1
|
Pernomian L, Blascke de Mello MM, Parente JM, Sanches-Lopes JM, Tanus-Santos JE, Parreiras E Silva LT, Antunes-Rodrigues J, da Conceição Dos Santos R, Elias LLK, Fabro AT, Silva CAA, Fazan R, de Castro MM. The hydrogen sulfide donor 4-carboxyphenyl-isothiocyanate decreases blood pressure and promotes cardioprotective effect through reduction of oxidative stress and nuclear factor kappa B/matrix metalloproteinase (MMP)-2 axis in hypertension. Life Sci 2024; 351:122819. [PMID: 38857651 DOI: 10.1016/j.lfs.2024.122819] [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: 04/18/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
AIMS Our aim was to evaluate whether the hydrogen sulfide (H2S) donor, 4-carboxyphenyl-isothiocyanate (4-CPI), exerts cardioprotective effect in the two kidney- one clip (2K-1C) rats through oxidative stress and MMP-2 activity attenuation and compare it with the classical H2S donor, Sodium Hydrosulfide (NaHS). MATERIALS AND METHODS Renovascular hypertension (two kidneys-one clip; 2K-1C) was surgically induced in male Wistar rats. After two weeks, normotensive (2K) and hypertensive rats were intraperitoneally treated with vehicle (0.6 % dimethyl sulfoxide), NaHS (0.24 mg/Kg/day) or with 4-CPI (0.24 mg/Kg/day), for more 4 weeks. Systolic blood pressure (SBP) was evaluated weekly by tail-cuff plethysmography. Heart function was assessed by using the Millar catheter. Cardiac hypertrophy and fibrosis were evaluated by hematoxylin and eosin, and Picrosirius Red staining, respectively. The H2S was analyzed using WSP-1 fluorimetry and the cardiac oxidative stress was measured by lucigenin chemiluminescence and Amplex Red. MMP-2 activity was measured by in-gel gelatin or in situ zymography assays. Nox1, gp91phox, MMP-2 and the phospho-p65 subunit (Serine 279) nuclear factor kappa B (NF-κB) levels were evaluated by Western blotting. KEY FINDINGS 4-CPI reduced blood pressure in hypertensive rats, decreased cardiac remodeling and promoted cardioprotection through the enhancement of cardiac H2S levels. An attenuation of oxidative stress, with inactivation of the p65-NF-κB/MMP-2 axis was similarly observed after NaHS or 4-CPI treatment in 2K-1C hypertension. SIGNIFICANCE H2S is a mediator that promotes cardioprotective effects and decreases blood pressure, and 4-CPI seems to be a good candidate to reverse the maladaptive remodeling and cardiac dysfunction in renovascular hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alexandre Todorovic Fabro
- Department of Pathology and Legal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | | |
Collapse
|
2
|
Lou S, Jiang ZL, Zhu YW, Zhang RY, Wang Y, Chu T, Liu YF, Zhang YX, Zhang CH, Su YK, Liu HX, Ji XY, Wu DD. Exploring the impact of hydrogen sulfide on hematologic malignancies: A review. Cell Signal 2024; 120:111236. [PMID: 38810860 DOI: 10.1016/j.cellsig.2024.111236] [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: 04/27/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
Hydrogen sulfide (H2S) is one of the three most crucial gaseous messengers in the body. The discovery of H2S donors, coupled with its endogenous synthesis capability, has sparked hope for the treatment of hematologic malignancies. In the last decade, the investigation into the impact of H2S has expanded, particularly within the fields of cardiovascular function, inflammation, infection, and neuromodulation. Hematologic malignancies refer to a diverse group of cancers originating from abnormal proliferation and differentiation of blood-forming cells, including leukemia, lymphoma, and myeloma. In this review, we delve deeply into the complex interrelation between H2S and hematologic malignancies. In addition, we comprehensively elucidate the intricate molecular mechanisms by which both H2S and its donors intricately modulate the progression of tumor growth. Furthermore, we systematically examine their impact on pivotal aspects, encompassing the proliferation, invasion, and migration capacities of hematologic malignancies. Therefore, this review may contribute novel insights to our understanding of the prospective therapeutic significance of H2S and its donors within the realm of hematologic malignancies.
Collapse
Affiliation(s)
- Shang Lou
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Zhi-Liang Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Rui-Yu Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ya-Fang Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Chuan-Hao Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Kun Su
- School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Hong-Xia Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan 450064, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, China.
| |
Collapse
|
3
|
Kies PJ, Hammer ND. A Resourceful Race: Bacterial Scavenging of Host Sulfur Metabolism during Colonization. Infect Immun 2022; 90:e0057921. [PMID: 35315692 PMCID: PMC9119060 DOI: 10.1128/iai.00579-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sulfur is a requirement for life. Therefore, both the host and colonizing bacteria must regulate sulfur metabolism in a coordinated fashion to meet cellular demands. The host environment is a rich source of organic and inorganic sulfur metabolites that are utilized in critical physiological processes such as redox homeostasis and cellular signaling. As such, modulating enzymes dedicated to sulfur metabolite biosynthesis plays a vital role in host fitness. This is exemplified from a molecular standpoint through layered regulation of this machinery at the transcriptional, translational, and posttranslational levels. With such a diverse metabolite pool available, pathogens and symbionts have evolved multiple mechanisms to exploit sulfur reservoirs to ensure propagation within the host. Indeed, characterization of sulfur transporters has revealed that bacteria employ multiple tactics to acquire ideal sulfur sources, such as cysteine and its derivatives. However, bacteria that employ acquisition strategies targeting multiple sulfur sources complicate in vivo studies that investigate how specific sulfur metabolites support proliferation. Furthermore, regulatory systems controlling the bacterial sulfur regulon are also multifaceted. This too creates an interesting challenge for in vivo work focused on bacterial regulation of sulfur metabolism in response to the host. This review examines the importance of sulfur at the host-bacterium interface and the elegant studies conducted to define this interaction.
Collapse
Affiliation(s)
- Paige J. Kies
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Neal D. Hammer
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
4
|
Maiti BK. Cross‐talk Between (Hydrogen)Sulfite and Metalloproteins: Impact on Human Health. Chemistry 2022; 28:e202104342. [DOI: 10.1002/chem.202104342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Indexed: 12/28/2022]
Affiliation(s)
- Biplab K Maiti
- Department of Chemistry National Institute of Technology Sikkim, Ravangla Campus Barfung Block, Ravangla Sub Division South Sikkim 737139 India
- Department of Chemistry Cluster University of Jammu Canal Road Jammu 180001
| |
Collapse
|
5
|
Boo YC. Metabolic Basis and Clinical Evidence for Skin Lightening Effects of Thiol Compounds. Antioxidants (Basel) 2022; 11:antiox11030503. [PMID: 35326153 PMCID: PMC8944565 DOI: 10.3390/antiox11030503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 12/17/2022] Open
Abstract
Melanin pigment is a major factor in determining the color of the skin, and its abnormal increase or decrease can cause serious pigmentation disorders. The melanin pigment of the skin is divided into light pheomelanin and dark eumelanin, and a big difference between them is whether they contain sulfur. Melanin synthesis starts from a common reaction in which tyrosine or dihydroxyphenylalanine (DOPA) is oxidized by tyrosinase (TYR) to produce dopaquinone (DQ). DQ is spontaneously converted to leukodopachrome and then oxidized to dopachrome, which enters the eumelanin synthesis pathway. When DQ reacts with cysteine, cysteinyl dopa is generated, which is oxidized to cysteinyl DQ and enters the pheomelanin synthesis pathway. Therefore, thiol compounds can influence the relative synthesis of eumelanin and pheomelanin. In addition, thiol compounds can inhibit enzymatic activity by binding to copper ions at the active site of TYR, and act as an antioxidant scavenging reactive oxygen species and free radicals or as a modulator of redox balance, thereby inhibiting overall melanin synthesis. This review will cover the metabolic aspects of thiol compounds, the role of thiol compounds in melanin synthesis, comparison of the antimelanogenic effects of various thiol compounds, and clinical trials on the skin lightening efficacy of thiol compounds. We hope that this review will help identify the advantages and disadvantages of various thiol compounds as modulators of skin pigmentation and contribute to the development of safer and more effective strategies for the treatment of pigmentation disorders.
Collapse
Affiliation(s)
- Yong Chool Boo
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea;
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
| |
Collapse
|
6
|
Jiang X, MacArthur MR, Treviño-Villarreal JH, Kip P, Ozaki CK, Mitchell SJ, Mitchell JR. Intracellular H 2S production is an autophagy-dependent adaptive response to DNA damage. Cell Chem Biol 2021; 28:1669-1678.e5. [PMID: 34166610 PMCID: PMC8665944 DOI: 10.1016/j.chembiol.2021.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/08/2021] [Accepted: 05/26/2021] [Indexed: 12/21/2022]
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter with broad physiological activities, including protecting cells against stress, but little is known about the regulation of cellular H2S homeostasis. We have performed a high-content small-molecule screen and identified genotoxic agents, including cancer chemotherapy drugs, as activators of intracellular H2S levels. DNA damage-induced H2S in vitro and in vivo. Mechanistically, DNA damage elevated autophagy and upregulated H2S-generating enzyme CGL; chemical or genetic disruption of autophagy or CGL impaired H2S induction. Importantly, exogenous H2S partially rescued autophagy-deficient cells from genotoxic stress. Furthermore, stressors that are not primarily genotoxic (growth factor depletion and mitochondrial uncoupler FCCP) increased intracellular H2S in an autophagy-dependent manner. Our findings highlight the role of autophagy in H2S production and suggest that H2S generation may be a common adaptive response to DNA damage and other stressors.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| | - Michael R MacArthur
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | | | - Peter Kip
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Einthoven Laboratory for Experimental Vascular Medicine and Department of Surgery, Leiden University Medical Center, 2333 CC Leiden, the Netherlands
| | - C Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sarah J Mitchell
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland.
| | - James R Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
7
|
Malard E, Valable S, Bernaudin M, Pérès E, Chatre L. The Reactive Species Interactome in the Brain. Antioxid Redox Signal 2021; 35:1176-1206. [PMID: 34498917 DOI: 10.1089/ars.2020.8238] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: Redox pioneer Helmut Sies attempted to explain reactive species' challenges faced by organelles, cells, tissues, and organs via three complementary definitions: (i) oxidative stress, that is, the disturbance in the prooxidant-antioxidant defense balance in favor of the prooxidants; (ii) oxidative eustress, the low physiological exposure to prooxidants; and (iii) oxidative distress, the supraphysiological exposure to prooxidants. Recent Advances: Identification, concentration, and interactions are the most important elements to improve our understanding of reactive species in physiology and pathology. In this context, the reactive species interactome (RSI) is a new multilevel redox regulatory system that identifies reactive species families, reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species, and it integrates their interactions with their downstream biological targets. Critical Issues: We propose a united view to fully combine reactive species identification, oxidative eustress and distress, and the RSI system. In this view, we also propose including the forgotten reactive carbonyl species, an increasingly rediscovered reactive species family related to the other reactive families, and key enzymes within the RSI. We focus on brain physiology and pathology to demonstrate why this united view should be considered. Future Directions: More studies are needed for an improved understanding of the contributions of reactive species through their identification, concentration, and interactions, including in the brain. Appreciating the RSI in its entirety should unveil new molecular players and mechanisms in physiology and pathology in the brain and elsewhere.
Collapse
Affiliation(s)
- Elise Malard
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Elodie Pérès
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Laurent Chatre
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| |
Collapse
|
8
|
Scrivner O, Kumar MR, Sorokolet K, Wong A, Kebaara B, Farmer PJ. Characterization of Endogenous and Extruded H 2S and Small Oxoacids of Sulfur (SOS) in Cell Cultures. ACS Chem Biol 2021; 16:1413-1424. [PMID: 34374506 DOI: 10.1021/acschembio.1c00257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This report characterizes and quantifies endogenous hydrogen sulfide (H2S) and small oxoacids of sulfur (SOS = HOSH, HOSOH) in a panel of cell lines including human cancer (A375 melanoma cells, HeLa cervical cells) and noncancer (HEK293 embryonic kidney cells), as well as E. coli DH5α and S. cerevisiae S288C. The methodology used is a translation of well-studied nucleophilic and electrophilic traps for cysteine and oxidized cysteines residues to target small molecular weight sulfur species; mass spectrometric analysis allows for species quantification. The observed intracellular concentrations of H2S and SOS vary in different cell types, from nanomolar to femtomolar, typically with H2S > HOSOH > HOSH. We propose the term sulfome, a subset of the metabolome, describing the nonproteinaceous metabolites of H2S; the sulfomic index is as a measure of the S-oxide redox status, which gives a profile of endogenous sulfur at different oxidation states. An important observation is that H2S and SOS were found to be continuously extruded into surrounding media against a concentration gradient, implying an active efflux process. Small molecule inhibition of several H2S generating enzymes suggest that SOS are not derived solely from H2S oxidation. Even after successful inhibition of H2S production, cells maintain constant efflux and repopulate H2S and SOS over time. This work proves that these small sulfur oxoacids are generated in cells of all types, and their efflux implies that they play a role in cell signaling and possibly other vascular physiology attributed to H2S.
Collapse
Affiliation(s)
- Ottis Scrivner
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Murugaeson R. Kumar
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Kristina Sorokolet
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Angelo Wong
- Department of Biology, Baylor University, Waco, Texas 76898, United States
| | - Bessie Kebaara
- Department of Biology, Baylor University, Waco, Texas 76898, United States
| | - Patrick J. Farmer
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| |
Collapse
|
9
|
Peng SY, Wu X, Lu T, Cui G, Chen G. Research progress of hydrogen sulfide in Alzheimer's disease from laboratory to hospital: a narrative review. Med Gas Res 2021; 10:125-129. [PMID: 33004710 PMCID: PMC8086622 DOI: 10.4103/2045-9912.296043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease is a neurodegenerative disease that mainly occurs in old age and early stages. Its main manifestations are memory impairment, aphasia, apraxia, loss of identity, abstract thinking and impairment of computing power, personality and behavior changes, etc. At present, the treatment of Alzheimer's disease only stays on reducing the disease and delaying the development, which is also a difficult problem to overcome in clinical practice. Hydrogen sulfide, as a third gaseous signal molecule after carbon monoxide and nitrogen monoxide, has become very popular in recent years. It shows very promising prospects in the Alzheimer's disease model. It can protect the nerve function and prevent the progress of the disease by affecting the amyloid precursor protein metabolism, anti-apoptosis, anti-inflammatory, and antioxidant pathways. Therefore, this article summarizes the relevant basic and clinical research of hydrogen sulfide in Alzheimer's disease, and discusses its progress and findings and mechanism characteristics.
Collapse
Affiliation(s)
- Song-Yang Peng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xin Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ting Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
10
|
Pedre B, Barayeu U, Ezeriņa D, Dick TP. The mechanism of action of N-acetylcysteine (NAC): The emerging role of H 2S and sulfane sulfur species. Pharmacol Ther 2021; 228:107916. [PMID: 34171332 DOI: 10.1016/j.pharmthera.2021.107916] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022]
Abstract
Initially adopted as a mucolytic about 60 years ago, the cysteine prodrug N-acetylcysteine (NAC) is the standard of care to treat paracetamol intoxication, and is included on the World Health Organization's list of essential medicines. Additionally, NAC increasingly became the epitome of an "antioxidant". Arguably, it is the most widely used "antioxidant" in experimental cell and animal biology, as well as clinical studies. Most investigators use and test NAC with the idea that it prevents or attenuates oxidative stress. Conventionally, it is assumed that NAC acts as (i) a reductant of disulfide bonds, (ii) a scavenger of reactive oxygen species and/or (iii) a precursor for glutathione biosynthesis. While these mechanisms may apply under specific circumstances, they cannot be generalized to explain the effects of NAC in a majority of settings and situations. In most cases the mechanism of action has remained unclear and untested. In this review, we discuss the validity of conventional assumptions and the scope of a newly discovered mechanism of action, namely the conversion of NAC into hydrogen sulfide and sulfane sulfur species. The antioxidative and cytoprotective activities of per- and polysulfides may explain many of the effects that have previously been ascribed to NAC or NAC-derived glutathione.
Collapse
Affiliation(s)
- Brandán Pedre
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Daria Ezeriņa
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
11
|
Lignelli E, Palumbo F, Bayindir SG, Nagahara N, Vadász I, Herold S, Seeger W, Morty RE. The H 2S-generating enzyme 3-mercaptopyruvate sulfurtransferase regulates pulmonary vascular smooth muscle cell migration and proliferation but does not impact normal or aberrant lung development. Nitric Oxide 2021; 107:31-45. [PMID: 33338600 DOI: 10.1016/j.niox.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/07/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
Along with nitric oxide (NO), the gasotransmitters carbon monoxide (CO) and hydrogen sulfide (H2S) are emerging as potentially important players in newborn physiology, as mediators of newborn disease, and as new therapeutic modalities. Several recent studies have addressed H2S in particular in animal models of bronchopulmonary dysplasia (BPD), a common complication of preterm birth where oxygen toxicity stunts lung development. In those studies, exogenous H2S attenuated the impact of oxygen toxicity on lung development, and two H2S-generating enzymes were documented to affect pulmonary vascular development. H2S is directly generated endogenously by three enzymes, one of which, 3-mercaptopyruvate sulfurtransferase (MPST), has not been studied in the lung. In a hyperoxia-based animal model of BPD, oxygen exposure deregulated MPST expression during post-natal lung development, where MPST was localized to the smooth muscle layer of the pulmonary vessels in developing lungs. siRNA-mediated abrogation of MPST expression in human pulmonary artery smooth muscle cells in vitro limited baseline cell migration and cell proliferation, without affecting apoptosis or cell viability. In vivo, MPST was dispensable for normal lung development in Mpst-/-mice, and MPST did not contribute to stunted lung development driven by hyperoxia exposure, assessed by design-based stereology. These data demonstrate novel roles for MPST in pulmonary vascular smooth muscle cell physiology. The potential caveats of using Mpst-/- mice to study normal and aberrant lung development are also discussed, highlighting the possible confounding, compensatory effects of other H2S-generating enzymes that are present alongside MPST in the smooth muscle compartment of developing pulmonary vessels.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Selahattin Görkem Bayindir
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Noriyuki Nagahara
- Isotope Research Laboratory, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; Institute for Lung Health (ILH), Justus Liebig University Giessen, Aulweg 130, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany.
| |
Collapse
|
12
|
Yang B, Zhao W, Yin C, Bai Y, Wang S, Xing G, Li F, Bian J, Aschner M, Cai J, Shi H, Lu R. Acute acrylonitrile exposure inhibits endogenous H 2S biosynthesis in rat brain and liver: The role of CBS/3-MPST-H 2S pathway in its astrocytic toxicity. Toxicology 2021; 451:152685. [PMID: 33486070 DOI: 10.1016/j.tox.2021.152685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/12/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022]
Abstract
Hydrogen sulfide (H2S) as the third gasotransmitter molecule serves various biological regulatory roles in health and disease. Acrylonitrile (AN) is a common occupational toxicant and environmental pollutant, causing brain and liver damage in mammals. The biotransformation of AN is dependent-upon reduced glutathione (GSH), cysteine and other sulfur-containing compounds. However, the effects of AN on the endogenous H2S biosynthesis pathway have yet to be determined. Herein, we demonstrated that a single exposure to AN (at 25, 50, or 75 mg/kg for 1, 6 or 24 h) decreased the endogenous H2S content and H2S-producing capacity in a dose-dependent manner, both in the cerebral cortex and liver of rats in vivo. In addition, the inhibitory effects of AN (1, 2.5, 5, 10 mM for 12 h) on the H2S content and/or the expression of H2S-producing enzymes were also found both in primary rat astrocytes and rat liver cell line (BRL cells). Impairment in the H2S biosynthesis pathway was also assessed in primary rat astrocytes treated with AN. It was found that inhibition of the cystathionine-β-synthase (CBS)/3-mercaptopyruvate sulfurtransferase (3-MPST)-H2S pathway with the CBS inhibitor or 3-MPST-targeted siRNA significantly increased the AN-induced (5 mM for 12 h) cytotoxicity in astrocytes. In turn, CBS activation or 3-MPST overexpression as well as exogenous NaHS supplementation significantly attenuated AN-induced cytotoxicity. Taken together, endogenous H2S biosynthesis pathway was disrupted in rats acutely exposed to AN, which contributes to acute AN neurotoxicity in primary rat astrocytes.
Collapse
Affiliation(s)
- Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Wenjun Zhao
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China; Department of Clinical Laboratory, Affiliated People's Hospital to Jiangsu University School of Medicine, Zhenjiang, Jiangsu, 212002, China
| | - Changsheng Yin
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Yu Bai
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Jinsong Bian
- Department of Pharmacology, School of Medicine, National Singapore University, 117597, Singapore
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jiyang Cai
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Lindsay, Oklahoma City, OK, 73104, USA
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China; Center for Experimental Research, Affiliated Kunshan Hospital to Jiangsu University School of Medicine, Kunshan, Suzhou, Jiangsu, 215132, China.
| |
Collapse
|
13
|
Pedre B, Dick TP. 3-Mercaptopyruvate sulfurtransferase: an enzyme at the crossroads of sulfane sulfur trafficking. Biol Chem 2020; 402:223-237. [PMID: 33055309 DOI: 10.1515/hsz-2020-0249] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
3-Mercaptopyruvate sulfurtransferase (MPST) catalyzes the desulfuration of 3-mercaptopyruvate to generate an enzyme-bound hydropersulfide. Subsequently, MPST transfers the persulfide's outer sulfur atom to proteins or small molecule acceptors. MPST activity is known to be involved in hydrogen sulfide generation, tRNA thiolation, protein urmylation and cyanide detoxification. Tissue-specific changes in MPST expression correlate with ageing and the development of metabolic disease. Deletion and overexpression experiments suggest that MPST contributes to oxidative stress resistance, mitochondrial respiratory function and the regulation of fatty acid metabolism. However, the role and regulation of MPST in the larger physiological context remain to be understood.
Collapse
Affiliation(s)
- Brandán Pedre
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120Heidelberg, Germany
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120Heidelberg, Germany
| |
Collapse
|
14
|
Stipanuk MH. Metabolism of Sulfur-Containing Amino Acids: How the Body Copes with Excess Methionine, Cysteine, and Sulfide. J Nutr 2020; 150:2494S-2505S. [PMID: 33000151 DOI: 10.1093/jn/nxaa094] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/28/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolism of excess methionine (Met) to homocysteine (Hcy) by transmethylation is facilitated by the expression of methionine adenosyltransferase (MAT) I/III and glycine N-methyltransferase (GNMT) in liver, and a lack of either enzyme results in hypermethioninemia despite normal concentrations of MATII and methyltransferases other than GNMT. The further metabolism of Hcy by the transsulfuration pathway is facilitated by activation of cystathionine β-synthase (CBS) by S-adenosylmethionine (SAM) as well as the relatively high KM of CBS for Hcy. Transmethylation plus transsulfuration effects catabolism of the Met molecule along with transfer of the sulfur atom of Met to serine to synthesize cysteine (Cys). Oxidation and excretion of Met sulfur depend upon Cys catabolism and sulfur oxidation pathways. Excess Cys is oxidized by cysteine dioxygenase 1 (CDO1) and further metabolized to taurine or sulfate. Some Cys is normally metabolized by desulfhydration pathways, and the hydrogen sulfide (H2S) produced is further oxidized to sulfate. If Cys or Hcy concentrations are elevated, Cys or Hcy desulfhydration can result in excess H2S and thiosulfate production. Excess Cys or Met may also promote their limited metabolism by transamination pathways.
Collapse
Affiliation(s)
- Martha H Stipanuk
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| |
Collapse
|
15
|
Walsh BJC, Giedroc DP. H 2S and reactive sulfur signaling at the host-bacterial pathogen interface. J Biol Chem 2020; 295:13150-13168. [PMID: 32699012 PMCID: PMC7504917 DOI: 10.1074/jbc.rev120.011304] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial pathogens that cause invasive disease in the vertebrate host must adapt to host efforts to cripple their viability. Major host insults are reactive oxygen and reactive nitrogen species as well as cellular stress induced by antibiotics. Hydrogen sulfide (H2S) is emerging as an important player in cytoprotection against these stressors, which may well be attributed to downstream more oxidized sulfur species termed reactive sulfur species (RSS). In this review, we summarize recent work that suggests that H2S/RSS impacts bacterial survival in infected cells and animals. We discuss the mechanisms of biogenesis and clearance of RSS in the context of a bacterial H2S/RSS homeostasis model and the bacterial transcriptional regulatory proteins that act as "sensors" of cellular RSS that maintain H2S/RSS homeostasis. In addition, we cover fluorescence imaging- and MS-based approaches used to detect and quantify RSS in bacterial cells. Last, we discuss proteome persulfidation (S-sulfuration) as a potential mediator of H2S/RSS signaling in bacteria in the context of the writer-reader-eraser paradigm, and progress toward ascribing regulatory significance to this widespread post-translational modification.
Collapse
Affiliation(s)
- Brenna J C Walsh
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
| | - David P Giedroc
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana, USA.
| |
Collapse
|
16
|
Adams JC, Bell PD, Bodine SC, Brooks HL, Bunnett N, Joe B, Keehan KH, Kleyman TR, Marette A, Morty RE, Ramírez JM, Thomsen MB, Yates BJ, Zucker IH. An American Physiological Society cross-journal Call for Papers on "Deconstructing Organs: Single-Cell Analyses, Decellularized Organs, Organoids, and Organ-on-a-Chip Models". Am J Physiol Lung Cell Mol Physiol 2020; 319:L266-L272. [PMID: 32609556 PMCID: PMC7473938 DOI: 10.1152/ajplung.00311.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sue C Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Nigel Bunnett
- Department of Molecular Pathobiology, New York University, New York, New York
| | - Bina Joe
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | | | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Hôpital Laval, Quebec City, Quebec, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, Justus Liebig University Giessen, member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jan-Marino Ramírez
- Department of Neurological Surgery, University of Washington Medical Center, Seattle, Washington.,Center on Human Development and Disability, University of Washington, Seattle, Washington.,Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington, Seattle, Washington
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bill J Yates
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
17
|
Ahmad A, Druzhyna N, Szabo C. Effect of 3-mercaptopyruvate Sulfurtransferase Deficiency on the Development of Multiorgan Failure, Inflammation, and Wound Healing in Mice Subjected to Burn Injury. J Burn Care Res 2020; 40:148-156. [PMID: 30649358 DOI: 10.1093/jbcr/irz007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The gaseous transmitter hydrogen sulfide (H2S) has been implicated in various forms of critical illness. Here, we have compared the outcome of scald burn injury in wild-type mice and in mice deficient in 3-mercaptopyruvate sulfurtransferase (3-MST), a mammalian H2S-generating enzyme. Outcome variables included indices of organ injury, clinical chemistry parameters, and plasma levels of inflammatory mediators. Plasma levels of H2S significantly increased in response to burn in wild-type mice, but remained unchanged in 3-MST-/- mice. The capacity of tissue homogenates to produce H2S from 3-mercaptopyruvate was unaffected by burn injury. In 3-MST-/- mice, compared to wild-type controls, there was a significant enhancement in the accumulation of polymorphonuclear cells (as assessed by the quantification of myeloperoxidase) in the liver (but not heart, lung, or skin) at 7 days postburn. Oxidative tissue damage (as assessed by malon dialdehyde content) was comparable between wild-type and 3-MST-deficient mice in all tissues studied. 3-MST-/- and wild-type mice exhibited comparable burn-induced elevations in circulating plasma levels of hepatic injury; however, 3-MST-/- mice exhibited a higher degree of renal injury (as reflected by elevated blood urea nitrogen levels) at 7 days postburn. Inflammatory mediators (eg, TNF-α, IL-1β, IL-2, IL-6, IL-10, and IL-12) increased in burn injury, but without significant differences between the 3-MST-/- and wild-type groups. The healing of the burn wound was also unaffected by 3-MST deficiency. In conclusion, the absence of the H2S-producing enzyme 3-MST slightly exacerbates the development of multiorgan dysfunction but does not affect inflammatory mediator production or wound healing in a murine model of burn injury.
Collapse
Affiliation(s)
- Akbar Ahmad
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston
| | - Nadiya Druzhyna
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston.,Shriners Hospital for Children, Galveston, Texas.,Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Switzerland
| |
Collapse
|
18
|
Eleftheriadis T, Pissas G, Nikolaou E, Filippidis G, Liakopoulos V, Stefanidis I. Mistimed H 2S upregulation, Nrf2 activation and antioxidant proteins levels in renal tubular epithelial cells subjected to anoxia and reoxygenation. Biomed Rep 2020; 13:3. [PMID: 32509306 DOI: 10.3892/br.2020.1309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/08/2020] [Indexed: 12/28/2022] Open
Abstract
Ischemia-reperfusion (I-R) injury is involved in the pathogenesis of several human diseases. In the present study, the kinetics of the H2S producing enzymes-nuclear factor erythroid 2-like 2 (Nrf2)-antioxidant proteins axis under anoxia or reoxygenation was evaluated, as well as its effects on survival of mouse renal proximal tubular epithelial cells (RPTECs). In RPTECs subjected to anoxia and subsequent reoxygenation, reactive oxygen species (ROS) production, lipid peroxidation, ferroptotic cell death, the levels of the H2S producing enzymes and H2S, the expression of Nrf2 and its transcriptional targets superoxide dismutase-3, glutathione reductase, ferritin H and cystine-glutamate antiporter, as well as apoptosis, and the levels of p53, Bax and phosphorylated p53 were assessed. When needed, the H2S producing enzyme inhibitor aminooxyacetate, or the ferroptosis inhibitor α-tocopherol, were used. Reoxygenation induced ferroptosis, whereas anoxia activated the p53-Bax pathway and induced apoptosis. The H2S producing enzymes-Nrf2-antioxidant proteins axis was activated only during anoxia and not during reoxygenation, when cellular viability is threatened by ROS overproduction and the ensuing ferroptosis. The activation of the above axis during anoxia ameliorated the effects of the apoptotic p53-Bax pathway, but did not adequately protect against apoptosis. In conclusion, the H2S-Nrf2 axis is activated by anoxia, and although it reduces apoptosis, it does not completely prevent apoptotic cell death. Additionally, following reoxygenation, the above axis was not activated. This mistimed activation of the H2S producing enzymes-Nrf2-antioxidant proteins axis contributes to reoxygenation-induced cell death. Determining the exact molecular mechanisms involved in reoxygenation-induced cell death may assist in the development of clinically relevant interventions for preventing I-R injury.
Collapse
Affiliation(s)
- Theodoros Eleftheriadis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Georgios Pissas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Evdokia Nikolaou
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Georgios Filippidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Vassilios Liakopoulos
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece
| |
Collapse
|
19
|
Zuhra K, Augsburger F, Majtan T, Szabo C. Cystathionine-β-Synthase: Molecular Regulation and Pharmacological Inhibition. Biomolecules 2020; 10:E697. [PMID: 32365821 PMCID: PMC7277093 DOI: 10.3390/biom10050697] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Cystathionine-β-synthase (CBS), the first (and rate-limiting) enzyme in the transsulfuration pathway, is an important mammalian enzyme in health and disease. Its biochemical functions under physiological conditions include the metabolism of homocysteine (a cytotoxic molecule and cardiovascular risk factor) and the generation of hydrogen sulfide (H2S), a gaseous biological mediator with multiple regulatory roles in the vascular, nervous, and immune system. CBS is up-regulated in several diseases, including Down syndrome and many forms of cancer; in these conditions, the preclinical data indicate that inhibition or inactivation of CBS exerts beneficial effects. This article overviews the current information on the expression, tissue distribution, physiological roles, and biochemistry of CBS, followed by a comprehensive overview of direct and indirect approaches to inhibit the enzyme. Among the small-molecule CBS inhibitors, the review highlights the specificity and selectivity problems related to many of the commonly used "CBS inhibitors" (e.g., aminooxyacetic acid) and provides a comprehensive review of their pharmacological actions under physiological conditions and in various disease models.
Collapse
Affiliation(s)
- Karim Zuhra
- Chair of Pharmacology, Section of Medicine, University of Fribourg, 1702 Fribourg, Switzerland; (K.Z.); (F.A.)
| | - Fiona Augsburger
- Chair of Pharmacology, Section of Medicine, University of Fribourg, 1702 Fribourg, Switzerland; (K.Z.); (F.A.)
| | - Tomas Majtan
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, 1702 Fribourg, Switzerland; (K.Z.); (F.A.)
| |
Collapse
|
20
|
Structure-activity relationships study of isothiocyanates for H 2S releasing properties: 3-Pyridyl-isothiocyanate as a new promising cardioprotective agent. J Adv Res 2020; 27:41-53. [PMID: 33318865 PMCID: PMC7728584 DOI: 10.1016/j.jare.2020.02.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/27/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction The gasotransmitter hydrogen sulphide (H2S), an endogenous ubiquitous signalling molecule, is known for its beneficial effects on different mammalian systems. H2S exhibits cardioprotective activity against ischemia/reperfusion (I/R) or hypoxic injury. Methods A library of forty-five isothiocyanates, selected for their different chemical properties, has been evaluated for its hydrogen sulfide (H2S) releasing capacity. The obtained results allowed to correlate several factors such as steric hindrance, electronic effects and position of the substituents to the observed H2S production. Moreover, the chemical-physical profiles of the selected compounds have been studied by an in silico approach and from a combination of the obtained results, 3-pyridyl-isothiocyanate (25) has been selected as the most promising one. A detailed pharmacological characterization of its cardioprotective action has been performed. Results The results herein obtained strongly indicate 3-pyridyl-isothiocyanate (25) as a suitable pharmacological option in anti-ischemic therapy. The cardioprotective effects of compound 25 were tested in vivo and found to exhibit a positive effect. Conclusion Results strongly suggest that isothiocyanate-based H2S-releasing drugs, such as compound 25, can trigger a ‘‘pharmacological pre-conditioning” and could represent a suitable pharmacological option in antiischemic therapy.
Collapse
|
21
|
Zhang J, Shi C, Wang H, Gao C, Chang P, Chen X, Shan H, Zhang M, Tao L. Hydrogen sulfide protects against cell damage through modulation of PI3K/Akt/Nrf2 signaling. Int J Biochem Cell Biol 2019; 117:105636. [PMID: 31654751 DOI: 10.1016/j.biocel.2019.105636] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/01/2023]
Abstract
Hydrogen sulfide as the third endogenous gaseous mediator had protective effects against traumatic brain injury-induced neuronal damage in mice. However, the exact pathophysiological mechanism underlying traumatic brain injury is complicated and the protective role of H2S is not yet fully known. Therefore, we combined the mechanical injury (scratch) with secondary injury including metabolic impairment (no glucose) together to investigate the underlying cellular mechanism of hydrogen sulfide in vitro models of traumatic brain injury. In the present study, we found that H2S could prevent the scratch-induced decrease in the expression of cystathionine-β-synthetase, a key enzyme involved in the source of hydrogen sulfide, and endogenous hydrogen sulfide generation in PC12 cells. We also found that hydrogen sulfide could prevent scratch-induced cellular injury, alteration of mitochondrial membrane potential, intracellular accumulation of reactive oxygen species and cell death (autophagic cell death and apoptosis) in PC12 cells. It was also found that blocking PI3K/AKT pathway by LY294002, abolished the protection of H2S against scratch-induced cellular reactive oxygen species level and NRF2 accumulation and function in the nucleus. These results suggest that hydrogen sulfide protects against cell damage induced by scratch injury through modulation of the PI3K/Akt/Nrf2 pathway. This study raises the possibility that hydrogen sulfide may have therapeutic efficacy in traumatic brain injury.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Institute of Forensic Sciences, Soochow University, Suzhou, 215123, China
| | - Chaoqun Shi
- Institute of Forensic Sciences, Soochow University, Suzhou, 215123, China
| | - Haochen Wang
- Institute of Forensic Sciences, Soochow University, Suzhou, 215123, China
| | - Cheng Gao
- Institute of Forensic Sciences, Soochow University, Suzhou, 215123, China
| | - Pan Chang
- Central Laboratory, The Second Affiliated Hospital of Xi'an Medical College, Xi'an, Shaanxi, 710038, China
| | - Xiping Chen
- Institute of Forensic Sciences, Soochow University, Suzhou, 215123, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215000, China.
| | - Mingyang Zhang
- Institute of Forensic Sciences, Soochow University, Suzhou, 215123, China; School of Pharmacy, Soochow University, Suzhou, 215000, China.
| | - Luyang Tao
- Institute of Forensic Sciences, Soochow University, Suzhou, 215123, China
| |
Collapse
|
22
|
Eleftheriadis T, Pissas G, Nikolaou E, Liakopoulos V, Stefanidis I. The H2S-Nrf2-Antioxidant Proteins Axis Protects Renal Tubular Epithelial Cells of the Native Hibernator Syrian Hamster from Reoxygenation-Induced Cell Death. BIOLOGY 2019; 8:biology8040074. [PMID: 31574983 PMCID: PMC6955957 DOI: 10.3390/biology8040074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/31/2022]
Abstract
During hibernation, repeated cycles of ischemia-reperfusion (I-R) leave vital organs without injury. Studying this phenomenon may reveal pathways applicable to improving outcomes in I-R injury-induced human diseases. We evaluated whether the H2S–nuclear factor erythroid 2-like 2 (Nrf2)–antioxidant proteins axis protects renal proximal tubular epithelial cells (RPTECs) of the native hibernator, the Syrian hamster, from reperfusion-induced cell death. To imitate I-R, the hamsters’, and control mice’s RPTECs were subjected to warm anoxia, washed, and then subjected to reoxygenation in fresh culture medium. Whenever required, the H2S-producing enzymes inhibitor aminooxyacetate or the lipid peroxidation inhibitor α-tocopherol were used. A handmade H2S detection methylene blue assay, a reactive oxygen species (ROS) detection kit, a LDH release cytotoxicity assay kit, and western blotting were used. Reoxygenation upregulated the H2S-producing enzymes cystathionine beta-synthase, cystathionine γ-lyase, and 3-mercaptopyruvate sulfurtransferase in the hamster, but not in mouse RPTECs. As a result, H2S production increased only in the hamster RPTECs under reoxygenation conditions. Nrf2 expression followed the alterations of H2S production leading to an enhanced level of the antioxidant enzymes superoxide dismutase 3 and glutathione reductase, and anti-ferroptotic proteins ferritin H and cystine-glutamate antiporter. The upregulated antioxidant enzymes and anti-ferroptotic proteins controlled ROS production and rescued hamster RPTECs from reoxygenation-induced, lipid peroxidation-mediated cell death. In conclusion, in RPTECs of the native hibernator Syrian hamster, reoxygenation activates the H2S–Nrf2–antioxidant proteins axis, which rescues cells from reoxygenation-induced cell death. Further studies may reveal that the therapeutic activation of this axis in non-hibernating species, including humans, may be beneficial in I-R injury-induced diseases.
Collapse
Affiliation(s)
- Theodoros Eleftheriadis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece.
| | - Georgios Pissas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece.
| | - Evdokia Nikolaou
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece.
| | - Vassilios Liakopoulos
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece.
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Biopolis, Mezourlo Hill, 41110 Larissa, Greece.
| |
Collapse
|
23
|
Yuan C, Hou HT, Chen HX, Wang J, Wang ZQ, Chen TN, Novakovic A, Marinko M, Yang Q, Liu ZG, He GW. Hydrogen sulfide-mediated endothelial function and the interaction with eNOS and PDE5A activity in human internal mammary arteries. J Int Med Res 2019; 47:3778-3791. [PMID: 31155983 PMCID: PMC6726794 DOI: 10.1177/0300060519847386] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/09/2019] [Indexed: 12/20/2022] Open
Abstract
Objective To investigate the role of hydrogen sulfide (H2S) in human internal mammary arteries (IMA) and its interaction with endothelial nitric oxide synthase (eNOS) and phosphodiesterase (PDE)5A activity. Methods Human IMA segments from patients undergoing coronary artery bypass grafting (CABG) were studied by myography for acetylcholine and sodium hydrosulfide (NaHS)-induced relaxation. Locations of 3-mercaptopyruvate sulfurtransferase (3-MPST) and cysteine aminotransferase (CAT) were examined immunohistochemically. Levels of H2S, eNOS, phosphorylated-eNOSser1177, and PDE5A were measured. Results In IMA segments from 47 patients, acetylcholine-induced relaxation (resistant to NG-nitro-L-arginine and indomethacin) was significantly attenuated by aminooxyacetic acid or L-aspartate (CAT inhibitors), iberiotoxin (large-conductance calcium-activated K+ channel blocker), TRAM-34 plus apamin (intermediate- and small-conductance Ca2+-activated K+ channel blockers) or glibenclamide (ATP-sensitive K+ channel blocker). 3-MPST and mitochondrial CAT were found in endothelial and smooth muscle cells while cytosolic CAT was located only in endothelial cells. Acetylcholine significantly increased the H2S levels. The H2S donor, NaHS, increased eNOS phosphorylation and down-regulated PDE5A. Conclusions Human conduit artery endothelium releases H2S under basal and stimulated conditions, involving the 3-MPST/CAT pathway, eNOS phosphorylation, PDE5A activity, and potassium channels. These findings may provide new therapeutic targets for treating vasospasm in CABG grafts and facilitate the development of new vasodilator drugs.
Collapse
Affiliation(s)
- Chao Yuan
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
- Postdoctoral Station, Medical College, Nankai University, Tianjin, China
| | - Hai-Tao Hou
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
- Medical College, Zhejiang University, Hangzhou, China
| | - Huan-Xin Chen
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Jun Wang
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Zheng-Qing Wang
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Tie-Nan Chen
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Aleksandra Novakovic
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Marija Marinko
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Qin Yang
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Zhi-Gang Liu
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Guo-Wei He
- Center for Basic Medical Research and Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin, China
- Medical College, Zhejiang University, Hangzhou, China
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
24
|
Luo H, Wu PF, Han QQ, Cao Y, Deng SL, Wang J, Deng Q, Wang F, Chen JG. Reactive Sulfur Species Emerge as Gliotransmitters to Support Memory via Sulfuration-Dependent Gating of NR2A-Containing N-Methyl-d-Aspartate Subtype Glutamate Receptor Function. Antioxid Redox Signal 2019; 30:1880-1899. [PMID: 30187770 DOI: 10.1089/ars.2018.7503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS Astrocytes have been revealed as a controller of synaptic plasticity and memory via releasing gliotransmitters. Our recent findings showed that reactive sulfur species (RSS), including hydrogen sulfide (H2S) and polysulfide (H2Sn), regulated the availability of d-serine, which is a well-known gliotransmitter that is involved in synaptic plasticity. An interesting question is whether RSS, which are small molecules, can function as direct gliotransmitters to integrate astrocyte-neuron interactions throughout the memory process. RESULTS We found that hippocampal RSS level increased significantly in response to learning. We further demonstrated that the activity-triggered RSS signal controlled memory formation by using pharmacological and genetic approaches. The RSS-supporting memory was primarily conferred by enzymes that were mainly located in astrocytes, including cystathionine β-synthase (CBS) and mercaptopyruvate sulfurtransferase (3-MST), and the memory-promoting effects were mostly dependent on sulfration of the NR2A subunit of N-methyl-d-aspartate subtype glutamate receptors (NMDARs). Further, RSS were demonstrated to buffer the strong inhibitory effect of synaptically released zinc on NR2A-containing NMDARs. Innovation and Conclusion: These results suggest that glial-derived RSS signals can serve as direct gliotransmitters that regulate memory formation through the redox modulation of postsynaptic receptors; this conclusion will enrich the gliotransmission hypothesis.
Collapse
Affiliation(s)
- Han Luo
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng-Fei Wu
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Han
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Cao
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Long Deng
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji Wang
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiao Deng
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wang
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2 Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.,3 The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.,4 Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China.,5 The Collaborative-Innovation Center for Brain Science, Wuhan, China
| | - Jian-Guo Chen
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2 Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.,3 The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.,4 Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China.,5 The Collaborative-Innovation Center for Brain Science, Wuhan, China
| |
Collapse
|
25
|
Pilkington LI, Deed RC, Parish-Virtue K, Huang CW, Walker ME, Jiranek V, Barker D, Fedrizzi B. Iterative synthetic strategies and gene deletant experiments enable the first identification of polysulfides in Saccharomyces cerevisiae. Chem Commun (Camb) 2019; 55:8868-8871. [DOI: 10.1039/c9cc03020d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Polysulfides, potential signalling molecules, were synthesised and then found and explored for the first time in yeast.
Collapse
Affiliation(s)
- Lisa I. Pilkington
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Rebecca C. Deed
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
- School of Biological Sciences
| | - Katie Parish-Virtue
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Chien-Wei Huang
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Michelle E. Walker
- Department of Wine and Food Science
- The University of Adelaide
- Wine Innovation Central
- Adelaide
- Australia
| | - Vladimir Jiranek
- Department of Wine and Food Science
- The University of Adelaide
- Wine Innovation Central
- Adelaide
- Australia
| | - David Barker
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| | - Bruno Fedrizzi
- School of Chemical Sciences
- The University of Auckland
- Auckland 1142
- New Zealand
| |
Collapse
|
26
|
Tsubota M, Okawa Y, Irie Y, Maeda M, Ozaki T, Sekiguchi F, Ishikura H, Kawabata A. Involvement of the cystathionine-γ-lyase/Ca v3.2 pathway in substance P-induced bladder pain in the mouse, a model for nonulcerative bladder pain syndrome. Neuropharmacology 2018; 133:254-263. [PMID: 29407215 DOI: 10.1016/j.neuropharm.2018.01.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 11/29/2017] [Accepted: 01/25/2018] [Indexed: 11/28/2022]
Abstract
Hydrogen sulfide (H2S) formed by cystathionine-γ-lyase (CSE) enhances the activity of Cav3.2 T-type Ca2+ channels, contributing to the bladder pain accompanying hemorrhagic cystitis caused by systemic administration of cyclophosphamide (CPA) in mice. Given clinical and fundamental evidence for the involvement of the substance P/NK1 receptor systems in bladder pain syndrome (BPS)/interstitial cystitis (IC), we created an intravesical substance P-induced bladder pain model in mice and analyzed the possible involvement of the CSE/Cav3.2 pathway. Bladder pain/cystitis was induced by i.p. CPA or intravesical substance P in female mice. Bladder pain was evaluated by counting nociceptive behavior and by detecting referred hyperalgesia in the lower abdomen and hindpaw. The isolated bladder tissue was weighed to estimate bladder swelling and subjected to histological observation and Western blotting. Intravesical substance P caused profound referred hyperalgesia accompanied by little bladder swelling or edema 6-24 h after the administration, in contrast to i.p. CPA-induced nociceptive behavior/referred hyperalgesia with remarkable bladder swelling/edema and urothelial damage. The bladder pain and/or cystitis symptoms caused by substance P or CPA were prevented by the NK1 receptor antagonist. CSE in the bladder was upregulated by substance P or CPA, and the NK1 antagonist prevented the CPA-induced CSE upregulation. A CSE inhibitor, a T-type Ca2+ channel blocker and gene silencing of Cav3.2 abolished the intravesical substance P-induced referred hyperalgesia. The intravesical substance P-induced pain in mice is useful as a model for nonulcerative BPS, and involves the activation of the NK1 receptor/CSE/H2S/Cav3.2 cascade.
Collapse
Affiliation(s)
- Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Yasumasa Okawa
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Yuhei Irie
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan; Division of Emergency and Critical Care Medicine, Fukuoka University, Hospital, Fukuoka 814-0180, Japan
| | - Mariko Maeda
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Tomoka Ozaki
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Hiroyasu Ishikura
- Division of Emergency and Critical Care Medicine, Fukuoka University, Hospital, Fukuoka 814-0180, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan.
| |
Collapse
|
27
|
Ujike A, Kuraishi T, Yamaguchi S, Eguchi R, Kitano T, Kamise J, Ito S, Otsuguro KI. IL-1β augments H 2S-induced increase in intracellular Ca 2+ through polysulfides generated from H 2S/NO interaction. Eur J Pharmacol 2018; 821:88-96. [PMID: 29337193 DOI: 10.1016/j.ejphar.2018.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/07/2017] [Accepted: 01/10/2018] [Indexed: 10/18/2022]
Abstract
H2S has excitatory and inhibitory effects on Ca2+ signals via transient receptor potential ankyrin 1 (TRPA1) and ATP-sensitive K+ channels, respectively. H2S converts intracellularly to polysulfides, which are more potent agonists for TRPA1 than H2S. Under inflammatory conditions, changes in the expression and activity of these H2S target channels and/or the conversion of H2S to polysulfides may modulate H2S effects. Effects of proinflammatory cytokines on H2S-induced Ca2+ signals and polysulfide production in RIN14B cells were examined using fluorescence imaging with fura-2 and SSP4, respectively. Na2S, a H2S donor, induced 1) the inhibition of spontaneous Ca2+ signals, 2) inhibition followed by [Ca2+]i increase, and 3) rapid [Ca2+]i increase without inhibition in 50% (23/46), 22% (10/46), and 17% (8/46) of cells tested, respectively. IL-1β augmented H2S-induced [Ca2+]i increases, which were inhibited by TRPA1 and voltage-dependent L-type Ca2+ channel blockers. However, IL-1β treatment did not affect [Ca2+]i increases evoked by a TRPA1 agonist or high concentration of KCl. Na2S increased intracellular polysulfide levels, which were enhanced by IL-1β treatment. A NOS inhibitor suppressed the increased polysulfide production and [Ca2+]i increase in IL-1β-treated cells. These results suggest that IL-1β augments H2S-induced [Ca2+]i increases via the conversion of H2S to polysulfides through NO synthesis, but not via changes in the activity and expression of target channels. Polysulfides may play an important role in the effects of H2S during inflammation.
Collapse
Affiliation(s)
- Ayako Ujike
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Tomoki Kuraishi
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Soichiro Yamaguchi
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Ryota Eguchi
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Taisuke Kitano
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Jumpei Kamise
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Shigeo Ito
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Ken-Ichi Otsuguro
- Laboratory of Pharmacology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.
| |
Collapse
|
28
|
Shan H, Chu Y, Chang P, Yang L, Wang Y, Zhu S, Zhang M, Tao L. Neuroprotective effects of hydrogen sulfide on sodium azide‑induced autophagic cell death in PC12 cells. Mol Med Rep 2017; 16:5938-5946. [PMID: 28849152 PMCID: PMC5865772 DOI: 10.3892/mmr.2017.7363] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Sodium azide (NaN3) is a chemical of rapidly growing commercial importance. It is very acutely toxic and inhibits cytochrome oxidase (COX) by binding irreversibly to the heme cofactor. A previous study from our group demonstrated that hydrogen sulfide (H2S), the third endogenous gaseous mediator identified, had protective effects against neuronal damage induced by traumatic brain injury (TBI). It is well‑known that TBI can reduce the activity of COX and have detrimental effects on the central nervous system metabolism. Therefore, in the present study, it was hypothesized that H2S may provide neuroprotection against NaN3 toxicity. The current results revealed that NaN3 treatment induced non‑apoptotic cell death, namely autophagic cell death, in PC12 cells. Expression of the endogenous H2S‑producing enzymes, cystathionine‑β‑synthase and 3‑mercaptopyruvate sulfurtransferase, decreased in a dose‑dependent manner following NaN3 treatment. Pretreatment with H2S markedly attenuated the NaN3‑induced cell viability loss and autophagic cell death in a dose‑dependent manner. The present study suggests that H2S‑based strategies may have future potential in the prevention and/or therapy of neuronal damage following NaN3 exposure.
Collapse
Affiliation(s)
- Haiyan Shan
- Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Science, Ministry of Justice, Shanghai 200063, P.R. China
| | - Yang Chu
- Institute of Forensic Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Pan Chang
- Central Laboratory, Second Affiliated Hospital of Xi'an Medical College, Xi'an, Shaanxi 710038, P.R. China
| | - Lijun Yang
- Institute of Forensic Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Yi Wang
- Institute of Forensic Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Shaohua Zhu
- Institute of Forensic Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Mingyang Zhang
- Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Science, Ministry of Justice, Shanghai 200063, P.R. China
| | - Luyang Tao
- Institute of Forensic Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
29
|
Hartle MD, Tillotson MR, Prell JS, Pluth MD. Spectroscopic investigation of the reaction of metallo-protoporphyrins with hydrogen sulfide. J Inorg Biochem 2017; 173:152-157. [PMID: 28551529 DOI: 10.1016/j.jinorgbio.2017.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/10/2017] [Accepted: 04/23/2017] [Indexed: 11/16/2022]
Abstract
Hydrogen sulfide (H2S) is the most recently discovered gasotransmitter molecule joining nitric oxide and carbon monoxide. In addition to being biologically important gases, these gasotransmitters also provide distinct modes of reactivity with biomimetic metal complexes. The majority of previous investigations on the reactivity of H2S with bioinorganic models have focused on Fe-based porphyrin systems, whereas investigations with other metals remains underinvestigated. To address this gap, we report here an examination of the reactions of H2S, HS-, and S8 with MgII, CuII, CoII, ZnII, CrII, SnIV, and MnII/III protoporphyrins.
Collapse
Affiliation(s)
- Matthew D Hartle
- Department of Chemistry & Biochemistry, Materials Science Institute, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403-1253, USA
| | - McKinna R Tillotson
- Department of Chemistry & Biochemistry, Materials Science Institute, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403-1253, USA
| | - James S Prell
- Department of Chemistry & Biochemistry, Materials Science Institute, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403-1253, USA
| | - Michael D Pluth
- Department of Chemistry & Biochemistry, Materials Science Institute, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403-1253, USA.
| |
Collapse
|
30
|
Possible Involvement of Hydrosulfide in B 12-Dependent Methyl Group Transfer. Molecules 2017; 22:molecules22040582. [PMID: 28379205 PMCID: PMC6154648 DOI: 10.3390/molecules22040582] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/22/2017] [Accepted: 03/30/2017] [Indexed: 11/29/2022] Open
Abstract
Evidence from several fields of investigation lead to the hypothesis that the sulfur atom is involved in vitamin B12-dependent methyl group transfer. To compile the evidence, it is necessary to briefly review the following fields: methylation, the new field of sulfane sulfur/hydrogen sulfide (S°/H2S), hydrosulfide derivatives of cobalamins, autoxidation of hydrosulfide radical, radical S-adenosylmethionine methyl transfer (RSMT), and methionine synthase (MS). Then, new reaction mechanisms for B12-dependent methyl group transfer are proposed; the mechanisms are facile and overcome difficulties that existed in previously-accepted mechanisms. Finally, the theory is applied to the effect of S°/H2S in nerve tissue involving the “hypomethylation theory” that was proposed 50 years ago to explain the neuropathology resulting from deficiency of vitamin B12 or folic acid. The conclusions are consistent with emerging evidence that sulfane sulfur/hydrogen sulfide may be beneficial in treating Alzheimer’s disease.
Collapse
|
31
|
Gonzalez-Carter DA, Leo BF, Ruenraroengsak P, Chen S, Goode AE, Theodorou IG, Chung KF, Carzaniga R, Shaffer MSP, Dexter DT, Ryan MP, Porter AE. Silver nanoparticles reduce brain inflammation and related neurotoxicity through induction of H 2S-synthesizing enzymes. Sci Rep 2017; 7:42871. [PMID: 28251989 PMCID: PMC5333087 DOI: 10.1038/srep42871] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 01/12/2017] [Indexed: 02/07/2023] Open
Abstract
Silver nanoparticles (AgNP) are known to penetrate into the brain and cause neuronal death. However, there is a paucity in studies examining the effect of AgNP on the resident immune cells of the brain, microglia. Given microglia are implicated in neurodegenerative disorders such as Parkinson's disease (PD), it is important to examine how AgNPs affect microglial inflammation to fully assess AgNP neurotoxicity. In addition, understanding AgNP processing by microglia will allow better prediction of their long term bioreactivity. In the present study, the in vitro uptake and intracellular transformation of citrate-capped AgNPs by microglia, as well as their effects on microglial inflammation and related neurotoxicity were examined. Analytical microscopy demonstrated internalization and dissolution of AgNPs within microglia and formation of non-reactive silver sulphide (Ag2S) on the surface of AgNPs. Furthermore, AgNP-treatment up-regulated microglial expression of the hydrogen sulphide (H2S)-synthesizing enzyme cystathionine-γ-lyase (CSE). In addition, AgNPs showed significant anti-inflammatory effects, reducing lipopolysaccharide (LPS)-stimulated ROS, nitric oxide and TNFα production, which translated into reduced microglial toxicity towards dopaminergic neurons. Hence, the present results indicate that intracellular Ag2S formation, resulting from CSE-mediated H2S production in microglia, sequesters Ag+ ions released from AgNPs, significantly limiting their toxicity, concomitantly reducing microglial inflammation and related neurotoxicity.
Collapse
Affiliation(s)
- Daniel A. Gonzalez-Carter
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
- Parkinson’s Disease Research Unit, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Bey Fen Leo
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
- Central Unit for Advanced Research Imaging, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Pakatip Ruenraroengsak
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, UK
| | - Shu Chen
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Angela E. Goode
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Ioannis G. Theodorou
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, UK
| | - Raffaella Carzaniga
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, Lincoln’s Inn Fields Laboratory, 44 Lincoln’s Inn Fields, London, WC2A 3LY, UK
| | - Milo S. P. Shaffer
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
- Department of Chemistry and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - David T. Dexter
- Parkinson’s Disease Research Unit, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Mary P. Ryan
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Alexandra E. Porter
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| |
Collapse
|
32
|
Olson KR, Gao Y, DeLeon ER, Arif M, Arif F, Arora N, Straub KD. Catalase as a sulfide-sulfur oxido-reductase: An ancient (and modern?) regulator of reactive sulfur species (RSS). Redox Biol 2017; 12:325-339. [PMID: 28285261 PMCID: PMC5350573 DOI: 10.1016/j.redox.2017.02.021] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/11/2017] [Accepted: 02/24/2017] [Indexed: 11/19/2022] Open
Abstract
Catalase is well-known as an antioxidant dismutating H2O2 to O2 and H2O. However, catalases evolved when metabolism was largely sulfur-based, long before O2 and reactive oxygen species (ROS) became abundant, suggesting catalase metabolizes reactive sulfide species (RSS). Here we examine catalase metabolism of H2Sn, the sulfur analog of H2O2, hydrogen sulfide (H2S) and other sulfur-bearing molecules using H2S-specific amperometric electrodes and fluorophores to measure polysulfides (H2Sn; SSP4) and ROS (dichlorofluorescein, DCF). Catalase eliminated H2Sn, but did not anaerobically generate H2S, the expected product of dismutation. Instead, catalase concentration- and oxygen-dependently metabolized H2S and in so doing acted as a sulfide oxidase with a P50 of 20mmHg. H2O2 had little effect on catalase-mediated H2S metabolism but in the presence of the catalase inhibitor, sodium azide (Az), H2O2 rapidly and efficiently expedited H2S metabolism in both normoxia and hypoxia suggesting H2O2 is an effective electron acceptor in this reaction. Unexpectedly, catalase concentration-dependently generated H2S from dithiothreitol (DTT) in both normoxia and hypoxia, concomitantly oxidizing H2S in the presence of O2. H2S production from DTT was inhibited by carbon monoxide and augmented by NADPH suggesting that catalase heme-iron is the catalytic site and that NADPH provides reducing equivalents. Catalase also generated H2S from garlic oil, diallyltrisulfide, thioredoxin and sulfur dioxide, but not from sulfite, metabisulfite, carbonyl sulfide, cysteine, cystine, glutathione or oxidized glutathione. Oxidase activity was also present in catalase from Aspergillus niger. These results show that catalase can act as either a sulfide oxidase or sulfur reductase and they suggest that these activities likely played a prominent role in sulfur metabolism during evolution and may continue do so in modern cells as well. This also appears to be the first observation of catalase reductase activity independent of peroxide dismutation.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine - South Bend, South Bend, IN 46617, USA.
| | - Yan Gao
- Indiana University School of Medicine - South Bend, South Bend, IN 46617, USA
| | - Eric R DeLeon
- Indiana University School of Medicine - South Bend, South Bend, IN 46617, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Maaz Arif
- Indiana University School of Medicine - South Bend, South Bend, IN 46617, USA
| | - Faihaan Arif
- Indiana University School of Medicine - South Bend, South Bend, IN 46617, USA
| | - Nitin Arora
- Indiana University School of Medicine - South Bend, South Bend, IN 46617, USA
| | - Karl D Straub
- Central Arkansas Veteran's Healthcare System, Little Rock, AR 72205, USA; Departments of Medicine and Biochemistry, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| |
Collapse
|
33
|
Thirumalaivasan N, Venkatesan P, Wu SP. Highly selective turn-on probe for H2S with imaging applications in vitro and in vivo. NEW J CHEM 2017. [DOI: 10.1039/c7nj02869e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A pyrene-based chemosensor, PyN3, has been developed as a H2S turn-on sensor via reduction of azide to amine.
Collapse
Affiliation(s)
| | - Parthiban Venkatesan
- Department of Applied Chemistry, National Chiao Tung University
- Hsinchu 300
- Taiwan
| | - Shu-Pao Wu
- Department of Applied Chemistry, National Chiao Tung University
- Hsinchu 300
- Taiwan
| |
Collapse
|
34
|
Tomita M, Nagahara N, Ito T. Expression of 3-Mercaptopyruvate Sulfurtransferase in the Mouse. Molecules 2016; 21:molecules21121707. [PMID: 27973427 PMCID: PMC6273466 DOI: 10.3390/molecules21121707] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 01/16/2023] Open
Abstract
3-Mercaptopyruvate sulfurtransferase (MST) is one of the principal enzymes for the production of hydrogen sulfide and polysulfides in mammalians, and emerging evidence supports the physiological significance of MST. As a fundamental study of the physiology and pathobiology of MST, it is necessary to establish the tissue distribution of MST in mice. In the present study, the expression of MST in various organs of adult and fetal mice was analyzed by Western blotting and enzyme-immunohistochemistry. Moreover, the histology of MST gene-deficient mice was examined. Western blotting revealed that all organs examined had MST. The brain, liver, kidneys testes, and endocrine organs contained large amounts of MST, but the lungs, spleen, thymus, and small intestine did not. Immunohistochemically, the MST expression pattern varies in a cell-specific manner. In the brain, neural and glial cells are positively stained; in the lung, bronchiolar cells are preferentially stained; in the liver, hepatocytes around central veins are more strongly stained; renal convoluted cells are strongly stained; and pancreatic islets are strongly stained. Fetal tissues were studied, and MST expression was found to be similar before and after birth. Histological observation revealed no remarkable findings in MST gene-deficient mice. The present study revealed fundamental information regarding the MST expression of various organs in adult and fetal mice, and the morphological phenotype of MST gene-deficient mice.
Collapse
Affiliation(s)
- Masahiro Tomita
- Department of Pathology and Experimental Medicine, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo Chuo-ku, Kumamoto 860-8556, Japan.
| | - Noriyuki Nagahara
- Isotope Research Center, Nippon Medical School; Tokyo 113-8602, Japan.
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo Chuo-ku, Kumamoto 860-8556, Japan.
| |
Collapse
|
35
|
Cupp-Sutton KA, Ashby MT. Biological Chemistry of Hydrogen Selenide. Antioxidants (Basel) 2016; 5:E42. [PMID: 27879667 PMCID: PMC5187540 DOI: 10.3390/antiox5040042] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/04/2016] [Accepted: 11/08/2016] [Indexed: 12/13/2022] Open
Abstract
There are no two main-group elements that exhibit more similar physical and chemical properties than sulfur and selenium. Nonetheless, Nature has deemed both essential for life and has found a way to exploit the subtle unique properties of selenium to include it in biochemistry despite its congener sulfur being 10,000 times more abundant. Selenium is more easily oxidized and it is kinetically more labile, so all selenium compounds could be considered to be "Reactive Selenium Compounds" relative to their sulfur analogues. What is furthermore remarkable is that one of the most reactive forms of selenium, hydrogen selenide (HSe- at physiologic pH), is proposed to be the starting point for the biosynthesis of selenium-containing molecules. This review contrasts the chemical properties of sulfur and selenium and critically assesses the role of hydrogen selenide in biological chemistry.
Collapse
Affiliation(s)
- Kellye A Cupp-Sutton
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA.
| | - Michael T Ashby
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA.
| |
Collapse
|
36
|
An early dysregulation of FAK and MEK/ERK signaling pathways precedes the β-amyloid deposition in the olfactory bulb of APP/PS1 mouse model of Alzheimer's disease. J Proteomics 2016; 148:149-58. [PMID: 27498392 DOI: 10.1016/j.jprot.2016.07.032] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/28/2016] [Accepted: 07/31/2016] [Indexed: 01/25/2023]
Abstract
UNLABELLED Olfactory dysfunction is an early event of Alzheimer's disease (AD). However, the mechanisms associated to AD neurodegeneration in olfactory areas are unknown. Here we used double-transgenic amyloid precursor protein/presenilin 1 (APPswe/PS1dE9) mice and label-free quantitative proteomics to analyze early pathological effects on the olfactory bulb (OB) during AD progression. Prior to β-amyloid plaque formation, 9 modulated proteins were detected on 3-month-old APP/PS1 mice while 16 differential expressed proteins were detected at 6months, when β-amyloid plaques appear, indicating a moderate imbalance in cytoskeletal rearrangement, and synaptic plasticity in APP/PS1 OBs. Moreover, β-amyloid induced an inactivation of focal adhesion kinase (FAK) together with a transient activation of MEK1/2, leading to inactivation of ERK1/2 in 6-months APP/PS1 OBs. In contrast, the analysis of human OBs revealed a late activation of FAK in advanced AD stages, whereas ERK1/2 activation was enhanced across AD staging respect to controls. This survival potential was accompanied by the inhibition of the proapototic factor BAD in the OB across AD phenotypes. Our data contribute to a better understanding of the early molecular mechanisms that are modulated in AD neurodegeneration, highlighting significant differences in the regulation of survival pathways between APP/PS1 mice and sporadic human AD. SIGNIFICANCE Loss of smell is involved in early stages of Alzheimer's disease (AD), usually preceding classic disease symptoms. However, the mechanisms governing this dysfunction are still poorly understood, losing its potential as a useful tool for clinical diagnosis. Our study characterizes potential AD-associated molecular changes in APP/PS1 mice olfactory bulb (OB) using MS-quantitative proteomics, revealing early cytoskeletal disruption and synaptic plasticity impairment. Moreover, an opposite pattern was found when comparing the activation status of specific survival pathways between APP/PS1 OBs and OBs derived from sAD subjects with different neuropathological grading. Our data reflect, in part, the progressive effect of APP overproduction and Aβ accumulation on the OB proteome during AD progression.
Collapse
|
37
|
Decreased Endogenous Hydrogen Sulfide Generation in Penile Tissues of Diabetic Rats With Erectile Dysfunction. J Sex Med 2016; 13:350-60. [DOI: 10.1016/j.jsxm.2016.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/04/2015] [Accepted: 01/06/2016] [Indexed: 11/19/2022]
|
38
|
AP39, a Mitochondria-Targeted Hydrogen Sulfide Donor, Supports Cellular Bioenergetics and Protects against Alzheimer's Disease by Preserving Mitochondrial Function in APP/PS1 Mice and Neurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8360738. [PMID: 27057285 PMCID: PMC4753001 DOI: 10.1155/2016/8360738] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/11/2015] [Accepted: 12/15/2015] [Indexed: 01/11/2023]
Abstract
Increasing evidence suggests that mitochondrial functions are altered in AD and play an important role in AD pathogenesis. It has been established that H2S homeostasis is balanced in AD. The emerging mitochondrial roles of H2S include antioxidation, antiapoptosis, and the modulation of cellular bioenergetics. Here, using primary neurons from the well-characterized APP/PS1 transgenic mouse model, we studied the effects of AP39 (a newly synthesized mitochondrially targeted H2S donor) on mitochondrial function. AP39 increased intracellular H2S levels, mainly in mitochondrial regions. AP39 exerted dose-dependent effects on mitochondrial activity in APP/PS1 neurons, including increased cellular bioenergy metabolism and cell viability at low concentrations (25–100 nM) and decreased energy production and cell viability at a high concentration (250 nM). Furthermore, AP39 (100 nM) increased ATP levels, protected mitochondrial DNA, and decreased ROS generation. AP39 regulated mitochondrial dynamics, shifting from fission toward fusion. After 6 weeks, AP39 administration to APP/PS1 mice significantly ameliorated their spatial memory deficits in the Morris water maze and NORT and reduced Aβ deposition in their brains. Additionally, AP39 inhibited brain atrophy in APP/PS1 mice. Based on these results, AP39 was proposed as a promising drug candidate for AD treatment, and its anti-AD mechanism may involve protection against mitochondrial damage.
Collapse
|
39
|
Neuronal regulation of expression of hydrogen sulfide-producing enzyme cystathionine β-synthase in rat spinal cord astrocytes. Neurosci Res 2015; 97:52-9. [PMID: 25797494 DOI: 10.1016/j.neures.2015.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 12/13/2022]
Abstract
Cystathionine β-synthase (CBS), expressed in astrocytes, generates a gaseous neuromodulator, hydrogen sulfide (H2S) in the central nervous system (CNS). However, little is known about the regulatory mechanisms of astrocytic CBS expression and activity. This study evaluated the influence of neurons on astrocytic CBS expression by employing multiple culture systems. Substantial CBS expression was observed in the intact neonatal rat spinal cord, while CBS content was markedly reduced in an astrocyte-enriched culture prepared from the neonatal spinal cord. Immunofluorescence analysis confirmed the localization of spinal cord CBS in astrocytes, but not in neurons. Although CBS expression was weak in the embryonic rat spinal cord, enzyme levels were time-dependently increased in a neuron/astrocyte mixed culture originating from embryonic spinal cord. The reduced CBS expression in isolated neonatal astrocytes was restored by co-culture with embryonic neurons. Together with the observed CBS expression levels, H2S production was relatively low in astrocytes cultured alone, but was considerably higher in astrocytes cultured with neurons. These results indicate that neurons are essential for maintaining the expression and H2S-producing activity of astrocytic CBS in the rat spinal cord.
Collapse
|
40
|
Coletta C, Módis K, Szczesny B, Brunyánszki A, Oláh G, Rios ECS, Yanagi K, Ahmad A, Papapetropoulos A, Szabo C. Regulation of Vascular Tone, Angiogenesis and Cellular Bioenergetics by the 3-Mercaptopyruvate Sulfurtransferase/H2S Pathway: Functional Impairment by Hyperglycemia and Restoration by DL-α-Lipoic Acid. Mol Med 2015; 21:1-14. [PMID: 25715337 DOI: 10.2119/molmed.2015.00035] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/18/2015] [Indexed: 01/05/2023] Open
Abstract
Hydrogen sulfide (H2S), as a reducing agent and an antioxidant molecule, exerts protective effects against hyperglycemic stress in the vascular endothelium. The mitochondrial enzyme 3-mercaptopyruvate sulfurtransferase (3-MST) is an important biological source of H2S. We have recently demonstrated that 3-MST activity is inhibited by oxidative stress in vitro and speculated that this may have an adverse effect on cellular homeostasis. In the current study, given the importance of H2S as a vasorelaxant, angiogenesis stimulator and cellular bioenergetic mediator, we first determined whether the 3-MST/H2S system plays a physiological regulatory role in endothelial cells. Next, we tested whether a dysfunction of this pathway develops during the development of hyperglycemia and μmol/L to diabetes-associated vascular complications. Intraperitoneal (IP) 3-MP (1 mg/kg) raised plasma H2S levels in rats. 3-MP (10 1 mmol/L) promoted angiogenesis in vitro in bEnd3 microvascular endothelial cells and in vivo in a Matrigel assay in mice (0.3-1 mg/kg). In vitro studies with bEnd3 cell homogenates demonstrated that the 3-MP-induced increases in H2S production depended on enzymatic activity, although at higher concentrations (1-3 mmol/L) there was also evidence for an additional nonenzymatic H2S production by 3-MP. In vivo, 3-MP facilitated wound healing in rats, induced the relaxation of dermal microvessels and increased mitochondrial bioenergetic function. In vitro hyperglycemia or in vivo streptozotocin diabetes impaired angiogenesis, attenuated mitochondrial function and delayed wound healing; all of these responses were associated with an impairment of the proangiogenic and bioenergetic effects of 3-MP. The antioxidants DL-α-lipoic acid (LA) in vivo, or dihydrolipoic acid (DHLA) in vitro restored the ability of 3-MP to stimulate angiogenesis, cellular bioenergetics and wound healing in hyperglycemia and diabetes. We conclude that diabetes leads to an impairment of the 3-MST/H2S pathway, and speculate that this may contribute to the pathogenesis of hyperglycemic endothelial cell dysfunction. We also suggest that therapy with H2S donors, or treatment with the combination of 3-MP and lipoic acid may be beneficial in improving angiogenesis and bioenergetics in hyperglycemia.
Collapse
Affiliation(s)
- Ciro Coletta
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Katalin Módis
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bartosz Szczesny
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Attila Brunyánszki
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Gábor Oláh
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ester C S Rios
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kazunori Yanagi
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Akbar Ahmad
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | | | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
41
|
Yuan S, Patel RP, Kevil CG. Working with nitric oxide and hydrogen sulfide in biological systems. Am J Physiol Lung Cell Mol Physiol 2014; 308:L403-15. [PMID: 25550314 DOI: 10.1152/ajplung.00327.2014] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are gasotransmitter molecules important in numerous physiological and pathological processes. Although these molecules were first known as environmental toxicants, it is now evident that that they are intricately involved in diverse cellular functions with impact on numerous physiological and pathogenic processes. NO and H2S share some common characteristics but also have unique chemical properties that suggest potential complementary interactions between the two in affecting cellular biochemistry and metabolism. Central among these is the interactions between NO, H2S, and thiols that constitute new ways to regulate protein function, signaling, and cellular responses. In this review, we discuss fundamental biochemical principals, molecular functions, measurement methods, and the pathophysiological relevance of NO and H2S.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; and
| | - Rakesh P Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; and
| |
Collapse
|
42
|
Cocaine exposure alters H2S tissue concentrations in peripheral mouse organs. Pharmacol Rep 2014; 67:421-5. [PMID: 25933948 DOI: 10.1016/j.pharep.2014.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/23/2014] [Accepted: 11/07/2014] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) is well-known as a physiological mediator in the mammalian brain and peripheral tissues. Among several factors that change the concentration of H2S, oxidative stress and generation of reactive oxygen species, which accompany neurochemical actions of drugs of abuse, are of recent interest. OBJECTIVE Limited data on the connection of cocaine and H2S levels prompted us to investigate the effect of this psychostimulant on the H2S concentration in the mouse brain and peripheral organs. METHODS Male BALB/C mice were given several cocaine dosage and treatment regimens, and the free and acid-labile H2S tissue concentrations were determined with a modified spectrophotometric method of Siegel. RESULTS We demonstrated the dose- and treatment-dependent decreases in the H2S level in the heart (83% of control level), and in the liver and kidney (17-34% of control levels) homogenates, but no changes were seen in the mouse brain. The strongest effect occurred after repeated administration of cocaine (20mg/kg) in all peripheral tissues. CONCLUSION A reduction in the peripheral tissue H2S level in the heart, liver and kidney homogenates after repeated injections of cocaine may be the result of a strong toxic effect of the drug.
Collapse
|
43
|
Fräsdorf B, Radon C, Leimkühler S. Characterization and interaction studies of two isoforms of the dual localized 3-mercaptopyruvate sulfurtransferase TUM1 from humans. J Biol Chem 2014; 289:34543-56. [PMID: 25336638 DOI: 10.1074/jbc.m114.605733] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human tRNA thiouridine modification protein (TUM1), also designated as 3-mercaptopyruvate sulfurtransferase (MPST), has been implicated in a wide range of physiological processes in the cell. The roles range from an involvement in thiolation of cytosolic tRNAs to the generation of H2S as signaling molecule both in mitochondria and the cytosol. TUM1 is a member of the sulfurtransferase family and catalyzes the conversion of 3-mercaptopyruvate to pyruvate and protein-bound persulfide. Here, we purified and characterized two novel TUM1 splice variants, designated as TUM1-Iso1 and TUM1-Iso2. The purified proteins showed similar kinetic behavior and comparable pH and temperature dependence. Cellular localization studies, however, showed a different localization pattern between the isoforms. TUM1-Iso1 is exclusively localized in the cytosol, whereas TUM1-Iso2 showed a dual localization both in the cytosol and mitochondria. Interaction studies were performed with the isoforms both in vitro using the purified proteins and in vivo by fluorescence analysis in human cells, using the split-EGFP system. The studies showed that TUM1 interacts with the l-cysteine desulfurase NFS1 and the rhodanese-like protein MOCS3, suggesting a dual function of TUM1 both in sulfur transfer for the biosynthesis of the molybdenum cofactor, and for the thiolation of tRNA. Our studies point to distinct roles of each TUM1 isoform in the sulfur transfer processes in the cell, with different compartmentalization of the two splice variants of TUM1.
Collapse
Affiliation(s)
- Benjamin Fräsdorf
- From the University of Potsdam, Institute of Biochemistry and Biology, D-14476 Potsdam, Germany
| | - Christin Radon
- From the University of Potsdam, Institute of Biochemistry and Biology, D-14476 Potsdam, Germany
| | - Silke Leimkühler
- From the University of Potsdam, Institute of Biochemistry and Biology, D-14476 Potsdam, Germany
| |
Collapse
|