Hydrogen Sulfide in Biology

A special issue of Antioxidants (ISSN 2076-3921). This special issue belongs to the section "ROS, RNS and RSS".

Deadline for manuscript submissions: closed (30 June 2021) | Viewed by 54570

Special Issue Editors

Instituto de Tecnologia Quimica e Biologica Antonio Xavier, NOVA University of Lisbon, Oeiras, Portugal
Interests: hydrogen sulfide biochemistry; nitric oxide reductases; gasotransmitters
CNR Institute of Molecular Biology and Pathology, Rome, Italy
Interests: Biochemistry; hydrogen sulfide; Human Hydrogen Sulfide-Synthesizing Enzyme Cystathionine β -Synthase

Special Issue Information

Dear Colleagues,

Hydrogen sulfide (H2S) is a versatile player in biology, acting as an antioxidant or prooxidant, an energy metabolite, a signaling molecule, or simply a poisonous toxicant, depending on the biological system and the physiological context. The chemical versatility of H2S has been exploited along evolution in increasingly complex biological processes since primordial times. H2S is an intermediate or end-product of various prokaryotic metabolic pathways. Moreover, it acts as a source of reducing power in prokaryotic and mitochondrial electron transport chains. As a signaling molecule, H2S exerts its regulatory function via chemical modification (persulfidation and polysulfidation) of cysteine residues or binding to metal centers in protein targets. The recognition over the past three decades of H2S as a fundamental second messenger in mammalian physiology has raised a great deal of attention to the association between dysregulation of H2S homeostasis and human pathologies, from neurological and cardiovascular diseases to different types of cancer.

In this Special Issue, we welcome contributions covering different aspects of H2S biochemistry and physiology, underlining how H2S evolved as an inorganic building block of life to an energy metabolite in prokaryotic and eukaryotic cells up to a multifaceted signaling molecule in human physiology and pathophysiology.

Dr. João Vicente
Dr. Alessandro Giuffré
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Antioxidants is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Chemical biology of H2S and reactive sulfide species (RSS)
  • H2S and life evolution
  • H2S metabolism in microorganisms and mammals
  • Regulation of H2S biosynthesis and catabolism
  • H2S, microbial physiology and the gut microbiota
  • H2S as a signaling molecule
  • Role of H2S in redox homeostasis and bioenergetics
  • Interplay between gasotransmitters (H2S, NO and CO)
  • Biological roles of persulfides and polysulfides
  • Detection of H2S and RSS in biological samples
  • Impact of H2S and RSS on human health and disease
  • H2S-based pharmacological approaches
  • Sulfur-containing bioactive molecules
  • H2S-related species and nutrition
  • Toxicology of H2S

Published Papers (15 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

16 pages, 3149 KiB  
Article
Sulfane Sulfur Regulates LasR-Mediated Quorum Sensing and Virulence in Pseudomonas aeruginosa PAO1
by Guanhua Xuan, Chuanjuan Lv, Huangwei Xu, Kai Li, Huaiwei Liu, Yongzhen Xia and Luying Xun
Antioxidants 2021, 10(9), 1498; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10091498 - 21 Sep 2021
Cited by 17 | Viewed by 3178
Abstract
Sulfane sulfur, such as inorganic and organic polysulfide (HSn and RSn, n > 2), is a common cellular component, produced either from hydrogen sulfide oxidation or cysteine metabolism. In Pseudomonas aeruginosa PAO1, LasR is a quorum sensing master [...] Read more.
Sulfane sulfur, such as inorganic and organic polysulfide (HSn and RSn, n > 2), is a common cellular component, produced either from hydrogen sulfide oxidation or cysteine metabolism. In Pseudomonas aeruginosa PAO1, LasR is a quorum sensing master regulator. After binding its autoinducer, LasR binds to its target DNA to activate the transcription of a suite of genes, including virulence factors. Herein, we report that the production of hydrogen sulfide and sulfane sulfur were positively correlated in P. aeruginosa PAO1, and sulfane sulfur was able to modify LasR, which generated Cys188 persulfide and trisulfide and produced a pentasulfur link between Cys201 and Cys203. The modifications did not affect LasR binding to its target DNA site, but made it several-fold more effective than unmodified LasR in activating transcription in both in vitro and in vivo assays. On the contrary, H2O2 inactivates LasR via producing a disulfide bond between Cys201 and Cys203. P. aeruginosa PAO1 had a high cellular sulfane sulfur and high LasR activity in the mid log phase and early stationary phase, but a low sulfane sulfur and low LasR activity in the declination phase. Thus, sulfane sulfur is a new signaling factor in the bacterium, adding another level of control over LasR-mediated quorum sensing and turning down the activity in old cells. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

19 pages, 3261 KiB  
Article
Human Cystathionine γ-Lyase Is Inhibited by s-Nitrosation: A New Crosstalk Mechanism between NO and H2S
by Dalila G. F. Fernandes, João Nunes, Catarina S. Tomé, Karim Zuhra, João M. F. Costa, Alexandra M. M. Antunes, Alessandro Giuffrè and João B. Vicente
Antioxidants 2021, 10(9), 1391; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10091391 - 30 Aug 2021
Cited by 6 | Viewed by 2142
Abstract
The ‘gasotransmitters’ hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO) act as second messengers in human physiology, mediating signal transduction via interaction with or chemical modification of protein targets, thereby regulating processes such as neurotransmission, blood flow, immunomodulation, or [...] Read more.
The ‘gasotransmitters’ hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO) act as second messengers in human physiology, mediating signal transduction via interaction with or chemical modification of protein targets, thereby regulating processes such as neurotransmission, blood flow, immunomodulation, or energy metabolism. Due to their broad reactivity and potential toxicity, the biosynthesis and breakdown of H2S, NO, and CO are tightly regulated. Growing evidence highlights the active role of gasotransmitters in their mutual cross-regulation. In human physiology, the transsulfuration enzymes cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE) are prominent H2S enzymatic sources. While CBS is known to be inhibited by NO and CO, little is known about CSE regulation by gasotransmitters. Herein, we investigated the effect of s-nitrosation on CSE catalytic activity. H2S production by recombinant human CSE was found to be inhibited by the physiological nitrosating agent s-nitrosoglutathione (GSNO), while reduced glutathione had no effect. GSNO-induced inhibition was partially reverted by ascorbate and accompanied by the disappearance of one solvent accessible protein thiol. By combining differential derivatization procedures and mass spectrometry-based analysis with functional assays, seven out of the ten protein cysteine residues, namely Cys84, Cys109, Cys137, Cys172, Cys229, Cys307, and Cys310, were identified as targets of s-nitrosation. By generating conservative Cys-to-Ser variants of the identified s-nitrosated cysteines, Cys137 was identified as most significantly contributing to the GSNO-mediated CSE inhibition. These results highlight a new mechanism of crosstalk between gasotransmitters. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

32 pages, 5582 KiB  
Article
Mycobacterium tuberculosis H2S Functions as a Sink to Modulate Central Metabolism, Bioenergetics, and Drug Susceptibility
by Tafara T. R. Kunota, Md. Aejazur Rahman, Barry E. Truebody, Jared S. Mackenzie, Vikram Saini, Dirk A. Lamprecht, John H. Adamson, Ritesh R. Sevalkar, Jack R. Lancaster, Jr., Michael Berney, Joel N. Glasgow and Adrie J. C. Steyn
Antioxidants 2021, 10(8), 1285; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10081285 - 13 Aug 2021
Cited by 8 | Viewed by 3578
Abstract
H2S is a potent gasotransmitter in eukaryotes and bacteria. Host-derived H2S has been shown to profoundly alter M. tuberculosis (Mtb) energy metabolism and growth. However, compelling evidence for endogenous production of H2S and its role [...] Read more.
H2S is a potent gasotransmitter in eukaryotes and bacteria. Host-derived H2S has been shown to profoundly alter M. tuberculosis (Mtb) energy metabolism and growth. However, compelling evidence for endogenous production of H2S and its role in Mtb physiology is lacking. We show that multidrug-resistant and drug-susceptible clinical Mtb strains produce H2S, whereas H2S production in non-pathogenic M. smegmatis is barely detectable. We identified Rv3684 (Cds1) as an H2S-producing enzyme in Mtb and show that cds1 disruption reduces, but does not eliminate, H2S production, suggesting the involvement of multiple genes in H2S production. We identified endogenous H2S to be an effector molecule that maintains bioenergetic homeostasis by stimulating respiration primarily via cytochrome bd. Importantly, H2S plays a key role in central metabolism by modulating the balance between oxidative phosphorylation and glycolysis, and it functions as a sink to recycle sulfur atoms back to cysteine to maintain sulfur homeostasis. Lastly, Mtb-generated H2S regulates redox homeostasis and susceptibility to anti-TB drugs clofazimine and rifampicin. These findings reveal previously unknown facets of Mtb physiology and have implications for routine laboratory culturing, understanding drug susceptibility, and improved diagnostics. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

22 pages, 3851 KiB  
Article
Characterization of the Inducible and Slow-Releasing Hydrogen Sulfide and Persulfide Donor P*: Insights into Hydrogen Sulfide Signaling
by Modesta Trummer, Erwan Galardon, Anita Fischer, Stefan Toegel, Bernd Mayer, Guenter Steiner and Burkhard Kloesch
Antioxidants 2021, 10(7), 1049; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10071049 - 29 Jun 2021
Cited by 7 | Viewed by 3216
Abstract
Hydrogen sulfide (H2S) is an important mediator of inflammatory processes. However, controversial findings also exist, and its underlying molecular mechanisms are largely unknown. Recently, the byproducts of H2S, per-/polysulfides, emerged as biological mediators themselves, highlighting the complex chemistry of [...] Read more.
Hydrogen sulfide (H2S) is an important mediator of inflammatory processes. However, controversial findings also exist, and its underlying molecular mechanisms are largely unknown. Recently, the byproducts of H2S, per-/polysulfides, emerged as biological mediators themselves, highlighting the complex chemistry of H2S. In this study, we characterized the biological effects of P*, a slow-releasing H2S and persulfide donor. To differentiate between H2S and polysulfide-derived effects, we decomposed P* into polysulfides. P* was further compared to the commonly used fast-releasing H2S donor sodium hydrogen sulfide (NaHS). The effects on oxidative stress and interleukin-6 (IL-6) expression were assessed in ATDC5 cells using superoxide measurement, qPCR, ELISA, and Western blotting. The findings on IL-6 expression were corroborated in primary chondrocytes from osteoarthritis patients. In ATDC5 cells, P* not only induced the expression of the antioxidant enzyme heme oxygenase-1 via per-/polysulfides, but also induced activation of Akt and p38 MAPK. NaHS and P* significantly impaired menadione-induced superoxide production. P* reduced IL-6 levels in both ATDC5 cells and primary chondrocytes dependent on H2S release. Taken together, P* provides a valuable research tool for the investigation of H2S and per-/polysulfide signaling. These data demonstrate the importance of not only H2S, but also per-/polysulfides as bioactive signaling molecules with potent anti-inflammatory and, in particular, antioxidant properties. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

13 pages, 2010 KiB  
Article
The Mechanisms of Thiosulfate Toxicity against Saccharomyces cerevisiae
by Zhigang Chen, Yongzhen Xia, Huaiwei Liu, Honglei Liu and Luying Xun
Antioxidants 2021, 10(5), 646; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10050646 - 22 Apr 2021
Cited by 12 | Viewed by 2935
Abstract
Elemental sulfur and sulfite have been used to inhibit the growth of yeasts, but thiosulfate has not been reported to be toxic to yeasts. We observed that thiosulfate was more inhibitory than sulfite to Saccharomyces cerevisiae growing in a common yeast medium. At [...] Read more.
Elemental sulfur and sulfite have been used to inhibit the growth of yeasts, but thiosulfate has not been reported to be toxic to yeasts. We observed that thiosulfate was more inhibitory than sulfite to Saccharomyces cerevisiae growing in a common yeast medium. At pH < 4, thiosulfate was a source of elemental sulfur and sulfurous acid, and both were highly toxic to the yeast. At pH 6, thiosulfate directly inhibited the electron transport chain in yeast mitochondria, leading to reductions in oxygen consumption, mitochondrial membrane potential and cellular ATP. Although thiosulfate was converted to sulfite and H2S by the mitochondrial rhodanese Rdl1, its toxicity was not due to H2S as the rdl1-deletion mutant that produced significantly less H2S was more sensitive to thiosulfate than the wild type. Evidence suggests that thiosulfate inhibits cytochrome c oxidase of the electron transport chain in yeast mitochondria. Thus, thiosulfate is a potential agent against yeasts. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Graphical abstract

16 pages, 2754 KiB  
Article
Selenium-Binding Protein 1 (SELENBP1) Supports Hydrogen Sulfide Biosynthesis and Adipogenesis
by Elisa B. Randi, Giovanna Casili, Simona Jacquemai and Csaba Szabo
Antioxidants 2021, 10(3), 361; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10030361 - 27 Feb 2021
Cited by 24 | Viewed by 3528
Abstract
Hydrogen sulfide (H2S), a mammalian gasotransmitter, is involved in the regulation of a variety of fundamental processes including intracellular signaling, cellular bioenergetics, cell proliferation, and cell differentiation. Cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS), and 3-mercaptopyruvate sulfurtransferase (3-MST) are currently considered the [...] Read more.
Hydrogen sulfide (H2S), a mammalian gasotransmitter, is involved in the regulation of a variety of fundamental processes including intracellular signaling, cellular bioenergetics, cell proliferation, and cell differentiation. Cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS), and 3-mercaptopyruvate sulfurtransferase (3-MST) are currently considered the three principal mammalian H2S-generating enzymes. However, recently, a fourth H2S-producing enzyme, selenium-binding-protein 1 (SELENBP1), has also been identified. The cellular regulatory role(s) of SELENBP1 are incompletely understood. The current study investigated whether SELENBP1 plays a role in the regulation of adipocyte differentiation in vitro. 3T3-L1 preadipocytes with or without SELENBP1 knock-down were subjected to differentiation-inducing conditions, and H2S production, cellular lipid accumulation, cell proliferation, and mitochondrial activity were quantified. Adipocyte differentiation was associated with an upregulation of H2S biosynthesis. SELENBP1 silencing decreased cellular H2S levels, suppressed the expression of the three “classical” H2S-producing enzymes (CBS, CSE, and 3-MST) and significantly suppressed adipocyte differentiation. Treatment of SELENBP1 knock-down cells with the H2S donor GYY4137 partially restored lipid accumulation, increased cellular H2S levels, and exerted a bell-shaped effect on cellular bioenergetics (enhancement at 1 and 3 mM, and inhibition at 6 mM). We conclude that SELENBP1 in adipocytes (1) contributes to H2S biosynthesis and (2) acts as an endogenous stimulator of adipocyte differentiation. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

13 pages, 1390 KiB  
Article
ΔMST and the Regulation of Cardiac CSE and OTR Expression in Trauma and Hemorrhage
by Britta Trautwein, Tamara Merz, Nicole Denoix, Csaba Szabo, Enrico Calzia, Peter Radermacher and Oscar McCook
Antioxidants 2021, 10(2), 233; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10020233 - 03 Feb 2021
Cited by 6 | Viewed by 2066
Abstract
Genetic deletion of 3-mercaptopyruvate sulfurtransferase (MST) is known to result in hypertension and cardiac hypertrophy in older mice, and is associated with increased anxiety-like behaviors. Endogenous hydrogen sulfide (H2S) produced by MST in the mitochondria is also known to be involved [...] Read more.
Genetic deletion of 3-mercaptopyruvate sulfurtransferase (MST) is known to result in hypertension and cardiac hypertrophy in older mice, and is associated with increased anxiety-like behaviors. Endogenous hydrogen sulfide (H2S) produced by MST in the mitochondria is also known to be involved in physiological and cellular bioenergetics, and its dysfunction associated with depressive behavior and increased cardiovascular morbidity. Interestingly, early life stress has been shown to lead to a significant loss of cystathionine-γ-lyase (CSE) and oxytocin receptor (OTR) expression in the heart. Thus, we were interested in testing the hypothesis of whether genetic MST mutation (ΔMST) would affect cardiac CSE and OTR expression and affect the mitochondrial respiration in a clinically relevant, resuscitated, mouse model of trauma and hemorrhagic shock. In ΔMST mice, we found a reduction of CSE and OTR in both the naive as well as injured state, in contrast to the wild type (wt) controls. Interestingly, the ΔMST showed a different complex IV response to injury than the wt controls, although our claims are based on the non-demonstrated assumption that naive wt and naive ΔMST mice have comparable complex IV activity. Finally, hemorrhagic shock led to a reduction of CSE and OTR, confirming previous results in the injured mouse heart. To date, the exact mechanisms of the cardiac interaction between H2S and OT are not clear, but they point the way to potential cardioprotective therapies. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

12 pages, 2408 KiB  
Article
The Protective Role of Hydrogen Sulfide Against Obesity-Associated Cellular Stress in Blood Glucose Regulation
by Ania Mezouari, Radhika Nangia and Jeffrey Gagnon
Antioxidants 2020, 9(11), 1038; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9111038 - 23 Oct 2020
Cited by 6 | Viewed by 1871
Abstract
Circulating palmitic acid (PA) is increased in obesity and causes metabolic stress, leading to diabetes. This includes the impairment of the glucoregulatory hormone glucagon-like peptide-1 (GLP-1) secreted from intestinal L-cells. Recently, the anti-inflammatory gasotransmitter hydrogen sulfide (H2S) has been implicated in [...] Read more.
Circulating palmitic acid (PA) is increased in obesity and causes metabolic stress, leading to diabetes. This includes the impairment of the glucoregulatory hormone glucagon-like peptide-1 (GLP-1) secreted from intestinal L-cells. Recently, the anti-inflammatory gasotransmitter hydrogen sulfide (H2S) has been implicated in the enhancement of GLP-1 secretion. We hypothesized that H2S can reduce the oxidative stress caused by palmitate and play a protective role in L-cell function. This study was conducted on both human and mouse L-cells and a mouse model of Western diet (WD)-induced obesity. PA-induced L-cell stress was assessed using DCF-DA. H2S was delivered using the donor GYY4137. C57BL/6 mice were fed either chow diet or PA-enriched WD for 20 weeks with ongoing measurements of glycemia and GLP-1 secretion. In both L-cell models, we demonstrated that PA caused an increase in reactive oxygen species (ROS). This ROS induction was partially blocked by the H2S administration. In mice, the WD elevated body weight in both sexes and elevated fasting blood glucose and lipid peroxidation in males. Additionally, a single GYY4137 injection improved oral glucose tolerance in WD-fed male mice and also enhanced glucose-stimulated GLP-1 release. To conclude, H2S reduces oxidative stress in GLP-1 cells and can improve glucose clearance in mice. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

13 pages, 2921 KiB  
Article
A Red Fluorescent Protein-Based Probe for Detection of Intracellular Reactive Sulfane Sulfur
by Zimai Li, Qingda Wang, Yongzhen Xia, Luying Xun and Huaiwei Liu
Antioxidants 2020, 9(10), 985; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9100985 - 13 Oct 2020
Cited by 5 | Viewed by 2352
Abstract
Reactive sulfane sulfur, including persulfide and polysulfide, is a type of regular cellular component, playing an antioxidant role. Its function may be organelle-dependent; however, the shortage of probes for detecting organellar reactive sulfane sulfur has hindered further investigation. Herein, we reported a red [...] Read more.
Reactive sulfane sulfur, including persulfide and polysulfide, is a type of regular cellular component, playing an antioxidant role. Its function may be organelle-dependent; however, the shortage of probes for detecting organellar reactive sulfane sulfur has hindered further investigation. Herein, we reported a red fluorescent protein (mCherry)-based probe for specifically detecting intracellular reactive sulfane sulfur. By mutating two amino acid residues of mCherry (A150 and S151) to cysteine residues, we constructed a mCherry mutant, which reacted with reactive sulfane sulfur to form an intramolecular –Sn– bond (n ≥ 3). The bond largely decreased the intensity of 610 nm emission (excitation at 587 nm) and slightly increased the intensity of 466 nm emission (excitation at 406 nm). The 466/610 nm emission ratio was used to indicate the relative abundance of reactive sulfane sulfur. We then expressed this mutant in the cytoplasm and mitochondria of Saccharomyces cerevisiae. The 466/610 nm emission ratio revealed that mitochondria had a higher level of reactive sulfane sulfur than cytoplasm. Thus, the mCherry mutant can be used as a specific probe for detecting reactive sulfane sulfur in vivo. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Graphical abstract

Review

Jump to: Research, Other

22 pages, 3042 KiB  
Review
Sulfur Administration in Fe–S Cluster Homeostasis
by Leszek Rydz, Maria Wróbel and Halina Jurkowska
Antioxidants 2021, 10(11), 1738; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10111738 - 29 Oct 2021
Cited by 16 | Viewed by 2878
Abstract
Mitochondria are the key organelles of Fe–S cluster synthesis. They contain the enzyme cysteine desulfurase, a scaffold protein, iron and electron donors, and specific chaperons all required for the formation of Fe–S clusters. The newly formed cluster can be utilized by mitochondrial Fe–S [...] Read more.
Mitochondria are the key organelles of Fe–S cluster synthesis. They contain the enzyme cysteine desulfurase, a scaffold protein, iron and electron donors, and specific chaperons all required for the formation of Fe–S clusters. The newly formed cluster can be utilized by mitochondrial Fe–S protein synthesis or undergo further transformation. Mitochondrial Fe–S cluster biogenesis components are required in the cytosolic iron–sulfur cluster assembly machinery for cytosolic and nuclear cluster supplies. Clusters that are the key components of Fe–S proteins are vulnerable and prone to degradation whenever exposed to oxidative stress. However, once degraded, the Fe–S cluster can be resynthesized or repaired. It has been proposed that sulfurtransferases, rhodanese, and 3-mercaptopyruvate sulfurtransferase, responsible for sulfur transfer from donor to nucleophilic acceptor, are involved in the Fe–S cluster formation, maturation, or reconstitution. In the present paper, we attempt to sum up our knowledge on the involvement of sulfurtransferases not only in sulfur administration but also in the Fe–S cluster formation in mammals and yeasts, and on reconstitution-damaged cluster or restoration of enzyme’s attenuated activity. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

41 pages, 4353 KiB  
Review
Biosynthesis, Quantification and Genetic Diseases of the Smallest Signaling Thiol Metabolite: Hydrogen Sulfide
by Joanna Myszkowska, Ilia Derevenkov, Sergei V. Makarov, Ute Spiekerkoetter and Luciana Hannibal
Antioxidants 2021, 10(7), 1065; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10071065 - 01 Jul 2021
Cited by 12 | Viewed by 4286
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter and the smallest signaling thiol metabolite with important roles in human health. The turnover of H2S in humans is mainly governed by enzymes of sulfur amino acid metabolism and also by the microbiome. [...] Read more.
Hydrogen sulfide (H2S) is a gasotransmitter and the smallest signaling thiol metabolite with important roles in human health. The turnover of H2S in humans is mainly governed by enzymes of sulfur amino acid metabolism and also by the microbiome. As is the case with other small signaling molecules, disease-promoting effects of H2S largely depend on its concentration and compartmentalization. Genetic defects that impair the biogenesis and catabolism of H2S have been described; however, a gap in knowledge remains concerning physiological steady-state concentrations of H2S and their direct clinical implications. The small size and considerable reactivity of H2S renders its quantification in biological samples an experimental challenge. A compilation of methods currently employed to quantify H2S in biological specimens is provided in this review. Substantial discrepancy exists in the concentrations of H2S determined by different techniques. Available methodologies permit end-point measurement of H2S concentration, yet no definitive protocol exists for the continuous, real-time measurement of H2S produced by its enzymatic sources. We present a summary of available animal models, monogenic diseases that impair H2S metabolism in humans including structure-function relationships of pathogenic mutations, and discuss possible approaches to overcome current limitations of study. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Graphical abstract

11 pages, 458 KiB  
Review
Anoxygenic Photosynthesis in Photolithotrophic Sulfur Bacteria and Their Role in Detoxication of Hydrogen Sulfide
by Ivan Kushkevych, Veronika Bosáková, Monika Vítězová and Simon K.-M. R. Rittmann
Antioxidants 2021, 10(6), 829; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10060829 - 22 May 2021
Cited by 6 | Viewed by 4077
Abstract
Hydrogen sulfide is a toxic compound that can affect various groups of water microorganisms. Photolithotrophic sulfur bacteria including Chromatiaceae and Chlorobiaceae are able to convert inorganic substrate (hydrogen sulfide and carbon dioxide) into organic matter deriving energy from photosynthesis. This process takes place [...] Read more.
Hydrogen sulfide is a toxic compound that can affect various groups of water microorganisms. Photolithotrophic sulfur bacteria including Chromatiaceae and Chlorobiaceae are able to convert inorganic substrate (hydrogen sulfide and carbon dioxide) into organic matter deriving energy from photosynthesis. This process takes place in the absence of molecular oxygen and is referred to as anoxygenic photosynthesis, in which exogenous electron donors are needed. These donors may be reduced sulfur compounds such as hydrogen sulfide. This paper deals with the description of this metabolic process, representatives of the above-mentioned families, and discusses the possibility using anoxygenic phototrophic microorganisms for the detoxification of toxic hydrogen sulfide. Moreover, their general characteristics, morphology, metabolism, and taxonomy are described as well as the conditions for isolation and cultivation of these microorganisms will be presented. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

17 pages, 722 KiB  
Review
Hydrogen Sulfide and Carbon Monoxide Tolerance in Bacteria
by Sofia S. Mendes, Vanessa Miranda and Lígia M. Saraiva
Antioxidants 2021, 10(5), 729; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10050729 - 05 May 2021
Cited by 22 | Viewed by 3683
Abstract
Hydrogen sulfide and carbon monoxide share the ability to be beneficial or harmful molecules depending on the concentrations to which organisms are exposed. Interestingly, humans and some bacteria produce small amounts of these compounds. Since several publications have summarized the recent knowledge of [...] Read more.
Hydrogen sulfide and carbon monoxide share the ability to be beneficial or harmful molecules depending on the concentrations to which organisms are exposed. Interestingly, humans and some bacteria produce small amounts of these compounds. Since several publications have summarized the recent knowledge of its effects in humans, here we have chosen to focus on the role of H2S and CO on microbial physiology. We briefly review the current knowledge on how bacteria produce and use H2S and CO. We address their potential antimicrobial properties when used at higher concentrations, and describe how microbial systems detect and survive toxic levels of H2S and CO. Finally, we highlight their antimicrobial properties against human pathogens when endogenously produced by the host and when released by external chemical donors. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

24 pages, 1612 KiB  
Review
Hydrogen Sulfide: From a Toxic Molecule to a Key Molecule of Cell Life
by Angeles Aroca, Cecilia Gotor, Diane C. Bassham and Luis C. Romero
Antioxidants 2020, 9(7), 621; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9070621 - 15 Jul 2020
Cited by 88 | Viewed by 9474
Abstract
Hydrogen sulfide (H2S) has always been considered toxic, but a huge number of articles published more recently showed the beneficial biochemical properties of its endogenous production throughout all regna. In this review, the participation of H2S in many [...] Read more.
Hydrogen sulfide (H2S) has always been considered toxic, but a huge number of articles published more recently showed the beneficial biochemical properties of its endogenous production throughout all regna. In this review, the participation of H2S in many physiological and pathological processes in animals is described, and its importance as a signaling molecule in plant systems is underlined from an evolutionary point of view. H2S quantification methods are summarized and persulfidation is described as the underlying mechanism of action in plants, animals and bacteria. This review aims to highlight the importance of its crosstalk with other signaling molecules and its fine regulation for the proper function of the cell and its survival. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

Other

Jump to: Research, Review

14 pages, 1649 KiB  
Perspective
The Interaction of the Endogenous Hydrogen Sulfide and Oxytocin Systems in Fluid Regulation and the Cardiovascular System
by Nicole Denoix, Oscar McCook, Sarah Ecker, Rui Wang, Christiane Waller, Peter Radermacher and Tamara Merz
Antioxidants 2020, 9(8), 748; https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9080748 - 14 Aug 2020
Cited by 9 | Viewed by 3452
Abstract
The purpose of this review is to explore the parallel roles and interaction of hydrogen sulfide (H2S) and oxytocin (OT) in cardiovascular regulation and fluid homeostasis. Their interaction has been recently reported to be relevant during physical and psychological trauma. However, [...] Read more.
The purpose of this review is to explore the parallel roles and interaction of hydrogen sulfide (H2S) and oxytocin (OT) in cardiovascular regulation and fluid homeostasis. Their interaction has been recently reported to be relevant during physical and psychological trauma. However, literature reports on H2S in physical trauma and OT in psychological trauma are abundant, whereas available information regarding H2S in psychological trauma and OT in physical trauma is much more limited. This review summarizes recent direct and indirect evidence of the interaction of the two systems and their convergence in downstream nitric oxide-dependent signaling pathways during various types of trauma, in an effort to better understand biological correlates of psychosomatic interdependencies. Full article
(This article belongs to the Special Issue Hydrogen Sulfide in Biology)
Show Figures

Figure 1

Back to TopTop