1
|
Zhang J, Lv W, Liu X, Sun Z, Zeng M, Kang J, Zhang Q, Liu F, Ma S, Su J, Cao K, Liu J. Ginsenoside Rh4 prevents endothelial dysfunction as a novel AMPK activator. Br J Pharmacol 2024; 181:3346-3363. [PMID: 38757416 DOI: 10.1111/bph.16403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND AND PURPOSE The AMP-activated protein kinase (AMPK) signalling pathway is a desirable target for various cardiovascular diseases (CVD), while the involvement of AMPK-mediated specific downstream pathways and effective interventions in hyperlipidaemia-induced endothelial dysfunction remain largely unknown. Herein, we aim to identify an effective AMPK activator and to explore its efficacy and mechanism against endothelial dysfunction. EXPERIMENTAL APPROACH Molecular docking technique was adopted to screen for the potent AMPK activator among 11 most common rare ginsenosides. In vivo, poloxamer 407 (P407) was used to induce acute hyperlipidaemia in C57BL/6J mice. In vitro, palmitic acid (PA) was used to induce lipid toxicity in HAEC cells. KEY RESULTS We discovered the strongest binding of ginsenoside Rh4 to AMPKα1 and confirmed the action of Rh4 on AMPK activation. Rh4 effectively attenuated hyperlipidaemia-related endothelial injury and oxidative stress both in vivo and in vitro and restored cell viability, mitochondrial membrane potential and mitochondrial oxygen consumption rate in HAEC cells. Mechanistically, Rh4 bound to AMPKα1 and simultaneously up-regulated AKT/eNOS-mediated NO release, promoted PGC-1α-mediated mitochondrial biogenesis and inhibited P38 MAPK/NFκB-mediated inflammatory responses in both P407-treated mice and PA-treated HAEC cells. The AMPK inhibitor Compound C treatment completely abrogated the regulation of Rh4 on the above pathways and weakened the lowering effect of Rh4 on endothelial impairment markers, suggesting that the beneficial effects of Rh4 are AMPK dependent. CONCLUSION AND IMPLICATIONS Rh4 may serve as a novel AMPK activator to protect against hyperlipidaemia-induced endothelial dysfunction, providing new insights into the prevention and treatment of endothelial injury-associated CVD.
Collapse
Affiliation(s)
- Jiawei Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Weiqiang Lv
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuyun Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhenyu Sun
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mengqi Zeng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Jiahao Kang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qi Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fuying Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shaozhou Ma
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiacan Su
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ke Cao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| |
Collapse
|
2
|
Silva-Cunha M, Lacchini R, Tanus-Santos JE. Facilitating Nitrite-Derived S-Nitrosothiol Formation in the Upper Gastrointestinal Tract in the Therapy of Cardiovascular Diseases. Antioxidants (Basel) 2024; 13:691. [PMID: 38929130 PMCID: PMC11200996 DOI: 10.3390/antiox13060691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are often associated with impaired nitric oxide (NO) bioavailability, a critical pathophysiological alteration in CVDs and an important target for therapeutic interventions. Recent studies have revealed the potential of inorganic nitrite and nitrate as sources of NO, offering promising alternatives for managing various cardiovascular conditions. It is now becoming clear that taking advantage of enzymatic pathways involved in nitrite reduction to NO is very relevant in new therapeutics. However, recent studies have shown that nitrite may be bioactivated in the acidic gastric environment, where nitrite generates NO and a variety of S-nitrosating compounds that result in increased circulating S-nitrosothiol concentrations and S-nitrosation of tissue pharmacological targets. Moreover, transnitrosation reactions may further nitrosate other targets, resulting in improved cardiovascular function in patients with CVDs. In this review, we comprehensively address the mechanisms and relevant effects of nitrate and nitrite-stimulated gastric S-nitrosothiol formation that may promote S-nitrosation of pharmacological targets in various CVDs. Recently identified interfering factors that may inhibit these mechanisms and prevent the beneficial responses to nitrate and nitrite therapy were also taken into consideration.
Collapse
Affiliation(s)
- Mila Silva-Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil;
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto 14040-902, Brazil;
| | - Jose E. Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil;
| |
Collapse
|
3
|
Rao J, Mou X, Mo Y, Bei HP, Wang L, Tang CY, Yiu KH, Yang Z, Zhao X. Gas station in blood vessels: An endothelium mimicking, self-sustainable nitric oxide fueling stent coating for prevention of thrombosis and restenosis. Biomaterials 2023; 302:122311. [PMID: 37677916 DOI: 10.1016/j.biomaterials.2023.122311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Stenting is the primary treatment for vascular obstruction-related cardiovascular diseases, but it inevitably causes endothelial injury which may lead to severe thrombosis and restenosis. Maintaining nitric oxide (NO, a vasoactive mediator) production and grafting endothelial glycocalyx such as heparin (Hep) onto the surface of cardiovascular stents could effectively reconstruct the damaged endothelium. However, insufficient endogenous NO donors may impede NO catalytic generation and fail to sustain cardiovascular homeostasis. Here, a dopamine-copper (DA-Cu) network-based coating armed with NO precursor L-arginine (Arg) and Hep (DA-Cu-Arg-Hep) is prepared using an organic solvent-free dipping technique to form a nanometer-thin coating onto the cardiovascular stents. The DA-Cu network adheres tightly to the surface of stents and confers excellent NO catalytic activity in the presence of endogenous NO donors. The immobilized Arg functions as a NO fuel to generate NO via endothelial nitric oxide synthase (eNOS), while Hep works as eNOS booster to increase the level of eNOS to decompose Arg into NO, ensuring a sufficient supply of NO even when endogenous donors are insufficient. The synergistic interaction between Cu and Arg is analogous to a gas station to fuel NO production to compensate for the insufficient endogenous NO donor in vivo. Consequently, it promotes the reconstruction of natural endothelium, inhibits smooth muscle cell (SMC) migration, and suppresses cascading platelet adhesion, preventing stent thrombosis and restenosis. We anticipate that our DA-Cu-Arg-Hep coating will improve the quality of life of cardiovascular patients through improved surgical follow-up, increased safety, and decreased medication, as well as revitalize the stenting industry through durable designs.
Collapse
Affiliation(s)
- Jingdong Rao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Xiaohui Mou
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong 523000, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, Guangdong, China
| | - Yongyi Mo
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Ho-Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Li Wang
- Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Island, Hong Kong SAR, China
| | - Chuyang Y Tang
- Department of Civil Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Island, Hong Kong SAR, China
| | - Kai-Hang Yiu
- Cardiology Division, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong Island, Hong Kong SAR, China
| | - Zhilu Yang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong 523000, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, Guangdong, China.
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Applied Biology and Chemical Technology, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China.
| |
Collapse
|
4
|
Phua TJ. Understanding human aging and the fundamental cell signaling link in age-related diseases: the middle-aging hypovascularity hypoxia hypothesis. FRONTIERS IN AGING 2023; 4:1196648. [PMID: 37384143 PMCID: PMC10293850 DOI: 10.3389/fragi.2023.1196648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 06/30/2023]
Abstract
Aging-related hypoxia, oxidative stress, and inflammation pathophysiology are closely associated with human age-related carcinogenesis and chronic diseases. However, the connection between hypoxia and hormonal cell signaling pathways is unclear, but such human age-related comorbid diseases do coincide with the middle-aging period of declining sex hormonal signaling. This scoping review evaluates the relevant interdisciplinary evidence to assess the systems biology of function, regulation, and homeostasis in order to discern and decipher the etiology of the connection between hypoxia and hormonal signaling in human age-related comorbid diseases. The hypothesis charts the accumulating evidence to support the development of a hypoxic milieu and oxidative stress-inflammation pathophysiology in middle-aged individuals, as well as the induction of amyloidosis, autophagy, and epithelial-to-mesenchymal transition in aging-related degeneration. Taken together, this new approach and strategy can provide the clarity of concepts and patterns to determine the causes of declining vascularity hemodynamics (blood flow) and physiological oxygenation perfusion (oxygen bioavailability) in relation to oxygen homeostasis and vascularity that cause hypoxia (hypovascularity hypoxia). The middle-aging hypovascularity hypoxia hypothesis could provide the mechanistic interface connecting the endocrine, nitric oxide, and oxygen homeostasis signaling that is closely linked to the progressive conditions of degenerative hypertrophy, atrophy, fibrosis, and neoplasm. An in-depth understanding of these intrinsic biological processes of the developing middle-aged hypoxia could provide potential new strategies for time-dependent therapies in maintaining healthspan for healthy lifestyle aging, medical cost savings, and health system sustainability.
Collapse
Affiliation(s)
- Teow J. Phua
- Molecular Medicine, NSW Health Pathology, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
5
|
Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol 2023; 20:109-125. [PMID: 35931887 PMCID: PMC9362470 DOI: 10.1038/s41569-022-00741-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 01/21/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as a gaseous signalling molecule with crucial implications for cardiovascular health. H2S is involved in many biological functions, including interactions with nitric oxide, activation of molecular signalling cascades, post-translational modifications and redox regulation. Various preclinical and clinical studies have shown that H2S and its synthesizing enzymes - cystathionine γ-lyase, cystathionine β-synthase and 3-mercaptosulfotransferase - can protect against cardiovascular pathologies, including arrhythmias, atherosclerosis, heart failure, myocardial infarction and ischaemia-reperfusion injury. The bioavailability of H2S and its metabolites, such as hydropersulfides and polysulfides, is substantially reduced in cardiovascular disease and has been associated with single-nucleotide polymorphisms in H2S synthesis enzymes. In this Review, we highlight the role of H2S, its synthesizing enzymes and metabolites, their roles in the cardiovascular system, and their involvement in cardiovascular disease and associated pathologies. We also discuss the latest clinical findings from the field and outline areas for future study.
Collapse
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Paari Dominic
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
6
|
Zhang Y, Jing M, Cai C, Zhu S, Zhang C, Wang Q, Zhai Y, Ji X, Wu D. Role of hydrogen sulphide in physiological and pathological angiogenesis. Cell Prolif 2022; 56:e13374. [PMID: 36478328 PMCID: PMC9977675 DOI: 10.1111/cpr.13374] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The role of hydrogen sulphide (H2 S) in angiogenesis has been widely demonstrated. Vascular endothelial growth factor (VEGF) plays an important role in H2 S-induced angiogenesis. H2 S promotes angiogenesis by upregulating VEGF via pro-angiogenic signal transduction. The involved signalling pathways include the mitogen-activated protein kinase pathway, phosphoinositide-3 kinase pathway, nitric oxide (NO) synthase/NO pathway, signal transducer and activator of transcription 3 (STAT3) pathway, and adenosine triphosphate (ATP)-sensitive potassium (KATP ) channels. H2 S has been shown to contribute to tumour angiogenesis, diabetic wound healing, angiogenesis in cardiac and cerebral ischaemic tissues, and physiological angiogenesis during the menstrual cycle and pregnancy. Furthermore, H2 S can exert an anti-angiogenic effect by inactivating Wnt/β-catenin signalling or blocking the STAT3 pathway in tumours. Therefore, H2 S plays a double-edged sword role in the process of angiogenesis. The regulation of H2 S production is a promising therapeutic approach for angiogenesis-associated diseases. Novel H2 S donors and/or inhibitors can be developed in the treatment of angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Yan‐Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Mi‐Rong Jing
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Chun‐Bo Cai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Shuai‐Gang Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Chao‐Jing Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Qi‐Meng Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Yuan‐Kun Zhai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,School of StomatologyHenan UniversityKaifengHenanChina
| | - Xin‐Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina,Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical SciencesHenan UniversityKaifengHenanChina
| | - Dong‐Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina,School of StomatologyHenan UniversityKaifengHenanChina
| |
Collapse
|
7
|
Macabrey D, Joniová J, Gasser Q, Bechelli C, Longchamp A, Urfer S, Lambelet M, Fu CY, Schwarz G, Wagnières G, Déglise S, Allagnat F. Sodium thiosulfate, a source of hydrogen sulfide, stimulates endothelial cell proliferation and neovascularization. Front Cardiovasc Med 2022; 9:965965. [PMID: 36262202 PMCID: PMC9575962 DOI: 10.3389/fcvm.2022.965965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Therapies to accelerate vascular repair are currently lacking. Pre-clinical studies suggest that hydrogen sulfide (H2S), an endogenous gasotransmitter, promotes angiogenesis. Here, we hypothesized that sodium thiosulfate (STS), a clinically relevant source of H2S, would stimulate angiogenesis and vascular repair. STS stimulated neovascularization in WT and LDLR receptor knockout mice following hindlimb ischemia as evidenced by increased leg perfusion assessed by laser Doppler imaging, and capillary density in the gastrocnemius muscle. STS also promoted VEGF-dependent angiogenesis in matrigel plugs in vivo and in the chorioallantoic membrane of chick embryos. In vitro, STS and NaHS stimulated human umbilical vein endothelial cell (HUVEC) migration and proliferation. Seahorse experiments further revealed that STS inhibited mitochondrial respiration and promoted glycolysis in HUVEC. The effect of STS on migration and proliferation was glycolysis-dependent. STS probably acts through metabolic reprogramming of endothelial cells toward a more proliferative glycolytic state. These findings may hold broad clinical implications for patients suffering from vascular occlusive diseases.
Collapse
Affiliation(s)
- Diane Macabrey
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Jaroslava Joniová
- Laboratory for Functional and Metabolic Imaging, LIFMET, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Quentin Gasser
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Clémence Bechelli
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Severine Urfer
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Martine Lambelet
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Chun-Yu Fu
- Institute of Biochemistry, Department of Chemistry & Center for Molecular Medicine, Cologne University, Cologne, Germany
| | - Guenter Schwarz
- Institute of Biochemistry, Department of Chemistry & Center for Molecular Medicine, Cologne University, Cologne, Germany
| | - Georges Wagnières
- Laboratory for Functional and Metabolic Imaging, LIFMET, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland,*Correspondence: Florent Allagnat,
| |
Collapse
|
8
|
Chen Z, Haus JM, Chen L, Jiang Y, Sverdlov M, DiPietro LA, Xiong N, Wu SC, Koh TJ, Minshall RD. Inhibition of CCL28/CCR10-Mediated eNOS Downregulation Improves Skin Wound Healing in the Obesity-Induced Mouse Model of Type 2 Diabetes. Diabetes 2022; 71:2166-2180. [PMID: 35899992 PMCID: PMC9501665 DOI: 10.2337/db21-1108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/21/2022] [Indexed: 11/13/2022]
Abstract
Chronic, nonhealing skin wounds, such as diabetic foot ulcers (DFUs), are common in patients with type 2 diabetes. Here, we investigated the role of chemokine (C-C motif) ligand 28 (CCL28) and its receptor C-C chemokine receptor type 10 (CCR10) in downregulation of endothelial nitric (NO) oxide synthase (eNOS) in association with delayed skin wound healing in the db/db mouse model of type 2 diabetes. We observed reduced eNOS expression and elevated CCL28/CCR10 levels in dorsal skin of db/db mice and subdermal leg biopsy specimens from human subjects with type 2 diabetes. Further interrogation revealed that overexpression of CCR10 reduced eNOS expression, NO bioavailability, and tube formation of human dermal microvascular endothelial cells (HDMVECs) in vitro, which was recapitulated in mouse dorsal skin. In addition, incubation of HDMVECs with CCL28 led to internalization of the CCR10/eNOS complex and colocalization with lysosome-associated membrane protein 1. Finally, topical application of myristoylated CCR10 binding domain 7 amino acid (Myr-CBD7) peptide prevented CCR10-eNOS interaction and subsequent eNOS downregulation, enhanced eNOS/NO levels, eNOS/VEGF-R2+ microvessel density, and blood perfusion, reduced inflammatory cytokine levels, and importantly, decreased wound healing time in db/db mice. Thus, endothelial cell CCR10 activation in genetically obese mice with type 2 diabetes promotes eNOS depletion and endothelial dysfunction, and targeted disruption of CCR10/eNOS interaction improves wound healing.
Collapse
Affiliation(s)
- Zhenlong Chen
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL
| | - Jacob M. Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Lin Chen
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
| | - Ying Jiang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| | - Maria Sverdlov
- Research Resources Center, Research Histology and Tissue Imaging Collaborative, University of Illinois at Chicago, Chicago, IL
| | - Luisa A. DiPietro
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
| | - Na Xiong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Stephanie C. Wu
- Departments of Surgery and Stem Cell and Regenerative Medicine, Center for Lower Extremity Ambulatory Research, Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL
| | - Timothy J. Koh
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL
| | - Richard D. Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
9
|
He M, Wang D, Xu Y, Jiang F, Zheng J, Feng Y, Cao J, Zhou X. Nitric Oxide-Releasing Platforms for Treating Cardiovascular Disease. Pharmaceutics 2022; 14:pharmaceutics14071345. [PMID: 35890241 PMCID: PMC9317153 DOI: 10.3390/pharmaceutics14071345] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the first leading cause of death globally. Nitric oxide (NO) is an important signaling molecule that mediates diverse processes in the cardiovascular system, thereby providing a fundamental basis for NO-based therapy of CVD. At present, numerous prodrugs have been developed to release NO in vivo. However, the clinical application of these prodrugs still faces many problems, including the low payloads, burst release, and non-controlled delivery. To address these, various biomaterial-based platforms have been developed as the carriers to deliver NO to the targeted tissues in a controlled and sustained manner. This review aims to summarize recent developments of various therapeutic platforms, engineered to release NO for the treatment of CVD. In addition, two potential strategies to improve the effectiveness of existing NO therapy are also discussed, including the combination of NO-releasing platforms and either hydrogen sulfide-based therapy or stem cell therapy. Hopefully, some NO-releasing platforms may provide important therapeutic benefits for CVD.
Collapse
Affiliation(s)
- Mingyue He
- Department of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China; (M.H.); (Y.X.)
| | - Deping Wang
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, Shanxi Medical University, Taiyuan 030001, China; (D.W.); (F.J.); (J.Z.)
| | - Yumei Xu
- Department of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China; (M.H.); (Y.X.)
| | - Fangying Jiang
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, Shanxi Medical University, Taiyuan 030001, China; (D.W.); (F.J.); (J.Z.)
| | - Jian Zheng
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, Shanxi Medical University, Taiyuan 030001, China; (D.W.); (F.J.); (J.Z.)
- Department of Breast Surgery, Shanxi Provincial Cancer Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Yanlin Feng
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, Shanxi Medical University, Taiyuan 030001, China; (D.W.); (F.J.); (J.Z.)
- Correspondence: (Y.F.); (J.C.); (X.Z.)
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, Shanxi Medical University, Taiyuan 030001, China; (D.W.); (F.J.); (J.Z.)
- Correspondence: (Y.F.); (J.C.); (X.Z.)
| | - Xin Zhou
- Department of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China; (M.H.); (Y.X.)
- Key Laboratory of Cellular Physiology, Ministry of Education, The Department of Physiology, Shanxi Medical University, Taiyuan 030001, China; (D.W.); (F.J.); (J.Z.)
- Correspondence: (Y.F.); (J.C.); (X.Z.)
| |
Collapse
|
10
|
Long K, Yang Y, Du Z, Kang W, Lv W, Li Y, Xie Y, Sun H, Zhan C, Wang W. H
2
S‐Responsive Small‐Molecule Nanocarriers for Drug Delivery to Colorectal Tumors. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kaiqi Long
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak‐Sum Research Centre & Department of Pharmacology and Pharmacy Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Yang Yang
- School of Pharmacy & Key Laboratory of Smart Drug Delivery Ministry of Education Fudan University Shanghai China
| | - Zhanfeng Du
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak‐Sum Research Centre & Department of Pharmacology and Pharmacy Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Weirong Kang
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak‐Sum Research Centre & Department of Pharmacology and Pharmacy Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Wen Lv
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak‐Sum Research Centre & Department of Pharmacology and Pharmacy Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Yafei Li
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak‐Sum Research Centre & Department of Pharmacology and Pharmacy Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Yusheng Xie
- Department of Pharmacology School of Basic Medical Sciences Shandong University Jinan China
| | - Hongyan Sun
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong Kong Hong Kong SAR China
| | - Changyou Zhan
- School of Pharmacy & Key Laboratory of Smart Drug Delivery Ministry of Education Fudan University Shanghai China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak‐Sum Research Centre & Department of Pharmacology and Pharmacy Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| |
Collapse
|
11
|
H 2O 2-responsive VEGF/NGF gene co-delivery nano-system achieves stable vascularization in ischemic hindlimbs. J Nanobiotechnology 2022; 20:145. [PMID: 35305670 PMCID: PMC8934504 DOI: 10.1186/s12951-022-01328-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/24/2022] [Indexed: 12/02/2022] Open
Abstract
Peripheral vascular disease (PVD) is a common clinical manifestation of atherosclerosis. Vascular endothelial growth factor (VEGF) gene therapy is a promising approach for PVD treatment. However, due to single-gene therapy limitations and high H2O2 pathological microenvironment, VEGF gene therapy are not as expectations and its clinical application are limited. Synergistic effects of Nerve factors and vascular factors in angiogenesis have attracted attention in recent years. In this study, VEGF and nerve growth factor (NGF) genes co-delivery nanoparticles (VEGF/NGF-NPs) were prepared by using H2O2 responsive 6s-PLGA-Po-PEG as a carrier. 6s-PLGA-Po-PEG could react with H2O2 specifically due to the internal peroxalate bond. Angiogenic effects of VEGF/NGF-NPs has been evaluated in cells and hindlimb ischemia mice model. Results showed that VEGF/NGF-NPs promoted VEGF and NGF co-expression simultaneously, eliminated excessive H2O2, strengthened reactions between SH-SY5Ys and HUVECs, and finally enhanced migration, tube formation, proliferation and H2O2 damage resistance of HUVECs. VEGF/NGF-NPs also recovered blood perfusion, promoted the expression of VEGF, NGF, eNOS and NO, and enhanced vascular coverage of pericytes. Treatment effects of VEGF/NGF-NPs may related to VEGF/eNOS/NO pathway. Altogether, VEGF/NGF-NPs eliminated excessive H2O2 while achieving gene co-delivery, and promoted stable angiogenesis. It’s a promising way for PVD treatment by using VEGF/NGF-NPs.
Collapse
|
12
|
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
|
13
|
Scrivner O, Ismaeel A, Kumar MR, Sorokolet K, Koutakis P, Farmer PJ. Expanding the Reactive Sulfur Metabolome: Intracellular and Efflux Measurements of Small Oxoacids of Sulfur (SOS) and H 2S in Human Primary Vascular Cell Culture. Molecules 2021; 26:7160. [PMID: 34885743 PMCID: PMC8659008 DOI: 10.3390/molecules26237160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/29/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous signaling molecule which is important for cardiovascular health, but its mechanism of action remains poorly understood. Here, we report measurements of H2S as well as its oxidized metabolites, termed small oxoacids of sulfur (SOS = HSOH and HOSOH), in four human primary vascular cell lines: smooth muscle and endothelial cells derived from both human arterial and coronary tissues. We use a methodology that targets small molecular weight sulfur species; mass spectrometric analysis allows for species quantification to report cellular concentrations based on an H2S calibration curve. The production of H2S and SOS is orders of magnitude higher in smooth muscle (nanomolar) as compared to endothelial cell lines (picomolar). In all the primary lines measured, the distributions of these three species were HOSOH >H2S > HSOH, with much higher SOS than seen previously in non-vascular cell lines. H2S and SOS were effluxed from smooth muscle cells in higher concentrations than endothelial cells. Aortic smooth muscle cells were used to examine changes under hypoxic growth conditions. Hypoxia caused notable increases in HSOH and ROS, which we attribute to enhanced sulfide quinone oxidase activity that results in reverse electron transport.
Collapse
Affiliation(s)
- Ottis Scrivner
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (O.S.); (M.R.K.); (K.S.)
| | - Ahmed Ismaeel
- Department of Biology, Baylor University, Waco, TX 76798, USA; (A.I.); (P.K.)
| | - Murugaeson R. Kumar
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (O.S.); (M.R.K.); (K.S.)
| | - Kristina Sorokolet
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (O.S.); (M.R.K.); (K.S.)
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, Waco, TX 76798, USA; (A.I.); (P.K.)
| | - Patrick J. Farmer
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (O.S.); (M.R.K.); (K.S.)
| |
Collapse
|
14
|
Modulatory effects of BPC 157 on vasomotor tone and the activation of Src-Caveolin-1-endothelial nitric oxide synthase pathway. Sci Rep 2020; 10:17078. [PMID: 33051481 PMCID: PMC7555539 DOI: 10.1038/s41598-020-74022-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
BPC 157-activated endothelial nitric oxide synthase (eNOS) is associated with tissue repair and angiogenesis as reported in previous studies. However, how BPC 157 regulates the vasomotor tone and intracellular Src-Caveolin-1 (Cav-1)-eNOS signaling is not yet clear. The present study demonstrated a concentration-dependent vasodilation effect of BPC 157 in isolated rat aorta. Attenuation of this vasodilation effect in the absence of endothelium suggested an endothelium-dependent vasodilation effect of BPC 157. Although slightly increased vasorelaxation in aorta without endothelium was noticed at high concentration of BPC 157, there was no direct relaxation effect on three-dimensional model made of vascular smooth muscle cells. The vasodilation effect of BPC 157 was nitric oxide mediated because the addition of L-NAME or hemoglobin inhibited the vasodilation of aorta. Nitric oxide generation was induced by BPC 157 as detected by intracellular DFA-FM DA labeling which was capable of promoting the migration of vascular endothelial cells. BPC 157 enhanced the phosphorylation of Src, Cav-1 and eNOS which was abolished by pretreatment with Src inhibitor, confirming the upstream role of Src in this signal pathway. Activation of eNOS required the released binding with Cav-1 in advance. Co-immunoprecipitation analysis revealed that BPC 157 could reduce the binding between Cav-1 and eNOS. Together, the present study demonstrates that BPC 157 can modulate the vasomotor tone of an isolated aorta in a concentration- and nitric oxide-dependent manner. BPC 157 can induce nitric oxide generation likely through the activation of Src-Cav-1-eNOS pathway.
Collapse
|
15
|
Pardue S, Kolluru GK, Shen X, Lewis SE, Saffle CB, Kelley EE, Kevil CG. Hydrogen sulfide stimulates xanthine oxidoreductase conversion to nitrite reductase and formation of NO. Redox Biol 2020; 34:101447. [PMID: 32035920 PMCID: PMC7327988 DOI: 10.1016/j.redox.2020.101447] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular disease is the leading cause of death and disability worldwide with increased oxidative stress and reduced NO bioavailability serving as key risk factors. For decades, elevation in protein abundance and enzymatic activity of xanthine oxidoreductase (XOR) under hypoxic/inflammatory conditions has been associated with organ damage and vascular dysfunction. Recent reports have challenged this dogma by identifying a beneficial function for XOR, under similar hypoxic/acidic conditions, whereby XOR catalyzes the reduction of nitrite (NO2-) to nitric oxide (NO) through poorly defined mechanisms. We previously reported that hydrogen sulfide (H2S/sulfide) confers significant vascular benefit under these same conditions via NO2- mediated mechanisms independent of nitric oxide synthase (NOS). Here we report for the first time the convergence of H2S, XOR, and nitrite to form a concerted triad for NO generation. Specifically, hypoxic endothelial cells show a dose-dependent, sulfide and polysulfide (diallyl trisulfide (DATS)-induced, NOS-independent NO2- reduction to NO that is dependent upon the enzymatic activity of XOR. Interestingly, nitrite reduction to NO was found to be slower and more sustained with DATS compared to H2S. Capacity for sulfide/polysulfide to produce an XOR-dependent impact on NO generation translates to salutary actions in vivo as DATS administration in cystathionine-γ-lyase (CSE) knockout mice significantly improved hindlimb ischemia blood flow post ligation, while the XOR-specific inhibitor, febuxostat (Febx), abrogated this benefit. Moreover, flow-mediated vasodilation (FMD) in CSE knockout mice following administration of DATS resulted in greater than 4-fold enhancement in femoral artery dilation while co-treatment with Febx completely completely abrogated this effect. Together, these results identify XOR as a focal point of convergence between sulfide- and nitrite-mediated signaling, as well as affirm the critical need to reexamine current dogma regarding inhibition of XOR in the context of vascular dysfunction.
Collapse
Affiliation(s)
- Sibile Pardue
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Gopi K Kolluru
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Xinggui Shen
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Sara E Lewis
- Department of Physiology and Pharmacology, West Virginia University, United States
| | - Courtney B Saffle
- Department of Physiology and Pharmacology, West Virginia University, United States
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University, United States
| | - Christopher G Kevil
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA; Department of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport, LA, USA; Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
16
|
Morales-Loredo H, Barrera A, Garcia JM, Pace CE, Naik JS, Gonzalez Bosc LV, Kanagy NL. Hydrogen sulfide regulation of renal and mesenteric blood flow. Am J Physiol Heart Circ Physiol 2019; 317:H1157-H1165. [PMID: 31625777 DOI: 10.1152/ajpheart.00303.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hydrogen sulfide (H2S) dilates isolated arteries, and knockout of the H2S-synthesizing enzyme cystathionine γ-lyase (CSE) increases blood pressure. However, the contributions of endogenously produced H2S to blood flow regulation in specific vascular beds are unknown. Published studies in isolated arteries show that CSE production of H2S influences vascular tone more in small mesenteric arteries than in renal arteries or the aorta. Therefore, the goal of this study was to evaluate H2S regulation of blood pressure, vascular resistance, and regional blood flows using chronically instrumented rats. We hypothesized that during whole animal CSE inhibition, vascular resistance would increase more in the mesenteric than the renal circulation. Under anesthesia, CSE inhibition [β-cyanoalanine (BCA), 30 mg/kg bolus + 5 mg·kg-1·min-1 for 20 min iv) rapidly increased mean arterial pressure (MAP) more than saline administration (%Δ: saline -1.4 ± 0.75 vs. BCA 7.1 ± 1.69, P < 0.05) but did not change resistance (MAP/flow) in either the mesenteric or renal circulation. In conscious rats, BCA infusion similarly increased MAP (%Δ: saline -0.8 ± 1.18 vs. BCA 8.2 ± 2.6, P < 0.05, n = 7) and significantly increased mesenteric resistance (saline 0.9 ± 3.1 vs. BCA 15.6 ± 6.5, P < 0.05, n = 12). The H2S donor Na2S (50 mg/kg) decreased blood pressure and mesenteric resistance ,but the fall in resistance was not significant. Inhibiting CSE for multiple days with dl-proparglycine (PAG, 50 mg·kg-1·min-1 iv bolus for 5 days) significantly increased vascular resistance in both mesenteric (ratio of day 1: saline 0.86 ± 0.033 vs. PAG 1.79 ± 0.38) and renal circulations (ratio of day 1: saline 1.26 ± 0.22 vs. 1.98 ± 0.14 PAG). These results support our hypothesis that CSE-derived H2S is an important regulator of blood pressure and vascular resistance in both mesenteric and renal circulations. Furthermore, inhalation anesthesia diminishes the effect of CSE inhibition on vascular tone.NEW & NOTEWORTHY These results suggest that CSE-derived H2S has a prominent role in regulating blood pressure and blood flow under physiological conditions, which may have been underestimated in prior studies in anesthetized subjects. Therefore, enhancing substrate availability or enzyme activity or dosing with H2S donors could be a novel therapeutic approach to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Humberto Morales-Loredo
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Adelaeda Barrera
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Joshua M Garcia
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Carolyn E Pace
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Nancy L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
17
|
Rajendran S, Shen X, Glawe J, Kolluru GK, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Growth and Remodeling. Compr Physiol 2019; 9:1213-1247. [PMID: 31187898 DOI: 10.1002/cphy.c180026] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic vascular remodeling occurs in response to stenosis or arterial occlusion leading to a change in blood flow and tissue perfusion. Altered blood flow elicits a cascade of molecular and cellular physiological responses leading to vascular remodeling of the macro- and micro-circulation. Although cellular mechanisms of vascular remodeling such as arteriogenesis and angiogenesis have been studied, therapeutic approaches in these areas have had limited success due to the complexity and heterogeneous constellation of molecular signaling events regulating these processes. Understanding central molecular players of vascular remodeling should lead to a deeper understanding of this response and aid in the development of novel therapeutic strategies. Hydrogen sulfide (H2 S) and nitric oxide (NO) are gaseous signaling molecules that are critically involved in regulating fundamental biochemical and molecular responses necessary for vascular growth and remodeling. This review examines how NO and H2 S regulate pathophysiological mechanisms of angiogenesis and arteriogenesis, along with important chemical and experimental considerations revealed thus far. The importance of NO and H2 S bioavailability, their synthesis enzymes and cofactors, and genetic variations associated with cardiovascular risk factors suggest that they serve as pivotal regulators of vascular remodeling responses. © 2019 American Physiological Society. Compr Physiol 9:1213-1247, 2019.
Collapse
Affiliation(s)
| | - Xinggui Shen
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - John Glawe
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Gopi K Kolluru
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Christopher G Kevil
- Departments of Pathology, LSU Health Sciences Center, Shreveport.,Departments of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport.,Departments of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport
| |
Collapse
|
18
|
Grambow E, Augustin VA, Strüder D, Kundt G, Klar E, Vollmar B. The effects of hydrogen sulfide on microvascular circulation in the axial pattern flap ear model in hairless mice. Microvasc Res 2018; 120:74-83. [PMID: 29991448 DOI: 10.1016/j.mvr.2018.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 01/30/2023]
Affiliation(s)
- Eberhard Grambow
- Institute for Experimental Surgery, University Medical Center Rostock, Rostock, Germany; Department for General, Thoracic-, Vascular- and Transplantation Surgery, University Medical Center Rostock, Rostock, Germany.
| | - Vicky A Augustin
- Institute for Experimental Surgery, University Medical Center Rostock, Rostock, Germany
| | - Daniel Strüder
- Institute for Experimental Surgery, University Medical Center Rostock, Rostock, Germany; Department of Otorhinolaryngology, Head and Neck Surgery, Rostock University Medical Center, Rostock, Germany
| | - Günther Kundt
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Ernst Klar
- Department for General, Thoracic-, Vascular- and Transplantation Surgery, University Medical Center Rostock, Rostock, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, University Medical Center Rostock, Rostock, Germany
| |
Collapse
|
19
|
Cross-talk between endogenous H 2S and NO accounts for vascular protective activity of the metal-nonoate Zn(PipNONO)Cl. Biochem Pharmacol 2018; 152:143-152. [PMID: 29588193 DOI: 10.1016/j.bcp.2018.03.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/22/2018] [Indexed: 02/08/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are now recognized as gaseous transmitters with many cardiovascular protective properties. The present study concerns the possibility that NO donors can also function through endogenous activation of NO and H2S pathways. Based on the previous characterization of a novel metal-nonoate, Ni(PipNONO)Cl, our aim was: 1) to study the effects of a zinc based compound, Zn(PipNONO)Cl, on vascular endothelial and smooth muscle cells, and 2) to assess the role and interplay between endogenous NO and H2S promoted by the nonoate. Zn(PipNONO)Cl completely reproduced the vasodilation elicited by Ni(PipNONO)Cl. In the presence of endothelium, preincubation with Zn(PipNONO)Cl sensitized the intima to acetylcholine-induced vasodilation. When tested on cultured endothelial cells, Zn(PipNONO)Cl prompted PI-3K/Akt- and MAPK/ERK1/2-mediated survival. Nitrite levels indicated fast NO release (due to the molecule) and delayed (1-6 h) NO production linked to PI-3K/Akt-dependent eNOS activation. In the same time frame (1-6 h), significant CSE-dependent H2S levels were detected in response to Zn(PipNONO)Cl. The mechanisms responsible for H2S increase seemed to depend on the NONO moiety/sGC/cGMP pathway and zinc-associated ROS production. Our results indicate that endogenous H2S and NO were produced after fast NO release from Zn(PipNONO)Cl, contributing to the vascular endothelium protective effect. The effect was partially reproduced on smooth muscle cells, where Zn(PipNONO)Cl inhibited cell proliferation and migration. In conclusion, vasorelaxant effects, with complementary activities on endothelium and smooth muscle cells, are elicited by the novel metal-nonoate Zn(PipNONO)Cl.
Collapse
|
20
|
Khaddaj Mallat R, Mathew John C, Kendrick DJ, Braun AP. The vascular endothelium: A regulator of arterial tone and interface for the immune system. Crit Rev Clin Lab Sci 2017; 54:458-470. [PMID: 29084470 DOI: 10.1080/10408363.2017.1394267] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
As the primary interface between the blood and various tissues of the body, the vascular endothelium exhibits a diverse range of roles and activities, all of which contribute to the overall health and function of the cardiovascular system. In this focused review, we discuss several key aspects of endothelial function, how this may be compromised and subsequent consequences. Specifically, we examine the dynamic regulation of arterial contractility and distribution of blood flow through the generation of chemical and electrical signaling events that impinge upon vascular smooth muscle. The endothelium can generate a diverse range of vasoactive compounds and signals, most of which act locally to adjust blood flow in a dynamic fashion to match tissue metabolism. Disruption of these vascular signaling processes (e.g. reduced nitric oxide bioavailability) is typically referred to as endothelial dysfunction, which is a recognized risk factor for cardiovascular disease in patients and occurs early in the development and progression of hypertension, atherosclerosis and tissue ischemia. Endothelial dysfunction is also associated with type-2 Diabetes and aging and increased mechanistic knowledge of the cellular changes contributing to these effects may provide important clues for interventional strategies. The endothelium also serves as the initial site of interaction for immune cells entering tissues in response to damage and acts to facilitate the actions of both the innate and acquired immune systems to interact with the vascular wall. In addition to representing the main cell type responsible for the formation of new blood vessels (i.e. angiogenesis) within the vasculature, the endothelium is also emerging as a source of extracellular vesicle or microparticles for the transport of signaling molecules and other cellular materials to nearby, or remote, sites in the body. The characteristics of released microparticles appear to change with the functional status of the endothelium; thus, these microparticles may represent novel biomarkers of endothelial health and more serious cardiovascular disease.
Collapse
Affiliation(s)
- Rayan Khaddaj Mallat
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Cini Mathew John
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Dylan J Kendrick
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Andrew P Braun
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| |
Collapse
|
21
|
Szabo C, Papapetropoulos A. International Union of Basic and Clinical Pharmacology. CII: Pharmacological Modulation of H 2S Levels: H 2S Donors and H 2S Biosynthesis Inhibitors. Pharmacol Rev 2017; 69:497-564. [PMID: 28978633 PMCID: PMC5629631 DOI: 10.1124/pr.117.014050] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Over the last decade, hydrogen sulfide (H2S) has emerged as an important endogenous gasotransmitter in mammalian cells and tissues. Similar to the previously characterized gasotransmitters nitric oxide and carbon monoxide, H2S is produced by various enzymatic reactions and regulates a host of physiologic and pathophysiological processes in various cells and tissues. H2S levels are decreased in a number of conditions (e.g., diabetes mellitus, ischemia, and aging) and are increased in other states (e.g., inflammation, critical illness, and cancer). Over the last decades, multiple approaches have been identified for the therapeutic exploitation of H2S, either based on H2S donation or inhibition of H2S biosynthesis. H2S donation can be achieved through the inhalation of H2S gas and/or the parenteral or enteral administration of so-called fast-releasing H2S donors (salts of H2S such as NaHS and Na2S) or slow-releasing H2S donors (GYY4137 being the prototypical compound used in hundreds of studies in vitro and in vivo). Recent work also identifies various donors with regulated H2S release profiles, including oxidant-triggered donors, pH-dependent donors, esterase-activated donors, and organelle-targeted (e.g., mitochondrial) compounds. There are also approaches where existing, clinically approved drugs of various classes (e.g., nonsteroidal anti-inflammatories) are coupled with H2S-donating groups (the most advanced compound in clinical trials is ATB-346, an H2S-donating derivative of the non-steroidal anti-inflammatory compound naproxen). For pharmacological inhibition of H2S synthesis, there are now several small molecule compounds targeting each of the three H2S-producing enzymes cystathionine-β-synthase (CBS), cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase. Although many of these compounds have their limitations (potency, selectivity), these molecules, especially in combination with genetic approaches, can be instrumental for the delineation of the biologic processes involving endogenous H2S production. Moreover, some of these compounds (e.g., cell-permeable prodrugs of the CBS inhibitor aminooxyacetate, or benserazide, a potentially repurposable CBS inhibitor) may serve as starting points for future clinical translation. The present article overviews the currently known H2S donors and H2S biosynthesis inhibitors, delineates their mode of action, and offers examples for their biologic effects and potential therapeutic utility.
Collapse
Affiliation(s)
- Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas (C.S.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece (A.P.); and Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Andreas Papapetropoulos
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas (C.S.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece (A.P.); and Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| |
Collapse
|
22
|
Yuan S, Shen X, Kevil CG. Beyond a Gasotransmitter: Hydrogen Sulfide and Polysulfide in Cardiovascular Health and Immune Response. Antioxid Redox Signal 2017; 27:634-653. [PMID: 28398086 PMCID: PMC5576200 DOI: 10.1089/ars.2017.7096] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Hydrogen sulfide (H2S) metabolism leads to the formation of oxidized sulfide species, including polysulfide, persulfide, and others. Evidence is emerging that many biological effects of H2S may indeed be due to polysulfide and persulfide activation of signaling pathways and reactivity with discrete small molecules. Recent Advances: Exogenous oxidized sulfide species, including polysulfides, are more reactive than H2S with a wide range of molecules. Importantly, endogenous polysulfide and persulfide formation has been reported to occur via transsulfuration enzymes, cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS). CRITICAL ISSUES In light of the recent understanding of oxidized sulfide metabolite formation and reactivity, comparatively few studies have been reported comparing cellular biological and in vivo effects of H2S donors versus polysulfide and persulfide donors. Likewise, it is equally unclear when, how, and to what extent persulfide and polysulfide formation occurs in vivo under pathophysiological conditions. FUTURE DIRECTIONS Additional studies regarding persulfide and polysulfide formation and molecular reactions are needed in nearly all aspects of biology to better understand how sulfide metabolites contribute to key chemical biology reactions involved in cardiovascular health and immune responses. Antioxid. Redox Signal. 27, 634-653.
Collapse
Affiliation(s)
- Shuai Yuan
- 1 Department of Cell Biology and Anatomy, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| | - Xinggui Shen
- 2 Department of Pathology and Translational Pathobiology, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| | - Christopher G Kevil
- 2 Department of Pathology and Translational Pathobiology, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| |
Collapse
|
23
|
H₂S-Mediated Protein S-Sulfhydration: A Prediction for Its Formation and Regulation. Molecules 2017; 22:molecules22081334. [PMID: 28800080 PMCID: PMC6152389 DOI: 10.3390/molecules22081334] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/26/2017] [Accepted: 08/04/2017] [Indexed: 11/27/2022] Open
Abstract
Protein S-sulfhydration is a newly discovered post-translational modification of specific cysteine residue(s) in target proteins, which is involved in a broad range of cellular functions and metabolic pathways. By changing local conformation and the final activity of target proteins, S-sulfhydration is believed to mediate most cellular responses initiated by H2S, a novel gasotransmitter. In comparison to protein S-sulfhydration, nitric oxide-mediated protein S-nitrosylation has been extensively investigated, including its formation, regulation, transfer and metabolism. Although the investigation on the regulatory mechanisms associated with protein S-sulfhydration is still in its infancy, accumulated evidence suggested that protein S-sulfhydration may share similar chemical features with protein S-nitrosylation. Glutathione persulfide acts as a major donor for protein S-sulfhydration. Here, we review the present knowledge on protein S-sulfhydration, and also predict its formation and regulation mechanisms based on the knowledge from protein S-nitrosylation.
Collapse
|
24
|
Manicam C, Ginter N, Li H, Xia N, Goloborodko E, Zadeh JK, Musayeva A, Pfeiffer N, Gericke A. Compensatory Vasodilator Mechanisms in the Ophthalmic Artery of Endothelial Nitric Oxide Synthase Gene Knockout Mice. Sci Rep 2017; 7:7111. [PMID: 28769073 PMCID: PMC5541003 DOI: 10.1038/s41598-017-07768-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/29/2017] [Indexed: 01/02/2023] Open
Abstract
Nitric oxide (NO) generated by endothelial nitric oxide synthase (eNOS) plays an important role in the maintenance of ocular vascular homeostasis. Therefore, perturbations in vascular NO synthesis have been implicated in the pathogenesis of several ocular diseases. We recently reported that eNOS contributes significantly to vasodilation of the mouse ophthalmic artery. Interestingly, dilatory responses were also retained in eNOS gene-deficient mice (eNOS-/-), indicating inherent endothelial adaptive mechanism(s) that act as back-up systems in chronic absence of eNOS to preserve vasorelaxation. Thus, this study endeavoured to identify the compensatory mechanism(s) in the ophthalmic artery of eNOS-/- mice employing isolated arterial segments and pharmacological inhibitors in vitro. Endothelium removal virtually abolished acetylcholine (ACh)-induced vasodilation, suggesting an obligatory involvement of the endothelium in cholinergic control of vascular tone. However, non-NOS and non-cyclooxygenase components compensate for eNOS deficiency via endothelium-derived hyperpolarizing factors (EDHFs). Notably, arachidonic acid-derived metabolites of the 12-lipoxygenase pathway were key mediators in activating the inwardly rectifying potassium channels to compensate for chronic lack of eNOS. Conclusively, endothelium-dependent cholinergic responses of the ophthalmic artery in the eNOS-/- mice are largely preserved and, this vascular bed has the ability to compensate for the loss of normal vasodilator responses solely via EDHFs.
Collapse
Affiliation(s)
- Caroline Manicam
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Natalja Ginter
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Huige Li
- Institute of Pharmacology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ning Xia
- Institute of Pharmacology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Evgeny Goloborodko
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jenia Kouchek Zadeh
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Aytan Musayeva
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
25
|
Prieto-Bermejo R, Hernández-Hernández A. The Importance of NADPH Oxidases and Redox Signaling in Angiogenesis. Antioxidants (Basel) 2017; 6:antiox6020032. [PMID: 28505091 PMCID: PMC5488012 DOI: 10.3390/antiox6020032] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/28/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
Eukaryotic cells have to cope with the constant generation of reactive oxygen species (ROS). Although the excessive production of ROS might be deleterious for cell biology, there is a plethora of evidence showing that moderate levels of ROS are important for the control of cell signaling and gene expression. The family of the nicotinamide adenine dinucleotide phosphate oxidases (NADPH oxidases or Nox) has evolved to produce ROS in response to different signals; therefore, they fulfil a central role in the control of redox signaling. The role of NADPH oxidases in vascular physiology has been a field of intense study over the last two decades. In this review we will briefly analyze how ROS can regulate signaling and gene expression. We will address the implication of NADPH oxidases and redox signaling in angiogenesis, and finally, the therapeutic possibilities derived from this knowledge will be discussed.
Collapse
Affiliation(s)
- Rodrigo Prieto-Bermejo
- Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain.
| | | |
Collapse
|
26
|
Candela J, Wang R, White C. Microvascular Endothelial Dysfunction in Obesity Is Driven by Macrophage-Dependent Hydrogen Sulfide Depletion. Arterioscler Thromb Vasc Biol 2017; 37:889-899. [DOI: 10.1161/atvbaha.117.309138] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 03/12/2017] [Indexed: 11/16/2022]
Abstract
Objective—
The function of perivascular adipose tissue as an anticontractile mediator in the microvasculature is lost during obesity. Obesity results in inflammation and recruitment of proinflammatory macrophages to the perivascular adipose tissue that is paralleled by depletion of the vasorelaxant signaling molecule hydrogen sulfide (H
2
S) in the vessel. The current objective was to assess the role of macrophages in determining vascular [H
2
S] and defining how this impinged on vasodilation.
Approach and Results—
Contractility and [H
2
S] were measured in mesenteric resistance arterioles from lean and obese mice by using pressure myography and confocal microscopy, respectively. Vasodilation was impaired and smooth muscle and endothelial [H
2
S] decreased in vessels from obese mice compared with those from lean controls. Coculturing vessels from lean mice with macrophages from obese mice, or macrophage-conditioned media, recapitulated obese phenotypes in vessels. These effects were mediated by low molecular weight species and dependent on macrophage inducible nitric oxide synthase activity.
Conclusions—
The inducible nitric oxide synthase activity of perivascular adipose tissue–resident proinflammatory macrophages promotes microvascular endothelial dysfunction by reducing the bioavailability of H
2
S in the vessel. These findings support a model in which vascular H
2
S depletion underpins the loss of perivascular adipose tissue anticontractile function in obesity.
Collapse
Affiliation(s)
- Joseph Candela
- From the Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL (J.C., C.W.); and Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada (R.W.)
| | - Rui Wang
- From the Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL (J.C., C.W.); and Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada (R.W.)
| | - Carl White
- From the Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL (J.C., C.W.); and Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada (R.W.)
| |
Collapse
|
27
|
Zhang T, Wei W, Dirsch O, Krüger T, Kan C, Xie C, Kniemeyer O, Fang H, Settmacher U, Dahmen U. Identification of Proteins Interacting with Cytoplasmic High-Mobility Group Box 1 during the Hepatocellular Response to Ischemia Reperfusion Injury. Int J Mol Sci 2017; 18:ijms18010167. [PMID: 28275217 PMCID: PMC5297800 DOI: 10.3390/ijms18010167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 01/22/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) occurs inevitably in liver transplantations and frequently during major resections, and can lead to liver dysfunction as well as systemic disorders. High-mobility group box 1 (HMGB1) plays a pathogenic role in hepatic IRI. In the normal liver, HMGB1 is located in the nucleus of hepatocytes; after ischemia reperfusion, it translocates to the cytoplasm and it is further released to the extracellular space. Unlike the well-explored functions of nuclear and extracellular HMGB1, the role of cytoplasmic HMGB1 in hepatic IRI remains elusive. We hypothesized that cytoplasmic HMGB1 interacts with binding proteins involved in the hepatocellular response to IRI. In this study, binding proteins of cytoplasmic HMGB1 during hepatic IRI were identified. Liver tissues from rats with warm ischemia reperfusion (WI/R) injury and from normal rats were subjected to cytoplasmic protein extraction. Co-immunoprecipitation using these protein extracts was performed to enrich HMGB1-protein complexes. To separate and identify the immunoprecipitated proteins in eluates, 2-dimensional electrophoresis and subsequent mass spectrometry detection were performed. Two of the identified proteins were verified using Western blotting: betaine–homocysteine S-methyltransferase 1 (BHMT) and cystathionine γ-lyase (CTH). Therefore, our results revealed the binding of HMGB1 to BHMT and CTH in cytoplasm during hepatic WI/R. This finding may help to better understand the cellular response to IRI in the liver and to identify novel molecular targets for reducing ischemic injury.
Collapse
Affiliation(s)
- Tianjiao Zhang
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Weiwei Wei
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Olaf Dirsch
- Institute of Pathology, Klinikum Chemnitz gGmbH, 09116 Chemnitz, Germany.
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany.
| | - Chunyi Kan
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
- Department of Obstetrics and Gynecology, Wuhan Central Hospital, Wuhan 430014, China.
| | - Chichi Xie
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany.
| | - Haoshu Fang
- Department of Pathophysiology, Anhui Medical University, Hefei 230032, China.
| | - Utz Settmacher
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|
28
|
Núñez-Gómez E, Pericacho M, Ollauri-Ibáñez C, Bernabéu C, López-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis 2016; 20:1-24. [PMID: 27943030 DOI: 10.1007/s10456-016-9535-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Following arterial occlusion, blood vessels respond by forming a new network of functional capillaries (angiogenesis), by reorganizing preexisting capillaries through the recruitment of smooth muscle cells to generate new arteries (arteriogenesis) and by growing and remodeling preexisting collateral arterioles into physiologically relevant arteries (collateral development). All these processes result in the recovery of organ perfusion. The importance of endoglin in post-occlusion reperfusion is sustained by several observations: (1) endoglin expression is increased in vessels showing active angiogenesis/remodeling; (2) genetic endoglin haploinsufficiency in humans causes deficient angiogenesis; and (3) the reduction of endoglin expression by gene disruption or the administration of endoglin-neutralizing antibodies reduces angiogenesis and revascularization. However, the precise role of endoglin in the several processes associated with revascularization has not been completely elucidated and, in some cases, the function ascribed to endoglin by different authors is controversial. The purpose of this review is to organize in a critical way the information available for the role of endoglin in several phenomena (angiogenesis, arteriogenesis and collateral development) associated with post-ischemic revascularization.
Collapse
Affiliation(s)
- Elena Núñez-Gómez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Spanish National Research Council (CIB, CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M López-Novoa
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain. .,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
29
|
Secomb TW, Pries AR. Microvascular Plasticity: Angiogenesis in Health and Disease--Preface. Microcirculation 2016; 23:93-4. [PMID: 26639099 DOI: 10.1111/micc.12262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 11/25/2015] [Indexed: 11/26/2022]
Abstract
This Special Topic Issue is concerned with the mechanisms that determine the structure of microvascular networks. The vast number of vessels and the highly plastic character of the microcirculation give evidence that microvascular network structures emerge as a result of responses of individual vessels and cells to the local stimuli that they experience, through a combination of angiogenesis, remodeling and pruning. The articles in this issue of Microcirculation address a range of cellular and molecular mechanisms involved in these processes.
Collapse
Affiliation(s)
- Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, Arizona, USA
| | - Axel R Pries
- Department of Physiology, Charité Berlin, Berlin, Germany.,Deutsches Herzzentrum Berlin, Berlin, Germany
| |
Collapse
|
30
|
Cheng Z, Garikipati VNS, Nickoloff E, Wang C, Polhemus DJ, Zhou J, Benedict C, Khan M, Verma SK, Rabinowitz JE, Lefer D, Kishore R. Restoration of Hydrogen Sulfide Production in Diabetic Mice Improves Reparative Function of Bone Marrow Cells. Circulation 2016; 134:1467-1483. [PMID: 27660293 DOI: 10.1161/circulationaha.116.022967] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/06/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Bone marrow cell (BMC)-based treatment for critical limb ischemia in diabetic patients yielded a modest therapeutic effect resulting from cell dysfunction. Therefore, approaches that improve diabetic stem/progenitor cell functions may provide therapeutic benefits. Here, we tested the hypothesis that restoration of hydrogen sulfide (H2S) production in diabetic BMCs improves their reparative capacities. METHODS Mouse BMCs were isolated by density-gradient centrifugation. Unilateral hind limb ischemia was conducted in 12- to 14-week-old db/+ and db/db mice by ligation of the left femoral artery. The H2S level was measured by either gas chromatography or staining with florescent dye sulfidefluor 7 AM. RESULTS Both H2S production and cystathionine γ-lyase (CSE), an H2S enzyme, levels were significantly decreased in BMCs from diabetic db/db mice. Administration of H2S donor diallyl trisulfide (DATS) or overexpression of CSE restored H2S production and enhanced cell survival and migratory capacity in high glucose (HG)-treated BMCs. Immediately after hind limb ischemia surgery, the db/+ and db/db mice were administered DATS orally and/or given a local intramuscular injection of green fluorescent protein-labeled BMCs or red fluorescent protein-CSE-overexpressing BMCs (CSE-BMCs). Mice with hind limb ischemia were divided into 6 groups: db/+, db/db, db/db+BMCs, db/db+DATS, db/db+DATS+BMCs, and db/db+CSE-BMCs. DATS and CSE overexpression greatly enhanced diabetic BMC retention in ischemic hind limbs followed by improved blood perfusion, capillary/arteriole density, skeletal muscle architecture, and cell survival and decreased perivascular CD68+ cell infiltration in the ischemic hind limbs of diabetic mice. It is interesting to note that DATS or CSE overexpression rescued high glucose-impaired migration, tube formation, and survival of BMCs or mature human cardiac microvascular endothelial cells. Moreover, DATS restored nitric oxide production and decreased endothelial nitric oxide synthase phosphorylation at threonine 495 levels in human cardiac microvascular endothelial cells and improved BMC angiogenic activity under high glucose condition. Last, silencing CSE by siRNA significantly increased endothelial nitric oxide synthase phosphorylation at threonine 495 levels in human cardiac microvascular endothelial cells. CONCLUSIONS Decreased CSE-mediated H2S bioavailability is an underlying source of BMC dysfunction in diabetes mellitus. Our data indicate that H2S and overexpression of CSE in diabetic BMCs may rescue their dysfunction and open novel avenues for cell-based therapeutics of critical limb ischemia in diabetic patients.
Collapse
Affiliation(s)
- Zhongjian Cheng
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Venkata Naga Srikanth Garikipati
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Emily Nickoloff
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Chunlin Wang
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - David J Polhemus
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Jibin Zhou
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Cynthia Benedict
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Mohsin Khan
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Suresh K Verma
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Joseph E Rabinowitz
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - David Lefer
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.)
| | - Raj Kishore
- From Center for Translational Medicine (Z.C., V.N.S.G., E.N., C.W., J.Z., C.B., M.K., S.K.V., J.E.R., R.K.) and Department of Pharmacology (J.E.R., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA; and Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA (D.J.P., D.L.).
| |
Collapse
|
31
|
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
|
32
|
Al-Samir S, Wang Y, Meissner JD, Gros G, Endeward V. Cardiac Morphology and Function, and Blood Gas Transport in Aquaporin-1 Knockout Mice. Front Physiol 2016; 7:181. [PMID: 27252655 PMCID: PMC4878313 DOI: 10.3389/fphys.2016.00181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 05/06/2016] [Indexed: 12/24/2022] Open
Abstract
We have studied cardiac and respiratory functions of aquaporin-1-deficient mice by the Pressure-Volume-loop technique and by blood gas analysis. In addition, the morphological properties of the animals' hearts were analyzed. In anesthesia under maximal dobutamine stimulation, the mice exhibit a moderately elevated heart rate of < 600 min−1 and an O2 consumption of ~0.6 ml/min/g, which is about twice the basal rate. In this state, which is similar to the resting state of the conscious animal, all cardiac functions including stroke volume and cardiac output exhibited resting values and were identical between deficient and wildtype animals. Likewise, pulmonary and peripheral exchange of O2 and CO2 were normal. In contrast, several morphological parameters of the heart tissue of deficient mice were altered: (1) left ventricular wall thickness was reduced by 12%, (2) left ventricular mass, normalized to tibia length, was reduced by 10–20%, (3) cardiac muscle fiber cross sectional area was decreased by 17%, and (4) capillary density was diminished by 10%. As the P-V-loop technique yielded normal end-diastolic and end-systolic left ventricular volumes, the deficient hearts are characterized by thin ventricular walls in combination with normal intraventricular volumes. The aquaporin-1-deficient heart thus seems to be at a disadvantage compared to the wild-type heart by a reduced left-ventricular wall thickness and an increased diffusion distance between blood capillaries and muscle mitochondria. While under the present quasi-resting conditions these morphological alterations have no consequences for cardiac function, we expect that the deficient hearts will show a reduced maximal cardiac output.
Collapse
Affiliation(s)
- Samer Al-Samir
- Abteilung Molekular- und Zellphysiologie, AG Vegetative Physiologie 4220, Medizinische Hochschule Hannover Hannover, Germany
| | - Yong Wang
- Division Molecular and Translational Cardiology, Department Cardiology and Angiology, Medizinische Hochschule Hannover Hannover, Germany
| | - Joachim D Meissner
- Abteilung Molekular- und Zellphysiologie, AG Vegetative Physiologie 4220, Medizinische Hochschule Hannover Hannover, Germany
| | - Gerolf Gros
- Abteilung Molekular- und Zellphysiologie, AG Vegetative Physiologie 4220, Medizinische Hochschule Hannover Hannover, Germany
| | - Volker Endeward
- Abteilung Molekular- und Zellphysiologie, AG Vegetative Physiologie 4220, Medizinische Hochschule Hannover Hannover, Germany
| |
Collapse
|