1
|
Kim DS, Firoz W, Santana Maldonado CM, Gauger PC, Weir A, Baumgarth N, Rumbeiha WK. One health: Subchronic exposure to low ambient hydrogen sulfide increases mortality of influenza A virus infection in mice. ENVIRONMENTAL RESEARCH 2024; 266:120536. [PMID: 39638025 DOI: 10.1016/j.envres.2024.120536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
The environment plays an important role in modulating susceptibility and severity of respiratory tract infections. Influenza is a significant zoonotic disease globally. Hydrogen sulfide (H2S), a respiratory tract irritant and toxic gas, is ubiquitous in the environment. The interaction of environmental H2S exposure and influenza is unknown. In this pilot study we tested the hypothesis that subchronic exposure to ambient H2S worsens the outcome of influenza A virus (IAV) infection in mice. Male C57BL6 mice were exposed either to room air (RA), or to 5 or 10 ppm H2S for 2 h, 5 days a week for 5 weeks, followed by a single exposure either to phosphate buffered saline (sham) or a sublethal IAV intranasal dose of 10 plaque-forming units and observed for up to 28 days post inoculation (DPI). 10 ppm H2S alone suppressed growth. Mice challenged with IAV following exposure to 5 or 10 ppm H2S were most severely affected and euthanized on DPI 6 to 7 or DPI 4, respectively. In contrast, mice exposed to RA and challenged with IAV only showed minor weight loss. Viral titer in lung homogenates was 11-fold higher in mice pre-exposed to 5 ppm H2S and challenged with IAV compared to the RA-IAV group on DPI 3. BALF concentrations of TNF-α, IL-6, and IL-10 cytokines were significantly higher in mice exposed to H2S and challenged with IAV compared to sham groups. Lung pathology was most severe in mice exposed to H2S and challenged with IAV. Collectively, the study shows that mice subchronically exposed to low levels of H2S overly reacted to a nonlethal dose of IAV, suffering severe lung injury and mortality. This suggests that communities and workers subchronically exposed to ambient H2S concentrations used in this study or higher are at higher risk for developing very severe IAV infections and mortality.
Collapse
Affiliation(s)
- Dong-Suk Kim
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, USA
| | - Wahed Firoz
- Center for Immunology and Infectious Diseases, Department of Pathology, Microbiology and Immunology, UC Davis, USA; Graduate Group in Immunology, UC Davis, USA
| | | | - Phillip C Gauger
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Abigail Weir
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, USA
| | - Nicole Baumgarth
- Center for Immunology and Infectious Diseases, Department of Pathology, Microbiology and Immunology, UC Davis, USA; Graduate Group in Immunology, UC Davis, USA; Lyme and Tickborne Diseases Research and Education Institute, W Harry Feinstone Dept. Molecular Microbiology and Immunology, Bloomberg School of Public Health, and Dept. Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, USA
| | - Wilson K Rumbeiha
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, USA.
| |
Collapse
|
2
|
Crucianelli S, Mariano A, Valeriani F, Cocomello N, Gianfranceschi G, Baseggio Conrado A, Moretti F, Scotto d'Abusco A, Mennuni G, Fraioli A, Del Ben M, Romano Spica V, Fontana M. Effects of sulphur thermal water inhalations in long-COVID syndrome: Spa-centred, double-blinded, randomised case-control pilot study. Clin Med (Lond) 2024; 24:100251. [PMID: 39370044 PMCID: PMC11570715 DOI: 10.1016/j.clinme.2024.100251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/09/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND The long-COVID syndrome is characterised by a plethora of symptoms. Given its social and economic impact, many studies have stressed the urgency of proposing innovative strategies other than hospital settings. In this double-blinded, randomised, case-control trial, we investigate the effects of sulphur thermal water inhalations, rich in H2S, compared to distilled water inhalations on symptoms, inflammatory markers and nasal microbiome in long-COVID patients. METHODS About 30 outpatients aged 18-75 with positive diagnosis for long-COVID were randomised in two groups undergoing 12 consecutive days of inhalations. The active group (STW) received sulphur thermal water inhalations whereas the placebo group received inhalations of sterile distilled non-pyrogenic water (SDW). Each participant was tested prior treatment at day 1 (T0), after the inhalations at day 14 (T1) and at 3 months follow-up (T2). At each time point, blood tests, nasal swabs for microbiome sampling, pulmonary functionality tests (PFTs) and pro-inflammatory marker measure were performed. RESULTS The scores obtained in the administered tests (6MWT, Borg score and SGRQ) at T0 showed a significant variation in the STW group, at T1 and T2. Serum cytokine levels and other inflammatory biomarkers reported a statistically significant decrease. Some specific parameters of PFTs showed ameliorations in the STW group only. Changes in the STW nasopharyngeal microbiota composition were noticed, especially from T0 to T2. CONCLUSIONS Inhalations of sulphur thermal water exerted objective and subjective improvements on participants affected by long-COVID. Significant reduction of inflammatory markers, dyspnoea scores and quantitative and qualitative changes in the nasopharyngeal microbiome were also assessed.
Collapse
Affiliation(s)
- Serena Crucianelli
- School of Thermal Medicine, Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Alessia Mariano
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Federica Valeriani
- Laboratory of Epidemiology and Biotechnologies, Department of Movement Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, Rome 00135, Italy
| | - Nicholas Cocomello
- School of Thermal Medicine, Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Gianluca Gianfranceschi
- Laboratory of Epidemiology and Biotechnologies, Department of Movement Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, Rome 00135, Italy
| | - Alessia Baseggio Conrado
- Department of Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Ferdinando Moretti
- School of Thermal Medicine, Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Anna Scotto d'Abusco
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Gioacchino Mennuni
- School of Thermal Medicine, Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Antonio Fraioli
- School of Thermal Medicine, Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Maria Del Ben
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Vincenzo Romano Spica
- Laboratory of Epidemiology and Biotechnologies, Department of Movement Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, Rome 00135, Italy
| | - Mario Fontana
- School of Thermal Medicine, Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy; Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy.
| |
Collapse
|
3
|
Perry BW, McGowan KL, Arias-Rodriguez L, Duttke SH, Tobler M, Kelley JL. Nascent transcription reveals regulatory changes in extremophile fishes inhabiting hydrogen sulfide-rich environments. Proc Biol Sci 2024; 291:20240412. [PMID: 38889788 PMCID: PMC11285508 DOI: 10.1098/rspb.2024.0412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/03/2024] [Indexed: 06/20/2024] Open
Abstract
Regulating transcription allows organisms to respond to their environment, both within a single generation (plasticity) and across generations (adaptation). We examined transcriptional differences in gill tissues of fishes in the Poecilia mexicana species complex (family Poeciliidae), which have colonized toxic springs rich in hydrogen sulfide (H2S) in southern Mexico. There are gene expression differences between sulfidic and non-sulfidic populations, yet regulatory mechanisms mediating this gene expression variation remain poorly studied. We combined capped-small RNA sequencing (csRNA-seq), which captures actively transcribed (i.e. nascent) transcripts, and messenger RNA sequencing (mRNA-seq) to examine how variation in transcription, enhancer activity, and associated transcription factor binding sites may facilitate adaptation to extreme environments. csRNA-seq revealed thousands of differentially initiated transcripts between sulfidic and non-sulfidic populations, many of which are involved in H2S detoxification and response. Analyses of transcription factor binding sites in promoter and putative enhancer csRNA-seq peaks identified a suite of transcription factors likely involved in regulating H2S-specific shifts in gene expression, including several key transcription factors known to respond to hypoxia. Our findings uncover a complex interplay of regulatory processes that reflect the divergence of extremophile populations of P. mexicana from their non-sulfidic ancestors and suggest shared responses among evolutionarily independent lineages.
Collapse
Affiliation(s)
- Blair W. Perry
- School of Biological Sciences, Washington State University, Pullman, WA 99164, USA
| | - Kerry L. McGowan
- School of Biological Sciences, Washington State University, Pullman, WA 99164, USA
| | - Lenin Arias-Rodriguez
- División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco 86150, México
| | - Sascha H. Duttke
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Michael Tobler
- Department of Biology, University of Missouri—St Louis, St Louis, MO 63121, USA
- Whitney R. Harris World Ecology Center, University of Missouri—St Louis, St Louis, MO 63121, USA
- WildCare Institute, Saint Louis Zoo, St Louis, MO 63110, USA
| | - Joanna L. Kelley
- Department of Ecology and Evolutionary Biology, University of California Santa Cruz, Santa Cruz, CA 95060, USA
| |
Collapse
|
4
|
Abolfazli S, Ebrahimi N, Morabi E, Asgari Yazdi MA, Zengin G, Sathyapalan T, Jamialahmadi T, Sahebkar A. Hydrogen Sulfide: Physiological Roles and Therapeutic Implications against COVID-19. Curr Med Chem 2024; 31:3132-3148. [PMID: 37138436 DOI: 10.2174/0929867330666230502111227] [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: 09/26/2022] [Revised: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 05/05/2023]
Abstract
The COVID-19 pandemic due to severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) poses a major menace to economic and public health worldwide. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) are two host proteins that play an essential function in the entry of SARS-- COV-2 into host cells. Hydrogen sulfide (H2S), a new gasotransmitter, has been shown to protect the lungs from potential damage through its anti-inflammatory, antioxidant, antiviral, and anti-aging effects. It is well known that H2S is crucial in controlling the inflammatory reaction and the pro-inflammatory cytokine storm. Therefore, it has been suggested that some H2S donors may help treat acute lung inflammation. Furthermore, recent research illuminates a number of mechanisms of action that may explain the antiviral properties of H2S. Some early clinical findings indicate a negative correlation between endogenous H2S concentrations and COVID-19 intensity. Therefore, reusing H2S-releasing drugs could represent a curative option for COVID-19 therapy.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Nima Ebrahimi
- Student Research Committee, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Etekhar Morabi
- Student Research Committee, School of Pharmacy, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | | | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya 42130, Turkey
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom of Great Britain and Northern Ireland
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Jiang S, Chen H, Shen P, Zhou Y, Li Q, Zhang J, Chen Y. Gasotransmitter Research Advances in Respiratory Diseases. Antioxid Redox Signal 2024; 40:168-185. [PMID: 37917094 DOI: 10.1089/ars.2023.0410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Significance: Gasotransmitters are small gas molecules that are endogenously generated and have well-defined physiological functions. The most well-defined gasotransmitters currently are nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), while other potent gasotransmitters include ammonia, methane, cyanide, hydrogen gas, and sulfur dioxide. Gasotransmitters play a role in various respiratory diseases such as asthma, chronic obstructive pulmonary disease, obstructive sleep apnea, lung infection, bronchiectasis, cystic fibrosis, primary ciliary dyskinesia, and COVID-19. Recent Advances: Gasotransmitters can act as biomarkers that facilitate disease diagnosis, indicate disease severity, predict disease exacerbation, and evaluate disease outcomes. They also have cell-protective properties, and many studies have been conducted to explore their pharmacological applications. Innovative drug donors and drug delivery methods have been invented to amplify their therapeutic effects. Critical Issues: In this article, we briefly reviewed the physiological and pathophysiological functions of some gasotransmitters in the respiratory system, the progress in detecting exhaled gasotransmitters, as well as innovative drugs derived from these molecules. Future Directions: The current challenge for gasotransmitter research includes further exploring their physiological and pathological functions, clarifying their complicated interactions, exploring suitable drug donors and delivery devices, and characterizing new members of gasotransmitters. Antioxid. Redox Signal. 40, 168-185.
Collapse
Affiliation(s)
- Simin Jiang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Haijie Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Pu Shen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yumou Zhou
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Qiaoyu Li
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
6
|
Kumar M. Hydrogen sulfide: From a toxic gas to a potential therapy for COVID-19 and inflammatory disorders. Nitric Oxide 2023; 140-141:8-15. [PMID: 37648016 DOI: 10.1016/j.niox.2023.08.002] [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: 05/15/2023] [Revised: 07/06/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
COVID-19 has been shown to induce inflammatory disorders and CNS manifestations. Swift and efficient treatment strategies are urgently warranted for the management of COVID, inflammatory and neurological disorders. Hydrogen sulfide (H2S) has been associated with several clinical disorders due to its potential to influence a broad range of biological signalling pathways. According to recent clinical studies, COVID patients with lower physiological H2S had higher fatality rates. These findings clearly demonstrate an inverse correlation between H2S levels and the severity of COVID-19. H2S has been proposed as a protective molecule because of its antioxidant, anti-inflammatory, and antiviral properties. Various H2S-releasing prodrugs, hybrids and natural compounds have been tested for their therapeutic efficacy in viral infections and inflammatory disorders. In this review, I am highlighting the rationale for using H2S-based interventions for the management of COVID-19 and post-infection inflammatory disorders including neuroinflammation. I am also proposing therepurposing of existing H2S-releasing prodrugs, developing new NO-H2S-hybrids, targeting H2S metabolic pathways, and using H2S-producing dietary supplements as viable defensive strategies against SARS-CoV-2 infection and COVID-19 pathologies.
Collapse
Affiliation(s)
- Mohit Kumar
- Centre for Excellence in Functional Foods, Food and Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Punjab, 140306, India.
| |
Collapse
|
7
|
Oza PP, Kashfi K. The Triple Crown: NO, CO, and H 2S in cancer cell biology. Pharmacol Ther 2023; 249:108502. [PMID: 37517510 PMCID: PMC10529678 DOI: 10.1016/j.pharmthera.2023.108502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) are three endogenously produced gases with important functions in the vasculature, immune defense, and inflammation. It is increasingly apparent that, far from working in isolation, these three exert many effects by modulating each other's activity. Each gas is produced by three enzymes, which have some tissue specificities and can also be non-enzymatically produced by redox reactions of various substrates. Both NO and CO share similar properties, such as activating soluble guanylate cyclase (sGC) to increase cyclic guanosine monophosphate (cGMP) levels. At the same time, H2S both inhibits phosphodiesterase 5A (PDE5A), an enzyme that metabolizes sGC and exerts redox regulation on sGC. The role of NO, CO, and H2S in the setting of cancer has been quite perplexing, as there is evidence for both tumor-promoting and pro-inflammatory effects and anti-tumor and anti-inflammatory activities. Each gasotransmitter has been found to have dual effects on different aspects of cancer biology, including cancer cell proliferation and apoptosis, invasion and metastasis, angiogenesis, and immunomodulation. These seemingly contradictory actions may relate to each gas having a dual effect dependent on its local flux. In this review, we discuss the major roles of NO, CO, and H2S in the context of cancer, with an effort to highlight the dual nature of each gas in different events occurring during cancer progression.
Collapse
Affiliation(s)
- Palak P Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York 10091, USA.
| |
Collapse
|
8
|
Escaffre O, Szaniszlo P, Törő G, Vilas CL, Servantes BJ, Lopez E, Juelich TL, Levine CB, McLellan SLF, Cardenas JC, Freiberg AN, Módis K. Hydrogen Sulfide Ameliorates SARS-CoV-2-Associated Lung Endothelial Barrier Disruption. Biomedicines 2023; 11:1790. [PMID: 37509430 PMCID: PMC10376201 DOI: 10.3390/biomedicines11071790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 07/30/2023] Open
Abstract
Recent studies have confirmed that lung microvascular endothelial injury plays a critical role in the pathophysiology of COVID-19. Our group and others have demonstrated the beneficial effects of H2S in several pathological processes and provided a rationale for considering the therapeutic implications of H2S in COVID-19 therapy. Here, we evaluated the effect of the slow-releasing H2S donor, GYY4137, on the barrier function of a lung endothelial cell monolayer in vitro, after challenging the cells with plasma samples from COVID-19 patients or inactivated SARS-CoV-2 virus. We also assessed how the cytokine/chemokine profile of patients' plasma, endothelial barrier permeability, and disease severity correlated with each other. Alterations in barrier permeability after treatments with patient plasma, inactivated virus, and GYY4137 were monitored and assessed by electrical impedance measurements in real time. We present evidence that GYY4137 treatment reduced endothelial barrier permeability after plasma challenge and completely reversed the endothelial barrier disruption caused by inactivated SARS-CoV-2 virus. We also showed that disease severity correlated with the cytokine/chemokine profile of the plasma but not with barrier permeability changes in our assay. Overall, these data demonstrate that treatment with H2S-releasing compounds has the potential to ameliorate SARS-CoV-2-associated lung endothelial barrier disruption.
Collapse
Affiliation(s)
- Olivier Escaffre
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Peter Szaniszlo
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gabor Törő
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Caitlyn L. Vilas
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Brenna J. Servantes
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ernesto Lopez
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Terry L. Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Corri B. Levine
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Susan L. F. McLellan
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jessica C. Cardenas
- The Center for Translational Injury Research, Department of Surgery, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Katalin Módis
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
9
|
Qu Y, Haas de Mello A, Morris DR, Jones-Hall YL, Ivanciuc T, Sattler RA, Paessler S, Menachery VD, Garofalo RP, Casola A. SARS-CoV-2 Inhibits NRF2-Mediated Antioxidant Responses in Airway Epithelial Cells and in the Lung of a Murine Model of Infection. Microbiol Spectr 2023; 11:e0037823. [PMID: 37022178 PMCID: PMC10269779 DOI: 10.1128/spectrum.00378-23] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
Several viruses have been shown to modulate the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2), the master regulator of redox homeostasis. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for the COVID-19 pandemic, also seems to disrupt the balance between oxidants and antioxidants, which likely contributes to lung damage. Using in vitro and in vivo models of infection, we investigated how SARS-CoV-2 modulates the transcription factor NRF2 and its dependent genes, as well as the role of NRF2 during SARS-CoV-2 infection. We found that SARS-CoV-2 infection downregulates NRF2 protein levels and NRF2-dependent gene expression in human airway epithelial cells and in lungs of BALB/c mice. Reductions in cellular levels of NRF2 seem to be independent of proteasomal degradation and the interferon/promyelocytic leukemia (IFN/PML) pathway. Furthermore, lack of the Nrf2 gene in SARS-CoV-2-infected mice exacerbates clinical disease, increases lung inflammation, and is associated with a trend toward increased lung viral titers, indicating that NRF2 has a protective role during this viral infection. In summary, our results suggest that SARS-CoV-2 infection alters the cellular redox balance by downregulating NRF2 and its dependent genes, which exacerbates lung inflammation and disease, therefore, suggesting that the activation of NRF2 could be explored as therapeutic approach during SARS-CoV-2 infection. IMPORTANCE The antioxidant defense system plays a major function in protecting the organism against oxidative damage caused by free radicals. COVID-19 patients often present with biochemical characteristics of uncontrolled pro-oxidative responses in the respiratory tract. We show herein that SARS-CoV-2 variants, including Omicron, are potent inhibitors of cellular and lung nuclear factor erythroid 2-related factor 2 (NRF2), the master transcription factor that controls the expression of antioxidant and cytoprotective enzymes. Moreover, we show that mice lacking the Nrf2 gene show increased clinical signs of disease and lung pathology when infected with a mouse-adapted strain of SARS-CoV-2. Overall, this study provides a mechanistic explanation for the observed unbalanced pro-oxidative response in SARS-CoV-2 infections and suggests that therapeutic strategies for COVID-19 may consider the use of pharmacologic agents that are known to boost the expression levels of cellular NRF2.
Collapse
Affiliation(s)
- Yue Qu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Aline Haas de Mello
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Dorothea R. Morris
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Yava L. Jones-Hall
- School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Teodora Ivanciuc
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Rachel A. Sattler
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Slobodan Paessler
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Vineet D. Menachery
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Roberto P. Garofalo
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Antonella Casola
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
10
|
Liu H, Sun J, Cheng X, Duan L, Guo S, Zhang Z, Wan J, Wang C, Zhi X, Yuan L, Wang H. Hydrogen sulfide inhibits human T-cell leukemia virus type-1 (HTLV-1) protein expression via regulation of ATG4B. J Med Virol 2023; 95:e28176. [PMID: 36163615 DOI: 10.1002/jmv.28176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide (H2 S) is a redox gasotransmitter. It has been shown that H2 S has a key role in host antiviral defense by inhibiting interleukin production and S-sulfhydrating Keap1 lead to Nrf2/ARE pathway activation. However, it is yet unclear whether H2 S can play an antiviral role by regulating autophagy. In this study, we found that exogenous H2 S decreased the expression of human T-cell leukemia virus type-1 (HTLV-1) protein and HTLV-1 induced autophagosomes accumulation. Transmission electron microscope assays indicated that autophagosomes accumulation decreased after H2 S administration. HTLV-1-transformed T-cell lines had a high level of CSE (H2 S endogenous enzyme) which could be induced in Hela by HTLV-1 infection. Immunoblot demonstrated that overexpression of CSE inhibited HTLV-1 protein expression and autophagy. And we got the opposite after CSE knockdown. Meanwhile, H2 S could not restrain the autophagy when ATG4B had a mutant at its site of 89. In a word, these results suggested that H2 S modulated HTLV-1 protein expression via ATG4B. Therefore, our findings suggested a new mechanism by which H2 S defended against virus infection.
Collapse
Affiliation(s)
- Huandi Liu
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Jiaxiang Sun
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Xuhong Cheng
- Department of Laboratory Medicine, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liangwei Duan
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Shuaifeng Guo
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Zhongxin Zhang
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Jia Wan
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Chunduo Wang
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Xiaoying Zhi
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Linghui Yuan
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical, University, Xinxiang, China
| |
Collapse
|
11
|
Ranjbar T, Oza PP, Kashfi K. The Renin-Angiotensin-Aldosterone System, Nitric Oxide, and Hydrogen Sulfide at the Crossroads of Hypertension and COVID-19: Racial Disparities and Outcomes. Int J Mol Sci 2022; 23:ijms232213895. [PMID: 36430371 PMCID: PMC9699619 DOI: 10.3390/ijms232213895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease 2019 is caused by SARS-CoV-2 and is more severe in the elderly, racial minorities, and those with comorbidities such as hypertension and diabetes. These pathologies are often controlled with medications involving the renin-angiotensin-aldosterone system (RAAS). RAAS is an endocrine system involved in maintaining blood pressure and blood volume through components of the system. SARS-CoV-2 enters the cells through ACE2, a membrane-bound protein related to RAAS. Therefore, the use of RAAS inhibitors could worsen the severity of COVID-19's symptoms, especially amongst those with pre-existing comorbidities. Although a vaccine is currently available to prevent and reduce the symptom severity of COVID-19, other options, such as nitric oxide and hydrogen sulfide, may also have utility to prevent and treat this virus.
Collapse
Affiliation(s)
- Tara Ranjbar
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Palak P. Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
- Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
12
|
Oza PP, Kashfi K. Utility of NO and H 2S donating platforms in managing COVID-19: Rationale and promise. Nitric Oxide 2022; 128:72-102. [PMID: 36029975 PMCID: PMC9398942 DOI: 10.1016/j.niox.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 01/08/2023]
Abstract
Viral infections are a continuing global burden on the human population, underscored by the ramifications of the COVID-19 pandemic. Current treatment options and supportive therapies for many viral infections are relatively limited, indicating a need for alternative therapeutic approaches. Virus-induced damage occurs through direct infection of host cells and inflammation-related changes. Severe cases of certain viral infections, including COVID-19, can lead to a hyperinflammatory response termed cytokine storm, resulting in extensive endothelial damage, thrombosis, respiratory failure, and death. Therapies targeting these complications are crucial in addition to antiviral therapies. Nitric oxide and hydrogen sulfide are two endogenous gasotransmitters that have emerged as key signaling molecules with a broad range of antiviral actions in addition to having anti-inflammatory properties and protective functions in the vasculature and respiratory system. The enhancement of endogenous nitric oxide and hydrogen sulfide levels thus holds promise for managing both early-stage and later-stage viral infections, including SARS-CoV-2. Using SARS-CoV-2 as a model for similar viral infections, here we explore the current evidence regarding nitric oxide and hydrogen sulfide's use to limit viral infection, resolve inflammation, and reduce vascular and pulmonary damage.
Collapse
Affiliation(s)
- Palak P Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, 10091, USA.
| |
Collapse
|
13
|
The Effects of H2S and Recombinant Human Hsp70 on Inflammation Induced by SARS and Other Agents In Vitro and In Vivo. Biomedicines 2022; 10:biomedicines10092155. [PMID: 36140256 PMCID: PMC9496158 DOI: 10.3390/biomedicines10092155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/27/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
The ongoing epidemic caused by SARS-CoV-2 infection led to the search for fundamentally new ways and means to combat inflammation and other pathologies caused by this virus. Using a cellular model of lipopolysaccharide (LPS)-induced sepsis (human promonocytes), we showed that both a hydrogen sulfide donor (sodium thiosulfate, STS) and a recombinant Heat shock protein 70 (rHsp70) effectively block all major inflammatory mediators when administrated before and after LPS challenge. The protective anti-inflammatory effect of rHsp70 and H2S was also confirmed in vivo using various animal models of pneumonia. Specifically, it was found that rHsp70 injections prevented the development of the acute respiratory distress syndrome in highly pathogenic pneumonia in mice, increased animal survival, and reduced the number of Programmed death-1 (PD-1)-positive T-lymphocytes in peripheral blood. Based on our model experiments we developed a combined two-phase therapeutic approach for the treatment of COVID-19 patients. This procedure includes the inhalation of hot helium–oxygen mixtures for induction of endogenous Hsp70 in the first phase and STS inhalation in the second phase. The use of this approach has yielded positive results in COVID-19 patients, reducing the area of lung lesions, restoring parameters of innate immunity and T-cell immune response against coronavirus infection, and preventing the development of pulmonary fibrosis and immune exhaustion syndrome.
Collapse
|
14
|
de Mello AH, Liu T, Garofalo RP, Casola A. Hydrogen Sulfide Donor GYY4137 Rescues NRF2 Activation in Respiratory Syncytial Virus Infection. Antioxidants (Basel) 2022; 11:1410. [PMID: 35883901 PMCID: PMC9311616 DOI: 10.3390/antiox11071410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) can cause severe respiratory illness in infants, immunocompromised, and older adults. Despite its burden, no vaccine or specific treatment is available. RSV infection is associated with increased reactive oxygen species (ROS) production, degradation of the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2), and decreased antioxidant enzymes (AOEs), leading to oxidative damage and lung injury. Hydrogen sulfide (H2S) is an endogenous gaseous molecule that plays a physiological role in numerous cellular processes and a protective role in multiple pathological conditions, displaying vasoactive, cytoprotective, anti-inflammatory, and antioxidant activities. H2S can promote NRF2 activation through the sulfhydration of Kelch-like ECH-associated protein 1, the cytoplasmic repressor of NRF2. Here we investigated whether increasing cellular H2S levels could rescue NRF2 and NRF2-dependent gene expression in RSV-infected primary airway epithelial cells. We found that treatment with the H2S donor GYY4137 significantly increased NRF2 levels and AOEs gene expression by decreasing KEAP1 levels, and by modulating pathways involved in RSV-induced NRF2 degradation, such as NRF2 ubiquitination, and promyelocytic leukemia (PML) protein levels. These results suggest that the administration of exogenous H2S can positively impact the altered redox balance associated with RSV infection, which represents an important determinant of RSV-induced lung disease.
Collapse
Affiliation(s)
- Aline Haas de Mello
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (A.H.d.M.); (T.L.); (R.P.G.)
| | - Tianshuang Liu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (A.H.d.M.); (T.L.); (R.P.G.)
| | - Roberto P. Garofalo
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (A.H.d.M.); (T.L.); (R.P.G.)
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Antonella Casola
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555, USA; (A.H.d.M.); (T.L.); (R.P.G.)
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
15
|
Iciek M, Bilska-Wilkosz A, Kozdrowicki M, Górny M. Reactive Sulfur Compounds in the Fight against COVID-19. Antioxidants (Basel) 2022; 11:antiox11061053. [PMID: 35739949 PMCID: PMC9220020 DOI: 10.3390/antiox11061053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The SARS-CoV-2 coronavirus pandemic outbreak in 2019 resulted in the need to search for an effective and safe strategy for treating infected patients, relieving symptoms, and preventing severe disease. SARS-CoV-2 is an RNA virus that can cause acute respiratory failure and thrombosis, as well as impair circulatory system function. Permanent damage to the heart muscle or other cardiovascular disorders may occur during or after the infection. The severe course of the disease is associated with the release of large amounts of pro-inflammatory cytokines. Due to their documented anti-inflammatory, antioxidant, and antiviral effects, reactive sulfur compounds, including hydrogen sulfide (H2S), lipoic acid (LA), N-acetylcysteine (NAC), glutathione (GSH), and some other lesser-known sulfur compounds, have attracted the interest of scientists for the treatment and prevention of the adverse effects of diseases caused by SARS-CoV-2. This article reviews current knowledge about various endogenous or exogenous reactive sulfur compounds and discusses the possibility, or in some cases the results, of their use in the treatment or prophylaxis of COVID-19.
Collapse
|
16
|
Refractory Mycoplasma pneumoniae Pneumonia in Children: Early Recognition and Management. J Clin Med 2022; 11:jcm11102824. [PMID: 35628949 PMCID: PMC9144103 DOI: 10.3390/jcm11102824] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Refractory Mycoplasma pneumoniae pneumonia (RMPP) is a severe state of M. pneumoniae infection that has attracted increasing universal attention in recent years. The pathogenesis of RMPP remains unknown, but the excessive host immune responses as well as macrolide resistance of M. pneumoniae might play important roles in the development of RMPP. To improve the prognosis of RMPP, it is mandatory to recognize RMPP in the early stages, and the detection of macrolide-resistant MP, clinical unresponsiveness to macrolides and elevated proinflammatory cytokines might be clues. Timely and effective anti-mycoplasmal therapy and immunomodulating therapy are the main strategies for RMPP.
Collapse
|
17
|
Bailly M, Evrard B, Coudeyre E, Rochette C, Meriade L, Blavignac C, Fournier AC, Bignon YJ, Dutheil F, Duclos M, Thivel D. Health management of patients with COVID-19: is there a room for hydrotherapeutic approaches? INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2022; 66:1031-1038. [PMID: 35079866 PMCID: PMC8789204 DOI: 10.1007/s00484-022-02246-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 05/05/2023]
Abstract
With highly variable types of coronavirus disease 2019 (COVID-19) symptoms in both severity and duration, there is today an important need for early, individualized, and multidisciplinary strategies of rehabilitation. Some patients present persistent affections of the respiratory function, digestive system, cardiovascular function, locomotor system, mental health, sleep, nervous system, immune system, taste, smell, metabolism, inflammation, and skin. In this context, we highlight here that hydrothermal centers should be considered today as medically and economically relevant alternatives to face the urgent need for interventions among COVID-19 patients. We raise the potential benefits of hydrotherapy programs already existing which combine alternative medicine with respiratory care, physical activity, nutritional advice, psychological support, and physiotherapy, in relaxing environments and under medical supervision. Beyond the virtues of thermal waters, many studies reported medical benefits of natural mineral waters through compressing, buoyancy, resistance, temperature changes, hydrostatic pressure, inhalations, or drinking. Thermal institutions might offer individualized follow-up helping to unclog hospitals while ensuring the continuity of health care for the different clinical manifestations of COVID-19 in both post-acute and chronic COVID-19 patients. Our present review underlines the need to further explore the medical effectiveness, clinical and territorial feasibility, and medico-economic impacts of the implementation of post-COVID-19 patient management in hydrotherapeutic establishments.
Collapse
Affiliation(s)
- Mélina Bailly
- Université Clermont Auvergne, CRNH, AME2P, F-63000 Clermont-Ferrand, France
| | - Bertrand Evrard
- CHU Clermont-Ferrand, Service d’Immunologie, CHU Gabriel-Montpied, Clermont-Ferrand, France
- Université Clermont Auvergne, INRA, UMR 1019, Clermont-Ferrand, France
| | - Emmanuel Coudeyre
- Service de Médecine Physique Et de Réadaptation, INRAE, UNH, CHU Clermont-Ferrand, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Corinne Rochette
- Université Clermont Auvergne, Health and Terrirory Chair, CleRMa, 63000 Clermont-Ferrand, France
| | - Laurent Meriade
- Université Clermont Auvergne, Health and Terrirory Chair, CleRMa, 63000 Clermont-Ferrand, France
| | - Christelle Blavignac
- Centre Imagerie Cellulaire Santé, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Anne-Cécile Fournier
- Cluster Auvergne-Rhône-Alpes Innovation Innovatherm, 63000 Clermont-Ferrand, France
| | - Yves-Jean Bignon
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand, France
- Département d’Oncogénétique, Centre Jean Perrin, Clermont-Ferrand, France
| | - Frédéric Dutheil
- Preventive and Occupational Medicine, Université Clermont Auvergne, CNRS, LaPSCo, Physiological and Psychosocial Stress, CHU Clermont-Ferrand, University Hospital of Clermont-Ferrand, Witty Fit, 63000 Clermont-Ferrand, France
| | - Martine Duclos
- Service de Médecine du Sport Et Des Explorations Fonctionnelles, CHU de Clermont-Ferrand, Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - David Thivel
- Université Clermont Auvergne, CRNH, AME2P, F-63000 Clermont-Ferrand, France
| |
Collapse
|
18
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
19
|
Tesei D, Jewczynko A, Lynch AM, Urbaniak C. Understanding the Complexities and Changes of the Astronaut Microbiome for Successful Long-Duration Space Missions. Life (Basel) 2022; 12:life12040495. [PMID: 35454986 PMCID: PMC9031868 DOI: 10.3390/life12040495] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
During space missions, astronauts are faced with a variety of challenges that are unique to spaceflight and that have been known to cause physiological changes in humans over a period of time. Several of these changes occur at the microbiome level, a complex ensemble of microbial communities residing in various anatomic sites of the human body, with a pivotal role in regulating the health and behavior of the host. The microbiome is essential for day-to-day physiological activities, and alterations in microbiome composition and function have been linked to various human diseases. For these reasons, understanding the impact of spaceflight and space conditions on the microbiome of astronauts is important to assess significant health risks that can emerge during long-term missions and to develop countermeasures. Here, we review various conditions that are caused by long-term space exploration and discuss the role of the microbiome in promoting or ameliorating these conditions, as well as space-related factors that impact microbiome composition. The topics explored pertain to microgravity, radiation, immunity, bone health, cognitive function, gender differences and pharmacomicrobiomics. Connections are made between the trifecta of spaceflight, the host and the microbiome, and the significance of these interactions for successful long-term space missions.
Collapse
Affiliation(s)
- Donatella Tesei
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria;
| | - Anna Jewczynko
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Anne M. Lynch
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Camilla Urbaniak
- ZIN Technologies Inc., Middleburg Heights, OH 44130, USA
- NASA Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
- Correspondence:
| |
Collapse
|
20
|
Role of Hydrogen Sulfide and Polysulfides in the Regulation of Lipolysis in the Adipose Tissue: Possible Implications for the Pathogenesis of Metabolic Syndrome. Int J Mol Sci 2022; 23:ijms23031346. [PMID: 35163277 PMCID: PMC8836184 DOI: 10.3390/ijms23031346] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Hydrogen sulfide (H2S) and inorganic polysulfides are important signaling molecules; however, little is known about their role in the adipose tissue. We examined the effect of H2S and polysulfides on adipose tissue lipolysis. H2S and polysulfide production by mesenteric adipose tissue explants in rats was measured. The effect of Na2S and Na2S4, the H2S and polysulfide donors, respectively, on lipolysis markers, plasma non-esterified fatty acids (NEFA) and glycerol, was examined. Na2S but not Na2S4 increased plasma NEFA and glycerol in a time- and dose-dependent manner. Na2S increased cyclic AMP but not cyclic GMP concentration in the adipose tissue. The effect of Na2S on NEFA and glycerol was abolished by the specific inhibitor of protein kinase A, KT5720. The effect of Na2S on lipolysis was not abolished by propranolol, suggesting no involvement of β-adrenergic receptors. In addition, Na2S had no effect on phosphodiesterase activity in the adipose tissue. Obesity induced by feeding rats a highly palatable diet for 1 month was associated with increased plasma NEFA and glycerol concentrations, as well as greater H2S production in the adipose tissue. In conclusion, H2S stimulates lipolysis and may contribute to the enhanced lipolysis associated with obesity.
Collapse
|
21
|
Dugbartey GJ, Alornyo KK, Ohene BO, Boima V, Antwi S, Sener A. Renal consequences of the novel coronavirus disease 2019 (COVID-19) and hydrogen sulfide as a potential therapy. Nitric Oxide 2022; 120:16-25. [PMID: 35032641 PMCID: PMC8755416 DOI: 10.1016/j.niox.2022.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
The novel coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, is a global pandemic which is primarily considered a respiratory illness. However, emerging reports show that the virus exhibits both pulmonary and extra-pulmonary manifestations in humans, with the kidney as a major extra-pulmonary target due to its abundant expression of angiotensin-converting enzyme 2 and transmembrane protease serine 2, which facilitate entry of the virus into cells. Acute kidney injury has become prevalent in COVID-19 patients without prior any history of kidney dysfunction. In addition, the virus also worsens kidney conditions and increases mortality of COVID-19 patients with pre-existing chronic kidney disease, renal cancer, diabetic nephropathy, end-stage kidney disease as well as dialysis and kidney transplant patients. In the search for antiviral agents for the treatment of COVID-19, hydrogen sulfide (H2S), the third established member of gasotransmitter family, is emerging as a potential candidate, possessing important therapeutic properties including antiviral, anti-inflammatory, anti-thrombotic and antioxidant properties. A recent clinical study revealed higher serum H2S levels in survivors of COVID-19 pneumonia with reduced interleukin-6 levels compared to fatal cases. In this review, we summarize the global impact of COVID-19 on kidney conditions and discuss the emerging role of H2S as a potential COVID-19 therapy.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana.
| | - Karl K Alornyo
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Bright O Ohene
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Vincent Boima
- Department of Medicine and Therapeutics, University of Ghana Medical School, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Sampson Antwi
- Department of Child Health, School of Medical Sciences, Kwame Nkrumah University of Science and Technology and Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Alp Sener
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, Ontario, Canada; Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Multi-organ Transplant Program, London Health Sciences Center, Ontario, Canada; Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
22
|
Bourgonje AR, Offringa AK, van Eijk LE, Abdulle AE, Hillebrands JL, van der Voort PHJ, van Goor H, van Hezik EJ. N-Acetylcysteine and Hydrogen Sulfide in Coronavirus Disease 2019. Antioxid Redox Signal 2021; 35:1207-1225. [PMID: 33607929 DOI: 10.1089/ars.2020.8247] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Hydrogen sulfide (H2S) is one of the three main gasotransmitters that are endogenously produced in humans and are protective against oxidative stress. Recent findings from studies focusing on coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), shifted our attention to a potentially modulatory role of H2S in this viral respiratory disease. Recent Advances: H2S levels at hospital admission may be of importance since this gasotransmitter has been shown to be protective against lung damage through its antiviral, antioxidant, and anti-inflammatory actions. Furthermore, many COVID-19 cases have been described demonstrating remarkable clinical improvement upon administration of high doses of N-acetylcysteine (NAC). NAC is a renowned pharmacological antioxidant substance acting as a source of cysteine, thereby promoting endogenous glutathione (GSH) biosynthesis as well as generation of sulfane sulfur species when desulfurated to H2S. Critical Issues: Combining H2S physiology and currently available knowledge of COVID-19, H2S is hypothesized to target three main vulnerabilities of SARS-CoV-2: (i) cell entry through interfering with functional host receptors, (ii) viral replication through acting on RNA-dependent RNA polymerase (RdRp), and (iii) the escalation of inflammation to a potentially lethal hyperinflammatory cytokine storm (toll-like receptor 4 [TLR4] pathway and NLR family pyrin domain containing 3 [NLRP3] inflammasome). Future Directions: Dissecting the breakdown of NAC reveals the possibility of increasing endogenous H2S levels, which may provide a convenient rationale for the application of H2S-targeted therapeutics. Further randomized-controlled trials are warranted to investigate its definitive role.
Collapse
Affiliation(s)
- Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Annette K Offringa
- Microbiology and System Biology, Netherlands Organisation for Applied Scientific Research, Zeist, the Netherlands
| | - Larissa E van Eijk
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Amaal E Abdulle
- Division of Vascular Medicine, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter H J van der Voort
- Department of Critical Care Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ed J van Hezik
- Visiting Consultant Chest Physician, formerly Walcheren Hospital, Vlissingen, the Netherlands
| |
Collapse
|
23
|
Ali Qaba MAM, Saleem MK, Ali Qaba NK, Alani MA, Ahmed MM, Sabry SM. Assessment of Inhaled Hydrogen Sulfide in Suppressing Deterioration in Patients With COVID-19. Shock 2021; 56:868-869. [PMID: 34652343 PMCID: PMC8518203 DOI: 10.1097/shk.0000000000001722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 11/25/2022]
Affiliation(s)
| | | | - Nahla Kh Ali Qaba
- Trainning and Development Center Ninawa Health Directorate Mosul, Iraq
| | - Muataz A Alani
- Alkhansaa Teaching Hospital, Ninawa Health Directorate, Mosul, Iraq
| | - Muna Muneer Ahmed
- Department of Family and Community Medicine, College of Medicine, University of Mosul, Mosul, Iraq
| | - Salih M Sabry
- Al-Shifa Hospital, Ninawa Health Directorate, Mosul, Iraq
| |
Collapse
|
24
|
Affiliation(s)
- Thomas Datzmann
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Csaba Szabo
- Chair of Pharmacology, OMI Department, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| |
Collapse
|
25
|
Lisle JT, Lukasik G. Natural inactivation of MS2, poliovirus type 1 and Cryptosporidium parvum in an anaerobic and reduced aquifer. J Appl Microbiol 2021; 132:2464-2474. [PMID: 34724290 DOI: 10.1111/jam.15349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 11/29/2022]
Abstract
AIMS The study of microbial inactivation rates in aquifer systems has most often been determined in aerobic and oxidized systems. This study examined the inactivation (i.e. loss of infectivity) of MS2, poliovirus type 1 (PV1) and Cryptosporidium parvum in an anaerobic and reduced groundwater system that has been identified as storage zones for aquifer storage and recovery (ASR) facilities. METHODS AND RESULTS Anaerobic and reduced (ORP < - 250 mV) groundwater from an artesian well was diverted to an above-ground, flow-through mesocosm that contained diffusion chambers filled with MS2, PV1 or Cryptosporidium parvum. The respective infectivity assays were performed on microorganisms recovered from the diffusion chambers during 30- to 58-day experiments. The net reduction in infectivity was 5.73 log10 over 30 days for MS2, 5.00 log10 over 58 days for PV1 and 4.07 log10 over 37 days for C. parvum. The best fit inactivation model for PV1 was the log-linear model and the Weibull model for MS2 and C. parvum, with respective inactivation rates (95% confidence interval) of 0.19 (0.17-0.21) log10 day-1 , 0.31 (0.19-0.89) log10 day-1 and 0.20 (0.14-0.37) log10 day-1 . CONCLUSIONS The groundwater geochemical conditions in this aquifer enhanced the inactivation of MS2, PV1, and C. parvum at rates approximately 2.0-5.3-fold, 1.2-17.0-fold, and 4.5-5.6-fold greater, respectively, than those from published studies that used diffusion chambers in aerobic-to-anoxic groundwater systems, with positive redox potentials. SIGNIFICANCE AND IMPACT OF THE STUDY Geochemical conditions like those in the aquifer zone in this study can naturally and significantly reduce concentrations of microbial indicators and pathogens of human health concern in injected surface water. Appropriate storage times for injected surface water could complement above-ground engineered processes for microorganism removal and inactivation (e.g. filtration, disinfection) by naturally increasing overall microorganism log-inactivation rates of ASR facilities.
Collapse
Affiliation(s)
- John T Lisle
- St. Petersburg Coastal and Marine Science Center, U.S. Geological Survey, St. Petersburg, Florida, USA
| | | |
Collapse
|
26
|
Pozzi G, Masselli E, Gobbi G, Mirandola P, Taborda-Barata L, Ampollini L, Carbognani P, Micheloni C, Corazza F, Galli D, Carubbi C, Vitale M. Hydrogen Sulfide Inhibits TMPRSS2 in Human Airway Epithelial Cells: Implications for SARS-CoV-2 Infection. Biomedicines 2021; 9:1273. [PMID: 34572459 PMCID: PMC8469712 DOI: 10.3390/biomedicines9091273] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 12/23/2022] Open
Abstract
The COVID-19 pandemic has now affected around 190 million people worldwide, accounting for more than 4 million confirmed deaths. Besides ongoing global vaccination, finding protective and therapeutic strategies is an urgent clinical need. SARS-CoV-2 mostly infects the host organism via the respiratory system, requiring angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) to enter target cells. Therefore, these surface proteins are considered potential druggable targets. Hydrogen sulfide (H2S) is a gasotransmitter produced by several cell types and is also part of natural compounds, such as sulfurous waters that are often inhaled as low-intensity therapy and prevention in different respiratory conditions. H2S is a potent biological mediator, with anti-oxidant, anti-inflammatory, and, as more recently shown, also anti-viral activities. Considering that respiratory epithelial cells can be directly exposed to H2S by inhalation, here we tested the in vitro effects of H2S-donors on TMPRSS2 and ACE2 expression in human upper and lower airway epithelial cells. We showed that H2S significantly reduces the expression of TMPRSS2 without modifying ACE2 expression both in respiratory cell lines and primary human upper and lower airway epithelial cells. Results suggest that inhalational exposure of respiratory epithelial cells to natural H2S sources may hinder SARS-CoV-2 entry into airway epithelial cells and, consequently, potentially prevent the virus from spreading into the lower respiratory tract and the lung.
Collapse
Affiliation(s)
- Giulia Pozzi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Elena Masselli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Giuliana Gobbi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Prisco Mirandola
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Luis Taborda-Barata
- CICS-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal;
| | - Luca Ampollini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Paolo Carbognani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Cristina Micheloni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Francesco Corazza
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Daniela Galli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Cecilia Carubbi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
| | - Marco Vitale
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.P.); (E.M.); (G.G.); (P.M.); (L.A.); (P.C.); (C.M.); (F.C.); (D.G.); (M.V.)
- Italian Foundation for Research in Balneotherapy (FoRST), 00198 Rome, Italy
| |
Collapse
|
27
|
Pacitti D, Scotton CJ, Kumar V, Khan H, Wark PAB, Torregrossa R, Hansbro PM, Whiteman M. Gasping for Sulfide: A Critical Appraisal of Hydrogen Sulfide in Lung Disease and Accelerated Aging. Antioxid Redox Signal 2021; 35:551-579. [PMID: 33736455 DOI: 10.1089/ars.2021.0039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hydrogen sulfide (H2S) is a gaseous signaling molecule involved in a plethora of physiological and pathological processes. It is primarily synthesized by cystathionine-β-synthase, cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase as a metabolite of the transsulfuration pathway. H2S has been shown to exert beneficial roles in lung disease acting as an anti-inflammatory and antiviral and to ameliorate cell metabolism and protect from oxidative stress. H2S interacts with transcription factors, ion channels, and a multitude of proteins via post-translational modifications through S-persulfidation ("sulfhydration"). Perturbation of endogenous H2S synthesis and/or levels have been implicated in the development of accelerated lung aging and diseases, including asthma, chronic obstructive pulmonary disease, and fibrosis. Furthermore, evidence indicates that persulfidation is decreased with aging. Here, we review the use of H2S as a biomarker of lung pathologies and discuss the potential of using H2S-generating molecules and synthesis inhibitors to treat respiratory diseases. Furthermore, we provide a critical appraisal of methods of detection used to quantify H2S concentration in biological samples and discuss the challenges of characterizing physiological and pathological levels. Considerations and caveats of using H2S delivery molecules, the choice of generating molecules, and concentrations are also reviewed. Antioxid. Redox Signal. 35, 551-579.
Collapse
Affiliation(s)
- Dario Pacitti
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Chris J Scotton
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Vinod Kumar
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, Australia
| | - Haroon Khan
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, Australia
| | - Roberta Torregrossa
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, Australia
| | - Matthew Whiteman
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
28
|
Gorini F, Del Turco S, Sabatino L, Gaggini M, Vassalle C. H 2S as a Bridge Linking Inflammation, Oxidative Stress and Endothelial Biology: A Possible Defense in the Fight against SARS-CoV-2 Infection? Biomedicines 2021; 9:biomedicines9091107. [PMID: 34572292 PMCID: PMC8472626 DOI: 10.3390/biomedicines9091107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 12/17/2022] Open
Abstract
The endothelium controls vascular homeostasis through a delicate balance between secretion of vasodilators and vasoconstrictors. The loss of physiological homeostasis leads to endothelial dysfunction, for which inflammatory events represent critical determinants. In this context, therapeutic approaches targeting inflammation-related vascular injury may help for the treatment of cardiovascular disease and a multitude of other conditions related to endothelium dysfunction, including COVID-19. In recent years, within the complexity of the inflammatory scenario related to loss of vessel integrity, hydrogen sulfide (H2S) has aroused great interest due to its importance in different signaling pathways at the endothelial level. In this review, we discuss the effects of H2S, a molecule which has been reported to demonstrate anti-inflammatory activity, in addition to many other biological functions related to endothelium and sulfur-drugs as new possible therapeutic options in diseases involving vascular pathobiology, such as in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Francesca Gorini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (L.S.); (M.G.)
- Correspondence: (F.G.); (S.D.T.); (C.V.)
| | - Serena Del Turco
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (L.S.); (M.G.)
- Correspondence: (F.G.); (S.D.T.); (C.V.)
| | - Laura Sabatino
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (L.S.); (M.G.)
| | - Melania Gaggini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy; (L.S.); (M.G.)
| | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G. Monasterio, 56124 Pisa, Italy
- Correspondence: (F.G.); (S.D.T.); (C.V.)
| |
Collapse
|
29
|
Paul BD, Lemle MD, Komaroff AL, Snyder SH. Redox imbalance links COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome. Proc Natl Acad Sci U S A 2021; 118:e2024358118. [PMID: 34400495 PMCID: PMC8403932 DOI: 10.1073/pnas.2024358118] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Although most patients recover from acute COVID-19, some experience postacute sequelae of severe acute respiratory syndrome coronavirus 2 infection (PASC). One subgroup of PASC is a syndrome called "long COVID-19," reminiscent of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). ME/CFS is a debilitating condition, often triggered by viral and bacterial infections, leading to years-long debilitating symptoms including profound fatigue, postexertional malaise, unrefreshing sleep, cognitive deficits, and orthostatic intolerance. Some are skeptical that either ME/CFS or long COVID-19 involves underlying biological abnormalities. However, in this review, we summarize the evidence that people with acute COVID-19 and with ME/CFS have biological abnormalities including redox imbalance, systemic inflammation and neuroinflammation, an impaired ability to generate adenosine triphosphate, and a general hypometabolic state. These phenomena have not yet been well studied in people with long COVID-19, and each of them has been reported in other diseases as well, particularly neurological diseases. We also examine the bidirectional relationship between redox imbalance, inflammation, energy metabolic deficits, and a hypometabolic state. We speculate as to what may be causing these abnormalities. Thus, understanding the molecular underpinnings of both PASC and ME/CFS may lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205;
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - Anthony L Komaroff
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02120
| | - Solomon H Snyder
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205;
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
30
|
Padmanabhan N, Kyon HK, Boot A, Lim K, Srivastava S, Chen S, Wu Z, Lee HO, Mukundan VT, Chan C, Chan YK, Xuewen O, Pitt JJ, Isa ZFA, Xing M, Lee MH, Tan ALK, Ting SHW, Luftig MA, Kappei D, Kruger WD, Bian J, Ho YS, Teh M, Rozen SG, Tan P. Highly recurrent CBS epimutations in gastric cancer CpG island methylator phenotypes and inflammation. Genome Biol 2021; 22:167. [PMID: 34074348 PMCID: PMC8170989 DOI: 10.1186/s13059-021-02375-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023] Open
Abstract
Background CIMP (CpG island methylator phenotype) is an epigenetic molecular subtype, observed in multiple malignancies and associated with the epigenetic silencing of tumor suppressors. Currently, for most cancers including gastric cancer (GC), mechanisms underlying CIMP remain poorly understood. We sought to discover molecular contributors to CIMP in GC, by performing global DNA methylation, gene expression, and proteomics profiling across 14 gastric cell lines, followed by similar integrative analysis in 50 GC cell lines and 467 primary GCs. Results We identify the cystathionine beta-synthase enzyme (CBS) as a highly recurrent target of epigenetic silencing in CIMP GC. Likewise, we show that CBS epimutations are significantly associated with CIMP in various other cancers, occurring even in premalignant gastroesophageal conditions and longitudinally linked to clinical persistence. Of note, CRISPR deletion of CBS in normal gastric epithelial cells induces widespread DNA methylation changes that overlap with primary GC CIMP patterns. Reflecting its metabolic role as a gatekeeper interlinking the methionine and homocysteine cycles, CBS loss in vitro also causes reductions in the anti-inflammatory gasotransmitter hydrogen sulfide (H2S), with concomitant increase in NF-κB activity. In a murine genetic model of CBS deficiency, preliminary data indicate upregulated immune-mediated transcriptional signatures in the stomach. Conclusions Our results implicate CBS as a bi-faceted modifier of aberrant DNA methylation and inflammation in GC and highlights H2S donors as a potential new therapy for CBS-silenced lesions. Supplementary Information The online version contains supplementary material available at 10.1186/s13059-021-02375-2.
Collapse
Affiliation(s)
- Nisha Padmanabhan
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Huang Kie Kyon
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Arnoud Boot
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Kevin Lim
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Supriya Srivastava
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Shuwen Chen
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Zhiyuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Hyung-Ok Lee
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Vineeth T Mukundan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Charlene Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Yarn Kit Chan
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Ong Xuewen
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Jason J Pitt
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Zul Fazreen Adam Isa
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Manjie Xing
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Ming Hui Lee
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Angie Lay Keng Tan
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Shamaine Ho Wei Ting
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore
| | - Micah A Luftig
- Department of Molecular Genetics and Microbiology, Duke Centre for Virology, Duke University School of Medicine, Durham, NC, USA
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Warren D Kruger
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jinsong Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.,National University of Singapore (Suzhou) Research Institute, Suzhou, 215123, China
| | - Ying Swan Ho
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Ming Teh
- Department of Pathology, National University of Singapore, Singapore, 119228, Singapore
| | - Steve George Rozen
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Patrick Tan
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8, College road, Singapore, 169857, Singapore. .,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore. .,Genome Institute of Singapore, Singapore, 138672, Singapore. .,SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, 169856, Singapore. .,Singapore Gastric Cancer Consortium, Singapore, 119074, Singapore. .,Department of Physiology, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
31
|
Sueiro-Olivares M, Scott J, Gago S, Petrovic D, Kouroussis E, Zivanovic J, Yu Y, Strobel M, Cunha C, Thomson D, Fortune-Grant R, Thusek S, Bowyer P, Beilhack A, Carvalho A, Bignell E, Filipovic MR, Amich J. Fungal and host protein persulfidation are functionally correlated and modulate both virulence and antifungal response. PLoS Biol 2021; 19:e3001247. [PMID: 34061822 PMCID: PMC8168846 DOI: 10.1371/journal.pbio.3001247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Aspergillus fumigatus is a human fungal pathogen that can cause devastating pulmonary infections, termed "aspergilloses," in individuals suffering immune imbalances or underlying lung conditions. As rapid adaptation to stress is crucial for the outcome of the host-pathogen interplay, here we investigated the role of the versatile posttranslational modification (PTM) persulfidation for both fungal virulence and antifungal host defense. We show that an A. fumigatus mutant with low persulfidation levels is more susceptible to host-mediated killing and displays reduced virulence in murine models of infection. Additionally, we found that a single nucleotide polymorphism (SNP) in the human gene encoding cystathionine γ-lyase (CTH) causes a reduction in cellular persulfidation and correlates with a predisposition of hematopoietic stem cell transplant recipients to invasive pulmonary aspergillosis (IPA), as correct levels of persulfidation are required for optimal antifungal activity of recipients' lung resident host cells. Importantly, the levels of host persulfidation determine the levels of fungal persulfidation, ultimately reflecting a host-pathogen functional correlation and highlighting a potential new therapeutic target for the treatment of aspergillosis.
Collapse
Affiliation(s)
- Monica Sueiro-Olivares
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jennifer Scott
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Sara Gago
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Dunja Petrovic
- Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et Genetique Cellulaires (IBGC), Bordeaux, France
- Université de Bordeaux, Institut de Biochimie et Genetique Cellulaires (IBGC), Bordeaux, France
| | - Emilia Kouroussis
- Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et Genetique Cellulaires (IBGC), Bordeaux, France
- Université de Bordeaux, Institut de Biochimie et Genetique Cellulaires (IBGC), Bordeaux, France
| | - Jasmina Zivanovic
- Centre National de la Recherche Scientifique (CNRS), Institut de Biochimie et Genetique Cellulaires (IBGC), Bordeaux, France
- Université de Bordeaux, Institut de Biochimie et Genetique Cellulaires (IBGC), Bordeaux, France
| | - Yidong Yu
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany
| | - Marlene Strobel
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute (ICVS)/Biomaterials, Biodegradables and Biomimetics (3B’s)—PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Darren Thomson
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rachael Fortune-Grant
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Sina Thusek
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany
| | - Paul Bowyer
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute (ICVS)/Biomaterials, Biodegradables and Biomimetics (3B’s)—PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Elaine Bignell
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | | | - Jorge Amich
- Manchester Fungal Infection Group (MFIG), School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
32
|
Hewawaduge C, Senevirathne A, Jawalagatti V, Kim JW, Lee JH. Copper-impregnated three-layer mask efficiently inactivates SARS-CoV2. ENVIRONMENTAL RESEARCH 2021; 196:110947. [PMID: 33662346 PMCID: PMC7919544 DOI: 10.1016/j.envres.2021.110947] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 05/09/2023]
Abstract
The present study investigates the potential of SARS-CoV-2 inactivation by a copper sulfide (CuS) incorporated three-layer mask design. The mask consisted of the outer, middle, and inner layers to give comfort, strength, shape, and safety. The outer layer contained a total of 4.4% CuS (w/w) (2.2% CuS coated & 2.2% CuS impregnated) nylon fibers and the middle entrapment area contain a total of 17.6% CuS (w/w) impregnated nylon. No CuS was present in the inner layer. The antiviral efficacy assessment revealed, CuS incorporated mask is highly effective in inactivating SARS-CoV-2 within 30 min exposure. After, 1h and 2 h exposure, near-complete elimination of virus were observed by cytopathy, fluorescence, and viral copy number. The antiviral activity of the mask material was derived by incorporated solid-state CuS. Noticeably, the antiviral activity of CuS against SARS-CoV-2 was in the form of solid-state CuS, but not as Cu2+ ionic form derived by dissolved CuSO4. The kinetics of droplet entrapment revealed, that the three-layered mask almost completely block virus-containing droplet pass-through for short exposure periods of 1-2 min, and 80% efficacy for longer exposure times of 5-10 min. We also demonstrated the incorporated CuS is evenly distributed all over the fibers assuring the uniformity of potential antiviral activity and proves, CuS particles are not easily shed out of the fabric fibers. The inactivation efficacy demonstrated against SARS-CoV-2 proves that the CuS incorporated three-layer mask will be a lifesaver during the present intense global pandemic.
Collapse
Affiliation(s)
- Chamith Hewawaduge
- College of Veterinary Medicine and Korea Zoonosis Research Institute, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine and Korea Zoonosis Research Institute, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Vijayakumar Jawalagatti
- College of Veterinary Medicine and Korea Zoonosis Research Institute, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Jang Whan Kim
- LSK Finetex Co., Ltd, Goyang-Si, Gyeonggi-Do, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine and Korea Zoonosis Research Institute, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea.
| |
Collapse
|
33
|
Santana CM, Gauger P, Vetger A, Magstadt D, Kim DS, Shrestha D, Charavaryamath C, Rumbeiha WK. Ambient hydrogen sulfide exposure increases the severity of influenza A virus infection in swine. ARCHIVES OF ENVIRONMENTAL & OCCUPATIONAL HEALTH 2021; 76:526-538. [PMID: 33750267 DOI: 10.1080/19338244.2021.1896986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Hydrogen sulfide (H2S) is common in concentrated pig feed operations from the decomposition of manure. Ambient H2S is a respiratory tract irritant and an environmental stressor for caretakers and pigs. Influenza A virus (IAV), a zoonotic pathogen, has caused prior pandemics. The effects of H2S or IAV alone on the respiratory system have been investigated, but their interaction has not. We hypothesized that exposure to environmentally-relevant H2S concentrations increases the pathogenicity of IAV infection in swine. Thirty-five, three-week old pigs of mixed sex were exposed to breathing air or H2S via inhalation 6 hours daily for 12 days. After 7 days, pigs were inoculated with H3N2 IAV (or a placebo). Results showed that ambient H2S increased the severity of respiratory distress and lung pathology. H2S also suppressed IL-IL-1β, IL-6 and IL-8 cytokine response in BALF and increased viral loads and nasal shedding.
Collapse
Affiliation(s)
- Cristina M Santana
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Phillip Gauger
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Amber Vetger
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Drew Magstadt
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Dong-Suk Kim
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Denusha Shrestha
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | | | - Wilson K Rumbeiha
- Department of Molecular Biosciences, University of California, Davis, CA, USA
| |
Collapse
|
34
|
Mendoza CA, Yamaoka S, Tsuda Y, Matsuno K, Weisend CM, Ebihara H. The NF-κB inhibitor, SC75741, is a novel antiviral against emerging tick-borne bandaviruses. Antiviral Res 2020; 185:104993. [PMID: 33296695 DOI: 10.1016/j.antiviral.2020.104993] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/12/2020] [Accepted: 12/03/2020] [Indexed: 11/28/2022]
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) and Heartland virus (HRTV) cause viral hemorrhagic fever-like illnesses in humans due to an aberrant host inflammatory response, which contributes to pathogenesis. Here, we established two separate minigenome (MG) systems based on the M-segment of SFTSV and HRTV. Following characterization of both systems for SFTSV and HRTV, we used them as a platform to screen potential compounds that inhibit viral RNA synthesis. We demonstrated that the NF-κB inhibitor, SC75741, reduces viral RNA synthesis of SFTSV and HRTV using our MG platform and validated these results using infectious SFTSV and HRTV. These results may lead to the use of MG systems as potential screening systems for the identification of antiviral compounds and yield novel insights into host-factors that could play role in bandavirus transcription and replication.
Collapse
Affiliation(s)
- Crystal A Mendoza
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
| | - Satoko Yamaoka
- Mayo Clinic, Department of Molecular Medicine, Rochester, MN, 55905, USA
| | - Yoshimi Tsuda
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, 060-8638, Japan
| | - Keita Matsuno
- Unit of Risk Analysis and Management, Hokkaido University Research Center for Zoonosis Control, Sapporo, Hokkaido, 001-0020, Japan; International Collaboration Unit, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, 001-0020, Japan
| | - Carla M Weisend
- Mayo Clinic, Department of Molecular Medicine, Rochester, MN, 55905, USA
| | - Hideki Ebihara
- Mayo Clinic, Department of Molecular Medicine, Rochester, MN, 55905, USA.
| |
Collapse
|
35
|
Gojon G, Morales GA. SG1002 and Catenated Divalent Organic Sulfur Compounds as Promising Hydrogen Sulfide Prodrugs. Antioxid Redox Signal 2020; 33:1010-1045. [PMID: 32370538 PMCID: PMC7578191 DOI: 10.1089/ars.2020.8060] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/15/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022]
Abstract
Significance: Sulfur has a critical role in protein structure/function and redox status/signaling in all living organisms. Although hydrogen sulfide (H2S) and sulfane sulfur (SS) are now recognized as central players in physiology and pathophysiology, the full scope and depth of sulfur metabolome's impact on human health and healthy longevity has been vastly underestimated and is only starting to be grasped. Since many pathological conditions have been related to abnormally low levels of H2S/SS in blood and/or tissues, and are amenable to treatment by H2S supplementation, development of safe and efficacious H2S donors deserves to be undertaken with a sense of urgency; these prodrugs also hold the promise of becoming widely used for disease prevention and as antiaging agents. Recent Advances: Supramolecular tuning of the properties of well-known molecules comprising chains of sulfur atoms (diallyl trisulfide [DATS], S8) was shown to lead to improved donors such as DATS-loaded polymeric nanoparticles and SG1002. Encouraging results in animal models have been obtained with SG1002 in heart failure, atherosclerosis, ischemic damage, and Duchenne muscular dystrophy; with TC-2153 in Alzheimer's disease, schizophrenia, age-related memory decline, fragile X syndrome, and cocaine addiction; and with DATS in brain, colon, gastric, and breast cancer. Critical Issues: Mode-of-action studies on allyl polysulfides, benzyl polysulfides, ajoene, and 12 ring-substituted organic disulfides and thiosulfonates led several groups of researchers to conclude that the anticancer effect of these compounds is not mediated by H2S and is only modulated by reactive oxygen species, and that their central model of action is selective protein S-thiolation. Future Directions: SG1002 is likely to emerge as the H2S donor of choice for acquiring knowledge on this gasotransmitter's effects in animal models, on account of its unique ability to efficiently generate H2S without byproducts and in a slow and sustained mode that is dose independent and enzyme independent. Efficient tuning of H2S donation characteristics of DATS, dibenzyl trisulfide, and other hydrophobic H2S prodrugs for both oral and parenteral administration will be achieved not only by conventional structural modification of a lead molecule but also through the new "supramolecular tuning" paradigm.
Collapse
|
36
|
Rahman MA, Glasgow JN, Nadeem S, Reddy VP, Sevalkar RR, Lancaster JR, Steyn AJC. The Role of Host-Generated H 2S in Microbial Pathogenesis: New Perspectives on Tuberculosis. Front Cell Infect Microbiol 2020; 10:586923. [PMID: 33330130 PMCID: PMC7711268 DOI: 10.3389/fcimb.2020.586923] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
For centuries, hydrogen sulfide (H2S) was considered primarily as a poisonous gas and environmental hazard. However, with the discovery of prokaryotic and eukaryotic enzymes for H2S production, breakdown, and utilization, H2S has emerged as an important signaling molecule in a wide range of physiological and pathological processes. Hence, H2S is considered a gasotransmitter along with nitric oxide (•NO) and carbon monoxide (CO). Surprisingly, despite having overlapping functions with •NO and CO, the role of host H2S in microbial pathogenesis is understudied and represents a gap in our knowledge. Given the numerous reports that followed the discovery of •NO and CO and their respective roles in microbial pathogenesis, we anticipate a rapid increase in studies that further define the importance of H2S in microbial pathogenesis, which may lead to new virulence paradigms. Therefore, this review provides an overview of sulfide chemistry, enzymatic production of H2S, and the importance of H2S in metabolism and immunity in response to microbial pathogens. We then describe our current understanding of the role of host-derived H2S in tuberculosis (TB) disease, including its influences on host immunity and bioenergetics, and on Mycobacterium tuberculosis (Mtb) growth and survival. Finally, this review discusses the utility of H2S-donor compounds, inhibitors of H2S-producing enzymes, and their potential clinical significance.
Collapse
Affiliation(s)
| | - Joel N Glasgow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sajid Nadeem
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vineel P Reddy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ritesh R Sevalkar
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jack R Lancaster
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Adrie J C Steyn
- Africa Health Research Institute, Durban, South Africa.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States.,Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
37
|
Citi V, Martelli A, Brancaleone V, Brogi S, Gojon G, Montanaro R, Morales G, Testai L, Calderone V. Anti-inflammatory and antiviral roles of hydrogen sulfide: Rationale for considering H 2 S donors in COVID-19 therapy. Br J Pharmacol 2020; 177:4931-4941. [PMID: 32783196 PMCID: PMC7436626 DOI: 10.1111/bph.15230] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/19/2020] [Accepted: 08/02/2020] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic caused by SARS-Cov-2 demands rapid, safe and effective therapeutic options. In the last decades, the endogenous gasotransmitter hydrogen sulfide (H2 S) has emerged as modulator of several biological functions and its deficiency has been associated with different disorders. Therefore, many H2 S-releasing agents have been developed as potential therapeutic tools for diseases related with impaired H2 S production and/or activity. Some of these compounds are in advanced clinical trials. Presently, the pivotal role of H2 S in modulating the inflammatory response and pro-inflammatory cytokine cascade is well recognized, and the usefulness of some H2 S-donors for the treatment of acute lung inflammation has been reported. Recent data is elucidating several mechanisms of action, which may account for antiviral effects of H2 S. Noteworthy, some preliminary clinical results suggest an inverse relationship between endogenous H2 S levels and severity of COVID-19. Therefore, repurposing of H2 S-releasing drugs may be a potential therapeutic opportunity for treatment of COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
| | | | | | - Simone Brogi
- Department of PharmacyUniversity of PisaPisaItaly
| | | | | | | | - Lara Testai
- Department of PharmacyUniversity of PisaPisaItaly
| | | |
Collapse
|
38
|
Dattilo M. The role of host defences in Covid 19 and treatments thereof. Mol Med 2020; 26:90. [PMID: 32993497 PMCID: PMC7522454 DOI: 10.1186/s10020-020-00216-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/07/2020] [Indexed: 01/08/2023] Open
Abstract
Hydrogen sulfide (H2S) is a natural defence against the infections from enveloped RNA viruses and is likely involved also in Covid 19. It was already shown to inhibit growth and pathogenic mechanisms of a variety of enveloped RNA viruses and it was now found that circulating H2S is higher in Covid 19 survivors compared to fatal cases. H2S release is triggered by carbon monoxide (CO) from the catabolism of heme by inducible heme oxygenase (HO-1) and heme proteins possess catalytic activity necessary for the H2S signalling by protein persulfidation. Subjects with a long promoter for the HMOX1 gene, coding for HO-1, are predicted for lower efficiency of this mechanism. SARS-cov-2 exerts ability to attack the heme of hemoglobin and other heme-proteins thus hampering both release and signalling of H2S. Lack of H2S-induced persulfidation of the KATP channels of leucocytes causes adhesion and release of the inflammatory cytokines, lung infiltration and systemic endothelial damage with hyper-coagulability. These events largely explain the sex and age distribution, clinical manifestations and co-morbidities of Covid-19. The understanding of this mechanism may be of guidance in re-evaluating the ongoing therapeutic strategies, with special attention to the interaction with mechanical ventilation, paracetamol and chloroquine use, and in the individuation of genetic traits causing increased susceptibility to the disruption of these physiologic processes and to a critical Covid 19. Finally, an array of therapeutic interventions with the potential to clinically modulate the HO-1/CO/H2S axis is already available or under development. These include CO donors and H2S donors and a boost to the endogenous production of H2S is also possible.
Collapse
|
39
|
Evgen'ev MB, Frenkel A. Possible application of H 2S-producing compounds in therapy of coronavirus (COVID-19) infection and pneumonia. Cell Stress Chaperones 2020; 25:713-715. [PMID: 32409956 PMCID: PMC7221330 DOI: 10.1007/s12192-020-01120-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Mikhail B Evgen'ev
- Laboratory of Molecular Mechanisms of Biological Adaptations, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia, 119991.
| | | |
Collapse
|
40
|
Zinovkin RA, Grebenchikov OA. Transcription Factor Nrf2 as a Potential Therapeutic Target for Prevention of Cytokine Storm in COVID-19 Patients. BIOCHEMISTRY. BIOKHIMIIA 2020; 85:833-837. [PMID: 33040727 PMCID: PMC7356136 DOI: 10.1134/s0006297920070111] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022]
Abstract
Nrf2 is a key transcription factor responsible for antioxidant defense in many tissues and cells, including alveolar epithelium, endothelium, and macrophages. Furthermore, Nrf2 functions as a transcriptional repressor that inhibits expression of the inflammatory cytokines in macrophages. Critically ill patients with COVID-19 infection often present signs of high oxidative stress and systemic inflammation - the leading causes of mortality. This article suggests rationale for the use of Nrf2 inducers to prevent development of an excessive inflammatory response in COVID-19 patients.
Collapse
Affiliation(s)
- R A Zinovkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, 119992, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| | - O A Grebenchikov
- Negovsky Research Institute of General Reanimatology, Russian Academy of Medical Sciences, Moscow, 107031, Russia
| |
Collapse
|
41
|
Abstract
The outbreak of COVID-19 pneumonia caused by a new coronavirus (severe acute respiratory syndrome coronavirus 2, SARS-CoV-2) is posing a global health emergency and has led to more than 380,000 deaths worldwide. The cell entry of SARS-CoV-2 depends on two host proteins angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2). There is currently no vaccine available and also no effective drug for the treatment of COVID-19. Hydrogen sulfide (H2S) as a novel gasotransmitter has been shown to protect against lung damage via its anti-inflammation, antioxidative stress, antiviral, prosurvival, and antiaging effects. In light of the research advances on H2S signaling in biology and medicine, this review proposed H2S as a potential defense against COVID-19. It is suggested that H2S may block SARS-CoV-2 entry into host cells by interfering with ACE2 and TMPRSS2, inhibit SARS-CoV-2 replication by attenuating virus assembly/release, and protect SARS-CoV-2-induced lung damage by suppressing immune response and inflammation development. Preclinical studies and clinical trials with slow-releasing H2S donor(s) or the activators of endogenous H2S-generating enzymes should be considered as a preventative treatment or therapy for COVID-19.
Collapse
Affiliation(s)
- Guangdong Yang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Ontario, Canada.,Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
42
|
Urbaniak C, Lorenzi H, Thissen J, Jaing C, Crucian B, Sams C, Pierson D, Venkateswaran K, Mehta S. The influence of spaceflight on the astronaut salivary microbiome and the search for a microbiome biomarker for viral reactivation. MICROBIOME 2020; 8:56. [PMID: 32312311 PMCID: PMC7171750 DOI: 10.1186/s40168-020-00830-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/18/2020] [Indexed: 05/05/2023]
Abstract
BACKGROUND Spaceflight impacts astronauts in many ways but little is known on how spaceflight affects the salivary microbiome and the consequences of these changes on astronaut health, such as viral reactivation. In order to understand this, the salivary microbiome was analyzed with 16S rRNA gene amplicon sequencing, and saliva viral titers were analyzed with quantitative polymerase chain reaction (qPCR) with primers specific for Epstein-Barr virus (EBV), herpes simplex virus (HSV), and varicella zoster virus (VZV) from 10 astronauts pre-flight, in-flight, and post-flight. RESULTS Streptococcus was the most abundant organism in the saliva, making up 8% of the total organisms detected, and their diversity decreased during spaceflight. Other organisms that had statistically significant changes were Proteobacteria and Fusobacteria which increased during flight and Actinobacteria which decreased during flight. At the genus level, Catonella, Megasphera, and Actinobacillus were absent in more than half of saliva samples collected pre-flight but were then detected during flight. In those subjects that already had these genera pre-flight, their relative abundances increased during flight. Correlation analyses between the microbiome and viral titers revealed a positive correlation with Gracilibacteria, Absconditabacteria, and Abiotrophia and a negative correlation between Oribacterium, Veillonella, and Haemophilus. There was also a significant positive correlation between microbiome richness and EBV viral titers. CONCLUSIONS This is the first study to look at how the salivary microbiome changes as a result of spaceflight and the search for bacterial biomarkers for viral reactivation. Further studies examining the role of specific organisms that were shown to be correlative and predictive in viral reactivation, a serious problem in astronauts during spaceflight, could lead to mitigation strategies to help prevent disease during both short and long duration space missions. Video abstract.
Collapse
Affiliation(s)
- Camilla Urbaniak
- NASA Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA, USA
| | - Hernan Lorenzi
- Department of Infectious Diseases, J. Craig Venter Institute, Rockville, MD, USA
| | - James Thissen
- Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Crystal Jaing
- Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | | | | | | | - Satish Mehta
- JES Tech, NASA Johnson Space Center, Houston, TX, USA
| |
Collapse
|
43
|
Nin DS, Idres SB, Song ZJ, Moore PK, Deng LW. Biological Effects of Morpholin-4-Ium 4 Methoxyphenyl (Morpholino) Phosphinodithioate and Other Phosphorothioate-Based Hydrogen Sulfide Donors. Antioxid Redox Signal 2020; 32:145-158. [PMID: 31642346 DOI: 10.1089/ars.2019.7896] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Significance: Hydrogen sulfide (H2S) is regarded as the third gasotransmitter along with nitric oxide and carbon monoxide. Extensive studies have demonstrated a variety of biological roles for H2S in neurophysiology, cardiovascular disease, endocrine regulation, and other physiological and pathological processes. Recent Advances: Novel H2S donors have proved useful in understanding the biological functions of H2S, with morpholin-4-ium 4 methoxyphenyl (morpholino) phosphinodithioate (GYY4137) being one of the most common pharmacological tools used. One advantage of GYY4137 over sulfide salts is its ability to release H2S in a slow and sustained manner akin to endogenous H2S production, rather than the delivery of H2S as a single concentrated burst. Critical Issues: Here, we summarize recent progress made in the characterization of the biological activities and pharmacological effects of GYY4137 in a range of in vitro and in vivo systems. Recent developments in the structural modification of GYY4137 to generate new compounds and their biological effects are also discussed. Future Directions: Slow-releasing H2S donor, GYY4137, and other phosphorothioate-based H2S donors are potent tools to study the biological functions of H2S. Despite recent progress, more work needs to be performed on these new compounds to unravel the mechanisms behind H2S release and pace of its discharge, as well as to define the effects of by-products of donors after H2S liberation. This will not only lead to better in-depth understanding of the biological effects of H2S but will also shed light on the future development of a new class of therapeutic agents with potential to treat a wide range of human diseases.
Collapse
Affiliation(s)
- Dawn Sijin Nin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shabana Binte Idres
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhi Jian Song
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Philip K Moore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lih-Wen Deng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore, Singapore
| |
Collapse
|
44
|
Escaffre O, Juelich TL, Freiberg AN. Polyphenylene carboxymethylene (PPCM) in vitro antiviral efficacy against Ebola virus in the context of a sexually transmitted infection. Antiviral Res 2019; 170:104567. [PMID: 31351092 DOI: 10.1016/j.antiviral.2019.104567] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
Ebola virus disease (EVD) is caused by Ebola virus (EBOV) and characterized in humans by hemorrhagic fever with high fatality rates. Human-to-human EBOV transmission occurs by physical contact with infected body fluids, or indirectly by contaminated surfaces. Sexual transmission is a route of infection only recently documented despite isolating EBOV virus or genome in the semen since 1976. Data on dissemination of EBOV from survivors remain limited and EBOV pathogenesis in humans following sexual transmission is unknown. The in vitro antiviral efficacy of polyphenylene carboxymethylene (PPCM) against EBOV was investigated considering the limited countermeasures available to block infection through sexual intercourse. PPCM is a vaginal topical contraceptive microbicide shown to prevent sexual transmission of HIV, herpes virus, and bacterial infections in several different models. Here we demonstrate its antiviral activity against EBOV. No viral replication was detected in the presence of PPCM in cell culture, including vaginal epithelial (VK2/E6E7) cells. Specifically, PPCM reduced viral attachment to cells by interfering with EBOV glycoprotein, and possibly through binding the cell surface glycosaminoglycan heparan sulfate important in the infection process. EBOV-infected VK2/E6E7 cells were found to secrete type III interferon (IFN), suggesting activation of distinct PRRs or downstream signaling factors from those required for type I and II IFN. The addition of PPCM following cell infection prevented notably the increase of these inflammation markers. Therefore, PPCM could potentially be used as a topical microbicide to reduce transmission by EBOV-positive survivors during sexual intercourse.
Collapse
Affiliation(s)
| | | | - Alexander N Freiberg
- Department of Pathology, Galveston, TX, 77555, USA; Center for Biodefense and Emerging Infectious Diseases, Galveston, TX, 77555, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
45
|
The Mycoplasma pneumoniae HapE alters the cytokine profile and growth of human bronchial epithelial cells. Biosci Rep 2019; 39:BSR20182201. [PMID: 30573530 PMCID: PMC6340952 DOI: 10.1042/bsr20182201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022] Open
Abstract
Mycoplasma pneumoniae is one of the most common pathogenic causes of community-acquired pneumonia. Hydrogen sulfide, alanine, and pyruvate producing enzyme (HapE) is a recently discovered M. pneumoniae virulence factor that can produce H2S to promote erythrocyte lysis. However, other cytotoxic effects of HapE have not been explored. The present study examined the effects of this enzyme on normal human bronchial epithelial (NHBE) cells, in an attempt to identify additional mechanisms of M. pneumoniae pathogenesis. Recombinant HapE was purified for use in downstream assays. MTT and colony formation assays were conducted to determine the effects of HapE on cell viability and growth, while flow cytometry was used to examine changes in cell proliferation and cell cycle function. ELISA was performed to examine changes in the cytokine profile of HapE-treated cells. HapE treatment arrested NHBE cells in S phase and inhibited cell proliferation in a concentration-dependent manner. The anti-inflammatory factors interleukin (IL)-4 and IL-6 were significantly enhanced following HapE treatment. Increased secretion of pro-inflammatory factors was not observed. The effects of HapE on the respiratory epithelium may have an impact on the efficiency of host immune surveillance and pathogen elimination, and contribute to the pathogenesis of M. pneumoniae.
Collapse
|
46
|
Bazhanov N, Ivanciuc T, Wu H, Garofalo M, Kang J, Xian M, Casola A. Thiol-Activated Hydrogen Sulfide Donors Antiviral and Anti-Inflammatory Activity in Respiratory Syncytial Virus Infection. Viruses 2018; 10:E249. [PMID: 29747463 PMCID: PMC5977242 DOI: 10.3390/v10050249] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
We have recently shown that endogenous hydrogen sulfide (H₂S), an important cellular gaseous mediator, exerts an antiviral and anti-inflammatory activity in vitro and in vivo, and that exogenous H₂S delivered via the synthetic H₂S-releasing compound GYY4137 also has similar properties. In this study, we sought to extend our findings to a novel class of H₂S donors, thiol-activated gem-dithiol-based (TAGDDs). In an in vitro model of human respiratory syncytial virus (RSV) infection, TAGDD-1 treatment significantly reduced viral replication, even when added up to six hours after infection. Using a mouse model of RSV infection, intranasal delivery of TAGDD-1 to infected mice significantly reduced viral replication and lung inflammation, markedly improving clinical disease parameters and pulmonary dysfunction, compared to vehicle treated controls. Overall our results indicate that this novel synthetic class of H₂S-releasing compounds exerts antiviral and anti-inflammatory activity in the context of RSV infection and represents a potential novel pharmacological approach to ameliorate viral-induced lung disease.
Collapse
Affiliation(s)
- Nikolay Bazhanov
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | - Haotian Wu
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | - Matteo Garofalo
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | - Jianming Kang
- Department of Chemistry, Washington State University, Pullman, WA 99164, USA.
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, WA 99164, USA.
| | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| |
Collapse
|
47
|
Pal VK, Bandyopadhyay P, Singh A. Hydrogen sulfide in physiology and pathogenesis of bacteria and viruses. IUBMB Life 2018; 70:393-410. [PMID: 29601123 PMCID: PMC6029659 DOI: 10.1002/iub.1740] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/14/2018] [Accepted: 03/02/2018] [Indexed: 12/18/2022]
Abstract
An increasing number of studies have established hydrogen sulfide (H2S) gas as a major cytoprotectant and redox modulator. Following its discovery, H2S has been found to have pleiotropic effects on physiology and human health. H2S acts as a gasotransmitter and exerts its influence on gastrointestinal, neuronal, cardiovascular, respiratory, renal, and hepatic systems. Recent discoveries have clearly indicated the importance of H2S in regulating vasorelaxation, angiogenesis, apoptosis, ageing, and metabolism. Contrary to studies in higher organisms, the role of H2S in the pathophysiology of infectious agents such as bacteria and viruses has been less studied. Bacterial and viral infections are often accompanied by changes in the redox physiology of both the host and the pathogen. Emerging studies indicate that bacterial-derived H2S constitutes a defense system against antibiotics and oxidative stress. The H2S signaling pathway also seems to interfere with redox-based events affected on infection with viruses. This review aims to summarize recent advances on the emerging role of H2S gas in the bacterial physiology and viral infections. Such studies have opened up new research avenues exploiting H2S as a potential therapeutic intervention.
Collapse
Affiliation(s)
- Virender Kumar Pal
- Department of Microbiology and Cell Biology, Centre for Infectious Disease Research, Indian Institute of Science (IISc), Bangalore, India
| | - Parijat Bandyopadhyay
- Department of Microbiology and Cell Biology, Centre for Infectious Disease Research, Indian Institute of Science (IISc), Bangalore, India
| | - Amit Singh
- Department of Microbiology and Cell Biology, Centre for Infectious Disease Research, Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
48
|
Atkins C, Freiberg AN. Recent advances in the development of antiviral therapeutics for Rift Valley fever virus infection. Future Virol 2017; 12:651-665. [PMID: 29181086 DOI: 10.2217/fvl-2017-0060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/26/2017] [Indexed: 12/25/2022]
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus endemic to sub-Saharan Africa and the Arabian Peninsula and the etiological agent of Rift Valley fever. Rift Valley fever is a disease of major public health and economic concern, affecting livestock and humans. In ruminants, RVFV infection is characterized by high mortality rates in newborns and near 100% abortion rates in pregnant animals. Infection in humans is typically manifested as a self-limiting febrile illness, but can lead to severe and fatal hepatitis, encephalitis, hemorrhagic fever or retinitis with partial or complete blindness. Currently, there are no specific treatment options available for RVFV infection. This review presents a summary of the therapeutic approaches that have been explored on the treatment of RVFV infection.
Collapse
Affiliation(s)
- Colm Atkins
- Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Alexander N Freiberg
- Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Sealy Center for Vaccine Development, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Center for Biodefense & Emerging Infectious Diseases, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,Department of Pathology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Sealy Center for Vaccine Development, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.,The Center for Biodefense & Emerging Infectious Diseases, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| |
Collapse
|
49
|
Szabo C, Papapetropoulos A. International Union of Basic and Clinical Pharmacology. CII: Pharmacological Modulation of H 2S Levels: H 2S Donors and H 2S Biosynthesis Inhibitors. Pharmacol Rev 2017; 69:497-564. [PMID: 28978633 PMCID: PMC5629631 DOI: 10.1124/pr.117.014050] [Citation(s) in RCA: 278] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Over the last decade, hydrogen sulfide (H2S) has emerged as an important endogenous gasotransmitter in mammalian cells and tissues. Similar to the previously characterized gasotransmitters nitric oxide and carbon monoxide, H2S is produced by various enzymatic reactions and regulates a host of physiologic and pathophysiological processes in various cells and tissues. H2S levels are decreased in a number of conditions (e.g., diabetes mellitus, ischemia, and aging) and are increased in other states (e.g., inflammation, critical illness, and cancer). Over the last decades, multiple approaches have been identified for the therapeutic exploitation of H2S, either based on H2S donation or inhibition of H2S biosynthesis. H2S donation can be achieved through the inhalation of H2S gas and/or the parenteral or enteral administration of so-called fast-releasing H2S donors (salts of H2S such as NaHS and Na2S) or slow-releasing H2S donors (GYY4137 being the prototypical compound used in hundreds of studies in vitro and in vivo). Recent work also identifies various donors with regulated H2S release profiles, including oxidant-triggered donors, pH-dependent donors, esterase-activated donors, and organelle-targeted (e.g., mitochondrial) compounds. There are also approaches where existing, clinically approved drugs of various classes (e.g., nonsteroidal anti-inflammatories) are coupled with H2S-donating groups (the most advanced compound in clinical trials is ATB-346, an H2S-donating derivative of the non-steroidal anti-inflammatory compound naproxen). For pharmacological inhibition of H2S synthesis, there are now several small molecule compounds targeting each of the three H2S-producing enzymes cystathionine-β-synthase (CBS), cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase. Although many of these compounds have their limitations (potency, selectivity), these molecules, especially in combination with genetic approaches, can be instrumental for the delineation of the biologic processes involving endogenous H2S production. Moreover, some of these compounds (e.g., cell-permeable prodrugs of the CBS inhibitor aminooxyacetate, or benserazide, a potentially repurposable CBS inhibitor) may serve as starting points for future clinical translation. The present article overviews the currently known H2S donors and H2S biosynthesis inhibitors, delineates their mode of action, and offers examples for their biologic effects and potential therapeutic utility.
Collapse
Affiliation(s)
- Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas (C.S.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece (A.P.); and Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Andreas Papapetropoulos
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas (C.S.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece (A.P.); and Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| |
Collapse
|
50
|
Bazhanov N, Ansar M, Ivanciuc T, Garofalo RP, Casola A. Hydrogen Sulfide: A Novel Player in Airway Development, Pathophysiology of Respiratory Diseases, and Antiviral Defenses. Am J Respir Cell Mol Biol 2017; 57:403-410. [PMID: 28481637 PMCID: PMC5650090 DOI: 10.1165/rcmb.2017-0114tr] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/04/2017] [Indexed: 12/28/2022] Open
Abstract
Hydrogen sulfide (H2S) is a biologically relevant signaling molecule in mammals. Along with the volatile substances nitric oxide (NO) and carbon monoxide (CO), H2S is defined as a gasotransmitter. It plays a physiological role in a variety of functions, including synaptic transmission, vascular tone, angiogenesis, inflammation, and cellular signaling. The generation of H2S is catalyzed by cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3-MST). The expression of CBS and CSE is tissue specific, with CBS being expressed predominantly in the brain, and CSE in peripheral tissues, including lungs. CSE expression and activity are developmentally regulated, and recent studies suggest that CSE plays an important role in lung alveolarization during fetal development. In the respiratory tract, endogenous H2S has been shown to participate in the regulation of important functions such as airway tone, pulmonary circulation, cell proliferation or apoptosis, fibrosis, oxidative stress, and inflammation. In the past few years, changes in the generation of H2S have been linked to the pathogenesis of a variety of acute and chronic inflammatory lung diseases, including asthma and chronic obstructive pulmonary disease. Recently, our laboratory made the critical discovery that cellular H2S exerts broad-spectrum antiviral activity both in vitro and in vivo, in addition to independent antiinflammatory activity. These findings have important implications for the development of novel therapeutic strategies for viral respiratory infections, as well as other inflammatory lung diseases, especially in light of recent significant efforts to generate controlled-release H2S donors for clinical therapeutic applications.
Collapse
Affiliation(s)
| | | | | | - Roberto P. Garofalo
- Departments of Pediatrics and
- Microbiology and Immunology, and
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Antonella Casola
- Departments of Pediatrics and
- Microbiology and Immunology, and
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|