1
|
Zhang J, Xu H, Li Z, Feng F, Wang S, Li Y. Frequency of autoantibodies and their associated clinical characteristics and outcomes in patients with dilated cardiomyopathy: A systematic review and meta-analysis. Autoimmun Rev 2025; 24:103755. [PMID: 39855285 DOI: 10.1016/j.autrev.2025.103755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/24/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a prevalent myocardial disorder characterized by impaired cardiac function affecting either the left ventricle or both ventricles. Accumulating evidence suggests that autoimmunity represents a key mechanism implicated in its pathogenesis, as several abundant autoantibodies have been identified in patients with the condition. However, the prevalence of these antibodies (Abs) in patients with DCM compared to that in both healthy controls (HCs) and those with ischemic cardiomyopathy (ICM), as well as their potential association with DCM, remains unclear. This study aimed to elucidate the prevalence of certain autoantibodies in patients with DCM compared to that in HCs and patients with ICM, as well as to evaluate their correlation with clinical characteristics and outcomes. METHODS A comprehensive literature search of the PubMed, Web of Science, EMBASE, the Cochrane Library, and Scopus was conducted up to March 26, 2024, and any article that fulfilled our inclusion criteria was reviewed. A meta-analysis was then conducted, using both random- and fixed-effects models. RESULTS A total of 38 studies met the inclusion criteria and were pulled for this analysis. Significantly higher prevalence rates of autoantibodies targeting the anti-β1 adrenergic receptor (β1-AR; odds ratio [OR] = 4.96, p = 0.000), M2 muscarinic receptor (M2-R; OR = 4.07, p = 0.000), adenine nucleotide translocator (ANT; OR = 21.18, p = 0.001) and myosin (OR = 12.26, p = 0.000) were observed in patients with DCM compared to HCs. Moreover, patients with DCM exhibited a significantly higher frequency of positive ANT Abs (OR = 34.52, p = 0.005) compared to those with ICM. Regarding clinical characteristics and outcomes, seropositivity for β1-AR Abs was found to be significantly correlated with New York Heart Association (NYHA) classification (standardized mean difference [SMD] = 0.78, p = 0.006), left ventricular ejection fraction (LVEF) (SMD = -1.38, p = 0.001), and heart rate (HR) (SMD = 1.505, p = 0.022). Seropositivity for anti‑calcium channel Abs was significantly associated with sudden cardiac death (SCD; OR = 3.17, p = 0.000) and all-cause mortality (OR = 2.06, p = 0.008), while anti-troponin I (TnI) Abs were associated with atrial fibrillation (OR = 0.21, p = 0.042). In terms of Ab prevalence rates, significant heterogeneity in the frequency of anti-β1-AR Abs between studies investigating DCM and ICM may be partially explained by the detection methods used and the mean ages of the patients. Meta-regression analysis suggested that the patients' ages may partially explain the observed heterogeneity between studies regarding β1-AR Ab seropositivity and HR. However, the heterogeneity observed in the studies comparing the prevalences of Abs in patients with DCM vs HCs and ICM, as well as their associated clinical characteristics, could not be explained by subgroup analyses or demographic factors such as age and sex-nor by cardiac function. CONCLUSIONS Patients with DCM are more likely to have elevated levels of anti- β1-AR, M2-R, ANT and myosin autoantibodies compared to HCs, as well as higher levels of ANT Abs compared to patients with ICM. Anti-β1-AR, calcium channel, and TnI Abs may play an essential role in DCM severity and poor prognosis. This study represents the first comprehensive meta-analysis regarding autoantibody prevalence and DCM and may thus potentially guide the clinical management of such patients. However, further research is warranted to evaluate the accuracy of this presumed role of autoantibodies in DCM, owing to the small number of the studies included and their high degree of heterogeneity.
Collapse
Affiliation(s)
- Jingdi Zhang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Honglin Xu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhan Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Futai Feng
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Siyu Wang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China,.
| |
Collapse
|
2
|
Cardarelli S, Biglietto M, Orsini T, Fustaino V, Monaco L, de Oliveira do Rêgo AG, Liccardo F, Masciarelli S, Fazi F, Naro F, De Angelis L, Pellegrini M. Modulation of cAMP/cGMP signaling as prevention of congenital heart defects in Pde2A deficient embryos: a matter of oxidative stress. Cell Death Dis 2024; 15:169. [PMID: 38395995 PMCID: PMC10891154 DOI: 10.1038/s41419-024-06549-1] [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: 07/01/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
Phosphodiesterase 2A (Pde2A) is a dual-specific PDE that breaks down both cAMP and cGMP cyclic nucleotides. We recently highlighted a direct relationship between Pde2A impairment, a consequent increase of cAMP, and the appearance of mouse congenital heart defects (CHDs). Here we aimed to characterize the pathways involved in the development of CHDs and in their prevention by pharmacological approaches targeting cAMP and cGMP signaling. Transcriptome analysis revealed a modulation of more than 500 genes affecting biological processes involved in the immune system, cardiomyocyte development and contractility, angiogenesis, transcription, and oxidative stress in hearts from Pde2A-/- embryos. Metoprolol and H89 pharmacological administration prevented heart dilatation and hypertabeculation in Pde2A-/- embryos. Metoprolol was also able to partially impede heart septum defect and oxidative stress at tissue and molecular levels. Amelioration of cardiac defects was also observed by using the antioxidant NAC, indicating oxidative stress as one of the molecular mechanisms underpinning the CHDs. In addition, Sildenafil treatment recovered cardiac defects suggesting the requirement of cAMP/cGMP nucleotides balance for the correct heart development.
Collapse
Affiliation(s)
- Silvia Cardarelli
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Martina Biglietto
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161, Rome, Italy
- Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015, Monterotondo Scalo, Rome, Italy
| | - Tiziana Orsini
- Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015, Monterotondo Scalo, Rome, Italy
| | - Valentina Fustaino
- Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015, Monterotondo Scalo, Rome, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | | | - Francesca Liccardo
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Silvia Masciarelli
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Luciana De Angelis
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Manuela Pellegrini
- Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015, Monterotondo Scalo, Rome, Italy.
| |
Collapse
|
3
|
Kawai A, Nagatomo Y, Yukino-Iwashita M, Nakazawa R, Taruoka A, Yumita Y, Takefuji A, Yasuda R, Toya T, Ikegami Y, Masaki N, Ido Y, Adachi T. β 1 Adrenergic Receptor Autoantibodies and IgG Subclasses: Current Status and Unsolved Issues. J Cardiovasc Dev Dis 2023; 10:390. [PMID: 37754819 PMCID: PMC10531529 DOI: 10.3390/jcdd10090390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
A wide range of anti-myocardial autoantibodies have been reported since the 1970s. Among them, autoantibodies against the β1-adrenergic receptor (β1AR-AAb) have been the most thoroughly investigated, especially in dilated cardiomyopathy (DCM). Β1AR-Aabs have agonist effects inducing desensitization of β1AR, cardiomyocyte apoptosis, and sustained calcium influx which lead to cardiac dysfunction and arrhythmias. Β1AR-Aab has been reported to be detected in approximately 40% of patients with DCM, and the presence of the antibody has been associated with worse clinical outcomes. The removal of anti-myocardial autoantibodies including β1AR-AAb by immunoadsorption is beneficial for the improvement of cardiac function for DCM patients. However, several studies have suggested that its efficacy depended on the removal of AAbs belonging to the IgG3 subclass, not total IgG. IgG subclasses differ in the structure of the Fc region, suggesting that the mechanism of action of β1AR-AAb differs depending on the IgG subclasses. Our previous clinical research demonstrated that the patients with β1AR-AAb better responded to β-blocker therapy, but the following studies found that its response also differed among IgG subclasses. Further studies are needed to elucidate the possible pathogenic role of IgG subclasses of β1AR-AAbs in DCM, and the broad spectrum of cardiovascular diseases including HF with preserved ejection fraction.
Collapse
Affiliation(s)
- Akane Kawai
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Yuji Nagatomo
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Midori Yukino-Iwashita
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Ryota Nakazawa
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Akira Taruoka
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Yusuke Yumita
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Asako Takefuji
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Risako Yasuda
- Department of Intensive Care, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Takumi Toya
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Yukinori Ikegami
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Nobuyuki Masaki
- Department of Intensive Care, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Yasuo Ido
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| | - Takeshi Adachi
- Department of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan; (A.K.)
| |
Collapse
|
4
|
Markousis-Mavrogenis G, Minich WB, Al-Mubarak AA, Anker SD, Cleland JGF, Dickstein K, Lang CC, Ng LL, Samani NJ, Zannad F, Metra M, Seemann P, Hoeg A, Lopez P, van Veldhuisen DJ, de Boer RA, Voors AA, van der Meer P, Schomburg L, Bomer N, The BIOSTAT-CHF Consortium. Clinical and prognostic associations of autoantibodies recognizing adrenergic/muscarinic receptors in patients with heart failure. Cardiovasc Res 2023; 119:1690-1705. [PMID: 36883593 PMCID: PMC10325696 DOI: 10.1093/cvr/cvad042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/11/2023] [Accepted: 01/21/2023] [Indexed: 03/09/2023] Open
Abstract
AIMS The importance of autoantibodies (AABs) against adrenergic/muscarinic receptors in heart failure (HF) is not well-understood. We investigated the prevalence and clinical/prognostic associations of four AABs recognizing the M2-muscarinic receptor or the β1-, β2-, or β3-adrenergic receptor in a large and well-characterized cohort of patients with HF. METHODS AND RESULTS Serum samples from 2256 patients with HF from the BIOSTAT-CHF cohort and 299 healthy controls were analysed using newly established chemiluminescence immunoassays. The primary outcome was a composite of all-cause mortality and HF rehospitalization at 2-year follow-up, and each outcome was also separately investigated. Collectively, 382 (16.9%) patients and 37 (12.4%) controls were seropositive for ≥1 AAB (P = 0.045). Seropositivity occurred more frequently only for anti-M2 AABs (P = 0.025). Amongst patients with HF, seropositivity was associated with the presence of comorbidities (renal disease, chronic obstructive pulmonary disease, stroke, and atrial fibrillation) and with medication use. Only anti-β1 AAB seropositivity was associated with the primary outcome [hazard ratio (95% confidence interval): 1.37 (1.04-1.81), P = 0.024] and HF rehospitalization [1.57 (1.13-2.19), P = 0.010] in univariable analyses but remained associated only with HF rehospitalization after multivariable adjustment for the BIOSTAT-CHF risk model [1.47 (1.05-2.07), P = 0.030]. Principal component analyses showed considerable overlap in B-lymphocyte activity between seropositive and seronegative patients, based on 31 circulating biomarkers related to B-lymphocyte function. CONCLUSIONS AAB seropositivity was not strongly associated with adverse outcomes in HF and was mostly related to the presence of comorbidities and medication use. Only anti-β1 AABs were independently associated with HF rehospitalization. The exact clinical value of AABs remains to be elucidated.
Collapse
Affiliation(s)
- George Markousis-Mavrogenis
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Waldemar B Minich
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straß0065 4A, CCM, Berlin D-10115, Germany
- ImmunometriX GmbH i.L, Brandenburgische Str. 83, D-10713 Berlin, Germany
| | - Ali A Al-Mubarak
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité; Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Charitépl. 1, 10117 Berlin, Germany
| | - John G F Cleland
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
- National Heart & Lung Institute, Imperial College, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Kenneth Dickstein
- University of Bergen, Stavanger University Hospital, Gerd-Ragna Bloch Thorsens gate 8, 4011 Stavanger, Norway
| | - Chim C Lang
- Division of Molecular & Clinical Medicine, University of Dundee, Nethergate, Dundee DD1 4HN, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Groby Rd, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Rd, Leicester LE3 9QP, UK
| | - Nilesh J Samani
- University of Bergen, Stavanger University Hospital, Gerd-Ragna Bloch Thorsens gate 8, 4011 Stavanger, Norway
| | - Faiez Zannad
- Université de Lorraine, Inserm CIC 1403, CHRU, Cité Universitaire, 57000 Metz, France
| | - Marco Metra
- Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza del Mercato, 15, 25121 Brescia BS, Italy
| | - Petra Seemann
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straß0065 4A, CCM, Berlin D-10115, Germany
- ImmunometriX GmbH i.L, Brandenburgische Str. 83, D-10713 Berlin, Germany
| | - Antonia Hoeg
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straß0065 4A, CCM, Berlin D-10115, Germany
| | - Patricio Lopez
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straß0065 4A, CCM, Berlin D-10115, Germany
- ImmunometriX GmbH i.L, Brandenburgische Str. 83, D-10713 Berlin, Germany
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straß0065 4A, CCM, Berlin D-10115, Germany
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
5
|
Wang E, Zhou R, Li T, Hua Y, Zhou K, Li Y, Luo S, An Q. The Molecular Role of Immune Cells in Dilated Cardiomyopathy. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1246. [PMID: 37512058 PMCID: PMC10385992 DOI: 10.3390/medicina59071246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Dilated cardiomyopathy (DCM) is a rare and severe condition characterized by chamber dilation and impaired contraction of the left ventricle. It constitutes a fundamental etiology for profound heart failure and abrupt cardiac demise, rendering it a prominent clinical indication for heart transplantation (HTx) among both adult and pediatric populations. DCM arises from various etiologies, including genetic variants, epigenetic disorders, infectious insults, autoimmune diseases, and cardiac conduction abnormalities. The maintenance of cardiac function involves two distinct types of immune cells: resident immune cells and recruited immune cells. Resident immune cells play a crucial role in establishing a harmonious microenvironment within the cardiac tissue. Nevertheless, in response to injury, cardiomyocytes initiate a cytokine cascade that attracts peripheral immune cells, thus perturbing this intricate equilibrium and actively participating in the initiation and pathological remodeling of dilated cardiomyopathy (DCM), particularly during the progression of myocardial fibrosis. Additionally, immune cells assume a pivotal role in orchestrating the inflammatory processes, which are intimately linked to the prognosis of DCM. Consequently, understanding the molecular role of various immune cells and their regulation mechanisms would provide an emerging era for managing DCM. In this review, we provide a summary of the most recent advancements in our understanding of the molecular mechanisms of immune cells in DCM. Additionally, we evaluate the effectiveness and limitations of immunotherapy approaches for the treatment of DCM, with the aim of optimizing future immunotherapeutic strategies for this condition.
Collapse
Affiliation(s)
- Enping Wang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Ruofan Zhou
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Tiange Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yifei Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuhua Luo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Low-level tragus stimulation improves autoantibody-induced hyperadrenergic postural tachycardia syndrome in rabbits. Heart Rhythm O2 2023; 4:127-133. [PMID: 36873318 PMCID: PMC9975011 DOI: 10.1016/j.hroo.2022.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Recent studies have demonstrated that antiadrenergic autoantibodies are involved in the pathophysiology of postural orthostatic tachycardia syndrome (POTS). Objective The purpose of this study was to test the hypothesis that transcutaneous low-level tragus stimulation (LLTS) ameliorates autoantibody-induced autonomic dysfunction and inflammation in a rabbit model of autoimmune POTS. Methods Six New Zealand white rabbits were co-immunized with peptides from the α1-adrenergic and β1-adrenergic receptors to produce sympathomimetic antibodies. The tilt test was performed on conscious rabbits before immunization, 6 weeks after immunization, and 10 weeks after immunization with 4-week daily LLTS treatment. Each rabbit served as its own control. Results An enhanced postural heart rate increase in the absence of significant change in blood pressure was observed in immunized rabbits, confirming our previous report. Power spectral analysis of heart rate variability during the tilt test showed a predominance of sympathetic over parasympathetic activity in immunized rabbits as reflected by markedly increased low-frequency power, decreased high-frequency power, and increased low-to-high-frequency ratio. Serum inflammatory cytokines were also significantly increased in immunized rabbits. LLTS suppressed the postural tachycardia, improved the sympathovagal balance with increased acetylcholine secretion, and attenuated the inflammatory cytokine expression. Antibody production and activity were confirmed with in vitro assays, and no antibody suppression by LLTS was found in this short-term study. Conclusion LLTS improves cardiac autonomic imbalance and inflammation in a rabbit model of autoantibody-induced hyperadrenergic POTS, suggesting that LLTS may be used as a novel neuromodulation therapy for POTS.
Collapse
|
7
|
Autoimmune autonomic nervous system imbalance and conditions: Chronic fatigue syndrome, fibromyalgia, silicone breast implants, COVID and post-COVID syndrome, sick building syndrome, post-orthostatic tachycardia syndrome, autoimmune diseases and autoimmune/inflammatory syndrome induced by adjuvants. Clin Exp Rheumatol 2023; 22:103230. [PMID: 36347462 DOI: 10.1016/j.autrev.2022.103230] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Chronic fatigue syndrome (CFS), fibromyalgia, silicone breast implants syndrome (SBIs), COVID and post-COVID syndrome (PCS), sick building syndrome (SBS), post-orthostatic tachycardia syndrome (POTS), autoimmune diseases and autoimmune/inflammatory syndrome induced by adjuvants (ASIA) are frequently accompanied by clinical symptoms characteristic for dysautonomia: severe fatigue, dizziness, fogginess, memory loss, dry mouth and eyes, hearing dysfunction, tachycardia etc. The recent discovery of an imbalance of autoantibodies against G protein-coupled receptors (GPCR) in some autoimmune diseases, post-COVID syndrome, SBIs allowed researchers to assume the novel mechanism in these conditions - autoimmune autonomic nervous system imbalance. In this review, all data published on an imbalance of autoantibodies against GPCR, clinical symptoms and pathogenic mechanisms in CFS, Fibromyalgia, SBIs, COVID and PCS, SBS, POTS, and some autoimmune diseases were analyzed. Possible criteria to diagnose the autoimmune autonomic nervous system imbalance were created.
Collapse
|
8
|
Malkova AM, Shoenfeld Y. WITHDRAWN: Autoimmune autonomic nervous system imbalance and conditions: Chronic fatigue syndrome, fibromyalgia, silicone breast implants, COVID and post-COVID syndrome, sick building syndrome, post-orthostatic tachycardia syndrome, autoimmune diseases and autoimmune/inflammatory syndrome induced by adjuvants. Autoimmun Rev 2022:103231. [PMID: 36356798 DOI: 10.1016/j.autrev.2022.103231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, https://doi.org/10.1016/j.autrev.2022.103230. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- A M Malkova
- Zabludowicz Center of autoimmunity, Sheba Medical Center, Tel Hashomer, Israel.
| | - Y Shoenfeld
- Zabludowicz Center of autoimmunity, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
9
|
Macrophage polarization is involved in liver fibrosis induced by β 1-adrenoceptor autoantibody. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1100-1112. [PMID: 35983976 PMCID: PMC9828683 DOI: 10.3724/abbs.2022102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Accumulating evidence suggests that liver injury can be induced by the over-expression of β 1-adrenergic receptors (β 1-ARs). High titers of autoantibodies specific to β 1-adrenergic receptors (β 1-AA) are detected in the sera of heart failure patients, potentially playing agonist-like roles. However, the role of β 1-AA in liver function has not been characterized. In this study, we collect the sera of primary biliary cholangitis (PBC) patients, a condition which easily develops into liver fibrosis, and analyze the relationship between PBC and β 1-AA. A passive immunization model is established to assess the effect of β 1-AA on the liver. Subsequently, the effect of β 1-AA on macrophages is investigated in vitro. Results show that PBC patients have a high titer and ratio of β 1-AA, compared to controls. Liver injury and fibrosis are induced by β 1-AA. In vitro experiments with ROS probe demonstrate that β 1-AA induces macrophages to produce ROS and secrete TNFα. These effects can be partially reversed by metoprolol, a blocker for β 1-AR. Results from the transwell and phagocytosis assays show that β 1-AA promotes macrophage migration and phagocytosis. FCM tests suggest that β 1-AA induces the alteration of M1 rather than M2 markers in macrophages. Finally, the Annexin V/PI assay indicates that macrophage culture supernatants stimulated by β 1-AA cause hepatocyte apoptosis. Overall, these results suggest that β 1-AA is involved in PBC. The β 1-AA-induced activation, phagocytosis and phenotypic modification of macrophages may play an important role in the development of hepatic fibrosis and injury.
Collapse
|
10
|
Devi ML, Thorat SS, Devi KK, Sharma KC, Singh YD, Mishra A, Das S. Internal Transcribed Spacer (ITS) Region of Nuclear Ribosomal DNA as a Suitable DNA Barcode for Identification of Zanthoxylum armatum DC. from Manipur. Mol Biotechnol 2022; 64:1454-1467. [PMID: 35763198 DOI: 10.1007/s12033-022-00518-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 05/20/2022] [Indexed: 10/17/2022]
Abstract
Zanthoxylum armatum DC. is a plant with many medicinal values which is extensively used in traditional system of medicine for curing various diseases and ailments, including cancer. The aim of the present study is to identify Zanthoxylum armatum collected from different parts of Manipur, India, at molecular level. Molecular markers like internal transcribed spacer (ITS) region and other DNA barcoding genes such as matK, rbcL, psbA-trnH and trnL-trnF were targeted to find out the most suitable DNA barcode for identifying this species. Sequences obtained using the five primer pairs-ITS An5 and ITS An4, matK-413f-1 and matK-1227r-1, rbcL-1F and rbcL-724R, psbA-F and trnH-R and trnL-F and trnF-R were submitted to GenBank, NCBI. Amongst the five DNA barcoding targets, one nuclear and four chloroplast genes were successfully amplified by PCR (100%) and sequencing (100%) in all the eight plant samples. Sequence similarity of total ITS region (620 bp) when compared to the reference sequence were found to be between 98.55 and 99.68%. In our study, ITS sequence in combination with DNA barcoding sequences of rbcL, trnH-psbA and trnL-trnF was very successful in identification of Z. armatum and differentiate other species clearly in the phylogeny analysis. Our work shows ITS region to be the most suitable DNA barcode which formed a monophyletic group of the species in the phylogenetic tree analysis. The sequences of the barcoding genes of Z. armatum DC. obtained from this study adds to the already available resources which will be helpful in the future research endeavours.
Collapse
Affiliation(s)
- Moirangthem Lakshmipriyari Devi
- Plant Molecular Genetics and Genomics Laboratory, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, 795001, India.,School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha, 751024, India
| | - Sunil S Thorat
- Bioresources Database and Bioinformatics, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, 795001, India
| | - Khomdram Khedashwori Devi
- Plant Molecular Genetics and Genomics Laboratory, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, 795001, India
| | | | - Yumnam Dhaneshwor Singh
- Plant Molecular Genetics and Genomics Laboratory, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, 795001, India
| | - Amrita Mishra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha, 751024, India
| | - Sudripta Das
- Plant Molecular Genetics and Genomics Laboratory, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, 795001, India.
| |
Collapse
|
11
|
Tang Q, Cen Z, Lu J, Dong J, Qin L, Lu F, Wu W. The abnormal distribution of peripheral B1 cells and transition B cells in patients with idiopathic dilated cardiomyopathy: a pilot study. BMC Cardiovasc Disord 2022; 22:78. [PMID: 35246021 PMCID: PMC8895850 DOI: 10.1186/s12872-022-02461-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background The aberrant distribution of peripheral B cell subsets is associated with the pathogenesis of a variety of inflammatory and autoimmune diseases. However, the distribution of peripheral B cell subsets in patients with idiopathic dilated cardiomyopathy (DCM) remains to be elucidated.
Methods Twenty-seven patients with idiopathic DCM (DCM group), 18 control patients with heart failure (HF group) and 21 healthy individuals (HC group) were included in this study. Peripheral B cell subsets were analysed using multicolour flow cytometry. The plasma β1 adrenergic receptor (β1-AR) autoantibody titre was determined using ELISA. Additionally, clinical features were also collected. Results Compared with the HF and HC groups, the percentage of B1 cells was significantly decreased, whereas the percentage of transitional B cells (Tr) was significantly increased in the DCM group. Notably, the percentage of B1 cells was significantly lower in patients with β1-AR autoantibody-positive DCM than in β1-AR autoantibody-negative patients. The correlation analysis showed that the percentage of B1 cells was negatively correlated with N-terminal pro-brain natriuretic peptide (NT-proBNP) levels and positively correlated with the left ventricular ejection fraction in patients with DCM. Conclusion As shown in the present study, the percentage of B1 cells in the peripheral blood of patients with idiopathic DCM is abnormally decreased, especially in β1-AR autoantibody-positive patients, while the percentage of Tr cells is significantly increased, indicating that B1 cells and Tr cells may be implicated in the pathogenesis of idiopathic DCM. The decrease in the percentage of B1 cells is directly related to the severity of DCM. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02461-8.
Collapse
Affiliation(s)
- Quan Tang
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Shuangyong Road 6, Nanning, 530021, Guangxi, People's Republic of China
| | - Zhihong Cen
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Shuangyong Road 6, Nanning, 530021, Guangxi, People's Republic of China
| | - Jing Lu
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Shuangyong Road 6, Nanning, 530021, Guangxi, People's Republic of China
| | - Jingwei Dong
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Shuangyong Road 6, Nanning, 530021, Guangxi, People's Republic of China
| | - Lin Qin
- Department of Cardiology, Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, People's Republic of China
| | - Feiyu Lu
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Shuangyong Road 6, Nanning, 530021, Guangxi, People's Republic of China
| | - Weifeng Wu
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Shuangyong Road 6, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
12
|
Xie X, Ji J, Chen X, Xu W, Chen H, Zhu S, Wu J, Wu Y, Sun Y, Sai W, Liu Z, Xiao M, Bao B. Human umbilical cord mesenchymal stem cell-derived exosomes carrying hsa-miRNA-128-3p suppress pancreatic ductal cell carcinoma by inhibiting Galectin-3. Clin Transl Oncol 2022; 24:517-531. [PMID: 34811696 DOI: 10.1007/s12094-021-02705-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/31/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal malignant tumors of the digestive system. Many patients are diagnosed at an advanced stage and lose eligibility for surgery. Moreover, there are few effective methods for treating pancreatic ductal cell carcinoma. Increasing attention has been given to microRNAs (miRNAs) and their regulatory roles in tumor progression. In this study, we investigated the effects of exosomes extracted from human umbilical cord mesenchymal stem cells (HUCMSCs) carrying hsa-miRNA-128-3p on pancreatic cancer cells. METHODS Based on existing experimental and database information, we selected Galectin-3, which is associated with pancreatic cancer, and the corresponding upstream hsa-miRNA-128-3p. We extracted HUCMSCs from a fresh umbilical cord, hsa-miRNA-128-3p was transfected into HUCMSCs, and exosomes containing hsa-miRNA-128-3p were extracted and collected. The effect of exosomes rich in hsa-miRNA-128-3p on pancreatic cancer cells was analyzed. RESULTS The expression of Galectin-3 in normal pancreatic duct epithelial cells was significantly lower than that in PDAC cell lines. We successfully extracted HUCMSCs from the umbilical cord and transfected hsa-miRNA-128-3p into HUCMSCs. Then we demonstrated that HUCMSC-derived exosomes with hsa-miRNA-128-3p could suppress the proliferation, invasion, and migration of PANC-1 cells in vitro by targeting Galectin-3. CONCLUSION Hsa-miRNA-128-3p could be considered as a potential therapy for pancreatic cancer. We provided a new idea for targeted therapy of PDAC.
Collapse
Affiliation(s)
- X Xie
- Department of Gastroenterology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - J Ji
- Department of Gastroenterology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - X Chen
- Office of Infection Management, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - W Xu
- Department of Gastroenterology, Second People's Hospital of Nantong, Nantong, 226001, China
| | - H Chen
- Office of Infection Management, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - S Zhu
- Medical School of Nantong University Oral Medicine, Nantong, 226001, Jiangsu, China
| | - J Wu
- Department of Gastroenterology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Y Wu
- Department of Gastroenterology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Y Sun
- Blood Center of Jiangsu Province, Nanjing, 210000, Jiangsu, China
| | - W Sai
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Z Liu
- Department of Gastroenterology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - M Xiao
- Department of Gastroenterology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - B Bao
- Department of Gastroenterology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
13
|
Sun C, Lu J, Long Y, Guo S, Jia W, Ning N, Hao H, Wang X, Bian Y, Liu H, Wang L. Adiponectin up-regulates the decrease of myocardial autophagic flux induced by β 1 -adrenergic receptor autoantibody partly dependent on AMPK. J Cell Mol Med 2021; 25:8464-8478. [PMID: 34322993 PMCID: PMC8419161 DOI: 10.1111/jcmm.16807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocytes autophagy is essential for maintaining cardiac function. Our previous studies have found that β1‐adrenergic receptor autoantibody (β1‐AA) induced the decreased myocardial autophagic flux, which resulted in cardiomyocyte death and cardiac dysfunction. And other studies demonstrated that β1‐AA induced the decrease of AMPK phosphorylation, the key hub of autophagy pathway, while adiponectin up‐regulated autophagic flux mediated by AMPK. However, it is not clear whether adiponectin improves the inhibition of myocardial autophagic flux induced by β1‐AA by up‐regulating the level of AMPK phosphorylation. In this study, it has been confirmed that β1‐AA induced the decrease of AMPK phosphorylation level in both vivo and vitro. Moreover, pretreatment of cardiomyocytes with AMPK inhibitor Compound C could further reduce the autophagic flux induced by β1‐AA. Adiponectin deficiency could aggravate the decrease of myocardial AMPK phosphorylation level, autophagic flux and cardiac function induced by β1‐AA. Further, exogenous adiponectin could reverse the decline of AMPK phosphorylation level and autophagic flux induced by β1‐AA and even reduce cardiomyocyte death. While pretreated with the Compound C, the adiponectin treatment did not improve the decreased autophagosome formation, but still improved the decreased autophagosome clearance induced by β1‐AA in cardiomyocytes. This study is the first time to confirm that β1‐AA could inhibit myocardial autophagic flux by down‐regulating AMPK phosphorylation level. Adiponectin could improve the inhibition of myocardial autophagic flux induced by β1‐AA partly dependent on AMPK, so as to provide an experimental basis for the treatment of patients with β1‐AA‐positive cardiac dysfunction.
Collapse
Affiliation(s)
- Cong Sun
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China.,Department of Pathology, Linfen Central Hospital, Linfen, China
| | - Jiebei Lu
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Yaolin Long
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Shuai Guo
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Weiwei Jia
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Na Ning
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Haihu Hao
- Department of Orthopedics, Shanxi Bethune Hospital & Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiaohui Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| | - Yunfei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| |
Collapse
|
14
|
Expression stability of internal reference gene in response to Trichoderma polysporum infection in Avena fatua L. Curr Genet 2021; 67:909-918. [PMID: 34287660 DOI: 10.1007/s00294-021-01200-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
Trichoderma polysporum was a pathogenic fungi which showed strong pathogenicity to Avena fatua L. in recent study. The stress response of A. fatua to T. polysporum is mediated by the regulation of gene expression. Quantification of the expression of genes requires normalizing RT-qPCR data using reference genes with stable expression in the system studied as internal standards. To construct a RT-qPCR system suitable for response of A. fatua to T. polysporum, and screen stable internal reference genes, GeNorm, NormFinder, BestKeeper and RefFinde were used to perform SYBR Green-based RT-qPCR analysis on eight candidate internal reference genes (18S, 28S, TUA, UBC, ACT, GAPDH, TBP and EF-1α) in A. fatua samples after inoculation of T. polysporum Strain HZ-31. The results showed that TBP, 18S and UBC were the most stable internal reference genes, TBP and TUA, TBP and GAPDH, 18S and TBP, UBC and 18S were the most suitable combination of the two internal reference genes, which could be used as internal reference genes for functional gene expression analysis during the interaction between T. polysporum and A. fatua. This is the first study describing a set of reference genes with a stable expression under fungi stress in A. fatua. These genes are also candidate reference genes of choice for studies seeking to identify stress-responsive genes in A. fatua.
Collapse
|
15
|
Sex Differences, Genetic and Environmental Influences on Dilated Cardiomyopathy. J Clin Med 2021; 10:jcm10112289. [PMID: 34070351 PMCID: PMC8197492 DOI: 10.3390/jcm10112289] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/11/2021] [Accepted: 05/18/2021] [Indexed: 12/15/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is characterized by dilatation of the left ventricle and impaired systolic function and is the second most common cause of heart failure after coronary heart disease. The etiology of DCM is diverse including genetic pathogenic variants, infection, inflammation, autoimmune diseases, exposure to chemicals/toxins as well as endocrine and neuromuscular causes. DCM is inherited in 20–50% of cases where more than 30 genes have been implicated in the development of DCM with pathogenic variants in TTN (Titin) most frequently associated with disease. Even though male sex is a risk factor for heart failure, few studies have examined sex differences in the pathogenesis of DCM. We searched the literature for studies examining idiopathic or familial/genetic DCM that reported data by sex in order to determine the sex ratio of disease. We found 31 studies that reported data by sex for non-genetic DCM with an average overall sex ratio of 2.5:1 male to female and 7 studies for familial/genetic DCM with an overall average sex ratio of 1.7:1 male to female. No manuscripts that we found had more females than males in their studies. We describe basic and clinical research findings that may explain the increase in DCM in males over females based on sex differences in basic physiology and the immune and fibrotic response to damage caused by mutations, infections, chemotherapy agents and autoimmune responses.
Collapse
|
16
|
Jafari A, Babajani A, Abdollahpour-Alitappeh M, Ahmadi N, Rezaei-Tavirani M. Exosomes and cancer: from molecular mechanisms to clinical applications. Med Oncol 2021; 38:45. [PMID: 33743101 DOI: 10.1007/s12032-021-01491-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/04/2021] [Indexed: 12/15/2022]
Abstract
Exosomes are extracellular nanovesicles secreted from almost all types of normal and cancer cells. Collective evidence suggests that exosomes participate in cell-cell communication via transmitting their cargo, including nucleic acids, proteins, and metabolites to recipient cells. Tumor-derived exosomes (TEXs) play prominent roles in the regulation of molecular pathways in malignancies. Internalization of exosomes by tumor cells affects cellular pathways and several cancer hallmarks, including reprogramming of stromal cells, modulating immune responses, reconstructing extracellular matrix architecture, or even endowing tumor cells with drug features resistance. The unique biogenesis pathways of exosomes, their composition, low immunogenicity, and nontoxicity, together with their ability to target tumor cells, bring them up as an attractive vesicles for cancer therapy. Thus, understanding the molecular mechanisms of exosomes' participation in tumorigenesis will be critical for the next generation of cancer therapeutics. This review aims to summarize the exosomes' roles in different mechanisms underlying cancer progression for the rational design of tailored strategies against this illness. The present study also highlights the new findings on using these smart vesicles as therapeutic targets and potential biomarkers. Recent advances in exosome biology will open up new, more effective, less invasive, and more individualized clinical applications for treating cancer patients.
Collapse
Affiliation(s)
- Ameneh Jafari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nayebali Ahmadi
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Proteomics Research Center, Department of Medical Lab Technology, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Tsilika M, Antonakos N, Gkavogianni T, Karageorgos A, Kyriazopoulou E, Netea MG, Giamarellos-Bourboulis EJ. Impact of comorbidities on the performance of interferon-gamma release assay in an elderly Greek population without overt immunodeficiency. Int J Antimicrob Agents 2020; 56:106073. [PMID: 32629117 DOI: 10.1016/j.ijantimicag.2020.106073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/09/2020] [Accepted: 06/28/2020] [Indexed: 12/13/2022]
Abstract
Background The prevalence of latent tuberculosis infection (LTBI) increases with age. Interferon-gamma release assay (IGRA) is a T-cell based assay widely used for the detection of LTBI. Objectives To identify the prevalence of LTBI among an elderly Greek population using IGRA and to evaluate comorbidities associated with LTBI. Methods Individuals aged at least 65 years who were non-immunocompromised and had no history of active tuberculosis infection (TBI) underwent IGRA to identify LTBI. Participant characteristics were compared between the LTBI and non-LTBI groups. Interferon-gamma (INFγ) levels were analysed in each group. Results A total of 130 (38.7%) participants with LTBI and 206 (61.3%) participants without LTBI were included. Multivariate logistic regression analysis identified the following features that were independently associated with a positive IGRA result: female sex (odds ratio [OR]: 0.45; 95% confidence interval [CI]: 0.28-0.72; P=0.001), chronic heart failure (OR: 0.41; 95% CI: 0.22-0.77; P=0.005), history of major surgery (OR: 0.55; 95% CI: 0.33-0.92; P=0.022) and Charlson Comorbidity Index >3 (OR: 3.06; 95% CI: 1.46-6.40; P=0.003). Production of stimulated INFγ was significantly lower in the non-LTBI group. Conclusions Female sex, history of chronic heart failure and history of any surgical intervention were independently associated with a negative IGRA result, and CCI >3 was associated with a positive IGRA result. These results indicate careful interpretation of IGRA is required among elderly individuals with these characteristics.
Collapse
Affiliation(s)
- Maria Tsilika
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Greece
| | - Nikolaos Antonakos
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Greece
| | - Theologia Gkavogianni
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Greece
| | - Athanassios Karageorgos
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Greece
| | - Evdoxia Kyriazopoulou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Greece
| | - Mihai G Netea
- Department of Internal Medicine and Infectious Diseases, Radboud University Nijmegen, The Netherlands
| | | |
Collapse
|
18
|
Robles-Vera I, Toral M, de la Visitación N, Aguilera-Sánchez N, Redondo JM, Duarte J. Protective Effects of Short-Chain Fatty Acids on Endothelial Dysfunction Induced by Angiotensin II. Front Physiol 2020; 11:277. [PMID: 32372967 PMCID: PMC7176911 DOI: 10.3389/fphys.2020.00277] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/12/2020] [Indexed: 02/03/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are among the main classes of bacterial metabolic products and are mainly synthesized in the colon through bacterial fermentation. Short-chain fatty acids, such as acetate, butyrate, and propionate, reduce endothelial activation induced by proinflammatory mediators, at least in part, by activation of G protein–coupled receptors (GPRs): GPR41 and GPR43. The objective of the study was to analyze the possible protective effects of SCFAs on endothelial dysfunction induced by angiotensin II (AngII). Rat aortic endothelial cells (RAECs) and rat aortas were incubated with AngII (1 μM) for 6 h in the presence or absence of SCFAs (5–10 mM). In RAECs, we found that AngII reduces the production of nitric oxide (NO) stimulated by calcium ionophore A23187; increases the production of reactive oxygen species (ROS), both from the nicotinamide adenine dinucleotide phosphate oxidase system and the mitochondria; diminishes vasodilator-stimulated phosphoprotein (VASP) phosphorylation at Ser239; reduces GPR41 and GPR43 mRNA level; and reduces the endothelium-dependent relaxant response to acetylcholine in aorta. Coincubation with butyrate and acetate, but not with propionate, increases both NO production and pSer239-VASP, reduces the concentration of intracellular ROS, and improves relaxation to acetylcholine. The beneficial effects of butyrate were inhibited by the GPR41 receptor antagonist, β-hydroxybutyrate, and by the GPR43 receptor antagonist, GLPG0794. Butyrate inhibited the down-regulation of GPR41 and GPR43 induced by AngII, being without effect acetate and propionate. Neither β-hydroxybutyrate nor GLPG0794 affects the protective effect of acetate in endothelial dysfunction. In conclusion, acetate and butyrate improve endothelial dysfunction induced by AngII by increasing the bioavailability of NO. The effect of butyrate seems to be related to GPR41/43 activation, whereas acetate effects were independent of GPR41/43.
Collapse
Affiliation(s)
- Iñaki Robles-Vera
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Marta Toral
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Nazaret Aguilera-Sánchez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain.,CIBERCV, Madrid, Spain.,Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| |
Collapse
|
19
|
Du Y, Zhang S, Yu H, Wu Y, Cao N, Wang W, Xu W, Li Y, Liu H. Autoantibodies Against β 1-Adrenoceptor Exaggerated Ventricular Remodeling by Inhibiting CTRP9 Expression. J Am Heart Assoc 2020; 8:e010475. [PMID: 30764693 PMCID: PMC6405676 DOI: 10.1161/jaha.118.010475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Autoantibodies against the second extracellular loop of the β1‐adrenoceptor (β1‐AA) act similarly to agonist of β1‐adrenergic receptor, which plays an important role in the pathophysiological characteristics of ventricular remodeling. Recently, considerable lines of evidence have suggested that CTRP9 (C1q tumor necrosis factor–related protein 9) is a potent cardioprotective cardiokine and protects the heart from ventricular remodeling. The aim of this study was to determine the role of CTRP9 in ventricular remodeling induced by β1‐AA. Methods and Results Blood samples were collected from 131 patients with coronary heart disease and 131 healthy subjects. The serum levels of β1‐AA and CTRP9 were detected using ELISA. The results revealed that CTRP9 levels in β1‐AA–positive patients were lower than those in β1‐AA–negative patients, and serum CTRP9 concentrations were inversely correlated with β1‐AA. β1‐AA monoclonal antibodies (β1‐AAmAbs) were administered in mice with and without rAAV9‐cTnT‐Full Ctrp9‐FLAG virus for 8 weeks. Reverse transcription–polymerase chain reaction/Western analysis showed that cardiomyocyte CTRP9 expression was significantly reduced in β1‐AAmAb–treated mice. Moreover, compared with the β1‐AAmAb alone group, cardiac‐specific CTRP9 overexpression improved cardiac function, attenuated adverse remodeling, and ameliorated cardiomyocyte apoptosis and fibrosis. Mechanistic studies demonstrated that CTRP9 overexpression decreased the levels of G‐protein–coupled receptor kinase 2 and promoted the activation of AMP‐dependent kinase pathway. However, cardiac‐specific overexpression of CTRP9 had no effect on the levels of cAMP and protein kinase A activity elevated by β1‐AAmAb. Conclusions This study provides the first evidence that the long‐term existence of β1‐AAmAb suppresses cardiac CTRP9 expression and exaggerates cardiac remodeling, suggesting that CTRP9 may be a novel therapeutic target against pathologic remodeling in β1‐AA–positive patients with coronary heart disease.
Collapse
Affiliation(s)
- Yunhui Du
- 1 Beijing Anzhen Hospital Capital Medical University Beijing Institute of Heart, Lung and Blood Vessel Diseases Beijing China
| | - Shihan Zhang
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Haicun Yu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China.,4 Zhengzhou Central Hospital affiliated of Zhengzhou University Henan Province China
| | - Ye Wu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Ning Cao
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Wen Wang
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Wenli Xu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Yuming Li
- 3 Department of Basic Medical Sciences Yanjing Medical College Capital Medical University Beijing China
| | - Huirong Liu
- 2 Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| |
Collapse
|
20
|
Wang X, Han M, He S, Zhang Y, Xu X, Wang Y, Dang C, Zhang J, Wang H, Chen M, Liu J, Hou D, Zhao W, Xu L, Zhang L. Diagnostic and prognostic value of autoantibodies against β 1-adrenoreceptors in patients with heart failure following acute myocardial infarction: A 5-year prospective study. Exp Ther Med 2020; 19:1259-1266. [PMID: 32010297 PMCID: PMC6966159 DOI: 10.3892/etm.2019.8331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/15/2019] [Indexed: 11/18/2022] Open
Abstract
A number of studies have suggested that autoantibodies against β1-adrenoreceptors (β1R-AAbs) have an important role in pathophysiological processes of heart failure. The aim of the present study was to determine whether β1R-AAbs are implicated in cardiac dysfunction following acute myocardial infarction (AMI) and their association with prognosis. A total of 33 cases with systolic heart failure (SHF), 49 with diastolic heart failure (DHF) and 44 with normal heart function following AMI were recruited. β1R-AAbs were detected by ELISA and major adverse cardiac events (MACEs) were recorded during the 5-year follow-up. The positive rate of β1R-AAbs in the SHF group (45.5%) was significantly higher compared with that in the DHF (22.4%; P<0.05) and normal (15.9%; P<0.05) groups. The area under the receiver operating characteristics curve for the diagnosis of SHF was 0.630 (95% CI: 0.514–0.747, P=0.026). During a median follow-up period of 51.0±15.4 months, the positive rate of β1R-AAbs in the MACEs group was significantly higher compared with that in the non-MACEs group (P<0.05). Multivariate logistic regression analysis indicated that the left ventricular ejection fraction and diabetes were independent predictors of 5-year MACEs following AMI, whereas β1R-AAbs were not. Kaplan-Meier analysis revealed that the cumulative MACEs-free survival rate was the lowest in the SHF group, followed by the DHF and normal groups (P<0.05). Therefore, β1R-AAbs were indicated to be of value for early diagnosis of SHF after AMI but not as independent predictors for the prognosis of patients with AMI.
Collapse
Affiliation(s)
- Xin Wang
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Mengmeng Han
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Shan He
- Heart Center, Beijing Chaoyang Hospital Jingxi Branch, Capital Medical University, Beijing 100020, P.R. China
| | - Yuan Zhang
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiaorong Xu
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Yuxing Wang
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Caijing Dang
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Juan Zhang
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Hua Wang
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Mulei Chen
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Jiamei Liu
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Dongyan Hou
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Wenshu Zhao
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Lin Xu
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Lin Zhang
- Heart Center and Beijing Key Laboratory of Hypertension Research, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
21
|
Cabral-Marques O, Schimke LF, de Oliveira EB, El Khawanky N, Ramos RN, Al-Ramadi BK, Segundo GRS, Ochs HD, Condino-Neto A. Flow Cytometry Contributions for the Diagnosis and Immunopathological Characterization of Primary Immunodeficiency Diseases With Immune Dysregulation. Front Immunol 2019; 10:2742. [PMID: 31849949 PMCID: PMC6889851 DOI: 10.3389/fimmu.2019.02742] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Almost 70 years after establishing the concept of primary immunodeficiency disorders (PIDs), more than 320 monogenic inborn errors of immunity have been identified thanks to the remarkable contribution of high-throughput genetic screening in the last decade. Approximately 40 of these PIDs present with autoimmune or auto-inflammatory symptoms as the primary clinical manifestation instead of infections. These PIDs are now recognized as diseases of immune dysregulation. Loss-of function mutations in genes such as FOXP3, CD25, LRBA, IL-10, IL10RA, and IL10RB, as well as heterozygous gain-of-function mutations in JAK1 and STAT3 have been reported as causative of these disorders. Identifying these syndromes has considerably contributed to expanding our knowledge on the mechanisms of immune regulation and tolerance. Although whole exome and whole genome sequencing have been extremely useful in identifying novel causative genes underlying new phenotypes, these approaches are time-consuming and expensive. Patients with monogenic syndromes associated with autoimmunity require faster diagnostic tools to delineate therapeutic strategies and avoid organ damage. Since these PIDs present with severe life-threatening phenotypes, the need for a precise diagnosis in order to initiate appropriate patient management is necessary. More traditional approaches such as flow cytometry are therefore a valid option. Here, we review the application of flow cytometry and discuss the relevance of this powerful technique in diagnosing patients with PIDs presenting with immune dysregulation. In addition, flow cytometry represents a fast, robust, and sensitive approach that efficiently uncovers new immunopathological mechanisms underlying monogenic PIDs.
Collapse
Affiliation(s)
- Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Nadia El Khawanky
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg im Breisgau, Germany.,Precision Medicine Theme, The South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Rodrigo Nalio Ramos
- INSERM U932, SiRIC Translational Immunotherapy Team, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Basel K Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | | | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, United States
| | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
22
|
Xu W, Wu Y, Wang L, Bai Y, Du Y, Li Y, Cao N, Zhao Y, Zhang Y, Liu H. Autoantibody against β 1-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4 + T cells by up-regulating AMPK-mediated fatty acid oxidation. Cell Death Dis 2019; 10:158. [PMID: 30770790 PMCID: PMC6377640 DOI: 10.1038/s41419-018-1209-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022]
Abstract
Therapeutic adoptive transfer of natural regulatory T cells (nTreg, CD4+ CD25+ Foxp3+ T cells) or in vivo selective expansion of nTreg cells has been demonstrated to improve the cardiac function in various cardiovascular disease models. The differentiation of nTreg cells is mediated by catecholamines via β1-adrenergic receptor (β1-AR) activation. Autoantibody against β1-adrenoceptor (β1-AA) as a β1-AR agonist is closely associated with the occurrence and deterioration of cardiac dysfunction. However, whether β1-AA has any impact on nTreg cells has not been reported. The aim of the present study was intended to assess the potential impact of β1-AA on nTreg cell differentiation and explore the underlying mechanism. It was found that the expression of multiple proteins involved in nTreg cell differentiation, immunosuppressive function, and migration was up-regulated in mice after β1-AA administration, suggesting that β1-AA may promote nTreg cell activation. In vitro, β1-AA promoted nTreg cell differentiation by up-regulating mitochondrial fatty acid oxidation (FAO) in activated CD4+ T cells via AMP-activated protein kinase (AMPK) activation and mitochondrial membrane potential reduction. In addition, the AMPK agonist facilitated β1-AA-mediated FAO and nTreg cell differentiation. To further confirm the role of AMPK in β1-AA-mediated nTreg cell differentiation, β1-AA was acted on the CD4+ T cells isolated from AMPK-deficient (AMPK−/−) mice. The result showed that the effect of β1-AA on nTreg cell differentiation was attenuated markedly after AMPK knockout. In conclusion, AMPK-mediated metabolic regulation targeting for nTreg cell restoration may be a promising therapeutic target for β1-AA-positive patients with cardiac dysfunction.
Collapse
Affiliation(s)
- Wenli Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Li Wang
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, 030001, Taiyuan, China
| | - Yan Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Yunhui Du
- Beijing Anzhen Hospital, Capital Medical University, 100029, Beijing, China
| | - Yang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Ning Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Yuhui Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, 100191, Beijing, China.,Beijing Key Laboratory of Cardiovascular Receptors Research, 100191, Beijing, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 100069, Beijing, China. .,Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, 100069, Beijing, China.
| |
Collapse
|
23
|
Activation of T Lymphocytes as a Novel Mechanism in Beta1-Adrenergic Receptor Autoantibody-Induced Cardiac Remodeling. Cardiovasc Drugs Ther 2019; 33:149-161. [DOI: 10.1007/s10557-019-06856-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
24
|
Du Y, Zhang S, Hao W, Xu W, Yan L, Liu H. Pro-remodeling effect of autoantibody against β 1-adrenoceptor on cardiomyocytes involves T cells dysfunction under the pathological condition of heart failure. Biochem Biophys Res Commun 2019; 510:163-170. [PMID: 30678811 DOI: 10.1016/j.bbrc.2019.01.072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 11/24/2022]
Abstract
Autoantibody against β1-adrenoceptor (β1-AA) has been shown to be closely linked to the aggravation of heart failure. Removal of β1-AA remarkably attenuated patients' cardiac dysfunction. We found that β1-AA induced rat heart failure with increased CD4+ T cells. However, whether or not β1-AA interacts with T cells isolated from heart failure patients remains unknown. Twenty-one β1-AA-negative heart failure patients were divided into those taking β-adrenergic blocker and those not. The effects of β1-AA monoclonal antibodies (β1-AAmAb) on T cells proliferation were detected using the CCK-8 assay. IFN-γ and IL-4 production by human T cells were measured by after the administration of β1-AAmAb. The levels of cardiomyocyte apoptosis and hypertrophy were detected after co-cultured with the supernatant of T cells pre-stimulated by β1-AAmAb. It was found that β1-AAmAb promoted T cell proliferation via the β1-AR/cAMP/PKA pathway in patients who not take β-blocker. β1-AAmAb inhibited the characteristic cytokine secretion of Th1, IFN-γ, but had no significant effect on the Th2 cytokine IL-4. Supernatant resulted from the T cells pre-treated with β1-AAmAb induced cardiomyocytes remodeling, as evidenced by increased levels of cardiomyocytes apoptosis and hypertrophy. We propose that heart failure is likely to be an interference factor for Th-mediated immunity, and the presence of β1-AAmAb may aggravate this effect and deteriorate concomitant inflammatory injury in cardiomyocytes, partially via β1-AR/cAMP/PKA pathway.
Collapse
Affiliation(s)
- Yunhui Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Shihan Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wenjing Hao
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Wenli Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Li Yan
- Department of Pathophysiology, Institute of Basic Medical Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
25
|
Dong Y, Bai Y, Zhang S, Xu W, Xu J, Zhou Y, Zhang S, Wu Y, Yu H, Cao N, Liu H, Wang W. Cyclic peptide RD808 reduces myocardial injury induced by β 1-adrenoreceptor autoantibodies. Heart Vessels 2018; 34:1040-1051. [PMID: 30554265 DOI: 10.1007/s00380-018-1321-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 12/07/2018] [Indexed: 10/27/2022]
Abstract
Autoantibodies against the second extracellular loop of β1-adrenergic receptor (β1-AA) have been shown to be involved in the development of cardiovascular diseases. Recently, there has been considerable interest in strategies to remove these autoantibodies, particularly therapeutic peptides to neutralize β1-AA. Researchers are investigating the roles of cyclic peptides that mimic the structure of relevant epitopes on the β1-AR-ECII in a number of immune-mediated diseases. Here, we used a cyclic peptide, namely, RD808, to neutralize β1-AA, consequently alleviating β1-AA-induced myocardial injury. We investigated the protective effects of RD808 on the myocardium both in vitro and in vivo. RD808 was found to increase the survival rate of cardiomyocytes; furthermore, it decreased myocardial necrosis and apoptosis and improved the cardiac function of BalB/c mice in a β1-AA transfer model. In vitro and in vivo experiments showed that myocardial autophagy was increased in the presence of RD808, which might contribute to its cardioprotective effects. Our findings indicate that RD808 reduced myocardial injury induced by β1-AA.
Collapse
Affiliation(s)
- Yu Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Yan Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Shangyue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Wenli Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Jiahui Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Yi Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Ye Wu
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Haicun Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Ning Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China. .,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China.
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China. .,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Saad NS, Elnakish MT, Ahmed AAE, Janssen PML. Protein Kinase A as a Promising Target for Heart Failure Drug Development. Arch Med Res 2018; 49:530-537. [PMID: 30642654 PMCID: PMC6451668 DOI: 10.1016/j.arcmed.2018.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/13/2018] [Indexed: 12/24/2022]
Abstract
Heart failure (HF) is a clinical syndrome characterized by impaired ability of the heart to fill or eject blood. HF is rather prevalent and it represents the foremost reason of hospitalization in the United States. The costs linked to HF overrun those of all other causes of disabilities, and death in the United States and all over the developed as well as the developing countries which amplify the supreme significance of its prevention. Protein kinase (PK) A plays multiple roles in heart functions including, contraction, metabolism, ion fluxes, and gene transcription. Altered PKA activity is likely to cause the progression to cardiomyopathy and HF. Thus, this review is intended to focus on the roles of PKA and PKA-mediated signal transduction in the healthy heart as well as during the development of HF. Furthermore, the impact of cardiac PKA inhibition/activation will be highlighted to identify PKA as a potential target for the HF drug development.
Collapse
Affiliation(s)
- Nancy S Saad
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohammad T Elnakish
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Amany A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
27
|
Simpson RJ, Bigley AB, Agha N, Hanley PJ, Bollard CM. Mobilizing Immune Cells With Exercise for Cancer Immunotherapy. Exerc Sport Sci Rev 2017; 45:163-172. [PMID: 28418996 DOI: 10.1249/jes.0000000000000114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hematopoietic stem cell (HSC) transplantation and adoptive transfer immunotherapy are effective in treating blood cancers and posttransplant infections, but low-circulating cell numbers in patients and donors are oftentimes a limiting factor. We postulate that a single exercise bout will increase the yield of patient- and donor-derived HSCs and cytotoxic lymphocytes to improve this form of treatment for cancer patients.
Collapse
Affiliation(s)
- Richard J Simpson
- 1Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston; 2Department of Behavioral Sciences, University of Texas MD Anderson Cancer Center, Houston, TX; and 3Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Health System and The George Washington University, Washington, DC
| | | | | | | | | |
Collapse
|
28
|
Functional autoantibodies targeting G protein-coupled receptors in rheumatic diseases. Nat Rev Rheumatol 2017; 13:648-656. [PMID: 28855694 DOI: 10.1038/nrrheum.2017.134] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
G protein-coupled receptors (GPCRs) comprise the largest and most diverse family of integral membrane proteins that participate in different physiological processes such as the regulation of the nervous and immune systems. Besides the endogenous ligands of GPCRs, functional autoantibodies are also able to bind GPCRs to trigger or block intracellular signalling pathways, resulting in agonistic or antagonistic effects, respectively. In this Review, the effects of functional GPCR-targeting autoantibodies on the pathogenesis of autoimmune diseases, including rheumatic diseases, are discussed. Autoantibodies targeting β1 and β2 adrenergic receptors, which are expressed by cardiac and airway smooth muscle cells, respectively, have an important role in the development of asthma and cardiovascular diseases. In addition, high levels of autoantibodies against the muscarinic acetylcholine receptor M3 as well as those targeting endothelin receptor type A and type 1 angiotensin II receptor have several implications in the pathogenesis of rheumatic diseases such as Sjögren syndrome and systemic sclerosis. Expanding the knowledge of the pathophysiological roles of autoantibodies against GPCRs will shed light on the biology of these receptors and open avenues for new therapeutic approaches.
Collapse
|
29
|
Nagatomo Y, McNamara DM, Alexis JD, Cooper LT, Dec GW, Pauly DF, Sheppard R, Starling RC, Tang WHW. Myocardial Recovery in Patients With Systolic Heart Failure and Autoantibodies Against β 1-Adrenergic Receptors. J Am Coll Cardiol 2017; 69:968-977. [PMID: 28231950 DOI: 10.1016/j.jacc.2016.11.067] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/15/2016] [Accepted: 11/29/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Among various cardiac autoantibodies (AAbs), those recognizing the β1-adrenergic receptor (β1AR) demonstrate agonist-like effects and induce myocardial damage that can be reversed by β-blockers and immunoglobulin G3 (IgG3) immunoadsorption. OBJECTIVES The goal of this study was to investigate the role of β1AR-AAbs belonging to the IgG3 subclass in patients with recent-onset cardiomyopathy. METHODS Peripheral blood samples were drawn at enrollment in patients with recent-onset cardiomyopathy (left ventricular ejection fraction [LVEF] ≤0.40; <6 months). The presence of IgG and IgG3-β1AR-AAb was determined, and echocardiograms were assessed, at baseline and 6 months. Patients were followed up for ≤48 months. RESULTS Among the 353 patients who had blood samples adequate for the analysis, 62 (18%) were positive for IgG3-β1AR-AAbs (IgG3 group), 58 (16%) were positive for IgG but not IgG3 (non-IgG3 group), and the remaining were negative. There were no significant differences in baseline systolic blood pressure, heart rate, or LVEF among the groups at baseline. Left ventricular end-diastolic and end-systolic diameters were significantly larger in the non-IgG3 group compared with the other groups (left ventricular end-diastolic diameter, p < 0.01; left ventricular end-systolic diameter, p = 0.03). At 6 months, LVEF was significantly higher in the IgG3 group (p = 0.007). Multiple regression analysis showed that IgG3-β1AR-AAb was an independent predictor of LVEF at 6 months and change in LVEF over 6 months, even after multivariable adjustment (LVEF at 6 months, β = 0.20, p = 0.01; change in LVEF, β = 0.20, p = 0.008). In patients with high New York Heart Association functional class (III or IV) at baseline, the IgG3 group had a lower incidence of the composite endpoint of all-cause death, cardiac transplantation, and hospitalization due to heart failure, whereas the non-IgG3 group had the highest incidence of the composite endpoint. CONCLUSIONS IgG3-β1AR-AAbs were associated with more favorable myocardial recovery in patients with recent-onset cardiomyopathy.
Collapse
Affiliation(s)
- Yuji Nagatomo
- Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, Ohio; Sakakibara Heart Institute, Fuchu, Japan
| | - Dennis M McNamara
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jeffrey D Alexis
- University of Rochester Medical Center School of Medicine and Dentistry, Rochester, New York
| | | | - G William Dec
- Massachusetts General Hospital, Boston, Massachusetts
| | - Daniel F Pauly
- Truman Medical Centers, University of Missouri-Kansas City, Kansas City, Missouri
| | | | - Randall C Starling
- Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - W H Wilson Tang
- Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, Ohio.
| | | |
Collapse
|
30
|
Sheng X, Yan J, Meng Y, Kang Y, Han Z, Tai G, Zhou Y, Cheng H. Immunomodulatory effects of Hericium erinaceus derived polysaccharides are mediated by intestinal immunology. Food Funct 2017; 8:1020-1027. [PMID: 28266682 DOI: 10.1039/c7fo00071e] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study was aimed at investigating the immunomodulating activity of Hericium erinaceus polysaccharide (HEP) in mice, by assessing splenic lymphocyte proliferation (cell-mediated immunity), serum hemolysin levels (humoral immunity), phagocytic capacity of peritoneal cavity phagocytes (macrophage phagocytosis), and NK cell activity. ELISA of immunoglobulin A (SIgA) in the lamina propria, and western blotting of small intestinal proteins were also performed to gain insight into the mechanism by which HEP affects the intestinal immune system. Here, we report that HEP improves immune function by functionally enhancing cell-mediated and humoral immunity, macrophage phagocytosis, and NK cell activity. In addition, HEP was found to upregulate the secretion of SIgA and activate the MAPK and AKT cellular signaling pathways in the intestine. In conclusion, all these results allow us to postulate that the immunomodulatory effects of HEP are most likely attributed to the effective regulation of intestinal mucosal immune activity.
Collapse
Affiliation(s)
- Xiaotong Sheng
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| | - Jingmin Yan
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| | - Yue Meng
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| | - Yuying Kang
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| | - Zhen Han
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| | - Guihua Tai
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| | - Yifa Zhou
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| | - Hairong Cheng
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, PR China.
| |
Collapse
|
31
|
Cardiomyopathy - An approach to the autoimmune background. Autoimmun Rev 2017; 16:269-286. [PMID: 28163240 DOI: 10.1016/j.autrev.2017.01.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/20/2016] [Indexed: 12/15/2022]
Abstract
Autoimmunity is increasingly accepted as the origin or amplifier of various diseases. In contrast to classic autoantibodies (AABs), which induce immune responses resulting in the destruction of the affected tissue, an additional class of AABs is directed against G-protein-coupled receptors (GPCRs; GPCR-AABs). GPCR-AABs functionally affect their related GPCRs for activation of receptor mediated signal cascades. Diseases which are characterized by the presence of GPCR-AABs with evidence for disease-specific pathogenic activity could be named "functional autoantibody disease". We briefly summarize here the historical view on autoimmunity in cardiomyopathy, followed by an approach to the mechanistic autoimmunity background. Furthermore, autoantibodies with outstanding importance for cardiomyopathies as a functional autoantibody disease, such as GPCR-AABs, and mainly those directed against the beta1-adrenergic and muscarinic 2 receptor autoantibodies, are introduced. Anti-cardiac myosin and anti-cardiac troponin autoantibodies, as further potential players in autoimmune cardiomyopathy, are additionally taken into account. The basic view on the autoantibodies, their related receptor interactions and pathogenic consequences are presented. Focused specifically on GPCR-AABs, "pros and cons" of assays such as indirect assays (functional changes of cell preparations are monitored after GPCR-AAB receptor binding) and direct assays based on the ELISA technologies (GPCR epitope mimics for GPCR-AAB binding) are critically discussed. Last but not least, treatment strategies for "functional autoantibody disease", such as for GPCR-AAB removal (therapeutic plasma exchange, immunoadsorption) and in vivo GPCR-AAB attack such as intravenous IgG treatment (IVIG), B-cell depletion and GPCR-AAB binding and neutralization, are critically reflected with respect to their patient benefits.
Collapse
|
32
|
Agapova OY, Skoblov YS, Tkachev GA, Mironova NA, Golitsyn SP, Masenko VP, Chazova IE, Zykov KA. Changes in the receptor activity of β2-adrenoreceptors of human T-lymphocytes under the effect of β2-agonists. Mol Biol 2016. [DOI: 10.1134/s0026893316050022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
Autoantibodies against β1-adrenoceptor induce blood glucose enhancement and insulin insufficient via T lymphocytes. Immunol Res 2016; 64:584-93. [PMID: 26639354 PMCID: PMC4788697 DOI: 10.1007/s12026-015-8757-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Diabetes mellitus is a chronic metabolic disorder with a high morbidity and mortality, but its pathogenesis is not fully understood. An increasing amount of evidence indicates that an immune mechanism plays an important role in the pathogenesis of diabetes. We demonstrated previously that the long-term presence of autoantibodies against the second extracellular loop of the β1-adrenoceptor (β1-AA) could change the ratio of peripheral CD4+T/CD8+T in rats, which was accompanied by lymphocytes infiltration in the rat heart, liver, and kidneys. To investigate whether β1-AA is involved in the pathogenesis of diabetes, BALB/c or nude mice were passively immunized with monoclonal antibodies against β1-AR (β1-AR mAb). Compared with vehicle control mice, β1-AA-positive BALB/c mice exhibited significantly increased blood glucose (P < 0.01) and increased fasting insulin (P < 0.05). However, the same changes did not occur in the nude mice. And altered islet morphology was found at week 28 in β1-AA immunization group compared with vehicle control. The basal insulin level of NIT-1 β-cells was decreased markedly (P < 0.01), and the lactate dehydrogenase level was increased (P < 0.01) after the administration of conditioned media from T lymphocytes that had been treated with β1-AA alone. However, these effects were reversed by treatment with metoprolol or peptides of the second extracellular loop of β1-adrenoceptor (β1-AR-ECII). These results suggest that β1-AA could induce hyperglycemia in both rats and mice, and also impair insulin secretion and change islet structure. T lymphocytes may play a key role in the pathogenesis of these changes in the islets.
Collapse
|
34
|
Hu B, Sun Y, Li S, Sun J, Liu T, Wu Z, Feng LI. Association of β1-Adrenergic, M2-Muscarinic Receptor Autoantibody with Occurrence and Development of Nonvalvular Atrial Fibrillation. Pacing Clin Electrophysiol 2016; 39:1379-1387. [PMID: 27862036 DOI: 10.1111/pace.12976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 10/06/2016] [Accepted: 10/28/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common sustained arrhythmia and is independently associated with increased risk of stroke and death. Although the exact mechanisms of AF are not completely elucidated, a large number of evidences demonstrate that autoimmunity may play an important role in the initiation, the progression, and the maintenance of AF. In this study, we aimed to compare anti-β1-adrenergic receptor autoantibody (anti-β1-R) and anti-M2-muscarinic receptor autoantibody (anti-M2-R) levels between nonvalvular AF patients and healthy control subjects. METHODS The levels of serum anti-β1-R, antinuclear antibodies, and anti-M2-R were measured in both groups by enzyme-linked immunosorbent assay (ELISA). High-sensitivity C-reactive protein (hs-CRP) and interleukin (IL)-6 concentration were measured, respectively, by immunoturbidimetry and chemiluminescence. RESULTS Anti-β1-R and anti-M2-R levels were significantly higher in patients with nonvalvular AF than in healthy controls (anti-β1-R 221.11 [132.38-291.69] ng/mL vs 198.14 [125.70-278.40] ng/mL, P < 0.01; anti-M2-R 271.81 [144.99-378.20] ng/mL vs 235.01 [121.53-358.99] ng/mL, P < 0.01). Compared with the control group, the serum levels of IL-6 and hs-CRP were higher in the nonvalvular AF group (IL-6 19.65 ± 5.6 pg/mL vs 6.79 ± 1.09 pg/mL, hs-CRP 6.03 ± 1.35 mg/L vs 2.73 ± 0.63 mg/L, P < 0.05). Antinuclear antibody (ANA) levels were similar between two groups (ANA 10.55 [1.86-271.8] U/mL vs 10.49 [1.303-161.7] U/mL, P > 0.05). The baseline value of serum anti-β1-R (odds ratio [OR]: 13.176, P < 0.001), anti-M2-R (OR: 4.41, P < 0.001), IL-6 (OR: 6.126, P < 0.05) levels, and left atrial diameter (OR: 5.781, P < 0.05) were independent predictors of nonvalvular AF by multivariable analyses. CONCLUSION We found a significant association between circulating serum anti-β1-R, anti-M2-R, IL-6 levels, and nonvalvular AF. We presume that the autoimmunity and inflammation might take part in electrical remodeling and structural remodeling of left atrium.
Collapse
Affiliation(s)
- Bing Hu
- Department of Cardiology, Zhongshan Affiliated Hospital of Sun Yat-sen University, Zhongshan, Guangdong, China
| | - Yanxiang Sun
- Department of Cardiology, Zhongshan Affiliated Hospital of Sun Yat-sen University, Zhongshan, Guangdong, China
| | - Sha Li
- Department of Laboratory Diagnosis, Zhongshan Affiliated Hospital of Sun Yat-sen University, Zhongshan, Guangdong, China
| | - Jie Sun
- Department of Cardiology, Zhongshan Affiliated Hospital of Sun Yat-sen University, Zhongshan, Guangdong, China
| | - Tong Liu
- Department of Cardiology, Zhongshan Affiliated Hospital of Sun Yat-sen University, Zhongshan, Guangdong, China
| | - Zidi Wu
- Department of Cardiology, Zhongshan Affiliated Hospital of Sun Yat-sen University, Zhongshan, Guangdong, China
| | - L I Feng
- Department of Cardiology, Zhongshan Affiliated Hospital of Sun Yat-sen University, Zhongshan, Guangdong, China
| |
Collapse
|
35
|
Lv T, Du Y, Cao N, Zhang S, Gong Y, Bai Y, Wang W, Liu H. Proliferation in cardiac fibroblasts induced by β1-adrenoceptor autoantibody and the underlying mechanisms. Sci Rep 2016; 6:32430. [PMID: 27577254 PMCID: PMC5006240 DOI: 10.1038/srep32430] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/09/2016] [Indexed: 12/25/2022] Open
Abstract
Chronic sustained stimulation of β-adrenoceptor is closely related to cardiac fibrosis which is bad for cardiac function. Growing evidence showed that the high prevalence of β1-adrenoceptor autoantibody (β1-AA) in the sera of patients with various types of cardiovascular diseases decreased cardiac function. In the current study, we demonstrated that β1-AA impaired the cardiac function evaluated by echocardiography and that β1-AA triggered cardiac fibrosis in terms of increased expression of α-smooth muscle actin as the marker of myofibroblast and collagen deposition in a passive β1-AA immunized mice model during 16 weeks. Further, we showed that β1-AA activated β1-AR/cAMP/PKA pathway and promoted proliferation in primary cardiac fibroblasts through specific binding to β1-AR but not to β2-AR. Moreover, β1-AA was also likely to promote proliferation in cardiac fibroblasts through activating p38MAPK and ERK1/2 as p38MAPK inhibitor SB203580 and ERK1/2 inhibitor PD98059 partially reversed the proliferative effect. The persistent activating signalling of PKA and P38MAPK in 1 h induced by β1-AA was associated with lacking agonist-induced desensitization phenomena. The conditioned medium from β1-AA-stimulated cardiac fibroblasts induced cardiomyocyte apoptosis, which indicated that β1-AA changed the secretion of cardiac fibroblasts contributing to cardiac injury. These findings will contribute to our understanding of the pathological mechanisms of β1-AA.
Collapse
Affiliation(s)
- Tingting Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Yunhui Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Ning Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Yulin Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Yan Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| |
Collapse
|
36
|
Loebel M, Grabowski P, Heidecke H, Bauer S, Hanitsch LG, Wittke K, Meisel C, Reinke P, Volk HD, Fluge Ø, Mella O, Scheibenbogen C. Antibodies to β adrenergic and muscarinic cholinergic receptors in patients with Chronic Fatigue Syndrome. Brain Behav Immun 2016; 52:32-39. [PMID: 26399744 DOI: 10.1016/j.bbi.2015.09.013] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/18/2015] [Accepted: 09/19/2015] [Indexed: 10/23/2022] Open
Abstract
Infection-triggered disease onset, chronic immune activation and autonomic dysregulation in CFS point to an autoimmune disease directed against neurotransmitter receptors. Autoantibodies against G-protein coupled receptors were shown to play a pathogenic role in several autoimmune diseases. Here, serum samples from a patient cohort from Berlin (n=268) and from Bergen with pre- and post-treatment samples from 25 patients treated within the KTS-2 rituximab trial were analysed for IgG against human α and β adrenergic, muscarinic (M) 1-5 acetylcholine, dopamine, serotonin, angiotensin, and endothelin receptors by ELISA and compared to a healthy control cohort (n=108). Antibodies against β2, M3 and M4 receptors were significantly elevated in CFS patients compared to controls. In contrast, levels of antibodies against α adrenergic, dopamine, serotonin, angiotensin, and endothelin receptors were not different between patients and controls. A high correlation was found between levels of autoantibodies and elevated IgG1-3 subclasses, but not with IgG4. Further patients with high β2 antibodies had significantly more frequently activated HLA-DR+ T cells and more frequently thyreoperoxidase and anti-nuclear antibodies. In patients receiving rituximab maintenance treatment achieving prolonged B-cell depletion, elevated β2 and M4 receptor autoantibodies significantly declined in clinical responder, but not in non-responder. We provide evidence that 29.5% of patients with CFS had elevated antibodies against one or more M acetylcholine and β adrenergic receptors which are potential biomarkers for response to B-cell depleting therapy. The association of autoantibodies with immune markers suggests that they activate B and T cells expressing β adrenergic and M acetylcholine receptors. Dysregulation of acetylcholine and adrenergic signalling could also explain various clinical symptoms of CFS.
Collapse
Affiliation(s)
- Madlen Loebel
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany.
| | - Patricia Grabowski
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany
| | | | - Sandra Bauer
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Leif G Hanitsch
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Kirsten Wittke
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Christian Meisel
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany; Labor Berlin GmbH, Immunology Department, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Petra Reinke
- Department of Nephrology, Charité University Medicine Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Germany
| | - Hans-Dieter Volk
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Germany
| | - Øystein Fluge
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Olav Mella
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Carmen Scheibenbogen
- Institute for Medical Immunology, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Germany
| |
Collapse
|
37
|
Pavlovic V, Cekic S, Ciric M, Krtinic D, Jovanovic J. Curcumin attenuates Mancozeb-induced toxicity in rat thymocytes through mitochondrial survival pathway. Food Chem Toxicol 2016; 88:105-11. [DOI: 10.1016/j.fct.2015.12.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 12/20/2015] [Accepted: 12/31/2015] [Indexed: 12/21/2022]
|
38
|
Agapova OY, Skoblov YS, Zykov KA, Rvacheva AV, Beilina VB, Masenko VP, Chazova IE. [Radioligand Method of Assessment of β-Adrenoceptor's Activity on Human T-Lymphocytes]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2016; 41:592-8. [PMID: 26762097 DOI: 10.1134/s1068162015050027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We proposed a new method of evaluation of beta-receptor's activity on the surface of human T-lymphocytes based on the radioligand method. Optimal conditions for evaluation of specific binding to β2-adrenoceptors of 0.5 fmol ligand per 1 million cells using [125I]-cyanopindolol were found. The possibility of using of β2-adrenoceptor's activity assessment in clinical settings was demonstrated on human T-lymphocyte.
Collapse
|
39
|
Jiang Y, Yang S, Tao J, Lin Z, Ye X, You Y, Peng J, Hong Z, Chen L. Opposing needling promotes behavior recovery and exerts neuroprotection via the cAMP/PKA/CREB signal transduction pathway in transient MCAO rats. Mol Med Rep 2016; 13:2060-70. [PMID: 26780954 PMCID: PMC4768950 DOI: 10.3892/mmr.2016.4773] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 10/28/2015] [Indexed: 01/02/2023] Open
Abstract
The aim of the present study was to investigate whether the cyclic adenosine 3′,5′-monophosphate (cAMP)/protein kinase A(PKA)/cAMP-responsive element binding protein (CREB) signal transduction pathway triggered by γ-aminobutyric acid class B (GABAB) receptor activation is involved in neuroprotection against ischemia and behavioral recovery induced by opposing needling (ON). A total of 80 rats were randomly divided into four groups: A sham operation group, an ischemia group, an ON group and an ON group effectively inhibited by the GABAB receptor antagonist, CGP35384 (n=20/group). The behavior of the rats was assessed by their neurological deficit score, whereas the impairment of gait was examined using the CatWalk system. The volume of cerebral infarction was examined upon treatment with 2,3,5-triphenyltetrazolium chloride. The expression levels of CREB, GABAB1 and GABAB2 were examined by western blotting and reverse transcription-quantitative polymerase chain reaction, and the activity of adenylyl cyclase (AC), cAMP and PKA in the serum was detected using an enzyme-linked immunosorbent assay. In the present study, in comparison with other groups, the ON group exhibited a reduced score for the neurological deficit, the stride length and swing speed were improved, and the volume of infarction was reduced. However, these effects were reversed upon administration of CGP35384. Additionally, the expression levels of CREB, GABAB1 and GABAB2 were increased in the ON group. The levels of AC, cAMP and PKA in the serum were also increased in the ON group, whereas the addition of CGP35384 reversed these effects. The results of the present study demonstrated that ON markedly protected the brain against transient cerebral ischemic injury, and this effect was possibly mediated by the activation of the GABAB/cAMP/PKA/CREB signal transduction pathway. These findings implied that ON may be a potential therapeutic method for treating stroke.
Collapse
Affiliation(s)
- Yijing Jiang
- Department of Rehabilitation Medicine, Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350001, P.R. China
| | - Shanli Yang
- Department of Rehabilitation Medicine, Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350001, P.R. China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Zhicheng Lin
- Department of Rehabilitation Medicine, Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350001, P.R. China
| | - Xiaoqian Ye
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yongmei You
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jun Peng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Zhenfeng Hong
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
40
|
Zhao Y, Huang H, Du Y, Li X, Lv T, Zhang S, Wei H, Shang J, Liu P, Liu H. β1-Adrenoceptor autoantibodies affect action potential duration and delayed rectifier potassium currents in guinea pigs. Cardiovasc Toxicol 2015; 15:1-9. [PMID: 24894912 DOI: 10.1007/s12012-014-9261-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
β1-Adrenoceptor autoantibodies (β1-AAs) affect the action potential duration (APD) in cardiomyocytes and are related to ventricular arrhythmias. The delayed rectifier potassium current (I K) plays a crucial role in APD, but the effects of β1-AAs on I K have not been completely illuminated. This work aimed to observe the effects of β1-AAs on I K and APD and further explore the mechanisms of β1-AA-mediated ventricular arrhythmias. β1-AAs were obtained from sera of patients with coronary heart disease (CHD) and nonsustained ventricular tachycardia. With whole-cell patch clamp technique, action potentials and I K were recorded. The results illustrated 0.1 μmol/L β1-AAs shortened APD at 50 % (APD50) and 90 % (APD90) of the repolarization. However, at 0.01 μmol/L, β1-AAs had no effects on either APD90 or APD50 (P > 0.05). At 0.001 μmol/L, β1-AAs significantly prolonged APD90 and APD50. Moreover, β1-AAs (0.001, 0.01, 0.1 μmol/L) dose-dependently increased the rapidly activating delayed rectifier potassium current (I Kr), but similarly decreased the slowly activating delayed rectifier potassium current (I Ks) and increased L-type calcium currents at the different concentrations. Taken together, the IKr increase induced by high β1-AA concentrations is responsible for a significant APD reduction which would contribute to repolarization changes and trigger the malignant ventricular arrhythmias in CHD patients.
Collapse
Affiliation(s)
- Yuhui Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pro-arrhythmic action of autoantibodies against the second extracellular loop of β1-adrenoceptor and its underlying molecular mechanisms. Int J Cardiol 2015; 198:251-8. [PMID: 26241168 DOI: 10.1016/j.ijcard.2015.06.144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/19/2015] [Accepted: 06/27/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The incidence of arrhythmia is associated with autoantibodies against the second extracellular loop of β1-adrenergic receptor (β1-AAbs). The current study was designed to determine the mechanisms by which arrhythmia experimentally might be induced by β1-AAbs. METHODS Blood samples were collected from patients with varied arrhythmias or coronary heart disease (CHD) and healthy subjects. The titer of β1-AAbs was assessed. Passive immunization rat models with β1-AAbs were established to determine whether β1-AAbs induced arrhythmia. Conventional intracellular microelectrode technique and whole cell patch clamp were employed to record action potential duration (APD), resting potential (RP), L-type calcium current (ICa-L), sodium-calcium exchange current (INCX), transient outward potassium current (Ito), inward rectifier potassium current (Ik1) and delayed rectifier potassium current (Ik). RESULTS High levels of β1-AAbs were found in the sera of heart disease patients, especially in ventricular arrhythmia (VA). Transfusion with β1-AAbs could induce arrhythmias in normal rats in vivo. β1-AAbs purified from the sera of active immunized rats induced triggered activity (TA), delayed after depolarization (DAD), and prolonged APD in the papillary muscles of rats. β1-AAbs prolonged QT interval, increased ICa-L and decreased IK1, Ito and INa-Ca in rat ventricular myocytes in vitro. All these effects can be inhibited by β1-AR blocker metoprolol. CONCLUSIONS These results demonstrate for the first time that β1-AAbs could directly induce ventricular arrhythmia by prolonging QT interval.
Collapse
|
42
|
Angiotensin II type 1 receptor autoantibody as a novel regulator of aldosterone independent of preeclampsia. J Hypertens 2015; 33:1046-56. [DOI: 10.1097/hjh.0000000000000521] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
43
|
Nagatomo Y, Yoshikawa T, Okamoto H, Kitabatake A, Hori M. Presence of Autoantibody Directed Against β1-Adrenergic Receptors Is Associated With Amelioration of Cardiac Function in Response to Carvedilol: Japanese Chronic Heart Failure (J-CHF) Study. J Card Fail 2015; 21:198-207. [DOI: 10.1016/j.cardfail.2014.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/08/2014] [Accepted: 12/10/2014] [Indexed: 11/27/2022]
|
44
|
Barreiro-Costa O, Tobío A, Alfonso A, Botana LM. Different role of cAMP pathway on the human mast cells HMC-1(560) and HMC-1(560,816) activation. J Cell Biochem 2014; 115:896-909. [PMID: 24307603 DOI: 10.1002/jcb.24732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/02/2013] [Indexed: 01/07/2023]
Abstract
HMC-1 are inflammatory cells that release vasoactive substances such as histamine. These cells have the c-kit receptor permanently activated in the membrane due to mutations in the proto-oncogene c-kit: Val-560 → Gly and Asp-816 → Val. Thus, there are two known cellular lines: HMC-1(560) and HMC-1(560,816) . These mutations are involved in a disease called mastocitosys. In the present paper both lines were used to study the influence of cAMP/PKA/PDEs pathway on the histamine release and Ca(2+) signaling since this pathway is often involved in these process. For this, the cells were preincubated with cAMP/PKA/PDEs modulators such as dibutyryl cAMP (dbcAMP), forskolin, H89, rolipram, IBMX, or imidazole and then stimulated with ionomycin. When cells were stimulated with agents that increase cAMP levels, the histamine release was not modified in HMC-1(560) but decreased in HMC-1(560,816) cells. The same happened when PKA was blocked. Furthermore, PDEs role on histamine release was independent of cAMP in HMC-1(560) cells and possibly also in HMC-1(560,816) cells. By contrast, the modulation of PKA and PDEs together changed the response in both cellular lines, therefore a relationship between them was suggested. All these modulatory effects on histamine release are Ca(2+) -independent. On the other hand, the effect of c-kit modulation on the cAMP/PKA/PDEs pathway was also checked. This receptor was blocked with STI571 (imatinib) and BMS-354825 (dasatinib), but only the last one caused a decrease in the cellular response to ionomycin. This article demonstrates for the first time than the cAMP/PKA/PDEs pathway is involved in the activation of HMC-1(560) and HMC-1(560,816) cells.
Collapse
|
45
|
Francone M. Role of cardiac magnetic resonance in the evaluation of dilated cardiomyopathy: diagnostic contribution and prognostic significance. ISRN RADIOLOGY 2014; 2014:365404. [PMID: 24967294 PMCID: PMC4045555 DOI: 10.1155/2014/365404] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 11/05/2013] [Indexed: 01/07/2023]
Abstract
Dilated cardiomyopathy (DCM) represents the final common morphofunctional pathway of various pathological conditions in which a combination of myocyte injury and necrosis associated with tissue fibrosis results in impaired mechanical function. Recognition of the underlying aetiology of disease and accurate disease monitoring may be crucial to individually optimize therapeutic strategies and stratify patient's prognosis. In this regard, CMR has emerged as a new reference gold standard providing important information for differential diagnosis and new insight about individual risk stratification. The present review article will focus on the role of CMR in the evaluation of present condition, analysing respective strengths and limitations in the light of current literature and technological developments.
Collapse
Affiliation(s)
- Marco Francone
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena, 324 00161 Rome, Italy
| |
Collapse
|