover, GMPS levels are increased in metastatic human melanoma specimens compared with primary melanomas arguing that GMPS is an attractive candidate for anti-melanoma therapy. Accordingly, for the first time we demonstrate that angustmycin A, a nucleoside-analog inhibitor of GMPS produced by Streptomyces hygroscopius efficiently suppresses melanoma cell invasion in vitro and tumorigenicity in immunocompromised mice. Our data identify GMPS as a powerful driver of melanoma cell invasion and warrant further investigation of angustmycin A as a novel anti-melanoma agent.
Collapse
Affiliation(s)
- A Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - J A Wawrzyniak
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - A Bagati
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - E K Marvin
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - J Ackroyd
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - S Moparthy
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - W Bshara
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - E E Fink
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - C E Foley
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - G E Morozevich
- Orekhovich Institute of Biomedical Chemistry, Moscow 119121, Russia
| | - A E Berman
- Orekhovich Institute of Biomedical Chemistry, Moscow 119121, Russia
| | - D S Shewach
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - M A Nikiforov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
14
|
Gu L, Gao W, Yang HM, Wang BB, Wang XN, Xu J, Zhang H. Control of Trx1 redox state modulates protection against methyl methanesulfonate-induced DNA damage via stabilization of p21. J Biochem 2015; 159:101-10. [PMID: 26276860 DOI: 10.1093/jb/mvv080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 07/02/2015] [Indexed: 12/13/2022] Open
Abstract
Thioredoxin 1 (Trx1) is known to play an important role in protecting against cell death. However, the mechanism for control of Trx1 in cell death resulting from DNA damage has not been fully investigated. In this study, we used the DNA-damaging agent methyl methanesulfonate (MMS) to investigate the protective effects of Trx1 against DNA damage and cell death in HEK293 cells. We found that MMS application caused dose-dependent changes in the Trx1 redox state determined by redox western blotting. At lower concentrations, both reduced and oxidized Trx1 were observed, whereas the reduced band was fully oxidized at the higher concentration. Trx1 overexpression and small interfering RNA knockdown in cells revealed that reduced Trx1 after exposure to lower doses of MMS attenuated DNA damage, assessed by comet assay, and level of the DNA-damage marker histone γ-H2AX, possibly through scavenging intracellular ROS and an increase in p21 protein level via enhancing its stability. However, oxidized Trx1 lost its protective ability to DNA damage in response to higher concentration of MMS. Corresponding to the redox state control of Trx1, cell death induced by different dose of MMS was also found, by inhibiting phosphorylations of p38 and 4E-BP1. These results indicate that reduced Trx1 plays important protective roles against MMS-induced DNA damage and cell death, suggesting that cell protection is regulated by the intracellular redox state. Control of the redox state of Trx1 and its regulating proteins may offer a novel therapeutic strategy for the control of cancer.
Collapse
Affiliation(s)
- Li Gu
- Department of Neurobiology, Capital Medical University, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorder, Ministry of Education
| | - Wei Gao
- Department of Neurobiology, Capital Medical University, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorder, Ministry of Education
| | - Hui Min Yang
- Department of Neurobiology, Capital Medical University, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorder, Ministry of Education
| | - Bei Bei Wang
- School of Basic Medical Science, Capital Medical University, Beijing 100069 and
| | - Xiao Na Wang
- School of Basic Medical Science, Capital Medical University, Beijing 100069 and
| | - Jianguo Xu
- Department of Internal Medicine, Shaoxing Second Hospital, Zhejiang 312000, China
| | - Hong Zhang
- Department of Neurobiology, Capital Medical University, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorder, Ministry of Education,
| |
Collapse
|
15
|
Abstract
Thioredoxin (Trx) is an inflammation-inducible small oxidoreductase protein ubiquitously expressed in all organisms. Trx acts both intracellularly and extracellularly and is involved in a wide range of physiological cellular responses. Inside the cell, Trx alleviates oxidative stress by scavenging reactive oxygen species (ROS), regulates a variety of redox-sensitive signaling pathways as well as ROS-independent genes, and exerts cytoprotective effects. Outside the cell, Trx acts as growth factors or cytokines and promotes cell growth and many other cellular responses. Trx is also implicated in tumorigenesis. Trx is a proto-oncogene and is overexpressed in many cancers and correlates with poor prognosis. Trx stimulates cancer cell survival, promotes tumor angiogenesis, and inhibits both spontaneous apoptosis and drug-induced apoptosis. Inhibitors targeting Trx pathway provide a promising therapeutic strategy for cancer prevention and intervention. More recently, data from our laboratory demonstrate an important role of Trx in expanding long-term repopulating hematopoietic stem cells. In this chapter, we first provide an overview of Trx including its isoforms, compartmentation, and functions. We then discuss the roles of Trx in hematologic malignancies. Finally, we summarize the most recent findings from our lab on the use of Trx to enhance hematopoietic reconstitution following hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Ningfei An
- Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yubin Kang
- Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA; Current address: Division of Hematologic Malignancy and Cellular Therapy/Adult BMT, Department of Medicine, Duke University Medical Center, North Carolina, USA.
| |
Collapse
|