1
|
Wang YW, Chu T, Wang XL, Fan YQ, Cao L, Chen YH, Zhu YW, Liu HX, Ji XY, Wu DD. The role of cystathionine β-synthase in cancer. Cell Signal 2024; 124:111406. [PMID: 39270916 DOI: 10.1016/j.cellsig.2024.111406] [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: 07/12/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Cystathionine β-synthase (CBS) occupies a key position as the initiating and rate-limiting enzyme in the sulfur transfer pathway and plays a vital role in health and disease. CBS is responsible for regulating the metabolism of cysteine, the precursor of glutathione (GSH), an important antioxidant in the body. Additionally, CBS is one of the three enzymes that produce hydrogen sulfide (H2S) in mammals through a variety of mechanisms. The dysregulation of CBS expression in cancer cells affects H2S production through direct or indirect pathways, thereby influencing cancer growth and metastasis by inducing angiogenesis, facilitating proliferation, migration, and invasion, modulating cellular energy metabolism, promoting cell cycle progression, and inhibiting apoptosis. It is noteworthy that CBS expression exhibits complex changes in different cancer models. In this paper, we focus on the CBS synthesis and metabolism, tissue distribution, potential mechanisms influencing tumor growth, and relevant signaling pathways. We also discuss the impact of pharmacological CBS inhibitors and silencing CBS in preclinical cancer models, supporting their potential as targeted cancer therapies.
Collapse
Affiliation(s)
- Yan-Wen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Xue-Li Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yong-Qi Fan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Lei Cao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yu-Hang Chen
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Hong-Xia Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan 450064, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
2
|
Feng H, Sun Z, Han B, Xia H, Chen L, Tian C, Yan S, Shi Y, Yin J, Song W, Gong P, Wang S, Li Y. Miro2 sulfhydration by CBS/H 2S promotes human trophoblast invasion and migration via regulating mitochondria dynamics. Cell Death Dis 2024; 15:776. [PMID: 39461943 PMCID: PMC11513031 DOI: 10.1038/s41419-024-07167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Insufficient cytotrophoblast (CTB) migration and invasion into the maternal myometrium leads to pregnancy related complications like Intra-uterus Growth Restriction (IUGR), and pre-eclampsia (PE). We previously found that hydrogen sulfide (H2S) enhanced CTB migration without knowing the mechanism(s) and the pathophysiological significance. By studying human samples and cell line, we found that H2S levels were lower in PE patients' plasma; H2S synthetic enzyme cystathionine β-synthetase (CBS) was reduced in PE extravillious invasive trophoblasts. GYY4137 (H2S donor, 1 µM) promoted CBS/H2S translocation onto mitochondria, preserved mitochondria functions, enhanced cell invasion and migration. CBS knockdown hindered the above functions which were rescued by GYY4137, indicating the vital roles of CBS/H2S signal. Disturbance of mitochondria dynamics inhibited cell invasion and migration. The 185 and 504 cysteines of Mitochondrial Rho GTPase 2 (Miro2C185/C504) were highly sulfhydrated by H2S. Knockdown Miro2 or double mutation of Miro2C185/C504 to serine fragmented mitochondria, and inhibited cell invasion and migration which can't be rescued by H2S. The present study showed that human cytotrophoblast receives low dose H2S regulation; CBS/H2S sustained mitochondria functions via Miro2C185/C504 sulfhydration to enhance cytotrophoblast mobility. These findings established a new regulatory pathway for cytotrophoblast functions, and provided new targets for IUGR and PE.
Collapse
Affiliation(s)
- Hao Feng
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Zongxin Sun
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
- Department of Emergency, Affiliated Hospital of Chifeng University, Shifeng, 024001, China
| | - Baoshi Han
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
| | - Huitang Xia
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong, 250014, PR China
| | - Lumei Chen
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Chunlei Tian
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Suhua Yan
- Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Yugen Shi
- Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Jie Yin
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong, 250014, PR China
| | - Wengang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250012, China
| | - Peipei Gong
- Department of Rehabilitation Medicine, the Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Shuanglian Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250012, China.
| | - Yan Li
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250012, China.
- Translational Medical Research Centre, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China.
| |
Collapse
|
3
|
Shahid A, Bhatia M. Hydrogen Sulfide: A Versatile Molecule and Therapeutic Target in Health and Diseases. Biomolecules 2024; 14:1145. [PMID: 39334911 PMCID: PMC11430449 DOI: 10.3390/biom14091145] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
In recent years, research has unveiled the significant role of hydrogen sulfide (H2S) in many physiological and pathological processes. The role of endogenous H2S, H2S donors, and inhibitors has been the subject of studies that have aimed to investigate this intriguing molecule. The mechanisms by which H2S contributes to different diseases, including inflammatory conditions, cardiovascular disease, viral infections, and neurological disorders, are complex. Despite noteworthy progress, several questions remain unanswered. H2S donors and inhibitors have shown significant therapeutic potential for various diseases. This review summarizes our current understanding of H2S-based therapeutics in inflammatory conditions, cardiovascular diseases, viral infections, and neurological disorders.
Collapse
Affiliation(s)
- Aqsa Shahid
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand
| |
Collapse
|
4
|
Ju W, Zhao Y, Yu Y, Zhao S, Xiang S, Lian F. Mechanisms of mitochondrial dysfunction in ovarian aging and potential interventions. Front Endocrinol (Lausanne) 2024; 15:1361289. [PMID: 38694941 PMCID: PMC11061492 DOI: 10.3389/fendo.2024.1361289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/22/2024] [Indexed: 05/04/2024] Open
Abstract
Mitochondria plays an essential role in regulating cellular metabolic homeostasis, proliferation/differentiation, and cell death. Mitochondrial dysfunction is implicated in many age-related pathologies. Evidence supports that the dysfunction of mitochondria and the decline of mitochondrial DNA copy number negatively affect ovarian aging. However, the mechanism of ovarian aging is still unclear. Treatment methods, including antioxidant applications, mitochondrial transplantation, emerging biomaterials, and advanced technologies, are being used to improve mitochondrial function and restore oocyte quality. This article reviews key evidence and research updates on mitochondrial damage in the pathogenesis of ovarian aging, emphasizing that mitochondrial damage may accelerate and lead to cellular senescence and ovarian aging, as well as exploring potential methods for using mitochondrial mechanisms to slow down aging and improve oocyte quality.
Collapse
Affiliation(s)
- Wenhan Ju
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuewen Zhao
- CReATe Fertility Centre, Toronto, ON, Canada
| | - Yi Yu
- Department of Reproduction and Genetics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuai Zhao
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shan Xiang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fang Lian
- Department of Reproduction and Genetics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Dey A, Pramanik PK, Dwivedi SKD, Neizer-Ashun F, Kiss T, Ganguly A, Rice H, Mukherjee P, Xu C, Ahmad M, Csiszar A, Bhattacharya R. A role for the cystathionine-β-synthase /H 2S axis in astrocyte dysfunction in the aging brain. Redox Biol 2023; 68:102958. [PMID: 37948927 PMCID: PMC10663824 DOI: 10.1016/j.redox.2023.102958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
Astrocytic dysfunction is central to age-related neurodegenerative diseases. However, the mechanisms leading to astrocytic dysfunction are not well understood. We identify that among the diverse cellular constituents of the brain, murine and human astrocytes are enriched in the expression of CBS. Depleting CBS in astrocytes causes mitochondrial dysfunction, increases the production of reactive oxygen species (ROS) and decreases cellular bioenergetics that can be partially rescued by exogenous H2S supplementation or by re-expressing CBS. Conversely, the CBS/H2S axis, associated protein persulfidation and proliferation are decreased in astrocytes upon oxidative stress which can be rescued by exogenous H2S supplementation. Here we reveal that in the aging brain, the CBS/H2S axis is downregulated leading to decreased protein persulfidation, together augmenting oxidative stress. Our findings uncover an important protective role of the CBS/H2S axis in astrocytes that may be disrupted in the aged brain.
Collapse
Affiliation(s)
- Anindya Dey
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Pijush Kanti Pramanik
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Fiifi Neizer-Ashun
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Abhrajit Ganguly
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Heather Rice
- Department of Biochemistry & Molecular Biology, Oklahoma Center for Geroscience & Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mohiuddin Ahmad
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
6
|
Chen H, Li K, Qin Y, Zhou J, Li T, Qian L, Yang C, Ji X, Wu D. Recent advances in the role of endogenous hydrogen sulphide in cancer cells. Cell Prolif 2023; 56:e13449. [PMID: 36929586 PMCID: PMC10472536 DOI: 10.1111/cpr.13449] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/16/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Hydrogen sulphide (H2 S) is a gaseous neurotransmitter that can be self-synthesized by living organisms. With the deepening of research, the pathophysiological mechanisms of endogenous H2 S in cancer have been increasingly elucidated: (1) promote angiogenesis, (2) stimulate cell bioenergetics, (3) promote migration and proliferation thereby invasion, (4) inhibit apoptosis and (5) activate abnormal cell cycle. However, the increasing H2 S levels via exogenous sources show the opposite trend. This phenomenon can be explained by the bell-shaped pharmacological model of H2 S, that is, the production of endogenous (low concentration) H2 S promotes tumour growth while the exogenous (high concentration) H2 S inhibits tumour growth. Here, we review the impact of endogenous H2 S synthesis and metabolism on tumour progression, summarize the mechanism of action of H2 S in tumour growth, and discuss the possibility of H2 S as a potential target for tumour treatment.
Collapse
Affiliation(s)
- Hao‐Jie Chen
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
| | - Ke Li
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
| | - Yang‐Zhe Qin
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
| | - Jing‐Jing Zhou
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
| | - Tao Li
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
| | - Lei Qian
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
| | - Chang‐Yong Yang
- School of Nursing and HealthHenan UniversityKaifengHenan475004China
| | - Xin‐Ying Ji
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
| | - Dong‐Dong Wu
- School of Basic Medical SciencesHenan UniversityKaifengHenan475004China
- Henan International Joint Laboratory for Nuclear Protein RegulationHenan UniversityKaifengHenan475004China
- School of StomatologyHenan UniversityKaifengHenan475004China
| |
Collapse
|
7
|
Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol 2023; 20:109-125. [PMID: 35931887 PMCID: PMC9362470 DOI: 10.1038/s41569-022-00741-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 01/21/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as a gaseous signalling molecule with crucial implications for cardiovascular health. H2S is involved in many biological functions, including interactions with nitric oxide, activation of molecular signalling cascades, post-translational modifications and redox regulation. Various preclinical and clinical studies have shown that H2S and its synthesizing enzymes - cystathionine γ-lyase, cystathionine β-synthase and 3-mercaptosulfotransferase - can protect against cardiovascular pathologies, including arrhythmias, atherosclerosis, heart failure, myocardial infarction and ischaemia-reperfusion injury. The bioavailability of H2S and its metabolites, such as hydropersulfides and polysulfides, is substantially reduced in cardiovascular disease and has been associated with single-nucleotide polymorphisms in H2S synthesis enzymes. In this Review, we highlight the role of H2S, its synthesizing enzymes and metabolites, their roles in the cardiovascular system, and their involvement in cardiovascular disease and associated pathologies. We also discuss the latest clinical findings from the field and outline areas for future study.
Collapse
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Paari Dominic
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
8
|
De Rasmo D, Cormio A, Cormio G, Signorile A. Ovarian Cancer: A Landscape of Mitochondria with Emphasis on Mitochondrial Dynamics. Int J Mol Sci 2023; 24:ijms24021224. [PMID: 36674740 PMCID: PMC9865899 DOI: 10.3390/ijms24021224] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Ovarian cancer (OC) represents the main cause of death from gynecological malignancies in western countries. Altered cellular and mitochondrial metabolism are considered hallmarks in cancer disease. Several mitochondrial aspects have been found altered in OC, such as the oxidative phosphorylation system, oxidative stress and mitochondrial dynamics. Mitochondrial dynamics includes cristae remodeling, fusion, and fission processes forming a dynamic mitochondrial network. Alteration of mitochondrial dynamics is associated with metabolic change in tumour development and, in particular, the mitochondrial shaping proteins appear also to be responsible for the chemosensitivity and/or chemoresistance in OC. In this review a focus on the mitochondrial dynamics in OC cells is presented.
Collapse
Affiliation(s)
- Domenico De Rasmo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), 70124 Bari, Italy
| | - Antonella Cormio
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Gennaro Cormio
- IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Anna Signorile
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124 Bari, Italy
- Correspondence:
| |
Collapse
|
9
|
Zhang Q, Gao Y, Zhang Y, Jing M, Wang D, Wang Y, Khattak S, Qi H, Cai C, Zhang J, Ngowi EE, Khan NH, Li T, Ji A, Jiang Q, Ji X, Li Y, Wu D. Cystathionine γ-lyase mediates cell proliferation, migration, and invasion of nasopharyngeal carcinoma. Oncogene 2022; 41:5238-5252. [PMID: 36310322 DOI: 10.1038/s41388-022-02512-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelia-derived malignancy with a distinctive geographic distribution. Cystathionine γ-lyase (CSE) is involved in cancer development and progression. Nevertheless, the role of CSE in the growth of NPC is unknown. In this study, we found that CSE levels in human NPC cells were higher than those in normal nasopharyngeal cells. CSE overexpression enhanced the proliferative, migrative, and invasive abilities of NPC cells and CSE downregulation exerted reverse effects. Overexpression of CSE decreased the expressions of cytochrome C, cleaved caspase (cas)-3, cleaved cas-9, and cleaved poly-ADP-ribose polymerase, whereas CSE knockdown exhibited reverse effects. CSE overexpression decreased reactive oxygen species (ROS) levels and the expressions of phospho (p)-extracellular signal-regulated protein kinase 1/2, p-c-Jun N-terminal kinase, and p-p38, but promoted the expressions of p-phosphatidylinositol 3-kinase (PI3K), p-AKT, and p-mammalian target of rapamycin (mTOR), whereas CSE knockdown showed oppose effects. In addition, CSE overexpression promoted NPC xenograft tumor growth and CSE knockdown decreased tumor growth by modulating proliferation, angiogenesis, cell cycle, and apoptosis. Furthermore, DL-propargylglycine (an inhibitor of CSE) dose-dependently inhibited NPC cell growth via ROS-mediated mitogen-activated protein kinase (MAPK) and PI3K/AKT/mTOR pathways without significant toxicity. In conclusion, CSE could regulate the growth of NPC cells through ROS-mediated MAPK and PI3K/AKT/mTOR cascades. CSE might be a novel tumor marker for the diagnosis and prognosis of NPC. Novel donors/drugs that inhibit the expression/activity of CSE can be developed in the treatment of NPC.
Collapse
Affiliation(s)
- Qianqian Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Yingran Gao
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Yanxia Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Mirong Jing
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Di Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Yizhen Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China.,School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Huiwen Qi
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Chunbo Cai
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Jing Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, 475004, China.,Department of Biological Sciences, Faculty of Science, Dar es Salaam University College of Education, Dar es Salaam, 2329, Tanzania
| | - Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China.,School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Tao Li
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Ailing Ji
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Qiying Jiang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China
| | - Xinying Ji
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China. .,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China. .,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, 475004, China.
| | - Yanzhang Li
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China. .,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China.
| | - Dongdong Wu
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China. .,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan, 475004, China. .,School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
10
|
Shiau JP, Chuang YT, Tang JY, Yang KH, Chang FR, Hou MF, Yen CY, Chang HW. The Impact of Oxidative Stress and AKT Pathway on Cancer Cell Functions and Its Application to Natural Products. Antioxidants (Basel) 2022; 11:1845. [PMID: 36139919 PMCID: PMC9495789 DOI: 10.3390/antiox11091845] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress and AKT serine-threonine kinase (AKT) are responsible for regulating several cell functions of cancer cells. Several natural products modulate both oxidative stress and AKT for anticancer effects. However, the impact of natural product-modulating oxidative stress and AKT on cell functions lacks systemic understanding. Notably, the contribution of regulating cell functions by AKT downstream effectors is not yet well integrated. This review explores the role of oxidative stress and AKT pathway (AKT/AKT effectors) on ten cell functions, including apoptosis, autophagy, endoplasmic reticulum stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, migration, and cell-cycle progression. The impact of oxidative stress and AKT are connected to these cell functions through cell function mediators. Moreover, the AKT effectors related to cell functions are integrated. Based on this rationale, natural products with the modulating abilities for oxidative stress and AKT pathway exhibit the potential to regulate these cell functions, but some were rarely reported, particularly for AKT effectors. This review sheds light on understanding the roles of oxidative stress and AKT pathway in regulating cell functions, providing future directions for natural products in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
| | - Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
11
|
Panza E, Bello I, Smimmo M, Brancaleone V, Mitidieri E, Bucci M, Cirino G, Sorrentino R, D Emmanuele di Villa Bianca R. Endogenous and exogenous hydrogen sulfide modulates urothelial bladder carcinoma development in human cell lines. Biomed Pharmacother 2022; 151:113137. [PMID: 35605291 DOI: 10.1016/j.biopha.2022.113137] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/15/2022] [Indexed: 11/25/2022] Open
Abstract
The role of H2S in urothelial carcinoma (UC) is still unclear. Here we have evaluated the expression of H2S producing enzymes as well as the effect of endogenous and exogenous H2S on human bladder UC cells. In human UC cells the expression of cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3-MST); is significantly lower as compared to healthy cells. A modulatory role for the H2S pathway is supported by the finding that, the overexpression of CSE or CBS, but not 3-MST, inhibits cell proliferation and promotes apoptosis. A similar effect is obtained by using exogenous H2S. Diallyl trisulfide (DATS), which is a fully characterized H2S donor, inhibits the proliferation of UC cells in a time and concentration-dependent manner as well as promotes apoptosis. Moreover, DATS also induces autophagy, as determined by transcriptomic and western blot analysis. Finally, DATS inhibits mRNA expression levels of canonical markers of epithelial-mesenchymal transition by limiting migration and clonogenic ability of human UC cells in vitro. In conclusion, in urothelial carcinoma, there is an impairment of H2S pathway that involves CSE and CBS- derived hydrogen sulfide. Thus, targeting H2S signaling pathway in urothelial carcinoma could represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - Ivana Bello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Martina Smimmo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples, Federico II, Italy
| | | |
Collapse
|
12
|
Khattak S, Rauf MA, Khan NH, Zhang QQ, Chen HJ, Muhammad P, Ansari MA, Alomary MN, Jahangir M, Zhang CY, Ji XY, Wu DD. Hydrogen Sulfide Biology and Its Role in Cancer. Molecules 2022; 27:3389. [PMID: 35684331 PMCID: PMC9181954 DOI: 10.3390/molecules27113389] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 02/07/2023] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous biologically active gas produced in mammalian tissues. It plays a very critical role in many pathophysiological processes in the body. It can be endogenously produced through many enzymes analogous to the cysteine family, while the exogenous source may involve inorganic sulfide salts. H2S has recently been well investigated with regard to the onset of various carcinogenic diseases such as lung, breast, ovaries, colon cancer, and neurodegenerative disorders. H2S is considered an oncogenic gas, and a potential therapeutic target for treating and diagnosing cancers, due to its role in mediating the development of tumorigenesis. Here in this review, an in-detail up-to-date explanation of the potential role of H2S in different malignancies has been reported. The study summarizes the synthesis of H2S, its roles, signaling routes, expressions, and H2S release in various malignancies. Considering the critical importance of this active biological molecule, we believe this review in this esteemed journal will highlight the oncogenic role of H2S in the scientific community.
Collapse
Affiliation(s)
- Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (S.K.); (N.H.K.); (Q.-Q.Z.); (H.-J.C.)
| | - Mohd Ahmar Rauf
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (S.K.); (N.H.K.); (Q.-Q.Z.); (H.-J.C.)
| | - Qian-Qian Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (S.K.); (N.H.K.); (Q.-Q.Z.); (H.-J.C.)
| | - Hao-Jie Chen
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (S.K.); (N.H.K.); (Q.-Q.Z.); (H.-J.C.)
| | - Pir Muhammad
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng 475004, China;
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Mohammad N. Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia;
| | - Muhammad Jahangir
- Department of Psychiatric and Mental Health, Central South University, Changsha 410078, China;
| | - Chun-Yang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Department of General Thoracic Surgery, Hami Central Hospital, Hami 839000, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (S.K.); (N.H.K.); (Q.-Q.Z.); (H.-J.C.)
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (S.K.); (N.H.K.); (Q.-Q.Z.); (H.-J.C.)
- School of Stomatology, Henan University, Kaifeng 475004, China
| |
Collapse
|
13
|
Ascenção K, Szabo C. Emerging roles of cystathionine β-synthase in various forms of cancer. Redox Biol 2022; 53:102331. [PMID: 35618601 PMCID: PMC9168780 DOI: 10.1016/j.redox.2022.102331] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
The expression of the reverse transsulfuration enzyme cystathionine-β-synthase (CBS) is markedly increased in many forms of cancer, including colorectal, ovarian, lung, breast and kidney, while in other cancers (liver cancer and glioma) it becomes downregulated. According to the clinical database data in high-CBS-expressor cancers (e.g. colon or ovarian cancer), high CBS expression typically predicts lower survival, while in the low-CBS-expressor cancers (e.g. liver cancer), low CBS expression is associated with lower survival. In the high-CBS expressing tumor cells, CBS, and its product hydrogen sulfide (H2S) serves as a bioenergetic, proliferative, cytoprotective and stemness factor; it also supports angiogenesis and epithelial-to-mesenchymal transition in the cancer microenvironment. The current article reviews the various tumor-cell-supporting roles of the CBS/H2S axis in high-CBS expressor cancers and overviews the anticancer effects of CBS silencing and pharmacological CBS inhibition in various cancer models in vitro and in vivo; it also outlines potential approaches for biomarker identification, to support future targeted cancer therapies based on pharmacological CBS inhibition.
Collapse
|
14
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
15
|
The CBS-H 2S axis promotes liver metastasis of colon cancer by upregulating VEGF through AP-1 activation. Br J Cancer 2022; 126:1055-1066. [PMID: 34952931 PMCID: PMC8979992 DOI: 10.1038/s41416-021-01681-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/04/2021] [Accepted: 12/16/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The main therapy for colon cancer with liver metastasis is chemotherapy based on 5-fluorouracil combined with targeted drugs. However, acquired drug resistance and severe adverse reactions limit patients' benefit from standard chemotherapy. Here, we investigate the involvement of endogenous hydrogen sulfide (H2S) in liver metastasis of colon cancer and its potential value as a novel therapeutic target. METHODS We used the CRISPR/Cas9 system to knockdown CBS gene expression in colon cancer cell lines. PCR arrays and proteome arrays were applied to detect the transcription and protein expression levels, respectively, of angiogenesis-related genes after knockdown. The molecular mechanism was investigated by western blot analysis, RT-qPCR, immunofluorescence staining, ChIP assays and dual-luciferase reporter assays. A liver metastasis mouse model was adopted to investigate the effect of targeting CBS on tumour metastasis in vivo. RESULTS Knockdown of CBS decreased the metastasis and invasion of colon cancer cells and inhibited angiogenesis both in vivo and in vitro. Tissue microarray analysis showed a positive correlation between CBS and VEGF expression in colon cancer tissues. Further analysis at the molecular level validated a positive feedback loop between the CBS-H2S axis and VEGF. CONCLUSIONS Endogenous H2S promotes angiogenesis and metastasis in colon cancer, and targeting the positive feedback loop between the CBS-H2S axis and VEGF can effectively intervene in liver metastasis of colon cancer.
Collapse
|
16
|
Shackelford RE, Li Y, Ghali GE, Kevil CG. Bad Smells and Broken DNA: A Tale of Sulfur-Nucleic Acid Cooperation. Antioxidants (Basel) 2021; 10:1820. [PMID: 34829691 PMCID: PMC8614844 DOI: 10.3390/antiox10111820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 12/19/2022] Open
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter that exerts numerous physiologic and pathophysiologic effects. Recently, a role for H2S in DNA repair has been identified, where H2S modulates cell cycle checkpoint responses, the DNA damage response (DDR), and mitochondrial and nuclear genomic stability. In addition, several DNA repair proteins modulate cellular H2S concentrations and cellular sulfur metabolism and, in turn, are regulated by cellular H2S concentrations. Many DDR proteins are now pharmacologically inhibited in targeted cancer therapies. As H2S and the enzymes that synthesize it are increased in many human malignancies, it is likely that H2S synthesis inhibition by these therapies is an underappreciated aspect of these cancer treatments. Moreover, both H2S and DDR protein activities in cancer and cardiovascular diseases are becoming increasingly apparent, implicating a DDR-H2S signaling axis in these pathophysiologic processes. Taken together, H2S and DNA repair likely play a central and presently poorly understood role in both normal cellular function and a wide array of human pathophysiologic processes. Here, we review the role of H2S in DNA repair.
Collapse
Affiliation(s)
- Rodney E. Shackelford
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA; (Y.L.); (C.G.K.)
| | - Yan Li
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA; (Y.L.); (C.G.K.)
| | - Ghali E. Ghali
- Head & Neck Oncologic/Microvascular Reconstructive Surgery Department of Oral & Maxillofacial/Head & Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA;
| | - Christopher G. Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA; (Y.L.); (C.G.K.)
| |
Collapse
|
17
|
Molecular Functions of Hydrogen Sulfide in Cancer. PATHOPHYSIOLOGY 2021; 28:437-456. [PMID: 35366284 PMCID: PMC8830448 DOI: 10.3390/pathophysiology28030028] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 12/30/2022] Open
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter that exerts a multitude of functions in both physiologic and pathophysiologic processes. H2S-synthesizing enzymes are increased in a variety of human malignancies, including colon, prostate, breast, renal, urothelial, ovarian, oral squamous cell, and thyroid cancers. In cancer, H2S promotes tumor growth, cellular and mitochondrial bioenergetics, migration, invasion, angiogenesis, tumor blood flow, metastasis, epithelia–mesenchymal transition, DNA repair, protein sulfhydration, and chemotherapy resistance Additionally, in some malignancies, increased H2S-synthesizing enzyme expression correlates with a worse prognosis and a higher tumor stage. Here we review the role of H2S in cancer, with an emphasis on the molecular mechanisms by which H2S promotes cancer development, progression, dedifferentiation, and metastasis.
Collapse
|
18
|
Wang T, Ashrafi A, Modareszadeh P, Deese AR, Chacon Castro MDC, Alemi PS, Zhang L. An Analysis of the Multifaceted Roles of Heme in the Pathogenesis of Cancer and Related Diseases. Cancers (Basel) 2021; 13:4142. [PMID: 34439295 PMCID: PMC8393563 DOI: 10.3390/cancers13164142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Heme is an essential prosthetic group in proteins and enzymes involved in oxygen utilization and metabolism. Heme also plays versatile and fascinating roles in regulating fundamental biological processes, ranging from aerobic respiration to drug metabolism. Increasing experimental and epidemiological data have shown that altered heme homeostasis accelerates the development and progression of common diseases, including various cancers, diabetes, vascular diseases, and Alzheimer's disease. The effects of heme on the pathogenesis of these diseases may be mediated via its action on various cellular signaling and regulatory proteins, as well as its function in cellular bioenergetics, specifically, oxidative phosphorylation (OXPHOS). Elevated heme levels in cancer cells intensify OXPHOS, leading to higher ATP generation and fueling tumorigenic functions. In contrast, lowered heme levels in neurons may reduce OXPHOS, leading to defects in bioenergetics and causing neurological deficits. Further, heme has been shown to modulate the activities of diverse cellular proteins influencing disease pathogenesis. These include BTB and CNC homology 1 (BACH1), tumor suppressor P53 protein, progesterone receptor membrane component 1 protein (PGRMC1), cystathionine-β-synthase (CBS), soluble guanylate cyclase (sGC), and nitric oxide synthases (NOS). This review provides an in-depth analysis of heme function in influencing diverse molecular and cellular processes germane to disease pathogenesis and the modes by which heme modulates the activities of cellular proteins involved in the development of cancer and other common diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (T.W.); (A.A.); (P.M.); (A.R.D.); (M.D.C.C.C.); (P.S.A.)
| |
Collapse
|
19
|
Nunes SC, Ramos C, Santos I, Mendes C, Silva F, Vicente JB, Pereira SA, Félix A, Gonçalves LG, Serpa J. Cysteine Boosts Fitness Under Hypoxia-Mimicked Conditions in Ovarian Cancer by Metabolic Reprogramming. Front Cell Dev Biol 2021; 9:722412. [PMID: 34458274 PMCID: PMC8386479 DOI: 10.3389/fcell.2021.722412] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/13/2021] [Indexed: 01/20/2023] Open
Abstract
Among gynecologic malignancies, ovarian cancer is the third most prevalent and the most common cause of death, especially due to diagnosis at an advanced stage together with resistance to therapy. As a solid tumor grows, cancer cells in the microenvironment are exposed to regions of hypoxia, a selective pressure prompting tumor progression and chemoresistance. We have previously shown that cysteine contributes to the adaptation to this hypoxic microenvironment, but the mechanisms by which cysteine protects ovarian cancer cells from hypoxia-induced death are still to be unveiled. Herein, we hypothesized that cysteine contribution relies on cellular metabolism reprogramming and energy production, being cysteine itself a metabolic source. Our results strongly supported a role of xCT symporter in energy production that requires cysteine metabolism instead of hydrogen sulfide (H2S) per se. Cysteine degradation depends on the action of the H2S-synthesizing enzymes cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and/or 3-mercaptopyruvate sulfurtransferase (MpST; together with cysteine aminotransferase, CAT). In normoxia, CBS and CSE inhibition had a mild impact on cysteine-sustained ATP production, pointing out the relevance of CAT + MpST pathway. However, in hypoxia, the concomitant inhibition of CBS and CSE had a stronger impact on ATP synthesis, thus also supporting a role of their hydrogen sulfide and/or cysteine persulfide-synthesizing activity in this stressful condition. However, the relative contributions of each of these enzymes (CBS/CSE/MpST) on cysteine-derived ATP synthesis under hypoxia remains unclear, due to the lack of specific inhibitors. Strikingly, NMR analysis strongly supported a role of cysteine in the whole cellular metabolism rewiring under hypoxia. Additionally, the use of cysteine to supply biosynthesis and bioenergetics was reinforced, bringing cysteine to the plateau of a main carbon sources in cancer. Collectively, this work supports that sulfur and carbon metabolism reprogramming underlies the adaptation to hypoxic microenvironment promoted by cysteine in ovarian cancer.
Collapse
Affiliation(s)
- Sofia C. Nunes
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Cristiano Ramos
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Inês Santos
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Cindy Mendes
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Fernanda Silva
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - João B. Vicente
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Sofia A. Pereira
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ana Félix
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Luís G. Gonçalves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Jacinta Serpa
- Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| |
Collapse
|
20
|
Roth HE, Bhinderwala F, Franco R, Zhou Y, Powers R. DNAJA1 Dysregulates Metabolism Promoting an Antiapoptotic Phenotype in Pancreatic Ductal Adenocarcinoma. J Proteome Res 2021; 20:3925-3939. [PMID: 34264680 DOI: 10.1021/acs.jproteome.1c00233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The cochaperone protein DNAJA1 (HSP40) is downregulated four-fold in pancreatic cancer cells. The impact of DNAJA1 expression on pancreatic ductal adenocarcinoma (PDAC) progression remains unclear. The metabolic impacts of increased DNAJA1 expression were evaluated using a combination of untargeted metabolomics, stable isotope-resolved metabolomics (SIRM), confocal microscopy, flow cytometry, and cell-based assays. Differential Warburg glycolysis, an increase in redox currency, and alterations in amino acid levels were observed in both overexpression cell lines. DNAJA1 overexpression also led to mitochondrial fusion, an increase in the expression of Bcl-2, a modest protection from redox-induced cell death, a loss of structural integrity due to the loss of actin fibers, and an increase in cell invasiveness in BxPC-3. These differences were more pronounced in BxPC-3, which contains a loss-of-function mutation in the tumor-suppressing gene SMAD4. These findings suggest a proto-oncogenic role of DNAJA1 in PDAC progression and suggest DNAJA1 may function synergistically with other proteins with altered activities in pancreatic cancer cell lines.
Collapse
Affiliation(s)
- Heidi E Roth
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States.,Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States
| | - Rodrigo Franco
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68583-0905, United States.,Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States
| | - You Zhou
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States.,Morrison Microscopy Core Research Facility, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0664, United States
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States.,Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States.,Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska 68588-0304, United States
| |
Collapse
|
21
|
Roubenne L, Marthan R, Le Grand B, Guibert C. Hydrogen Sulfide Metabolism and Pulmonary Hypertension. Cells 2021; 10:cells10061477. [PMID: 34204699 PMCID: PMC8231487 DOI: 10.3390/cells10061477] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a severe and multifactorial disease characterized by a progressive elevation of pulmonary arterial resistance and pressure due to remodeling, inflammation, oxidative stress, and vasoreactive alterations of pulmonary arteries (PAs). Currently, the etiology of these pathological features is not clearly understood and, therefore, no curative treatment is available. Since the 1990s, hydrogen sulfide (H2S) has been described as the third gasotransmitter with plethoric regulatory functions in cardiovascular tissues, especially in pulmonary circulation. Alteration in H2S biogenesis has been associated with the hallmarks of PH. H2S is also involved in pulmonary vascular cell homeostasis via the regulation of hypoxia response and mitochondrial bioenergetics, which are critical phenomena affected during the development of PH. In addition, H2S modulates ATP-sensitive K+ channel (KATP) activity, and is associated with PA relaxation. In vitro or in vivo H2S supplementation exerts antioxidative and anti-inflammatory properties, and reduces PA remodeling. Altogether, current findings suggest that H2S promotes protective effects against PH, and could be a relevant target for a new therapeutic strategy, using attractive H2S-releasing molecules. Thus, the present review discusses the involvement and dysregulation of H2S metabolism in pulmonary circulation pathophysiology.
Collapse
Affiliation(s)
- Lukas Roubenne
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut-Lévêque, F-33604 Pessac, France; (L.R.); (R.M.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ Bordeaux, U1045, 146 Rue Léo Saignat, F-33000 Bordeaux, France
- OP2 Drugs, Avenue du Haut Lévêque, F-33604 Pessac, France;
| | - Roger Marthan
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut-Lévêque, F-33604 Pessac, France; (L.R.); (R.M.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ Bordeaux, U1045, 146 Rue Léo Saignat, F-33000 Bordeaux, France
- CHU de Bordeaux, Avenue du Haut Lévêque, F-33604 Pessac, France
| | - Bruno Le Grand
- OP2 Drugs, Avenue du Haut Lévêque, F-33604 Pessac, France;
| | - Christelle Guibert
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut-Lévêque, F-33604 Pessac, France; (L.R.); (R.M.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ Bordeaux, U1045, 146 Rue Léo Saignat, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
22
|
Shukla P, Singh KK. The mitochondrial landscape of ovarian cancer: emerging insights. Carcinogenesis 2021; 42:663-671. [PMID: 33928357 PMCID: PMC8163040 DOI: 10.1093/carcin/bgab033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/18/2021] [Accepted: 04/20/2021] [Indexed: 02/02/2023] Open
Abstract
Ovarian cancer (OC) is known to be the most lethal cancer in women worldwide, and its etiology is poorly understood. Recent studies show that mitochondrial DNA (mtDNA) content as well as mtDNA and nuclear genes encoding mitochondrial proteins influence OC risk. This review presents an overview of role of mitochondrial genetics in influencing OC development and discusses the contribution of mitochondrial proteome in OC development, progression and therapy. A role of mitochondrial genetics in racial disparity is also highlighted. In-depth understanding of role of mitochondria in OC will help develop strategies toward prevention and treatment and improving overall survival in women with OC.
Collapse
Affiliation(s)
- Pallavi Shukla
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Molecular Endocrinology, Indian Council of Medical Research-National Institute for Research in Reproductive Health (ICMR-NIRRH), Mumbai, India
| | - Keshav K Singh
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
23
|
Szabo C. Hydrogen Sulfide, an Endogenous Stimulator of Mitochondrial Function in Cancer Cells. Cells 2021; 10:cells10020220. [PMID: 33499368 PMCID: PMC7911547 DOI: 10.3390/cells10020220] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) has a long history as toxic gas and environmental hazard; inhibition of cytochrome c oxidase (mitochondrial Complex IV) is viewed as a primary mode of its cytotoxic action. However, studies conducted over the last two decades unveiled multiple biological regulatory roles of H2S as an endogenously produced mammalian gaseous transmitter. Cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST) are currently viewed as the principal mammalian H2S-generating enzymes. In contrast to its inhibitory (toxicological) mitochondrial effects, at lower (physiological) concentrations, H2S serves as a stimulator of electron transport in mammalian mitochondria, by acting as an electron donor—with sulfide:quinone oxidoreductase (SQR) being the immediate electron acceptor. The mitochondrial roles of H2S are significant in various cancer cells, many of which exhibit high expression and partial mitochondrial localization of various H2S producing enzymes. In addition to the stimulation of mitochondrial ATP production, the roles of endogenous H2S in cancer cells include the maintenance of mitochondrial organization (protection against mitochondrial fission) and the maintenance of mitochondrial DNA repair (via the stimulation of the assembly of mitochondrial DNA repair complexes). The current article overviews the state-of-the-art knowledge regarding the mitochondrial functions of endogenously produced H2S in cancer cells.
Collapse
Affiliation(s)
- Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
24
|
Ascenção K, Dilek N, Augsburger F, Panagaki T, Zuhra K, Szabo C. Pharmacological induction of mesenchymal-epithelial transition via inhibition of H2S biosynthesis and consequent suppression of ACLY activity in colon cancer cells. Pharmacol Res 2021; 165:105393. [PMID: 33484818 DOI: 10.1016/j.phrs.2020.105393] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/12/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023]
Abstract
Hydrogen sulfide (H2S) is an important endogenous gaseous transmitter mediator, which regulates a variety of cellular functions in autocrine and paracrine manner. The enzymes responsible for the biological generation of H2S include cystathionine-β-synthase (CBS), cystathionine-γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST). Increased expression of these enzymes and overproduction of H2S has been implicated in essential processes of various cancer cells, including the stimulation of metabolism, maintenance of cell proliferation and cytoprotection. Cancer cell identity is characterized by so-called "transition states". The progression from normal (epithelial) to transformed (mesenchymal) state is termed epithelial-to-mesenchymal transition (EMT) whereby epithelial cells lose their cell-to-cell adhesion capacity and gain mesenchymal characteristics. The transition process can also proceed in the opposite direction, and this process is termed mesenchymal-to-epithelial transition (MET). The current project was designed to determine whether inhibition of endogenous H2S production in colon cancer cells affects the EMT/MET balance in vitro. Inhibition of H2S biosynthesis in HCT116 human colon cancer cells was achieved either with aminooxyacetic acid (AOAA) or 2-[(4-hydroxy-6-methylpyrimidin-2-yl)sulfanyl]-1-(naphthalen-1-yl)ethan-1-one (HMPSNE). These inhibitors induced an upregulation of E-cadherin and Zonula occludens-1 (ZO-1) expression and downregulation of fibronectin expression, demonstrating that H2S biosynthesis inhibitors can produce a pharmacological induction of MET in colon cancer cells. These actions were functionally reflected in an inhibition of cell migration, as demonstrated in an in vitro "scratch wound" assay. The mechanisms involved in the action of endogenously produced H2S in cancer cells in promoting (or maintaining) EMT (or tonically inhibiting MET) relate, at least in part, in the induction of ATP citrate lyase (ACLY) protein expression, which occurs via upregulation of ACLY mRNA (via activation of the ACLY promoter). ACLY in turn, regulates the Wnt-β-catenin pathway, an essential regulator of the EMT/MET balance. Taken together, pharmacological inhibition of endogenous H2S biosynthesis in cancer cells induces MET. We hypothesize that this may contribute to anti-cancer / anti-metastatic effects of H2S biosynthesis inhibitors.
Collapse
Affiliation(s)
- Kelly Ascenção
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| | - Nahzli Dilek
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| | - Fiona Augsburger
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| | - Theodora Panagaki
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| | - Karim Zuhra
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
25
|
The multifaceted roles of sulfane sulfur species in cancer-associated processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2020; 1862:148338. [PMID: 33212042 DOI: 10.1016/j.bbabio.2020.148338] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/30/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023]
Abstract
Sulfane sulfur species comprise a variety of biologically relevant hydrogen sulfide (H2S)-derived species, including per- and poly-sulfidated low molecular weight compounds and proteins. A growing body of evidence suggests that H2S, currently recognized as a key signaling molecule in human physiology and pathophysiology, plays an important role in cancer biology by modulating cell bioenergetics and contributing to metabolic reprogramming. This is accomplished through functional modulation of target proteins via H2S binding to heme iron centers or H2S-mediated reversible per- or poly-sulfidation of specific cysteine residues. Since sulfane sulfur species are increasingly viewed not only as a major source of H2S but also as key mediators of some of the biological effects commonly attributed to H2S, the multifaceted role of these species in cancer biology is reviewed here with reference to H2S, focusing on their metabolism, signaling function, impact on cell bioenergetics and anti-tumoral properties.
Collapse
|
26
|
Ren L, Chen X, Chen X, Li J, Cheng B, Xia J. Mitochondrial Dynamics: Fission and Fusion in Fate Determination of Mesenchymal Stem Cells. Front Cell Dev Biol 2020; 8:580070. [PMID: 33178694 PMCID: PMC7593605 DOI: 10.3389/fcell.2020.580070] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are pivotal to tissue homeostasis, repair, and regeneration due to their potential for self-renewal, multilineage differentiation, and immune modulation. Mitochondria are highly dynamic organelles that maintain their morphology via continuous fission and fusion, also known as mitochondrial dynamics. MSCs undergo specific mitochondrial dynamics during proliferation, migration, differentiation, apoptosis, or aging. Emerging evidence suggests that mitochondrial dynamics are key contributors to stem cell fate determination. The coordination of mitochondrial fission and fusion is crucial for cellular function and stress responses, while abnormal fission and/or fusion causes MSC dysfunction. This review focuses on the role of mitochondrial dynamics in MSC commitment under physiological and stress conditions. We highlight mechanistic insights into modulating mitochondrial dynamics and mitochondrial strategies for stem cell-based regenerative medicine. These findings shed light on the contribution of mitochondrial dynamics to MSC fate and MSC-based tissue repair.
Collapse
Affiliation(s)
- Lin Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaodan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaobing Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jiayan Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Rao G, Murphy B, Dey A, Dhar Dwivedi SK, Zhang Y, Roy RV, Chakraborty P, Bhattacharya R, Mukherjee P. Cystathionine beta synthase regulates mitochondrial dynamics and function in endothelial cells. FASEB J 2020; 34:9372-9392. [PMID: 32463541 PMCID: PMC7675787 DOI: 10.1096/fj.202000173r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/19/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Mutations in the human cystathionine beta synthase (CBS) gene are known to cause endothelial dysfunction responsible for cardiovascular and neurovascular diseases. CBS is the predominant hydrogen sulfide (H2 S)-producing enzyme in endothelial cells (ECs). Recently, H2 S was shown to attenuate ROS and improve mitochondrial function. Mitochondria are metabolic organelles that actively transform their ultrastructure to mediate their function. Therefore, we questioned whether perturbation of CBS/H2 S activity could drive mitochondrial dysfunction via mitochondrial dynamics in ECs. Here we demonstrate that silencing CBS induces mitochondria fragmentation, attenuates efficient oxidative phosphorylation, and decreases EC function. Mechanistically, CBS silencing significantly elevates ROS production, thereby leading to reduced mitofusin 2 (MFN2) expression, decouple endoplasmic reticulum-mitochondria contacts, increased mitochondria fission, enhanced receptor-mediated mitophagy, and increased EC death. These defects were significantly rescued by the treatment of H2 S donors. Taken together our data highlights a novel signaling axis that mechanistically links CBS with mitochondrial function and ER-mitochondrial tethering and could be considered as a new therapeutic approach for the intervention of EC dysfunction-related pathologies.
Collapse
Affiliation(s)
- Geeta Rao
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Brennah Murphy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anindya Dey
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Yushan Zhang
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ram Vinod Roy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Prabir Chakraborty
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
28
|
Zuhra K, Augsburger F, Majtan T, Szabo C. Cystathionine-β-Synthase: Molecular Regulation and Pharmacological Inhibition. Biomolecules 2020; 10:E697. [PMID: 32365821 PMCID: PMC7277093 DOI: 10.3390/biom10050697] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Cystathionine-β-synthase (CBS), the first (and rate-limiting) enzyme in the transsulfuration pathway, is an important mammalian enzyme in health and disease. Its biochemical functions under physiological conditions include the metabolism of homocysteine (a cytotoxic molecule and cardiovascular risk factor) and the generation of hydrogen sulfide (H2S), a gaseous biological mediator with multiple regulatory roles in the vascular, nervous, and immune system. CBS is up-regulated in several diseases, including Down syndrome and many forms of cancer; in these conditions, the preclinical data indicate that inhibition or inactivation of CBS exerts beneficial effects. This article overviews the current information on the expression, tissue distribution, physiological roles, and biochemistry of CBS, followed by a comprehensive overview of direct and indirect approaches to inhibit the enzyme. Among the small-molecule CBS inhibitors, the review highlights the specificity and selectivity problems related to many of the commonly used "CBS inhibitors" (e.g., aminooxyacetic acid) and provides a comprehensive review of their pharmacological actions under physiological conditions and in various disease models.
Collapse
Affiliation(s)
- Karim Zuhra
- Chair of Pharmacology, Section of Medicine, University of Fribourg, 1702 Fribourg, Switzerland; (K.Z.); (F.A.)
| | - Fiona Augsburger
- Chair of Pharmacology, Section of Medicine, University of Fribourg, 1702 Fribourg, Switzerland; (K.Z.); (F.A.)
| | - Tomas Majtan
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, 1702 Fribourg, Switzerland; (K.Z.); (F.A.)
| |
Collapse
|
29
|
Murphy B, Bhattacharya R, Mukherjee P. Hydrogen sulfide signaling in mitochondria and disease. FASEB J 2019; 33:13098-13125. [PMID: 31648556 PMCID: PMC6894098 DOI: 10.1096/fj.201901304r] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/17/2019] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide can signal through 3 distinct mechanisms: 1) reduction and/or direct binding of metalloprotein heme centers, 2) serving as a potent antioxidant through reactive oxygen species/reactive nitrogen species scavenging, or 3) post-translational modification of proteins by addition of a thiol (-SH) group onto reactive cysteine residues: a process known as persulfidation. Below toxic levels, hydrogen sulfide promotes mitochondrial biogenesis and function, thereby conferring protection against cellular stress. For these reasons, increases in hydrogen sulfide and hydrogen sulfide-producing enzymes have been implicated in several human disease states. This review will first summarize our current understanding of hydrogen sulfide production and metabolism, as well as its signaling mechanisms; second, this work will detail the known mechanisms of hydrogen sulfide in the mitochondria and the implications of its mitochondrial-specific impacts in several pathologic conditions.-Murphy, B., Bhattacharya, R., Mukherjee, P. Hydrogen sulfide signaling in mitochondria and disease.
Collapse
Affiliation(s)
- Brennah Murphy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
30
|
Santos I, Ramos C, Mendes C, Sequeira CO, Tomé CS, Fernandes DGH, Mota P, Pires RF, Urso D, Hipólito A, Antunes AMM, Vicente JB, Pereira SA, Bonifácio VDB, Nunes SC, Serpa J. Targeting Glutathione and Cystathionine β-Synthase in Ovarian Cancer Treatment by Selenium-Chrysin Polyurea Dendrimer Nanoformulation. Nutrients 2019; 11:E2523. [PMID: 31635026 PMCID: PMC6836284 DOI: 10.3390/nu11102523] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is the main cause of death from gynecological cancer, with its poor prognosis mainly related to late diagnosis and chemoresistance (acquired or intrinsic) to conventional alkylating and reactive oxygen species (ROS)-generating drugs. We and others reported that the availability of cysteine and glutathione (GSH) impacts the mechanisms of resistance to carboplatin in ovarian cancer. Different players in cysteine metabolism can be crucial in chemoresistance, such as the cystine/glutamate antiporter system Xc (xCT) and the H2S-synthesizing enzyme cystathionine β-synthase (CBS) in the pathway of cysteine catabolism. We hypothesized that, by disrupting cysteine metabolic flux, chemoresistance would be reverted. Since the xCT transporter is also able to take up selenium, we used selenium-containing chrysin (SeChry) as a plausible competitive inhibitor of xCT. For that, we tested the effects of SeChry on three different ovarian cancer cell lines (ES2, OVCAR3, and OVCAR8) and in two non-malignant cell lines (HaCaT and HK2). Results showed that, in addition to being highly cytotoxic, SeChry does not affect the uptake of cysteine, although it increases GSH depletion, indicating that SeChry might induce oxidative stress. However, enzymatic assays revealed an inhibitory effect of SeChry toward CBS, thus preventing production of the antioxidant H2S. Notably, our data showed that SeChry and folate-targeted polyurea dendrimer generation four (SeChry@PUREG4-FA) nanoparticles increased the specificity for SeChry delivery to ovarian cancer cells, reducing significantly the toxicity against non-malignant cells. Collectively, our data support SeChry@PUREG4-FA nanoparticles as a targeted strategy to improve ovarian cancer treatment, where GSH depletion and CBS inhibition underlie SeChry cytotoxicity.
Collapse
Affiliation(s)
- Inês Santos
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal.
| | - Cristiano Ramos
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal.
| | - Cindy Mendes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal.
| | - Catarina O Sequeira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
| | - Catarina S Tomé
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| | - Dalila G H Fernandes
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| | - Pedro Mota
- CQFM-IN and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Rita F Pires
- CQFM-IN and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Donato Urso
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal.
| | - Ana Hipólito
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal.
| | - Alexandra M M Antunes
- Centro de Química Estrutural, Instituto Superior Técnico, ULisboa, Avenida Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - João B Vicente
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
| | - Vasco D B Bonifácio
- CQFM-IN and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Sofia C Nunes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal.
| | - Jacinta Serpa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal.
| |
Collapse
|
31
|
Chen S, Yue T, Huang Z, Zhu J, Bu D, Wang X, Pan Y, Liu Y, Wang P. Inhibition of hydrogen sulfide synthesis reverses acquired resistance to 5-FU through miR-215-5p-EREG/TYMS axis in colon cancer cells. Cancer Lett 2019; 466:49-60. [PMID: 31542354 DOI: 10.1016/j.canlet.2019.09.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/03/2019] [Accepted: 09/14/2019] [Indexed: 01/05/2023]
Abstract
Acquired resistance to 5-fluorouracil (5-FU) is a major barrier to benefit from chemotherapy in colon cancer patients. Hydrogen sulfide (H2S), mainly produced by cystathionine-β-synthase (CBS), has been reported to promote the proliferation and migration of colon cancer cells. In this study, the effect of inhibiting H2S synthesis on the sensitivity of colon cancer cell lines to 5-FU was investigated. Increased expression of CBS was validated in online database and tissue microarrays. Inhibiting H2S synthesis significantly sensitized colon cancer cell lines to 5-FU both in vitro and in vivo. Decreasing H2S synthesis utilizing shRNA lentiviruses significantly reversed the acquired resistance to 5-FU. MicroRNA sequencing was performed and miR-215-5p was revealed as one of the miRNAs with most significantly altered expression levels after CBS knock down. Epiregulin (EREG) and thymidylate synthetase (TYMS) were predicted to be potential targets of miR-215-5p. Decreasing H2S synthesis significantly decreased the expression of EREG and TYMS. These results demonstrate that inhibiting H2S synthesis can reverse the acquired resistance to 5-FU in colon cancer cells.
Collapse
Affiliation(s)
- Shanwen Chen
- Division of General Surgery, Peking University First Hospital, Beijing, China
| | - Taohua Yue
- Division of General Surgery, Peking University First Hospital, Beijing, China
| | - Zhihao Huang
- Division of General Surgery, Peking University First Hospital, Beijing, China
| | - Jing Zhu
- Division of General Surgery, Peking University First Hospital, Beijing, China
| | - Dingfang Bu
- Central Laboratory, Peking University First Hospital, Beijing, China
| | - Xin Wang
- Division of General Surgery, Peking University First Hospital, Beijing, China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Beijing, China
| | - Yucun Liu
- Division of General Surgery, Peking University First Hospital, Beijing, China
| | - Pengyuan Wang
- Division of General Surgery, Peking University First Hospital, Beijing, China.
| |
Collapse
|
32
|
Mitophagy in Cancer: A Tale of Adaptation. Cells 2019; 8:cells8050493. [PMID: 31121959 PMCID: PMC6562743 DOI: 10.3390/cells8050493] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/18/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
:In the past years, we have learnt that tumors co-evolve with their microenvironment, and that the active interaction between cancer cells and stromal cells plays a pivotal role in cancer initiation, progression and treatment response. Among the players involved, the pathways regulating mitochondrial functions have been shown to be crucial for both cancer and stromal cells. This is perhaps not surprising, considering that mitochondria in both cancerous and non-cancerous cells are decisive for vital metabolic and bioenergetic functions and to elicit cell death. The central part played by mitochondria also implies the existence of stringent mitochondrial quality control mechanisms, where a specialized autophagy pathway (mitophagy) ensures the selective removal of damaged or dysfunctional mitochondria. Although the molecular underpinnings of mitophagy regulation in mammalian cells remain incomplete, it is becoming clear that mitophagy pathways are intricately linked to the metabolic rewiring of cancer cells to support the high bioenergetic demand of the tumor. In this review, after a brief introduction of the main mitophagy regulators operating in mammalian cells, we discuss emerging cell autonomous roles of mitochondria quality control in cancer onset and progression. We also discuss the relevance of mitophagy in the cellular crosstalk with the tumor microenvironment and in anti-cancer therapy responses.
Collapse
|
33
|
Dai W, Jiang L. Dysregulated Mitochondrial Dynamics and Metabolism in Obesity, Diabetes, and Cancer. Front Endocrinol (Lausanne) 2019; 10:570. [PMID: 31551926 PMCID: PMC6734166 DOI: 10.3389/fendo.2019.00570] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022] Open
Abstract
Metabolism describes the life-sustaining chemical reactions in organisms that provide both energy and building blocks for cellular survival and proliferation. Dysregulated metabolism leads to many life-threatening diseases including obesity, diabetes, and cancer. Mitochondria, subcellular organelles, contain the central energy-producing metabolic pathway, the tricarboxylic acid (TCA) cycle. Also, mitochondria exist in a dynamic network orchestrated by extracellular nutrient levels and intracellular energy needs. Upon stimulation, mitochondria undergo consistent interchange through fusion (small to big) and fission (big to small) processes. Mitochondrial fusion is primarily controlled by three GTPases, mitofusin 1 (Mfn1), Mfn2, and optic atrophy 1 (Opa1), while mitochondrial fission is primarily regulated by GTPase dynamin-related protein 1 (Drp1). Dysregulated activity of these GTPases results in disrupted mitochondrial dynamics and cellular metabolism. This review will update the metabolic roles of these GTPases in obesity, diabetes, and cancer.
Collapse
Affiliation(s)
- Wenting Dai
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute, Duarte, CA, United States
| | - Lei Jiang
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute, Duarte, CA, United States
- Comprehensive Cancer Center, City of Hope Medical Center, Duarte, CA, United States
- *Correspondence: Lei Jiang
| |
Collapse
|