1
|
Wu Z, Wang Y, Gao R, Chen J, Chen Y, Li M, Gao Y. Potential therapeutic effects of traditional Chinese medicine in acute mountain sickness: pathogenesis, mechanisms and future directions. Front Pharmacol 2024; 15:1393209. [PMID: 38895636 PMCID: PMC11183292 DOI: 10.3389/fphar.2024.1393209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Background and objectives Acute mountain sickness (AMS) is a pathology with different symptoms in which the organism is not adapted to the environment that occurs under the special environment of high altitude. Its main mechanism is the organism's tissue damage caused by acute hypobaric hypoxia. Traditional Chinese medicine (TCM) theory focuses on the holistic concept. TCM has made remarkable achievements in the treatment of many mountain sicknesses. This review outlines the pathogenesis of AMS in modern and traditional medicine, the progress of animal models of AMS, and summarizes the therapeutic effects of TCM on AMS. Methods Using the keywords "traditional Chinese medicine," "herbal medicine," "acute mountain sickness," "high-altitude pulmonary edema," "high-altitude cerebral edema," "acute hypobaric hypoxia," and "high-altitude," all relevant TCM literature published up to November 2023 were collected from Scopus, Web of Science, PubMed, and China National Knowledge Infrastructure databases, and the key information was analyzed. Results We systematically summarised the effects of acute hypobaric hypoxia on the tissues of the organism, the study of the methodology for the establishment of an animal model of AMS, and retrieved 18 proprietary Chinese medicines for the clinical treatment of AMS. The therapeutic principle of medicines is mainly invigorating qi, activating blood and removing stasis. The components of botanical drugs mainly include salidroside, ginsenoside Rg1, and tetrahydrocurcumin. The mechanism of action of TCM in the treatment of AMS is mainly through the regulation of HIF-1α/NF-κB signaling pathway, inhibition of inflammatory response and oxidative stress, and enhancement of energy metabolism. Conclusion The main pathogenesis of AMS is unclear. Still, TCM formulas and components have been used to treat AMS through multifaceted interventions, such as compound danshen drip pills, Huangqi Baihe granules, salidroside, and ginsenoside Rg1. These components generally exert anti-AMS pharmacological effects by inhibiting the expression of VEGF, concentration of MDA and pro-inflammatory factors, down-regulating NF-κB/NLRP3 pathway, and promoting SOD and Na + -K + -ATPase activities, which attenuates acute hypobaric hypoxia-induced tissue injury. This review comprehensively analyses the application of TCM in AMS and makes suggestions for more in-depth studies in the future, aiming to provide some ideas and insights for subsequent studies.
Collapse
Affiliation(s)
- Zhenhui Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
- Department of Hematology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yihao Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Rong Gao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Junru Chen
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingfan Chen
- Department of Traditional Chinese Medicine, The Sixth Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Maoxing Li
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yue Gao
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
2
|
Ali Akbari Ghavimi S, Faulkner TJ, Tata RR, Hemmerla AJ, Huddleston SE, Rezaei F, Lungren ES, Zhang R, Bumann EE, Ulery BD. Hydrogen Sulfide Delivery to Enhance Bone Tissue Engineering Cell Survival. Pharmaceuticals (Basel) 2024; 17:585. [PMID: 38794155 PMCID: PMC11124412 DOI: 10.3390/ph17050585] [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: 02/15/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 05/26/2024] Open
Abstract
Though crucial for natural bone healing, local calcium ion (Ca2+) and phosphate ion (Pi) concentrations can exceed the cytotoxic limit leading to mitochondrial overload, oxidative stress, and cell death. For bone tissue engineering applications, H2S can be employed as a cytoprotective molecule to enhance mesenchymal stem cell (MSC) tolerance to cytotoxic Ca2+/Pi concentrations. Varied concentrations of sodium hydrogen sulfide (NaSH), a fast-releasing H2S donor, were applied to assess the influence of H2S on MSC proliferation. The results suggested a toxicity limit of 4 mM for NaSH and that 1 mM of NaSH could improve cell proliferation and differentiation in the presence of cytotoxic levels of Ca2+ (32 mM) and/or Pi (16 mM). To controllably deliver H2S over time, a novel donor molecule (thioglutamic acid-GluSH) was synthesized and evaluated for its H2S release profile. Excitingly, GluSH successfully maintained cytoprotective level of H2S over 7 days. Furthermore, MSCs exposed to cytotoxic Ca2+/Pi concentrations in the presence of GluSH were able to thrive and differentiate into osteoblasts. These findings suggest that the incorporation of a sustained H2S donor such as GluSH into CaP-based bone graft substitutes can facilitate considerable cytoprotection, making it an attractive option for complex bone regenerative engineering applications.
Collapse
Affiliation(s)
- Soheila Ali Akbari Ghavimi
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
| | - Trent J. Faulkner
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
| | - Rama Rao Tata
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
| | - August J. Hemmerla
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
| | - Samantha E. Huddleston
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
| | - Farnoushsadat Rezaei
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
| | - Ethan S. Lungren
- Department of Chemistry, University of Missouri, Columbia, MO 65211, USA
| | - Rui Zhang
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
| | - Erin E. Bumann
- Department of Oral and Craniofacial Sciences, University of Missouri, Kansas City, MO 64110, USA;
| | - Bret D. Ulery
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, USA; (S.A.A.G.); (R.R.T.); (S.E.H.)
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
- Materials Science & Engineering Institute, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
3
|
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: 12.0] [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
|
4
|
Olson KR. A Case for Hydrogen Sulfide Metabolism as an Oxygen Sensing Mechanism. Antioxidants (Basel) 2021; 10:antiox10111650. [PMID: 34829521 PMCID: PMC8615108 DOI: 10.3390/antiox10111650] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022] Open
Abstract
The ability to detect oxygen availability is a ubiquitous attribute of aerobic organisms. However, the mechanism(s) that transduce oxygen concentration or availability into appropriate physiological responses is less clear and often controversial. This review will make the case for oxygen-dependent metabolism of hydrogen sulfide (H2S) and polysulfides, collectively referred to as reactive sulfur species (RSS) as a physiologically relevant O2 sensing mechanism. This hypothesis is based on observations that H2S and RSS metabolism is inversely correlated with O2 tension, exogenous H2S elicits physiological responses identical to those produced by hypoxia, factors that affect H2S production or catabolism also affect tissue responses to hypoxia, and that RSS efficiently regulate downstream effectors of the hypoxic response in a manner consistent with a decrease in O2. H2S-mediated O2 sensing is then compared to the more generally accepted reactive oxygen species (ROS) mediated O2 sensing mechanism and a number of reasons are offered to resolve some of the confusion between the two.
Collapse
Affiliation(s)
- Kenneth R Olson
- Department of Physiology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, USA
| |
Collapse
|
5
|
Huang Y, Wang G, Zhou Z, Tang Z, Zhang N, Zhu X, Ni X. Endogenous Hydrogen Sulfide Is an Important Factor in Maintaining Arterial Oxygen Saturation. Front Pharmacol 2021; 12:677110. [PMID: 34135757 PMCID: PMC8200772 DOI: 10.3389/fphar.2021.677110] [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: 03/07/2021] [Accepted: 05/18/2021] [Indexed: 12/30/2022] Open
Abstract
The gasotransmitter H2S is involved in various physiological and pathophysiological processes. The aim of this study was to investigate the physiological functions of H2S in the lungs. In the model of mouse with genetic deficiency in a H2S natural synthesis enzyme cystathionine-γ-lyase (CSE), we found that arterial oxygen saturation (SaO2) was decreased compared with wild type mice. Hypoxyprobe test showed that mild hypoxia occurred in the tissues of heart, lungs and kidneys in Cse-/- mice. H2S donor GYY4137 treatment increased SaO2 and ameliorated hypoxia state in cardiac and renal tissues. Further, we revealed that lung blood perfusion and airway responsiveness were not linked to reduced SaO2 level. Lung injury was found in Cse-/- mice as evidenced by alveolar wall thickening, diffuse interstitial edema and leukocyte infiltration in pulmonary tissues. IL-8, IL-1β, and TNF-α levels were markedly increased and oxidative stress levels were also significantly higher with increased levels of the pro-oxidative biomarker, MDA, decreased levels of the anti-oxidative biomarkers, T-AOC and GSH/GSSG, and reduced superoxide dismutase (SOD) activity in lung tissues of Cse-/- mice compared with those of wild type mice. GYY4137 treatment ameliorated lung injury and suppressed inflammatory state and oxidative stress in lung tissues of Cse-/- mice. A decrease in SaO2 was found in normal mice under hypoxia. These mice displayed lung injury as evidenced by alveolar wall thickening, interstitial edema and leukocyte infiltration. Increased levels of inflammatory cytokines and oxidative stress were also found in lung tissues of the mice with hypoxia insult. GYY4137 treatment increased SaO2 and ameliorated lung injury, inflammation and oxidative stress. Our data indicate that endogenous H2S is an important factor in maintaining normal SaO2 by preventing oxidative stress and inflammation in the lungs.
Collapse
Affiliation(s)
- Yan Huang
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China.,Reproductive medicine center, Department of obstetrics and Gynecology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Gang Wang
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhan Zhou
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengshan Tang
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China
| | - Ningning Zhang
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China
| |
Collapse
|
6
|
Sokolov AS, Nekrasov PV, Shaposhnikov MV, Moskalev AA. Hydrogen sulfide in longevity and pathologies: Inconsistency is malodorous. Ageing Res Rev 2021; 67:101262. [PMID: 33516916 DOI: 10.1016/j.arr.2021.101262] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) is one of the biologically active gases (gasotransmitters), which plays an important role in various physiological processes and aging. Its production in the course of methionine and cysteine catabolism and its degradation are finely balanced, and impairment of H2S homeostasis is associated with various pathologies. Despite the strong geroprotective action of exogenous H2S in C. elegans, there are controversial effects of hydrogen sulfide and its donors on longevity in other models, as well as on stress resistance, age-related pathologies and aging processes, including regulation of senescence-associated secretory phenotype (SASP) and senescent cell anti-apoptotic pathways (SCAPs). Here we discuss that the translation potential of H2S as a geroprotective compound is influenced by a multiplicity of its molecular targets, pleiotropic biological effects, and the overlapping ranges of toxic and beneficial doses. We also consider the challenges of the targeted delivery of H2S at the required dose. Along with this, the complexity of determining the natural levels of H2S in animal and human organs and their ambiguous correlations with longevity are reviewed.
Collapse
|
7
|
Wang G, Huang Y, Zhang N, Liu W, Wang C, Zhu X, Ni X. Hydrogen Sulfide Is a Regulator of Hemoglobin Oxygen-Carrying Capacity via Controlling 2,3-BPG Production in Erythrocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8877691. [PMID: 33628390 PMCID: PMC7896853 DOI: 10.1155/2021/8877691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/18/2022]
Abstract
Hydrogen sulfide (H2S) is naturally synthesized in a wide range of mammalian tissues. Whether H2S is involved in the regulation of erythrocyte functions remains unknown. Using mice with a genetic deficiency in a H2S natural synthesis enzyme cystathionine-γ-lyase (CSE) and high-throughput metabolomic profiling, we found that levels of erythrocyte 2,3-bisphosphoglycerate (2,3-BPG), an erythroid-specific metabolite negatively regulating hemoglobin- (Hb-) oxygen (O2) binding affinity, were increased in CSE knockout (Cse -/-) mice under normoxia. Consistently, the 50% oxygen saturation (P50) value was increased in erythrocytes of Cse -/- mice. These effects were reversed by treatment with H2S donor GYY4137. In the models of cultured mouse and human erythrocytes, we found that H2S directly acts on erythrocytes to decrease 2,3-BPG production, thereby enhancing Hb-O2 binding affinity. Mouse genetic studies showed that H2S produced by peripheral tissues has a tonic inhibitory effect on 2,3-BPG production and consequently maintains Hb-O2 binding affinity in erythrocytes. We further revealed that H2S promotes Hb release from the membrane to the cytosol and consequently enhances bisphosphoglycerate mutase (BPGM) anchoring to the membrane. These processes might be associated with S-sulfhydration of Hb. Moreover, hypoxia decreased the circulatory H2S level and increased the erythrocyte 2,3-BPG content in mice, which could be reversed by GYY4137 treatment. Altogether, our study revealed a novel signaling pathway that regulates oxygen-carrying capacity in erythrocytes and highlights a previously unrecognized role of H2S in erythrocyte 2,3-BPG production.
Collapse
Affiliation(s)
- Gang Wang
- National Clinical Research Center for Geriatric Disorders and National International Joint Research Center for Medical Metabolomics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Yan Huang
- National Clinical Research Center for Geriatric Disorders and National International Joint Research Center for Medical Metabolomics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
- General Hospital of Southern Theater Command, Guangzhou, 510010 Guangdong, China
| | - Ningning Zhang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Wenhu Liu
- National Clinical Research Center for Geriatric Disorders and National International Joint Research Center for Medical Metabolomics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Changnan Wang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders and National International Joint Research Center for Medical Metabolomics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
8
|
Cheng MHY, Mo Y, Zheng G. Nano versus Molecular: Optical Imaging Approaches to Detect and Monitor Tumor Hypoxia. Adv Healthc Mater 2021; 10:e2001549. [PMID: 33241672 DOI: 10.1002/adhm.202001549] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/21/2020] [Indexed: 12/18/2022]
Abstract
Hypoxia is a ubiquitous feature of solid tumors, which plays a key role in tumor angiogenesis and resistance development. Conventional hypoxia detection methods lack continuous functional detection and are generally less suitable for dynamic hypoxia measurement. Optical sensors hereby provide a unique opportunity to noninvasively image hypoxia with high spatiotemporal resolution and enable real-time detection. Therefore, these approaches can provide a valuable tool for personalized treatment planning against this hallmark of aggressive cancers. Many small optical molecular probes can enable analyte triggered response and their photophysical properties can also be fine-tuned through structural modification. On the other hand, optical nanoprobes can acquire unique intrinsic optical properties through nanoconfinement as well as enable simultaneous multimodal imaging and drug delivery. Furthermore, nanoprobes provide biological advantages such as improving bioavailability and systemic delivery of the sensor to enhance bioavailability. This review provides a comprehensive overview of the physical, chemical, and biological analytes for cancer hypoxia detection and focuses on discussing the latest nano- and molecular developments in various optical imaging approaches (fluorescence, phosphorescence, and photoacoustic) in vivo. Finally, this review concludes with a perspective toward the potentials of these optical imaging approaches in hypoxia detection and the challenges with molecular and nanotechnology design strategies.
Collapse
Affiliation(s)
- Miffy Hok Yan Cheng
- Princess Margaret Cancer Centre University Health Network 101 College Street, PMCRT 5–354 Toronto Ontario M5G 1L7 Canada
| | - Yulin Mo
- Princess Margaret Cancer Centre University Health Network 101 College Street, PMCRT 5–354 Toronto Ontario M5G 1L7 Canada
- Institute of Medical Science University of Toronto 101 College Street Toronto Ontario M5G 1L7 Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre University Health Network 101 College Street, PMCRT 5–354 Toronto Ontario M5G 1L7 Canada
- Institute of Medical Science University of Toronto 101 College Street Toronto Ontario M5G 1L7 Canada
- Department of Medical Biophysics University of Toronto 101 College Street Toronto Ontario M5G 1L7 Canada
| |
Collapse
|
9
|
Skandalis DA, Dobell CD, Shaw JC, Tattersall GJ. Hydrogen sulfide exposure reduces thermal set point in zebrafish. ROYAL SOCIETY OPEN SCIENCE 2020; 7:200416. [PMID: 33391778 PMCID: PMC7735326 DOI: 10.1098/rsos.200416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 10/06/2020] [Indexed: 05/26/2023]
Abstract
Behavioural flexibility allows ectotherms to exploit the environment to govern their metabolic physiology, including in response to environmental stress. Hydrogen sulfide (H2S) is a widespread environmental toxin that can lethally inhibit metabolism. However, H2S can also alter behaviour and physiology, including a hypothesized induction of hibernation-like states characterized by downward shifts of the innate thermal set point (anapyrexia). Support for this hypothesis has proved controversial because it is difficult to isolate active and passive components of thermoregulation, especially in animals with high resting metabolic heat production. Here, we directly test this hypothesis by leveraging the natural behavioural thermoregulatory drive of fish to move between environments of different temperatures in accordance with their current physiological state and thermal preference. We observed a decrease in adult zebrafish (Danio rerio) preferred body temperature with exposure to 0.02% H2S, which we interpret as a shift in the thermal set point. Individuals exhibited consistent differences in shuttling behaviour and preferred temperatures, which were reduced by a constant temperature magnitude during H2S exposure. Seeking lower temperatures alleviated H2S-induced metabolic stress, as measured by reduced rates of aquatic surface respiration. Our findings highlight the interactions between individual variation and sublethal impacts of environmental toxins on behaviour.
Collapse
Affiliation(s)
| | | | | | - Glenn J. Tattersall
- Department of Biological Sciences, Brock University, St Catharines, 500 Glenridge Avenue, St Catharines, Ontario, CanadaL2S 3A1
| |
Collapse
|
10
|
Liu H, Perumal N, Manicam C, Mercieca K, Prokosch V. Proteomics Reveals the Potential Protective Mechanism of Hydrogen Sulfide on Retinal Ganglion Cells in an Ischemia/Reperfusion Injury Animal Model. Pharmaceuticals (Basel) 2020; 13:ph13090213. [PMID: 32867129 PMCID: PMC7557839 DOI: 10.3390/ph13090213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Glaucoma is the leading cause of irreversible blindness and is characterized by progressive retinal ganglion cell (RGC) degeneration. Hydrogen sulfide (H2S) is a potent neurotransmitter and has been proven to protect RGCs against glaucomatous injury in vitro and in vivo. This study is to provide an overall insight of H2S’s role in glaucoma pathophysiology. Ischemia-reperfusion injury (I/R) was induced in Sprague-Dawley rats (n = 12) by elevating intraocular pressure to 55 mmHg for 60 min. Six of the animals received intravitreal injection of H2S precursor prior to the procedure and the retina was harvested 24 h later. Contralateral eyes were assigned as control. RGCs were quantified and compared within the groups. Retinal proteins were analyzed via label-free mass spectrometry based quantitative proteomics approach. The pathways of the differentially expressed proteins were identified by ingenuity pathway analysis (IPA). H2S significantly improved RGC survival against I/R in vivo (p < 0.001). In total 1115 proteins were identified, 18 key proteins were significantly differentially expressed due to I/R and restored by H2S. Another 11 proteins were differentially expressed following H2S. IPA revealed a significant H2S-mediated activation of pathways related to mitochondrial function, iron homeostasis and vasodilation. This study provides first evidence of the complex role that H2S plays in protecting RGC against I/R.
Collapse
Affiliation(s)
- Hanhan Liu
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Caroline Manicam
- Experimental and Translational Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (H.L.); (N.P.); (C.M.)
| | - Karl Mercieca
- Royal Eye Hospital, School of Medicine, University of Manchester, Manchester M13 9WH, UK;
| | - Verena Prokosch
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-1703862250
| |
Collapse
|
11
|
Hydrogen Sulfide: Emerging Role in Bladder, Kidney, and Prostate Malignancies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2360945. [PMID: 31781328 PMCID: PMC6875223 DOI: 10.1155/2019/2360945] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/22/2019] [Accepted: 09/30/2019] [Indexed: 12/23/2022]
Abstract
Hydrogen sulfide (H2S) is the latest member of the gasotransmitter family and known to play essential roles in cancer pathophysiology. H2S is produced endogenously and can be administered exogenously. Recent studies showed that H2S in cancers has both pro- and antitumor roles. Understanding the difference in the expression and localization of tissue-specific H2S-producing enzymes in healthy and cancer tissues allows us to develop tools for cancer diagnosis and treatment. Urological malignancies are some of the most common cancers in both men and women, and their early detection is vital since advanced cancers are recurrent, metastatic, and often resistant to treatment. This review summarizes the roles of H2S in cancer and looks at current studies investigating H2S activity and expression of H2S-producing enzymes in urinary cancers. We specifically focused on urothelial carcinoma, renal cell carcinoma, and prostate cancer, as they form the majority of newly diagnosed urinary cancers. Recent studies show that besides the physiological activity of H2S in cancer cells, there are patterns between the development and prognosis of urinary cancers and the expression of H2S-producing enzymes and indirectly the H2S levels. Though controversial and not completely understood, studying the expression of H2S-producing enzymes in cancer tissue may represent an avenue for novel diagnostic and therapeutic strategies for addressing urological malignancies.
Collapse
|
12
|
Hydrogen Sulfide Oxidation: Adaptive Changes in Mitochondria of SW480 Colorectal Cancer Cells upon Exposure to Hypoxia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8102936. [PMID: 30838088 PMCID: PMC6374825 DOI: 10.1155/2019/8102936] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide (H2S), a known inhibitor of cytochrome c oxidase (CcOX), plays a key signaling role in human (patho)physiology. H2S is synthesized endogenously and mainly metabolized by a mitochondrial sulfide-oxidizing pathway including sulfide:quinone oxidoreductase (SQR), whereby H2S-derived electrons are injected into the respiratory chain stimulating O2 consumption and ATP synthesis. Under hypoxic conditions, H2S has higher stability and is synthesized at higher levels with protective effects for the cell. Herein, working on SW480 colon cancer cells, we evaluated the effect of hypoxia on the ability of cells to metabolize H2S. The sulfide-oxidizing activity was assessed by high-resolution respirometry, measuring the stimulatory effect of sulfide on rotenone-inhibited cell respiration in the absence or presence of antimycin A. Compared to cells grown under normoxic conditions (air O2), cells exposed for 24 h to hypoxia (1% O2) displayed a 1.3-fold reduction in maximal sulfide-oxidizing activity and 2.7-fold lower basal O2 respiration. Based on citrate synthase activity assays, mitochondria of hypoxia-treated cells were 1.8-fold less abundant and displayed 1.4-fold higher maximal sulfide-oxidizing activity and 2.6-fold enrichment in SQR as evaluated by immunoblotting. We speculate that under hypoxic conditions mitochondria undergo these adaptive changes to protect cell respiration from H2S poisoning.
Collapse
|
13
|
Greco V, Spalloni A, Corasolla Carregari V, Pieroni L, Persichilli S, Mercuri NB, Urbani A, Longone P. Proteomics and Toxicity Analysis of Spinal-Cord Primary Cultures upon Hydrogen Sulfide Treatment. Antioxidants (Basel) 2018; 7:antiox7070087. [PMID: 29996549 PMCID: PMC6070951 DOI: 10.3390/antiox7070087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/05/2018] [Accepted: 07/07/2018] [Indexed: 12/25/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous gasotransmitter recognized as an essential body product with a dual, biphasic action. It can function as an antioxidant and a cytoprotective, but also as a poison with a high probability of causing brain damage when present at noxious levels. In a previous study, we measured toxic liquoral levels of H2S in sporadic amyotrophic lateral sclerosis (ALS) patients and in the familial ALS (fALS) mouse model, SOD1G93A. In addition, we experimentally demonstrated that H2S is extremely and selectively toxic to motor neurons, and that it is released by glial cells and increases Ca2+ concentration in motor neurons due to a lack of ATP. The presented study further examines the effect of toxic concentrations of H2S on embryonic mouse spinal-cord cultures. We performed a proteomic analysis that revealed a significant H2S-mediated activation of pathways related to oxidative stress and cell death, particularly the Nrf-2-mediated oxidative stress response and peroxiredoxins. Furthermore, we report that Na2S (a stable precursor of H2S) toxicity is, at least in part, reverted by the Bax inhibitor V5 and by necrostatin, a potent necroptosis inhibitor.
Collapse
Affiliation(s)
- Viviana Greco
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy.
| | - Alida Spalloni
- Department of Experimental Neuroscience, Molecular Neurobiology Unit, Fondazione Santa Lucia, 00143 Rome, Italy.
| | - Victor Corasolla Carregari
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy.
| | - Luisa Pieroni
- Department of Experimental Neuroscience, Proteomics and Metabonomics Unit, Fondazione Santa Lucia-IRCCS, 00143 Rome, Italy.
| | - Silvia Persichilli
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy.
| | - Nicola B Mercuri
- Department of Systems Medicine, Policlinico Universitario "Tor Vergata", University of Rome "Tor Vergata", 00133 Rome, Italy.
- Department of Experimental Neuroscience, Experimental Neurology Unit, 00143 Rome, Italy.
| | - Andrea Urbani
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy.
| | - Patrizia Longone
- Department of Experimental Neuroscience, Molecular Neurobiology Unit, Fondazione Santa Lucia, 00143 Rome, Italy.
| |
Collapse
|
14
|
Gorr TA. Hypometabolism as the ultimate defence in stress response: how the comparative approach helps understanding of medically relevant questions. Acta Physiol (Oxf) 2017; 219:409-440. [PMID: 27364602 DOI: 10.1111/apha.12747] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/28/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022]
Abstract
First conceptualized from breath-hold diving mammals, later recognized as the ultimate cell autonomous survival strategy in anoxia-tolerant vertebrates and burrowing or hibernating rodents, hypometabolism is typically recruited by resilient organisms to withstand and recover from otherwise life-threatening hazards. Through the coordinated down-regulation of biosynthetic, proliferative and electrogenic expenditures at times when little ATP can be generated, a metabolism turned 'down to the pilot light' allows the re-balancing of energy demand with supply at a greatly suppressed level in response to noxious exogenous stimuli or seasonal endogenous cues. A unifying hallmark of stress-tolerant organisms, the adaptation effectively prevents lethal depletion of ATP, thus delineating a marked contrast with susceptible species. Along with disengaged macromolecular syntheses, attenuated transmembrane ion shuttling and PO2 -conforming respiration rates, the metabolic slowdown in tolerant species usually culminates in a non-cycling, quiescent phenotype. However, such a reprogramming also occurs in leading human pathophysiologies. Ranging from microbial infections through ischaemia-driven infarcts to solid malignancies, cells involved in these disorders may again invoke hypometabolism to endure conditions non-permissive for growth. At the same time, their reduced activities underlie the frequent development of a general resistance to therapeutic interventions. On the other hand, a controlled induction of hypometabolic and/or hypothermic states by pharmacological means has recently stimulated intense research aimed at improved organ preservation and patient survival in situations requiring acutely administered critical care. The current review article therefore presents an up-to-date survey of concepts and applications of a coordinated and reversibly down-regulated metabolic rate as the ultimate defence in stress responses.
Collapse
Affiliation(s)
- T. A. Gorr
- Institute of Veterinary Physiology; Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| |
Collapse
|
15
|
Katsouda A, Bibli SI, Pyriochou A, Szabo C, Papapetropoulos A. Regulation and role of endogenously produced hydrogen sulfide in angiogenesis. Pharmacol Res 2016; 113:175-185. [PMID: 27569706 DOI: 10.1016/j.phrs.2016.08.026] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 08/05/2016] [Accepted: 08/24/2016] [Indexed: 01/16/2023]
Abstract
Recent studies have implicated endogenously produced H2S in the angiogenic process. On one hand, pharmacological inhibition and silencing of the enzymes involved in H2S synthesis attenuate the angiogenic properties of endothelial cells, including proliferation, migration and tube-like structure network formation. On the other hand, enhanced production of H2S by substrate supplementation or over-expression of H2S-producing enzymes leads to enhanced angiogenic responses in cultured endothelial cells. Importantly, H2S up-regulates expression of the key angiogenic factor vascular endothelial growth factor (VEGF) and contributes to the angiogenic signaling in response to VEGF. The signaling pathways mediating H2S-induced angiogenesis include mitogen-activated protein kinases, phosphoinositide-3 kinase, nitric oxide/cGMP-regulated cascades and ATP-sensitive potassium channels. Endogenously produced H2S has also been shown to facilitate neovascularization in prototypical model systems in vivo, and to contribute to wound healing, post-ischemic angiogenesis in the heart and other tissues, as well as in tumor angiogenesis. Targeting of H2S synthesizing enzymes might offer novel therapeutic opportunities for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Antonia Katsouda
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece, Greece; Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Sofia-Iris Bibli
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece, Greece
| | - Anastasia Pyriochou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece, Greece; Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Greece.
| |
Collapse
|