1
|
El-Naggar AM, Li Y, Turgu B, Ding Y, Wei L, Chen SY, Trigo-Gonzalez G, Kalantari F, Vallejos R, Lynch B, Senz J, Lum A, Douglas JM, Salamanca C, Thornton S, Qin Y, Parmar K, Spencer SE, Leung S, Woo MM, Yong PJ, Zhang HF, Hughes CS, Negri GL, Wang Y, Morin GB, Sorensen PH, Huntsman DG. Cystathionine gamma-lyase-mediated hypoxia inducible factor 1-alpha expression drives clear cell ovarian cancer progression. J Pathol 2025. [PMID: 40371821 DOI: 10.1002/path.6433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 05/16/2025]
Abstract
Clear cell ovarian cancer (CCOC) is the second most common ovarian cancer subtype, accounting for 5%-11% of ovarian cancers in North America. Late-stage CCOC is associated with a worse prognosis compared to other ovarian cancer histotypes, a challenge that has seen limited progress in recent decades. CCOC typically originates within the toxic microenvironment of endometriotic ovarian cysts and is characterized by its intrinsic chemoresistance, a strong hypoxic signature, and abundant expression of cystathionine gamma-lyase (CTH). CTH is a key enzyme in the transsulfuration pathway and serves as a marker of ciliated cells derived from the Müllerian tract. CTH plays a pivotal role in de novo cysteine synthesis, which is essential for glutathione (GSH) production and redox homeostasis. Using an array of molecular tools and cancer models, including in vivo studies, we demonstrated that CTH expression was induced under various stress conditions, such as exposure to endometriotic cyst content and hypoxia. This induction enables cell survival and creates a differentiation state manifested by CCOC that potentiates tumor progression and metastasis. In addition to regulating redox homeostasis, CTH enhances hypoxia inducible factor 1-alpha (HIF1α) expression, independently of hydrogen sulfide (H2S) production. Re-expression of HIF1α in CTH KO cells fully restored metastatic capacity in in vivo models. Co-expression of CTH and HIF1α proteins was also observed in human CCOC samples. Importantly, targeting CTH in CCOC significantly reduced its metastatic potential in in vivo models and enhanced sensitivity to chemotherapy. These findings underscore that CTH is both a defining feature of CCOC and a promising therapeutic target, not only for CCOC patients but also for those with other CTH-expressing cancers. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Amal M El-Naggar
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | - Yuqin Li
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Busra Turgu
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuchen Ding
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Longyijie Wei
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Shary Yuting Chen
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Genny Trigo-Gonzalez
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Forouh Kalantari
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Rodrigo Vallejos
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Branden Lynch
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Janine Senz
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Amy Lum
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - J Maxwell Douglas
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Clara Salamanca
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Shelby Thornton
- Molecular and Advanced Pathology Core (MAPCore), University of British Columbia, Vancouver, BC, Canada
| | - Yimei Qin
- Molecular and Advanced Pathology Core (MAPCore), University of British Columbia, Vancouver, BC, Canada
| | - Kiran Parmar
- BC Centre for Pelvic Pain and Endometriosis, BC Women's Hospital, Vancouver, BC, Canada
| | - Sandra E Spencer
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Samuel Leung
- Genetic Pathology Evaluation Centre, Vancouver, BC, Canada
| | - Michelle Mm Woo
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
- BC's Gynecologic Cancer Research Program, OVCARE, Vancouver, BC, Canada
| | - Paul J Yong
- BC Centre for Pelvic Pain and Endometriosis, BC Women's Hospital, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Hai-Feng Zhang
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Gian Luca Negri
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Yemin Wang
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gregg B Morin
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Molecular and Advanced Pathology Core (MAPCore), University of British Columbia, Vancouver, BC, Canada
- Genetic Pathology Evaluation Centre, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
- BC's Gynecologic Cancer Research Program, OVCARE, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Hydrogen Sulfide (H 2S- or H 2S n-Polysulfides) in Synaptic Plasticity: Modulation of NMDA Receptors and Neurotransmitter Release in Learning and Memory. Int J Mol Sci 2025; 26:3131. [PMID: 40243915 PMCID: PMC11988931 DOI: 10.3390/ijms26073131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Hydrogen sulfide (H2S) has emerged as a pivotal gaseous transmitter in the central nervous system, influencing synaptic plasticity, learning, and memory by modulating various molecular pathways. This review examines recent evidence regarding how H2S regulates NMDA receptor function and neurotransmitter release in neuronal circuits. By synthesizing findings from animal and cellular models, we investigate the impacts of enzymatic H2S production and exogenous H2S on excitatory synaptic currents, long-term potentiation, and intracellular calcium signaling. Data suggest that H2S interacts directly with NMDA receptor subunits, altering receptor function and modulating neuronal excitability. Simultaneously, H2S promotes the release of neurotransmitters such as glutamate and GABA, shaping synaptic dynamics and plasticity. Furthermore, reports indicate that disruptions in H2S metabolism contribute to cognitive impairments and neurodegenerative disorders, underscoring the potential therapeutic value of targeting H2S-mediated pathways. Although the precise mechanisms of H2S-induced changes in synaptic strength remain elusive, a growing body of evidence positions H2S as a significant regulator of memory formation processes. This review calls for more rigorous exploration into the molecular underpinnings of H2S in synaptic plasticity, paving the way for novel pharmacological interventions in cognitive dysfunction.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
3
|
Munteanu C, Popescu C, Vlădulescu-Trandafir AI, Onose G. Signaling Paradigms of H 2S-Induced Vasodilation: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1158. [PMID: 39456412 PMCID: PMC11505308 DOI: 10.3390/antiox13101158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Hydrogen sulfide (H2S), a gas traditionally considered toxic, is now recognized as a vital endogenous signaling molecule with a complex physiology. This comprehensive study encompasses a systematic literature review that explores the intricate mechanisms underlying H2S-induced vasodilation. The vasodilatory effects of H2S are primarily mediated by activating ATP-sensitive potassium (K_ATP) channels, leading to membrane hyperpolarization and subsequent relaxation of vascular smooth muscle cells (VSMCs). Additionally, H2S inhibits L-type calcium channels, reducing calcium influx and diminishing VSMC contraction. Beyond ion channel modulation, H2S profoundly impacts cyclic nucleotide signaling pathways. It stimulates soluble guanylyl cyclase (sGC), increasing the production of cyclic guanosine monophosphate (cGMP). Elevated cGMP levels activate protein kinase G (PKG), which phosphorylates downstream targets like vasodilator-stimulated phosphoprotein (VASP) and promotes smooth muscle relaxation. The synergy between H2S and nitric oxide (NO) signaling further amplifies vasodilation. H2S enhances NO bioavailability by inhibiting its degradation and stimulating endothelial nitric oxide synthase (eNOS) activity, increasing cGMP levels and potent vasodilatory responses. Protein sulfhydration, a post-translational modification, plays a crucial role in cell signaling. H2S S-sulfurates oxidized cysteine residues, while polysulfides (H2Sn) are responsible for S-sulfurating reduced cysteine residues. Sulfhydration of key proteins like K_ATP channels and sGC enhances their activity, contributing to the overall vasodilatory effect. Furthermore, H2S interaction with endothelium-derived hyperpolarizing factor (EDHF) pathways adds another layer to its vasodilatory mechanism. By enhancing EDHF activity, H2S facilitates the hyperpolarization and relaxation of VSMCs through gap junctions between endothelial cells and VSMCs. Recent findings suggest that H2S can also modulate transient receptor potential (TRP) channels, particularly TRPV4 channels, in endothelial cells. Activating these channels by H2S promotes calcium entry, stimulating the production of vasodilatory agents like NO and prostacyclin, thereby regulating vascular tone. The comprehensive understanding of H2S-induced vasodilation mechanisms highlights its therapeutic potential. The multifaceted approach of H2S in modulating vascular tone presents a promising strategy for developing novel treatments for hypertension, ischemic conditions, and other vascular disorders. The interaction of H2S with ion channels, cyclic nucleotide signaling, NO pathways, ROS (Reactive Oxygen Species) scavenging, protein sulfhydration, and EDHF underscores its complexity and therapeutic relevance. In conclusion, the intricate signaling paradigms of H2S-induced vasodilation offer valuable insights into its physiological role and therapeutic potential, promising innovative approaches for managing various vascular diseases through the modulation of vascular tone.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
| | - Cristina Popescu
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Andreea-Iulia Vlădulescu-Trandafir
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| |
Collapse
|
4
|
Munteanu C, Galaction AI, Poștaru M, Rotariu M, Turnea M, Blendea CD. Hydrogen Sulfide Modulation of Matrix Metalloproteinases and CD147/EMMPRIN: Mechanistic Pathways and Impact on Atherosclerosis Progression. Biomedicines 2024; 12:1951. [PMID: 39335465 PMCID: PMC11429404 DOI: 10.3390/biomedicines12091951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory condition marked by endothelial dysfunction, lipid accumulation, inflammatory cell infiltration, and extracellular matrix (ECM) remodeling within arterial walls, leading to plaque formation and potential cardiovascular events. Key players in ECM remodeling and inflammation are matrix metalloproteinases (MMPs) and CD147/EMMPRIN, a cell surface glycoprotein expressed on endothelial cells, vascular smooth muscle cells (VSMCs), and immune cells, that regulates MMP activity. Hydrogen sulfide (H₂S), a gaseous signaling molecule, has emerged as a significant modulator of these processes including oxidative stress mitigation, inflammation reduction, and vascular remodeling. This systematic review investigates the mechanistic pathways through which H₂S influences MMPs and CD147/EMMPRIN and assesses its impact on atherosclerosis progression. A comprehensive literature search was conducted across PubMed, Scopus, and Web of Science databases, focusing on studies examining H₂S modulation of MMPs and CD147/EMMPRIN in atherosclerosis contexts. Findings indicate that H₂S modulates MMP expression and activity through transcriptional regulation and post-translational modifications, including S-sulfhydration. By mitigating oxidative stress, H₂S reduces MMP activation, contributing to plaque stability and vascular remodeling. H₂S also downregulates CD147/EMMPRIN expression via transcriptional pathways, diminishing inflammatory responses and vascular cellular proliferation within plaques. The dual regulatory role of H₂S in inhibiting MMP activity and downregulating CD147 suggests its potential as a therapeutic agent in stabilizing atherosclerotic plaques and mitigating inflammation. Further research is warranted to elucidate the precise molecular mechanisms and to explore H₂S-based therapies for clinical application in atherosclerosis.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Mădălina Poștaru
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Corneliu Dan Blendea
- Department of Medical-Clinical Disciplines, General Surgery, Faculty of Medicine, "Titu Maiorescu" University of Bucharest, 0400511 Bucharest, Romania
| |
Collapse
|
5
|
Ye S, Jin N, Liu N, Cheng F, Hu L, Zhang G, Li Q, Jing J. Gases and gas-releasing materials for the treatment of chronic diabetic wounds. Biomater Sci 2024; 12:3273-3292. [PMID: 38727636 DOI: 10.1039/d4bm00351a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Chronic non-healing wounds are a common consequence of skin ulceration in diabetic patients, with severe cases such as diabetic foot even leading to amputations. The interplay between pathological factors like hypoxia-ischemia, chronic inflammation, bacterial infection, impaired angiogenesis, and accumulation of advanced glycosylation end products (AGEs), resulting from the dysregulation of the immune microenvironment caused by hyperglycemia, establishes an unending cycle that hampers wound healing. However, there remains a dearth of sufficient and effective approaches to break this vicious cycle within the complex immune microenvironment. Consequently, numerous scholars have directed their research efforts towards addressing chronic diabetic wound repair. In recent years, gases including Oxygen (O2), Nitric oxide (NO), Hydrogen (H2), Hydrogen sulfide (H2S), Ozone (O3), Carbon monoxide (CO) and Nitrous oxide (N2O), along with gas-releasing materials associated with them have emerged as promising therapeutic solutions due to their ability to regulate angiogenesis, intracellular oxygenation levels, exhibit antibacterial and anti-inflammatory effects while effectively minimizing drug residue-induced damage and circumventing drug resistance issues. In this review, we discuss the latest advances in the mechanisms of action and treatment of these gases and related gas-releasing materials in diabetic wound repair. We hope that this review can provide different ideas for the future design and application of gas therapy for chronic diabetic wounds.
Collapse
Affiliation(s)
- Shuming Ye
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Neng Jin
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Nan Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Feixiang Cheng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Liang Hu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Guiyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Qi Li
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Juehua Jing
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
6
|
Soldatov AA, Shalagina NE, Rychkova VN, Kukhareva TA. Membrane-Bound Ferric Hemoglobin in Nucleated Erythrocytes of the Black Scorpionfish Scorpaena porcus, Linnaeus 1758. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2024; 516:50-54. [PMID: 38700814 DOI: 10.1134/s0012496624700984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 05/26/2024]
Abstract
The content of membrane-bound methemoglobin (MtHb) in nucleated erythrocytes was studied in the black scorpionfish Scorpaena porcus (Linnaeus, 1758) in vitro. Spectral characteristics were determined for a whole hemolysate, a hemolysate obtained by stroma precipitation (a clarified hemolysate), and a resuspended stroma. The MtHb proportion in the erythrocyte stroma was found to exceed 80% (6.20 ± 0.59 µM). Clarified hemolysates were nearly free of MtHb (0.5 ± 0.2 µM). Membrane-bound ferric hemoglobin did not affect the erythrocyte resistance to osmotic shock. The osmotic fragility range was determined using a LaSca-TM laser microparticle analyzer (BioMedSystems, Russia) to be 102-136 mOsm/kg, much the same as in other bony fish species. A nitrite load (10 mg/L) significantly increased the MtHb content in the blood. However, the membrane-bound ferric hemoglobin content did not change significantly, amounting to 6.34 ± 1.09 µM (approximately 95%). The finding suggested a functional importance for MtHb present in the plasma membrane of nucleated erythrocytes. Membrane-bound MtHb was assumed to neutralize the external oxidative load and the toxic effect of hydrogen sulfide in bottom water layers, where the species lives.
Collapse
Affiliation(s)
- A A Soldatov
- Kovalevsky Institute of Biology of the South Seas, Russian Academy of Sciences, Sevastopol, Russia.
- Sevastopol State University, Sevastopol, Russia.
| | - N E Shalagina
- Kovalevsky Institute of Biology of the South Seas, Russian Academy of Sciences, Sevastopol, Russia
| | - V N Rychkova
- Kovalevsky Institute of Biology of the South Seas, Russian Academy of Sciences, Sevastopol, Russia
| | - T A Kukhareva
- Kovalevsky Institute of Biology of the South Seas, Russian Academy of Sciences, Sevastopol, Russia
| |
Collapse
|
7
|
Yang H, Tan M, Gao Z, Wang S, Lyu L, Ding H. Role of Hydrogen Sulfide and Hypoxia in Hepatic Angiogenesis of Portal Hypertension. J Clin Transl Hepatol 2023; 11:675-681. [PMID: 36969894 PMCID: PMC10037502 DOI: 10.14218/jcth.2022.00217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/23/2022] [Accepted: 09/21/2022] [Indexed: 01/05/2023] Open
Abstract
The pathogenesis of portal hypertension remains unclear, and is believed to involve dysfunction of liver sinusoidal endothelial cells (LSEC), activation of hepatic stellate cells (HSC), dysregulation of endogenous hydrogen sulfide (H2S) synthesis, and hypoxia-induced angiogenic responses. H2S, a novel gas transmitter, plays an important role in various pathophysiological processes, especially in hepatic angiogenesis. Inhibition of endogenous H2S synthase by pharmaceutical agents or gene silencing may enhance the angiogenic response of endothelial cells. Hypoxia-inducible factor-1 (HIF-1) is the main transcription factor of hypoxia, which induces hepatic angiogenesis through up-regulation of vascular endothelial growth factor (VEGF) in HSC and LSEC. H2S has also been shown to be involved in the regulation of VEGF-mediated angiogenesis. Therefore, H2S and HIF-1 may be potential therapeutic targets for portal hypertension. The effects of H2S donors or prodrugs on the hemodynamics of portal hypertension and the mechanism of H2S-induced angiogenesis are promising areas for future research.
Collapse
Affiliation(s)
- Huaxiang Yang
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Mingjie Tan
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhuqing Gao
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Shanshan Wang
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
- Cell Biology Laboratory, Beijing Institute of Hepatology, Beijing, China
| | - Lingna Lyu
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Gáll T, Nagy P, Garai D, Potor L, Balla GJ, Balla G, Balla J. Overview on hydrogen sulfide-mediated suppression of vascular calcification and hemoglobin/heme-mediated vascular damage in atherosclerosis. Redox Biol 2022; 57:102504. [PMID: 36240620 PMCID: PMC9576974 DOI: 10.1016/j.redox.2022.102504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 12/04/2022] Open
Abstract
Vulnerable atherosclerotic plaques with hemorrhage considerably contribute to cardiovascular morbidity and mortality. Calcification is the main characteristic of advanced atherosclerotic lesions and calcified aortic valve disease (CAVD). Lyses of red blood cells and hemoglobin (Hb) release occur in human hemorrhagic complicated lesions. During the interaction of cell-free Hb with plaque constituents, Hb is oxidized to ferric and ferryl states accompanied by oxidative changes of the globin moieties and heme release. Accumulation of both ferryl-Hb and metHb has been observed in atherosclerotic plaques. The oxidation hotspots in the globin chain are the cysteine and tyrosine amino acids associated with the generation of Hb dimers, tetramers and polymers. Moreover, fragmentation of Hb occurs leading to the formation of globin-derived peptides. A series of these pro-atherogenic cellular responses can be suppressed by hydrogen sulfide (H2S). Since H2S has been explored to exhibit a wide range of physiologic functions to maintain vascular homeostasis, it is not surprising that H2S may play beneficial effects in the progression of atherosclerosis. In the present review, we summarize the findings about the effects of H2S on atherosclerosis and CAVD with a special emphasis on the oxidation of Hb/heme in atherosclerotic plaque development and vascular calcification.
Collapse
Affiliation(s)
- Tamás Gáll
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary; Institute of Oncochemistry, University of Debrecen, Hungary
| | - Dorottya Garai
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary
| | - László Potor
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | | | - György Balla
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - József Balla
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
9
|
Xu YR, Wang AL, Li YQ. Hypoxia-inducible factor 1-alpha is a driving mechanism linking chronic obstructive pulmonary disease to lung cancer. Front Oncol 2022; 12:984525. [PMID: 36338690 PMCID: PMC9634253 DOI: 10.3389/fonc.2022.984525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/10/2022] [Indexed: 11/27/2022] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD), irrespective of their smoking history, are more likely to develop lung cancer than the general population. This is mainly because COPD is characterized by chronic persistent inflammation and hypoxia, which are the risk factors for lung cancer. However, the mechanisms underlying this observation are still unknown. Hypoxia-inducible factor 1-alpha (HIF-1α) plays an important role in the crosstalk that exists between inflammation and hypoxia. Furthermore, HIF-1α is the main regulator of somatic adaptation to hypoxia and is highly expressed in hypoxic environments. In this review, we discuss the molecular aspects of the crosstalk between hypoxia and inflammation, showing that HIF-1α is an important signaling pathway that drives COPD progression to lung cancer. Here, we also provide an overview of HIF-1α and its principal regulatory mechanisms, briefly describe HIF-1α-targeted therapy in lung cancer, and summarize substances that may be used to target HIF-1α at the level of COPD-induced inflammation.
Collapse
Affiliation(s)
- Yuan-rui Xu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - An-long Wang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Ya-qing Li
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- *Correspondence: Ya-qing Li,
| |
Collapse
|
10
|
Katsouda A, Valakos D, Dionellis VS, Bibli SI, Akoumianakis I, Karaliota S, Zuhra K, Fleming I, Nagahara N, Havaki S, Gorgoulis VG, Thanos D, Antoniades C, Szabo C, Papapetropoulos A. MPST sulfurtransferase maintains mitochondrial protein import and cellular bioenergetics to attenuate obesity. J Exp Med 2022; 219:e20211894. [PMID: 35616614 PMCID: PMC9143789 DOI: 10.1084/jem.20211894] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/16/2022] [Accepted: 04/27/2022] [Indexed: 11/04/2022] Open
Abstract
Given the clinical, economic, and societal impact of obesity, unraveling the mechanisms of adipose tissue expansion remains of fundamental significance. We previously showed that white adipose tissue (WAT) levels of 3-mercaptopyruvate sulfurtransferase (MPST), a mitochondrial cysteine-catabolizing enzyme that yields pyruvate and sulfide species, are downregulated in obesity. Here, we report that Mpst deletion results in fat accumulation in mice fed a high-fat diet (HFD) through transcriptional and metabolic maladaptation. Mpst-deficient mice on HFD exhibit increased body weight and inguinal WAT mass, reduced metabolic rate, and impaired glucose/insulin tolerance. At the molecular level, Mpst ablation activates HIF1α, downregulates subunits of the translocase of outer/inner membrane (TIM/TOM) complex, and impairs mitochondrial protein import. MPST deficiency suppresses the TCA cycle, oxidative phosphorylation, and fatty acid oxidation, enhancing lipid accumulation. Sulfide donor administration to obese mice reverses the HFD-induced changes. These findings reveal the significance of MPST for white adipose tissue biology and metabolic health and identify a potential new therapeutic target for obesity.
Collapse
Affiliation(s)
- Antonia Katsouda
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Valakos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Sofia-Iris Bibli
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Ioannis Akoumianakis
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sevasti Karaliota
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute/National Institutes of Health, Frederick, MD
| | - Karim Zuhra
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research Partner Site Rhein-Main, Frankfurt am Main, Germany
| | | | - Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis G. Gorgoulis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Thanos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
11
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
12
|
Arif HM, Qian Z, Wang R. Signaling Integration of Hydrogen Sulfide and Iron on Cellular Functions. Antioxid Redox Signal 2022; 36:275-293. [PMID: 34498949 DOI: 10.1089/ars.2021.0203] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Hydrogen sulfide (H2S) is an endogenous signaling molecule, regulating numerous physiological functions from vasorelaxation to neuromodulation. Iron is a well-known bioactive metal ion, being the central component of hemoglobin for oxygen transportation and participating in biomolecule degradation, redox balance, and enzymatic actions. The interplay between H2S and iron metabolisms and functions impacts significantly on the fate and wellness of different types of cells. Recent Advances: Iron level in vivo affects the production of H2S via nonenzymatic reactions. On the contrary, H2S quenches excessive iron inside the cells and regulates the redox status of iron. Critical Issues: Abnormal metabolisms of both iron and H2S are associated with various conditions and diseases such as iron overload, anemia, oxidative stress, and cardiovascular and neurodegenerative diseases. The molecular mechanisms for the interactions between H2S and iron are unsettled yet. Here we review signaling links of the production, metabolism, and their respective and integrative functions of H2S and iron in normalcy and diseases. Future Directions: Physiological and pathophysiological importance of H2S and iron as well as their therapeutic applications should be evaluated jointly, not separately. Future investigation should expand from iron-rich cells and tissues to the others, in which H2S and iron interaction has not received due attention. Antioxid. Redox Signal. 36, 275-293.
Collapse
Affiliation(s)
| | - Zhongming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, China
| | - Rui Wang
- Department of Biology, York University, Toronto, Canada
| |
Collapse
|
13
|
Lu X, Wu M, Wang S, Qin J, Li P. Synthesis and preliminary exploration of a NIR fluorescent probe for the evaluation of androgen dependence of prostate cancer. Talanta 2021; 239:123058. [PMID: 34823861 DOI: 10.1016/j.talanta.2021.123058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/03/2021] [Accepted: 11/10/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Castration resistance prostate cancer patients showing resistance to the androgen deprivation therapy always have low five-year survival rate and worse prognosis. A responsive NIR fluorescent probe was designed to report the androgen dependence and monitor the development of castration resistance for prostate cancer. METHODS Intratumoral H2S in prostate cancer was closely related to castration resistance. A H2S-responsive NIR probe (HM) was developed as a dependent indicator to report the androgen dependence of prostate cancer. The specificity of HM to H2S and the influence of normal intracellular substrates to the response between H2S and HM were determined. Cell/in vivo animal imaging were performed on PC-3 and LnCAP cell/tumor bearing mice, which presented with androgen independence and androgen dependence, respectively. RESULTS When HM responded to H2S, strong fluorescence at 770 nm could be rapidly turned on in 5 min with the stokes shift as large as 200 nm. The recognition between HM and H2S showed high specificity. Neither other common substrates showed capacity to turn on HM's fluorescence, nor their existence demonstrated competition. The fluorescence intensity was linearly dependent to the H2S concentration and the limited of detection was 0.15 μM. When HM was applied to PC-3/LNCaP prostate cancer cell and tumor, the intracellular and intratumoral H2S could be clearly imaged and monitored. CONCLUSION HM showing obvious fluorescent behaviors in androgen dependence and independence prostate tumor, which could work as an indicator to reported the androgen dependence of prostate cancer and monitor the development of castration resistance.
Collapse
Affiliation(s)
- Xinmiao Lu
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Muyu Wu
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Siwen Wang
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jingcan Qin
- School of Medicine, Shanghai Jiao Tong University, South Chongqing Road, Shanghai, 200025, China.
| | - Peiyong Li
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
14
|
Olson KR. A Case for Hydrogen Sulfide Metabolism as an Oxygen Sensing Mechanism. Antioxidants (Basel) 2021; 10:antiox10111650. [PMID: 34829521 PMCID: PMC8615108 DOI: 10.3390/antiox10111650] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022] Open
Abstract
The ability to detect oxygen availability is a ubiquitous attribute of aerobic organisms. However, the mechanism(s) that transduce oxygen concentration or availability into appropriate physiological responses is less clear and often controversial. This review will make the case for oxygen-dependent metabolism of hydrogen sulfide (H2S) and polysulfides, collectively referred to as reactive sulfur species (RSS) as a physiologically relevant O2 sensing mechanism. This hypothesis is based on observations that H2S and RSS metabolism is inversely correlated with O2 tension, exogenous H2S elicits physiological responses identical to those produced by hypoxia, factors that affect H2S production or catabolism also affect tissue responses to hypoxia, and that RSS efficiently regulate downstream effectors of the hypoxic response in a manner consistent with a decrease in O2. H2S-mediated O2 sensing is then compared to the more generally accepted reactive oxygen species (ROS) mediated O2 sensing mechanism and a number of reasons are offered to resolve some of the confusion between the two.
Collapse
Affiliation(s)
- Kenneth R Olson
- Department of Physiology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, USA
| |
Collapse
|
15
|
Dai J, Teng X, Jin S, Wu Y. The Antiviral Roles of Hydrogen Sulfide by Blocking the Interaction between SARS-CoV-2 and Its Potential Cell Surface Receptors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7866992. [PMID: 34497683 PMCID: PMC8421161 DOI: 10.1155/2021/7866992] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is posing a great threat to the global economy and public health security. Together with the acknowledged angiotensin-converting enzyme 2, glucose-regulated protein 78, transferrin receptor, AXL, kidney injury molecule-1, and neuropilin 1 are also identified as potential receptors to mediate SARS-CoV-2 infection. Therefore, how to inhibit or delay the binding of SARS-CoV-2 with the abovementioned receptors is a key step for the prevention and treatment of COVID-19. As the third gasotransmitter, hydrogen sulfide (H2S) plays an important role in many physiological and pathophysiological processes. Recently, survivors were reported to have significantly higher H2S levels in COVID-19 patients, and mortality was significantly greater among patients with decreased H2S levels. Considering that the beneficial role of H2S against COVID-19 and COVID-19-induced comorbidities and multiorgan damage has been well-examined and reported in some excellent reviews, this review will discuss the recent findings on the potential receptors of SARS-CoV-2 and how H2S modulates the above receptors, in turn blocking SARS-CoV-2 entry into host cells.
Collapse
Affiliation(s)
- Jing Dai
- Department of Clinical Diagnostics, Hebei Medical University, Hebei 050017, China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, Hebei 050017, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei 050017, China
- Key Laboratory of Vascular Medicine of Hebei Province, Hebei 050017, China
| |
Collapse
|
16
|
Kuschman HP, Palczewski MB, Thomas DD. Nitric oxide and hydrogen sulfide: Sibling rivalry in the family of epigenetic regulators. Free Radic Biol Med 2021; 170:34-43. [PMID: 33482335 DOI: 10.1016/j.freeradbiomed.2021.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 01/12/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) were previously only known for their toxic properties. Now they are regarded as potent gaseous messenger molecules (gasotransmitters) that rapidly transverse cell membranes and transduce cellular signals through their chemical reactions and modifications to protein targets. Both are known to regulate numerous physiological functions including angiogenesis, vascular tone, and immune response, to name a few. NO and H2S often work synergistically and in competition to regulate each other's synthesis, target protein activity via posttranslational modifications (PTMs), and chemical interactions. In addition to their canonical modes of action, increasing evidence has demonstrated that NO and H2S share another signaling mechanism: epigenetic regulation. This review will compare and contrast biosynthesis and metabolism of NO and H2S, their individual and shared interactions, and the growing body of evidence for their roles as endogenous epigenetic regulatory molecules.
Collapse
Affiliation(s)
- Hannah Petraitis Kuschman
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Marianne B Palczewski
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Douglas D Thomas
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States.
| |
Collapse
|
17
|
Rahman MA, Glasgow JN, Nadeem S, Reddy VP, Sevalkar RR, Lancaster JR, Steyn AJC. The Role of Host-Generated H 2S in Microbial Pathogenesis: New Perspectives on Tuberculosis. Front Cell Infect Microbiol 2020; 10:586923. [PMID: 33330130 PMCID: PMC7711268 DOI: 10.3389/fcimb.2020.586923] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
For centuries, hydrogen sulfide (H2S) was considered primarily as a poisonous gas and environmental hazard. However, with the discovery of prokaryotic and eukaryotic enzymes for H2S production, breakdown, and utilization, H2S has emerged as an important signaling molecule in a wide range of physiological and pathological processes. Hence, H2S is considered a gasotransmitter along with nitric oxide (•NO) and carbon monoxide (CO). Surprisingly, despite having overlapping functions with •NO and CO, the role of host H2S in microbial pathogenesis is understudied and represents a gap in our knowledge. Given the numerous reports that followed the discovery of •NO and CO and their respective roles in microbial pathogenesis, we anticipate a rapid increase in studies that further define the importance of H2S in microbial pathogenesis, which may lead to new virulence paradigms. Therefore, this review provides an overview of sulfide chemistry, enzymatic production of H2S, and the importance of H2S in metabolism and immunity in response to microbial pathogens. We then describe our current understanding of the role of host-derived H2S in tuberculosis (TB) disease, including its influences on host immunity and bioenergetics, and on Mycobacterium tuberculosis (Mtb) growth and survival. Finally, this review discusses the utility of H2S-donor compounds, inhibitors of H2S-producing enzymes, and their potential clinical significance.
Collapse
Affiliation(s)
| | - Joel N Glasgow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sajid Nadeem
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vineel P Reddy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ritesh R Sevalkar
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jack R Lancaster
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Adrie J C Steyn
- Africa Health Research Institute, Durban, South Africa.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States.,Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
18
|
Dey A, Prabhudesai S, Zhang Y, Rao G, Thirugnanam K, Hossen MN, Dwivedi SKD, Ramchandran R, Mukherjee P, Bhattacharya R. Cystathione β-synthase regulates HIF-1α stability through persulfidation of PHD2. SCIENCE ADVANCES 2020; 6:eaaz8534. [PMID: 32937467 PMCID: PMC7458453 DOI: 10.1126/sciadv.aaz8534] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 05/20/2020] [Indexed: 05/10/2023]
Abstract
The stringent expression of the hypoxia inducible factor-1α (HIF-1α) is critical to a variety of pathophysiological conditions. We reveal that, in normoxia, enzymatic action of cystathionine β-synthase (CBS) produces H2S, which persulfidates prolyl hydroxylase 2 (PHD2) at residues Cys21 and Cys33 (zinc finger motif), augmenting prolyl hydroxylase activity. Depleting endogenous H2S either by hypoxia or by inhibiting CBS via chemical or genetic means reduces persulfidation of PHD2 and inhibits activity, preventing hydroxylation of HIF-1α, resulting in stabilization. Our in vitro findings are further supported by the depletion of CBS in the zebrafish model that exhibits axis defects and abnormal intersegmental vessels. Exogenous H2S supplementation rescues both in vitro and in vivo phenotypes. We have identified the persulfidated residues and defined their functional significance in regulating the activity of PHD2 via point mutations. Thus, the CBS/H2S/PHD2 axis may provide therapeutic opportunities for pathologies associated with HIF-1α dysregulation in chronic diseases.
Collapse
Affiliation(s)
- Anindya Dey
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Shubhangi Prabhudesai
- Department of Pediatrics, Department of Obstetrics and Gynecology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yushan Zhang
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Geeta Rao
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Karthikeyan Thirugnanam
- Department of Pediatrics, Department of Obstetrics and Gynecology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Md Nazir Hossen
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Ramani Ramchandran
- Department of Pediatrics, Department of Obstetrics and Gynecology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA.
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
19
|
Hydrogen sulfide dysregulates the immune response by suppressing central carbon metabolism to promote tuberculosis. Proc Natl Acad Sci U S A 2020; 117:6663-6674. [PMID: 32139610 PMCID: PMC7104411 DOI: 10.1073/pnas.1919211117] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ubiquitous gasotransmitter hydrogen sulfide (H2S) has been recognized to play a crucial role in human health. Using cystathionine γ-lyase (CSE)-deficient mice, we demonstrate an unexpected role of H2S in Mycobacterium tuberculosis (Mtb) pathogenesis. We showed that Mtb-infected CSE-/- mice survive longer than WT mice, and support reduced pathology and lower bacterial burdens in the lung, spleen, and liver. Similarly, in vitro Mtb infection of macrophages resulted in reduced colony forming units in CSE-/- cells. Chemical complementation of infected WT and CSE-/- macrophages using the slow H2S releaser GYY3147 and the CSE inhibitor DL-propargylglycine demonstrated that H2S is the effector molecule regulating Mtb survival in macrophages. Furthermore, we demonstrate that CSE promotes an excessive innate immune response, suppresses the adaptive immune response, and reduces circulating IL-1β, IL-6, TNF-α, and IFN-γ levels in response to Mtb infection. Notably, Mtb infected CSE-/- macrophages show increased flux through glycolysis and the pentose phosphate pathway, thereby establishing a critical link between H2S and central metabolism. Our data suggest that excessive H2S produced by the infected WT mice reduce HIF-1α levels, thereby suppressing glycolysis and production of IL-1β, IL-6, and IL-12, and increasing bacterial burden. Clinical relevance was demonstrated by the spatial distribution of H2S-producing enzymes in human necrotic, nonnecrotic, and cavitary pulmonary tuberculosis (TB) lesions. In summary, CSE exacerbates TB pathogenesis by altering immunometabolism in mice and inhibiting CSE or modulating glycolysis are potential targets for host-directed TB control.
Collapse
|
20
|
Wang Y, Tang S, Wu Y, Wan X, Zhou M, Li H, Zha X. Upregulation of 6-phosphofructo-2-kinase (PFKFB3) by hyperactivated mammalian target of rapamycin complex 1 is critical for tumor growth in tuberous sclerosis complex. IUBMB Life 2020; 72:965-977. [PMID: 31958214 DOI: 10.1002/iub.2232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by the benign tumor formation in multiple organs. The main etiology of TSC is the loss-of-function mutation of TSC1 or TSC2 gene, which leads to aberrant activation of mammalian target of rapamycin complex 1 (mTORC1). In this research, we found a significant increase of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) expression in Tsc1-/- and Tsc2-/- mouse embryonic fibroblasts (MEFs) compared with the control cells. Inhibition of mTORC1 led to a dramatic decrease of PFKFB3 expression, indicating PFKFB3 regulation by mTORC1. Moreover, suppression of mTORC1 inhibited the expression of PFKFB3 in rat uterine leiomyoma-derived Tsc2-null ELT3 cells and human tumor cells. Furthermore, we identified hypoxia-inducible factor 1α (HIF-1α) as a mediator transmitting the signal from mTORC1 to PFKFB3. Depletion of PFKFB3 inhibited proliferation and tumorigenicity of Tsc1- or Tsc2-deficient cells. In addition, combination of rapamycin with PFK15, a PFKFB3 inhibitor, exerts a stronger inhibitory effect on cell proliferation of Tsc1- or Tsc2-null MEFs than treatment with single drug. We conclude that loss of TSC1 or TSC2 led to upregulated expression of PFKFB3 through activation of mTORC1/HIF-1α signaling pathway and co-administration of rapamycin and PFK15 may be a promising strategy for the treatment of TSC tumors as well as other hyperactivated mTORC1-related tumors.
Collapse
Affiliation(s)
- Yani Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Sisi Tang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Yuncui Wu
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Xiaofeng Wan
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Meng Zhou
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Hongwu Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Giuffrè A, Tomé CS, Fernandes DGF, Zuhra K, Vicente JB. Hydrogen Sulfide Metabolism and Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:335-353. [PMID: 32130707 DOI: 10.1007/978-3-030-34025-4_17] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogen sulfide (H2S), while historically perceived merely as a toxicant, has progressively emerged as a key regulator of numerous processes in mammalian physiology, exerting its signaling function essentially through interaction with and/or modification of proteins, targeting mainly cysteine residues and metal centers. As a gaseous signaling molecule that freely diffuses across aqueous and hydrophobic biological milieu, it has been designated the third 'gasotransmitter' in mammalian physiology. H2S is synthesized and detoxified by specialized endogenous enzymes that operate under a tight regulation, ensuring homeostatic levels of this otherwise toxic molecule. Indeed, imbalances in H2S levels associated with dysfunctional H2S metabolism have been growingly correlated with various human pathologies, from cardiovascular and neurodegenerative diseases to cancer. Several cancer cell lines and specimens have been shown to naturally overexpress one or more of the H2S-synthesizing enzymes. The resulting increased H2S levels have been proposed to promote cancer development through the regulation of various cancer-related processes, which led to the interest in pharmacological targeting of H2S metabolism. Herein are summarized some of the key observations that place H2S metabolism and signaling pathways at the forefront of the cellular mechanisms that support the establishment and development of a tumor within its complex and challenging microenvironment. Special emphasis is given to the mechanisms whereby H2S helps shaping cancer cell bioenergetic metabolism and affords resistance and adaptive mechanisms to hypoxia.
Collapse
Affiliation(s)
| | - Catarina S Tomé
- Instituto de Tecnologia Química e Biológica António Xavier, NOVA University of Lisbon, Oeiras, Portugal
| | - Dalila G F Fernandes
- Instituto de Tecnologia Química e Biológica António Xavier, NOVA University of Lisbon, Oeiras, Portugal
| | - Karim Zuhra
- CNR Institute of Molecular Biology and Pathology, Rome, Italy
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - João B Vicente
- Instituto de Tecnologia Química e Biológica António Xavier, NOVA University of Lisbon, Oeiras, Portugal.
| |
Collapse
|
22
|
Haouzi P, Sonobe T, Judenherc-Haouzi A. Hydrogen sulfide intoxication induced brain injury and methylene blue. Neurobiol Dis 2019; 133:104474. [PMID: 31103557 DOI: 10.1016/j.nbd.2019.05.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/16/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
Hydrogen sulfide (H2S) remains a chemical hazard in the gas and farming industry. It is easy to manufacture from common chemicals and thus represents a potential threat for the civilian population. It is also employed as a method of suicide, for which incidence has recently increased in the US. H2S is a mitochondrial poison and exerts its toxicity through mechanisms that are thought to result from its high affinity to various metallo-proteins (such as - but not exclusively- the mitochondrial cytochrome c oxidase) and interactions with cysteine residues of proteins. Ion channels with critical implications for the cardiac and the brain functions appear to be affected very early during and following H2S exposure, an effect which is rapidly reversible during a light intoxication. However, during severe H2S intoxication, a coma, associated with a reduction in cardiac contractility, develops within minutes or even seconds leading to death by complete electro-mechanical dissociation of the heart. If the level of intoxication is milder, a rapid and spontaneous recovery of the coma occurs as soon as the exposure stops. The risk, although probably very small, of developing long-term debilitating motor or cognitive deficits is present. One of the major challenges impeding our effort to offer an effective treatment against H2S intoxication after exposure is that the pool of free/soluble H2S almost immediately disappears from the body preventing agents trapping free H2S (cobalt or ferric compounds) to play their protective role. This paper (1) presents and discusses the neurological symptoms and lesions observed in various animals models and in humans following an acute exposure to sub-lethal or lethal levels of H2S, (2) reviews the potential interest of methylene blue (MB), a potent cyclic redox dye - currently used for the treatment of methemoglobinemia - which has potential rescuing effects on the mitochondrial activity, as an antidote against sulfide intoxication.
Collapse
Affiliation(s)
- Philippe Haouzi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Takashi Sonobe
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Annick Judenherc-Haouzi
- Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
23
|
Abdulle AE, van Goor H, Mulder DJ. Hydrogen Sulfide: A Therapeutic Option in Systemic Sclerosis. Int J Mol Sci 2018; 19:E4121. [PMID: 30572591 PMCID: PMC6320961 DOI: 10.3390/ijms19124121] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/07/2018] [Accepted: 12/17/2018] [Indexed: 12/11/2022] Open
Abstract
Systemic sclerosis (SSc) is a lethal disease that is characterized by auto-immunity, vascular injury, and progressive fibrosis of multiple organ systems. Despite the fact that the exact etiology of SSc remains unknown, oxidative stress has been associated with a large range of SSc-related complications. In addition to the well-known detrimental properties of reactive oxygen species (ROS), gasotransmitters (e.g., nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H₂S)) are also thought to play an important role in SSc. Accordingly, the diverse physiologic actions of NO and CO and their role in SSc have been previously studied. Recently, multiple studies have also shown the importance of the third gasotransmitter H₂S in both vascular physiology and pathophysiology. Interestingly, homocysteine (which is converted into H₂S through the transsulfuration pathway) is often found to be elevated in SSc patients; suggesting defects in the transsulfuration pathway. Hydrogen sulfide, which is known to have several effects, including a strong antioxidant and vasodilator effect, could potentially play a prominent role in the initiation and progression of vasculopathy. A better understanding of the actions of gasotransmitters, like H₂S, in the development of SSc-related vasculopathy, could help to create early interventions to attenuate the disease course. This paper will review the role of H₂S in vascular (patho-)physiology and potential disturbances in SSc. Moreover, current data from experimental animal studies will be reviewed. Lastly, we will evaluate potential interventional strategies.
Collapse
Affiliation(s)
- Amaal Eman Abdulle
- Department of Internal Medicine, Division Vascular Medicine, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Harry van Goor
- Department of Pathology and Medical Biology, Section Pathology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Douwe J Mulder
- Department of Internal Medicine, Division Vascular Medicine, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
24
|
Pozsgai G, Bátai IZ, Pintér E. Effects of sulfide and polysulfides transmitted by direct or signal transduction-mediated activation of TRPA1 channels. Br J Pharmacol 2018; 176:628-645. [PMID: 30292176 PMCID: PMC6346070 DOI: 10.1111/bph.14514] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/22/2018] [Accepted: 09/24/2018] [Indexed: 12/21/2022] Open
Abstract
Hydrogen sulfide (H2S) is a gaseous mediator in various physiological and pathological processes, including neuroimmune modulation, metabolic pathways, cardiovascular system, tumour growth, inflammation and pain. Now the hydrogen polysulfides (H2Sn) have been recognised as signalling molecules modulating ion channels, transcription factors and protein kinases. Transient receptor potential (TRP) cation channels can be activated by mechanical, thermal or chemical triggers. Here, we review the current literature regarding the biological actions of sulfide and polysulfide compounds mediated by TRP channels with special emphasis on the role of TRPA1, best known as ion channels in nociceptors. However, the non‐neuronal TRPA1 channels should also be considered to play regulatory roles. Although sulfide and polysulfide effects in different pathological circumstances and TRPA1‐mediated processes have been investigated intensively, our review attempts to present the first comprehensive overview of the potential crosstalk between TRPA1 channels and sulfide‐activated signalling pathways. Linked Articles This article is part of a themed section on Chemical Biology of Reactive Sulfur Species. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.4/issuetoc
Collapse
Affiliation(s)
- Gábor Pozsgai
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - István Zoárd Bátai
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
25
|
Role of Nitric Oxide and Hydrogen Sulfide in Ischemic Stroke and the Emergent Epigenetic Underpinnings. Mol Neurobiol 2018; 56:1749-1769. [PMID: 29926377 DOI: 10.1007/s12035-018-1141-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 05/22/2018] [Indexed: 02/06/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are the key gasotransmitters with an imperious role in the maintenance of cerebrovascular homeostasis. A decline in their levels contributes to endothelial dysfunction that portends ischemic stroke (IS) or cerebral ischemia/reperfusion (CI/R). Nevertheless, their exorbitant production during CI/R is associated with exacerbation of cerebrovascular injury in the post-stroke epoch. NO-producing nitric oxide synthases are implicated in IS pathology and their activity is regulated, inter alia, by various post-translational modifications and chromatin-based mechanisms. These account for heterogeneous alterations in NO production in a disease setting like IS. Interestingly, NO per se has been posited as an endogenous epigenetic modulator. Further, there is compelling evidence for an ingenious crosstalk between NO and H2S in effecting the canonical (direct) and non-canonical (off-target collateral) functions. In this regard, NO-mediated S-nitrosylation and H2S-mediated S-sulfhydration of specific reactive thiols in an expanding array of target proteins are the principal modalities mediating the all-pervasive influence of NO and H2S on cell fate in an ischemic brain. An integrated stress response subsuming unfolded protein response and autophagy to cellular stressors like endoplasmic reticulum stress, in part, is entrenched in such signaling modalities that substantiate the role of NO and H2S in priming the cells for stress response. The precis presented here provides a comprehension on the multifarious actions of NO and H2S and their epigenetic underpinnings, their crosstalk in maintenance of cerebrovascular homeostasis, and their "Janus bifrons" effect in IS milieu together with plausible therapeutic implications.
Collapse
|
26
|
Taylor CT, Colgan SP. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat Rev Immunol 2017; 17:774-785. [PMID: 28972206 PMCID: PMC5799081 DOI: 10.1038/nri.2017.103] [Citation(s) in RCA: 461] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunological niches are focal sites of immune activity that can have varying microenvironmental features. Hypoxia is a feature of physiological and pathological immunological niches. The impact of hypoxia on immunity and inflammation can vary depending on the microenvironment and immune processes occurring in a given niche. In physiological immunological niches, such as the bone marrow, lymphoid tissue, placenta and intestinal mucosa, physiological hypoxia controls innate and adaptive immunity by modulating immune cell proliferation, development and effector function, largely via transcriptional changes driven by hypoxia-inducible factor (HIF). By contrast, in pathological immunological niches, such as tumours and chronically inflamed, infected or ischaemic tissues, pathological hypoxia can drive tissue dysfunction and disease development through immune cell dysregulation. Here, we differentiate between the effects of physiological and pathological hypoxia on immune cells and the consequences for immunity and inflammation in different immunological niches. Furthermore, we discuss the possibility of targeting hypoxia-sensitive pathways in immune cells for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, 80045 Colorado, USA
| |
Collapse
|
27
|
Dugbartey GJ. The smell of renal protection against chronic kidney disease: Hydrogen sulfide offers a potential stinky remedy. Pharmacol Rep 2017; 70:196-205. [PMID: 29471067 DOI: 10.1016/j.pharep.2017.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/28/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
Chronic kidney disease (CKD) is a common global health challenge characterized by irreversible pathological processes that reduce kidney function and culminates in development of end-stage renal disease. It is associated with increased morbidity and mortality in addition to increased caregiver burden and higher financial cost. A central player in CKD pathogenesis and progression is renal hypoxia. Renal hypoxia stimulates induction of oxidative and endoplasmic reticulum stress, inflammation and tubulointerstitial fibrosis, which in turn, promote cellular susceptibility and further aggravate hypoxia, thus forming a pathological vicious cycle in CKD progression. Although the importance of CKD is widely appreciated, including improvements in the quality of existing therapies such as dialysis and transplantation, new therapeutic options are limited, as there is still increased morbidity, mortality and poor quality of life among CKD patients. Growing evidence indicates that hydrogen sulfide (H2S), a small gaseous signaling molecule with an obnoxious smell, accumulates in the renal medulla under hypoxic conditions, and functions as an oxygen sensor that restores oxygen balance and increases medullary flow. Moreover, plasma H2S level has been recently reported to be markedly reduced in CKD patients and animal models. Also, H2S has been established to possess potent antioxidant, anti-inflammatory, and anti-fibrotic properties in several experimental models of kidney diseases, suggesting that its supplementation could protect against CKD and retard its progression. The purpose of this review is to discuss current clinical and experimental developments regarding CKD, its pathophysiology, and potential cellular and molecular mechanisms of protection by H2S in experimental models of CKD.
Collapse
Affiliation(s)
- George J Dugbartey
- Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
28
|
Yuan S, Shen X, Kevil CG. Beyond a Gasotransmitter: Hydrogen Sulfide and Polysulfide in Cardiovascular Health and Immune Response. Antioxid Redox Signal 2017; 27:634-653. [PMID: 28398086 PMCID: PMC5576200 DOI: 10.1089/ars.2017.7096] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Hydrogen sulfide (H2S) metabolism leads to the formation of oxidized sulfide species, including polysulfide, persulfide, and others. Evidence is emerging that many biological effects of H2S may indeed be due to polysulfide and persulfide activation of signaling pathways and reactivity with discrete small molecules. Recent Advances: Exogenous oxidized sulfide species, including polysulfides, are more reactive than H2S with a wide range of molecules. Importantly, endogenous polysulfide and persulfide formation has been reported to occur via transsulfuration enzymes, cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS). CRITICAL ISSUES In light of the recent understanding of oxidized sulfide metabolite formation and reactivity, comparatively few studies have been reported comparing cellular biological and in vivo effects of H2S donors versus polysulfide and persulfide donors. Likewise, it is equally unclear when, how, and to what extent persulfide and polysulfide formation occurs in vivo under pathophysiological conditions. FUTURE DIRECTIONS Additional studies regarding persulfide and polysulfide formation and molecular reactions are needed in nearly all aspects of biology to better understand how sulfide metabolites contribute to key chemical biology reactions involved in cardiovascular health and immune responses. Antioxid. Redox Signal. 27, 634-653.
Collapse
Affiliation(s)
- Shuai Yuan
- 1 Department of Cell Biology and Anatomy, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| | - Xinggui Shen
- 2 Department of Pathology and Translational Pathobiology, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| | - Christopher G Kevil
- 2 Department of Pathology and Translational Pathobiology, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| |
Collapse
|
29
|
Fagundes RR, Taylor CT. Determinants of hypoxia-inducible factor activity in the intestinal mucosa. J Appl Physiol (1985) 2017; 123:1328-1334. [PMID: 28408694 DOI: 10.1152/japplphysiol.00203.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/28/2017] [Accepted: 04/07/2017] [Indexed: 12/16/2022] Open
Abstract
The intestinal mucosa is exposed to fluctuations in oxygen levels due to constantly changing rates of oxygen demand and supply and its juxtaposition with the anoxic environment of the intestinal lumen. This frequently results in a state of hypoxia in the healthy mucosa even in the physiologic state. Furthermore, pathophysiologic hypoxia (which is more severe and extensive) is associated with chronic inflammatory diseases including inflammatory bowel disease (IBD). The hypoxia-inducible factor (HIF), a ubiquitously expressed regulator of cellular adaptation to hypoxia, is central to both the adaptive and the inflammatory responses of cells of the intestinal mucosa in IBD patients. In this review, we discuss the microenvironmental factors which influence the level of HIF activity in healthy and inflamed intestinal mucosae and the consequences that increased HIF activity has for tissue function and disease progression.
Collapse
Affiliation(s)
- Raphael R Fagundes
- Graduate School of Medical Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and.,UCD Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
30
|
|
31
|
Zheng Y, Yu B, De La Cruz LK, Roy Choudhury M, Anifowose A, Wang B. Toward Hydrogen Sulfide Based Therapeutics: Critical Drug Delivery and Developability Issues. Med Res Rev 2017; 38:57-100. [PMID: 28240384 DOI: 10.1002/med.21433] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/10/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022]
Abstract
Hydrogen sulfide (H2 S), together with nitric oxide (NO) and carbon monoxide (CO), belongs to the gasotransmitter family and plays important roles in mammals as a signaling molecule. Many studies have also shown the various therapeutic effects of H2 S, which include protection against myocardial ischemia injury, cytoprotection against oxidative stress, mediation of neurotransmission, inhibition of insulin signaling, regulation of inflammation, inhibition of the hypoxia-inducible pathway, and dilation of blood vessels. One major challenge in the development of H2 S-based therapeutics is its delivery. In this manuscript, we assess the various drug delivery strategies in the context of being used research tools and eventual developability as therapeutic agents.
Collapse
Affiliation(s)
- Yueqin Zheng
- Department of Chemistry, Georgia State University, Atlanta, Georgia
| | - Bingchen Yu
- Department of Chemistry, Georgia State University, Atlanta, Georgia
| | | | | | | | - Binghe Wang
- Department of Chemistry, Georgia State University, Atlanta, Georgia
| |
Collapse
|
32
|
Gorr TA. Hypometabolism as the ultimate defence in stress response: how the comparative approach helps understanding of medically relevant questions. Acta Physiol (Oxf) 2017; 219:409-440. [PMID: 27364602 DOI: 10.1111/apha.12747] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/28/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022]
Abstract
First conceptualized from breath-hold diving mammals, later recognized as the ultimate cell autonomous survival strategy in anoxia-tolerant vertebrates and burrowing or hibernating rodents, hypometabolism is typically recruited by resilient organisms to withstand and recover from otherwise life-threatening hazards. Through the coordinated down-regulation of biosynthetic, proliferative and electrogenic expenditures at times when little ATP can be generated, a metabolism turned 'down to the pilot light' allows the re-balancing of energy demand with supply at a greatly suppressed level in response to noxious exogenous stimuli or seasonal endogenous cues. A unifying hallmark of stress-tolerant organisms, the adaptation effectively prevents lethal depletion of ATP, thus delineating a marked contrast with susceptible species. Along with disengaged macromolecular syntheses, attenuated transmembrane ion shuttling and PO2 -conforming respiration rates, the metabolic slowdown in tolerant species usually culminates in a non-cycling, quiescent phenotype. However, such a reprogramming also occurs in leading human pathophysiologies. Ranging from microbial infections through ischaemia-driven infarcts to solid malignancies, cells involved in these disorders may again invoke hypometabolism to endure conditions non-permissive for growth. At the same time, their reduced activities underlie the frequent development of a general resistance to therapeutic interventions. On the other hand, a controlled induction of hypometabolic and/or hypothermic states by pharmacological means has recently stimulated intense research aimed at improved organ preservation and patient survival in situations requiring acutely administered critical care. The current review article therefore presents an up-to-date survey of concepts and applications of a coordinated and reversibly down-regulated metabolic rate as the ultimate defence in stress responses.
Collapse
Affiliation(s)
- T. A. Gorr
- Institute of Veterinary Physiology; Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| |
Collapse
|
33
|
Yuan S, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Remodeling. Microcirculation 2016; 23:134-45. [PMID: 26381654 DOI: 10.1111/micc.12248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 09/13/2015] [Indexed: 12/22/2022]
Abstract
Blockage or restriction of blood flow through conduit arteries results in tissue ischemia downstream of the disturbed area. Local tissues can adapt to this challenge by stimulating vascular remodeling through angiogenesis and arteriogenesis thereby restoring blood perfusion and removal of wastes. Multiple molecular mechanisms of vascular remodeling during ischemia have been identified and extensively studied. However, therapeutic benefits from these findings and insights are limited due to the complexity of various signaling networks and a lack of understanding central metabolic regulators governing these responses. The gasotransmitters NO and H2 S have emerged as master regulators that influence multiple molecular targets necessary for ischemic vascular remodeling. In this review, we discuss how NO and H2 S are individually regulated under ischemia, what their roles are in angiogenesis and arteriogenesis, and how their interaction controls ischemic vascular remodeling.
Collapse
Affiliation(s)
- Shuai Yuan
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| | - Christopher G Kevil
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
34
|
Jang Y, Han J, Kim SJ, Kim J, Lee MJ, Jeong S, Ryu MJ, Seo KS, Choi SY, Shong M, Lim K, Heo JY, Kweon GR. Suppression of mitochondrial respiration with auraptene inhibits the progression of renal cell carcinoma: involvement of HIF-1α degradation. Oncotarget 2016; 6:38127-38. [PMID: 26474388 PMCID: PMC4741988 DOI: 10.18632/oncotarget.5511] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/29/2015] [Indexed: 12/14/2022] Open
Abstract
Renal cell carcinoma (RCC) progression resulting from the uncontrolled migration and enhanced angiogenesis is an obstacle to effective therapeutic intervention. Tumor metabolism has distinctive feature called Warburg effect, which enhances the aerobic glycolysis rapidly supplying the energy for migration of tumor. To manipulate this metabolic change characteristic of aggressive tumors, we utilized the citrus extract, auraptene, known as a mitochondrial inhibitor, testing its anticancer effects against the RCC4 cell line. We found that auraptene impaired RCC4 cell motility through reduction of mitochondrial respiration and glycolytic pathway-related genes. It also strongly disrupted VEGF-induced angiogenesis in vitro and in vivo. Hypoxia-inducible factor 1a (HIF-1a), a key regulator of cancer metabolism, migration and angiogenesis that is stably expressed in RCCs by virtue of a genetic mutation in the von Hippel–Lindau (VHL) tumor-suppressor protein, was impeded by auraptene, which blocked HIF-1a translation initiation without causing cytotoxicity. We suggest that blockade HIF-1a and reforming energy metabolism with auraptene is an effective approach for suspension RCC progression.
Collapse
Affiliation(s)
- Yunseon Jang
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Jeongsu Han
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Soo Jeong Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Jungim Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Soyeon Jeong
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Kang-Sik Seo
- R&D Center, KT&G Life Sciences, Suwon, Republic of Korea, 443-702
| | - Song-Yi Choi
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Minho Shong
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Kyu Lim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Brain research institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| |
Collapse
|
35
|
Leigh J, Saha MN, Mok A, Champsi O, Wang R, Lobb I, Sener A. Hydrogen Sulfide Induced Erythropoietin Synthesis is Regulated by HIF Proteins. J Urol 2016; 196:251-60. [PMID: 26880412 DOI: 10.1016/j.juro.2016.01.113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2016] [Indexed: 12/27/2022]
Abstract
PURPOSE Anemia of end stage renal disease affects 90% of patients on hemodialysis and it is a tremendous concern of patients and health care providers. Renal disease creates a state of renal hypoxia, which may contribute to a lack of erythropoietin production from the kidney when low oxygen levels are sensed. This necessitates the use of exogenous erythropoietin preparations. MATERIALS AND METHODS Recent evidence suggests that endogenously derived hydrogen sulfide may mediate oxygen sensing in tissues. Given the known involvement of other small molecules such as nitric oxide in erythropoietin production and the observation of decreased urinary H2S levels in patients with renal failure, we postulated that H2S may be the primary mediator of erythropoietin production during hypoxia. PK1, 786-O and Hep3B cells were incubated in hypoxia (1% O2) for 24 hours. Hypoxic cells were treated with the H2S donor GYY 4137 and the H2S inhibitor hydroxylamine. Following hypoxia erythropoietin, HIF-1α, HIF-2α and CBS expression was measured by quantitative real-time polymerase chain reaction and Western blot. RESULTS Hydroxylamine administration led to a significant decrease in erythropoietin, HIF-1α, HIF-2α and CBS protein levels during hypoxia. This was rescued by administration of GYY 4137 for erythropoietin, CBS and HIF-2α. Additionally, CSE -/- mice placed in hypoxia for 72 hours showed decreased renal erythropoietin production compared to wild-type mice. CONCLUSIONS These data suggest previously undocumented interplay of the production and action of H2S during hypoxia with subsequent erythropoietin production. The use of novel hydrogen sulfide donors could represent an alternative to standard therapies of anemia of renal failure.
Collapse
Affiliation(s)
- Jennifer Leigh
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Manujendra N Saha
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Amy Mok
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Omar Champsi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Ontario, Canada
| | - Ian Lobb
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Alp Sener
- Department of Surgery, Western University, London, Ontario, Canada; Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada.
| |
Collapse
|
36
|
Lencesova L, Vlcek M, Krizanova O, Hudecova S. Hypoxic conditions increases H₂S-induced ER stress in A2870 cells. Mol Cell Biochem 2016; 414:67-76. [PMID: 26868821 DOI: 10.1007/s11010-016-2659-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/03/2016] [Indexed: 12/13/2022]
Abstract
Hypoxia - a state of lower oxygen demand-is responsible for a higher aggressiveness of tumors and therefore a worse prognosis. During hypoxia, several metabolic pathways are re-organized, e.g., energetic metabolism, modulation of pH, and calcium transport. Calcium is an important second messenger that regulates variety of processes in the cell. Thus, aim of this work was to compare H2S modulation of the intracellular calcium transport systems in hypoxia and in cells grown in standard culture conditions. For all experiments, we used ovarian cancer cell line (A2780). H2S is a novel gasotransmitter, known to be involved in a modulation of several calcium transport systems, thus resulting in altered calcium signaling. Two models of hypoxia were used in our study-chemical (induced by dimethyloxallyl glycine) and 2 % O2 hypoxia, both combined with a treatment using a slow H2S donor GYY4137. In hypoxia, we observed rapid changes in cytosolic and reticular calcium levels compared to cells grown in standard culture conditions, and these changes were even more exagerrated when combined with the GYY4137. Changes in a calcium homeostasis result from IP3 receptor´s up-regulation and down-regulation of the SERCA 2, which leads to a development of the endoplasmic reticulum stress. Based on our results, we propose a higher vulnerability of calcium transport systems to H2S regulation under hypoxia.
Collapse
Affiliation(s)
- Lubomira Lencesova
- Biomedical Research Center, Institute for Clinical and Translational Research, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, Slovak Republic
| | - Miroslav Vlcek
- Biomedical Research Center, Institute for Clinical and Translational Research, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, Slovak Republic
| | - Olga Krizanova
- Biomedical Research Center, Institute for Clinical and Translational Research, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, Slovak Republic
| | - Sona Hudecova
- Biomedical Research Center, Institute for Clinical and Translational Research, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, Slovak Republic.
| |
Collapse
|
37
|
Sonobe T, Chenuel B, Cooper TK, Haouzi P. Immediate and Long-Term Outcome of Acute H2S Intoxication Induced Coma in Unanesthetized Rats: Effects of Methylene Blue. PLoS One 2015; 10:e0131340. [PMID: 26115032 PMCID: PMC4482667 DOI: 10.1371/journal.pone.0131340] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 06/01/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acute hydrogen sulfide (H2S) poisoning produces a coma, the outcome of which ranges from full recovery to severe neurological deficits. The aim of our study was to 1--describe the immediate and long-term neurological effects following H2S-induced coma in un-anesthetized rats, and 2--determine the potential benefit of methylene blue (MB), a compound we previously found to counteract acute sulfide cardiac toxicity. METHODS NaHS was administered IP in un-sedated rats to produce a coma (n = 34). One minute into coma, the rats received MB (4 mg/kg i.v.) or saline. The surviving rats were followed clinically and assigned to Morris water maze (MWM) and open field testing then sacrificed at day 7. RESULTS Sixty percent of the non-treated comatose rats died by pulseless electrical activity. Nine percent recovered with neurological deficits requiring euthanasia, their brain examination revealed major neuronal necrosis of the superficial and middle layers of the cerebral cortex and the posterior thalamus, with variable necrosis of the caudate putamen, but no lesions of the hippocampus or the cerebellum, in contrast to the typical distribution of post-ischemic lesions. The remaining animals displayed, on average, a significantly less effective search strategy than the control rats (n = 21) during MWM testing. Meanwhile, 75% of rats that received MB survived and could perform the MWM test (P<0.05 vs non-treated animals). The treated animals displayed a significantly higher occurrence of spatial search than the non-treated animals. However, a similar proportion of cortical necrosis was observed in both groups, with a milder clinical presentation following MB. CONCLUSION In conclusion, in rats surviving H2S induced coma, spatial search patterns were used less frequently than in control animals. A small percentage of rats presented necrotic neuronal lesions, which distribution differed from post-ischemic lesions. MB dramatically improved the immediate survival and spatial search strategy in the surviving rats.
Collapse
Affiliation(s)
- Takashi Sonobe
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Pennsylvania State University, College of Medicine, Hershey, PA, United States of America
| | - Bruno Chenuel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Pennsylvania State University, College of Medicine, Hershey, PA, United States of America
| | - Timothy K. Cooper
- Department of Comparative Medicine, Pennsylvania State University, College of Medicine, Hershey, PA, United States of America
- Department of Pathology, Pennsylvania State University, College of Medicine, Hershey, PA, United States of America
| | - Philippe Haouzi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Pennsylvania State University, College of Medicine, Hershey, PA, United States of America
- * E-mail:
| |
Collapse
|
38
|
Inhibition of endogenous hydrogen sulfide production in clear-cell renal cell carcinoma cell lines and xenografts restricts their growth, survival and angiogenic potential. Nitric Oxide 2015; 49:26-39. [PMID: 26068241 DOI: 10.1016/j.niox.2015.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 05/20/2015] [Accepted: 06/01/2015] [Indexed: 11/21/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by Von Hippel-Lindau (VHL)-deficiency, resulting in pseudohypoxic, angiogenic and glycolytic tumours. Hydrogen sulfide (H2S) is an endogenously-produced gasotransmitter that accumulates under hypoxia and has been shown to be pro-angiogenic and cytoprotective in cancer. It was hypothesized that H2S levels are elevated in VHL-deficient ccRCC, contributing to survival, metabolism and angiogenesis. Using the H2S-specific probe MeRhoAz, it was found that H2S levels were higher in VHL-deficient ccRCC cell lines compared to cells with wild-type VHL. Inhibition of H2S-producing enzymes could reduce the proliferation, metabolism and survival of ccRCC cell lines, as determined by live-cell imaging, XTT/ATP assay, and flow cytometry respectively. Using the chorioallantoic membrane angiogenesis model, it was found that systemic inhibition of endogenous H2S production was able to decrease vascularization of VHL-deficient ccRCC xenografts. Endogenous H2S production is an attractive new target in ccRCC due to its involvement in multiple aspects of disease.
Collapse
|
39
|
Interaction of Hydrogen Sulfide with Oxygen Sensing under Hypoxia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:758678. [PMID: 26078818 PMCID: PMC4442289 DOI: 10.1155/2015/758678] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/22/2015] [Indexed: 01/18/2023]
Abstract
Based on the discovery of endogenous H2S production, many in depth studies show this gasotransmitter with a variety of physiological and pathological functions. Three enzymes, cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (MST), are involved in enzymatic production of H2S. Emerging evidence has elucidated an important protective role of H2S in hypoxic conditions in many mammalian systems. However, the mechanisms by which H2S senses and responses to hypoxia are largely elusive. Hypoxia-inducible factors (HIFs) function as key regulators of oxygen sensing, activating target genes expression under hypoxia. Recent studies have shown that exogenous H2S regulates HIF action in different patterns. The activation of carotid bodies is a sensitive and prompt response to hypoxia, rapidly enhancing general O2 supply. H2S has been identified as an excitatory mediator of hypoxic sensing in the carotid bodies. This paper presents a brief review of the roles of these two pathways which contribute to hypoxic sensing of H2S.
Collapse
|
40
|
Lohninger L, Tomasova L, Praschberger M, Hintersteininger M, Erker T, Gmeiner BMK, Laggner H. Hydrogen sulphide induces HIF-1α and Nrf2 in THP-1 macrophages. Biochimie 2015; 112:187-95. [PMID: 25795259 DOI: 10.1016/j.biochi.2015.03.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 03/09/2015] [Indexed: 12/30/2022]
Abstract
The transcription factor HIF-1α regulates the adaptive response of cells to hypoxia and oxidative stress. In addition, an important regulatory role for HIF-1α in immune reactions and inflammation is suggested. The present study attempts to investigate the effect of the gaseous signalling molecule hydrogen sulphide (H2S) on HIF-1α in THP-1 macrophages using the slow H2S releasing donor GYY4137. We found that H2S induced HIF-1α protein accumulation in THP-1 macrophages in a concentration-dependent manner. Western blot analysis of cell fractions showed that HIF-1α protein translocates into the nucleus and leads to an increase of its target protein glucose transporter-1 (GLUT-1). Activation of nuclear factor-κB (NF-κB), as well as secretion of the pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), were reduced in the presence of H2S. These findings indicate that HIF-1α accumulation due to H2S was not triggered by the NF-κB pathway. The antioxidant pathway Nrf2/HO-1 (nuclear factor erythroid 2-related factor 2/heme oxygenase-1) was activated by H2S. Inhibition of the p38 mitogen-activated protein kinase (MAPK) reversed H2S mediated effects, suggesting that the p38 MAPK pathway may be involved in H2S induced HIF-1α/Nrf2 signalling pathways.
Collapse
Affiliation(s)
- Lilian Lohninger
- Center of Pathobiochemistry and Genetics, Department of Medical Chemistry and Pathobiochemistry, Medical University of Vienna, Vienna, Austria
| | - Lenka Tomasova
- Center of Pathobiochemistry and Genetics, Department of Medical Chemistry and Pathobiochemistry, Medical University of Vienna, Vienna, Austria; Faculty of Pharmacy, Comenius University, Bratislava, Slovak Republic; Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Monika Praschberger
- Center of Pathobiochemistry and Genetics, Department of Medical Chemistry and Pathobiochemistry, Medical University of Vienna, Vienna, Austria
| | - Michael Hintersteininger
- Division of Drug Design and Medicinal Chemistry, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Thomas Erker
- Division of Drug Design and Medicinal Chemistry, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Bernhard M K Gmeiner
- Center of Pathobiochemistry and Genetics, Department of Medical Chemistry and Pathobiochemistry, Medical University of Vienna, Vienna, Austria
| | - Hilde Laggner
- Center of Pathobiochemistry and Genetics, Department of Medical Chemistry and Pathobiochemistry, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
41
|
Cao J, Lopez R, Thacker JM, Moon JY, Jiang C, Morris SNS, Bauer JH, Tao P, Mason RP, Lippert AR. Chemiluminescent Probes for Imaging H 2S in Living Animals. Chem Sci 2015; 6:1979-1985. [PMID: 25709805 PMCID: PMC4335805 DOI: 10.1039/c4sc03516j] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/31/2014] [Indexed: 12/18/2022] Open
Abstract
Hydrogen sulphide (H2S) is an endogenous mediator of human health and disease, but precise measurement in living cells and animals remains a considerable challenge. We report the total chemical synthesis and characterization of three 1,2-dioxetane chemiluminescent reaction-based H2S probes, CHS-1, CHS-2, and CHS-3. Upon treatment with H2S at physiological pH, these probes display instantaneous light emission that is sustained for over an hour with high selectivity against other reactive sulphur, oxygen, and nitrogen species. Analysis of the phenol/phenolate equilibrium and atomic charges has provided a generally applicable predictive model to design improved chemiluminescent probes. The utility of these chemiluminescent reagents was demonstrated by applying CHS-3 to detect cellularly generated H2S using a multi-well plate reader and to image H2S in living mice using CCD camera technology.
Collapse
Affiliation(s)
- J. Cao
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA .
- Center for Drug Discovery , Design, and Delivery (CD4) , Southern Methodist University , Dallas , TX 75275-0314 , USA
| | - R. Lopez
- Laboratory of Prognostic Radiology , Pre-clinical Imaging Section , Department of Radiology , UT Southwestern Medical Center , Dallas , TX 75390-9058 , USA
| | - J. M. Thacker
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA .
| | - J. Y. Moon
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA .
| | - C. Jiang
- Hockaday School , Dallas , TX 75229 , USA
| | - S. N. S. Morris
- Department of Biological Sciences , Southern Methodist University , Dallas , TX 75275-0314 , USA
| | - J. H. Bauer
- Center for Drug Discovery , Design, and Delivery (CD4) , Southern Methodist University , Dallas , TX 75275-0314 , USA
- Department of Biological Sciences , Southern Methodist University , Dallas , TX 75275-0314 , USA
| | - P. Tao
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA .
- Center for Drug Discovery , Design, and Delivery (CD4) , Southern Methodist University , Dallas , TX 75275-0314 , USA
| | - R. P. Mason
- Laboratory of Prognostic Radiology , Pre-clinical Imaging Section , Department of Radiology , UT Southwestern Medical Center , Dallas , TX 75390-9058 , USA
| | - A. R. Lippert
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA .
- Center for Drug Discovery , Design, and Delivery (CD4) , Southern Methodist University , Dallas , TX 75275-0314 , USA
| |
Collapse
|
42
|
Abstract
SIGNIFICANCE Although oxygen (O2)-sensing cells and tissues have been known for decades, the identity of the O2-sensing mechanism has remained elusive. Evidence is accumulating that O2-dependent metabolism of hydrogen sulfide (H2S) is this enigmatic O2 sensor. RECENT ADVANCES The elucidation of biochemical pathways involved in H2S synthesis and metabolism have shown that reciprocal H2S/O2 interactions have been inexorably linked throughout eukaryotic evolution; there are multiple foci by which O2 controls H2S inactivation, and the effects of H2S on downstream signaling events are consistent with those activated by hypoxia. H2S-mediated O2 sensing has been demonstrated in a variety of O2-sensing tissues in vertebrate cardiovascular and respiratory systems, including smooth muscle in systemic and respiratory blood vessels and airways, carotid body, adrenal medulla, and other peripheral as well as central chemoreceptors. CRITICAL ISSUES Information is now needed on the intracellular location and stoichometry of these signaling processes and how and which downstream effectors are activated by H2S and its metabolites. FUTURE DIRECTIONS Development of specific inhibitors of H2S metabolism and effector activation as well as cellular organelle-targeted compounds that release H2S in a time- or environmentally controlled way will not only enhance our understanding of this signaling process but also provide direction for future therapeutic applications.
Collapse
Affiliation(s)
- Kenneth R Olson
- Department of Physiology, Indiana University School of Medicine-South Bend , South Bend, India na
| |
Collapse
|
43
|
Abstract
The physiological and biomedical importance of hydrogen sulfide (H2S) has been fully recognized in the cardiovascular system as well as in the rest of the body. In blood vessels, cystathionine γ-lyase (CSE) is a major H2S-producing enzyme expressed in both smooth muscle and endothelium as well as periadventitial adipose tissues. Regulation of H2S production from CSE is controlled by a complex integration of transcriptional, posttranscriptional, and posttranslational mechanisms in blood vessels. In smooth muscle cells, H2S regulates cell apoptosis, phenotypic switch, relaxation and contraction, and calcification. In endothelial cells, H2S controls cell proliferation, cellular senescence, oxidative stress, inflammation, etc. H2S interacts with nitric oxide and acts as an endothelium-derived relaxing factor and an endothelium-derived hyperpolarizing factor. H2S generated from periadventitial adipose tissues acts as an adipocyte-derived relaxing factor and modulates the vascular tone. Extensive evidence has demonstrated the beneficial roles of the CSE/H2S system in various blood vessel diseases, such as hypertension, atherosclerosis, and aortic aneurysm. The important roles signaling in the cardiovascular system merit further intensive and extensive investigation. H2S-releasing agents and CSE activators will find their great applications in the prevention and treatment of blood vessel-related disorders.
Collapse
Affiliation(s)
- Guangdong Yang
- Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada
| | | |
Collapse
|
44
|
Abstract
Hydrogen sulfide (H2S) has emerged as a key regulator of cardiovascular function. This gasotransmitter is produced in the vasculature and is involved in numerous processes that promote vascular homeostasis, including vasodilation and endothelial cell proliferation. Although H2S plays a role under physiological conditions, it has become clear in recent years that hypoxia modulates the production and action of H2S. Furthermore, there is growing evidence that H2S is cytoprotective in the face of hypoxic insults. This review focuses on the synthesis and signaling of H2S in hypoxic conditions in the vasculature, and highlights recent studies providing evidence that H2S is a potential therapy for preventing tissue damage in hypoxic conditions.
Collapse
Affiliation(s)
- Jessica M Osmond
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Nancy L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
45
|
Potenza DM, Guerra G, Avanzato D, Poletto V, Pareek S, Guido D, Gallanti A, Rosti V, Munaron L, Tanzi F, Moccia F. Hydrogen sulphide triggers VEGF-induced intracellular Ca²⁺ signals in human endothelial cells but not in their immature progenitors. Cell Calcium 2014; 56:225-34. [PMID: 25113159 DOI: 10.1016/j.ceca.2014.07.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/09/2014] [Accepted: 07/18/2014] [Indexed: 10/25/2022]
Abstract
Hydrogen sulphide (H2S) is a newly discovered gasotransmitter that regulates multiple steps in VEGF-induced angiogenesis. An increase in intracellular Ca(2+) concentration ([Ca(2+)]i) is central to endothelial proliferation and may be triggered by both VEGF and H2S. Albeit VEGFR-2 might serve as H2S receptor, the mechanistic relationship between VEGF- and H2S-induced Ca(2+) signals in endothelial cells is unclear. The present study aimed at assessing whether and how NaHS, a widely employed H2S donor, stimulates pro-angiogenic Ca(2+) signals in Ea.hy926 cells, a suitable surrogate for mature endothelial cells, and human endothelial progenitor cells (EPCs). We found that NaHS induced a dose-dependent increase in [Ca(2+)]i in Ea.hy926 cells. NaHS-induced Ca(2+) signals in Ea.hy926 cells did not require extracellular Ca(2+) entry, while they were inhibited upon pharmacological blockade of the phospholipase C/inositol-1,4,5-trisphosphate (InsP3) signalling pathway. Moreover, the Ca(2+) response to NaHS was prevented by genistein, but not by SU5416, which selectively inhibits VEGFR-2. However, VEGF-induced Ca(2+) signals were suppressed by dl-propargylglycine (PAG), which blocks the H2S-producing enzyme, cystathionine γ-lyase. Consistent with these data, VEGF-induced proliferation and migration were inhibited by PAG in Ea.hy926 cells, albeit NaHS alone did not influence these processes. Conversely, NaHS elevated [Ca(2+)]i only in a modest fraction of circulating EPCs, whereas neither VEGF-induced Ca(2+) oscillations nor VEGF-dependent proliferation were affected by PAG. Therefore, H2S-evoked elevation in [Ca(2+)]i is essential to trigger the pro-angiogenic Ca(2+) response to VEGF in mature endothelial cells, but not in their immature progenitors.
Collapse
Affiliation(s)
- Duilio Michele Potenza
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | - Germano Guerra
- Department of Health Sciences, University of Molise, Via F. De Santis, 86100 Campobasso, Italy
| | - Daniele Avanzato
- Department of Life Sciences and Systems Biology, Centre for Nanostructured Interfaces and Surfaces, Centre for Complex Systems in Molecular Biology and Medicine, University of Torino, 10123 Torino, Italy
| | - Valentina Poletto
- Center for the Study of Myelofibrosis, Research Laboratory of Biotechnology, IRCCS Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100 Pavia, Italy
| | - Sumedha Pareek
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | - Daniele Guido
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | - Angelo Gallanti
- Department of Molecular Medicine, University of Pavia, Via Taramelli 10, 27100 Pavia, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Research Laboratory of Biotechnology, IRCCS Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100 Pavia, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, Centre for Nanostructured Interfaces and Surfaces, Centre for Complex Systems in Molecular Biology and Medicine, University of Torino, 10123 Torino, Italy
| | - Franco Tanzi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Via Forlanini 6, 27100 Pavia, Italy.
| |
Collapse
|
46
|
Takano N, Sarfraz Y, Gilkes DM, Chaturvedi P, Xiang L, Suematsu M, Zagzag D, Semenza GL. Decreased expression of cystathionine β-synthase promotes glioma tumorigenesis. Mol Cancer Res 2014; 12:1398-406. [PMID: 24994751 DOI: 10.1158/1541-7786.mcr-14-0184] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
UNLABELLED Cystathionine β-synthase (CBS) catalyzes metabolic reactions that convert homocysteine to cystathionine. To assess the role of CBS in human glioma, cells were stably transfected with lentiviral vectors encoding shRNA targeting CBS or a nontargeting control shRNA, and subclones were injected into immunodeficient mice. Interestingly, decreased CBS expression did not affect proliferation in vitro but decreased the latency period before rapid tumor xenograft growth after subcutaneous injection and increased tumor incidence and volume following orthotopic implantation into the caudate-putamen. In soft-agar colony formation assays, CBS knockdown subclones displayed increased anchorage-independent growth. Molecular analysis revealed that CBS knockdown subclones expressed higher basal levels of the transcriptional activator hypoxia-inducible factor 2α (HIF2α/EPAS1). HIF2α knockdown counteracted the effect of CBS knockdown on anchorage-independent growth. Bioinformatic analysis of mRNA expression data from human glioma specimens revealed a significant association between low expression of CBS mRNA and high expression of angiopoietin-like 4 (ANGPTL4) and VEGF transcripts, which are HIF2 target gene products that were also increased in CBS knockdown subclones. These results suggest that decreased CBS expression in glioma increases HIF2α protein levels and HIF2 target gene expression, which promotes glioma tumor formation. IMPLICATIONS CBS loss-of-function promotes glioma growth.
Collapse
Affiliation(s)
- Naoharu Takano
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Biochemistry, School of Medicine, Keio University, Tokyo, Japan
| | - Yasmeen Sarfraz
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York
| | - Daniele M Gilkes
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pallavi Chaturvedi
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lisha Xiang
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Makoto Suematsu
- Department of Biochemistry, School of Medicine, Keio University, Tokyo, Japan
| | - David Zagzag
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York. Division of Neuropathology, Department of Pathology and Neurosurgery, New York University School of Medicine, New York, New York
| | | |
Collapse
|
47
|
Hypoxia-inducible factors regulate human and rat cystathionine β-synthase gene expression. Biochem J 2014; 458:203-11. [PMID: 24328859 DOI: 10.1042/bj20131350] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Increased catalytic activity of CBS (cystathionine β-synthase) was recently shown to mediate vasodilation of the cerebral microcirculation, which is initiated within minutes of the onset of acute hypoxia. To test whether chronic hypoxia was a stimulus for increased CBS expression, U87-MG human glioblastoma and PC12 rat phaeochromocytoma cells were exposed to 1% or 20% O2 for 24-72 h. CBS mRNA and protein expression were increased in hypoxic cells. Hypoxic induction of CBS expression was abrogated in cells transfected with vector encoding shRNA targeting HIF (hypoxia-inducible factor) 1α or 2α. Exposure of rats to hypobaric hypoxia (0.35 atm; 1 atm=101.325 kPa) for 3 days induced increased CBS mRNA, protein and catalytic activity in the cerebral cortex and cerebellum, which was blocked by administration of the HIF inhibitor digoxin. HIF-binding sites, located 0.8 and 1.2 kb 5' to the transcription start site of the human CBS and rat Cbs genes respectively, were identified by ChIP assays. A 49-bp human sequence, which encompassed an inverted repeat of the core HIF-binding site, functioned as a hypoxia-response element in luciferase reporter transcription assays. Thus HIFs mediate tissue-specific CBS expression, which may augment cerebral vasodilation as an adaptive response to chronic hypoxia.
Collapse
|
48
|
LI YUNQUAN, LIU GUOHUI, CAI DIANQI, PAN BAOYING, LIN YUESE, LI XUANDI, LI SHUJUAN, ZHU LING, LIAO XINXUE, WANG HUISHEN. H2S inhibition of chemical hypoxia-induced proliferation of HPASMCs is mediated by the upregulation of COX-2/PGI2. Int J Mol Med 2013; 33:359-66. [DOI: 10.3892/ijmm.2013.1579] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 12/02/2013] [Indexed: 11/06/2022] Open
|
49
|
Bekpinar S, Develi-Is S, Unlucerci Y, Kusku-Kiraz Z, Uysal M, Gurdol F. Modulation of arginine and asymmetric dimethylarginine concentrations in liver and plasma by exogenous hydrogen sulfide in LPS-induced endotoxemia. Can J Physiol Pharmacol 2013; 91:1071-5. [PMID: 24289078 DOI: 10.1139/cjpp-2013-0114] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Plasma levels of asymmetric dimethylarginine (ADMA) are known to be elevated under pathological conditions, but reports on intracellular ADMA levels are scarce. In this study, we investigated whether lipopolysaccharide (LPS)-induced endotoxemia alters the intra- and extra-cellular partition of l-arginine and ADMA. The effect of H2S pretreatment was also researched. Wistar rats were given sodium hydrogen sulfide (NaHS, 1 mg·(kg body mass)(-1)) one hour before the LPS injections (20 mg·kg(-1)). Six hours after the LPS treatment, the animals were sacrificed. Myeloperoxidase (MPO) and dimethylarginine dimethylaminohydrolase (DDAH) activities and levels of hypoxia-inducible factor (HIF)-1α were measured in the liver. ADMA and arginine levels were determined using HPLC. LPS injection caused liver injury, as evidenced by the activities of alanine transaminase, aspartate transaminase, and arginase. LPS increased l-arginine content and decreased DDAH activity in the rat liver. MPO activity and HIF-1α levels indicated inflammation and hypoxia. Despite the accumulation of ADMA in the plasma, the level remained unchanged in the liver. NaHS pretreatment restored both the DDAH activity and intracellular l-arginine levels. It is concluded that increased H2S generation has a potency to restore hepatic l-arginine levels and ADMA handling in endotoxemia. Extra- and intra-cellular partitions of ADMA seem to depend on transport proteins as well as the DDAH activity.
Collapse
Affiliation(s)
- Seldag Bekpinar
- Department of Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Capa 34093, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|