51
|
The Role of Exhaled Hydrogen Sulfide in the Diagnosis of Colorectal Adenoma. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2021; 2021:8046368. [PMID: 34900068 PMCID: PMC8654565 DOI: 10.1155/2021/8046368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/27/2021] [Accepted: 11/11/2021] [Indexed: 12/24/2022]
Abstract
Purpose Exhaled determination can detect metabolite hydrogen sulfide in the intestine. We aim to analyze the predictive value of hydrogen sulfide in the diagnosis of colorectal adenoma. Methods We recruited seventy patients diagnosed with colorectal adenoma as the observation group and sixty-six healthy subjects as the control group. The colorectal adenoma was diagnosed by colonoscopy at the Endoscopy Center of Huashan Hospital affiliated to Fudan University from June 2018 to November 2019. Exhaled gas was collected through the nose and mouth, respectively, and hydrogen sulfide in exhaled gas was determined according to the manufacturer's instructions. Results Receiver operating characteristic (ROC) curve was analyzed based on the exhaled data of the observation group and the control group. The ROC curve showed an area under ROC curve (AUC) 0.724 for nasal exhaled H2S, which had a diagnostic value. When nasal exhaled H2S was >13.3 part per billion (ppb), the sensitivity and the specificity of predicting colorectal adenoma were 57% and 78%, respectively. The exhaled H2S of the observation group was significantly different from that of the control group. The AUC value was 0.716 as a prognostic factor of colorectal adenoma. As exhaled H2S was >28.8 ppb, the sensitivity and the specificity of predicting colorectal adenoma were 63% and 77%, respectively. Conclusion Exhaled and nasal H2S determination has a predictive value for colorectal adenoma as a novel and noninvasive method. Therefore, it is worth conducting more research to analyze exhaled and nasal H2S.
Collapse
|
52
|
Zhao H, Li C, Zhu S, Zhao Q, Dong H, Huang B, Han H. Molecular characterization and immune protection by cystathionine β-synthase from Eimeria tenella. J Eukaryot Microbiol 2021; 69:e12876. [PMID: 34850487 DOI: 10.1111/jeu.12876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Eimeria tenella is an obligate intracellular apicomplexan parasite that causes avian coccidiosis and leads to severe economic losses in the global poultry industry. Cystathionine β-synthase (CBS) and cystathionine γ-lyase (CGL) act together to generate H2S in the reverse transsulfuration pathway. In this study, E. tenella Cystathionine β-synthase (EtCBS) was cloned using rapid amplification of cDNA 5'-ends (5'RACE) and characterized, and its immunoprotective effects were evaluated. The recombinant EtCBS protein (rEtCBS) was expressed and successfully recognized by anti-sporozoites (Spz) protein rabbit serum. EtCBS mRNA levels were highest in Spz by qPCR, and the protein expression levels were higher in unsporulated oocysts (UO) than in other stages by Western blot. Indirect immunofluorescence showed that EtCBS protein was found on the surface of Spz and second-generation merozoites (Mrz). The invasion inhibition assays showed that rabbit anti-rEtCBS polyclonal antibodies effectively inhibited parasite invasion host cells. Chickens immunized with rEtCBS protein showed prominently increased weight gains and decreased oocyst output compared to nonimmunized and infected control group. The results suggest that EtCBS could be a potential vaccine candidate against E. tenella.
Collapse
Affiliation(s)
- Huanzhi Zhao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Cong Li
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Shunhai Zhu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Qiping Zhao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Hui Dong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Bing Huang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| | - Hongyu Han
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, CAAS, Shanghai, China
| |
Collapse
|
53
|
Kalushkova A, Nylund P, Párraga AA, Lennartsson A, Jernberg-Wiklund H. One Omics Approach Does Not Rule Them All: The Metabolome and the Epigenome Join Forces in Haematological Malignancies. EPIGENOMES 2021; 5:epigenomes5040022. [PMID: 34968247 PMCID: PMC8715477 DOI: 10.3390/epigenomes5040022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 02/01/2023] Open
Abstract
Aberrant DNA methylation, dysregulation of chromatin-modifying enzymes, and microRNAs (miRNAs) play a crucial role in haematological malignancies. These epimutations, with an impact on chromatin accessibility and transcriptional output, are often associated with genomic instability and the emergence of drug resistance, disease progression, and poor survival. In order to exert their functions, epigenetic enzymes utilize cellular metabolites as co-factors and are highly dependent on their availability. By affecting the expression of metabolic enzymes, epigenetic modifiers may aid the generation of metabolite signatures that could be utilized as targets and biomarkers in cancer. This interdependency remains often neglected and poorly represented in studies, despite well-established methods to study the cellular metabolome. This review critically summarizes the current knowledge in the field to provide an integral picture of the interplay between epigenomic alterations and the cellular metabolome in haematological malignancies. Our recent findings defining a distinct metabolic signature upon response to enhancer of zeste homolog 2 (EZH2) inhibition in multiple myeloma (MM) highlight how a shift of preferred metabolic pathways may potentiate novel treatments. The suggested link between the epigenome and the metabolome in haematopoietic tumours holds promise for the use of metabolic signatures as possible biomarkers of response to treatment.
Collapse
Affiliation(s)
- Antonia Kalushkova
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
- Correspondence:
| | - Patrick Nylund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Alba Atienza Párraga
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, NEO, Karolinska Institutet, 14157 Huddinge, Sweden;
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| |
Collapse
|
54
|
N1, N12-Diacetylspermine Is Elevated in Colorectal Cancer and Promotes Proliferation through the miR-559/CBS Axis in Cancer Cell Lines. JOURNAL OF ONCOLOGY 2021; 2021:6665704. [PMID: 34603448 PMCID: PMC8486517 DOI: 10.1155/2021/6665704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 08/06/2021] [Accepted: 08/15/2021] [Indexed: 01/05/2023]
Abstract
N1, N12-Diacetylspermine (DiAcSpm) has been reported to be upregulated in the urine of cancer patients. Mass spectrometry has shown elevated DiAcSpm expressions in colorectal cancer (CRC) tissues. However, the diagnostic application of DiAcSpm is not available due to a lack of diagnostic grade antibodies. Also, its biological roles in CRC cells remain unexplored. In the present study, we developed an antibody that directly detected DiAcSpm expression in paraffin-embedded tissues. We also characterized its biological characteristics and underlying mechanisms. Polyclonal antibodies were generated by immunizing animals with a synthetic product of DiAcSpm. Antibody DAS AB016 showed strong sensitivity against DiAcSpm in CRC tissues. Immunohistochemistry results showed that DiAcSpm expression was significantly elevated in CRC tissues. High levels of DiAcSpm correlated with the clinical stage and Ki67 index. DiAcSpm treatment increased levels of proliferation, cell cycle progression, and cyclin D1 and cyclin E proteins in CRC cell lines, SW480 and Caco-2. DiAcSpm also upregulated ATP production in these two cell lines. RNA-sequencing showed that DiAcSpm downregulated miR-559, which was confirmed using RT-qPCR. The luciferase reporter assay, western blotting, and RT-qPCR showed that cystathionine β-synthase (CBS) was the target of miR-559. miR-559 inhibited, while CBS accelerated, CRC proliferation. In addition, CBS siRNA knockdown blocked the biological effects of DiAcSpm on CRC cells. In conclusion, DiAcSpm was found to be increased in CRC tissues using a newly developed antibody. DiAcSpm accelerated CRC proliferation by regulating the miR-559/CBS axis.
Collapse
|
55
|
Yang H, Hu Y, Weng M, Liu X, Wan P, Hu Y, Ma M, Zhang Y, Xia H, Lv K. Hypoxia inducible lncRNA-CBSLR modulates ferroptosis through m6A-YTHDF2-dependent modulation of CBS in gastric cancer. J Adv Res 2021; 37:91-106. [PMID: 35499052 PMCID: PMC9039740 DOI: 10.1016/j.jare.2021.10.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/30/2021] [Accepted: 10/03/2021] [Indexed: 01/17/2023] Open
Abstract
1. The hypoxic microenvironment is a common hallmark of solid tumors and is strongly associated with therapy resistance and poor prognosis. 2. CBSLR, a long noncoding RNA transactivated by HIF-1α, is upregulated in GC and associated with poor prognosis. 3. CBSLR inhibition induces ferroptosis under hypoxic conditions and contributes to chemoresistance. 4. lncRNA-CBSLR recruits YTHDF2 protein and CBS mRNA to form CBSLR/ YTHDF2/CBS complex, which in turn decreases CBS mRNA stability in an m6A dependent manner. 5. CBSLR/CBS inhibits ferroptosis by modulating ACSL4 methylation to be polyubiquitinated.
Introduction Tumors are usually refractory to anti-cancer therapeutics under hypoxic conditions. However, the underlying molecular mechanism remains to be elucidated. Objectives Our study intended to identify hypoxia inducible lncRNAs and their biological function in gastric cancer (GC). Methods Differentially expressed lncRNAs were determined by microarray analysis between GC cells exposed to hypoxia (1% O2) and normoxia (21% O2) for 24 h. The expression level of CBSLR was manipulated in several GC cell lines to perform molecular and biological analyses both in vitro and in vivo. Results We identified a hypoxia-induced lncRNA-CBSLR that protected GC cells from ferroptosis, leading to chem-resistance. Mechanically, CBSLR interacted with YTHDF2 to form a CBSLR/YTHDF2/CBS signaling axis that decreased the stability of CBS mRNA by enhancing the binding of YTHDF2 with the m6A-modified coding sequence (CDS) of CBS mRNA. Furthermore, under decreased CBS levels, the methylation of the ACSL4 protein was reduced, leading to protein polyubiquitination and degradation of ACSL4. This, in turn, decreased the pro-ferroptosis phosphatidylethanolamine (PE) (18:0/20:4) and PE (18:0/22:4) content and contributed to ferroptosis resistance. Notably, CBSLR is upregulated, whereas CBS is downregulated in GC tissues compared to matched normal tissues; and GC patients with high CBSLR/low CBS levels have a worse clinical outcome and a poorer response to chemotherapy. Conclusion Our study reveals a novel mechanism in how HIF1α/CBSLR modulates ferroptosis/chemoresistance in GC, illuminating potential therapeutic targets for refractory hypoxic tumors.
Collapse
|
56
|
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
|
57
|
Lei T, Qian H, Lei P, Hu Y. Ferroptosis-related gene signature associates with immunity and predicts prognosis accurately in patients with osteosarcoma. Cancer Sci 2021; 112:4785-4798. [PMID: 34506683 PMCID: PMC8586685 DOI: 10.1111/cas.15131] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/04/2021] [Accepted: 09/04/2021] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma has been the most common malignant bone tumor in children and adolescents, while the 5‐y survival of osteosarcoma patients gained no significant improvement over the past decades. This study aimed to explore the role of ferroptosis‐related genes (FRGs) in the development and prognosis of osteosarcoma. The datasets of osteosarcoma patients including RNA sequencing data and clinical information were acquired from the TRGET and Gene Expression Omnibus (GEO) databases. The identification of molecular subgroups with different FRG expression patterns was achieved through nonnegative matrix factorization (NMF) clustering. The prognostic model was constructed using the least absolute shrinkage and selection operator (LASSO) algorithm and multivariate Cox regression analysis. The ESTIMATE algorithm was applied for determining the stromal score, immune score, ESTIMA score, and tumor purity of osteosarcoma patients. Functional analyses including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, Gene Set Enrichment Analysis (GSEA), and Gene Set Variation Analysis (GSVA) were conducted to explore the underlying mechanisms in the development and prognosis of osteosarcoma. Two molecular subgroups with different FRGs expression patterns were identified. The molecular subgroups with higher immune score and more active immune status showed better prognostic survival. On the basis of FRGs, a prognostic model and a nomogram integrating clinical characteristics were constructed and their prediction efficiency for osteosarcoma prognosis were well validated. Gene functional enrichment analysis showed that these differentially expressed FRGs were mainly enriched in immunity‐related signaling pathways, indicating that FRGs may affect the development and prognosis of osteosarcoma by regulating the immune microenvironment. The expression profiles of FRGs were closely related to the immunity status and prognostic survival of osteosarcoma patients. The interaction between ferroptosis and immunity in the development of osteosarcoma could provide a new insight into the exploration of molecular mechanisms and targeted therapies of osteosarcoma patients.
Collapse
Affiliation(s)
- Ting Lei
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
| | - Hu Qian
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Lei
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China.,Department of Orthopedic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Yihe Hu
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China.,Department of Orthopedic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
58
|
The Role of H 2S in the Gastrointestinal Tract and Microbiota. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1315:67-98. [PMID: 34302689 DOI: 10.1007/978-981-16-0991-6_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pathways and mechanisms of the production of H2S in the gastrointestinal tract are briefly described, including endogenous H2S produced by the organism and H2S from microorganisms in the gastrointestinal tract. In addition, the physiological regulatory functions of H2S on gastrointestinal motility, sensation, secretion and absorption, endocrine system, proliferation and differentiation of stem cells, and the possible mechanisms involved are introduced. In view of the complexity of biosynthesis, physiological roles, and the mechanism of H2S, this chapter focuses on the interactions and dynamic balance among H2S, gastrointestinal microorganisms, and the host. Finally, we focus on some clinical gastrointestinal diseases, such as inflammatory bowel disease, colorectal cancer, functional gastrointestinal disease, which might occur or develop when the above balance is broken. Pharmacological regulation of H2S or the intestinal microorganisms related to H2S might provide new therapeutic approaches for some gastrointestinal diseases.
Collapse
|
59
|
Hellmich MR, Chao C, Módis K, Ding Y, Zatarain JR, Thanki K, Maskey M, Druzhyna N, Untereiner AA, Ahmad A, Xue Y, Chen H, Russell WK, Wang J, Zhou J, Szabo C. Efficacy of Novel Aminooxyacetic Acid Prodrugs in Colon Cancer Models: Towards Clinical Translation of the Cystathionine β-Synthase Inhibition Concept. Biomolecules 2021; 11:biom11081073. [PMID: 34439739 PMCID: PMC8394431 DOI: 10.3390/biom11081073] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022] Open
Abstract
Upregulation of hydrogen sulfide (H2S) biosynthesis, at least in part related to the upregulation of cystathionine β-synthetase (CBS) in cancer cells, serves as a tumor-promoting factor and has emerged as a possible molecular target for antitumor drug development. To facilitate future clinical translation, we have synthesized a variety of novel CBS-targeting, esterase-cleavable prodrugs based on the structure of the prototypical CBS inhibitor aminooxyacetic acid (AOAA). The pharmacological properties of these compounds were evaluated in cell-free assays with recombinant human CBS protein, the human colon cancer cell line HCT116, and in vivo using various tumor-bearing mice models. The prodrug YD0251 (the isopropyl ester derivative of AOAA) was selected for detailed characterization. YD0251 exhibits improved antiproliferative efficacy in cell culture models when compared to AOAA. It is up to 18 times more potent than AOAA at suppressing HCT116 tumor growth in vivo and is effective when administered to tumor-bearing mice either via subcutaneous injection or oral gavage. Patient-derived xenografts (PDTXs) with higher levels of CBS protein grew significantly larger than tumors with lower levels, and YD0251 treatment inhibited the growth of PDTXs with elevated CBS, whereas it had no significant effect on PDTXs with low CBS protein levels. The toxicity of YD0251 was assessed in mice subjected to subchronic administration of supratherapeutic doses the inhibitor; no significant alteration in circulating markers of organ injury or histopathological alterations were noted, up to 60 mg/kg/day × 5 days. In preparation to a future theranostic concept (to match CBS inhibitor therapy to high-CBS expressors), we identified a potential plasma marker of CBS-expressing tumors. Colon cancer cells produced significant levels of lanthionine, a rare metabolic intermediate of CBS-mediated H2S biosynthesis; forced expression of CBS into non-transformed epithelial cells increased lanthionine biogenesis in vitro and in vivo (measured in the urine of tumor-bearing mice). These current results may be useful to facilitate the translation of a CBS inhibition-based antitumor concept into the clinical space.
Collapse
Affiliation(s)
- Mark R. Hellmich
- Department of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USA; (C.C.); (K.M.); (J.R.Z.); (K.T.); (M.M.)
- Correspondence: (M.R.H.); (J.Z.); (C.S.)
| | - Celia Chao
- Department of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USA; (C.C.); (K.M.); (J.R.Z.); (K.T.); (M.M.)
| | - Katalin Módis
- Department of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USA; (C.C.); (K.M.); (J.R.Z.); (K.T.); (M.M.)
- Department of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (N.D.); (A.A.U.); (A.A.)
| | - Ye Ding
- Department of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (Y.D.); (Y.X.); (H.C.)
| | - John R. Zatarain
- Department of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USA; (C.C.); (K.M.); (J.R.Z.); (K.T.); (M.M.)
| | - Ketan Thanki
- Department of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USA; (C.C.); (K.M.); (J.R.Z.); (K.T.); (M.M.)
| | - Manjit Maskey
- Department of Surgery, University of Texas, Medical Branch, Galveston, TX 77555, USA; (C.C.); (K.M.); (J.R.Z.); (K.T.); (M.M.)
| | - Nadiya Druzhyna
- Department of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (N.D.); (A.A.U.); (A.A.)
| | - Ashley A. Untereiner
- Department of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (N.D.); (A.A.U.); (A.A.)
| | - Akbar Ahmad
- Department of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (N.D.); (A.A.U.); (A.A.)
| | - Yu Xue
- Department of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (Y.D.); (Y.X.); (H.C.)
| | - Haiying Chen
- Department of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (Y.D.); (Y.X.); (H.C.)
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, University of Texas, Medical Branch, Galveston, TX 77555, USA;
| | - Jianmei Wang
- College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (Y.D.); (Y.X.); (H.C.)
- Correspondence: (M.R.H.); (J.Z.); (C.S.)
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas, Medical Branch, Galveston, TX 77555, USA; (N.D.); (A.A.U.); (A.A.)
- Chair of Pharmacology, Section of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
- Correspondence: (M.R.H.); (J.Z.); (C.S.)
| |
Collapse
|
60
|
Sabino JPJ, Oliveira LVDC, Soriano RN, Kwiatkoski M, Branco LGS, da Silva GSF. Role of hydrogen sulfide in ventilatory responses to hypercapnia in the medullary raphe of adult rats. Exp Physiol 2021; 106:1992-2001. [PMID: 34159656 DOI: 10.1113/ep089335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/18/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? There is evidence that H2 S plays a role in the control of breathing: what are its actions on the ventilatory and thermoregulatory responses to hypercapnia via effects in the medullary raphe, a brainstem region that participates in the ventilatory adjustments to hypercapnia? What is the main finding and its importance? Hypercapnia increased the endogenous production of H2 S in the medullary raphe. Inhibition of endogenous H2 S attenuated the ventilatory response to hypercapnia in unanaesthetized rats, suggesting its excitatory action via the cystathionine β-synthase-H2 S pathway in the medullary raphe. ABSTRACT Hydrogen sulfide (H2 S) has been recently recognized as a gasotransmitter alongside carbon monoxide (CO) and nitric oxide (NO). H2 S seems to modulate the ventilatory and thermoregulatory responses to hypoxia and hypercapnia. However, the action of the H2 S in the medullary raphe (MR) on the ventilatory responses to hypercapnia remains to be elucidated. The present study aimed to assess the role of H2 S in the MR (a brainstem region that contains CO2 -sensitive cells and participates in the ventilatory adjustments to hypercapnia) in the ventilatory responses to hypercapnia in adult unanaesthetized Wistar rats. To do so, aminooxyacetic acid (AOA; a cystathionine β-synthase (CBS) enzyme inhibitor), propargylglycine (PAG; a cystathionine γ-lyase enzyme inhibitor) and sodium sulfide (Na2 S; an H2 S donor) were microinjected into the MR. Respiratory frequency (fR ), tidal volume (VT ), ventilation ( V ̇ E ), oxygen consumption ( V ̇ O 2 ) and body temperature (Tb ) were measured under normocapnic (room air) and hypercapnic (7% CO2 ) conditions. H2 S concentration within the MR was determined. Microinjection of the drugs did not affect fR , VT and V ̇ E during normocapnia when compared to the control group. However, the microinjection of AOA, but not PAG, attenuated fR and V ̇ E during hypercapnia in comparison to the vehicle group, but had no effects on Tb . In addition, we observed an increase in the endogenous production of H2 S in the MR during hypercapnia. Our findings indicate that endogenously produced H2 S in the MR plays an excitatory role in the ventilatory response to hypercapnia, acting through the CBS-H2 S pathway.
Collapse
Affiliation(s)
- João Paulo Jacob Sabino
- Department of Biophysics and Physiology, Graduate Program in Pharmaceutical Sciences, Federal University of Piaui, Teresina, PI, Brazil
| | - Lucas Vaz de Castro Oliveira
- Department of Biophysics and Physiology, Graduate Program in Pharmaceutical Sciences, Federal University of Piaui, Teresina, PI, Brazil
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | | | - Luiz G S Branco
- Department of Morphology, Physiology and Basic Pathology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Glauber S F da Silva
- Department of Physiology and Biophysics, Institute of Biological Science Federal, University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
61
|
Jiang C, Huang H, Kang X, Yang L, Xi Z, Sun H, Pluth MD, Yi L. NBD-based synthetic probes for sensing small molecules and proteins: design, sensing mechanisms and biological applications. Chem Soc Rev 2021; 50:7436-7495. [PMID: 34075930 PMCID: PMC8763210 DOI: 10.1039/d0cs01096k] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Compounds with a nitrobenzoxadiazole (NBD) skeleton exhibit prominent useful properties including environmental sensitivity, high reactivity toward amines and biothiols (including H2S) accompanied by distinct colorimetric and fluorescent changes, fluorescence-quenching ability, and small size, all of which facilitate biomolecular sensing and self-assembly. Amines are important biological nucleophiles, and the unique activity of NBD ethers with amines has allowed for site-specific protein labelling and for the detection of enzyme activities. Both H2S and biothiols are involved in a wide range of physiological processes in mammals, and misregulation of these small molecules is associated with numerous diseases including cancers. In this review, we focus on NBD-based synthetic probes as advanced chemical tools for biomolecular sensing. Specifically, we discuss the sensing mechanisms and selectivity of the probes, the design strategies for multi-reactable multi-quenching probes, and the associated biological applications of these important constructs. We also highlight self-assembled NBD-based probes and outline future directions for NBD-based chemosensors. We hope that this comprehensive review will facilitate the development of future probes for investigating and understanding different biological processes and aid the development of potential theranostic agents.
Collapse
Affiliation(s)
- Chenyang Jiang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Haojie Huang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Xueying Kang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Liu Yang
- Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
| | - Zhen Xi
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, College of Chemistry, National Pesticide Engineering Research Center, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Hongyan Sun
- Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China. and Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Michael D Pluth
- Department of Chemistry and Biochemistry, Materials Science Institute, Knight Campus for Accelerating Scientific Impact, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA.
| | - Long Yi
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| |
Collapse
|
62
|
Wang RH, Chu YH, Lin KT. The Hidden Role of Hydrogen Sulfide Metabolism in Cancer. Int J Mol Sci 2021; 22:ijms22126562. [PMID: 34207284 PMCID: PMC8235762 DOI: 10.3390/ijms22126562] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022] Open
Abstract
Hydrogen Sulfide (H2S), an endogenously produced gasotransmitter, is involved in various important physiological and disease conditions, including vasodilation, stimulation of cellular bioenergetics, anti-inflammation, and pro-angiogenesis. In cancer, aberrant up-regulation of H2S-producing enzymes is frequently observed in different cancer types. The recognition that tumor-derived H2S plays various roles during cancer development reveals opportunities to target H2S-mediated signaling pathways in cancer therapy. In this review, we will focus on the mechanism of H2S-mediated protein persulfidation and the detailed information about the dysregulation of H2S-producing enzymes and metabolism in different cancer types. We will also provide an update on mechanisms of H2S-mediated cancer progression and summarize current options to modulate H2S production for cancer therapy.
Collapse
Affiliation(s)
- Rong-Hsuan Wang
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan; (R.-H.W.); (Y.-H.C.)
| | - Yu-Hsin Chu
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan; (R.-H.W.); (Y.-H.C.)
- Department of Life Science, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Kai-Ti Lin
- Institute of Biotechnology, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan; (R.-H.W.); (Y.-H.C.)
- Department of Medical Science, College of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
- Correspondence:
| |
Collapse
|
63
|
Hidalgo-Gutiérrez A, González-García P, Díaz-Casado ME, Barriocanal-Casado E, López-Herrador S, Quinzii CM, López LC. Metabolic Targets of Coenzyme Q10 in Mitochondria. Antioxidants (Basel) 2021; 10:520. [PMID: 33810539 PMCID: PMC8066821 DOI: 10.3390/antiox10040520] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/14/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is classically viewed as an important endogenous antioxidant and key component of the mitochondrial respiratory chain. For this second function, CoQ molecules seem to be dynamically segmented in a pool attached and engulfed by the super-complexes I + III, and a free pool available for complex II or any other mitochondrial enzyme that uses CoQ as a cofactor. This CoQ-free pool is, therefore, used by enzymes that link the mitochondrial respiratory chain to other pathways, such as the pyrimidine de novo biosynthesis, fatty acid β-oxidation and amino acid catabolism, glycine metabolism, proline, glyoxylate and arginine metabolism, and sulfide oxidation metabolism. Some of these mitochondrial pathways are also connected to metabolic pathways in other compartments of the cell and, consequently, CoQ could indirectly modulate metabolic pathways located outside the mitochondria. Thus, we review the most relevant findings in all these metabolic functions of CoQ and their relations with the pathomechanisms of some metabolic diseases, highlighting some future perspectives and potential therapeutic implications.
Collapse
Affiliation(s)
- Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Catarina M. Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Luis C. López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| |
Collapse
|
64
|
Li M, Liu Y, Deng Y, Pan L, Fu H, Han X, Li Y, Shi H, Wang T. Therapeutic potential of endogenous hydrogen sulfide inhibition in breast cancer (Review). Oncol Rep 2021; 45:68. [PMID: 33760221 PMCID: PMC8020202 DOI: 10.3892/or.2021.8019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Hydrogen sulfide (H2S), the third gas signal molecule, is associated with the modulation of various physiological and pathological processes. Recent studies have reevealed that endogenous H2S may promote proliferation, induce angiogenesis and inhibit apoptosis, thereby stimulating oncogenesis. Conversely, decreased endogenous H2S release suppresses growth of various tumors including breast cancer. This observation suggests an alternative tumor therapy strategy by inhibiting H2S-producing enzymes to reduce the release of endogenous H2S. Breast cancer is the most common type of cancer in women. Due to the lack of approved targeted therapy, its recurrence and metastasis still affect its clinical treatment. In recent years, significant progress has been made in the control of breast cancer by using inhibitors on H2S-producing enzymes. This review summarized the roles of endogenous H2S-producing enzymes in breast cancer and the effects of the enzyme inhibitors on anticancer and anti-metastasis, with the aim of providing new insights for the treatment of breast cancer.
Collapse
Affiliation(s)
- Ming Li
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Ya Liu
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuying Deng
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Limin Pan
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Han Fu
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Xue Han
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuxi Li
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Haimei Shi
- Department of Anesthesiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Tianxiao Wang
- School of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
65
|
Bonifácio VDB, Pereira SA, Serpa J, Vicente JB. Cysteine metabolic circuitries: druggable targets in cancer. Br J Cancer 2021; 124:862-879. [PMID: 33223534 PMCID: PMC7921671 DOI: 10.1038/s41416-020-01156-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/03/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
To enable survival in adverse conditions, cancer cells undergo global metabolic adaptations. The amino acid cysteine actively contributes to cancer metabolic remodelling on three different levels: first, in its free form, in redox control, as a component of the antioxidant glutathione or its involvement in protein s-cysteinylation, a reversible post-translational modification; second, as a substrate for the production of hydrogen sulphide (H2S), which feeds the mitochondrial electron transfer chain and mediates per-sulphidation of ATPase and glycolytic enzymes, thereby stimulating cellular bioenergetics; and, finally, as a carbon source for epigenetic regulation, biomass production and energy production. This review will provide a systematic portrayal of the role of cysteine in cancer biology as a source of carbon and sulphur atoms, the pivotal role of cysteine in different metabolic pathways and the importance of H2S as an energetic substrate and signalling molecule. The different pools of cysteine in the cell and within the body, and their putative use as prognostic cancer markers will be also addressed. Finally, we will discuss the pharmacological means and potential of targeting cysteine metabolism for the treatment of cancer.
Collapse
Affiliation(s)
- Vasco D B Bonifácio
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001, Lisboa, 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
| | - 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.
| | - João B Vicente
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), Avenida da República (EAN), 2780-157, Oeiras, Portugal
| |
Collapse
|
66
|
Nylund P, Atienza Párraga A, Haglöf J, De Bruyne E, Menu E, Garrido-Zabala B, Ma A, Jin J, Öberg F, Vanderkerken K, Kalushkova A, Jernberg-Wiklund H. A distinct metabolic response characterizes sensitivity to EZH2 inhibition in multiple myeloma. Cell Death Dis 2021; 12:167. [PMID: 33579905 PMCID: PMC7881125 DOI: 10.1038/s41419-021-03447-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) is a heterogeneous haematological disease that remains clinically challenging. Increased activity of the epigenetic silencer EZH2 is a common feature in patients with poor prognosis. Previous findings have demonstrated that metabolic profiles can be sensitive markers for response to treatment in cancer. While EZH2 inhibition (EZH2i) has proven efficient in inducing cell death in a number of human MM cell lines, we hereby identified a subset of cell lines that despite a global loss of H3K27me3, remains viable after EZH2i. By coupling liquid chromatography-mass spectrometry with gene and miRNA expression profiling, we found that sensitivity to EZH2i correlated with distinct metabolic signatures resulting from a dysregulation of genes involved in methionine cycling. Specifically, EZH2i resulted in a miRNA-mediated downregulation of methionine cycling-associated genes in responsive cells. This induced metabolite accumulation and DNA damage, leading to G2 arrest and apoptosis. Altogether, we unveiled that sensitivity to EZH2i in human MM cell lines is associated with a specific metabolic and gene expression profile post-treatment.
Collapse
Affiliation(s)
- Patrick Nylund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alba Atienza Párraga
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Jakob Haglöf
- Department of Medicinal Chemistry, Analytical Pharmaceutical Chemistry, Uppsala University, Uppsala, Sweden
| | - Elke De Bruyne
- Department of Haematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Eline Menu
- Department of Haematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Berta Garrido-Zabala
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Anqi Ma
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Fredrik Öberg
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Vanderkerken
- Department of Haematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Antonia Kalushkova
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
67
|
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
|
68
|
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
|
69
|
Blachier F, Andriamihaja M, Larraufie P, Ahn E, Lan A, Kim E. Production of hydrogen sulfide by the intestinal microbiota and epithelial cells and consequences for the colonic and rectal mucosa. Am J Physiol Gastrointest Liver Physiol 2021; 320:G125-G135. [PMID: 33084401 DOI: 10.1152/ajpgi.00261.2020] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Among bacterial metabolites, hydrogen sulfide (H2S) has received increasing attention. The epithelial cells of the large intestine are exposed to two sources of H2S. The main one is the luminal source that results from specific bacteria metabolic activity toward sulfur-containing substrates. The other source in colonocytes is from the intracellular production mainly through cystathionine β-synthase (CBS) activity. H2S is oxidized by the mitochondrial sulfide oxidation unit, resulting in ATP synthesis, and, thus, establishing this compound as the first mineral energy substrate in colonocytes. However, when the intracellular H2S concentration exceeds the colonocyte capacity for its oxidation, it inhibits the mitochondrial respiratory chain, thus affecting energy metabolism. Higher luminal H2S concentration affects the integrity of the mucus layer and displays proinflammatory effects. However, a low/minimal amount of endogenous H2S exerts an anti-inflammatory effect on the colon mucosa, pointing out the ambivalent effect of H2S depending on its intracellular concentration. Regarding colorectal carcinogenesis, forced CBS expression in late adenoma-like colonocytes increased their proliferative activity, bioenergetics capacity, and tumorigenicity; whereas, genetic ablation of CBS in mice resulted in a reduced number of mutagen-induced aberrant crypt foci. Activation of endogenous H2S production and low H2S extracellular concentration enhance cancerous colorectal cell proliferation. Higher exogenous H2S concentrations markedly reduce mitochondrial ATP synthesis and proliferative capacity in cancerous cells and enhance glycolysis but do not affect their ATP cell content or viability. Thus, it appears that, notably through an effect on colonocyte energy metabolism, endogenous and microbiota-derived H2S are involved in the host intestinal physiology and physiopathology.
Collapse
Affiliation(s)
- François Blachier
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Mireille Andriamihaja
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Pierre Larraufie
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Eunyeong Ahn
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, South Korea
| | - Annaïg Lan
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Eunjung Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, South Korea
| |
Collapse
|
70
|
González-García P, Hidalgo-Gutiérrez A, Mascaraque C, Barriocanal-Casado E, Bakkali M, Ziosi M, Abdihankyzy UB, Sánchez-Hernández S, Escames G, Prokisch H, Martín F, Quinzii CM, López LC. Coenzyme Q10 modulates sulfide metabolism and links the mitochondrial respiratory chain to pathways associated to one carbon metabolism. Hum Mol Genet 2020; 29:3296-3311. [PMID: 32975579 PMCID: PMC7724311 DOI: 10.1093/hmg/ddaa214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/03/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023] Open
Abstract
Abnormalities of one carbon, glutathione and sulfide metabolisms have recently emerged as novel pathomechanisms in diseases with mitochondrial dysfunction. However, the mechanisms underlying these abnormalities are not clear. Also, we recently showed that sulfide oxidation is impaired in Coenzyme Q10 (CoQ10) deficiency. This finding leads us to hypothesize that the therapeutic effects of CoQ10, frequently administered to patients with primary or secondary mitochondrial dysfunction, might be due to its function as cofactor for sulfide:quinone oxidoreductase (SQOR), the first enzyme in the sulfide oxidation pathway. Here, using biased and unbiased approaches, we show that supraphysiological levels of CoQ10 induces an increase in the expression of SQOR in skin fibroblasts from control subjects and patients with mutations in Complex I subunits genes or CoQ biosynthetic genes. This increase of SQOR induces the downregulation of the cystathionine β-synthase and cystathionine γ-lyase, two enzymes of the transsulfuration pathway, the subsequent downregulation of serine biosynthesis and the adaptation of other sulfide linked pathways, such as folate cycle, nucleotides metabolism and glutathione system. These metabolic changes are independent of the presence of sulfur aminoacids, are confirmed in mouse models, and are recapitulated by overexpression of SQOR, further proving that the metabolic effects of CoQ10 supplementation are mediated by the overexpression of SQOR. Our results contribute to a better understanding of how sulfide metabolism is integrated in one carbon metabolism and may explain some of the benefits of CoQ10 supplementation observed in mitochondrial diseases.
Collapse
Affiliation(s)
- Pilar González-García
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Agustín Hidalgo-Gutiérrez
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Cristina Mascaraque
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Eliana Barriocanal-Casado
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Mohammed Bakkali
- Departamento de Genética, Facultad de Ciencias, Universidad de Granada, Granada 18071, Spain
| | - Marcello Ziosi
- Department of Neurology, Columbia University Medical Center, New York 10032, NY, USA
| | | | | | - Germaine Escames
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Holger Prokisch
- Institute of Human Genetics, Technische Universität München, München 81675, Germany
| | - Francisco Martín
- Genomic Medicine Department, Centre for Genomics and Oncological Research, Granada 18007, Spain
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York 10032, NY, USA
| | - Luis C López
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| |
Collapse
|
71
|
Abstract
Aims: Cysteine persulfidation (also called sulfhydration or sulfuration) has emerged as a potential redox mechanism to regulate protein functions and diverse biological processes in hydrogen sulfide (H2S) signaling. Due to its intrinsically unstable nature, working with this modification has proven to be challenging. Although methodological progress has expanded the inventory of persulfidated proteins, there is a continued need to develop methods that can directly and unequivocally identify persulfidated cysteine residues in complex proteomes. Results: A quantitative chemoproteomic method termed as low-pH quantitative thiol reactivity profiling (QTRP) was developed to enable direct site-specific mapping and reactivity profiling of proteomic persulfides and thiols in parallel. The method was first applied to cell lysates treated with NaHS, resulting in the identification of overall 1547 persulfidated sites on 994 proteins. Structural analysis uncovered unique consensus motifs that might define this distinct type of modification. Moreover, the method was extended to profile endogenous protein persulfides in cells expressing H2S-generating enzyme, mouse tissues, and human serum, which led to additional insights into mechanistic, structural, and functional features of persulfidation events, particularly on human serum albumin. Innovation and Conclusion: Low-pH QTRP represents the first method that enables direct and unbiased proteomic mapping of cysteine persulfidation. Our method allows to generate the most comprehensive inventory of persulfidated targets of NaHS so far and to perform the first analysis of in vivo persulfidation events, providing a valuable tool to dissect the biological functions of this important modification.
Collapse
Affiliation(s)
- Ling Fu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences • Beijing, Beijing Institute of Lifeomics, Beijing, China
| | - Keke Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences • Beijing, Beijing Institute of Lifeomics, Beijing, China
| | - Jingyang He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences • Beijing, Beijing Institute of Lifeomics, Beijing, China
| | - Caiping Tian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences • Beijing, Beijing Institute of Lifeomics, Beijing, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences • Beijing, Beijing Institute of Lifeomics, Beijing, China
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences • Beijing, Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
72
|
Ostrakhovitch EA, Akakura S, Tabibzadeh S. Hydrogen sulfide facilitates reprogramming and trans-differentiation in 3D dermal fibroblast. PLoS One 2020; 15:e0241685. [PMID: 33180827 PMCID: PMC7660576 DOI: 10.1371/journal.pone.0241685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022] Open
Abstract
The efficiency of cell reprogramming in two-dimensional (2D) cultures is limited. Given that cellular stemness is intimately related to microenvironmental changes, 3D cell cultures have the potential of overcoming this limited capacity by allowing cells to self-organize by aggregation. In 3D space, cells interact more efficiently, modify their cellular topology, gene expression, signaling, and metabolism. It is yet not clear as how 3D culture environments modify the reprogramming potential of fibroblasts. We demonstrate that 3D spheroids from dermal fibroblasts formed under ultra-low attachment conditions showed increased lactate production. This is a requisite for cell reprogramming, increase their expression of pluripotency genes, such as OCT4, NANOG and SOX2, and display upregulated cystathionine-β-synthase (CBS) and hydrogen sulfide (H2S) production. Knockdown of CBS by RNAi suppresses lactic acid and H2S production and concomitantly decreases the expression of OCT4 and NANOG. On the contrary, H2S donors, NaHS and garlic-derived diallyl trisulfide (DATS), promote the expression of OCT4, and support osteogenic trans-differentiation of fibroblasts. These results demonstrate that CBS mediated release of H2S regulates the reprogramming of dermal fibroblasts grown in 3D cultures and supports their trans-differentiation.
Collapse
Affiliation(s)
- Elena A. Ostrakhovitch
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA, United States of America
| | - Shin Akakura
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA, United States of America
| | - Siamak Tabibzadeh
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA, United States of America
- * E-mail:
| |
Collapse
|
73
|
Wu Q, Ma J, Wei J, Meng W, Wang Y, Shi M. lncRNA SNHG11 Promotes Gastric Cancer Progression by Activating the Wnt/β-Catenin Pathway and Oncogenic Autophagy. Mol Ther 2020; 29:1258-1278. [PMID: 33068778 DOI: 10.1016/j.ymthe.2020.10.011] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/22/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are under active investigation in the development of cancers, including gastric cancer (GC). Oncogenic autophagy is required for cancer cell survival. The present study aimed to investigate the regulatory role of lncRNA small nucleolar host gene 11 (SNHG11) in GC. We show that SNHG11 is upregulated in GC, and that its upregulation correlated with dismal patient outcomes. Functionally, SNHG11 aggravated oncogenic autophagy to facilitate cell proliferation, stemness, migration, invasion, and epithelial-to-mesenchymal transition (EMT) in GC. Mechanistically, SNHG11 post-transcriptionally upregulated catenin beta 1 (CTNNB1) and autophagy related 12 (ATG12) through miR-483-3p/miR-1276, while the processing of precursor (pre-)miR-483/pre-miR-1276 was hindered by SNHG11. SNHG11 induced GSK-3β ubiquitination through interacting with Cullin 4A (CUL4A) to further activate the Wnt/β-catenin pathway. Intriguingly, SNHG11 regulated autophagy in a manner dependent on ATG12 rather than the Wnt/β-catenin pathway, whereas SNHG11 contributed to the malignant behaviors of GC cells via both pathways. Finally, SNHG11 upregulation in GC cells was shown to be transcriptionally induced by TCF7L2. In conclusion, we reveal that SNHG11 is an onco-lncRNA in GC and might be a promising prognostic and therapeutic target for GC.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Jiali Ma
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Jue Wei
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Wenying Meng
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China.
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China.
| |
Collapse
|
74
|
Zuhra K, Panagaki T, Randi EB, Augsburger F, Blondel M, Friocourt G, Herault Y, Szabo C. Mechanism of cystathionine-β-synthase inhibition by disulfiram: The role of bis(N,N-diethyldithiocarbamate)-copper(II). Biochem Pharmacol 2020; 182:114267. [PMID: 33035509 DOI: 10.1016/j.bcp.2020.114267] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) is an endogenous mammalian gasotransmitter. Cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST) are the principal enzymes responsible for its biogenesis. A recent yeast screen suggested that disulfiram (a well-known inhibitor of aldehyde dehydrogenase and a clinically used drug in the treatment of alcoholism) may inhibit CBS in a cell-based environment. However, prior studies have not observed any direct inhibition of CBS by disulfiram. We investigated the potential role of bioconversion of disulfiram to bis(N,N-diethyldithiocarbamate)-copper(II) complex (CuDDC) in the inhibitory effect of disulfiram on H2S production and assessed its effect in two human cell types with high CBS expression: HCT116 colon cancer cells and Down syndrome (DS) fibroblasts. METHODS H2S production from recombinant human CBS, CSE and 3-MST was measured using the fluorescent H2S probe AzMC. Mouse liver homogenate (a rich source of CBS) was also employed to measure H2S biosynthesis. The interaction of copper with accessible protein cysteine residues was evaluated using the DTNB method. Cell proliferation and viability were measured using the BrdU and MTT methods. Cellular bioenergetics was evaluated by Extracellular Flux Analysis. RESULTS While disulfiram did not exert any significant direct inhibitory effect on any of the H2S-producing enzymes, its metabolite, CuDDC was a potent inhibitor of CBS and CSE. The mode of its action is likely related to the complexed copper molecule. In cell-based systems, the effects of disulfiram were variable. In colon cancer cells, no significant effect of disulfiram was observed on H2S production or proliferation or viability. In contrast, in DS fibroblasts, disulfiram inhibited H2S production and improved proliferation and viability. Copper, on its own, failed to have any effects on either cell type, likely due to its low cell penetration. CuDDC inhibited H2S production in both cell types studied and exerted the functional effects that would be expected from a CBS inhibitor: inhibition of cell proliferation of cancer cells and a bell-shaped effect (stimulation of proliferation at low concentration and inhibition of these responses at higher concentration) in DS cells. Control experiments using a chemical H2S donor showed that, in addition to inhibiting CBS and CSE, part of the biological effects of CuDDC relates to a direct reaction with H2S, which occurs through its complexed copper. CONCLUSIONS Disulfiram, via its metabolite CuDDC acts as an inhibitor of CBS and a scavenger of H2S, which, in turn, potently suppresses H2S levels in various cell types. Inhibition of H2S biosynthesis may explain some of the previously reported actions of disulfiram and CuDDC in vitro and in vivo. Disulfiram or CuDDC may be considered as potential agents for the experimental therapy of various pathophysiological conditions associated with H2S overproduction.
Collapse
Affiliation(s)
- Karim Zuhra
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Theodora Panagaki
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Elisa B Randi
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Fiona Augsburger
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Marc Blondel
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Gaelle Friocourt
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67404 Illkirch, France
| | - Csaba Szabo
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
75
|
Chang M, Hou Z, Jin D, Zhou J, Wang M, Wang M, Shu M, Ding B, Li C, Lin J. Colorectal Tumor Microenvironment-Activated Bio-Decomposable and Metabolizable Cu 2 O@CaCO 3 Nanocomposites for Synergistic Oncotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2004647. [PMID: 32945002 DOI: 10.1002/adma.202004647] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/13/2020] [Indexed: 06/11/2023]
Abstract
Rational design of tumor microenvironment (TME)-activated nanocomposites provides an innovative strategy to construct responsive oncotherapy. In colorectal cancer (CRC), the specific physiological features are the overexpressed endogenous H2 S and slightly acidic microenvironment. Here, a core-shell Cu2 O@CaCO3 nanostructure for CRC "turn-on" therapy is reported. With CaCO3 responsive to pH decomposition and Cu2 O responsive to H2 S sulfuration, Cu2 O@CaCO3 can be triggered "on" into the therapeutic mode by the colorectal TME. When the CaCO3 shell decomposes and releases calcium in acidic colorectal TME, the loss of protection from the CaCO3 shell exposes the Cu2 O core to be sulfuretted by H2 S to form metabolizable Cu31 S16 nanocrystals that gain remarkably strong near-infrared absorption. After modifying hyaluronic acid, Cu2 O@CaCO3 can achieve synergistic CRC-targeted and TME-triggered photothermal/photodynamic/chemodynamic/calcium-overload-mediated therapy. Moreover, it is found that the generation of hyperthermia and oxidative stress from Cu2 O@CaCO3 nanocomposites can efficiently reprogram the macrophages from the M2 phenotype to the M1 phenotype and initiate a vaccine-like immune effect after primary tumor removal, which further induces an immune-favorable TME and intense immune responses for anti-CD47 antibody to simultaneously inhibit CRC distant metastasis and recurrence by immunotherapy.
Collapse
Affiliation(s)
- Mengyu Chang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| | - Zhiyao Hou
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangdong, 511436, P. R. China
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China
| | - Dayong Jin
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, New South Wales, 2007, Australia
- UTS-SUStech Joint Research Centre for Biomedical Materials & Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Jiajia Zhou
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, New South Wales, 2007, Australia
| | - Man Wang
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Meifang Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| | - Mengmeng Shu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
76
|
Chen M, Liu LX. MiR-525-5p Repressed Metastasis and Anoikis Resistance in Cervical Cancer via Blocking UBE2C/ZEB1/2 Signal Axis. Dig Dis Sci 2020; 65:2442-2451. [PMID: 31679088 DOI: 10.1007/s10620-019-05916-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Accumulating evidence indicated that miRNAs are important regulators involved in cancer biology. AIMS We aimed to investigate the biological functions and potentially underlying molecular mechanism of miR-525-5p in CC. METHODS RT-PCR and Western blot assay were performed to detect mRNA and protein expression. Cell proliferation, anoikis resistance, and cell invasion were analyzed. RESULTS We observed that the expression of miR-525-5p was declined in several CC cell lines. Additionally, introduction of miR-525-5p dramatically hampered cell viability, invasiveness, and migration ability through modulating epithelial-to-mesenchymal transition (EMT) marked genes as reflected by the upregulation of E-cadherin, as well as the downregulation of vimentin and N-cadherin. Furthermore, administration of miR-525-5p markedly reduced anchorage-independent growth and anoikis resistance accompanied by a decrease in the expression of anti-apoptotic protein Bcl-2 and an increase in the expression of pro-apoptotic protein Bax, C-caspase 3, and C-PARP1. Most importantly, analysis using publicly available algorithms predicted that UBE2C was a direct and functional target of miR-525-5p. Luciferase assays coupled with RT-PCR and Western blot analysis further verified that miR-525-5p negatively regulated UBE2C expression. Interestingly, miR-525-5p modulated ZEB1/2 expression via targeting UBE2C. Mechanically, administration of UBE2C partially blunted the salutary effects of miR-525-5p on invasive ability, EMT, and anoikis resistance, indicating that miR-525-5p acts as a tumor suppressor in CC largely through repression of UBE2C/ZEB1/2 signaling. CONCLUSIONS Taken together, our data identify a novel signaling axis of miR-525-5p/UBE2C/ZEB1/2 in repressing EMT and anoikis resistance, and likely serve as a potential therapeutic target for CC metastasis and prognosis as well as a therapeutic application.
Collapse
Affiliation(s)
- Mei Chen
- Department of Gynecology, Affiliated Hospital of Shaanxi University of Chinese Medicine, No. 2, Weiyang West Road, Xianyang, 712021, Shaanxi, People's Republic of China
| | - Li-Xiu Liu
- Department of Gynecology, Affiliated Hospital of Shaanxi University of Chinese Medicine, No. 2, Weiyang West Road, Xianyang, 712021, Shaanxi, People's Republic of China.
| |
Collapse
|
77
|
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
|
78
|
Faubert B, Solmonson A, DeBerardinis RJ. Metabolic reprogramming and cancer progression. Science 2020; 368:368/6487/eaaw5473. [PMID: 32273439 DOI: 10.1126/science.aaw5473] [Citation(s) in RCA: 1220] [Impact Index Per Article: 244.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
Metabolic reprogramming is a hallmark of malignancy. As our understanding of the complexity of tumor biology increases, so does our appreciation of the complexity of tumor metabolism. Metabolic heterogeneity among human tumors poses a challenge to developing therapies that exploit metabolic vulnerabilities. Recent work also demonstrates that the metabolic properties and preferences of a tumor change during cancer progression. This produces distinct sets of vulnerabilities between primary tumors and metastatic cancer, even in the same patient or experimental model. We review emerging concepts about metabolic reprogramming in cancer, with particular attention on why metabolic properties evolve during cancer progression and how this information might be used to develop better therapeutic strategies.
Collapse
Affiliation(s)
- Brandon Faubert
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ashley Solmonson
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA. .,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
79
|
Role of 3-Mercaptopyruvate Sulfurtransferase in the Regulation of Proliferation, Migration, and Bioenergetics in Murine Colon Cancer Cells. Biomolecules 2020; 10:biom10030447. [PMID: 32183148 PMCID: PMC7175125 DOI: 10.3390/biom10030447] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023] Open
Abstract
3-mercaptopyruvate sulfurtransferase (3-MST) has emerged as one of the significant sources of biologically active sulfur species in various mammalian cells. The current study was designed to investigate the functional role of 3-MST’s catalytic activity in the murine colon cancer cell line CT26. The novel pharmacological 3-MST inhibitor HMPSNE was used to assess cancer cell proliferation, migration and bioenergetics in vitro. Methods included measurements of cell viability (MTT and LDH assays), cell proliferation and in vitro wound healing (IncuCyte) and cellular bioenergetics (Seahorse extracellular flux analysis). 3-MST expression was detected by Western blotting; H2S production was measured by the fluorescent dye AzMC. The results show that CT26 cells express 3-MST protein and mRNA, as well as several enzymes involved in H2S degradation (TST, ETHE1). Pharmacological inhibition of 3-MST concentration-dependently suppressed H2S production and, at 100 and 300 µM, attenuated CT26 proliferation and migration. HMPSNE exerted a bell-shaped effect on several cellular bioenergetic parameters related to oxidative phosphorylation, while other bioenergetic parameters were either unaffected or inhibited at the highest concentration of the inhibitor tested (300 µM). In contrast to 3-MST, the expression of CBS (another H2S producing enzyme which has been previously implicated in the regulation of various biological parameters in other tumor cells) was not detectable in CT26 cells and pharmacological inhibition of CBS exerted no significant effects on CT26 proliferation or bioenergetics. In summary, 3-MST catalytic activity significantly contributes to the regulation of cellular proliferation, migration and bioenergetics in CT26 murine colon cancer cells. The current studies identify 3-MST as the principal source of biologically active H2S in this cell line.
Collapse
|
80
|
Abstract
In the past, hydrogen sulfide (H2S) was considered as a poisonous gas or waste of the body. Later, researchers found that H2S-producing enzymes exist in mammals. Moreover, their findings indicated that endogenous H2S was associated with the occurrence of many diseases. Therefore, endogenous H2S is able to participate in the regulation of physiological and pathological functions of the body as a gas signaling molecule. In this review, we summarize the regulation mechanism of endogenous H2S on the body, such as proliferation, apoptosis, migration, angiogenesis, as well as vasodilation/vasoconstriction. Furthermore, we also analyze the relationship between H2S and some chronic diseases, including hypoxic pulmonary hypertension, myocardial infarction, ischemic perfusion kidney injury, diabetes, and chronic intestinal diseases. Finally, we discuss dietary restriction and drugs that target for H2S. Hence, H2S is expected to become a potential target for treatment of these chronic diseases.
Collapse
Affiliation(s)
- Na Yang
- Office of Educational Administration, Hunan Polytechnic of Environment and Biology, Hengyang, China
| | - Yuan Liu
- Medical College, Hunan Polytechnic of Environment and Biology, Hengyang, China
| | - Tianping Li
- Office of Educational Administration, Hunan Polytechnic of Environment and Biology, Hengyang, China
| | - Qinhui Tuo
- Medical College, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
81
|
Liu N, Lin X, Huang C. Activation of the reverse transsulfuration pathway through NRF2/CBS confers erastin-induced ferroptosis resistance. Br J Cancer 2019; 122:279-292. [PMID: 31819185 PMCID: PMC7052275 DOI: 10.1038/s41416-019-0660-x] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/02/2019] [Accepted: 11/08/2019] [Indexed: 12/26/2022] Open
Abstract
Background Ferroptosis is an iron-dependent, lipid peroxide-mediated cell death that may be exploited to selective elimination of damaged and malignant cells. Recent studies have identified that small-molecule erastin specifically inhibits transmembrane cystine–glutamate antiporter system xc−, prevents extracellular cystine import and ultimately causes ferroptosis in certain cancer cells. In this study, we aimed to investigate the molecular mechanism underlying erastin-induced ferroptosis resistance in ovarian cancer cells. Methods We treated ovarian cancer cells with erastin and examined cell viability, cellular ROS and metabolites of the transsulfuration pathway. We also depleted cystathionine β-synthase (CBS) and NRF2 to investigate the CBS and NRF2 dependency in erastin-resistant cells. Results We found that prolonged erastin treatment induced ferroptosis resistance. Upon exposure to erastin, cells gradually adapted to cystine deprivation via sustained activation of the reverse transsulfuration pathway, allowing the cells to bypass erastin insult. CBS, the biosynthetic enzyme for cysteine, was constantly upregulated and was critical for the resistance. Knockdown of CBS by RNAi in erastin-resistant cells caused ferroptotic cell death, while CBS overexpression conferred ferroptosis resistance. We determined that the antioxidant transcriptional factor, NRF2 was constitutively activated in erastin-resistant cells and NRF2 transcriptionally upregulated CBS. Genetically repression of NRF2 enhanced ferroptosis susceptibility. Conclusions Based on these results, we concluded that constitutive activation of NRF2/CBS signalling confers erastin-induced ferroptosis resistance. This study demonstrates a new mechanism underlying ferroptosis resistance, and has implications for the therapeutic response to erastin-induced ferroptosis.
Collapse
Affiliation(s)
- Nan Liu
- Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xiaoli Lin
- Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chengying Huang
- Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
82
|
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
|
83
|
Wang Y, Huang J, Chen W, Wang R, Kao M, Pan Y, Chan S, Tsai K, Kung H, Lin K, Wang L. Dysregulation of cystathionine γ-lyase promotes prostate cancer progression and metastasis. EMBO Rep 2019; 20:e45986. [PMID: 31468690 PMCID: PMC6776913 DOI: 10.15252/embr.201845986] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2 S), an endogenous signaling gaseous molecule, is involved in various physiological activities, including vessel relaxation, regulation of cellular bioenergetics, inflammation, and angiogenesis. By using xenograft orthotopic implantation of prostate cancer PC3 cells and subsequently comparing bone metastatic with primary tumor-derived cancer cells, we find that H2 S-producing enzyme cystathionine γ-lyase (CTH) is upregulated in bone-metastatic PC3 cells. Clinical data further reveal that the expression of CTH is elevated in late-stage prostate cancer patients, and higher CTH expression correlates with poor survival from The Cancer Genome Atlas (TCGA) prostate cancer RNA-seq datasets. CTH promotes NF-κB nuclear translocation through H2 S-mediated sulfhydration on cysteine-38 of the NF-κB p65 subunit, resulting in increased IL-1β expression and H2 S-induced cell invasion. Knockdown of CTH in PC3 cells results in the suppression of tumor growth and distant metastasis, while overexpression of CTH in DU145 cells promotes primary tumor growth and lymph node metastasis in the orthotopic implanted xenograft mouse model. Together, our findings provide evidence that CTH generated H2 S promotes prostate cancer progression and metastasis through IL-1β/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yi‐Hsiang Wang
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Institute of Molecular MedicineCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Jo‐Ting Huang
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
| | - Wen‐Ling Chen
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
| | - Rong‐Hsuan Wang
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Chien Kao
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Yan‐Ru Pan
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Shih‐Hsuan Chan
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Institute of Molecular MedicineCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
- Chiese Medicine Research CenterInstitute of Integrated MedicineChina Medical UniversityTaichung CityTaiwan
| | - Kuo‐Wang Tsai
- Department of Medical Education and ResearchKaohsiung Veterans General HospitalKaohsiungTaiwan
- Institute of Biomedical SciencesNational Sun Yat‐Sen UniversityKaohsiungTaiwan
- Department of Chemical BiologyNational Pingtung University of EducationPingtungTaiwan
| | - Hsing‐Jien Kung
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- PhD Program for Cancer Biology and Drug DiscoveryTaipei Medical UniversityTaipeiTaiwan
| | - Kai‐Ti Lin
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Institute of BiotechnologyCollege of Life ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Lu‐Hai Wang
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanMiaoli CountyTaiwan
- Chiese Medicine Research CenterInstitute of Integrated MedicineChina Medical UniversityTaichung CityTaiwan
| |
Collapse
|
84
|
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
|
85
|
Damba T, Zhang M, Buist-Homan M, van Goor H, Faber KN, Moshage H. Hydrogen sulfide stimulates activation of hepatic stellate cells through increased cellular bio-energetics. Nitric Oxide 2019; 92:26-33. [PMID: 31401106 DOI: 10.1016/j.niox.2019.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/28/2022]
Abstract
Hepatic fibrosis is caused by chronic inflammation and characterized as the excessive accumulation of extracellular matrix (ECM) by activated hepatic stellate cells (HSCs). Gasotransmitters like NO and CO are known to modulate inflammation and fibrosis, however, little is known about the role of the gasotransmitter hydrogen sulfide (H2S) in liver fibrogenesis and stellate cell activation. Endogenous H2S is produced by the enzymes cystathionine β-synthase (CBS), cystathionine γ-lyase (CTH) and 3-mercaptopyruvate sulfur transferase (MPST) [1]. The aim of this study was to elucidate the role of endogenously produced and/or exogenously administered H2S on rat hepatic stellate cell activation and fibrogenesis. Primary rat HSCs were culture-activated for 7 days and treated with different H2S releasing donors (slow releasing donor GYY4137, fast releasing donor NaHS) or inhibitors of the H2S producing enzymes CTH and CBS (DL-PAG, AOAA). The main message of our study is that mRNA and protein expression level of H2S synthesizing enzymes are low in HSCs compared to hepatocytes and Kupffer cells. However, H2S promotes hepatic stellate cell activation. This conclusion is based on the fact that production of H2S and mRNA and protein expression of its producing enzyme CTH are increased during hepatic stellate cell activation. Furthermore, exogenous H2S increased HSC proliferation while inhibitors of endogenous H2S production reduce proliferation and fibrotic makers of HSCs. The effect of H2S on stellate cell activation correlated with increased cellular bioenergetics. Our results indicate that the H2S generation in hepatic stellate cells is a target for anti-fibrotic intervention and that systemic interventions with H2S should take into account cell-specific effects of H2S.
Collapse
Affiliation(s)
- Turtushikh Damba
- Dept. Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mengfan Zhang
- Dept. Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Manon Buist-Homan
- Dept. Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Dept. Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Harry van Goor
- Dept. Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Klaas Nico Faber
- Dept. Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Dept. Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Han Moshage
- Dept. Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Dept. Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
86
|
Tang D, Lin T, Wang Y, Cao H. High expression of proenkephalin is associated with favorable outcomes in patients with gastrointestinal stromal tumors. Cancer Manag Res 2019; 11:6681-6690. [PMID: 31410059 PMCID: PMC6643514 DOI: 10.2147/cmar.s202044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/11/2019] [Indexed: 01/01/2023] Open
Abstract
Purpose The aim of this study was to elucidate the prognostic value of proenkephalin (PENK) in gastrointestinal stromal tumors (GISTs). Patients and methods We collected data on 268 eligible postoperative patients diagnosed with GIST between January 1, 2002, and December 31, 2011. PENK expression was detected in GIST tissues classified using the United States National Institutes of Health (NIH) risk classification system. The associations between high PENK expression and the clinicopathological characteristics were assessed. Overall survival (OS) and recurrence-free survival (RFS) were estimated by Kaplan–Meier analysis, and the log-rank test was used to compare the differences between groups. Univariate and multivariate Cox regression analyses were conducted to assess the prognostic value of PENK in GIST patients. Results High PENK expression was more common in the low- and intermediate-risk GIST groups compared with the high-risk group (P<0.05). Additionally, PENK expression was associated with tumor size, mitosis count per 50 high-power fields, and tumor rupture (P<0.05). Kaplan–Meier analysis revealed that high PENK expression was associated with superior OS and RFS, while low PENK expression was associated with worse OS and RFS. Furthermore, PENK was shown to be an independent predictor of OS and RFS in the overall population (for OS, hazard ratio [HR], 1.596, 95% confidence interval [CI], 1.006–2.914, P<0.001; for RFS, HR, 1.910, 95% CI, 0.977–3.089, P<0.001). Conclusion PENK expression in GIST is closely associated with NIH risk grade and prognosis, indicating that PENK may act as a tumor suppressor and may serve as a new biomarker for predicting prognosis in postoperative GIST patients.
Collapse
Affiliation(s)
- Defeng Tang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, People's Republic of China.,Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, People's Republic of China
| | - Tianlong Lin
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, People's Republic of China
| | - Yangyang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, People's Republic of China
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, People's Republic of China
| |
Collapse
|
87
|
Wang R, Tao B, Fan Q, Wang S, Chen L, Zhang J, Hao Y, Dong S, Wang Z, Wang W, Cai Y, Li X, Bao T, Wang X, Qiu X, Wang K, Mo X, Kang Y, Wang Z. Fatty-acid receptor CD36 functions as a hydrogen sulfide-targeted receptor with its Cys333-Cys272 disulfide bond serving as a specific molecular switch to accelerate gastric cancer metastasis. EBioMedicine 2019; 45:108-123. [PMID: 31262715 PMCID: PMC6642364 DOI: 10.1016/j.ebiom.2019.06.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Hydrogen Sulfide (H2S), a third member of gasotransmitter family along with nitric oxide (NO) and carbon monoxide (CO), exerts a wide range of cellular and molecular actions in our body. There is a large body of evidence suggesting that H2S plays an important role in cancer metastasis; however, the molecular mechanisms of H2S-mediated acceleration of cancer metastasis remain unknown. METHODS We examined the promote effects of H2S on phenotype of gastric cancer (GC) cells (including those of express wild type CD36 and mutant CD36) in vitro and in vivo. GC patients' samples were used for clinical translational significance evaluation. FINDINGS H2S triggered lipid metabolism reprogramming by significantly up-regulating the expression of the fatty-acid receptor CD36 (CD36) and directly activating CD36 in GC cells. Mechanistically, a disulfide bond located between cysteine (Cys)333 and Cys272 within the CD36 protein structure that was labile to H2S-mediated modification. The long chain-fatty acid (LC-FA) binding pocket was capped by a turn in the CD36 protein, located between helical and sheet structures that were stabilized by the Cys333-Cys272. This limited the secondary binding between LC-FAs and lysine (Lys)334. Breaking the Cys333-Cys272 disulfide bond restored the second LC-FA binding conformation of CD36. Targeting CD36 in vivo blocked H2S-promoted metastasis and improved animal survival. INTERPRETATION These findings identify that the Cys333-Cys272 disulfide bond disrupted the integrity of the second LC-FA binding conformation of CD36. Therefore, CD36 can directly activate LC-FA access to the cytoplasm by acting as a direct target molecule for H2S.
Collapse
Affiliation(s)
- Rui Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Beibei Tao
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fudan University, Shanghai 200032, China
| | - Qilin Fan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shengyue Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Li Chen
- Department of Gastroenterology, Baoshan Branch, Renji Hospital, Affiliated to Shanghai Jiaotong University, Shanghai 200444, China
| | - Junjie Zhang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Yinfang Hao
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Shuang Dong
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Zhe Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Wei Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Yixi Cai
- Department of Pediatrics, First People's Hospital of Liangjiang New District, Chongqing 401121, China
| | - Xutong Li
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Tuvshin Bao
- Department of Anesthesia, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaohui Wang
- Department of Pancreatic Surgery, State Key Laboratory of Oncology in South China, Zhongshan University, Guangzhou 510001, China
| | - Xiaoming Qiu
- Department of Orthopedics, Provincial Hospital of Gansu Province, Lanzhou 730001, China
| | - Kekun Wang
- School of Health Science, Wuhan University, Wuhan 430030, China
| | - Xinyu Mo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300001, China
| | - Yuqi Kang
- Department of Oncology, Oncology Hospital of Guizhou Province, Guiyang 550001, China
| | - Zhirong Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China.
| |
Collapse
|
88
|
Bobba KN, Binoy A, Koo S, Nedungadi D, Podder A, Sharma A, Mishra N, Kim JS, Bhuniya S. Direct readout protonophore induced selective uncoupling and dysfunction of individual mitochondria within cancer cells. Chem Commun (Camb) 2019; 55:6429-6432. [PMID: 31094377 DOI: 10.1039/c9cc01483g] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Concurrently, manipulation of mitochondrial activity and its monitoring have enormous significance in cancer therapy and diagnosis. In this context, a fluorescent probe MitoDP has been developed for validating H2S mediated protonophore (2,4-dinitrophenol, DNP) induced mitochondrial membrane potential change, ROS formation and ATP depletion in cancer cells. The extent of protonophore activation for mitochondrial dysfunction is monitored through fluorescence signalling at 450 nm. The current study provides a proof for the concept of endogenous H2S-mediated controlled and spatial release of bioactive agents, or toxins specifically in mitochondria of cancer cells.
Collapse
Affiliation(s)
- Kondapa Naidu Bobba
- Amrita Centre for Industrial Research and Innovation, Amrita School of engineering, Amrita Vishwa Vidyapeetham, Coimbatore, 641-112, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
La Vecchia S, Sebastián C. Metabolic pathways regulating colorectal cancer initiation and progression. Semin Cell Dev Biol 2019; 98:63-70. [PMID: 31129171 DOI: 10.1016/j.semcdb.2019.05.018] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is one of the most common types of cancer worldwide. Despite recent advances in the molecular genetics of CRC, poor treatment outcomes highlight the need for a better understanding of the underlying mechanisms accounting for tumor initiation and progression. Recently, deregulation of cellular metabolism has emerged as a key hallmark of cancer. Reprogramming of core cellular metabolic pathways by cancer cells provides energy, anaplerotic precursors and reducing equivalents required to support tumor growth. Here, we review key findings implicating cancer metabolism as a major contributor of tumor initiation, growth and metastatic dissemination in CRC. We summarize the metabolic pathways governing stem cell fate in the intestine, the metabolic adaptations of proliferating colon cancer cells and their crosstalk with oncogenic signaling, and how they fulfill the energetic demands imposed by the metastatic cascade. Lastly, we discuss how some of these metabolic pathways could represent new vulnerabilities of CRC cells with the potential to be targeted.
Collapse
Affiliation(s)
- Sofia La Vecchia
- Laboratory of Metabolic Dynamics in Cancer, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo TO, Italy
| | - Carlos Sebastián
- Laboratory of Metabolic Dynamics in Cancer, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo TO, Italy.
| |
Collapse
|
90
|
Yang Z, Sun Q, Guo J, Wang S, Song G, Liu W, Liu M, Tang H. GRSF1-mediated MIR-G-1 promotes malignant behavior and nuclear autophagy by directly upregulating TMED5 and LMNB1 in cervical cancer cells. Autophagy 2019; 15:668-685. [PMID: 30394198 PMCID: PMC6526811 DOI: 10.1080/15548627.2018.1539590] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 10/14/2018] [Accepted: 10/17/2018] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence has revealed that miRNAs could upregulate the expression levels of target genes. However, the molecular mechanism underlying upregulation of targets mediated by miRNAs remains unclear. In this study, we found a novel miRNA named MIR-G-1 by GRSF1-RNA immunoprecipitation (RIP)-deep sequencing, which could directly target and upregulate LMNB1 and TMED5 in a GRSF1-dependent manner in cervical cancer cells. In addition, upregulated MIR-G-1 in cervical cancer promoted a malignant phenotype in vitro and in vivo. TMED5 could interact with WNT7B and thus activated the canonical WNT-CTNNB1/β-catenin signaling pathway. MIR-G-1 mediated the activation of this pathway. Furthermore, MIR-G-1 promoted serum starvation-induced nuclear macroautophagy/autophagy, and accelerated taxol (TAX)-induced DNA-damage repair in cervical cancer cells. Collectively, these findings may provide a new insight into the upregulation mechanism and nuclear autophagy mediated by miRNAs and provide a potential biomarker for cervical cancer. Abbreviations: 3'UTR: 3' untranslated region; EMSA: electrophoretic mobility shift assay; EMT: epithelial-mesenchymal transition; GRSF1: G-rich RNA sequence binding factor 1; IF: immunofluorescence; IP: immunoprecipitation; IHC: immunohistochemistry; lnc: long noncoding; miRNA:microRNA; TAX: taxol; TMED5: transmembrane p24 trafficking protein 5.
Collapse
Affiliation(s)
- Zhen Yang
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qi Sun
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junfei Guo
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shixing Wang
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ge Song
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weiying Liu
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Min Liu
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hua Tang
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
91
|
Abdollahi Govar A, Törő G, Szaniszlo P, Pavlidou A, Bibli SI, Thanki K, Resto VA, Chao C, Hellmich MR, Szabo C, Papapetropoulos A, Módis K. 3-Mercaptopyruvate sulfurtransferase supports endothelial cell angiogenesis and bioenergetics. Br J Pharmacol 2019; 177:866-883. [PMID: 30644090 DOI: 10.1111/bph.14574] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/26/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE During angiogenesis, quiescent endothelial cells (ECs) are activated by various stimuli to form new blood vessels from pre-existing ones in physiological and pathological conditions. Many research groups have shown that hydrogen sulfide (H2 S), the newest member of the gasotransmitter family, acts as a proangiogenic factor. To date, very little is known about the regulatory role of 3-mercaptopyruvate sulfurtransferase (3-MST), an important H2 S-producing enzyme in ECs. The aim of our study was to explore the potential role of 3-MST in human EC bioenergetics, metabolism, and angiogenesis. EXPERIMENTAL APPROACH To assess in vitro angiogenic responses, we used EA.hy926 human vascular ECs subjected to shRNA-mediated 3-MST attenuation and pharmacological inhibition of proliferation, migration, and tube-like network formation. To evaluate bioenergetic parameters, cell respiration, glycolysis, glucose uptake, and mitochondrial/glycolytic ATP production were measured. Finally, global metabolomic profiling was performed to determine the level of 669 metabolic compounds. KEY RESULTS 3-MST-attenuated ECs subjected to shRNA or pharmacological inhibition of 3-MST significantly reduced EC proliferation, migration, and tube-like network formation. 3-MST silencing also suppressed VEGF-induced EC migration. From bioenergetic and metabolic standpoints, 3-MST attenuation decreased mitochondrial respiration and mitochondrial ATP production, increased glucose uptake, and perturbed the entire EC metabolome. CONCLUSION AND IMPLICATIONS 3-MST regulates bioenergetics and morphological angiogenic functions in human ECs. The data presented in the current report support the view that 3-MST pathway may be a potential candidate for therapeutic modulation of angiogenesis. LINKED ARTICLES This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
| | - Gábor Törő
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Peter Szaniszlo
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Athanasia Pavlidou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia-Iris Bibli
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Ketan Thanki
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Vicente A Resto
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Celia Chao
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Mark R Hellmich
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, USA.,Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, 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, Athens, Greece
| | - Katalin Módis
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, USA.,Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
92
|
Blachier F, Beaumont M, Kim E. Cysteine-derived hydrogen sulfide and gut health: a matter of endogenous or bacterial origin. Curr Opin Clin Nutr Metab Care 2019; 22:68-75. [PMID: 30461448 DOI: 10.1097/mco.0000000000000526] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Hydrogen sulfide (H2S) is produced in the gut from cysteine by epithelial cells and by the intestinal microbiota. Initially considered as a toxic gas, the pleiotropic effects of H2S are now recognized, especially in the colonic mucosa. The aim of this review is to present new experimental data indicating that cysteine-derived H2S is emerging as a key regulator of gut health. RECENT FINDINGS Cysteine degradation by the microbiota emerged as a dominant pathway for H2S production. Among bacteria producing H2S from cysteine, Fusobacterium appears as a pivotal genus associated with digestive diseases. H2S promotes or alleviates mucosal inflammation, mostly according to its high (high micromolar to millimolar) or low (nanomolar to low micromolar) concentration, respectively. H2S maintains the integrity of the mucus layer when derived from endogenous metabolism but is detrimental for this parameter when produced in excess by gut microbes. In inflammatory bowel diseases, an upregulation of H2S production from cysteine by the gut microbiota is observed concomitantly with a downregulation of enzymes implicated in its mucosal detoxification. In colorectal cancer patients, an upregulation of both endogenous and microbial H2S production from cysteine are observed at tumor site that might contribute to disease progression. SUMMARY H2S is a double-edge sword for the intestinal epithelium. This is related to the bell-shaped effects of H2S, with protective effect at low concentration but deleterious effects at higher concentrations. As the gut microbiota produces much more H2S from cysteine than endogenous metabolism, we consider that the bacterial or epithelial source of H2S is a major determinant of its effects for intestinal health.
Collapse
Affiliation(s)
| | - Martin Beaumont
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Toulouse, France
| | - Eunjung Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, South Korea
| |
Collapse
|
93
|
Figliuolo VR, Coutinho-Silva R, Coutinho CMLM. Contribution of sulfate-reducing bacteria to homeostasis disruption during intestinal inflammation. Life Sci 2018; 215:145-151. [PMID: 30414430 DOI: 10.1016/j.lfs.2018.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022]
Abstract
Alteration in microbial populations and metabolism are key events associated with disruption of intestinal homeostasis and immune tolerance during intestinal inflammation. A substantial imbalance in bacterial populations in the intestine and their relationships with the host have been observed in patients with inflammatory bowel disease (IBD), believed to be part of an intricate mechanism of triggering and progression of intestinal inflammation. Because elevated numbers of sulfate-reducing bacteria (SRB) have been found in the intestines of patients with IBD, the study of their interaction with intestinal cells and their potential involvement in IBD has been the focus of investigation to better understand the intestinal pathology during IBD, as well as to find new ways to treat the disease. SRB not only directly interact with intestinal epithelial cells during intestinal inflammation but may also promote intestinal damage through generation of hydrogen sulfide at high levels. Herein we review the literature to discuss the various aspects of SRB interaction with host intestinal tissue, focusing on their interaction with intestinal epithelial and immune cells during intestinal inflammation.
Collapse
Affiliation(s)
- Vanessa Ribeiro Figliuolo
- Instituto de Biofísica Carlos Chagas Filho - IBCCF, Universidade Federal do Rio de Janeiro, RJ, Brazil; LITEB, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil; Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho - IBCCF, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Claudia Mara Lara Melo Coutinho
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niteroi, RJ, Brazil; LITEB, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
94
|
A pharmacological probe identifies cystathionine β-synthase as a new negative regulator for ferroptosis. Cell Death Dis 2018; 9:1005. [PMID: 30258181 PMCID: PMC6158189 DOI: 10.1038/s41419-018-1063-2] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/26/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022]
Abstract
Cystathionine β-synthase (CBS) is responsible for the first enzymatic reaction in the transsulfuration pathway of sulfur amino acids. The molecular function and mechanism of CBS as well as that of transsulfuration pathway remain ill-defined in cell proliferation and death. In the present study, we designed, synthesized and obtained a bioactive inhibitor CH004 for human CBS, which functions in vitro and in vivo. CH004 inhibits CBS activity, elevated the cellular homocysteine and suppressed the production of hydrogen sulfide in a dose-dependent manner in cells or in vivo. Chemical or genetic inhibition of CBS demonstrates that endogenous CBS is closely coupled with cell proliferation and cell cycle. Moreover, CH004 substantially retarded in vivo tumor growth in a xenograft mice model of liver cancer. Importantly, inhibition of CBS triggers ferroptosis in hepatocellular carcinoma. Overall, the study provides several clues for studying the interplays amongst transsulfuration pathway, ferroptosis and liver cancer.
Collapse
|
95
|
Wang G, Wang JJ, Yin PH, Xu K, Wang YZ, Shi F, Gao J, Fu XL. New strategies for targeting glucose metabolism-mediated acidosis for colorectal cancer therapy. J Cell Physiol 2018; 234:348-368. [PMID: 30069931 DOI: 10.1002/jcp.26917] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/13/2018] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is a heterogeneous group of diseases that are the result of abnormal glucose metabolism alterations with high lactate production by pyruvate to lactate conversion, which remodels acidosis and offers an evolutional advantage for tumor cells, even enhancing their aggressive phenotype. This review summarizes recent findings that involve multiple genes, molecules, and downstream signaling in the dysregulated glycolytic pathway, which can allow a tumor to initiate acid byproducts and to progress, thereby resulting in acidosis commonly found in the tumor microenvironment of CRC. Moreover, the relationship between CRC cells and the tumor acidic microenvironment, especially for regulating lactate production and lactate dehydrogenase A levels, is also discussed, as well as comprehensively defining different aspects of glycolytic pathways that affect cancer cell proliferation, invasion, and migration. Furthermore, this review concentrates on glucose metabolism-mediated transduction factors in CRC, which include acid-sensing ion channels, triosephosphate isomerase and key glycolysis-related enzymes that regulate glycolytic metabolites, coupled with the effect on tumor cell glycolysis as well as signaling pathways. In conclusion, glucose metabolism mediated by glycolytic pathways that are integral to tumor acidosis in CRC is demonstrated. Therefore, selective metabolic inhibitors or agents against these targets in glucose metabolism through glycolytic pathways may be clinically useful to regulate the tumor's acidic microenvironment for CRC treatment and to identify specific targets that regulate tumor acidosis through a cancer patient-personalized approach. Furthermore, strategies for modifying the metabolic processes that effectively inhibit cancer cell growth and tumor progression and activate potent anticancer effects may provide more effective antitumor prospects for CRC therapy.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Pei-Hao Yin
- Department of Cancer, Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Xu
- Department of Cancer, Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu-Zhu Wang
- Department of Medicine, Jiangsu University, Zhenjiang, China
| | - Feng Shi
- Department of Medicine, Jiangsu University, Zhenjiang, China
| | - Jing Gao
- Department of Medicine, Jiangsu University, Zhenjiang, China
| | - Xing-Li Fu
- Department of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
96
|
Cystathionine β-Synthase in Physiology and Cancer. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3205125. [PMID: 30050925 PMCID: PMC6046153 DOI: 10.1155/2018/3205125] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/29/2018] [Indexed: 01/20/2023]
Abstract
Cystathionine β-synthase (CBS) regulates homocysteine metabolism and contributes to hydrogen sulfide (H2S) biosynthesis through which it plays multifunctional roles in the regulation of cellular energetics, redox status, DNA methylation, and protein modification. Inactivating mutations in CBS contribute to the pathogenesis of the autosomal recessive disease CBS-deficient homocystinuria. Recent studies demonstrating that CBS promotes colon and ovarian cancer growth in preclinical models highlight a newly identified oncogenic role for CBS. On the contrary, tumor-suppressive effects of CBS have been reported in other cancer types, suggesting context-dependent roles of CBS in tumor growth and progression. Here, we review the physiological functions of CBS, summarize the complexities regarding CBS research in oncology, and discuss the potential of CBS and its key metabolites, including homocysteine and H2S, as potential biomarkers for cancer diagnosis or therapeutic targets for cancer treatment.
Collapse
|
97
|
Pan Y, Lu L, Chen J, Zhong Y, Dai Z. Identification of potential crucial genes and construction of microRNA-mRNA negative regulatory networks in osteosarcoma. Hereditas 2018; 155:21. [PMID: 29760609 PMCID: PMC5941338 DOI: 10.1186/s41065-018-0061-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/02/2018] [Indexed: 12/25/2022] Open
Abstract
Background This study aimed to identify potential crucial genes and construction of microRNA-mRNA negative regulatory networks in osteosarcoma by comprehensive bioinformatics analysis. Methods Data of gene expression profiles (GSE28424) and miRNA expression profiles (GSE28423) were downloaded from GEO database. The differentially expressed genes (DEGs) and miRNAs (DEMIs) were obtained by R Bioconductor packages. Functional and enrichment analyses of selected genes were performed using DAVID database. Protein-protein interaction (PPI) network was constructed by STRING and visualized in Cytoscape. The relationships among the DEGs and module in PPI network were analyzed by plug-in NetworkAnalyzer and MCODE seperately. Through the TargetScan and comparing target genes with DEGs, the miRNA-mRNA regulation network was established. Results Totally 346 DEGs and 90 DEMIs were found to be differentially expressed. These DEGs were enriched in biological processes and KEGG pathway of inflammatory immune response. 25 genes in the PPI network were selected as hub genes. Top 10 hub genes were TYROBP, HLA-DRA, VWF, PPBP, SERPING1, HLA-DPA1, SERPINA1, KIF20A, FERMT3, HLA-E. PPI network of DEGs followed a pattern of power law network and met the characteristics of small-world network. MCODE analysis identified 4 clusters and the most significant cluster consisted of 11 nodes and 55 edges. SEPP1, CKS2, TCAP, BPI were identified as the seed genes in their own clusters, respectively. The miRNA-mRNA regulation network which was composed of 89 pairs was established. MiR-210 had the highest connectivity with 12 target genes. Among the predicted target of MiR-96, HLA-DPA1 and TYROBP were the hub genes. Conclusion Our study indicated possible differentially expressed genes and miRNA, and microRNA-mRNA negative regulatory networks in osteosarcoma by bioinformatics analysis, which may provide novel insights for unraveling pathogenesis of osteosarcoma.
Collapse
Affiliation(s)
- Yue Pan
- 1Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410011 China
| | - Lingyun Lu
- Department of Orthopaedics, the Fifth Hospital of Xiamen, Xiamen, 361101 China
| | - Junquan Chen
- 1Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410011 China
| | - Yong Zhong
- 3Department of Nephrology, Xiangya Hospital of Central South University, Changsha, 410008 China
| | - Zhehao Dai
- 1Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410011 China
| |
Collapse
|
98
|
Myte R, Gylling B, Häggström J, Schneede J, Löfgren-Burström A, Huyghe JR, Hallmans G, Meyer K, Johansson I, Ueland PM, Palmqvist R, Van Guelpen B. One-carbon metabolism biomarkers and genetic variants in relation to colorectal cancer risk by KRAS and BRAF mutation status. PLoS One 2018; 13:e0196233. [PMID: 29694444 PMCID: PMC5919009 DOI: 10.1371/journal.pone.0196233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/09/2018] [Indexed: 12/20/2022] Open
Abstract
Disturbances in one-carbon metabolism, intracellular reactions involved in nucleotide synthesis and methylation, likely increase the risk of colorectal cancer (CRC). However, results have been inconsistent. To explore whether this inconsistency could be explained by intertumoral heterogeneity, we evaluated a comprehensive panel of one-carbon metabolism biomarkers and some single nucleotide polymorphisms (SNPs) in relation to the risk of molecular subtypes of CRC defined by mutations in the KRAS and BRAF oncogenes. This nested case-control study included 488 CRC cases and 947 matched controls from two population-based cohorts in the Northern Sweden Health and Disease Study. We analyzed 14 biomarkers and 17 SNPs in prediagnostic blood and determined KRAS and BRAF mutation status in tumor tissue. In a multivariate network analysis, no variable displayed a strong association with the risk of specific CRC subtypes. A non-synonymous SNP in the CTH gene, rs1021737, had a stronger association compared with other variables. In subsequent univariate analyses, participants with variant rs1021737 genotype had a decreased risk of KRAS-mutated CRC (OR per allele = 0.72, 95% CI = 0.50, 1.05), and an increased risk of BRAF-mutated CRC (OR per allele = 1.56, 95% CI = 1.07, 2.30), with weak evidence for heterogeneity (Pheterogeneity = 0.01). This subtype-specific SNP association was not replicated in a case-case analysis of 533 CRC cases from The Cancer Genome Atlas (P = 0.85). In conclusion, we found no support for clear subtype-specific roles of one-carbon metabolism biomarkers and SNPs in CRC development, making differences in CRC molecular subtype distributions an unlikely explanation for the varying results on the role of one-carbon metabolism in CRC development across previous studies. Further investigation of the CTH gene in colorectal carcinogenesis with regards to KRAS and BRAF mutations or other molecular characteristics of the tumor may be warranted.
Collapse
Affiliation(s)
- Robin Myte
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
- * E-mail:
| | - Björn Gylling
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Jenny Häggström
- Department of Statistics, Umeå School of Business and Economics, Umeå University, Umeå, Sweden
| | - Jörn Schneede
- Department of Clinical Pharmacology, Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden
| | | | - Jeroen R. Huyghe
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Göran Hallmans
- Department of Biobank Research, Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | | | - Per Magne Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | |
Collapse
|
99
|
Oláh G, Módis K, Törö G, Hellmich MR, Szczesny B, Szabo C. Role of endogenous and exogenous nitric oxide, carbon monoxide and hydrogen sulfide in HCT116 colon cancer cell proliferation. Biochem Pharmacol 2018; 149:186-204. [PMID: 29074106 PMCID: PMC5866187 DOI: 10.1016/j.bcp.2017.10.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/20/2017] [Indexed: 02/06/2023]
Abstract
The role of the three gasotransmitter systems - nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) - in cancer cells has not yet been studied simultaneously in the same experimental system. We measured the expression of NO and CO and H2S generating enzymes in primary colon cancer tissues and HCT116 colon cancer cells, and evaluated the effect of their pharmacological inhibition or pharmacological donation on cell proliferation. Increased expression of iNOS, nNOS, HO-1, CBS and 3-MST was detected in colon cancer. Inhibitors of NOS, HO-1/2, CBS/CSE and 3-MST, at lower concentrations, slightly stimulated HCT116 cell proliferation, but inhibited proliferation at higher concentrations. Donors of NO, CO or H2S inhibited HCT116 proliferation in a concentration-dependent manner. Inhibition of the cGMP/VASP pathway, Akt and p44/42 MAPK (Erk1/2) inhibited HCT116 cell proliferation. Endogenous NO and H2S biosynthesis were found to play a role in the maintenance of the activity of the cGMP/VASP pathway in HCT116 cells. We conclude that each of the three gasotransmitters play similar, bell-shaped roles in the control of HCT116 cell proliferation: endogenously produced NO, CO and H2S, at an optimal concentration, support HCT116 proliferation; inhibition of their production (which decreases gasotransmitter levels below optimal concentrations) as well as exogenous delivery of these gasotransmitters (which increases gasotransmitter levels above optimal concentrations) suppresses colon cancer cell proliferation. The current data give a mechanistic explanation for the paradoxical finding that both inhibitors and donors of NO, CO and H2S exert anticancer actions in cancer cells.
Collapse
Affiliation(s)
- Gabor Oláh
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Katalin Módis
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Gabor Törö
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mark R Hellmich
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Bartosz Szczesny
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|