1
|
Song Y, Cao S, Sun X, Chen G. The interplay of hydrogen sulfide and microRNAs in cardiovascular diseases: insights and future perspectives. Mamm Genome 2024; 35:309-323. [PMID: 38834923 DOI: 10.1007/s00335-024-10043-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024]
Abstract
Hydrogen sulfide (H2S) is recognized as the third gasotransmitter, after nitric oxide (NO) and carbon monoxide (CO). It is known for its cardioprotective properties, including the relaxation of blood vessels, promotion of angiogenesis, regulation of myocardial cell apoptosis, inhibition of vascular smooth muscle cell proliferation, and reduction of inflammation. Additionally, abnormal H2S generation has been linked to the development of cardiovascular diseases (CVD), such as pulmonary hypertension, hypertension, atherosclerosis, vascular calcification, and myocardial injury. MicroRNAs (miRNAs) are non-coding, conserved, and versatile molecules that primarily influence gene expression by repressing translation and have emerged as biomarkers for CVD diagnosis. Studies have demonstrated that H2S can ameliorate cardiac dysfunction by regulating specific miRNAs, and certain miRNAs can also regulate H2S synthesis. The crosstalk between miRNAs and H2S offers a novel perspective for investigating the pathophysiology, prevention, and treatment of CVD. The present analysis outlines the interactions between H2S and miRNAs and their influence on CVD, providing insights into their future potential and advancement.
Collapse
Affiliation(s)
- Yunjia Song
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shuo Cao
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xutao Sun
- Department of Typhoid, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China.
| | - Guozhen Chen
- Department of Pediatrics, The Affiliated Yantai Yuhuangding Hospital, Yantai, Shandong, China.
| |
Collapse
|
2
|
Sun HJ, Lu QB, Zhu XX, Ni ZR, Su JB, Fu X, Chen G, Zheng GL, Nie XW, Bian JS. Pharmacology of Hydrogen Sulfide and Its Donors in Cardiometabolic Diseases. Pharmacol Rev 2024; 76:846-895. [PMID: 38866561 DOI: 10.1124/pharmrev.123.000928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/13/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
Cardiometabolic diseases (CMDs) are major contributors to global mortality, emphasizing the critical need for novel therapeutic interventions. Hydrogen sulfide (H2S) has garnered enormous attention as a significant gasotransmitter with various physiological, pathophysiological, and pharmacological impacts within mammalian cardiometabolic systems. In addition to its roles in attenuating oxidative stress and inflammatory response, burgeoning research emphasizes the significance of H2S in regulating proteins via persulfidation, a well known modification intricately associated with the pathogenesis of CMDs. This review seeks to investigate recent updates on the physiological actions of endogenous H2S and the pharmacological roles of various H2S donors in addressing diverse aspects of CMDs across cellular, animal, and clinical studies. Of note, advanced methodologies, including multiomics, intestinal microflora analysis, organoid, and single-cell sequencing techniques, are gaining traction due to their ability to offer comprehensive insights into biomedical research. These emerging approaches hold promise in characterizing the pharmacological roles of H2S in health and diseases. We will critically assess the current literature to clarify the roles of H2S in diseases while also delineating the opportunities and challenges they present in H2S-based pharmacotherapy for CMDs. SIGNIFICANCE STATEMENT: This comprehensive review covers recent developments in H2S biology and pharmacology in cardiometabolic diseases CMDs. Endogenous H2S and its donors show great promise for the management of CMDs by regulating numerous proteins and signaling pathways. The emergence of new technologies will considerably advance the pharmacological research and clinical translation of H2S.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Qing-Bo Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xue-Xue Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Zhang-Rong Ni
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Jia-Bao Su
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xiao Fu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Guo Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Guan-Li Zheng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xiao-Wei Nie
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Jin-Song Bian
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| |
Collapse
|
3
|
Gonzalez AL, Dungan MM, Smart CD, Madhur MS, Doran AC. Inflammation Resolution in the Cardiovascular System: Arterial Hypertension, Atherosclerosis, and Ischemic Heart Disease. Antioxid Redox Signal 2024; 40:292-316. [PMID: 37125445 PMCID: PMC11071112 DOI: 10.1089/ars.2023.0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
Significance: Chronic inflammation has emerged as a major underlying cause of many prevalent conditions in the Western world, including cardiovascular diseases. Although targeting inflammation has emerged as a promising avenue by which to treat cardiovascular disease, it is also associated with increased risk of infection. Recent Advances: Though previously assumed to be passive, resolution has now been identified as an active process, mediated by unique immunoresolving mediators and mechanisms designed to terminate acute inflammation and promote tissue repair. Recent work has determined that failures of resolution contribute to chronic inflammation and the progression of human disease. Specifically, failure to produce pro-resolving mediators and the impaired clearance of dead cells from inflamed tissue have been identified as major mechanisms by which resolution fails in disease. Critical Issues: Drawing from a rapidly expanding body of experimental and clinical studies, we review here what is known about the role of inflammation resolution in arterial hypertension, atherosclerosis, myocardial infarction, and ischemic heart disease. For each, we discuss the involvement of specialized pro-resolving mediators and pro-reparative cell types, including T regulatory cells, myeloid-derived suppressor cells, and macrophages. Future Directions: Pro-resolving therapies offer the promise of limiting chronic inflammation without impairing host defense. Therefore, it is imperative to better understand the mechanisms underlying resolution to identify therapeutic targets. Antioxid. Redox Signal. 40, 292-316.
Collapse
Affiliation(s)
- Azuah L. Gonzalez
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew M. Dungan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - C. Duncan Smart
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Amanda C. Doran
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
4
|
Xia Y, Zhang W, He K, Bai L, Miao Y, Liu B, Zhang X, Jin S, Wu Y. Hydrogen sulfide alleviates lipopolysaccharide-induced myocardial injury through TLR4-NLRP3 pathway. Physiol Res 2023; 72:15-25. [PMID: 36545872 PMCID: PMC10069815 DOI: 10.33549/physiolres.934928] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023] Open
Abstract
To investigate the effect of hydrogen sulfide (H2S) on myocardial injury in sepsis-induced myocardial dysfunction (SIMD), male C57BL/6 mice were intraperitoneally injected with lipopolysaccharide (LPS) (10 mg/kg, i.p.) to induce cardiac dysfunction without or with the H2S donor sodium hydrosulfide (NaHS) (50 µmol/kg, i.p.) administration 3 h after LPS injection. Six hours after the LPS injection, echocardiography, cardiac hematoxylin and eosin (HE) staining, myocardial damage and inflammatory biomarkers and Western blot results were analyzed. In mice, the administration of LPS decreased left ventricular ejection fraction (LVEF) by 30 % along with lowered H2S levels (35 % reduction). It was observed that cardiac troponin I (cTnI), tumor necrosis factor-alpha (TNF-alpha), and interleukin-1beta (IL-1beta) levels were all increased (by 0.22-fold, 2000-fold and 0.66-fold respectively). HE staining revealed structural damage and inflammatory cell infiltration in the myocardial tissue after LPS administration. Moreover, after 6 h of LPS treatment, toll-like receptor 4 (TLR4) and nod-like receptor protein 3 (NLRP3) expressions were up-regulated 2.7-fold and 1.6-fold respectively. When compared to the septic mice, NaHS enhanced ventricular function (by 0.19-fold), decreased cTnI, TNF-alpha, and IL-1beta levels (by 11 %, 33 %, and 16 % respectively) and downregulated TLR4 and NLRP3 expressions (by 64 % and 31 % respectively). Furthermore, NaHS did not further improve cardiac function and inflammation in TLR4-/- mice or mice in which NLRP3 activation was inhibited by MCC950, after LPS injection. In conclusion, these findings imply that decreased endogenous H2S promotes the progression of SIMD, whereas exogenous H2S alleviates SIMD by inhibiting inflammation via the TLR4-NLRP3 pathway suppression.
Collapse
Affiliation(s)
- Y Xia
- Department of Physiology, Hebei Medical University, Hebei, China. ;
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Sykora M, Andelova K, Szeiffova Bacova B, Egan Benova T, Martiskova A, Knezl V, Tribulova N. Hypertension Induces Pro-arrhythmic Cardiac Connexome Disorders: Protective Effects of Treatment. Biomolecules 2023; 13:biom13020330. [PMID: 36830700 PMCID: PMC9953310 DOI: 10.3390/biom13020330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 02/11/2023] Open
Abstract
Prolonged population aging and unhealthy lifestyles contribute to the progressive prevalence of arterial hypertension. This is accompanied by low-grade inflammation and over time results in heart dysfunction and failure. Hypertension-induced myocardial structural and ion channel remodeling facilitates the development of both atrial and ventricular fibrillation, and these increase the risk of stroke and sudden death. Herein, we elucidate hypertension-induced impairment of "connexome" cardiomyocyte junctions. This complex ensures cell-to-cell adhesion and coupling for electrical and molecular signal propagation. Connexome dysfunction can be a key factor in promoting the occurrence of both cardiac arrhythmias and heart failure. However, the available literature indicates that arterial hypertension treatment can hamper myocardial structural remodeling, hypertrophy and/or fibrosis, and preserve connexome function. This suggests the pleiotropic effects of antihypertensive agents, including anti-inflammatory. Therefore, further research is required to identify specific molecular targets and pathways that will protect connexomes, and it is also necessary to develop new approaches to maintain heart function in patients suffering from primary or pulmonary arterial hypertension.
Collapse
|
6
|
Bełtowski J, Kowalczyk-Bołtuć J. Hydrogen sulfide in the experimental models of arterial hypertension. Biochem Pharmacol 2023; 208:115381. [PMID: 36528069 DOI: 10.1016/j.bcp.2022.115381] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Hydrogen sulfide (H2S) is the third member of gasotransmitter family together with nitric oxide and carbon monoxide. H2S is involved in the regulation of blood pressure by controlling vascular tone, sympathetic nervous system activity and renal sodium excretion. Moderate age-dependent hypertension and endothelial dysfunction develop in mice with knockout of cystathionine γ-lyase (CSE), the enzyme involved in H2S production in the cardiovascular system. Decreased H2S concentration as well as the expression and activities of H2S-producing enzymes have been observed in most commonly used animal models of hypertension such as spontaneously hypertensive rats, Dahl salt-sensitive rats, chronic administration of NO synthase inhibitors, angiotensin II infusion and two-kidney-one-clip hypertension, the model of renovascular hypertension. Administration of H2S donors decreases blood pressure in these models but has no major effects on blood pressure in normotensive animals. H2S donors not only reduce blood pressure but also end-organ injury such as vascular and myocardial hypertrophy and remodeling, hypertension-associated kidney injury or erectile dysfunction. H2S level and signaling are modulated by some antihypertensive medications as well as natural products with antihypertensive activity such as garlic polysulfides or plant-derived isothiocyanates as well as non-pharmacological interventions. Modifying H2S signaling is the potential novel therapeutic approach for the management of hypertension, however, more experimental clinical studies about the role of H2S in hypertension are required.
Collapse
Affiliation(s)
- Jerzy Bełtowski
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland.
| | - Jolanta Kowalczyk-Bołtuć
- Endocrinology and Metabolism Clinic, Internal Medicine Clinic with Hypertension Department, Medical Institute of Rural Health, Lublin, Poland.
| |
Collapse
|
7
|
Sodium Thiosulphate-Loaded Liposomes Control Hydrogen Sulphide Release and Retain Its Biological Properties in Hypoxia-like Environment. Antioxidants (Basel) 2022; 11:antiox11112092. [DOI: 10.3390/antiox11112092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/09/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Hypoxia, or insufficient oxygen availability is a common feature in the development of a myriad of cardiovascular-related conditions including ischemic disease. Hydrogen sulphide (H2S) donors, such as sodium thiosulphate (STS), are known for their cardioprotective properties. However, H2S due to its gaseous nature, is released and cleared rapidly, limiting its potential translation to clinical settings. For the first time, we developed and characterised liposome formulations encapsulating STS and explored their potential for modulating STS uptake, H2S release and the ability to retain pro-angiogenic and biological signals in a hypoxia-like environment mirroring oxygen insufficiency in vitro. Liposomes were prepared by varying lipid ratios and characterised for size, polydispersity and charge. STS liposomal encapsulation was confirmed by HPLC-UV detection and STS uptake and H2S release was assessed in vitro. To mimic hypoxia, cobalt chloride (CoCl2) was administered in conjunction with formulated and non-formulated STS, to explore pro-angiogenic and metabolic signals. Optimised liposomal formulation observed a liposome diameter of 146.42 ± 7.34 nm, a polydispersity of 0.22 ± 0.19, and charge of 3.02 ± 1.44 mV, resulting in 25% STS encapsulation. Maximum STS uptake (76.96 ± 3.08%) from liposome encapsulated STS was determined at 24 h. Co-exposure with CoCl2 and liposome encapsulated STS resulted in increased vascular endothelial growth factor mRNA as well as protein expression, enhanced wound closure and increased capillary-like formation. Finally, liposomal STS reversed metabolic switch induced by hypoxia by enhancing mitochondrial bioenergetics. These novel findings provide evidence of a feasible controlled-delivery system for STS, thus H2S, using liposome-based nanoparticles. Likewise, data suggests that in scenarios of hypoxia, liposomal STS is a good therapeutic candidate to sustain pro-angiogenic signals and retain metabolic functions that might be impaired by limited oxygen and nutrient availability.
Collapse
|
8
|
Estrogen Protects against Renal Ischemia-Reperfusion Injury by Regulating Th17/Treg Cell Immune Balance. DISEASE MARKERS 2022; 2022:7812099. [PMID: 36246554 PMCID: PMC9560860 DOI: 10.1155/2022/7812099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/01/2022] [Accepted: 09/23/2022] [Indexed: 12/31/2022]
Abstract
Inflammation is a critical mediator of renal ischemia-reperfusion (I/R) injury (IRI), and T lymphocytes exert a key role in the renal IRI-induced inflammation. Connexin 43 (Cx43) is related to the maintenance of T lymphocyte homeostasis. Various preclinical researches have reported that estrogen is a renoprotective agent based on its anti-inflammatory potential. The present research is aimed at studying the role of T lymphocytes activated by Cx43 in 17β-estradiol-mediated protection against renal IRI. Female rats were classified into six groups: control rats, I/R rats, ovariectomized rats, ovariectomized I/R rats, and ovariectomized rats treated with 17β-estradiol or gap27. Levels of serum creatinine (Scr) and blood urea nitrogen (BUN) and Paller scoring were dramatically increased in I/R rats, especially in ovariectomized rats. By contrast, these indicators were markedly decreased by administering estradiol or gap27. Immunofluorescence staining revealed that CD4+ T cells infiltrated kidney tissues in the early stage of IRI. In both peripheral blood and renal tissue, the proportion of CD3+CD4+ T cells and ratio of CD4+ to CD8+ were high in I/R rats, especially in ovariectomized rats. The proportion of CD3+CD8+ T cells was low in peripheral blood but high in renal tissues. Administration of estrogen or Gap27 reversed these effects. IL-17 levels in both serum and tissue homogenate were significantly increased in ovariectomized rats subjected to I/R but significantly decreased in estrogen or gap 27 treated rats. The opposite trend was observed for IL-10 levels. Correlation analysis demonstrated that IL-17 was correlated positively with BUN, Scr, and Paller scores, while IL-10 was negatively correlated with these indicators. Western blot showed that Cx43 expression was markedly increased in the peripheral blood T lymphocytes of I/R rats, especially ovariectomized rats. After intervention with estrogen and gap27, Cx43 expression was significantly downregulated. These findings indicate that Cx43 may participate in the regulation of Th17/Treg balance by estrogen against renal IRI.
Collapse
|
9
|
Ding H, Chang J, He F, Gai S, Yang P. Hydrogen Sulfide: An Emerging Precision Strategy for Gas Therapy. Adv Healthc Mater 2022; 11:e2101984. [PMID: 34788499 DOI: 10.1002/adhm.202101984] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/06/2021] [Indexed: 12/13/2022]
Abstract
Advances in nanotechnology have enabled the rapid development of stimuli-responsive therapeutic nanomaterials for precision gas therapy. Hydrogen sulfide (H2 S) is a significant gaseous signaling molecule with intrinsic biochemical properties, which exerts its various physiological effects under both normal and pathological conditions. Various nanomaterials with H2 S-responsive properties, as new-generation therapeutic agents, are explored to guide therapeutic behaviors in biological milieu. The cross disciplinary of H2 S is an emerging scientific hotspot that studies the chemical properties, biological mechanisms, and therapeutic effects of H2 S. This review summarizes the state-of-art research on H2 S-related nanomedicines. In particular, recent advances in H2 S therapeutics for cancer, such as H2 S-mediated gas therapy and H2 S-related synergistic therapies (combined with chemotherapy, photodynamic therapy, photothermal therapy, and chemodynamic therapy) are highlighted. Versatile imaging techniques for real-time monitoring H2 S during biological diagnosis are reviewed. Finally, the biosafety issues, current challenges, and potential possibilities in the evolution of H2 S-based therapy that facilitate clinical translation to patients are discussed.
Collapse
Affiliation(s)
- He Ding
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education College of Materials Science and Chemical Engineering Harbin Engineering University Harbin 150001 P. R. China
| | - Jinhu Chang
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education College of Materials Science and Chemical Engineering Harbin Engineering University Harbin 150001 P. R. China
| | - Fei He
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education College of Materials Science and Chemical Engineering Harbin Engineering University Harbin 150001 P. R. China
| | - Shili Gai
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education College of Materials Science and Chemical Engineering Harbin Engineering University Harbin 150001 P. R. China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education College of Materials Science and Chemical Engineering Harbin Engineering University Harbin 150001 P. R. China
| |
Collapse
|
10
|
Zhu C, Liu Q, Li X, Wei R, Ge T, Zheng X, Li B, Liu K, Cui R. Hydrogen sulfide: A new therapeutic target in vascular diseases. Front Endocrinol (Lausanne) 2022; 13:934231. [PMID: 36034427 PMCID: PMC9399516 DOI: 10.3389/fendo.2022.934231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Hydrogen sulfide (H2S) is one of most important gas transmitters. H2S modulates many physiological and pathological processes such as inflammation, oxidative stress and cell apoptosis that play a critical role in vascular function. Recently, solid evidence show that H2S is closely associated to various vascular diseases. However, specific function of H2S remains unclear. Therefore, in this review we systemically summarized the role of H2S in vascular diseases, including hypertension, atherosclerosis, inflammation and angiogenesis. In addition, this review also outlined a novel therapeutic perspective comprising crosstalk between H2S and smooth muscle cell function. Therefore, this review may provide new insight inH2S application clinically.
Collapse
Affiliation(s)
- Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Qing Liu
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Xin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ran Wei
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Xiufen Zheng
- Department of Surgery, Western University, London, ON, Canada
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Ranji Cui, ; Kexiang Liu,
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Ranji Cui, ; Kexiang Liu,
| |
Collapse
|
11
|
Donertas Ayaz B, Oliveira AC, Malphurs WL, Redler T, de Araujo AM, Sharma RK, Sirmagul B, Zubcevic J. Central Administration of Hydrogen Sulfide Donor NaHS Reduces Iba1-Positive Cells in the PVN and Attenuates Rodent Angiotensin II Hypertension. Front Neurosci 2021; 15:690919. [PMID: 34602965 PMCID: PMC8479468 DOI: 10.3389/fnins.2021.690919] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Hydrogen sulfide (H2S) is a gaseous signaling molecule with neuromodulatory, anti-inflammatory, and anti-hypertensive effects. Here, we investigate whether chronic intracerebroventricular (ICV) infusion of sodium hydrosulfide (NaHS), an H2S donor, can alleviate angiotensin II (Ang II)-induced hypertension (HTN), improve autonomic function, and impact microglia in the paraventricular nucleus (PVN) of the hypothalamus, a brain region associated with autonomic control of blood pressure (BP) and neuroinflammation in HTN. Chronic delivery of Ang II (200 ng/kg/min, subcutaneous) for 4 weeks produced a typical increase in BP and sympathetic drive and elevated the number of ionized calcium binding adaptor molecule 1-positive (Iba1+) cells in the PVN of male, Sprague-Dawley rats. ICV co-infusion of NaHS (at 30 and/or 60 nmol/h) significantly attenuated these effects of Ang II. Ang II also increased the abundance of cecal Deltaproteobacteria and Desulfovibrionales, among others, which was prevented by ICV NaHS co-infusion at 30 and 60 nmol/h. We observed no differences in circulating H2S between the groups. Our results suggest that central H2S may alleviate rodent HTN independently from circulating H2S via effects on autonomic nervous system and PVN microglia.
Collapse
Affiliation(s)
- Basak Donertas Ayaz
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
- Department of Pharmacology, College of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Aline C. Oliveira
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Wendi L. Malphurs
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Ty Redler
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Alan Moreira de Araujo
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Ravindra K. Sharma
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Basar Sirmagul
- Department of Pharmacology, College of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Jasenka Zubcevic
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
- Department of Physiology and Pharmacology, Center for Hypertension and Precision Medicine, College of Medicine, University of Toledo, Toledo, OH, United States
| |
Collapse
|
12
|
Abramavicius S, Petersen AG, Renaltan NS, Prat-Duran J, Torregrossa R, Stankevicius E, Whiteman M, Simonsen U. GYY4137 and Sodium Hydrogen Sulfide Relaxations Are Inhibited by L-Cysteine and K V7 Channel Blockers in Rat Small Mesenteric Arteries. Front Pharmacol 2021; 12:613989. [PMID: 33841145 PMCID: PMC8032876 DOI: 10.3389/fphar.2021.613989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/12/2021] [Indexed: 01/23/2023] Open
Abstract
Donors of H2S may be beneficial in treating cardiovascular diseases where the plasma levels of H2S are decreased. Therefore, we investigated the mechanisms involved in relaxation of small arteries induced by GYY4137 [(4-methoxyphenyl)-morpholin-4-yl-sulfanylidene-sulfido-λ5-phosphane;morpholin-4-ium], which is considered a slow-releasing H2S donor. Sulfides were measured by use of 5,5′-dithiobis-(2-nitro benzoic acid), and small rat mesenteric arteries with internal diameters of 200–250 µm were mounted in microvascular myographs for isometric tension recordings. GYY4137 produced similar low levels of sulfides in the absence and the presence of arteries. In U46619-contracted small mesenteric arteries, GYY4137 (10−6–10–3 M) induced concentration-dependent relaxations, while a synthetic, sulfur-free, GYY4137 did not change the vascular tone. L-cysteine (10−6–10–3 M) induced only small relaxations reaching 24 ± 6% at 10–3 M. Premixing L-cysteine (10–3 M) with Na2S and GYY4137 decreased Na2S relaxation and abolished GYY4137 relaxation, an effect prevented by an nitric oxide (NO) synthase inhibitor, L-NAME (Nω-nitro-L-arginine methyl ester). In arteries without endothelium or in the presence of L-NAME, relaxation curves for GYY4137 were rightward shifted. High extracellular K+ concentrations decreased Na2S and abolished GYY4137 relaxation suggesting potassium channel-independent mechanisms are also involved Na2S relaxation while potassium channel activation is pivotal for GYY4137 relaxation in small arteries. Blockers of large-conductance calcium-activated (BKCa) and voltage-gated type 7 (KV7) potassium channels also inhibited GYY4137 relaxations. The present findings suggest that L-cysteine by reaction with Na2S and GYY4137 and formation of sulfides, inhibits relaxations by these compounds. The low rate of release of H2S species from GYY4137 is reflected by the different sensitivity of these relaxations towards high K+ concentration and potassium channel blockers compared with Na2S. The perspective is that the rate of release of sulfides plays an important for the effects of H2S salt vs. donors in small arteries, and hence for a beneficial effect of GYY4137 for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Silvijus Abramavicius
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Asbjørn G Petersen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Nirthika S Renaltan
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Judit Prat-Duran
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | | | - Edgaras Stankevicius
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
13
|
Lv B, Chen S, Tang C, Jin H, Du J, Huang Y. Hydrogen sulfide and vascular regulation - An update. J Adv Res 2021; 27:85-97. [PMID: 33318869 PMCID: PMC7728588 DOI: 10.1016/j.jare.2020.05.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hydrogen sulfide (H2S) is considered to be the third gasotransmitter after carbon monoxide (CO) and nitric oxide (NO). It plays an important role in the regulation of vascular homeostasis. Vascular remodeling have has proved to be related to the impaired H2S generation. AIM OF REVIEW This study aimed to summarize and discuss current data about the function of H2S in vascular physiology and pathophysiology as well as the underlying mechanisms. KEY SCIENTIFIC CONCEPTS OF REVIEW Endogenous hydrogen sulfide (H2S) as a third gasotransmitter is primarily generated by the enzymatic pathways and regulated by several metabolic pathways. H2S as a physiologic vascular regulator, inhibits proliferation, regulates its apoptosis and autophagy of vascular cells and controls the vascular tone. Accumulating evidence shows that the downregulation of H2S pathway is involved in the pathogenesis of a variety of vascular diseases, such as hypertension, atherosclerosis and pulmonary hypertension. Alternatively, H2S supplementation may greatly help to prevent the progression of the vascular diseases by regulating vascular tone, inhibiting vascular inflammation, protecting against oxidative stress and proliferation, and modulating vascular cell apoptosis, which has been verified in animal and cell experiments and even in the clinical investigation. Besides, H2S system and angiotensin-converting enzyme (ACE) inhibitors play a vital role in alleviating ischemic heart disease and left ventricular dysfunction. Notably, sulfhydryl-containing ACEI inhibitor zofenopril is superior to other ACE inhibitors due to its capability of H2S releasing, in addition to ACE inhibition. The design and application of novel H2S donors have significant clinical implications in the treatment of vascular-related diseases. However, further research regarding the role of H2S in vascular physiology and pathophysiology is required.
Collapse
Affiliation(s)
- Boyang Lv
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Selena Chen
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Corresponding authors at: Department of Pediatrics, Peking University First Hospital, Beijing, China (J. Du).
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, China
- Corresponding authors at: Department of Pediatrics, Peking University First Hospital, Beijing, China (J. Du).
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Corresponding authors at: Department of Pediatrics, Peking University First Hospital, Beijing, China (J. Du).
| |
Collapse
|
14
|
Gorini F, Bustaffa E, Chatzianagnostou K, Bianchi F, Vassalle C. Hydrogen sulfide and cardiovascular disease: Doubts, clues, and interpretation difficulties from studies in geothermal areas. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 743:140818. [PMID: 32758850 DOI: 10.1016/j.scitotenv.2020.140818] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/10/2020] [Accepted: 07/06/2020] [Indexed: 06/11/2023]
Abstract
Hydrogen sulfide (H2S) represents one of the main pollutants originating from both geologic phenomena such as volcanoes, geysers, fumaroles and hot springs, and geothermal plants that produce heat and electricity. Many increasing data suggest that H2S retains a variety of biological properties, and modulates many pathways related to cardiovascular pathophysiology although its role as beneficial/adverse determinant on cardiovascular disease (CVD) is not clearly established. In this review, the current knowledge on the association between H2S exposure and risk of CVD in geothermal areas has been examined. The few epidemiological studies carried out in geothermal areas suggest, in some cases, a protective role of H2S towards CVD, while in others a positive association between exposure to H2S and increased incidence of CVD. Most of the studies have an ecological design that does not allow to produce evidence to support a causal relationship and also often lack for an adequate adjustment for individual CVD risk factors. The review has also considered the potential role of two other aspects not sufficiently explored in this relationship: the production of endogenous H2S that is a gasotransmitter producing beneficial effects on cardiovascular function at low concentration and the intake of H2S-releasing drugs for the treatment of patients affected by hypertension, inflammatory diseases, and CVD. Thus, a threshold effect of H2S and the shift of action as beneficial/adverse determinant given by the synergy of exogenous exposure and endogenous production cannot be excluded. In this complex scenario, an effort is warranted in the future to include a more comprehensive evaluation of risk for CVD in relation to H2S emissions, especially in geothermal areas.
Collapse
Affiliation(s)
- Francesca Gorini
- Unit of Environmental Epidemiology and Diseases Registries, Institute of Clinical Physiology, National Research Council, IFC-CNR, via Moruzzi 1, Pisa 56124, Italy.
| | - Elisa Bustaffa
- Unit of Environmental Epidemiology and Diseases Registries, Institute of Clinical Physiology, National Research Council, IFC-CNR, via Moruzzi 1, Pisa 56124, Italy
| | | | - Fabrizio Bianchi
- Unit of Environmental Epidemiology and Diseases Registries, Institute of Clinical Physiology, National Research Council, IFC-CNR, via Moruzzi 1, Pisa 56124, Italy
| | - Cristina Vassalle
- Gabriele Monasterio Foundation for the Medical and Public Health Research, via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
15
|
Okada M, Fukuyama K, Shiroyama T, Murata M. A Working Hypothesis Regarding Identical Pathomechanisms between Clinical Efficacy and Adverse Reaction of Clozapine via the Activation of Connexin43. Int J Mol Sci 2020; 21:ijms21197019. [PMID: 32987640 PMCID: PMC7583770 DOI: 10.3390/ijms21197019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/04/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Clozapine (CLZ) is an approved antipsychotic agent for the medication of treatment-resistant schizophrenia but is also well known as one of the most toxic antipsychotics. Recently, the World Health Organization’s (WHO) global database (VigiBase) reported the relative lethality of severe adverse reactions of CLZ. Agranulocytosis is the most famous adverse CLZ reaction but is of lesser lethality compared with the other adverse drug reactions of CLZ. Unexpectedly, VigiBase indicated that the prevalence and relative lethality of pneumonia, cardiotoxicity, and seizures associated with CLZ were more serious than that of agranulocytosis. Therefore, haematological monitoring in CLZ patients monitoring system provided success in the prevention of lethal adverse events from CLZ-induced agranulocytosis. Hereafter, psychiatrists must amend the CLZ patients monitoring system to protect patients with treatment-resistant schizophrenia from severe adverse CLZ reactions, such as pneumonia, cardiotoxicity, and seizures, according to the clinical evidence and pathophysiology. In this review, we discuss the mechanisms of clinical efficacy and the adverse reactions of CLZ based on the accumulating pharmacodynamic findings of CLZ, including tripartite synaptic transmission, and we propose suggestions for amending the monitoring and medication of adverse CLZ reactions associated with pneumonia, cardiotoxicity, and seizures.
Collapse
Affiliation(s)
- Motohiro Okada
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (K.F.); (T.S.)
- Correspondence: ; Tel.: +81-59-231-5018
| | - Kouji Fukuyama
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (K.F.); (T.S.)
| | - Takashi Shiroyama
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (K.F.); (T.S.)
| | - Masahiko Murata
- National Hospital Organization Sakakibara Hospital, 777 Sakakibara, Tsu, Mie 514-1292, Japan;
| |
Collapse
|
16
|
Zhang LΖ, Fan ZR, Wang L, Liu LQ, Li XZ, Li L, Si JQ, Ma KT. Carbenoxolone decreases monocrotaline‑induced pulmonary inflammation and pulmonary arteriolar remodeling in rats by decreasing the expression of connexins in T lymphocytes. Int J Mol Med 2019; 45:81-92. [PMID: 31746364 PMCID: PMC6889920 DOI: 10.3892/ijmm.2019.4406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022] Open
Abstract
The adaptive immune response mediated by T lymphocytes is a well-established factor in the pathogenesis of pulmonary inflammation. Changes in the expression of various connexins (Cxs) or disruption of connexin-mediated cellular communication in T lymphocytes contribute to inflammation or tissue remodeling. The aim of the present study was to investigate the potential therapeutic value of blocking Cxs in a monocrotaline (MCT)-induced pulmonary inflammation rat model. Carbenoxolone (CBX) was used to inhibit connexin-mediated cellular communication. An MCT rat model was established by intraperitoneal (i.p.) injection of a single dose of MCT (60 mg/kg), and CBX treatment (20 µg/kg/day, i.p.) was initiated on the day following MCT treatment for 28 days. Vehicle-treated male Sprague-Dawley rats were used as the negative control. The MCT rat model was evaluated by measuring the pulmonary artery flow acceleration time and right ventricular hypertrophy index (RVHI). Histopathological features of the lung tissues and pulmonary arteriolar remodeling were assessed. The proportions of T lymphocyte subtypes, Cx40/cx43 expression in the T cell subtypes and the cytokine levels in the plasma and the lung tissues were also analyzed. Pharmacological inhibition of Cxs using CBX attenuated MCT-induced right ventricular hypertrophy, pulmonary arteriolar remodeling, lung fibrosis and inflammatory cell infiltration by decreasing the RVHI, pulmonary arterial wall thickening, collagen deposition and pro-inflammatory cytokines production as well as CD3+ and CD4+ T cell accumulation in lung tissues of MCT-treated rats. Furthermore, flow cytometry analysis revealed that CBX may inhibit MCT-induced Cx40 and Cx43 expression in CD4+ and CD8+ T lymphocytes in lung tissues. The present study provides evidence that pharmacological inhibition of Cxs may attenuate MCT-induced pulmonary arteriolar remodeling and pulmonary inflammatory response, at least in part, by decreasing Cx expression. The results highlight the critical role of Cxs in T lymphocytes in the MCT-induced pulmonary inflammatory response and that targeting of Cxs may be a potential therapeutic method for treating pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Liang Ζ Zhang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Zhi-Ru Fan
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Lu Wang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Lu-Qian Liu
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Xin-Zhi Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Ke-Tao Ma
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
17
|
Martín Giménez VM, Díaz-Rodríguez P, Sanz RL, Vivero-Lopez M, Concheiro A, Diez E, Prado N, Enrique Kassuha D, Alvarez-Lorenzo C, Manucha W. Anandamide-nanoformulation obtained by electrospraying for cardiovascular therapy. Int J Pharm 2019; 566:1-10. [DOI: 10.1016/j.ijpharm.2019.05.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/20/2023]
|
18
|
Ni X, Zhang L, Ma X, Shan LY, Li L, Si JQ, Li XZ, Zhang YY, Ma KT. β‑estradiol alleviates hypertension‑ and concanavalin A‑mediated inflammatory responses via modulation of connexins in peripheral blood lymphocytes. Mol Med Rep 2019; 19:3743-3755. [PMID: 30896818 PMCID: PMC6471871 DOI: 10.3892/mmr.2019.10037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 03/08/2019] [Indexed: 12/24/2022] Open
Abstract
Gap junctions (GJs) formed by connexins (Cxs) in T lymphocytes have been reported to have important roles in the T lymphocyte-driven inflammatory response and hypertension-mediated inflammation. Estrogen has a protective effect on cardiovascular diseases, including hypertension and it attenuates excessive inflammatory responses in certain autoimmune diseases. However, the mechanisms involved in regulating the pro-inflammatory response are complex and poorly understood. The current study investigated whether β-estradiol suppresses hypertension and pro-inflammatory stimuli-mediated inflammatory responses by regulating Cxs and Cx-mediated GJs in peripheral blood lymphocytes. Male, 16-week-old spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY) rats were randomly divided into the following three groups: WKY rats, vehicle (saline)-treated SHRs, and β-estradiol (20 µg/kg/day)-treated SHRs. β-estradiol was administered subcutaneously for 5 weeks. Hematoxylin and eosin staining was performed to evaluate target organ injury. Flow cytometry and ELISA were used to measure the populations of T lymphocyte subtypes in the peripheral blood, and expression of Cx40/Cx43 in T cell subtypes, and pro-inflammation cytokines levels, respectively. ELISA, a dye transfer technique, immunofluorescence and immunoblotting were used to analyze the effect of β-estradiol on pro-inflammatory cytokine secretion, Cx-mediated GJs and the expression of Cxs in concanavalin A (Con A)-stimulated peripheral blood lymphocytes isolated from WKY rat. β-estradiol significantly decreased blood pressure and inhibited hypertension-induced target organ injury in SHRs. Additionally, β-estradiol treatment significantly improved the immune homeostasis of SHRs, as demonstrated by the decreased percentage of cluster of differentiation (CD)4+/CD8+ T-cell subset ratio, reduced serum levels of pro-inflammatory cytokines and increased the percentage of CD4+CD25+ T cells. β-estradiol also markedly reduced the expression of Cx40/Cx43 in T lymphocytes from SHRs. In vitro, β-estradiol significantly suppressed the production of pro-inflammatory cytokines, reduced communication via Cx-mediated gap junctions and decreased the expression of Cx40/Cx43 in Con A-stimulated lymphocytes. These results indicate that β-estradiol attenuates inflammation and end organ damage in hypertension, which may be partially mediated via downregulated expression of Cxs and reduced function of Cx-mediated GJ.
Collapse
Affiliation(s)
- Xin Ni
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Liang Zhang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Xin Ma
- Department of Anesthesiology, First Affiliated Hospital, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Li-Ya Shan
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Xin-Zhi Li
- Department of Pathophysiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - You-Yi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Ke-Tao Ma
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
19
|
Yu L, Hu P, Chen Y. Gas-Generating Nanoplatforms: Material Chemistry, Multifunctionality, and Gas Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801964. [PMID: 30066474 DOI: 10.1002/adma.201801964] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/26/2018] [Indexed: 05/16/2023]
Abstract
The fast advances of theranostic nanomedicine enable the rational design and construction of diverse functional nanoplatforms for versatile biomedical applications, among which gas-generating nanoplatforms (GGNs) have emerged very recently as unique theranostic nanoplatforms for broad gas therapies. Here, the recent developments of the rational design and chemical construction of versatile GGNs for efficient gas therapies by either exogenous physical triggers or endogenous disease-environment responsiveness are reviewed. These gases involve some therapeutic gases that can directly change disease status, such as oxygen (O2 ), nitric oxide (NO), carbon monoxide (CO), hydrogen (H2 ), hydrogen sulfide (H2 S) and sulfur dioxide (SO2 ), and other gases such as carbon dioxide (CO2 ), dl-menthol (DLM), and gaseous perfluorocarbon (PFC) for supplementary assistance of the theranostic process. Abundant nanocarriers have been adopted for gas delivery into lesions, including poly(d,l-lactic-co-glycolic acid), micelles, silica/mesoporous silica, organosilica, MnO2 , graphene, Bi2 Se3 , upconversion nanoparticles, CaCO3 , etc. Especially, these GGNs have been successfully developed for versatile biomedical applications, including diagnostic imaging and therapeutic use. The biosafety issue, challenges faced, and future developments on the rational construction of GGNs are also discussed for further promotion of their clinical translation to benefit patients.
Collapse
Affiliation(s)
- Luodan Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ping Hu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
20
|
Ni X, Li XZ, Fan ZR, Wang A, Zhang HC, Zhang L, LI L, Si JQ, Ma KT. Increased expression and functionality of the gap junction in peripheral blood lymphocytes is associated with hypertension-mediated inflammation in spontaneously hypertensive rats. Cell Mol Biol Lett 2018; 23:40. [PMID: 30151015 PMCID: PMC6102908 DOI: 10.1186/s11658-018-0106-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 08/06/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Imbalances in circulating T lymphocytes play critical roles in the pathogenesis of hypertension-mediated inflammation. Connexins (Cxs) in immune cells are involved in the maintenance of homeostasis of T lymphocytes. However, the association between Cxs in peripheral blood T lymphocytes and hypertension-mediated inflammation remains unknown. This study was designed to investigate the role of Cxs in T lymphocytes in hypertension-mediated inflammation in spontaneously hypertensive rats (SHRs). METHODS The systolic blood pressure (SBP) in Wistar-Kyoto (WKY) rats and SHRs was monitored using the tail-cuff method. The serum cytokine level was determined using ELISA. The proportions of different T-lymphocyte subtypes in the peripheral blood, the expressions of Cx40/Cx43 in the T-cell subtypes, and the gap junctional intracellular communication (GJIC) of peripheral blood lymphocytes were measured using flow cytometry (FC). The accumulations of Cx40/Cx43 at the plasma membrane and/or in the cytoplasm were determined using immunofluorescence staining. The in vitro mRNA levels of cytokines and GJIC in the peripheral blood lymphocytes were respectively examined using real-time PCR and FC after treatment with Gap27 and/or concanavalin A (Con A). RESULTS The percentage of CD4+ T cells and the CD4+/CD8+ ratio were high, and the accumulation or expressions of Cx40/Cx43 in the peripheral blood lymphocytes in SHRs were higher than in those of WKY rats. The percentage of CD8+ and CD4+CD25+ T cells was lower in SHRs. The serum levels of IL-2, IL-4 and IL-6 from SHRs were higher than those from WKY rats, and the serum levels of IL-2 and IL-6 positively correlated with the expression of Cx40/Cx43 in the peripheral blood T lymphocytes from SHRs. The peripheral blood lymphocytes of SHRs exhibited enhanced GJIC. Cx43-based channel inhibition, which was mediated by Gap27, remarkably reduced GJIC in lymphocytes, and suppressed IL-2 and IL-6 mRNA expressions in Con A stimulated peripheral blood lymphocytes. CONCLUSIONS Our data suggest that Cxs may be involved in the regulation of T-lymphocyte homeostasis and the production of cytokines. A clear association was found between alterations in Cxs expression or in Cx43-based GJIC and hypertension-mediated inflammation.
Collapse
Affiliation(s)
- Xin Ni
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Xin-zhi Li
- Department of Pathophysiology, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Zhi-ru Fan
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Ai Wang
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Hai-chao Zhang
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Liang Zhang
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Li LI
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Jun-qiang Si
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| | - Ke-tao Ma
- Department of Physiology, Medical College of Shihezi University, 59 North 2nd Road, Shihezi, Xinjiang 832002 People’s Republic of China
- Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang China
| |
Collapse
|