1
|
Ayari A, Dakhli N, Jedidi S, Sammari H, Arrari F, Sebai H. Laxative and purgative actions of phytoactive compounds from beetroot juice against loperamide-induced constipation, oxidative stress, and inflammation in rats. Neurogastroenterol Motil 2025; 37:e14935. [PMID: 39370602 DOI: 10.1111/nmo.14935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Chronic constipation is a gastrointestinal functional disorder which affects patient quality of life. Therefore, many studies were oriented to search herbal laxative agents. In this study, we investigated the phytochemical composition of beetroot juice (BJ) and its laxative potential in an experimental model of constipation and colonic dysmotility induced by loperamide (LOP) in Wistar rats. METHODS Animals were concurrently pretreated with LOP (3 mg/kg, b.w., i.p.) and BJ (5 and 10 mL/kg, b.w., p.o.), or yohimbine (2 mg/kg, b.w., i.p.), during 1 week. The laxative activity was determined based on the weight, frequency, and water content of the feces matter. The gastric-emptying test and intestinal transit were determined. Colon histology was examined, and oxidative status was evaluated using biochemical-colorimetric methods. KEY RESULTS The in vivo study revealed that LOP induced a significant inhibition of gastrointestinal motility, negative consequences on defecation parameters, oxidative stress, and colonic mucosa lesions. Conversely, administration of BJ reestablished these parameters and restored colonic oxidative balance. Importantly, BJ treatment protected against LOP-induced inflammatory markers (pro-inflammatory cytokines and WBC) and the increase in intracellular mediators such as hydrogen peroxide, free iron, and calcium levels. CONCLUSIONS & INFERENCES This study demonstrated that the bioactive compounds in BJ provided an anti-constipation effect by modulating intestinal motility and regulating oxidative stress and inflammation induced by LOP intoxication.
Collapse
Affiliation(s)
- Ala Ayari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Nouha Dakhli
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
- Sylvo-Pastoral Institute of Tabarka, Laboratory of Sylvo-Pastoral Resources, University of Jendouba, Tabarka, Tunisia
| | - Houcem Sammari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Fatma Arrari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| |
Collapse
|
2
|
Brinsi C, Jedidi S, Sammari H, Selmi H, Sebai H. Antidiarrheal, anti-inflammatory and antioxidant effects of Anethum graveolens L. fruit extract on castor oil-induced diarrhea in rats. Neurogastroenterol Motil 2024; 36:e14892. [PMID: 39115258 DOI: 10.1111/nmo.14892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Between food and medicine, nutraceuticals are widely used in human health for the prevention and treatment of various diseases. This study aims to determine the cytoprotective effects of Anethum gravelons fruit extract (AGFAE) on castor oil-induced diarrhea in rats due to its phytochemical and antioxidant properties. METHODS Male rats were divided into six groups of six animals each: Control (C), Castor oil (CO), CO + different doses of AGFAE (50, 100, and 200 mg/kg, b.w., p.o.), and the CO + loperamide group (LOP, 10 mg/kg, b.w., p.o.). KEY RESULTS In vitro, the chemical composition of aqueous Dill fruit extract showed strong antioxidant activity, with a high content of total polyphenols, flavonoids, and tannins. In our in vivo studies, pre-treatment with AGFAE reduced malondialdehyde and hydrogen peroxide levels and maintained normal activity of enzymatic and non-enzymatic antioxidants in the gastric and intestinal mucosa. In addition, we found that AGFAE prophylaxis improved the stability of many plasma biochemical parameters altered by castor oil intoxication, such as C-reactive protein concentrations and alkaline phosphatase activities. CONCLUSIONS & INFERENCES We suggest that AGFAE phenolic compounds had significant protection against diarrhea involving several mechanisms such as reducing hypersecretion, peristaltic, inflammation, and preserving the endogenous antioxidant levels.
Collapse
Affiliation(s)
- Chirine Brinsi
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
- Laboratory of Sylvo-Pastoral Resources, Institution of Agricultural Research and Higher Education (IRESA), University of Jendouba, Sylvo- Pastoral Institute of Tabarka, Tabarka, Tunisia
| | - Houcem Sammari
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Houcine Selmi
- Laboratory of Sylvo-Pastoral Resources, Institution of Agricultural Research and Higher Education (IRESA), University of Jendouba, Sylvo- Pastoral Institute of Tabarka, Tabarka, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| |
Collapse
|
3
|
Wang YX, Reyes-García J, Di Mise A, Zheng YM. Role of ryanodine receptor 2 and FK506-binding protein 12.6 dissociation in pulmonary hypertension. J Gen Physiol 2023; 155:e202213100. [PMID: 36625865 PMCID: PMC9836826 DOI: 10.1085/jgp.202213100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/29/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by a progressive increase in pulmonary arterial pressure leading to right ventricular failure and death. A major cellular response in this disease is the contraction of smooth muscle cells (SMCs) of the pulmonary vasculature. Cell contraction is determined by the increase in intracellular Ca2+ concentration ([Ca2+]i), which is generated and regulated by various ion channels. Several studies by us and others have shown that ryanodine receptor 2 (RyR2), a Ca2+-releasing channel in the sarcoplasmic reticulum (SR), is an essential ion channel for the control of [Ca2+]i in pulmonary artery SMCs (PASMCs), thereby mediating the sustained vasoconstriction seen in PH. FK506-binding protein 12.6 (FKBP12.6) strongly associates with RyR2 to stabilize its functional activity. FKBP12.6 can be dissociated from RyR2 by a hypoxic stimulus to increase channel function and Ca2+ release, leading to pulmonary vasoconstriction and PH. More specifically, dissociation of the RyR2-FKBP12.6 complex is a consequence of increased mitochondrial ROS generation mediated by the Rieske iron-sulfur protein (RISP) at the mitochondrial complex III after hypoxia. Overall, RyR2/FKBP12.6 dissociation and the corresponding signaling pathway may be an important factor in the development of PH. Novel drugs and biologics targeting RyR2, FKBP12.6, and related molecules may become unique effective therapeutics for PH.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Jorge Reyes-García
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México,Ciudad de México, México
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
4
|
Yan K, Zheng J, Kluth MA, Li L, Ganss C, Yard B, Magdeburg R, Frank MH, Pallavi P, Keese M. ABCB5 + mesenchymal stromal cells therapy protects from hypoxia by restoring Ca 2+ homeostasis in vitro and in vivo. Stem Cell Res Ther 2023; 14:24. [PMID: 36759868 PMCID: PMC9912525 DOI: 10.1186/s13287-022-03228-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Hypoxia in ischemic disease impairs Ca2+ homeostasis and may promote angiogenesis. The therapeutic efficacy of mesenchymal stromal cells (MSCs) in peripheral arterial occlusive disease is well established, yet its influence on cellular Ca2+ homeostasis remains to be elucidated. We addressed the influence of ATP-binding cassette subfamily B member 5 positive mesenchymal stromal cells (ABCB5+ MSCs) on Ca2+ homeostasis in hypoxic human umbilical vein endothelial cells (HUVECs) in vitro and in vivo. METHODS Hypoxia was induced in HUVECs by Cobalt (II) chloride (CoCl2) or Deferoxamine (DFO). Dynamic changes in the cytosolic- and endoplasmic reticulum (ER) Ca2+ and changes in reactive oxygen species were assessed by appropriate fluorescence-based sensors. Metabolic activity, cell migration, and tube formation were assessed by standard assays. Acute-on-chronic ischemia in Apolipoprotein E knock-out (ApoE-/-) mice was performed by double ligation of the right femoral artery (DFLA). ABCB5+ MSC cells were injected into the ischemic limb. Functional recovery after DFLA and histology of gastrocnemius and aorta were assessed. RESULTS Hypoxia-induced impairment of cytosolic and ER Ca2+ were restored by ABCB5+ MSCs or their conditioned medium. Similar was found for changes in intracellular ROS production, metabolic activity, migratory ability and tube formation. The restoration was paralleled by an increased expression of the Ca2+ transporter Sarco-/endoplasmic reticulum ATPase 2a (SERCA2a) and the phosphorylation of Phospholamban (PLN). In acute-on-chronic ischemia, ABCB5+ MSCs treated mice showed a higher microvascular density, increased SERCA2a expression and PLN phosphorylation relative to untreated controls. CONCLUSIONS ABCB5+ MSCs therapy can restore cellular Ca2+ homeostasis, which may beneficially affect the angiogenic function of endothelial cells under hypoxia in vitro and in vivo.
Collapse
Affiliation(s)
- Kaixuan Yan
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jiaxing Zheng
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Lin Li
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Ganss
- TICEBA GmbH, Heidelberg, Germany ,grid.476673.7RHEACELL GmbH & Co. KG, Heidelberg, Germany
| | - Benito Yard
- grid.7700.00000 0001 2190 4373V Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Richard Magdeburg
- grid.411778.c0000 0001 2162 1728Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161 Mannheim, Germany
| | - Markus H. Frank
- grid.38142.3c000000041936754XDepartment of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XTransplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Stem Cell Institute, Harvard University, Cambridge, MA USA ,grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia
| | - Prama Pallavi
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161, Mannheim, Germany.
| | - Michael Keese
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,Department for General and Visceral Surgery, Theresienkrankenhaus Mannheim, Mannheim, Germany. .,Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161, Mannheim, Germany.
| |
Collapse
|
5
|
Di Salvo E, Casciaro M, Giorgianni CM, Cicero N, Gangemi S. Age-Related Diseases and Foods Generating Chlorinative Stress. Antioxidants (Basel) 2023; 12:antiox12020249. [PMID: 36829808 PMCID: PMC9952263 DOI: 10.3390/antiox12020249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/08/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Aging is a slow and inexorable process affecting all life beings and is characterised by age-related worsening in adaptation to external changes. Several factors contribute to such a process, and oxidative stress due to external damages is one key player. Of particular interest is the oxidative stress generated from halogen compounds such as chloride. Hypochlorus acid is produced starting from MPO's interaction with hydrogen peroxide. We focused on the oxidation of tyrosine residues by HOCl, which leads as a result to the formation of 3-chlorotyrosine (3-ClTyr). This molecule, due to its stability, is considered a marker for MPO activity. RESULTS We collected data from literature research articles evaluating chlorinative stress and the effects of 3-ClTyr on chronic diseases linked to aging. As diseases are not the only source of 3-ClTyr in people, we also focused on other origins of chlorinative stress, such as food intake. DISCUSSION Oxidation and halogenation are caused by infectious diseases and by pathologies characterised by inflammation. Moreover, diet could negatively or positively influence chlorinative stress. Comparing 3-ClTyr levels in the oldest and youngest old with age-related diseases and comparing data between different geographic areas with different pesticide rules could be the next challenge.
Collapse
Affiliation(s)
- Eleonora Di Salvo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Marco Casciaro
- School and Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
- Correspondence: ; Tel.: +39-0902212013
| | - Concetto Mario Giorgianni
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Nicola Cicero
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy
- Science4life srl, Spin off Company, University of Messina, 98100 Messina, Italy
| | - Sebastiano Gangemi
- School and Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
6
|
Boecker D, Zhang Z, Breves R, Herth F, Kramer A, Bulitta C. Antimicrobial efficacy, mode of action and in vivo use of hypochlorous acid (HOCl) for prevention or therapeutic support of infections. GMS HYGIENE AND INFECTION CONTROL 2023; 18:Doc07. [PMID: 37034111 PMCID: PMC10073986 DOI: 10.3205/dgkh000433] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
The objective is to provide a comprehensive overview of the rapidly developing field of the current state of research on in vivo use of hypochlorous acid (HOCl) to aid infection prevention and control, including naso-pharyngeal, alveolar, topical, and systemic HOCl applications. Also, examples are provided of dedicated applications in COVID-19. A brief background of HOCl's biological and chemical specifics and its physiological role in the innate immune system is provided to understand the effect of in vivo applications in the context of the body's own physiological defense mechanisms.
Collapse
Affiliation(s)
- Dirk Boecker
- TOTO Consulting LLC, San Jose CA, USA
- *To whom correspondence should be addressed: Dirk Boecker, TOTO Consulting LLC, San Jose CA, USA, E-mail:
| | - Zhentian Zhang
- Institute for Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | | | - Felix Herth
- Thoraxklinik, University of Heidelberg, Heidelberg, Germany
| | - Axel Kramer
- Institut of Hygiene and Environmental Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Clemens Bulitta
- Institut für Medizintechnik, Ostbayerische Technische Hochschule (OTH) Amberg-Weiden, Amberg-Weiden, Germany
| |
Collapse
|
7
|
Ma L, Zou R, Shi W, Zhou N, Chen S, Zhou H, Chen X, Wu Y. SGLT2 inhibitor dapagliflozin reduces endothelial dysfunction and microvascular damage during cardiac ischemia/reperfusion injury through normalizing the XO-SERCA2-CaMKII-coffilin pathways. Am J Cancer Res 2022; 12:5034-5050. [PMID: 35836807 PMCID: PMC9274739 DOI: 10.7150/thno.75121] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/13/2022] [Indexed: 01/12/2023] Open
Abstract
Background: Given the importance of microvascular injury in infarct formation and expansion, development of therapeutic strategies for microvascular protection against myocardial ischemia/reperfusion injury (IRI) is of great interest. Here, we explored the molecular mechanisms underlying the protective effects of the SGLT2 inhibitor dapagliflozin (DAPA) against cardiac microvascular dysfunction mediated by IRI. Methods: DAPA effects were evaluated both in vivo, in mice subjected to IRI, and in vitro, in human coronary artery endothelial cells (HCAECs) exposed to hypoxia/reoxygenation (H/R). DAPA pretreatment attenuated luminal stenosis, endothelial swelling, and inflammation in cardiac microvessels of IRI-treated mice. Results: In H/R-challenged HCAECs, DAPA treatment improved endothelial barrier function, endothelial nitric oxide synthase (eNOS) activity, and angiogenic capacity, and inhibited H/R-induced apoptosis by preventing cofilin-dependent F-actin depolymerization and cytoskeletal degradation. Inhibition of H/R-induced xanthine oxidase (XO) activation and upregulation, sarco(endo)plasmic reticulum calcium-ATPase 2 (SERCA2) oxidation and inactivation, and cytoplasmic calcium overload was further observed in DAPA-treated HCAECs. DAPA also suppressed calcium/Calmodulin (CaM)-dependent kinase II (CaMKII) activation and cofilin phosphorylation, and preserved cytoskeleton integrity and endothelial cell viability following H/R. Importantly, the beneficial effects of DAPA on cardiac microvascular integrity and endothelial cell survival were largely prevented in IRI-treated SERCA2-knockout mice. Conclusions: These results indicate that DAPA effectively reduces cardiac microvascular damage and endothelial dysfunction during IRI through inhibition of the XO-SERCA2-CaMKII-cofilin pathway.
Collapse
Affiliation(s)
- Li Ma
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Rongjun Zou
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wanting Shi
- Department of Paediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Na Zhou
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shaoxian Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China.,✉ Corresponding authors: Hao Zhou, E-mail: ; Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China. Xinxin Chen, E-mail: ; Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China. Yueheng Wu, E-mail: ; Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xinxin Chen
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,✉ Corresponding authors: Hao Zhou, E-mail: ; Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China. Xinxin Chen, E-mail: ; Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China. Yueheng Wu, E-mail: ; Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yueheng Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China.,✉ Corresponding authors: Hao Zhou, E-mail: ; Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China. Xinxin Chen, E-mail: ; Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China. Yueheng Wu, E-mail: ; Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
8
|
Snell JA, Jandova J, Wondrak GT. Hypochlorous Acid: From Innate Immune Factor and Environmental Toxicant to Chemopreventive Agent Targeting Solar UV-Induced Skin Cancer. Front Oncol 2022; 12:887220. [PMID: 35574306 PMCID: PMC9106365 DOI: 10.3389/fonc.2022.887220] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022] Open
Abstract
A multitude of extrinsic environmental factors (referred to in their entirety as the 'skin exposome') impact structure and function of skin and its corresponding cellular components. The complex (i.e. additive, antagonistic, or synergistic) interactions between multiple extrinsic (exposome) and intrinsic (biological) factors are important determinants of skin health outcomes. Here, we review the role of hypochlorous acid (HOCl) as an emerging component of the skin exposome serving molecular functions as an innate immune factor, environmental toxicant, and topical chemopreventive agent targeting solar UV-induced skin cancer. HOCl [and its corresponding anion (OCl-; hypochlorite)], a weak halogen-based acid and powerful oxidant, serves two seemingly unrelated molecular roles: (i) as an innate immune factor [acting as a myeloperoxidase (MPO)-derived microbicidal factor] and (ii) as a chemical disinfectant used in freshwater processing on a global scale, both in the context of drinking water safety and recreational freshwater use. Physicochemical properties (including redox potential and photon absorptivity) determine chemical reactivity of HOCl towards select biochemical targets [i.e. proteins (e.g. IKK, GRP78, HSA, Keap1/NRF2), lipids, and nucleic acids], essential to its role in innate immunity, antimicrobial disinfection, and therapeutic anti-inflammatory use. Recent studies have explored the interaction between solar UV and HOCl-related environmental co-exposures identifying a heretofore unrecognized photo-chemopreventive activity of topical HOCl and chlorination stress that blocks tumorigenic inflammatory progression in UV-induced high-risk SKH-1 mouse skin, a finding with potential implications for the prevention of human nonmelanoma skin photocarcinogenesis.
Collapse
Affiliation(s)
| | | | - Georg T. Wondrak
- Department of Pharmacology and Toxicology, R.K. Coit College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
9
|
Ai X, Yu P, Peng L, Luo L, Liu J, Li S, Lai X, Luan F, Meng X. Berberine: A Review of its Pharmacokinetics Properties and Therapeutic Potentials in Diverse Vascular Diseases. Front Pharmacol 2022; 12:762654. [PMID: 35370628 PMCID: PMC8964367 DOI: 10.3389/fphar.2021.762654] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
Traditional Chinese medicine plays a significant role in the treatment of various diseases and has attracted increasing attention for clinical applications. Vascular diseases affecting vasculature in the heart, cerebrovascular disease, atherosclerosis, and diabetic complications have compromised quality of life for affected individuals and increase the burden on health care services. Berberine, a naturally occurring isoquinoline alkaloid form Rhizoma coptidis, is widely used in China as a folk medicine for its antibacterial and anti-inflammatory properties. Promisingly, an increasing number of studies have identified several cellular and molecular targets for berberine, indicating its potential as an alternative therapeutic strategy for vascular diseases, as well as providing novel evidence that supports the therapeutic potential of berberine to combat vascular diseases. The purpose of this review is to comprehensively and systematically describe the evidence for berberine as a therapeutic agent in vascular diseases, including its pharmacological effects, molecular mechanisms, and pharmacokinetics. According to data published so far, berberine shows remarkable anti-inflammatory, antioxidant, antiapoptotic, and antiautophagic activity via the regulation of multiple signaling pathways, including AMP-activated protein kinase (AMPK), nuclear factor κB (NF-κB), mitogen-activated protein kinase silent information regulator 1 (SIRT-1), hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), janus kinase 2 (JAK-2), Ca2+ channels, and endoplasmic reticulum stress. Moreover, we discuss the existing limitations of berberine in the treatment of vascular diseases, and give corresponding measures. In addition, we propose some research perspectives and challenges, and provide a solid evidence base from which further studies can excavate novel effective drugs from Chinese medicine monomers.
Collapse
Affiliation(s)
- Xiaopeng Ai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peiling Yu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Lixia Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liuling Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengqian Li
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xianrong Lai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Luan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
van Leeuwen E, Hampton MB, Smyth LCD. Hypothiocyanous Acid Disrupts the Barrier Function of Brain Endothelial Cells. Antioxidants (Basel) 2022; 11:antiox11040608. [PMID: 35453292 PMCID: PMC9030776 DOI: 10.3390/antiox11040608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a common feature of neurological diseases. During neuroinflammation, neutrophils are recruited to the brain vasculature, where myeloperoxidase can produce hypochlorous acid and the less well-studied oxidant hypothiocyanous acid (HOSCN). In this study, we exposed primary brain endothelial cells (BECs) to HOSCN and observed a rapid loss of transendothelial electrical resistance (TEER) at sublethal concentrations. Decreased barrier function was associated with a loss of tight junctions at cellular contacts and a concomitant loss of dynamic microtubules. Both tight junction and cytoskeletal disruptions were visible within 30 min of exposure, whereas significant loss of TEER took more than 1 h. The removal of the HOSCN after 30 min prevented subsequent barrier dysfunction. These results indicate that BECs are sensitive to HOSCN, resulting in the eventual loss of barrier function. We hypothesise that this mechanism may be relevant in neutrophil transmigration, with HOSCN facilitating blood–brain barrier opening at the sites of egress. Furthermore, this mechanism may be a way through which neutrophils, residing in the vasculature, can influence neuroinflammation in diseases.
Collapse
Affiliation(s)
- Eveline van Leeuwen
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (E.v.L.); (M.B.H.)
| | - Mark B. Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (E.v.L.); (M.B.H.)
| | - Leon C. D. Smyth
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (E.v.L.); (M.B.H.)
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Correspondence: ; Tel.: +64-3-378-6225
| |
Collapse
|
11
|
Wang Z, Chen J, Babicheva A, Jain PP, Rodriguez M, Ayon RJ, Ravellette KS, Wu L, Balistrieri F, Tang H, Wu X, Zhao T, Black SM, Desai AA, Garcia JGN, Sun X, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Endothelial upregulation of mechanosensitive channel Piezo1 in pulmonary hypertension. Am J Physiol Cell Physiol 2021; 321:C1010-C1027. [PMID: 34669509 PMCID: PMC8714987 DOI: 10.1152/ajpcell.00147.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Piezo is a mechanosensitive cation channel responsible for stretch-mediated Ca2+ and Na+ influx in multiple types of cells. Little is known about the functional role of Piezo1 in the lung vasculature and its potential pathogenic role in pulmonary arterial hypertension (PAH). Pulmonary arterial endothelial cells (PAECs) are constantly under mechanic stretch and shear stress that are sufficient to activate Piezo channels. Here, we report that Piezo1 is significantly upregulated in PAECs from patients with idiopathic PAH and animals with experimental pulmonary hypertension (PH) compared with normal controls. Membrane stretch by decreasing extracellular osmotic pressure or by cyclic stretch (18% CS) increases Ca2+-dependent phosphorylation (p) of AKT and ERK, and subsequently upregulates expression of Notch ligands, Jagged1/2 (Jag-1 and Jag-2), and Delta like-4 (DLL4) in PAECs. siRNA-mediated downregulation of Piezo1 significantly inhibited the stretch-mediated pAKT increase and Jag-1 upregulation, whereas downregulation of AKT by siRNA markedly attenuated the stretch-mediated Jag-1 upregulation in human PAECs. Furthermore, the mRNA and protein expression level of Piezo1 in the isolated pulmonary artery, which mainly contains pulmonary arterial smooth muscle cells (PASMCs), from animals with severe PH was also significantly higher than that from control animals. Intraperitoneal injection of a Piezo1 channel blocker, GsMTx4, ameliorated experimental PH in mice. Taken together, our study suggests that membrane stretch-mediated Ca2+ influx through Piezo1 is an important trigger for pAKT-mediated upregulation of Jag-1 in PAECs. Upregulation of the mechanosensitive channel Piezo1 and the resultant increase in the Notch ligands (Jag-1/2 and DLL4) in PAECs may play a critical pathogenic role in the development of pulmonary vascular remodeling in PAH and PH.
Collapse
Affiliation(s)
- Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Haiyang Tang
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Stephen M Black
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ankit A Desai
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Joe G N Garcia
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
12
|
Addis DR, Aggarwal S, Lazrak A, Jilling T, Matalon S. Halogen-Induced Chemical Injury to the Mammalian Cardiopulmonary Systems. Physiology (Bethesda) 2021; 36:272-291. [PMID: 34431415 DOI: 10.1152/physiol.00004.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The halogens chlorine (Cl2) and bromine (Br2) are highly reactive oxidizing elements with widespread industrial applications and a history of development and use as chemical weapons. When inhaled, depending on the dose and duration of exposure, they cause acute and chronic injury to both the lungs and systemic organs that may result in the development of chronic changes (such as fibrosis) and death from cardiopulmonary failure. A number of conditions, such as viral infections, coexposure to other toxic gases, and pregnancy increase susceptibility to halogens significantly. Herein we review their danger to public health, their mechanisms of action, and the development of pharmacological agents that when administered post-exposure decrease morbidity and mortality.
Collapse
Affiliation(s)
- Dylan R Addis
- Department of Anesthesiology and Perioperative Medicine, Division of Cardiothoracic Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Saurabh Aggarwal
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tamas Jilling
- Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Pediatrics, Division of Neonatology, Children's Hospital, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
13
|
Sammari H, Jedidi S, Selmi H, Rtibi K, Jabri MA, Jridi M, Zouari N, Toumi L, Sebai H. Protective effects of Crataegus azarolus L. berries aqueous extract against castor oil-induced diarrhea, oxidative stress, and inflammation in rat. Neurogastroenterol Motil 2021; 33:e14065. [PMID: 33320393 DOI: 10.1111/nmo.14065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/02/2020] [Accepted: 11/25/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Diarrhea is a multifactorial gastrointestinal disorder responsible for about 5 million deaths annually. The chemical composition, the antioxidant activity of Crataegus azarolus berries aqueous extract (CABAE) as well as its protective effects against castor oil-induced diarrhea, oxidative stress, and inflammation in rat were studied. METHODS Sixty male rats were used and divided into six groups of ten animals in each: Control (C), castor oil (CO), CO+various doses of CABAE (100, 200, and 400 mg/kg b.w., p.o.), and CO+loperamide (LOP, 10 mg/kg b.w., p.o.). KEY RESULTS The CABAE showed relatively high levels of total polyphenols, flavonoids, and tannins. The LC-HRESIMS technique allowed the identification of 5 phenolic compounds and the major component is quinic acid. In vivo studies showed that CABAE protected against castor oil-induced diarrhea and intestinal fluid accumulation. The CABAE counteracted castor oil-induced lipoperoxidation, preserved GSH and thiol groups levels, and prevented the depletion of antioxidant enzyme activities, such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). The CABAE administration also protected against castor oil-induced inflammatory markers (ALP and CRP) increase. More importantly, castor oil induced an increase of intracellular mediators, such as hydrogen peroxide, free iron, and calcium, while CABAE pretreatment significantly reversed them to near control levels. CONCLUSION The Crataegus azarolus berries aqueous extract significantly protected against diarrhea due in part to its antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Houcem Sammari
- Unité de Physiologie Fonctionnelle et Valorisation des Bio-Ressources, Institut Supérieur de Biotechnologie de Beja, Université de Jendouba, Beja, Tunisia.,Laboratoire des Ressources Sylvo-Pastorales, Institut Sylvo-Pastoral de Tabarka, Université de Jendouba, Tabarka, Tunisia.,Faculté des Sciences de Bizerte, Université de Carthage, Bizerte, Tunisia
| | - Saber Jedidi
- Unité de Physiologie Fonctionnelle et Valorisation des Bio-Ressources, Institut Supérieur de Biotechnologie de Beja, Université de Jendouba, Beja, Tunisia.,Laboratoire des Ressources Sylvo-Pastorales, Institut Sylvo-Pastoral de Tabarka, Université de Jendouba, Tabarka, Tunisia
| | - Houcine Selmi
- Laboratoire des Ressources Sylvo-Pastorales, Institut Sylvo-Pastoral de Tabarka, Université de Jendouba, Tabarka, Tunisia
| | - Kais Rtibi
- Unité de Physiologie Fonctionnelle et Valorisation des Bio-Ressources, Institut Supérieur de Biotechnologie de Beja, Université de Jendouba, Beja, Tunisia
| | - Mohamed-Amine Jabri
- Unité de Physiologie Fonctionnelle et Valorisation des Bio-Ressources, Institut Supérieur de Biotechnologie de Beja, Université de Jendouba, Beja, Tunisia
| | - Mourad Jridi
- Laboratoire de Génie Enzymatique et de Microbiologie, Ecole Nationale d'Ingénieurs de Sfax, Université de Sfax, Sfax, Tunisia
| | - Nacim Zouari
- Laboratoire de Génie Enzymatique et de Microbiologie, Ecole Nationale d'Ingénieurs de Sfax, Université de Sfax, Sfax, Tunisia
| | - Lamjed Toumi
- Laboratoire des Ressources Sylvo-Pastorales, Institut Sylvo-Pastoral de Tabarka, Université de Jendouba, Tabarka, Tunisia
| | - Hichem Sebai
- Unité de Physiologie Fonctionnelle et Valorisation des Bio-Ressources, Institut Supérieur de Biotechnologie de Beja, Université de Jendouba, Beja, Tunisia
| |
Collapse
|
14
|
Flouda K, Mercer J, Davies MJ, Hawkins CL. Role of myeloperoxidase-derived oxidants in the induction of vascular smooth muscle cell damage. Free Radic Biol Med 2021; 166:165-177. [PMID: 33631301 DOI: 10.1016/j.freeradbiomed.2021.02.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/14/2021] [Indexed: 01/12/2023]
Abstract
Myeloperoxidase (MPO) is released by activated immune cells and forms the oxidants hypochlorous acid (HOCl) and hypothiocyanous acid (HOSCN) from the competing substrates chloride and thiocyanate. MPO and the overproduction of HOCl are strongly linked with vascular cell dysfunction and inflammation in atherosclerosis. HOCl is highly reactive and causes marked cell dysfunction and death, whereas data with HOSCN are conflicting, and highly dependent on the nature of the cell type. In this study we have examined the reactivity of HOCl and HOSCN with human coronary artery smooth muscle cells (HCASMC), given the key role of this cell type in maintaining vascular function. HOCl reacts rapidly with the cells, resulting in extensive cell death by both necrosis and apoptosis, and increased levels of intracellular calcium. In contrast, HOSCN reacts more slowly, with cell death occurring only after prolonged incubation, and in the absence of the accumulation of intracellular calcium. Exposure of HCASMC to HOCl also influences mitochondrial respiration, decreases glycolysis, lactate release, the production of ATP, cellular thiols and glutathione levels. These changes occurred to varying extents on exposure of the cells to HOSCN, where evidence was also obtained for the reversible modification of cellular thiols. HOCl also induced alterations in the mRNA expression of multiple inflammatory and phenotypic genes. Interestingly, the extent and nature of these changes was highly dependent on the specific cell donor used, with more marked effects observed in cells isolated from diseased compared to healthy vessels. Overall, these data provide new insight into pathways promoting vascular dysfunction during chronic inflammation, support the use of thiocyanate as a means to modulate MPO-induced cellular damage in atherosclerosis.
Collapse
Affiliation(s)
- Konstantina Flouda
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - John Mercer
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, United Kingdom
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
15
|
Cheshchevik VT, Krylova NG, Сheshchevik NG, Lapshina EA, Semenkova GN, Zavodnik IB. Role of mitochondrial calcium in hypochlorite induced oxidative damage of cells. Biochimie 2021; 184:104-115. [PMID: 33607241 DOI: 10.1016/j.biochi.2021.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/09/2021] [Accepted: 02/10/2021] [Indexed: 11/29/2022]
Abstract
Hypochlorite (HOCl) is one of the most important mediators of inflammatory processes. Recent evidence demonstrates that changes in intracellular calcium pool play a significant role in the damaging effects of hypochlorite and other oxidants. Mitochondria are shown to be one of the intracellular targets of hypochlorite. But little is known about the mitochondrial calcium pool changes in HOCl-induced mitochondrial dysfunction. Using isolated rat liver mitochondria, we showed the oxidative damage of mitochondria (GSH oxidation and mixed protein-glutathione formation without membrane lipid peroxidation) and alterations in the mitochondrial functional parameters (decrease of respiratory activity and efficiency of oxidative phosphorylation, NADH and FADH coenzyme levels, and membrane potential) under hypochlorite action (50-300 μM). Simultaneously, the mitochondrial calcium release and swelling were demonstrated. In the presence of EGTA, the damaging effects of HOCl were less pronounced, reflecting direct involvement of mitochondrial Ca2+ in mechanisms of oxidant-induced injury. Furthermore, exposure of HeLa cells to hypochlorite resulted in a considerable increase in cytoplasmic calcium concentrations and a decrease in mitochondrial ones. Applying specific inhibitors of calcium transfer systems, we demonstrated that mitochondria play a key role in the redistribution of cytoplasmic Ca2+ ions under hypochlorite action and act as mediators of calcium release from the endoplasmic reticulum into the cytoplasm.
Collapse
Affiliation(s)
- Vitali T Cheshchevik
- Department of Biotechnology, Polessky State University, ulitsa Dnieprovskoy Flotilii, 23, 225710, Pinsk, Belarus.
| | - Nina G Krylova
- Department of Biophysics, Belarusian State University, Prospekt Nezavisimosti 4, 220030, Minsk, Belarus
| | - Nina G Сheshchevik
- Department of Biotechnology, Polessky State University, ulitsa Dnieprovskoy Flotilii, 23, 225710, Pinsk, Belarus
| | - Elena A Lapshina
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola 50, 230030, Grodno, Belarus
| | - Galina N Semenkova
- Department of Biophysics, Belarusian State University, Prospekt Nezavisimosti 4, 220030, Minsk, Belarus
| | - Ilya B Zavodnik
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola 50, 230030, Grodno, Belarus
| |
Collapse
|
16
|
Li Z, Guo J, Bian Y, Zhang M. Intermedin protects thapsigargin‑induced endoplasmic reticulum stress in cardiomyocytes by modulating protein kinase A and sarco/endoplasmic reticulum Ca 2+‑ATPase. Mol Med Rep 2020; 23:107. [PMID: 33300086 PMCID: PMC7723158 DOI: 10.3892/mmr.2020.11746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Intermedin (IMD) is a calcitonin/calcitonin-related peptide that elicits cardioprotective effects in a variety of heart diseases, such as cardiac hypertrophy and heart failure. However, the molecular mechanism of IMD remains unclear. The present study investigated the effects of IMD on neonatal rat ventricular myocytes treated with thapsigargin. The results of the present study demonstrated that thapsigargin induced apoptosis in cardiomyocytes in a dose- and time-dependent manner. Thapsigargin induced endoplasmic reticulum stress, as determined by increased expression levels of 78-kDa glucose-regulated protein, C/EBP-homologous protein and caspase-12, which were dose-dependently attenuated by pretreatment with IMD. In addition, IMD treatment counteracted the thapsigargin-induced suppression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity and protein expression levels, and cytoplasmic Ca2+ overload. IMD treatment also augmented the phosphorylation of phospholamban, which is a crucial regulator of SERCA. Additionally, treatment with the protein kinase A antagonist H-89 inhibited the IMD-mediated cardioprotective effects, including SERCA activity restoration, anti-Ca2+ overload, endoplasmic reticulum stress inhibition and antiapoptosis effects. In conclusion, the results of the present study suggested that IMD may protect cardiomyocytes against thapsigargin-induced endoplasmic reticulum stress and the associated apoptosis at least partly by activating the protein kinase A/SERCA pathway.
Collapse
Affiliation(s)
- Zhidong Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jia Guo
- Department of Cardiology, Shanxi Medical University First Hospital, Taiyuan, Shanxi 030001, P.R. China
| | - Yunfei Bian
- Department of Cardiology, Shanxi Medical University Second Hospital, Taiyuan, Shanxi 030001, P.R. China
| | - Mingsheng Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
17
|
El Kazzi M, Rayner BS, Chami B, Dennis JM, Thomas SR, Witting PK. Neutrophil-Mediated Cardiac Damage After Acute Myocardial Infarction: Significance of Defining a New Target Cell Type for Developing Cardioprotective Drugs. Antioxid Redox Signal 2020; 33:689-712. [PMID: 32517486 PMCID: PMC7475094 DOI: 10.1089/ars.2019.7928] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Significance: Acute myocardial infarction (AMI) is a leading cause of death worldwide. Post-AMI survival rates have increased with the introduction of angioplasty as a primary coronary intervention. However, reperfusion after angioplasty represents a clinical paradox, restoring blood flow to the ischemic myocardium while simultaneously inducing ion and metabolic imbalances that stimulate immune cell recruitment and activation, mitochondrial dysfunction and damaging oxidant production. Recent Advances: Preclinical data indicate that these metabolic imbalances contribute to subsequent heart failure through sustaining local recruitment of inflammatory leukocytes and oxidative stress, cardiomyocyte death, and coronary microvascular disturbances, which enhance adverse cardiac remodeling. Both left ventricular dysfunction and heart failure are strongly linked to inflammation and immune cell recruitment to the damaged myocardium. Critical Issues: Overall, therapeutic anti-inflammatory and antioxidant agents identified in preclinical trials have failed in clinical trials. Future Directions: The versatile neutrophil-derived heme enzyme, myeloperoxidase (MPO), is gaining attention as an important oxidative mediator of reperfusion injury, vascular dysfunction, adverse ventricular remodeling, and atrial fibrillation. Accordingly, there is interest in therapeutically targeting neutrophils and MPO activity in the setting of heart failure. Herein, we discuss the role of post-AMI inflammation linked to myocardial damage and heart failure, describe previous trials targeting inflammation and oxidative stress post-AMI, highlight the potential adverse impact of neutrophil and MPO, and detail therapeutic options available to target MPO clinically in AMI patients.
Collapse
Affiliation(s)
- Mary El Kazzi
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | | | - Belal Chami
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Joanne Marie Dennis
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Shane Ross Thomas
- Department of Pathology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Paul Kenneth Witting
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| |
Collapse
|
18
|
Guo C, Davies MJ, Hawkins CL. Role of thiocyanate in the modulation of myeloperoxidase-derived oxidant induced damage to macrophages. Redox Biol 2020; 36:101666. [PMID: 32781424 PMCID: PMC7417949 DOI: 10.1016/j.redox.2020.101666] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Myeloperoxidase (MPO) is a vital component of the innate immune system, which produces the potent oxidant hypochlorous acid (HOCl) to kill invading pathogens. However, an overproduction of HOCl during chronic inflammatory conditions causes damage to host cells, which promotes disease, including atherosclerosis. As such, there is increasing interest in the use of thiocyanate (SCN-) therapeutically to decrease inflammatory disease, as SCN- is the favoured substrate for MPO, and a potent competitive inhibitor of HOCl formation. Use of SCN- by MPO forms hypothiocyanous acid (HOSCN), which can be less damaging to mammalian cells. In this study, we examined the ability of SCN- to modulate damage to macrophages induced by HOCl, which is relevant to lesion formation in atherosclerosis. Addition of SCN- prevented HOCl-mediated cell death, altered the extent and nature of thiol oxidation and the phosphorylation of mitogen activated protein kinases. These changes were dependent on the concentration of SCN- and were observed in some cases, at a sub-stoichiometric ratio of SCN-: HOCl. Co-treatment with SCN- also modulated HOCl-induced perturbations in the expression of various antioxidant and inflammatory genes. In general, the data reflect the conversion of HOCl to HOSCN, which can induce reversible modifications that are repairable by cells. However, our data also highlight the ability of HOSCN to increase pro-inflammatory gene expression and cytokine/chemokine release, which may be relevant to the use of SCN- therapeutically in atherosclerosis. Overall, this study provides further insight into the cellular pathways by which SCN- could exert protective effects on supplementation to decrease the development of chronic inflammatory diseases, such as atherosclerosis.
Collapse
Affiliation(s)
- Chaorui Guo
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen, DK-2200, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen, DK-2200, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen, DK-2200, Denmark.
| |
Collapse
|
19
|
Love DT, Guo C, Nikelshparg EI, Brazhe NA, Sosnovtseva O, Hawkins CL. The role of the myeloperoxidase-derived oxidant hypothiocyanous acid (HOSCN) in the induction of mitochondrial dysfunction in macrophages. Redox Biol 2020; 36:101602. [PMID: 32570189 PMCID: PMC7315103 DOI: 10.1016/j.redox.2020.101602] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/20/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
A host of chronic inflammatory diseases are accelerated by the formation of the powerful oxidant hypochlorous acid (HOCl) by myeloperoxidase (MPO). In the presence of thiocyanate (SCN-), the production of HOCl by MPO is decreased in favour of the formation of a milder oxidant, hypothiocyanous acid (HOSCN). The role of HOSCN in disease has not been fully elucidated, though there is increasing interest in using SCN- therapeutically in different disease settings. Unlike HOCl, HOSCN can be detoxified by thioredoxin reductase, and reacts selectively with thiols to result in reversible modifications, which could potentially reduce the extent of MPO-induced damage during chronic inflammation. In this study, we show that exposure of macrophages, a key inflammatory cell type, to HOSCN results in the reversible modification of multiple mitochondrial proteins, leading to increased mitochondrial membrane permeability, decreased oxidative phosphorylation and reduced formation of ATP. The increased permeability and reduction in ATP could be reversed by pre-treatment of the macrophages with cyclosporine A, implicating a role for the mitochondrial permeability transition pore. HOSCN also drives cells to utilise fatty acids as an energetic substrate after the inhibition of oxidative phosphorylation. Raman imaging studies highlighted the ability of HOSCN to perturb the electron transport chain of mitochondria and redistribute these organelles within the cell. Taken together, these data provide new insight into the pathways by which HOSCN can induce cytotoxicity and cellular damage, which may have relevance for the development of inflammatory disease, and therapeutic strategies to reduce HOCl-induced damage by supplementation with SCN-. HOSCN induces the oxidation of mitochondrial thiol proteins and cytochromes. HOSCN alters mitochondrial permeability and ATP production via MPTP formation. HOSCN increases the capacity of cells to use fatty acids as an energetic substrate. Raman imaging shows redistribution of mitochondria after cell exposure to HOSCN.
Collapse
Affiliation(s)
- Dominic T Love
- The Heart Research Institute, 7 Eliza Street, Newtown, NSW, 2042, Australia; Sydney Medical School, University of Sydney, NSW, 2006, Australia
| | - Chaorui Guo
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Evelina I Nikelshparg
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie Gory 1/12, Moscow, 119234, Russia
| | - Nadezda A Brazhe
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie Gory 1/12, Moscow, 119234, Russia
| | - Olga Sosnovtseva
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Clare L Hawkins
- The Heart Research Institute, 7 Eliza Street, Newtown, NSW, 2042, Australia; Sydney Medical School, University of Sydney, NSW, 2006, Australia; Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
20
|
He P, Talukder MAH, Gao F. Oxidative Stress and Microvessel Barrier Dysfunction. Front Physiol 2020; 11:472. [PMID: 32536875 PMCID: PMC7268512 DOI: 10.3389/fphys.2020.00472] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Clinical and experimental evidence indicate that increased vascular permeability contributes to many disease-associated vascular complications. Oxidative stress with increased production of reactive oxygen species (ROS) has been implicated in a wide variety of pathological conditions, including inflammation and many cardiovascular diseases. It is thus important to identify the role of ROS and their mechanistic significance in microvessel barrier dysfunction under pathological conditions. The role of specific ROS and their cross talk in pathological processes is complex. The mechanisms of ROS-induced increases in vascular permeability remain poorly understood. The sources of ROS in diseases have been extensively reviewed at enzyme levels. This review will instead focus on the underlying mechanisms of ROS release by leukocytes, the differentiate effects and signaling mechanisms of individual ROS on endothelial cells, pericytes and microvessel barrier function, as well as the interplay of reactive oxygen species, nitric oxide, and nitrogen species in ROS-mediated vascular barrier dysfunction. As a counter balance of excessive ROS, nuclear factor erythroid 2 related factor 2 (Nrf2), a redox-sensitive cell-protective transcription factor, will be highlighted as a potential therapeutic target for antioxidant defenses. The advantages and limitations of different experimental approaches used for the study of ROS-induced endothelial barrier function are also discussed. This article will outline the advances emerged mainly from in vivo and ex vivo studies and attempt to consolidate some of the opposing views in the field, and hence provide a better understanding of ROS-mediated microvessel barrier dysfunction and benefit the development of therapeutic strategies.
Collapse
Affiliation(s)
- Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| | - M A Hassan Talukder
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| | - Feng Gao
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
21
|
Davies MJ, Hawkins CL. The Role of Myeloperoxidase in Biomolecule Modification, Chronic Inflammation, and Disease. Antioxid Redox Signal 2020; 32:957-981. [PMID: 31989833 DOI: 10.1089/ars.2020.8030] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Significance: The release of myeloperoxidase (MPO) by activated leukocytes is critical in innate immune responses. MPO produces hypochlorous acid (HOCl) and other strong oxidants, which kill bacteria and other invading pathogens. However, MPO also drives the development of numerous chronic inflammatory pathologies, including atherosclerosis, neurodegenerative disease, lung disease, arthritis, cancer, and kidney disease, which are globally responsible for significant patient mortality and morbidity. Recent Advances: The development of imaging approaches to precisely identify the localization of MPO and the molecular targets of HOCl in vivo is an important advance, as typically the involvement of MPO in inflammatory disease has been inferred by its presence, together with the detection of biomarkers of HOCl, in biological fluids or diseased tissues. This will provide valuable information in regard to the cell types responsible for releasing MPO in vivo, together with new insight into potential therapeutic opportunities. Critical Issues: Although there is little doubt as to the value of MPO inhibition as a protective strategy to mitigate tissue damage during chronic inflammation in experimental models, the impact of long-term inhibition of MPO as a therapeutic strategy for human disease remains uncertain, in light of the potential effects on innate immunity. Future Directions: The development of more targeted MPO inhibitors or a treatment regimen designed to reduce MPO-associated host tissue damage without compromising pathogen killing by the innate immune system is therefore an important future direction. Similarly, a partial MPO inhibition strategy may be sufficient to maintain adequate bacterial activity while decreasing the propagation of inflammatory pathologies.
Collapse
Affiliation(s)
- Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
22
|
Reyes L, Hawkins CL, Rayner BS. Characterization of the cellular effects of myeloperoxidase-derived oxidants on H9c2 cardiac myoblasts. Arch Biochem Biophys 2019; 665:132-142. [DOI: 10.1016/j.abb.2019.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
|
23
|
Etwebi Z, Landesberg G, Preston K, Eguchi S, Scalia R. Mechanistic Role of the Calcium-Dependent Protease Calpain in the Endothelial Dysfunction Induced by MPO (Myeloperoxidase). Hypertension 2019; 71:761-770. [PMID: 29507101 DOI: 10.1161/hypertensionaha.117.10305] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/02/2017] [Accepted: 01/12/2018] [Indexed: 01/07/2023]
Abstract
MPO (myeloperoxidase) is a peroxidase enzyme secreted by activated leukocytes that plays a pathogenic role in cardiovascular disease, mainly by initiating endothelial dysfunction. The molecular mechanisms of the endothelial damaging action of MPO remain though largely elusive. Calpain is a calcium-dependent protease expressed in the vascular wall. Activation of calpains has been implicated in inflammatory disorders of the vasculature. Using endothelial cells and genetically modified mice, this study identifies the µ-calpain isoform as novel downstream signaling target of MPO in endothelial dysfunction. Mouse lung microvascular endothelial cells were stimulated with 10 nmol/L MPO for 180 minutes. MPO denitrosylated µ-calpain C-terminus domain, and time dependently activated µ-calpain, but not the m-calpain isoform. MPO also reduced Thr172 AMPK (AMP-activated protein kinase) and Ser1177 eNOS (endothelial nitric oxide synthase) phosphorylation via upregulation of PP2A (protein phosphatase 2) expression. At the functional level, MPO increased endothelial VCAM-1 (vascular cell adhesion molecule 1) abundance and the adhesion of leukocytes to the mouse aorta. In MPO-treated endothelial cells, pharmacological inhibition of calpain activity attenuated expression of VCAM-1 and PP2A, and restored Thr172 AMPK and Ser1177 eNOS phosphorylation. Compared with wild-type mice, µ-calpain deficient mice experienced reduced leukocyte adhesion to the aortic endothelium in response to MPO. Our data first establish a role for calpain in the endothelial dysfunction and vascular inflammation of MPO. The MPO/calpain/PP2A signaling pathway may provide novel pharmacological targets for the treatment of inflammatory vascular disorders.
Collapse
Affiliation(s)
- Zienab Etwebi
- From the Department of Physiology and the Cardiovascular Research Center, Temple University, Philadelphia, PA
| | - Gavin Landesberg
- From the Department of Physiology and the Cardiovascular Research Center, Temple University, Philadelphia, PA
| | - Kyle Preston
- From the Department of Physiology and the Cardiovascular Research Center, Temple University, Philadelphia, PA
| | - Satoru Eguchi
- From the Department of Physiology and the Cardiovascular Research Center, Temple University, Philadelphia, PA
| | - Rosario Scalia
- From the Department of Physiology and the Cardiovascular Research Center, Temple University, Philadelphia, PA.
| |
Collapse
|
24
|
Brini M, Leanza L, Szabo I. Lipid-Mediated Modulation of Intracellular Ion Channels and Redox State: Physiopathological Implications. Antioxid Redox Signal 2018; 28:949-972. [PMID: 28679281 DOI: 10.1089/ars.2017.7215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Ion channels play an important role in the regulation of organelle function within the cell, as proven by increasing evidence pointing to a link between altered function of intracellular ion channels and different pathologies ranging from cancer to neurodegenerative diseases, ischemic damage, and lysosomal storage diseases. Recent Advances: A link between these pathologies and redox state as well as lipid homeostasis and ion channel function is in the focus of current research. Critical Issues: Ion channels are target of modulation by lipids and lipid messengers, although in most cases the mechanistic details have not been clarified yet. Ion channel function importantly impacts production of reactive oxygen species (ROS), especially in the case of mitochondria and lysosomes. ROS, in turn, may modulate the function of intracellular channels triggering thereby a feedback control under physiological conditions. If produced in excess, ROS can be harmful to lipids and may produce oxidized forms of these membrane constituents that ultimately affect ion channel function by triggering a "circulus vitiosus." Future Directions: The present review summarizes our current knowledge about the contribution of intracellular channels to oxidative stress and gives examples of how these channels are modulated by lipids and how this modulation may affect ROS production in ROS-related diseases. Future studies need to address the importance of the regulation of intracellular ion channels and related oxidative stress by lipids in various physiological and pathological contexts. Antioxid. Redox Signal. 28, 949-972.
Collapse
Affiliation(s)
- Marisa Brini
- Department of Biology, University of Padova, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy.,CNR Institute of Neuroscience, Padova, Italy
| |
Collapse
|
25
|
A pivotal role for NF-κB in the macrophage inflammatory response to the myeloperoxidase oxidant hypothiocyanous acid. Arch Biochem Biophys 2018; 642:23-30. [DOI: 10.1016/j.abb.2018.01.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/04/2018] [Accepted: 01/29/2018] [Indexed: 01/04/2023]
|
26
|
Casciaro M, Di Salvo E, Pace E, Ventura-Spagnolo E, Navarra M, Gangemi S. Chlorinative stress in age-related diseases: a literature review. IMMUNITY & AGEING 2017; 14:21. [PMID: 29163665 PMCID: PMC5686828 DOI: 10.1186/s12979-017-0104-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/07/2017] [Indexed: 12/27/2022]
Abstract
Aging is an agglomerate of biological long-lasting processes that result being inevitable. Main actors in this scenario are both long-term inflammation and oxidative stress. It has been proved that oxidative stress induce alteration in proteins and this fact itself is critically important in the pathophysiological mechanisms leading to diseases typical of aging. Among reactive species, chlorine ones such as hypochlorous acid (HOCl) are cytotoxic oxidants produced by activated neutrophils during chronic inflammation processes. HOCl can also cause damages by reacting with biological molecules. HOCl is generated by myeloperoxidase (MPO) and augmented serum levels of MPO have been described in acute and chronic inflammatory conditions in cardiovascular patients and has been implicated in many inflammatory diseases such as atherosclerosis, neurodegenerative conditions, and some cancers. Due to these data, we decided to conduct an up-to-date review evaluating chlorinative stress effects on every age-related disease linked; potential anti-oxidant countermeasures were also assessed. Results obtained associated HOCl generation to the aging processes and confirmed its connection with diseases like neurodegenerative and cardiovascular pathologies, atherosclerosis and cancer; chlorination was mainly linked to diseases where molecular (protein) alteration constitute the major suspected cause: i.e. inflammation, tissue lesions, DNA damages, apoptosis and oxidative stress itself. According data collected, a healthy lifestyle together with some dietary suggestion and/or the administration of nutracetical antioxidant integrators could balance the effects of chlorinative stress and, in some cases, slow down or prevent the onset of age-releated diseases.
Collapse
Affiliation(s)
- Marco Casciaro
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Policlinico "G. Martino", University of Messina, Messina, Italy
| | - Eleonora Di Salvo
- IBIM-CNR Institute of Biomedicine and Molecular Immunology, National Research Council, 90100 Palermo, Italy
| | - Elisabetta Pace
- IBIM-CNR Institute of Biomedicine and Molecular Immunology, National Research Council, 90100 Palermo, Italy
| | - Elvira Ventura-Spagnolo
- Legal Medicine Section, Department for Health Promotion and Mother-Child Care, University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Policlinico "G. Martino", University of Messina, Messina, Italy
| |
Collapse
|
27
|
Carlisle M, Lam A, Svendsen ER, Aggarwal S, Matalon S. Chlorine-induced cardiopulmonary injury. Ann N Y Acad Sci 2016; 1374:159-67. [PMID: 27303906 DOI: 10.1111/nyas.13091] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chlorine (Cl2 ) is utilized worldwide for a diverse range of industrial applications, including pulp bleaching, sanitation, and pharmaceutical development. Though Cl2 has widespread use, little is known regarding the mechanisms of toxicity associated with Cl2 exposure, which occurs during industrial accidents or acts of terrorism. Previous instances of Cl2 exposure have led to reported episodes of respiratory distress that result in high morbidity and mortality. Furthermore, studies suggest that acute Cl2 exposure also results in systemic vascular injury and subsequent myocardial contractile dysfunction. Here, we review both lung and cardiac pathology associated with acute Cl2 inhalation and discuss recently published data that suggest that mitochondrial dysfunction underlies the pathogenesis of Cl2 -induced toxicity. Last, we discuss our findings that suggest that upregulation of autophagy protects against Cl2 -induced lung inflammation and can be a potential therapeutic target for ameliorating the toxic effects of Cl2 exposure.
Collapse
Affiliation(s)
- Matthew Carlisle
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Division of Molecular and Translational Biomedicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Adam Lam
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Division of Molecular and Translational Biomedicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Erik R Svendsen
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina.,Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Saurabh Aggarwal
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Division of Molecular and Translational Biomedicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Division of Molecular and Translational Biomedicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
28
|
Love DT, Barrett TJ, White MY, Cordwell SJ, Davies MJ, Hawkins CL. Cellular targets of the myeloperoxidase-derived oxidant hypothiocyanous acid (HOSCN) and its role in the inhibition of glycolysis in macrophages. Free Radic Biol Med 2016; 94:88-98. [PMID: 26898502 DOI: 10.1016/j.freeradbiomed.2016.02.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/12/2016] [Accepted: 02/15/2016] [Indexed: 12/20/2022]
Abstract
Myeloperoxidase (MPO) released at sites of inflammation catalyzes the formation of the oxidants hypochlorous acid (HOCl) and hypothiocyanous acid (HOSCN) from H2O2 and halide and pseudo-halide ions. HOCl, a major oxidant produced under physiological conditions reacts rapidly with many biological molecules, and is strongly linked with tissue damage during inflammatory disease. The role of HOSCN in disease is less clear, though it can initiate cellular damage by pathways involving the selective oxidation of thiol-containing proteins. Utilizing a thiol-specific proteomic approach, we explored the cellular targets of HOSCN in macrophages (J774A.1). We report that multiple thiol-containing proteins involved in metabolism and glycolysis; fructose bisphosphate aldolase, triosephosphate isomerase, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and creatine kinase, together with a number of chaperone, antioxidant and structural proteins, were modified in a reversible manner in macrophages treated with HOSCN. The modification of the metabolic enzymes was associated with a decrease in basal glycolysis, glycolytic reserve, glycolytic capacity and lactate release, which was only partly reversible on further incubation in the absence of HOSCN. Inhibition of glycolysis preceded cell death and was seen in cells exposed to low concentrations (≤25µM) of HOSCN. The ability of HOSCN to inhibit glycolysis and perturb energy production is likely to contribute to the cell death seen in macrophages on further incubation after the initial treatment period, which may be relevant for the propagation of inflammatory disease in smokers, who have elevated plasma levels of the HOSCN precursor, thiocyanate.
Collapse
Affiliation(s)
- Dominic T Love
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Tessa J Barrett
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Melanie Y White
- School of Molecular Bioscience, School of Medical Sciences, and Charles Perkins Centre, University of Sydney, 2006, Australia
| | - Stuart J Cordwell
- School of Molecular Bioscience, School of Medical Sciences, and Charles Perkins Centre, University of Sydney, 2006, Australia
| | - Michael J Davies
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Blegdamsvej 3, Copenhagen 2200, Denmark
| | - Clare L Hawkins
- The Heart Research Institute, 7 Eliza St., Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
29
|
Ahmad S, Ahmad A, Hendry-Hofer TB, Loader JE, Claycomb WC, Mozziconacci O, Schöneich C, Reisdorph N, Powell RL, Chandler JD, Day BJ, Veress LA, White CW. Sarcoendoplasmic reticulum Ca(2+) ATPase. A critical target in chlorine inhalation-induced cardiotoxicity. Am J Respir Cell Mol Biol 2016; 52:492-502. [PMID: 25188881 DOI: 10.1165/rcmb.2014-0005oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autopsy specimens from human victims or experimental animals that die due to acute chlorine gas exposure present features of cardiovascular pathology. We demonstrate acute chlorine inhalation-induced reduction in heart rate and oxygen saturation in rats. Chlorine inhalation elevated chlorine reactants, such as chlorotyrosine and chloramine, in blood plasma. Using heart tissue and primary cardiomyocytes, we demonstrated that acute high-concentration chlorine exposure in vivo (500 ppm for 30 min) caused decreased total ATP content and loss of sarcoendoplasmic reticulum calcium ATPase (SERCA) activity. Loss of SERCA activity was attributed to chlorination of tyrosine residues and oxidation of an important cysteine residue, cysteine-674, in SERCA, as demonstrated by immunoblots and mass spectrometry. Using cardiomyocytes, we found that chlorine-induced cell death and damage to SERCA could be decreased by thiocyanate, an important biological antioxidant, and by genetic SERCA2 overexpression. We also investigated a U.S. Food and Drug Administration-approved drug, ranolazine, used in treatment of cardiac diseases, and previously shown to stabilize SERCA in animal models of ischemia-reperfusion. Pretreatment with ranolazine or istaroxime, another SERCA activator, prevented chlorine-induced cardiomyocyte death. Further investigation of responsible mechanisms showed that ranolazine- and istaroxime-treated cells preserved mitochondrial membrane potential and ATP after chlorine exposure. Thus, these studies demonstrate a novel critical target for chlorine in the heart and identify potentially useful therapies to mitigate toxicity of acute chlorine exposure.
Collapse
Affiliation(s)
- Shama Ahmad
- 1 Pediatric Airway Research Center, Department of Pediatrics, University of Colorado, Denver, Aurora, Colorado
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cook NL, Moeke CH, Fantoni LI, Pattison DI, Davies MJ. The myeloperoxidase-derived oxidant hypothiocyanous acid inhibits protein tyrosine phosphatases via oxidation of key cysteine residues. Free Radic Biol Med 2016; 90:195-205. [PMID: 26616646 DOI: 10.1016/j.freeradbiomed.2015.11.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 11/13/2015] [Accepted: 11/20/2015] [Indexed: 12/31/2022]
Abstract
Phosphorylation of protein tyrosine residues is critical to cellular processes, and is regulated by kinases and phosphatases (PTPs). PTPs contain a redox-sensitive active site Cys residue, which is readily oxidized. Myeloperoxidase, released from activated leukocytes, catalyzes thiocyanate ion (SCN(-)) oxidation by H2O2 to form hypothiocyanous acid (HOSCN), an oxidant that targets Cys residues. Dysregulated phosphorylation and elevated MPO levels have been associated with chronic inflammatory diseases where HOSCN can be generated. Previous studies have shown that HOSCN inhibits isolated PTP1B and induces cellular dysfunction in cultured macrophage-like cells. The present study extends this previous work and shows that physiologically-relevant concentrations of HOSCN alter the activity and structure of other members of the wider PTP family (including leukocyte antigen-related PTP, PTP-LAR; T-cell PTP, TC-PTP; CD45 and Src homology phosphatase-1, Shp-1) by targeting Cys residues. Isolated PTP activity, and activity in lysates of human monocyte-derived macrophages (HMDM) was inhibited by 0-100 µM HOSCN with this being accompanied by reversible oxidation of Cys residues, formation of sulfenic acids or sulfenyl-thiocyanates (detected by Western blotting, and LC-MS as dimedone adducts), and structural changes. LC-MS/MS peptide mass-mapping has provided data on the modified Cys residues in PTP-LAR. This study indicates that inflammation-induced oxidants, and particularly myeloperoxidase-derived species, can modulate the activity of multiple members of the PTP superfamily via oxidation of Cys residues to sulfenic acids. This alteration of the balance of PTP/kinase activity may perturb protein phosphorylation and disrupt cell signaling with subsequent induction of apoptosis at sites of inflammation.
Collapse
Affiliation(s)
- Naomi L Cook
- The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia
| | - Cassidy H Moeke
- The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Luca I Fantoni
- The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia
| | - David I Pattison
- The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Michael J Davies
- The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Building 4.5, Blegdamsvej 3, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
31
|
Zaky A, Ahmad A, Dell'Italia LJ, Jahromi L, Reisenberg LA, Matalon S, Ahmad S. Inhaled matters of the heart. ACTA ACUST UNITED AC 2015; 2. [PMID: 26665179 DOI: 10.14800/crm.997] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inhalations of atmospheric pollutants, especially particulate matters, are known to cause severe cardiac effects and to exacerbate preexisting heart disease. Heart failure is an important sequellae of gaseous inhalation such as that of carbon monoxide. Similarly, other gases such as sulphur dioxide are known to cause detrimental cardiovascular events. However, mechanisms of these cardiac toxicities are so far unknown. Increased susceptibility of the heart to oxidative stress may play a role. Low levels of antioxidants in the heart as compared to other organs and high levels of reactive oxygen species produced due to the high energetic demand and metabolic rate in cardiac muscle are important in rendering this susceptibility. Acute inhalation of high concentrations of halogen gases is often fatal. Severe respiratory injury and distress occurs upon inhalation of halogens gases, such as chlorine and bromine; however, studies on their cardiac effects are scant. We have demonstrated that inhalation of high concentrations of halogen gases cause significant cardiac injury, dysfunction, and failure that can be critical in causing mortalities following exposures. Our studies also demonstrated that cardiac dysfunction occurs as a result of a direct insult independent of coexisting hypoxia, since it is not fully reversed by oxygen supplementation. Therefore, studies on offsite organ effects of inhaled toxic gases can impact development of treatment strategies upon accidental or deliberate exposures to these agents. Here we summarize the knowledge of cardiovascular effects of common inhaled toxic gases with the intent to highlight the importance of consideration of cardiac symptoms while treating the victims.
Collapse
Affiliation(s)
- Ahmed Zaky
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Alabama ; Department of Medicine, Birmingham Veteran Affairs Medical Center, Birmingham, Alabama and Division of Cardiovascular Disease, University of Alabama Medical Center, Birmingham, Alabama
| | - Aftab Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Alabama
| | - Louis J Dell'Italia
- Department of Medicine, Birmingham Veteran Affairs Medical Center, Birmingham, Alabama and Division of Cardiovascular Disease, University of Alabama Medical Center, Birmingham, Alabama
| | - Leila Jahromi
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Alabama
| | - Lee Ann Reisenberg
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Alabama
| | - Shama Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Alabama
| |
Collapse
|
32
|
Görlach A, Bertram K, Hudecova S, Krizanova O. Calcium and ROS: A mutual interplay. Redox Biol 2015; 6:260-271. [PMID: 26296072 PMCID: PMC4556774 DOI: 10.1016/j.redox.2015.08.010] [Citation(s) in RCA: 1050] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 02/07/2023] Open
Abstract
Calcium is an important second messenger involved in intra- and extracellular signaling cascades and plays an essential role in cell life and death decisions. The Ca2+ signaling network works in many different ways to regulate cellular processes that function over a wide dynamic range due to the action of buffers, pumps and exchangers on the plasma membrane as well as in internal stores. Calcium signaling pathways interact with other cellular signaling systems such as reactive oxygen species (ROS). Although initially considered to be potentially detrimental byproducts of aerobic metabolism, it is now clear that ROS generated in sub-toxic levels by different intracellular systems act as signaling molecules involved in various cellular processes including growth and cell death. Increasing evidence suggests a mutual interplay between calcium and ROS signaling systems which seems to have important implications for fine tuning cellular signaling networks. However, dysfunction in either of the systems might affect the other system thus potentiating harmful effects which might contribute to the pathogenesis of various disorders. Calcium and ROS act as signaling molecules inside the cell and their pathways can interact. The mutual interplay of calcium and ROS is required for the fine tuning of signaling. Failure in the interplay results in dysfunction and pathologies.
Collapse
Affiliation(s)
- Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Katharina Bertram
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Germany
| | - Sona Hudecova
- Center for Molecular Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Olga Krizanova
- Center for Molecular Medicine, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
33
|
Yamaguchi R, Kawata J, Yamamoto T, Ishimaru Y, Sakamoto A, Ono T, Narahara S, Sugiuchi H, Hirose E, Yamaguchi Y. Mechanism of interferon-gamma production by monocytes stimulated with myeloperoxidase and neutrophil extracellular traps. Blood Cells Mol Dis 2015; 55:127-33. [PMID: 26142328 DOI: 10.1016/j.bcmd.2015.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/29/2015] [Indexed: 12/27/2022]
Abstract
Neutrophil extracellular traps (NETs) have an important role in antimicrobial innate immunity and release substances that may modulate the immune response. We investigated the effects of soluble factors from NETs and neutrophil granule proteins on human monocyte function by using the Transwell system to prevent cell-cell contact. NET formation was induced by exposing human neutrophils to phorbol myristate acetate (PMA). When monocytes were incubated with PMA alone, expression of interleukin (IL)-4, IL-6, IL-8, and tumor necrosis factor (TNF)-alpha mRNA was upregulated, but IL-10, IL-12, and interferon (IFN)-gamma mRNA were not detected. Incubation of monocytes with NETs enhanced the expression of IL-10 and IFN-gamma mRNA, but not IL-12 mRNA. Myeloperoxidase stimulated IFN-gamma production by monocytes in a dose-dependent manner. Both a nuclear factor-kappaB inhibitor (PDTC) and an intracellular calcium antagonist (TMB-8) prevented upregulation of IFN-gamma production. Neither a combined p38alpha and p38beta inhibitor (SB203580) nor an extracellular signal-regulated kinase inhibitor (PD98059) suppressed IFN-gamma production. Interestingly, a combined p38gamma and p38delta inhibitor (BIRB796) significantly decreased IFN-gamma production. These findings suggest that myeloperoxidase induces IFN-gamma production by monocytes via p38gamma/delta mitogen-activated protein kinase.
Collapse
Affiliation(s)
- Rui Yamaguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Jin Kawata
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Toshitaka Yamamoto
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Yasuji Ishimaru
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Arisa Sakamoto
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Tomomichi Ono
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Shinji Narahara
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Hiroyuki Sugiuchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Eiji Hirose
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Yasuo Yamaguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan.
| |
Collapse
|
34
|
Csató V, Pető A, Fülöp GÁ, Rutkai I, Pásztor ET, Fagyas M, Kalász J, Édes I, Tóth A, Papp Z. Myeloperoxidase evokes substantial vasomotor responses in isolated skeletal muscle arterioles of the rat. Acta Physiol (Oxf) 2015; 214:109-23. [PMID: 25760778 PMCID: PMC4654238 DOI: 10.1111/apha.12488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/19/2015] [Accepted: 03/09/2015] [Indexed: 12/20/2022]
Abstract
Aims Myeloperoxidase (MPO) catalyses the formation of a wide variety of oxidants, including hypochlorous acid (HOCl), and contributes to cardiovascular disease progression. We hypothesized that during its action MPO evokes substantial vasomotor responses. Methods Following exposure to MPO (1.92 mU mL−1) in the presence of increasing concentrations of hydrogen peroxide (H2O2), changes in arteriolar diameter of isolated gracilis skeletal muscle arterioles (SMAs) and coronary arterioles (CAs) and in the isometric force in basilar arteries (BAs) of the rat were monitored. Results Myeloperoxidase increased vascular tone to different degrees in CAs, SMAs and BAs. The mechanism of increased vasoconstriction was studied in detail in SMAs. MPO-evoked vasoconstrictions were prevented by the MPO inhibitor 4-aminobenzhydrazide (50 μm), by endothelium removal in the SMAs. Surprisingly, the HOCl scavenger L-methionine (100 μm), the thromboxane A2 (TXA2) antagonist SQ-29548 (1 μm) or the non-specific cyclooxygenase (COX) antagonist indomethacin (1 μm) converted the MPO-evoked vasoconstrictions to pronounced vasodilations in SMAs, not seen in the presence of H2O2. In contrast to noradrenaline-induced vasoconstrictions, the MPO-evoked vasoconstrictions were not accompanied by significant increases in arteriolar [Ca2+] levels in SMAs. Conclusion These data showed that H2O2-derived HOCl to be a potent vasoconstrictor upon MPO application. HOCl activated the COX pathway, causing the synthesis and release of a TXA2-like substance to increase the Ca2+ sensitivity of the contractile apparatus in vascular smooth muscle cells and thereby to augment H2O2-evoked vasoconstrictions. Nevertheless, inhibition of the HOCl–COX–TXA2 pathway unmasked the effects of additional MPO-derived radicals with a marked vasodilatory potential in SMAs.
Collapse
Affiliation(s)
- V. Csató
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - A. Pető
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - G. Á. Fülöp
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - I. Rutkai
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - E. T. Pásztor
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - M. Fagyas
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - J. Kalász
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - I. Édes
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - A. Tóth
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - Z. Papp
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| |
Collapse
|
35
|
Rayner BS, Love DT, Hawkins CL. Comparative reactivity of myeloperoxidase-derived oxidants with mammalian cells. Free Radic Biol Med 2014; 71:240-255. [PMID: 24632382 DOI: 10.1016/j.freeradbiomed.2014.03.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 12/21/2022]
Abstract
Myeloperoxidase is an important heme enzyme released by activated leukocytes that catalyzes the reaction of hydrogen peroxide with halide and pseudo-halide ions to form various hypohalous acids. Hypohalous acids are chemical oxidants that have potent antibacterial, antiviral, and antifungal properties and, as such, play key roles in the human immune system. However, increasing evidence supports an alternative role for myeloperoxidase-derived oxidants in the development of disease. Excessive production of hypohalous acids, particularly during chronic inflammation, leads to the initiation and accumulation of cellular damage that has been implicated in many human pathologies including atherosclerosis, neurodegenerative disease, lung disease, arthritis, inflammatory cancers, and kidney disease. This has sparked a significant interest in developing a greater understanding of the mechanisms involved in myeloperoxidase-derived oxidant-induced mammalian cell damage. This article reviews recent developments in our understanding of the cellular reactivity of hypochlorous acid, hypobromous acid, and hypothiocyanous acid, the major oxidants produced by myeloperoxidase under physiological conditions.
Collapse
Affiliation(s)
- Benjamin S Rayner
- Inflammation Group, The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Dominic T Love
- Inflammation Group, The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Clare L Hawkins
- Inflammation Group, The Heart Research Institute, Newtown, Sydney, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
36
|
Jin Y, He L, Wang Q, Chen Y, Ren X, Tang H, Song X, Ding L, Qi Q, Huang Z, Yu J, Yao Y. Serum calcium levels are not associated with coronary heart disease. Vasc Health Risk Manag 2013; 9:517-20. [PMID: 24043943 PMCID: PMC3772709 DOI: 10.2147/vhrm.s49136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background Numerous studies have reported that low calcium intake is related to a higher prevalence of cardiovascular disease. However, the relationship between serum calcium and coronary heart disease is unclear. The purpose of this study was to compare serum calcium levels in patients with coronary heart disease and those in healthy individuals. Methods This retrospective, case-control study conducted in the People’s Republic of China comprised 380 cases and 379 controls. Serum calcium levels, blood lipids, and anthropometric measurements were measured in both groups. The Student’s unpaired t-test or Chi-square test was used to compare differences between cases and controls. Pearson’s partial correlation coefficient was used to determine the association between serum calcium, blood lipids, and blood pressure in both groups. Results Our results indicate that the average level of serum calcium in cases was higher than in controls. Serum calcium levels showed no correlation with any parameter except for triglycerides in either group. Conclusion Overall, these data suggest that serum calcium has no influence on coronary heart disease or triglyceride levels in the general population.
Collapse
Affiliation(s)
- Yuelong Jin
- Department of Preventive Medicine, Wannan Medical College, Wuhu, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Methionine oxidation activates a transcription factor in response to oxidative stress. Proc Natl Acad Sci U S A 2013; 110:9493-8. [PMID: 23690622 DOI: 10.1073/pnas.1300578110] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oxidant-mediated antibacterial response systems are broadly used to control bacterial proliferation. Hypochlorite (HOCl) is an important component of the innate immune system produced in neutrophils and specific epithelia. Its antimicrobial activity is due to damaging cellular macromolecules. Little is known about how bacteria escape HOCl-inflicted damage. Recently, the transcription factor YjiE was identified that specifically protects Escherichia coli from HOCl killing. According to its function, YjiE is now renamed HypT (hypochlorite-responsive transcription factor). Here we unravel that HypT is activated by methionine oxidation to methionine sulfoxide. Interestingly, so far only inactivation of cellular proteins by methionine oxidation has been reported. Mutational analysis revealed three methionines that are essential to confer HOCl resistance. Their simultaneous substitution by glutamine, mimicking the methionine sulfoxide state, increased the viability of E. coli cells upon HOCl stress. Triple glutamine substitution generates a constitutively active HypT that regulates target genes independently of HOCl stress and permanently down-regulates intracellular iron levels. Inactivation of HypT depends on the methionine sulfoxide reductases A/B. Thus, microbial protection mechanisms have evolved along the evolution of antimicrobial control systems, allowing bacteria to survive within the host environment.
Collapse
|
38
|
Cline MR, Chavez TA, Toscano JP. Oxidation of N-hydroxy-l-arginine by hypochlorous acid to form nitroxyl (HNO). J Inorg Biochem 2013; 118:148-54. [DOI: 10.1016/j.jinorgbio.2012.09.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 09/01/2012] [Accepted: 09/01/2012] [Indexed: 01/15/2023]
|
39
|
Moccia F, Berra-Romani R, Tanzi F. Update on vascular endothelial Ca 2+ signalling: A tale of ion channels, pumps and transporters. World J Biol Chem 2012; 3:127-58. [PMID: 22905291 PMCID: PMC3421132 DOI: 10.4331/wjbc.v3.i7.127] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/04/2012] [Accepted: 07/11/2012] [Indexed: 02/05/2023] Open
Abstract
A monolayer of endothelial cells (ECs) lines the lumen of blood vessels and forms a multifunctional transducing organ that mediates a plethora of cardiovascular processes. The activation of ECs from as state of quiescence is, therefore, regarded among the early events leading to the onset and progression of potentially lethal diseases, such as hypertension, myocardial infarction, brain stroke, and tumor. Intracellular Ca2+ signals have long been know to play a central role in the complex network of signaling pathways regulating the endothelial functions. Notably, recent work has outlined how any change in the pattern of expression of endothelial channels, transporters and pumps involved in the modulation of intracellular Ca2+ levels may dramatically affect whole body homeostasis. Vascular ECs may react to both mechanical and chemical stimuli by generating a variety of intracellular Ca2+ signals, ranging from brief, localized Ca2+ pulses to prolonged Ca2+ oscillations engulfing the whole cytoplasm. The well-defined spatiotemporal profile of the subcellular Ca2+ signals elicited in ECs by specific extracellular inputs depends on the interaction between Ca2+ releasing channels, which are located both on the plasma membrane and in a number of intracellular organelles, and Ca2+ removing systems. The present article aims to summarize both the past and recent literature in the field to provide a clear-cut picture of our current knowledge on the molecular nature and the role played by the components of the Ca2+ machinery in vascular ECs under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Francesco Moccia
- Francesco Moccia, Franco Tanzi, Department of Biology and Biotechnologies "Lazzaro Spallanzani", Laboratory of Physiology, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | | | | |
Collapse
|
40
|
Friedrichs K, Baldus S, Klinke A. Fibrosis in Atrial Fibrillation - Role of Reactive Species and MPO. Front Physiol 2012; 3:214. [PMID: 22723783 PMCID: PMC3379725 DOI: 10.3389/fphys.2012.00214] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 05/30/2012] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrosis with enhanced turnover and deposition of matrix proteins leads to inhomogeneous atrial electrical conduction and gives rise to electrical reentry circuits resulting in atrial fibrillation. The multifactorial pathogenesis of atrial fibrosis involves resident cardiac cells as well as infiltrating leukocytes, both generating and sequestering matrix metalloproteinases (MMPs), a key enzyme family involved in fibrosis. A growing body of evidence points toward an important role of reactive oxygen species (ROS) in the release and activation of pro-MMPs and the stimulation of pro-fibrotic cascades. Myeloperoxidase (MPO), a bactericidal enzyme released from activated polymorphonuclear neutrophils (PMN) is not only associated with a variety of cardiovascular diseases, but has also been shown to be mechanistically linked to atrial fibrosis and fibrillation. MPO catalyzes the generation of reactive species like hypochlorous acid, which affect intracellular signaling cascades in various cells and advance activation of pro-MMPs and deposition of atrial collagen resulting in atrial arrhythmias. Thus, inflammatory mechanisms effectively promote atrial structural remodeling and importantly contribute to the initiation and perpetuation of atrial fibrillation.
Collapse
Affiliation(s)
- Kai Friedrichs
- Department of Electrophysiology, Cardiovascular Research Center, University Heart Center HamburgHamburg, Germany
| | - Stephan Baldus
- Department of General and Interventional Cardiology, Cardiovascular Research Center, University Heart Center HamburgHamburg, Germany
| | - Anna Klinke
- Department of General and Interventional Cardiology, Cardiovascular Research Center, University Heart Center HamburgHamburg, Germany
| |
Collapse
|