1
|
Li M, Bai Y, Wang Y, Xing H, Zhang Y, Ding W, Ye H, Ma L, Wang W, Bao T. A Review of Clinical Advances and Challenges in Clozapine-Induced Myocarditis. Neuropsychiatr Dis Treat 2025; 21:525-538. [PMID: 40070369 PMCID: PMC11895683 DOI: 10.2147/ndt.s502312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/27/2025] [Indexed: 03/14/2025] Open
Abstract
Currently, the treatment of schizophrenia remains primarily pharmacological, with approximately 30% of patients diagnosed with treatment-resistant schizophrenia (TRS). Clozapine continues to be the first choice treatment for this subgroup of patients. As the preferred treatment, clozapine offers clear advantages in efficacy; however, its complex and troublesome adverse effects pose significant challenges for psychiatrists. Common side effects include granulocytopenia, intestinal obstruction, myocarditis, cardiomyopathy, constipation, and seizures. The first two complications are easier to manage due to the availability of laboratory monitoring, and improved management strategies are now in place in clinical practice. In recent years, clozapine-induced myocarditis (CIM) has gained considerable attention because of its potentially severe outcomes. However, the mechanism behind its lethality remains unclear, and there is no widely accepted consensus or treatment guideline, which complicates the implementation of targeted prevention in clinical practice. This review aims to summarize the clinical manifestations of CIM, explore the underlying mechanisms, and discuss recent advances in monitoring, diagnosis, and treatment, with the goal of offering constructive recommendations for future clinical management.
Collapse
Affiliation(s)
- Mier Li
- Department of Science and Education, The Affiliated Mental Health Center of Kunming Medical University, Yunnan Mental Hospital, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Yuncheng Bai
- Department of Orthopedic Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Yanjun Wang
- Department of Science and Education, The Affiliated Mental Health Center of Kunming Medical University, Yunnan Mental Hospital, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Haoran Xing
- Department of Science and Education, The Affiliated Mental Health Center of Kunming Medical University, Yunnan Mental Hospital, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Yingying Zhang
- Department of Science and Education, The Affiliated Mental Health Center of Kunming Medical University, Yunnan Mental Hospital, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Wenjing Ding
- Department of Science and Education, The Affiliated Mental Health Center of Kunming Medical University, Yunnan Mental Hospital, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Hong Ye
- Kunming Medical University, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Li Ma
- Department of Science and Education, The Affiliated Mental Health Center of Kunming Medical University, Yunnan Mental Hospital, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Weiwei Wang
- Department of Geriatrics, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, People’s Republic of China
| | - Tianhao Bao
- Department of Science and Education, The Affiliated Mental Health Center of Kunming Medical University, Yunnan Mental Hospital, Kunming, Yunnan Province, 650032, People’s Republic of China
- The Second People’s Hospital of Honghe Hani and Yi Autonomous Prefecture, Honghe Hani and Yi Autonomous Prefecture, Yunnan Province, 661400, People’s Republic of China
| |
Collapse
|
2
|
Ji M, Ran X, Zuo H, Zhang Q. Novel Insights into the Kallikrein-Kinin System in Fulminant Myocarditis: Physiological Basis and Potential Therapeutic Advances. J Inflamm Res 2024; 17:7347-7360. [PMID: 39429854 PMCID: PMC11490248 DOI: 10.2147/jir.s488237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Fulminant myocarditis (FM) is characterized by rapid cardiac deterioration often instigated by an inflammatory cytokine storm. The kallikrein-kinin system (KKS) is a metabolic cascade known for releasing vasoactive kinins, such as bradykinin-related peptides, possessing diverse pharmacological activities that include inflammation, regulation of vascular permeability, endothelial barrier dysfunction, and blood pressure modulation. The type 1 and type 2 bradykinin receptors (B1R and B2R), integral components of the KKS system, mediate the primary biological effects of kinin peptides. This review aims to offer a comprehensive overview of the primary mechanisms of the KKS in FM, including an examination of the structural components, regulatory activation, and downstream signaling pathways of the KKS. Furthermore, it explores the involvement of the tissue kallikrein/B1R/inducible nitric oxide synthase (TK/B1R/iNOS) pathway in myocyte dysfunction, modulation of the immune response, and preservation of endothelial barrier integrity. The potential therapeutic advances targeting the inhibition of the KKS in managing FM will be discussed, providing valuable insights for the development of clinical treatment strategies.
Collapse
Affiliation(s)
- Mengmeng Ji
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xiao Ran
- Department of Critical-Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Houjuan Zuo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
3
|
Ząbczyk M, Natorska J, Matusik PT, Mołek P, Wojciechowska W, Rajzer M, Rajtar-Salwa R, Tokarek T, Lenart-Migdalska A, Olszowska M, Undas A. Neutrophil-activating Peptide 2 as a Novel Modulator of Fibrin Clot Properties in Patients with Atrial Fibrillation. Transl Stroke Res 2024; 15:773-783. [PMID: 37294500 PMCID: PMC10250863 DOI: 10.1007/s12975-023-01165-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023]
Abstract
Neutrophil-activating peptide 2 (NAP-2, CXCL7), a platelet-derived neutrophil chemoattractant, is involved in inflammation. We investigated associations between NAP-2 levels, neutrophil extracellular traps (NETs) formation, and fibrin clot properties in atrial fibrillation (AF). We recruited 237 consecutive patients with AF (mean age, 68 ± 11 years; median CHA2DS2VASc score of 3 [2-4]) and 30 apparently healthy controls. Plasma NAP-2 concentrations were measured, along with plasma fibrin clot permeability (Ks) and clot lysis time (CLT), thrombin generation, citrullinated histone H3 (citH3), as a marker of NETs formation, and 3-nitrotyrosine reflecting oxidative stress. NAP-2 levels were 89% higher in AF patients than in controls (626 [448-796] vs. 331 [226-430] ng/ml; p < 0.0001). NAP-2 levels were not associated with demographics, CHA2DS2-VASc score, or the AF manifestation. Patients with NAP-2 in the top quartile (> 796 ng/ml) were characterized by higher neutrophil count (+ 31.7%), fibrinogen (+ 20.8%), citH3 (+ 86%), and 3-nitrotyrosine (+ 111%) levels, along with 20.2% reduced Ks and 8.4% prolonged CLT as compared to the remaining subjects (all p < 0.05). NAP-2 levels were positively associated with fibrinogen in AF patients (r = 0.41, p = 0.0006) and controls (r = 0.65, p < 0.01), along with citH3 (r = 0.36, p < 0.0001) and 3-nitrotyrosine (r = 0.51, p < 0.0001) in the former group. After adjustment for fibrinogen, higher citH3 (per 1 ng/ml β = -0.046, 95% CI -0.029; -0.064) and NAP-2 (per 100 ng/ml β = -0.21, 95% CI -0.14; -0.28) levels were independently associated with reduced Ks. Elevated NAP-2, associated with increased oxidative stress, has been identified as a novel modulator of prothrombotic plasma fibrin clot properties in patients with AF.
Collapse
Affiliation(s)
- Michał Ząbczyk
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Pradnicka 80, 31-202, Krakow, Poland
- Krakow Centre for Medical Research and Technologies, the John Paul II Hospital, Pradnicka 80, Krakow, Poland
| | - Joanna Natorska
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Pradnicka 80, 31-202, Krakow, Poland
- Krakow Centre for Medical Research and Technologies, the John Paul II Hospital, Pradnicka 80, Krakow, Poland
| | - Paweł T Matusik
- Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, Pradnicka 80, Kraków, Poland
- Department of Electrocardiology, the John Paul II Hospital, Pradnicka 80, Kraków, Poland
| | - Patrycja Mołek
- Krakow Centre for Medical Research and Technologies, the John Paul II Hospital, Pradnicka 80, Krakow, Poland
| | - Wiktoria Wojciechowska
- 1st Department of Cardiology, Interventional Electrocardiology and Arterial Hypertension, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, Poland
| | - Marek Rajzer
- 1st Department of Cardiology, Interventional Electrocardiology and Arterial Hypertension, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, Poland
| | - Renata Rajtar-Salwa
- Department of Cardiology and Cardiovascular Interventions, University Hospital, Jakubowskiego 2, Krakow, Poland
| | - Tomasz Tokarek
- Center for Invasive Cardiology, Electrotherapy and Angiology, Kilinskiego 68, Nowy Sacz, Poland
- Center for Innovative Medical Education, Jagiellonian University Medical College, Medyczna 9, Krakow, Poland
| | - Aleksandra Lenart-Migdalska
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Institute of Cardiology, Jagiellonian University Medical College, John Paul II Hospital, Pradnicka 80, Kraków, Poland
| | - Maria Olszowska
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Institute of Cardiology, Jagiellonian University Medical College, John Paul II Hospital, Pradnicka 80, Kraków, Poland
| | - Anetta Undas
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Pradnicka 80, 31-202, Krakow, Poland.
- Krakow Centre for Medical Research and Technologies, the John Paul II Hospital, Pradnicka 80, Krakow, Poland.
| |
Collapse
|
4
|
Toita R, Kawano T, Murata M, Kang JH. Bioinspired macrophage-targeted anti-inflammatory nanomedicine: A therapeutic option for the treatment of myocarditis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112492. [PMID: 34857278 DOI: 10.1016/j.msec.2021.112492] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/29/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Myocarditis is a disease characterized by inflammation of the heart muscle, which increases the risk of dilated cardiomyopathy and heart failure. Macrophage migration is a major histopathological hallmark of myocarditis, making macrophages a potential therapeutic target for the management of this disease. In the present study, we synthesized a bioinspired anti-inflammatory nanomedicine conjugated with protein G (PSL-G) that could target macrophages and induce macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Notably, PSL-G exhibited a higher affinity for macrophages than non-macrophage cells. The addition of PSL-G decreased the levels of pro-inflammatory cytokines (e.g., IL-1α, IL-6, and TNF-α), but increased the level of the anti-inflammatory cytokine IL-10 in macrophages treated with lipopolysaccharide and/or interferon-γ. Furthermore, the lifetime of PSL-G in murine blood circulation was found to be significantly higher than that of PSL. Systemic injection of PSL-G into a mouse model of experimental autoimmune myocarditis remarkably reduced macrophage migration in the myocardium (16-fold compared with the positive control group) and myocardial fibrosis (8-fold). Based on these results and the fact that macrophages play a critical role in the pathogenesis of various diseases, we believe that bioinspired macrophage-targeted anti-inflammatory nanomedicines may be effective therapeutic options for the treatment of autoimmune and autoinflammatory diseases, especially myocarditis.
Collapse
Affiliation(s)
- Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jeong-Hun Kang
- Division of Biopharmaceutics and Pharmacokinetics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
5
|
D’Errico S, Russa RL, Maiese A, Santurro A, Scopetti M, Romano S, Zanon M, Frati P, Fineschi V. Atypical antipsychotics and oxidative cardiotoxicity: review of literature and future perspectives to prevent sudden cardiac death. J Geriatr Cardiol 2021; 18:663-685. [PMID: 34527032 PMCID: PMC8390928 DOI: 10.11909/j.issn.1671-5411.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidative stress is considered the principal mediator of myocardial injury under pathological conditions. It is well known that reactive oxygen (ROS) or nitrogen species (RNS) are involved in myocardial injury and repair at the same time and that cellular damage is generally due to an unbalance between generation and elimination of the free radicals due to an inadequate mechanism of antioxidant defense or to an increase in ROS and RNS. Major adverse cardiovascular events are often associated with drugs with associated findings such as fibrosis or inflammation of the myocardium. Despite efforts in the preclinical phase of the development of drugs, cardiotoxicity still remains a great concern. Cardiac toxicity due to second-generation antipsychotics (clozapine, olanzapine, quetiapine) has been observed in preclinical studies and described in patients affected with mental disorders. A role of oxidative stress has been hypothesized but more evidence is needed to confirm a causal relationship. A better knowledge of cardiotoxicity mechanisms should address in the future to establish the right dose and length of treatment without impacting the physical health of the patients.
Collapse
Affiliation(s)
- Stefano D’Errico
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Raffaele La Russa
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
| | - Aniello Maiese
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Surgical Pathology, Medical, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - Alessandro Santurro
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Matteo Scopetti
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Romano
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Martina Zanon
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Paola Frati
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Vittorio Fineschi
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Zhong C, Wu Y, Chang H, Liu C, Zhou L, Zou J, Qi Z. Effect of PKC inhibitor on experimental autoimmune myocarditis in Lewis rats. Oncotarget 2017; 8:54187-54198. [PMID: 28903333 PMCID: PMC5589572 DOI: 10.18632/oncotarget.17018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022] Open
Abstract
Myocarditis is a major cause of sudden, unexpected death in young people. However, it is still one of the most challenging diseases to treat in cardiology. In the present study, we showed that both expression level and activity of PKC-α were up-regulated in the rat heart of experimental autoimmune myocarditis (EAM). Intraperitoneal administration of PKC inhibitor (Ro-32-0432) at the end of the most severe inflammation period of EAM still significantly reduced the EAM induced expression of failure biomarkers. Furthermore, Ro-32-0432 reduced the ratio of Bax/Bcl-2 and suppressed the expression of cleaved caspase-3, both of which were increased in the heart of the EAM rats, suggesting an anti-apoptotic role of Ro-32-0432. Besides, Ro-32-0432 suppressed EAM-induced cardiac fibrosis and release of pro-inflammatory cytokines IL-1β and IL-17. These results suggest that inhibition of PKC may serve as a potential therapeutic strategy for the treatment of myocarditis.
Collapse
Affiliation(s)
- Chunlian Zhong
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiang'an Nan Lu, Xiamen, China
| | - Yang Wu
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiang'an Nan Lu, Xiamen, China.,Xiamen Cardiovascular Hospital, Medical College of Xiamen University, Xiamen, China
| | - He Chang
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiang'an Nan Lu, Xiamen, China.,Xiamen Cardiovascular Hospital, Medical College of Xiamen University, Xiamen, China
| | - Chunxiao Liu
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiang'an Nan Lu, Xiamen, China.,Xiamen Cardiovascular Hospital, Medical College of Xiamen University, Xiamen, China
| | - Li Zhou
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiang'an Nan Lu, Xiamen, China
| | - Jun Zou
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiang'an Nan Lu, Xiamen, China
| | - Zhi Qi
- Department of Basic Medical Sciences, Medical College of Xiamen University, Xiang'an Nan Lu, Xiamen, China
| |
Collapse
|
7
|
Zhou MS, Adam A, Raij L. Review: Interaction among angiotensin II, nitric oxide and oxidative stress. J Renin Angiotensin Aldosterone Syst 2016; 2:S59-S63. [DOI: 10.1177/14703203010020011001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Ming-Sheng Zhou
- Nephrology and Hypertension Section, Department of Veterans
Affairs Medical Center, and University of Minnesota, Minneapolis, Minnesota,
USA
| | - Ahmed Adam
- Nephrology and Hypertension Section, Department of Veterans
Affairs Medical Center, and University of Minnesota, Minneapolis, Minnesota,
USA
| | - Leopoldo Raij
- Nephrology and Hypertension Section, Department of Veterans
Affairs Medical Center, and University of Minnesota, Minneapolis, Minnesota,
USA,
| |
Collapse
|
8
|
Arif Z, Arfat MY, Neelofar K, Ahmad S, Badar A, Khan MA, Zaman A, Ahmad J. Effect of peroxynitrite on human serum albumin: a multi technique approach. J Biomol Struct Dyn 2016; 35:2066-2076. [PMID: 27346535 DOI: 10.1080/07391102.2016.1206489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In this study, human serum albumin (HSA), the most abundant protein of blood plasma, was modified with varying concentrations of peroxynitrite. The peroxynitrite-induced changes in HSA was monitored by spectroscopy, SDS-PAGE, 1-anilinonaphthalene-8-sulfonic acid (ANS), thermal denaturation studies, and matrix-assisted laser desorption/inonization-time of flight mass spectrometry (MALDI-TOF MS). Aggregate formation was studied by thioflavin T binding and scanning electron microscopy (SEM). The results indicated formation of 3-nitrotyrosine, 6-nitrotryptophan, dityrosine, and carbonyls in modified samples and showed retarded mobility in SDS-polyacrylamide gel. Reduction in α-helicity and surface protein hydrophobicity confirmed the secondary and tertiary structure alterations in peroxynitrite-modified-HSA. Also, attachment of nitro group and increase in melting temperature was observed in modified sample. Furthermore, significant enhancement in the fluorescence intensity of ThT upon binding with peroxynitrite-modified-HSA and images under scanning electron microscope are suggestive of protein aggregation. It is, therefore, speculated that HSA modified by endogenously formed peroxynitrite might act as a trigger for nitration/aggregation and suggested the role of peroxynitrite-modified-HSA in SLE.
Collapse
Affiliation(s)
- Zarina Arif
- a Facuty of Medicine, Department of Biochemistry , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| | - Mir Yasir Arfat
- a Facuty of Medicine, Department of Biochemistry , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| | - Km Neelofar
- b Faculty of Medicine, Rajiv Gandhi Centre for Diabetes and Endocrinology , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| | - Shafeeque Ahmad
- a Facuty of Medicine, Department of Biochemistry , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| | - Asim Badar
- a Facuty of Medicine, Department of Biochemistry , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| | - Md Adnan Khan
- a Facuty of Medicine, Department of Biochemistry , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| | - Asif Zaman
- a Facuty of Medicine, Department of Biochemistry , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| | - Jamal Ahmad
- b Faculty of Medicine, Rajiv Gandhi Centre for Diabetes and Endocrinology , J.N. Medical College, Aligarh Muslim University , Aligarh 202002 , UP , India
| |
Collapse
|
9
|
Protective Effects of Carvedilol and Vitamin C against Azithromycin-Induced Cardiotoxicity in Rats via Decreasing ROS, IL1-β, and TNF-α Production and Inhibiting NF-κB and Caspase-3 Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1874762. [PMID: 27274777 PMCID: PMC4871977 DOI: 10.1155/2016/1874762] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 03/27/2016] [Accepted: 04/12/2016] [Indexed: 01/24/2023]
Abstract
The Food and Drug Administration recently warned of the fatal cardiovascular risks of azithromycin in humans. In addition, a recently published study documented azithromycin-induced cardiotoxicity in rats. This study aimed to justify the exact cardiovascular events accompanying azithromycin administration in rats, focusing on electrocardiographic, biochemical, and histopathological changes. In addition, the underlying mechanisms were studied regarding reactive oxygen species production, cytokine release, and apoptotic cell-death. Finally, the supposed protective effects of both carvedilol and vitamin C were assessed. Four groups of rats were used: (1) control, (2) azithromycin, (3) azithromycin + carvedilol, and (4) azithromycin + vitamin C. Azithromycin resulted in marked atrophy of cardiac muscle fibers and electrocardiographic segment alteration. It increased the heart rate, lactate dehydrogenase, creatine phosphokinase, malondialdehyde, nitric oxide, interleukin-1 beta (IL1-β), tumor necrosis factor alpha (TNF-α), nuclear factor kappa beta (NF-κB), and caspase-3. It decreased reduced glutathione, glutathione peroxidase, and superoxide dismutase. Carvedilol and vitamin C prevented most of the azithromycin-induced electrocardiographic and histopathological changes. Carvedilol and vitamin C decreased lactate dehydrogenase, malondialdehyde, IL1-β, TNF-α, NF-κB, and caspase-3. Both agents increased glutathione peroxidase. This study shows that both carvedilol and vitamin C protect against azithromycin-induced cardiotoxicity through antioxidant, immunomodulatory, and antiapoptotic mechanisms.
Collapse
|
10
|
Yang S, Xu W, Dong Z, Zhou M, Lin C, Jin H, Su Y, Li Q, Wang X, Chang H, Han W. TPEN prevents rapid pacing-induced calcium overload and nitration stress in HL-1 myocytes. Cardiovasc Ther 2016; 33:200-8. [PMID: 25973665 DOI: 10.1111/1755-5922.12134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Atrial fibrillation (AF) is the most common cardiac arrhythmia. However, the current drug interference of antiarrhythmia has limited efficacy and off-target effects. Accumulating evidence has implicated a potential role of nitration stress in the pathogenesis of AF. The aim of the study was to determine whether TPEN provided antinitration effects on atrial myocytes during AF, especially under circumstances of nitration stress. METHODS We utilized a rapid paced HL-1 cells model for AF. The changes of electrophysiological characteristics and structure of paced HL-1 cells were determined by a patch clamp and a TEM method. The effects of TPEN on pacing and ONOO(-) pretreated HL-1 cells were examined using MTT assay, TUNEL technique, confocal microscope experiment, and Western blot analysis. RESULTS The results revealed that ONOO(-) reduced the viability of HL-1 cells in a dose-dependent manner, and 1 μmol/L TPEN significantly ameliorated the damage caused by 50 μmol/L ONOO(-) (P < 0.05). Pacing and/or ONOO(-) -induced marked shortening of APD, myolysis, and nuclear condensation. TPEN inhibited the Ca(2+) overload induced by rapid pacing (P < 0.05) and ONOO(-) stimulation (P < 0.05). The application of TPEN significantly prevented the protein nitration caused by pacing or pacing plus ONOO(-) (P < 0.05). Additionally, pacing in combination with ONOO(-) treatment led to increase in apoptosis in HL-1 cells (P < 0.01), which could be reduced by pretreatment with TPEN (P < 0.05). CONCLUSIONS TPEN prevents Ca(2+) overload and nitration stress in HL-1 atrial myocytes during rapid pacing and circumstances of nitration stress.
Collapse
Affiliation(s)
- Shusen Yang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjing Xu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zengxiang Dong
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mo Zhou
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaolan Lin
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongbo Jin
- Laboratory of Physiology, Harbin Medical University, Harbin, China
| | - Yafen Su
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingyu Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xu Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huiying Chang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Han
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Kertai MD, Li YJ, Ji Y, Qi W, Lombard FW, Shah SH, Kraus WE, Stafford-Smith M, Newman MF, Milano CA, Waldron N, Podgoreanu MV, Mathew JP. Genome-wide association study of new-onset atrial fibrillation after coronary artery bypass grafting surgery. Am Heart J 2015; 170:580-90.e28. [PMID: 26385043 DOI: 10.1016/j.ahj.2015.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/15/2015] [Indexed: 01/04/2023]
Abstract
BACKGROUND Postoperative atrial fibrillation (AF) is a potentially life-threatening complication after coronary artery bypass graft (CABG) surgery. Genetic predisposition may predict risk for developing postoperative AF. METHODS Study subjects underwent CABG surgery with cardiopulmonary bypass at Duke University Medical Center. In a discovery cohort of 877 individuals from the Perioperative Genetics and Safety Outcomes Study, we performed a genome-wide association study using a logistic regression model with a covariate adjustment for AF risk index. Single-nucleotide polymorphisms (SNPs) that met a P < 5 × 10(-5) were further tested using a replication dataset of 304 individuals from the CATHeterization GENetics biorepository, followed by meta-analysis. Potential pathways related to postoperative AF were identified through gene enrichment analysis using the top genome-wide association study SNPs (P < 10(-4)). RESULTS Nineteen SNPs met the a priori defined discovery threshold for replication, but only 3 met nominal significance (P < .05) in the CATHeterization GENetics group, with only one-rs10504554, in the intronic region in lymphocyte antigen 96 (LY96)-showing the same direction of the effect for postoperative AF (odds ratio [OR] 0.48, 95% CI 0.34-0.68, P = 2.9 × 10(-5) vs OR 0.55, 95% CI 0.31-0.99, P = .046) and strong overall association by meta-analysis (meta-P = 4.0 × 10(-6)). Gene enrichment analysis highlighted the role of LY96 in pathways of biologic relevance to activation and modulation of innate immune responses. Our analysis also showed potential association between LY96 and nuclear factor κ-B interaction and postoperative AF through their relevance to inflammatory signaling pathways. CONCLUSIONS In patients undergoing CABG surgery, we found genetic polymorphisms in LY96 associated with decreased risk of postoperative AF.
Collapse
|
12
|
Ma X, Yu Q, Guo X, Zeng K, Zhao M, Tu P, Jiang Y. Nitric oxide inhibitory flavonoids from traditional Chinese medicine formula Baoyuan Decoction. Fitoterapia 2015; 103:252-9. [PMID: 25889071 DOI: 10.1016/j.fitote.2015.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/09/2015] [Accepted: 04/12/2015] [Indexed: 10/23/2022]
Abstract
Three new flavonoid glycosides, (3R)-(+)-isomucronulatol-2'-O-β-D-glucopyranoside (1), (3R)-(-)-isomucronulatol-7-O-β-D-apiofuranosyl(1→2)-β-D-glucopyranoside (2), and (2S)-(-)-7,8-dihydroxylflavanone-4'-O-β-D-apiofuranosyl(1→2)-β-D-glucopyranoside (3), along with eight flavanones (4, 8, 10, 12, 15, 16, 21, and 24), four isoflavones (5, 11, 13, and 23), four chalcones (6, 14, 17, and 18), two isoflavans (19-20), one flavone (7), one flavonol (9), and one dihydrochalcone (22) were isolated from Baoyuan Decoction (BYD), a traditional Chinese medicine formula. The structures of the new compounds were established by detailed analysis of NMR and HRESIMS spectroscopic data, and their absolute configurations were determined by electronic circular dichroism (ECD) data. The inhibitory effects of the isolates were evaluated on nitric oxide production in lipopolysaccharide activated RAW 264.7 macrophage cells. Compounds 6, 9, and 10 showed the significant inhibitory activities, with IC50 values of 1.4, 13.8, and 9.3 μM, respectively, comparable to or even better than the positive control, quercetin (IC50, 16.5 μM). The assignment of these isolated flavonoids was achieved using UPLC-Q-trap-MS, and the results suggested that they were originated from Astragalus membranaceus and Glycyrrhiza uralensis.
Collapse
Affiliation(s)
- Xiaoli Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Qian Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Xiaoyu Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Mingbo Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China.
| |
Collapse
|
13
|
Clozapine-Induced Cardiotoxicity: Role of Oxidative Stress, Tumour Necrosis Factor Alpha and NF-κβ. Cardiovasc Toxicol 2014; 15:355-65. [DOI: 10.1007/s12012-014-9304-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
14
|
Abdel-Wahab BA, Metwally ME. Clozapine-induced cardiotoxicity in rats: Involvement of tumour necrosis factor alpha, NF-κβ and caspase-3. Toxicol Rep 2014; 1:1213-1223. [PMID: 28962331 PMCID: PMC5598316 DOI: 10.1016/j.toxrep.2014.11.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/17/2014] [Accepted: 11/12/2014] [Indexed: 12/18/2022] Open
Abstract
Clozapine, an ideal antipsychotic drug for the treatment of resistant schizophrenia, is considered the most underutilised treatment for schizophrenia. However, safety concerns have been raised about clozapine-induced cardiotoxicity, which may lead to sudden death, particularly in young patients. The exact mechanism of clozapine cardiotoxicity has not yet been thoroughly studied. This study aimed to investigate the possible mechanisms of clozapine-induced cardiotoxicity in a rat model. Young male Wistar rats were treated with clozapine (10, 15 and 25 mg/kg/day, i.p.) for 21 days. Haemodynamic and echocardiographic studies were performed for assessment of cardiac functions. Heart sections were studied histopathologically and immunohistochemically. Serum and cardiac markers of cardiotoxicity, oxidative stress, inflammation and apoptosis were evaluated. Heart sections of CLZ-treated animals showed increased cardiac inflammation that correlated with the clozapine dose. Serum levels of CK-MB and LDH levels increased, as did cardiac levels of TNF-α, MDA, NO, myeloperoxidase (MPO), 8-OHdG, caspase-3 and NF-κB p65. In contrast, GSH levels and GSH-Px activity decreased. Furthermore, immunohistochemical examination of the heart sections showed positive immunostaining for both 3-nitrotyrosine and caspase-3 in all clozapine-treated groups. Clozapine, particularly in relatively high doses, has a clear cardiotoxic effect. This cardiotoxicity is accompanied by increased myocardial oxidative stress, inflammatory cytokines, DNA damage and apoptosis with attenuation in antioxidant defences, thus explaining the previously reported myocarditis and pericarditis during clozapine therapy in clinical studies.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Pharmacology, College of Medicine, Assiut University, Assiut, Egypt
| | - Metwally E Metwally
- Department of Forensic Medicine and Toxicology, College of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
15
|
Oliveira LGR, Kuehn CC, dos Santos CD, Miranda MA, da Costa CMB, Mendonça VJ, do Prado JC. Protective actions of melatonin against heart damage during chronic Chagas disease. Acta Trop 2013; 128:652-8. [PMID: 24055715 DOI: 10.1016/j.actatropica.2013.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 09/06/2013] [Accepted: 09/10/2013] [Indexed: 01/05/2023]
Abstract
Chronic cardiomyopathy is the most important clinical form of Chagas disease, and it is characterised by myocarditis that is associated with fibrosis and organ dysfunction. Alternative treatment options are important tools to modulate host immune responses. The main goal of this work was to evaluate the anti-inflammatory actions of melatonin during the chronic phase of Chagas disease. TNF-α, IL-10 and nitrite concentrations were evaluated as predictive factors of immune modulation. Creatine phosphokinase-MB (CK-MB), cardiac inflammatory foci and heart weight were assessed to evaluate the efficacy of the melatonin treatment. Male Wistar rats were infected with 1×10(5) blood trypomastigotes of the Y strain of Trypanosoma cruzi and kept untreated for 60 days to mimic chronic infection. After this period, the rats were orally treated with melatonin 50mg/kg/day, and the experiments were performed 90, 120, and 180 days post-infection. Melatonin treatment significantly increased the concentration of IL-10 and reduced the concentrations of NO and TNF-α produced by cardiomyocytes. Furthermore, it led to decreased heart weight, serum CK-MB levels and inflammatory foci when compared to the untreated and infected control groups. We conclude that melatonin therapy is effective at protecting animals against the harmful cardiac inflammatory response that is characteristic of chronic T. cruzi infection.
Collapse
|
16
|
Barin JG, Čiháková D. Control of inflammatory heart disease by CD4+ T cells. Ann N Y Acad Sci 2013; 1285:80-96. [PMID: 23692566 DOI: 10.1111/nyas.12134] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review focuses on autoimmune myocarditis and its sequela, inflammatory dilated cardiomyopathy (DCMI), and the inflammatory and immune mechanisms underlying the pathogenesis of these diseases. Several mouse models of myocarditis and DCMI have improved our knowledge of the pathogenesis of these diseases, informing more general problems of cardiac remodeling and heart failure. CD4(+) T cells are critical in driving the pathogenesis of myocarditis. We discuss in detail the role of T helper cell subtypes in the pathogenesis of myocarditis, the biology of T cell-derived effector cytokines, and the participation of other leukocytic effectors in mediating disease pathophysiology. We discuss interactions between these subsets in both suppressive and collaborative fashions. These findings indicate that cardiac inflammatory disease, and autoimmunity in general, may be more diverse in divergent effector mechanisms than has previously been appreciated.
Collapse
Affiliation(s)
- Jobert G Barin
- Department of Pathology, Division of Immunology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
17
|
Ivabradine inhibits the production of proinflammatory cytokines and inducible nitric oxide synthase in acute coxsackievirus B3-induced myocarditis. Biochem Biophys Res Commun 2013; 431:450-5. [DOI: 10.1016/j.bbrc.2012.12.147] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 12/16/2012] [Indexed: 02/02/2023]
|
18
|
Arumugam S, Thandavarayan RA, Veeraveedu PT, Nakamura T, Arozal W, Sari FR, Giridharan VV, Soetikno V, Palaniyandi SS, Harima M, Suzuki K, Nagata M, Kodama M, Watanabe K. Beneficial effects of edaravone, a novel antioxidant, in rats with dilated cardiomyopathy. J Cell Mol Med 2013; 16:2176-85. [PMID: 22268705 PMCID: PMC3822987 DOI: 10.1111/j.1582-4934.2012.01526.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Edaravone, a novel antioxidant, acts by trapping hydroxyl radicals, quenching active oxygen and so on. Its cardioprotective activity against experimental autoimmune myocarditis (EAM) was reported. Nevertheless, it remains to be determined whether edaravone protects against cardiac remodelling in dilated cardiomyopathy (DCM). The present study was undertaken to assess whether edaravone attenuates myocardial fibrosis, and examine the effect of edaravone on cardiac function in rats with DCM after EAM. Rat model of EAM was prepared by injection with porcine cardiac myosin 28 days after immunization, we administered edaravone intraperitoneally at 3 and 10 mg/kg/day to rats for 28 days. The results were compared with vehicle-treated rats with DCM. Cardiac function, by haemodynamic and echocardiographic study and histopathology were performed. Left ventricular (LV) expression of NADPH oxidase subunits (p47phox, p67phox, gp91phox and Nox4), fibrosis markers (TGF-β1 and OPN), endoplasmic reticulum (ER) stress markers (GRP78 and GADD 153) and apoptosis markers (cytochrome C and caspase-3) were measured by Western blotting. Edaravone-treated DCM rats showed better cardiac function compared with those of the vehicle-treated rats. In addition, LV expressions of NADPH oxidase subunits levels were significantly down-regulated in edaravone-treated rats. Furthermore, the number of collagen-III positive cells in the myocardium of edaravone-treated rats was lower compared with those of the vehicle-treated rats. Our results suggest that edaravone ameliorated the progression of DCM by modulating oxidative and ER stress-mediated myocardial apoptosis and fibrosis.
Collapse
Affiliation(s)
- Somasundaram Arumugam
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata City, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Carvalho CME, Silverio JC, da Silva AA, Pereira IR, Coelho JMC, Britto CC, Moreira OC, Marchevsky RS, Xavier SS, Gazzinelli RT, da Glória Bonecini-Almeida M, Lannes-Vieira J. Inducible nitric oxide synthase in heart tissue and nitric oxide in serum of Trypanosoma cruzi-infected rhesus monkeys: association with heart injury. PLoS Negl Trop Dis 2012; 6:e1644. [PMID: 22590660 PMCID: PMC3348164 DOI: 10.1371/journal.pntd.0001644] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 03/30/2012] [Indexed: 12/19/2022] Open
Abstract
Background The factors contributing to chronic Chagas' heart disease remain unknown. High nitric oxide (NO) levels have been shown to be associated with cardiomyopathy severity in patients. Further, NO produced via inducible nitric oxide synthase (iNOS/NOS2) is proposed to play a role in Trypanosoma cruzi control. However, the participation of iNOS/NOS2 and NO in T. cruzi control and heart injury has been questioned. Here, using chronically infected rhesus monkeys and iNOS/NOS2-deficient (Nos2−/−) mice we explored the participation of iNOS/NOS2-derived NO in heart injury in T. cruzi infection. Methodology Rhesus monkeys and C57BL/6 and Nos2−/− mice were infected with the Colombian T. cruzi strain. Parasite DNA was detected by polymerase chain reaction, T. cruzi antigens and iNOS/NOS2+ cells were immunohistochemically detected in heart sections and NO levels in serum were determined by Griess reagent. Heart injury was assessed by electrocardiogram (ECG), echocardiogram (ECHO), creatine kinase heart isoenzyme (CK-MB) activity levels in serum and connexin 43 (Cx43) expression in the cardiac tissue. Results Chronically infected monkeys presented conduction abnormalities, cardiac inflammation and fibrosis, which resembled the spectrum of human chronic chagasic cardiomyopathy (CCC). Importantly, chronic myocarditis was associated with parasite persistence. Moreover, Cx43 loss and increased CK-MB activity levels were primarily correlated with iNOS/NOS2+ cells infiltrating the cardiac tissue and NO levels in serum. Studies in Nos2−/− mice reinforced that the iNOS/NOS2-NO pathway plays a pivotal role in T. cruzi-elicited cardiomyocyte injury and in conduction abnormalities that were associated with Cx43 loss in the cardiac tissue. Conclusion T. cruzi-infected rhesus monkeys reproduce features of CCC. Moreover, our data support that in T. cruzi infection persistent parasite-triggered iNOS/NOS2 in the cardiac tissue and NO overproduction might contribute to CCC severity, mainly disturbing of the molecular pathway involved in electrical synchrony. These findings open a new avenue for therapeutic tools in Chagas' heart disease. Chagas disease, a neglected tropical disease caused by the protozoan Trypanosoma cruzi, afflicts from 8 to 15 million people in the Latin America. Chronic chagasic cardiomyopathy (CCC) is the most frequent manifestation of Chagas disease. Currently, patient management only mitigates CCC symptoms. The pathogenic factors leading to CCC remain unknown; therefore their comprehension may contribute to develop more efficient therapies. In patients, high nitric oxide (NO) levels have been associated with CCC severity. In T. cruzi-infected mice, NO, mainly produced via inducible nitric oxide synthase (iNOS/NOS2), is proposed to work in parasite control. However, the participation of iNOS/NOS2 and NO in T. cruzi control and heart injury has been questioned. Here, infected rhesus monkeys and iNOS/NOS2-deficient mice were used to explore the participation of iNOS/NOS2-derived NO in heart injury in T. cruzi infection. Chronically infected monkeys presented electrical abnormalities, myocarditis and fibrosis, resembling the spectrum of human CCC. Moreover, cardiomyocyte lesion correlated with iNOS/NOS2+ cells infiltrating the cardiac tissue. Our findings support that parasite-driven iNOS/NOS2+ cells accumulation in the cardiac tissue and NO overproduction contribute to cardiomyopathy severity, mainly disturbing the pathway involved in electrical synchrony in T. cruzi infection.
Collapse
Affiliation(s)
- Cristiano Marcelo Espinola Carvalho
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz (IOC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
- Serviço de Imunologia, Instituto de Pesquisa Clínica Evandro Chagas (IPEC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jaline Coutinho Silverio
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz (IOC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea Alice da Silva
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz (IOC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Patologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Isabela Resende Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz (IOC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Constança Carvalho Britto
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Otacílio Cruz Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Sergio Salles Xavier
- Serviço de Imunologia, Instituto de Pesquisa Clínica Evandro Chagas (IPEC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Tostes Gazzinelli
- Laboratório de Imunoparasitologia, Instituto Rene Rachou/Fiocruz, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Imunologia e Bioquímica, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz (IOC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
20
|
Arumugam S, Thandavarayan RA, Arozal W, Sari FR, Giridharan VV, Soetikno V, Palaniyandi SS, Harima M, Suzuki K, Nagata M, Tagaki R, Kodama M, Watanabe K. Quercetin offers cardioprotection against progression of experimental autoimmune myocarditis by suppression of oxidative and endoplasmic reticulum stress via endothelin-1/MAPK signalling. Free Radic Res 2012; 46:154-63. [PMID: 22145946 DOI: 10.3109/10715762.2011.647010] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In order to test the hypothesis that treatment with quercetin at a dose of 10 mg/kg protects from the progression of experimental autoimmune myocarditis (EAM) to dilated cardiomyopathy (DCM), we have used the rat model of EAM induced by porcine cardiac myosin. Our results identified that the post-myocarditis rats suffered from elevated endoplasmic reticulum (ER) stress and adverse cardiac remodelling in the form of myocardial fibrosis, whereas the rats treated with quercetin have been protected from these changes as evidenced by the decreased myocardial levels of ER stress and fibrosis markers when compared with the vehicle-treated DCM rats. In addition, the myocardial dimensions and cardiac function were preserved significantly in the quercetin-treated rats in comparison with the DCM rats treated with vehicle alone. Interestingly, the rats treated with quercetin showed significant suppression of the myocardial endothelin-1 and also the mitogen activated protein kinases (MAPK) suggesting that the protection offered by quercetin treatment against progression of EAM involves the modulation of MAPK signalling cascade. Collectively, the present study provides data to support the role of quercetin in protecting the hearts of the rats with post myocarditis DCM.
Collapse
Affiliation(s)
- Somasundaram Arumugam
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata City, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Suzuki JI, Ogawa M, Watanabe R, Takayama K, Hirata Y, Nagai R, Isobe M. Roles of prostaglandin E2 in cardiovascular diseases. Int Heart J 2011; 52:266-9. [PMID: 22008433 DOI: 10.1536/ihj.52.266] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Prostaglandin E2 (PGE(2)) is produced in inflammatory responses and regulates a variety of immunological reactions through 4 different receptor subtypes; EP1, 2, 3 and 4. However, the precise role of each receptor in cardiovascular disease has not yet been elucidated. Enhanced expression of some EPs has been observed in clinical and experimental cardiovascular diseases. EP agonists have been developed to clarify the role of each receptor. Recently, we developed a novel selective agonist to examine the effects of EP4 on cardiac transplantation, myocardial ischemia, and myocarditis. Of note, a selective EP4 agonist attenuated inflammatory cytokines and chemokines via attenuation of macrophage activation in inflammatory heart diseases. In this review article, we discuss the effects of PGE(2) receptor agonists on the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Jun-ichi Suzuki
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
The anti-inflammatory mechanism of prostaglandin e2 receptor 4 activation in rat experimental autoimmune myocarditis. J Cardiovasc Pharmacol 2011; 57:365-72. [PMID: 21383594 DOI: 10.1097/fjc.0b013e31820b7be1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Prostaglandins (PG) and their specific receptors for E type PG (EP) play an important role in inflammatory diseases. Although myocarditis results in inflammation of the heart, roles of PG and EP in its pathophysiology is still controversial. To clarify the role of PG and EP on the progression of myocarditis, we used an experimental autoimmune myocarditis model. A selective EP4 (EP4RAG) agonist was administered into both early (Day 0 to 21) and late (Day 14 to 21) -treated groups and the animals were killed on Day 21. We found that improved cardiac function was detected in the EP4RAG-treated groups in comparison to the untreated group. The infiltration area ratio in the early-treated (16.6% ± 4.6%) group was lower than those in the untreated group (32.1% ± 3.5%) (P < 0.05). The fibrosis area ratios in the early-treated (19.2% ± 6.3%) and the late-treated groups (24.4% ± 5.1%) were lower than those in the untreated group (37.4% ± 2.6%), respectively (P < 0.05). Moreover, we found that EP4RAG decreased T-cell proliferation and monocyte chemoattractant protein-1 production in vitro. We concluded that a selective EP4 agonist inactivates T-cells, which turns out to moderate the progression of experimental autoimmune myocarditis. Therefore, EP4 can be an effective target for myocarditis treatment.
Collapse
|
23
|
Mito S, Thandavarayan RA, Ma M, Lakshmanan A, Suzuki K, Kodama M, Watanabe K. Inhibition of cardiac oxidative and endoplasmic reticulum stress-mediated apoptosis by curcumin treatment contributes to protection against acute myocarditis. Free Radic Res 2011; 45:1223-31. [DOI: 10.3109/10715762.2011.607252] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
Abstract
Over the past several decades, investigations in humans and animal models of heart failure (HF) have provided substantial evidence that oxidative stress is increased in HF and contributes to disease progression. The high metabolic activity of cardiac myocytes makes these cells active sources of reactive oxygen species. Work in cell and animal models clearly demonstrates that oxidative stress activates processes such as changes in gene expression and cell death that are now accepted components of myocardial remodeling and HF. Antioxidants prevent progressive remodeling and even improve cardiac function in animal models of HF. It is therefore disappointing that to date no antioxidant strategy has translated to a therapeutic in the HF clinic. Possible explanations, including inadequate appreciation of the critical disease-modifying sources of reactive oxygen species, the choice of the wrong antioxidant strategy, or incomplete understanding of individual variability in human antioxidant defenses in this brief review.
Collapse
Affiliation(s)
- Douglas B Sawyer
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA.
| |
Collapse
|
25
|
Barin JG, Rose NR, Ciháková D. Macrophage diversity in cardiac inflammation: a review. Immunobiology 2011; 217:468-75. [PMID: 21820754 DOI: 10.1016/j.imbio.2011.06.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 06/23/2011] [Indexed: 01/04/2023]
Abstract
Cardiac inflammatory disease represents a significant public health burden, and interesting questions of immunopathologic science and clinical inquiry. Novel insights into the diverse programming and functions within the macrophage lineages in recent years have yielded a view of these cells as dynamic effectors and regulators of immunity, host defense, and inflammatory disease. In this review, we examine and discuss recent investigations into the complex participation of mononuclear phagocytic cells in the pathology of animal models of myocarditis.
Collapse
Affiliation(s)
- Jobert G Barin
- The Johns Hopkins University School of Medicine, Training Program in Immunology, USA
| | | | | |
Collapse
|
26
|
Barin JG, Talor MV, Baldeviano GC, Kimura M, Rose NR, Čiháková D. Mechanisms of IFNγ regulation of autoimmune myocarditis. Exp Mol Pathol 2010; 89:83-91. [PMID: 20599938 PMCID: PMC4266481 DOI: 10.1016/j.yexmp.2010.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/25/2010] [Accepted: 06/04/2010] [Indexed: 12/21/2022]
Abstract
A protective effect of interferon-gamma (IFNγ) has been described in a number of models of autoimmune disease, including experimental autoimmune myocarditis (EAM). Some reports have suggested that regulation of apoptosis in autoreactive lymphocytes mediate these protective functions. We examined the potential of IFNγ to regulate apoptotic mechanisms in detail, both in vitro and in vivo in EAM. We observed multiple apoptotic defects in caspase activity, and the expression of TNF superfamily members on CD4(+) T cells. In addition, we observed selective defects in CD4(+) T cell activation in response to antigenic stimulation. These activation and apoptotic defects were CD4(+) cell autonomous, independent of the genotype of APCs. Inhibition of nitric oxide production in vivo did not reproduce the severe form of EAM of IFNγ-deficient mice, indicating that this pathway does not mediate the protective effect of IFNγ. Crosswise adoptive transfer of wild type, IFNγ(-/-), and IFNγR(-/-)EAM demonstrated that IFNγ signaling was critical in CD4(+) cells, but that non-CD4(+) sources of IFNγ production were also involved in the control of disease. Together, these data indicate multiple mechanisms of autonomous and non-autonomous CD4(+) T cell regulation mediated by IFNγ in the control of autoimmune heart disease.
Collapse
Affiliation(s)
- Jobert G. Barin
- Training Program in Immunology, Johns Hopkins University School of Medicine
| | - Monica V. Talor
- Department of Pathology, Division of Immunology, Johns Hopkins University School of Medicine
| | - G. Christian Baldeviano
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health
| | - Miho Kimura
- Department of Pathology, Division of Immunology, Johns Hopkins University School of Medicine
| | - Noel R. Rose
- Training Program in Immunology, Johns Hopkins University School of Medicine
- Department of Pathology, Division of Immunology, Johns Hopkins University School of Medicine
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health
| | - Daniela Čiháková
- Department of Pathology, Division of Immunology, Johns Hopkins University School of Medicine
| |
Collapse
|
27
|
Liu W, Shimada M, Xiao J, Hu D, Matsumori A. Nifedipine inhibits the activation of inflammatory and immune reactions in viral myocarditis. Life Sci 2009; 85:235-40. [PMID: 19520090 DOI: 10.1016/j.lfs.2009.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Revised: 04/22/2009] [Accepted: 05/28/2009] [Indexed: 11/18/2022]
Abstract
AIMS The aim of study is to investigate the effect of nifedipine on viral myocarditis in an animal model. MAIN METHODS Four-week-old male DBA/2 mice were inoculated with 2 pfu of encephalomyocarditis virus (EMCV) and randomized to nifedipine (n=10) or control (n=10) group. The control group was fed by regular chow and the nifedipine group contained 0.01% of nifedipine. Mast cell density was counted, and expressions of messenger RNAs of stem cell factor (SCF), matrix metalloproteinases (MMPs), pro-collagen I, mast cell proteases, tumor necrosis factor-alpha (TNF-alpha), and interleukin-6 (IL-6) were evaluated by RT-PCR. KEY FINDINGS The area of myocardial necrosis was smaller in the nifedipine vs the control group (mean+/-SD, 1.2+/-1.3% vs 3.8+/-1.8%, respectively, P<0.005). The mast cell density (count/mm(2)) was lower in the nifedipine vs the control group (mean+/-SD, 0.23+/-0.16 vs 1.08+/-0.45, respectively, P<0.0005). The expressions of MMPs, mast cell proteases, TNF-alpha, IL-6, SCF and pro-collagen I were lower in the nifedipine group than in the control group (P<0.05). SIGNIFICANCE Nifedipine inhibited the activation of various participants in inflammatory and immune reactions in EMCV myocarditis.
Collapse
Affiliation(s)
- Wenling Liu
- Peking University People's Hospital, Beijing, China
| | | | | | | | | |
Collapse
|
28
|
Barsheshet A, Barshack I, Keren P, Keren G, George J. Whole-body hyperthermia attenuates experimental autoimmune myocarditis in the rat. Cardiovasc Pathol 2008; 17:375-81. [DOI: 10.1016/j.carpath.2008.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 12/08/2007] [Accepted: 02/08/2008] [Indexed: 11/16/2022] Open
|
29
|
Nitric oxide overproduction derived from inducible nitric oxide synthase increases cardiomyocyte apoptosis in human atrial fibrillation. Int J Cardiol 2008; 130:165-73. [PMID: 18585800 DOI: 10.1016/j.ijcard.2008.02.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Revised: 11/13/2007] [Accepted: 02/11/2008] [Indexed: 11/23/2022]
Abstract
BACKGROUND Inflammation and oxidant stress have been suggested to be involved in the structural remodeling in atrial fibrillation (AF), and inducible nitric oxide synthase (iNOS) is associated with inflammation and oxidant stress. To study whether iNOS could contribute to atrial remodeling in AF, we investigated the relationship between inflammation, oxidant stress, nitric oxide (NO) and its synthase, and cardiomyocytic apoptosis in the right atrium in human AF. METHODS Fifteen patients with permanent AF (PmAF) and 17 patients with sinus rhythm (SR), who underwent mitral valve replacement surgery were investigated. Western blotting and immunohistochemical staining were used to detect the expression of endothelial nitric oxide synthase (eNOS), iNOS and 3-nitrotyrosine (3NT; a biological marker of peroxynitrite) in the right atrium. The occurrence of cardiomyocytic apoptosis was determined by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL). Caspase 3 staining for the activated, cleaved protease was also performed. In addition, concentrations of NO(2)(-)/NO(3)(-) (NO(X)) both in plasma and in the right atrium were measured by a NO(X) Detection Kit. Finally, plasma levels of high-sensitivity C-reactive protein (hs-CRP) were routinely measured. RESULTS AND CONCLUSIONS Levels of hs-CRP were far more enhanced in patients with PmAF compared to the controls. Plasma levels of NO(X) were significantly lower in PmAF patients than SR patients, but the production of NO(X) in the local right atrium increased obviously. Furthermore, iNOS and 3NT expressions increased dramatically in the right atrium in PmAF patients, whereas the expression of eNOS did not change apparently. In addition, when patients were further divided into a higher hs-CRP group (> or =5 mg/L) and a lower hs-CRP group (<5 mg/L) according to the hs-CRP level, significant upregulation of iNOS was found in the higher hs-CRP group. Apoptosis index and caspase 3 staining were also prominently enhanced in PmAF patients compared with SR patients. More importantly, we demonstrated in this study that a higher expression of 3NT was associated with an increased expression of iNOS/eNOS (r=0.74, P<0.05) and an enhanced apoptosis index (r=0.69, P<0.05). In conclusion, the results presented novel evidence that imbalanced expression of iNOS/eNOS could contribute to protein nitration and cardiomyocyte apoptosis in human AF, in which condition inflammation may be an important participant.
Collapse
|
30
|
Gutierrez FRS, Lalu MM, Mariano FS, Milanezi CM, Cena J, Gerlach RF, Santos JET, Torres-Dueñas D, Cunha FQ, Schulz R, Silva JS. Increased activities of cardiac matrix metalloproteinases matrix metalloproteinase (MMP)-2 and MMP-9 are associated with mortality during the acute phase of experimental Trypanosoma cruzi infection. J Infect Dis 2008; 197:1468-76. [PMID: 18444803 DOI: 10.1086/587487] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The strong inflammatory reaction that occurs in the heart during the acute phase of Trypanosoma cruzi infection is modulated by cytokines and chemokines produced by leukocytes and cardiomyocytes. Matrix metalloproteinases (MMPs) have recently emerged as modulators of cardiovascular inflammation. In the present study we investigated the role of MMP-2 and MMP-9 in T. cruzi-induced myocarditis, by use of immunohistochemical analysis, gelatin zymography, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction to analyze the cardiac tissues of T. cruzi-infected C57BL/6 mice. Increased transcripts levels, immunoreactivity, and enzymatic activity for MMP-2 and MMP-9 were observed by day 14 after infection. Mice treated with an MMP inhibitor showed significantly decreased heart inflammation, delayed peak in parasitemia, and improved survival rates, compared with the control group. Reduced levels of cardiac tumor necrosis factor-alpha, interferon-gamma, serum nitrite, and serum nitrate were also observed in the treated group. These results suggest an important role for MMPs in the induction of T. cruzi-induced acute myocarditis.
Collapse
|
31
|
Abstract
BACKGROUND Myosin-induced autoimmune myocarditis of rats is a model of human dilated cardiomyopathy. Resveratrol is a natural polyphenol found in grapes and wine that is reported to have cardioprotective and immunomodulatory effects. METHODS AND RESULTS To examine the effect of resveratrol on myocarditis, vehicle or resveratrol (50 mg/kg per day) was administered to cardiac myosin immunized rats 1 day before the immunization. At 14 days after immunization, resveratrol had preserved cardiac function of myosin-immunized rats according to echocardiographic analysis. The heart weight/tibial length ratio of vehicle-treated myosin-immunized rats was increased by 1.8-fold compared with unimmunized rats, and resveratrol attenuated the heart weight increase. Resveratrol significantly decreased cellular infiltration, fibrosis, and expression of inflammatory cytokines in the myocardium. Expressions of antioxidant genes were increased in myosin-immunized hearts, and resveratrol decreased those expressions. Resveratrol also attenuated myocarditis 21 days after immunization. SIRT1, a potential effector of resveratrol, was increased in the myocardium of myosin-immunized rats compared with unimmunized rats. The SIRT1 protein was localized mainly in infiltrating mononuclear cells. CONCLUSIONS Resveratrol significantly ameliorated myocardial injury and preserved cardiac function in a rat model of autoimmune myocarditis. Resveratrol may be a therapeutic modality for myocarditis.
Collapse
Affiliation(s)
- Yuki Yoshida
- Department of Internal Medicine and Cardiology, Kitasato University School of Medicine, Sagamihara, Japan
| | | | | |
Collapse
|
32
|
Kittleson MM, Lowenstein CJ, Hare JM. Novel pathogenetic mechanisms in myocarditis: nitric oxide signaling. Heart Fail Clin 2007; 1:345-61. [PMID: 17386859 DOI: 10.1016/j.hfc.2005.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
33
|
Abstract
The discovery that mammalian cells have the ability to synthesize the free radical nitric oxide (NO) has stimulated an extraordinary impetus for scientific research in all the fields of biology and medicine. Since its early description as an endothelial-derived relaxing factor, NO has emerged as a fundamental signaling device regulating virtually every critical cellular function, as well as a potent mediator of cellular damage in a wide range of conditions. Recent evidence indicates that most of the cytotoxicity attributed to NO is rather due to peroxynitrite, produced from the diffusion-controlled reaction between NO and another free radical, the superoxide anion. Peroxynitrite interacts with lipids, DNA, and proteins via direct oxidative reactions or via indirect, radical-mediated mechanisms. These reactions trigger cellular responses ranging from subtle modulations of cell signaling to overwhelming oxidative injury, committing cells to necrosis or apoptosis. In vivo, peroxynitrite generation represents a crucial pathogenic mechanism in conditions such as stroke, myocardial infarction, chronic heart failure, diabetes, circulatory shock, chronic inflammatory diseases, cancer, and neurodegenerative disorders. Hence, novel pharmacological strategies aimed at removing peroxynitrite might represent powerful therapeutic tools in the future. Evidence supporting these novel roles of NO and peroxynitrite is presented in detail in this review.
Collapse
Affiliation(s)
- Pál Pacher
- Section on Oxidative Stress Tissue Injury, Laboratory of Physiologic Studies, National Institutes of Health, National Institute of Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
34
|
Veeraveedu PT, Watanabe K, Ma M, Gurusamy N, Palaniyandi SS, Wen J, Prakash P, Wahed MII, Kamal FA, Mito S, Kunisaki M, Kodama M, Aizawa Y. Comparative Effects of Pranidipine with Amlodipine in Rats with Heart Failure. Pharmacology 2006; 77:1-10. [PMID: 16508340 DOI: 10.1159/000091746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2005] [Accepted: 01/12/2006] [Indexed: 11/19/2022]
Abstract
The aim of the present study was to compare the cardioprotective properties of long-acting calcium channel antagonist pranidipine with amlodipine in rat model of heart failure induced by autoimmune myocarditis. Twenty-eight days after immunization the surviving rats were randomized for the oral administration of low-dose amlodipine (1 mg/kg/day), high-dose amlodipine (5 mg/kg/day), pranidipine (0.3 mg/kg/day) or vehicle (0.5% methylcellulose). After oral administration for 1 month, the animals underwent echocardiography and hemodynamic analysis. Histopathology, immunohistochemistry, and Western immunoblotting were carried out in the heart samples. Both pranidipine and high-dose amlodipine increased survival rate. Although the heart rate did not differ among the four groups, left ventricular end-diastolic pressure was significantly decreased and +/-dP/dt was increased in the pranidipine- and high-dose amlodipine-treated rats, but not in low-dose amlodipine-treated rats. In comparison to amlodipine treatment, pranidipine treatment significantly reduced myocyte size and central venous pressure. Furthermore, both pranidipine and high-dose amlodipine treatment significantly reduced myocardial protein levels of atrial natriuretic peptide and inducible nitric oxide synthase, whereas pranidipine only significantly decreased tumor necrosis factor-alpha, and improved sarcoplasmic reticulum Ca2+ ATPase2 protein levels. We conclude that pranidipine ameliorates the progression of left ventricular dysfunction and cardiac remodeling in rats with heart failure after autoimmune myocarditis in a lower dose when compared to amlodipine and which may be a clinically potential therapeutic agent for the treatment of heart failure.
Collapse
Affiliation(s)
- Punniyakoti T Veeraveedu
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nimata M, Okabe TA, Hattori M, Yuan Z, Shioji K, Kishimoto C. MCI-186 (edaravone), a novel free radical scavenger, protects against acute autoimmune myocarditis in rats. Am J Physiol Heart Circ Physiol 2005; 289:H2514-8. [PMID: 16100244 DOI: 10.1152/ajpheart.00661.2005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In this study, we tested the hypothesis that MCI-186 (3-methyl-1-phenyl-2-pyrazolin-5-one; edaravone), a novel free radical scavenger, protects against acute experimental autoimmune myocarditis (EAM) in rats by the radical scavenging action associated with the suppression of cytotoxic myocardial injury. Recent evidence suggests that oxidative stress may play a role in myocarditis. We administered MCI-186 intraperitoneally at 1, 3, and 10 mg·kg−1·day−1 to rats with EAM for 3 wk. The results were compared with untreated rats with EAM. MCI-186 treatment did not affect hemodynamics. MCI-186 treatment (3 and 10 mg·kg−1·day−1) reduced the severity of myocarditis as assessed by comparing the heart-to-body weight ratio and pathological scores. Myocardial interleukin-1β (IL-1β)-positive cells and myocardial oxidative stress overload with DNA damage in rats with EAM given MCI-186 treatment were significantly less compared with those of the untreated rats with EAM. In addition, MCI-186 treatment decreased not only the myocardial protein carbonyl contents but also the myocardial thiobarbituric acid reactive substance products in rats with EAM. The formation of hydroxyl radicals in MCI-186-treated heart homogenates was decreased compared with untreated heart homogenates. Furthermore, cytotoxic activities of lymphocytes of rats with EAM treated with MCI-186 were significantly lower compared with those of the untreated rats with EAM. Hydroxyl radicals may be involved in the development of myocarditis. MCI-186 protects against acute EAM in rats associated with scavenging hydroxyl free radicals, resulting in the suppression of autoimmune-mediated myocardial damage associated with reduced oxidative stress state.
Collapse
Affiliation(s)
- Masaomi Nimata
- Dept. of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Univ., 54 Kawaracho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Nimata M, Kishimoto C, Yuan Z, Shioji K. Beneficial effects of olmesartan, a novel angiotensin II receptor type 1 antagonist, upon acute autoimmune myocarditis. Mol Cell Biochem 2005; 259:217-22. [PMID: 15124927 DOI: 10.1023/b:mcbi.0000021379.82282.53] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Excess amount of cytokine produced by inflammatory stimuli contributes to the progression of myocardial damage in myocarditis. Some angiotensin II receptor type 1 antagonists are reported to inhibit proinflammatory cytokine production in vitro and in vivo. We tested the hypothesis that olmesartan, a novel angiotensin II receptor type 1 antagonist, ameliorated experimental autoimmune myocarditis (EAM) in rats attributing to the suppression of inflammatory cytokines in the heart. We orally administered olmesartan 1, 3, and 10 mg/kg/day to rats with EAM for 3 weeks. The results showed that olmesartan decreased blood pressure significantly compared with the untreated group, but markedly reduced the severity of myocarditis by comparing the heart weight/body weight ratio, pericardial effusion scores, macroscopic scores and microscopic scores. Myocardial interleukin (IL)- 1beta expression by western blotting and IL-1beta-positive staining cells by immunohistochemistry were significantly lower in rats with EAM given olmesartan treatment compared with those of rats given vehicle. We conclude that Olmesartan ameliorates acute EAM in rats. The cardioprotection of olmesartan may be due to suppression of inflammatory cytokines dependent of the hemodynamic modifications.
Collapse
Affiliation(s)
- Masaomi Nimata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|
37
|
Maeda K, Shioi T, Kosugi R, Yoshida Y, Takahashi K, Machida Y, Izumi T. Rapamycin Ameliorates Experimental Autoimmune Myocarditis. Int Heart J 2005; 46:513-30. [PMID: 16043946 DOI: 10.1536/ihj.46.513] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Myosin-induced autoimmune myocarditis in rats is a model of human dilated cardiomyopathy. Rapamycin is a potent immunosuppressant and specifically inactivates the mammalian target of rapamycin (mTOR). To examine the role of mTOR in autoimmune myocarditis, we administered rapamycin to rats immunized with cardiac myosin. Phosphorylation of p70 ribosomal S6 kinase 1 (S6K1), a target of mTOR, was increased by 6.9 fold in the heart tissue of myosin immunized rats. Rapamycin (2 mg/kg/day) completely suppressed S6K1 and S6 phosphorylation. The amount of interleukin-1beta, interferon-gamma, interleukin-2, or tumor necrosis factor-alpha mRNA in the heart tissue was markedly increased in myosin-immunized rats, and rapamycin significantly attenuated the cytokine gene expressions. Rapamycin improved the survival of the rats and preserved cardiac function. The plasma level of brain natriuretic peptide increased by 4.7 fold in myosin-immunized rats, and rapamycin attenuated the increase in plasma brain natriuretic peptide. The heart weight/tibial length ratio of vehicle-treated myosin-immunized rats was increased by 1.81 +/- 0.06 fold compared with vehicle-treated unimmunized rats, and rapamycin suppressed the increase in heart weight. Rapamycin decreased the cellular infiltration and fibrosis of the myocardium. The amount of phosphorylated S6 was increased in the infiltrating mononuclear cells in vehicle-treated myosin-immunized rats. Rapamycin significantly ameliorated myocardial injury and preserved cardiac function in a rat model of autoimmune myocarditis.
Collapse
Affiliation(s)
- Kayo Maeda
- Department of Internal Medicine and Cardiology, Kitasato University School of Medicine, Sagamihara, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Abd El-Gawad HM, El-Sawalhi MM. Nitric oxide and oxidative stress in brain and heart of normal rats treated with doxorubicin: Role of aminoguanidine. J Biochem Mol Toxicol 2004; 18:69-77. [PMID: 15122648 DOI: 10.1002/jbt.20013] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Doxorubicin (DOX) is a potent antitumor antibiotic drug known to cause severe cardiac toxicity. Moreover, its adverse effects were found to be extended to the cerebral tissue. Several mechanisms for this toxicity have been ascribed. Currently, one of the most accepted mechanisms is through free radicals; however, the exact role of nitric oxide (NO) is still unclear. Accordingly, a NO-synthase inhibitor with some antioxidant property, aminoguanidine (AG), was selected to examine its potential protective effect against DOX-induced toxicity. Male Wistar albino rats (150-200 g) were allocated into a normal control group, DOX-induced toxicity group, and DOX + AG-treated group. DOX was injected i.p. at a dose of 10 mg/kg divided into four equal injections over a period of 2 weeks. AG was injected i.p. at a dose of 100 mg/kg 1 h before each DOX injection. The animals were sacrificed 24 h after the last DOX injection and the following parameters were measured: serum lactate dehydrogenase (LDH) and creatine phosphokinase (CPK) activities, cardiac and cerebral contents of malondialdehyde (MDA), conjugated diene (CD), glutathione (GSH), NO, and cytosolic calcium, as well as superoxide dismutase (SOD) and glutathione peroxidase (GSHP(X)) activities. Cardiotoxicity was manifested by a marked increase in serum LDH and CPK in addition to the sharp increase in MDA reaching eightfolds the basal level. This was accompanied by significant increase in CD, NO, cytosolic calcium, SOD, and GSHP(X) content/activity by 69, 85, 76, 125, and 41% respectively as compared to normal control. On the other hand, GSH was significantly depressed. In brain, only significant increase in MDA and GSHP(X) and decrease in GSH were obtained but to a lesser extent than the cardiac tissue. AG treatment failed to prevent the excessive release of cardiac enzymes; however, it alleviated the adverse effects of DOX in heart. AG administration resulted in marked decrease in the elevated levels of MDA, NO, SOD, and GSHP(X), however, MDA level was still pathological. The altered parameters in brain were restored by AG. It is concluded that, AG could not provide complete protection against DOX-induced toxicity. Therefore, it is recommended that, maintenance of the endogenous antioxidant, GSH, and regulation of calcium homeostasis must be considered, rather than NO formation, to guard against DOX-induced toxicity.
Collapse
|
39
|
Wheeler DS, Kooy NW. A formidable challenge: the diagnosis and treatment of viral myocarditis in children. Crit Care Clin 2003; 19:365-91. [PMID: 12848311 DOI: 10.1016/s0749-0704(03)00006-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
It is generally well accepted that one third of patients with viral myocarditis experience a complete recovery of normal cardiac function, one third improve clinically but show residual cardiac dysfunction, and one third experience chronic heart failure and die or require heart transplantation. It is hoped that a better understanding of the underlying cause and pathogenesis of this disease will increase the number of patients who experience a complete recovery. New advances in both the diagnosis and treatment of viral myocarditis continue to enter clinical practice at a rapid pace, and it is likely that a genomic approach to the diagnostic evaluation and treatment of this disease will become possible in the near future. Viral myocarditis, however, will remain a significant diagnosticand therapeutic challenge to both physicians and scientists alike.
Collapse
Affiliation(s)
- Derek S Wheeler
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | |
Collapse
|
40
|
Yuan Z, Kishimoto C, Shioji K. Beneficial effects of low-dose benidipine in acute autoimmune myocarditis: suppressive effects on inflammatory cytokines and inducible nitric oxide synthase. Circ J 2003; 67:545-50. [PMID: 12808275 DOI: 10.1253/circj.67.545] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Excessive production of nitric oxide (NO) by inducible NO synthase (iNOS) contributes to the progression of myocardial damage in myocarditis. Some dihydropyridine calcium channel blockers reportedly inhibit NO production and proinflammatory cytokines and the present study sought to clarify if a low dose of benidipine, a novel dihydropyridine calcium channel blocker, would ameliorate experimental autoimmune myocarditis (EAM). Rats with or without myocarditis were administered oral benidipine at a dose of 3 mg. kg(-1). day(-1) for 3 weeks. Low-dose benidipine did not decrease blood pressure significantly compared with the untreated group, but markedly reduced the severity of myocarditis. Myocardial interleukin-1beta (IL-1beta) expression and IL-1beta-positive cells were significantly less in rats with EAM that were treated with low-dose benidipine compared with untreated rats. Also, myocardial iNOS expression and iNOS-positive cells were markedly reduced in in the treated rats compared with the untreated group. Furthermore, myocardial NO production and nitrotyrosine expression were suppressed by the treatment in rats with EAM. The cardioprotection of low-dose benidipine may be caused by suppression of inflammatory cytokines and inhibition of NO production.
Collapse
Affiliation(s)
- Zuyi Yuan
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan
| | | | | |
Collapse
|
41
|
Matsui Y, Okamoto H, Inobe M, Jia N, Shimizu T, Akino M, Sugawara T, Tezuka K, Nakayama Y, Morimoto J, Kimura C, Kon S, Miyazaki T, Kitabatake A, Uede T. Adenovirus-mediated gene transfer of ICOSIg fusion protein ameliorates ongoing experimental autoimmune myocarditis. Hum Gene Ther 2003; 14:521-32. [PMID: 12718763 DOI: 10.1089/104303403764539314] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Experimental autoimmune myocarditis (EAM) has been used as a model for human myocarditis. We previously demonstrated that blockade of B7/CD28 or CD40/CD40 ligand (CD40L) had a potential preventive effect on EAM, but less therapeutic effect on ongoing EAM. Thus, we searched for the involvement of other costimulatory molecules in EAM. We demonstrated the expression of inducible costimulator (ICOS)/ICOSL molecules in the lymph nodes, spleen, and heart in the EAM rat. We constructed adenovirus vectors containing ICOSIg (Adex1CAICOSIg) to achieve effective inhibition of ICOS/ICOSL interaction, and examined the effects of Adex1CAICOSIg on EAM. Adex1CAICOSIg treatment shortly after the immunization did not inhibit the onset and severity of EAM compared to control rats. On the other hand, delayed treatment with Adex1CAICOSIg significantly inhibited ongoing EAM. The survival rate in rats treated with Adex1CAICOSIg was significantly higher than that of the control group. Furthermore, the affected area ratio of the Adex1CAICOSIg treatment group was significantly lower than that of the control group. This study indicates that ICOS/ICOSL costimulation makes an important contribution to the progression of EAM and that the blockade of this pathway by gene transfer has therapeutic potential for ongoing autoimmune myocarditis.
Collapse
Affiliation(s)
- Yutaka Matsui
- Division of Molecular Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The newest treatment strategies for pediatric myocarditis have evolved from an understanding of the pathophysiology of myocyte damage. Although the initial stages of viral myocarditis apparently result from the direct cytopathic effects on the atrial and ventricular myocardium, later stages of progressive decompensation result from immune-mediated myocyte destruction common to many forms of myocarditis. Despite advances in the understanding of the role of genetics, immunologic mechanisms, and infectious causes of myocarditis, supportive therapy continues to remain the cornerstone of treatment. Presently, therapies include supportive management with anticongestive agents, antiviral medications, and therapies that attempt to interrupt the immunologic cascade. Clinical studies have yet to provide convincing evidence that the use of immunosuppressants and gamma-globulin favorably alters the outcome for pediatric patients with acute myocarditis. Ventricular assist devices and heart transplantation remain as treatment options for all pediatric patients with severe myocarditis resistant to all other therapies. Although this review will focus on viral myocarditis, the supportive strategies and surgical treatment options apply to most forms of cardiomyopathy.
Collapse
Affiliation(s)
- Daniel Levi
- Department of Pediatrics, School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | |
Collapse
|
43
|
Ferdinandy P, Schulz R. Nitric oxide, superoxide, and peroxynitrite in myocardial ischaemia-reperfusion injury and preconditioning. Br J Pharmacol 2003; 138:532-43. [PMID: 12598407 PMCID: PMC1573696 DOI: 10.1038/sj.bjp.0705080] [Citation(s) in RCA: 330] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
There appears to be a controversy in the study of myocardial ischaemia-reperfusion injury and preconditioning whether nitric oxide (NO) plays a protective or detrimental role. A number of findings and the interpretation of the results to date do not support such a controversy. An understanding of the latest developments in NO, superoxide (O(2)(-)*) and peroxynitrite (ONOO(-)) biology, as well as the various ischaemic animal models utilized is necessary to resolve the apparent controversy. NO is an important cardioprotective molecule via its vasodilator, antioxidant, antiplatelet, and antineutrophil actions and it is essential for normal heart function. However, NO is detrimental if it combines with O(2)(-)* to form ONOO(-) which rapidly decomposes to highly reactive oxidant species. There is a critical balance between cellular concentrations of NO, O(2)(-)*, and superoxide dismutase which physiologically favour NO production but in pathological conditions such as ischaemia and reperfusion result in ONOO(-) formation. In contrast, exposure of the heart to brief episode(s) of ischaemia markedly enhances its ability to withstand a subsequent ischaemic injury. The triggering of this endogenous cardioprotective mechanism known as preconditioning requires both NO and O(2)(-)* synthesis. However, preconditioning in turn attenuates the overproduction of NO, O(2)(-)* and ONOO(-) during a subsequent episode of ischaemia and reperfusion, thereby protecting the heart. Here we review the roles of NO, O(2)(-)*, and ONOO(-) in both ischaemia-reperfusion injury and preconditioning.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720 Hungary.
| | | |
Collapse
|
44
|
Kobayashi N, Horinaka S, Mita SI, Yoshida K, Honda T, Kobayashi T, Hara K, Nishikimi T, Matsuoka H. Aminoguanidine inhibits mitogen-activated protein kinase and improves cardiac performance and cardiovascular remodeling in failing hearts of salt-sensitive hypertensive rats. J Hypertens 2002; 20:2475-85. [PMID: 12473873 DOI: 10.1097/00004872-200212000-00028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Congestive heart failure (CHF) is associated with inducible nitric oxide synthase (iNOS) expression in the failing human heart, and recently we have also demonstrated that iNOS expression was upregulated in Dahl salt-sensitive hypertensive (DS) rats with cardiac dysfunction and vascular remodeling. Thus, we evaluated whether aminoguanidine (AG), a selective iNOS inhibitor, protects against cardiac dysfunction and vascular remodeling in DS rats receiving a high-salt diet. METHODS AG (DSHF-AG, 150 mg/kg per day) or vehicle (DSHF-V) were given from left ventricular hypertrophy stage (11 weeks) to CHF stage (18 weeks) for 7 weeks. The left ventricular end-systolic pressure-volume relationship (contractility: E(es)) was measured by conductance catheter. RESULTS Decreased E(es) in DSHF-V was significantly ameliorated by AG treatment. The iNOS mRNA and protein expression and phospho-p42/p44 extracellular signal-regulated kinase (ERK) activities in the left ventricle were significantly upregulated in DSHF-V compared with control rats, and significantly suppressed in DSHF-AG compared with DSHF-V. DSHF-V showed a significant increase of perivascular fibrosis and myocardial fibrosis, with all these parameters being significantly improved by AG treatment. CONCLUSIONS We demonstrated that the selective iNOS inhibitor, AG, may be at least a potential therapeutic strategy for treating CHF and cardiovascular remodeling.
Collapse
Affiliation(s)
- Naohiko Kobayashi
- Department of Hypertension and Cardiorenal Medicine, Dokkyo University School of Medicine, Mibu, Tochigi, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Predescu D, Predescu S, Malik AB. Transport of nitrated albumin across continuous vascular endothelium. Proc Natl Acad Sci U S A 2002; 99:13932-7. [PMID: 12370442 PMCID: PMC129800 DOI: 10.1073/pnas.212253499] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Because modification of plasma albumin on tyrosine residues generates nitrated albumin (NOA) that may function as a mechanism of nitrogen monoxide clearance from microcirculation, we investigated biochemicaly and morphologically the cell surface binding and the transendothelial transport of NOA. An electron microscopic study was carried out with mouse lungs and hearts perfused in situ with NOA and NOA-Au complexes. The results indicate that NOA-Au can bind to the endothelial cell surface, and its binding can be blocked by albumin plus nitrotyrosine (NO-tyrosine) or abolished by excess NOA. We detected NOA-Au into perivascular spaces as early as 30 sec after the beginning of its perfusion. NOA, unlike native albumin, leaves the vascular lumina via both endothelial caveolae and open junctions. By cross-linking and ligand blotting analysis, we showed that NOA interacted with the same albumin binding proteins of 16-18, 30-32, 60, and 74 kDa as native albumin. ELISA performed on tissue homogenates obtained from the same specimens showed that NOA transport was 2- to 4-fold greater than native albumin. The augmented transendothelial transport of NOA reflects its transcytosis as well as its exit from the microcirculation via open junctions. The increased transport of NOA may serve as an important mechanism that protects a vascular bed against the damaging effects of nitrogen monoxide and peroxynitrite.
Collapse
Affiliation(s)
- Dan Predescu
- Department of Pharmacology, University of Illinois College of Medicine, 835 Wolcott Avenue, MC 868, Chicago, IL 60612, USA
| | | | | |
Collapse
|
46
|
Abstract
Myocarditis offers a unique opportunity to study the factors contributing to its transition from a viral infection to an autoimmune disease. In this article, we review recent studies on the role of nitric oxide (NO), gamma interferon (IFN-gamma) and interleukin 12 (IL-12) in the progression from early (viral) to late (autoimmune) phases of myocarditis induced by Coxsackievirus B3 (CB3) in highly susceptible (A.CA) and moderately susceptible (B10.M) mice. NO plays a paradoxical role, being protective in early stages but detrimental later in the course of disease. Treatment with antibody to IFN-gamma reduced early disease, but had little effect on the severity of cardiac lesions at later times. Treatment with recombinant (r) IL-12 significantly reduced the autoimmune cardiac lesions in moderately susceptible B10.M mice, but had no measurable effect in highly susceptible A.CA animals. These studies provide evidence that the profile of inflammatory mediators produced early in the course of viral infection determines the later development of autoimmune disease.
Collapse
Affiliation(s)
- S L Hill
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Medical Institutions, JHU School of Hygiene and Public Health, Baltimore, MD 21205, USA
| | | |
Collapse
|
47
|
Shiraishi H, Toyozaki T, Tsukamoto Y, Saito T, Masuda Y, Hiroshima K, Ohwada H, Kobayashi N, Hiroe M. Antibody binding to fas ligand attenuates inflammatory cell infiltration and cytokine secretion, leading to reduction of myocardial infarct areas and reperfusion injury. J Transl Med 2002; 82:1121-9. [PMID: 12218072 DOI: 10.1097/01.lab.0000029148.88524.ce] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Fas ligand (FasL) induces apoptotic cell death when bound to Fas antigen. The engagement of FasL has anti-inflammatory effects through the prevention of cell proliferation and cytokine secretion. However, the role of FasL in myocardial ischemia/reperfusion (MI/R) injury is unclear. We examined the expression of FasL mRNA in the myocardium of MI/R rats by ligating the left coronary artery for 30 minutes and allowing reperfusion to occur for 0, 1, 3, and 24 hours. The expression of FasL mRNA was enhanced 1 hour after reperfusion, and enhanced levels were consistently seen after 24 hours of reperfusion. FasL immunostaining was observed on neutrophils, macrophages, T cells, and vascular endothelial cells. We then assessed the potential role of FasL in the cell proliferation and cytokine production seen in MI/R injury after 24 hours of reperfusion. Rats were divided into three groups; Group A, without treatment; Group B, treated with nonspecific rabbit IgG; and Group C, treated with anti-FasL antibody. Anti-FasL antibody or rabbit IgG were administered intravenously before coronary artery occlusion. In Group C, interleukin-1beta and interleukin-2 mRNA levels were decreased, and neutrophil and T cell accumulation was attenuated. The infarct area determined by triphenyltetrazolium chloride staining was significantly smaller in Group C (18 +/- 4%) than in Group A (34 +/- 2%) or Group B (33 +/- 4%) (p< 0.0001). However, there was no significant difference in the prevalence of terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end-labeling-positive cardiomyocytes among the three groups. These findings suggest that the cardioprotective effect of anti-FasL antibody is due to its anti-inflammatory action, rather than antiapoptotic action. The Fas/FasL system may be involved in the development of MI/R injury.
Collapse
Affiliation(s)
- Hirokazu Shiraishi
- Department of Cardiovascular Science and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Giri SN, Biring I, Nguyen T, Wang Q, Hyde DM. Abrogation of bleomycin-induced lung fibrosis by nitric oxide synthase inhibitor, aminoguanidine in mice. Nitric Oxide 2002; 7:109-18. [PMID: 12223180 DOI: 10.1016/s1089-8603(02)00102-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The effects of aminoguanidine (AG), a specific inhibitor of inducible nitric oxide synthase, on the bleomycin (BL)-induced lung fibrosis was evaluated in mice. The animals were placed into five groups: saline (SA)-instilled drinking water (SA+H(2)O), saline-instilled drinking water containing 0.5%AG (SA+0.5%AG), BL-instilled drinking water (BL+H(2)O), BL-instilled drinking water containing 0.2%AG (BL+0.2%AG), and BL-instilled drinking water containing 0.5%AG (BL+0.5%AG). The mice had free access to H(2)O or H(2)O containing AG and lab chow ad lib 2 days prior to intratracheal (IT) instillation of BL (0.07U/mouse/100 microL) or an equivalent volume of sterile isotonic saline. The mice in the SA+0.5%AG group consumed the greatest amount of AG without any ill effects than the mice in any other group. There were no differences in any of the measured biochemical determinants between the SA+H(2)O and SA+0.5%AG control groups. The IT instillation of BL in the BL+H(2)O group caused significant increases in the lipid peroxidation, hydroxyproline content, and prolyl hydroxylase activity of lungs and influx of inflammatory cells in the broncheoalveolar lavage fluid (BALF) as compared to both control groups. The intake of aminoguanidine by mice in the BL+0.5%AG group caused significant reductions in the BL-induced increases in all measured biochemical indices of lung fibrosis without any effects on the influx of inflammatory cells in the BALF. In fact, AG in both BL-treated groups additionally increased the total cell counts in the BALF from mice in the BL+0.2%AG and BL+0.5%AG groups as compared to the BL+H(2)O group. Histopathological evaluation of the lungs revealed that the mice in the BL+0.5%AG group had markedly fewer fibrotic lesions than mice in the BL+H(2)O group. These results demonstrate that aminoguanidine minimizes the BL-induced lung fibrosis at both the biochemical and the morphological level and support our earlier hypothesis that the production of nitric oxide plays a significant role in the pathogenesis lung fibrosis caused by BL.
Collapse
Affiliation(s)
- S N Giri
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, CA 95616, USA.
| | | | | | | | | |
Collapse
|
49
|
Abstract
Myocarditis is an insidious inflammatory disorder of the myocardium. As a clinical entity, it has been recognized for two centuries, but it defies traditional diagnostic tests. A greater understanding of the immune response underlying the pathobiology of the disorder can lead to a more rational therapeutic approach. The presentation, course and therapeutic options appear to be different in the pediatric compared with the adult population. An understanding of the difference between fulminant and acute progressive myocarditis has led to successful treatment strategies. A variety of new therapies are available, including antiviral agents, immunosuppression, and modulation of the biological response to inflammation. The specific question for patients with myocarditis is whether regimens designed to reduce or eliminate inflammation can provide clinical benefits compared with conventional heart failure therapy. This review highlights pathological mechanisms, modalities of diagnosis, and novel therapies which may improve outcomes.
Collapse
Affiliation(s)
- Desmond Bohn
- Department of Critical Care Medicine and Pediatrics, The Hospital for Sick Children, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | | |
Collapse
|
50
|
Saito T, Hu F, Tayara L, Fahas L, Shennib H, Giaid A. Inhibition of NOS II prevents cardiac dysfunction in myocardial infarction and congestive heart failure. Am J Physiol Heart Circ Physiol 2002; 283:H339-45. [PMID: 12063307 DOI: 10.1152/ajpheart.00596.2001] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Strong expression of the inducible form of nitric oxide synthase (NOS II) has been shown in the myocardium of patients with myocardial infarction (MI). We hypothesized that NOS II plays an important role in the development of MI and subsequent heart failure and that inhibition of NOS II may beneficially alter the course of the disease. Long-term administration (2 mo) of the selective NOS II inhibitor S-methylisothiourea (SMT) to rats with MI significantly improved cardiac function. A significant drop in mortality, lung water content, infarct size, and cardiomyocyte hypertrophy was also associated with the use of SMT. Plasma concentration of nitrite and nitrate was also reduced by SMT. Short-term administration of SMT (first 2 wk only) significantly reduced infarct size; however, it did not improve cardiac dysfunction measured 2 mo after MI. These findings demonstrate that induction of NOS II during MI exerts negative effects on cardiac function and structure. Long-term administration of a selective NOS II inhibitor may prove to be beneficial in the treatment of MI and congestive heart failure.
Collapse
Affiliation(s)
- Takayuki Saito
- Department of Pathology, The Montreal General Hospital and McGill University, Montreal, Quebec, Canada H3G 1A4
| | | | | | | | | | | |
Collapse
|