1
|
Figueiredo Galvao HB, Lieu M, Moodley S, Diep H, Jelinic M, Bobik A, Sobey CG, Drummond GR, Vinh A. Depletion of follicular B cell-derived antibody secreting cells does not attenuate angiotensin II-induced hypertension or vascular compliance. Front Cardiovasc Med 2024; 11:1419958. [PMID: 38883991 PMCID: PMC11176447 DOI: 10.3389/fcvm.2024.1419958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Marginal zone and follicular B cells are known to contribute to the development of angiotensin II-induced hypertension in mice, but the effector function(s) mediating this effect (e.g., antigen presentation, antibody secretion and/or cytokine production) are unknown. B cell differentiation into antibody secreting cells (ASCs) requires the transcription factor Blimp-1. Here, we studied mice with a Blimp-1 deficiency in follicular B cells to evaluate whether antibody secretion underlies the pro-hypertensive action of B cells. Methods 10- to 14-week-old male follicular B cell Blimp-1 knockout (FoB-Blimp-1-KO) and floxed control mice were subcutaneously infused with angiotensin II (0.7 mg/kg/d) or vehicle (0.1% acetic acid in saline) for 28 days. BP was measured by tail-cuff plethysmography or radiotelemetry. Pulse wave velocity was measured by ultrasound. Aortic collagen was quantified by Masson's trichrome staining. Cell types and serum antibodies were quantified by flow cytometry and a bead-based multiplex assay, respectively. Results In control mice, angiotensin II modestly increased serum IgG3 levels and markedly increased BP, cardiac hypertrophy, aortic stiffening and fibrosis. FoB-Blimp-1-KO mice exhibited impaired IgG1, IgG2a and IgG3 production despite having comparable numbers of B cells and ASCs to control mice. Nevertheless, FoB-Blimp-1-KO mice still developed hypertension, cardiac hypertrophy, aortic stiffening and fibrosis following angiotensin II infusion. Conclusions Inhibition of follicular B cell differentiation into ASCs did not protect against angiotensin II-induced hypertension or vascular compliance. Follicular B cell functions independent of their differentiation into ASCs and ability to produce high-affinity antibodies, or other B cell subtypes, are likely to be involved in angiotensin II-induced hypertension.
Collapse
Affiliation(s)
- Hericka Bruna Figueiredo Galvao
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Maggie Lieu
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Seyuri Moodley
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Henry Diep
- Victorian Heart Institute, Monash University, Clayton, VIC, Australia
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Alexander Bobik
- Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Department of Immunology, Monash University, Clayton, VIC, Australia
- Center for Inflammatory Diseases, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, VIC, Australia
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, VIC, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Moleón J, González-Correa C, Robles-Vera I, Miñano S, de la Visitación N, Barranco AM, Martín-Morales N, O’Valle F, Mayo-Martínez L, García A, Toral M, Jiménez R, Romero M, Duarte J. Targeting the gut microbiota with dietary fibers: a novel approach to prevent the development cardiovascular complications linked to systemic lupus erythematosus in a preclinical study. Gut Microbes 2023; 15:2247053. [PMID: 37615336 PMCID: PMC10453983 DOI: 10.1080/19490976.2023.2247053] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/04/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
This study is to investigate whether dietary fiber intake prevents vascular and renal damage in a genetic mouse model of systemic lupus erythematosus (SLE), and the contribution of gut microbiota in the protective effects. Female NZBWF1 (SLE) mice were treated with resistant-starch (RS) or inulin-type fructans (ITF). In addition, inoculation of fecal microbiota from these experimental groups to recipient normotensive female C57Bl/6J germ-free (GF) mice was performed. Both fiber treatments, especially RS, prevented the development of hypertension, renal injury, improved the aortic relaxation induced by acetylcholine, and the vascular oxidative stress. RS and ITF treatments increased the proportion of acetate- and butyrate-producing bacteria, respectively, improved colonic inflammation and integrity, endotoxemia, and decreased helper T (Th)17 proportion in mesenteric lymph nodes (MLNs), blood, and aorta in SLE mice. However, disease activity (splenomegaly and anti-ds-DNA) was unaffected by both fibers. T cell priming and Th17 differentiation in MLNs and increased Th17 infiltration was linked to aortic endothelial dysfunction and hypertension after inoculation of fecal microbiota from SLE mice to GF mice, without changes in proteinuria and autoimmunity. All these effects were lower in GF mice after fecal inoculation from fiber-treated SLE mice. In conclusion, these findings support that fiber consumption prevented the development of hypertension by rebalancing of dysfunctional gut-immune system-vascular wall axis in SLE.
Collapse
Affiliation(s)
- Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Néstor de la Visitación
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Antonio Manuel Barranco
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Natividad Martín-Morales
- Department of Pathology, School of Medicine, Instituto de Biopatología y Medicina Regenerativa (IBIMER) University of Granada, Granada, Spain
| | - Francisco O’Valle
- Department of Pathology, School of Medicine, Instituto de Biopatología y Medicina Regenerativa (IBIMER) University of Granada, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Laura Mayo-Martínez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, CEU Universities. Campus Monteprincipe, Boadilla del Monte, San Pablo, Spain
| | - Antonia García
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, CEU Universities. Campus Monteprincipe, Boadilla del Monte, San Pablo, Spain
| | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
3
|
Zimering MB. Repeated Traumatic Brain Injury is Associated with Neurotoxic Plasma Autoantibodies Directed against the Serotonin 2A and Alpha 1 Adrenergic Receptors. ENDOCRINOLOGY, DIABETES AND METABOLISM JOURNAL 2023; 7:1-12. [PMID: 37560352 PMCID: PMC10411137 DOI: 10.31038/edmj.2023722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Objectives Traumatic brain injury (TBI) was associated with increased plasma agonist autoantibodies targeting the serotonin 2A receptor. Repeated TBI exposure is associated with high risk for neurodegenerative and neuropsychiatric complications. Here we tested a hypothesis that repeated TBI is associated with plasma agonist autoantibodies targeting more than one kind of catecholamine G-protein coupled receptor. Methods Protein-A affinity chromatography was used to isolate the IgG fraction of plasma in forty-two middle-aged and older adults who had experienced one or more TBI exposures. The Ig (1/40th dilution=7.5 ug/mL) were tested for neurotoxicity in mouse neuroblastoma cells using an acute neurite retraction assay indicative of Gq11/IP3/Ca2+ and RhoA/Rho kinase signaling pathways' activation. Three different linear synthetic peptides corresponding to the second extracellular loop of the alpha 1A, alpha 2A or serotonin 2A receptors were used as target antigen in different enzyme-linked immunoassays. The second extracellular loop receptor peptides themselves (alpha 1A, alpha 2A) or a fragment (serotonin 2A) were tested for ability to prevent Ig-induced neurite retraction. Results Patients who had experienced either repeated TBI (N=10) or a single TBI with a co-morbid autoimmune disease (N=5) were significantly more likely to harbor neurotoxic plasma autoantibodies targeting both alpha 1 adrenergic and serotonin 2A receptors vs. patients having only a single TBI. Ig-induced neurotoxicity was significantly prevented by co-incubation with either 850 nM prazosin (alpha 1 adrenergic receptor) and/or 500 nM M100907 (serotonin 2A receptor) antagonists. Alpha 1 adrenergic receptor and serotonin 2A receptor Ig immunoreactive level and titer were significantly increased in repeated TBI and single TBI/autoimmune patients (N=7-8) compared to age-matched TBI patients without neurotoxic plasma Ig (N=4). SN.8, a linear synthetic peptide corresponding to a conserved region of the second extracellular loop (ECL) of the serotonin 2A receptor completely prevented neurite retraction induced by repeated TBI plasma Ig. A repeated TBI patient harboring alpha adrenergic receptor AAB alone experienced prospective steep decline in cognitive function over two years. Conclusions Repeated TBI and TBI with associated autoimmunity harbored more than one kind of neurotoxic catecholaminergic agonist GPCR autoantibody each associated with high risk for steep rate of cognitive decline. Specific immunoassays using the second extracellular receptor loop as target antigen are needed to detect each specific different GPCR autoantibody. A fragment of the second ECL of the serotonin 2A receptor (SN.8) neutralized Ig-induced neurotoxicity in repeated TBI or TBI with associated systemic autoimmunity.
Collapse
Affiliation(s)
- Mark B Zimering
- Medical Service, Veterans Affairs New Jersey Healthcare System, East Orange, New Jersey, USA
- Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
4
|
Benvenga S, Antonelli A, Fallahi P, Bonanno C, Rodolico C, Guarneri F. Amino acid sequence homology between thyroid autoantigens and central nervous system proteins: Implications for the steroid-responsive encephalopathy associated with autoimmune thyroiditis. J Clin Transl Endocrinol 2021; 26:100274. [PMID: 34849350 PMCID: PMC8609095 DOI: 10.1016/j.jcte.2021.100274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 10/15/2021] [Accepted: 10/27/2021] [Indexed: 11/13/2022] Open
Abstract
Alpha-enolase, aldehyde reductase-I and dimethylargininase-I are SREAT autoantigens. Molecular mimicry between thyroid and CNS autoantigens is hypothesized in SREAT. Homology with TSH-R, Tg and TPO exists for 6, 27 and 47 of 46,809 CNS-proteins. The above homologies are often in epitope-containing parts of thyroid autoantigens. Most of the above proteins are expressed in CNS regions which are altered in SREAT.
A few patients with Hashimoto’s thyroiditis or Graves’ disease develop a multiform syndrome of the central nervous system (CNS) termed Hashimoto’s encephalopathy or steroid-responsive encephalopathy associated with autoimmune thyroid disease (HE/SREAT). They have high levels of thyroid autoantibodies (TgAb, TPOAb and/or TSH-R-Ab) in blood and cerebrospinal fluid. Autoantibodies against alpha-enolase, aldehyde reductase-I (AKRIA) and/or dimethylargininase-I (DDAHI), proteins expressed in the CNS among other tissues, were detected in the blood and, when searched, in the cerebrospinal fluid of HE/SREAT patients. Recently, we reported that alpha-enolase, AKRIA and DDAHI share local sequence homology with each of the three autoantigens (TgAb, TPOAb, TSH-R-Ab), often in epitope-containing segments of the thyroid autoantigens. We hypothesized that there might be additional CNS-expressed proteins homologous to thyroid autoantigens, possibly overlapping known epitopes of the thyroid autoantigens. We used bioinformatic methods to address this hypothesis. Six, 27 and 47 of 46,809 CNS-expressed proteins share homology with TSH-R, Tg and TPO, respectively. The homologous regions often contain epitopes, and some match regions of thyroid autoantigens which have homology with alpha-enolase, AKRIA and/or DDAHI. Several of the aforementioned proteins are present in CNS areas that show abnormalities at neuroimaging in HE/SREAT patients. Furthermore, autoantibodies against some of the said six, 27 and 47 proteins were reported to be associated with a number of autoimmune diseases. Not only we validated our hypothesis, but we think that such a variety of potential CNS targets for thyroid Ab against epitopes contained in regions that have local homology with CNS proteins may explain the polymorphic phenotypes of HE/SREAT. Only when elevated amounts of these Ab are synthesized and trespass the blood-brain barrier, HE/SREAT appears. This might explain why HE/SREAT is so relatively rare.
Collapse
|
5
|
Zhang L, Sun Y, Zhang X, Shan X, Li J, Yao Y, Shu Y, Lin K, Huang X, Yang Z, Chu J, Huang L, Sun H. Three Novel Genetic Variants in the FAM110D, CACNA1A, and NLRP12 Genes Are Associated With Susceptibility to Hypertension Among Dai People. Am J Hypertens 2021; 34:874-879. [PMID: 33621312 DOI: 10.1093/ajh/hpab040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/17/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Although the genetic factors associated with hypertension remain unknown, genetic variations in genes related to ion channels, inflammation, and the cell cycle may affect susceptibility to hypertension. In the present study, the association between hypertension and 10 candidate single-nucleotide polymorphisms (SNPs) was evaluated among Chinese Dai people, who have a smaller gene pool than Han individuals. METHODS A total of 1,193 samples from Dai people were collected, including 488 with hypertension and 705 with normal blood pressure. Based on the preliminary results of whole-genome sequencing among pools of individuals (Pool-seq), 10 candidate SNPs in 6 genes (FAM110D, ADD1, RAG1, CACNA1C, CACNA1A, and NLRP12) were genotyped in the case and control groups by multiplex PCR for SNP genotyping with next-generation sequencing (MultiPCR-NGS). The relationship between hypertension and each candidate SNP was evaluated using the χ 2 test and multiple logistic regression analysis. RESULTS The χ 2 test showed that the allele frequencies of rs3748856 in FAM110D, rs139118504 in CACNA1A, and rs34436714 in NLRP12 were significantly different between the case and control groups (P < 0.005). After adjusting for age, body mass index, total cholesterol, triglyceride, and low-density lipoprotein, logistic regression analyses revealed that the association between the 3 SNPs and hypertension among Dai people remained significant (P = 0.012, 2.71 × 10-4, and 0.017, respectively). CONCLUSIONS These findings indicate that there may be different molecular pathogeneses of hypertension among Dai people, which should be noted in future studies.
Collapse
Affiliation(s)
- Lin Zhang
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yun Sun
- Yunnan Key Laboratory of Southern Medicinal Resource, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaochao Zhang
- Key Laboratory of Pharmacology for Natural Products of Yunnan Province, Pharmaceutical College, Kunming Medical University, Kunming, Yunnan, China
| | - Xiyun Shan
- The Department of clinical laboratory, People’s Hospital of Xishuangbanna Dai Autonomous Prefecture, Kunming University of Science and Technology, Xishuangbanna, China
| | - Jianmei Li
- The Cardiovascular Department, The Second People’s Hospital of Yunnan Province, Affiliated Hospital of Yunnan University, Kunming, China
| | - Yao Yao
- The Department of clinical laboratory, People’s Hospital of Xishuangbanna Dai Autonomous Prefecture, Kunming University of Science and Technology, Xishuangbanna, China
| | - Yun Shu
- The Department of clinical laboratory, People’s Hospital of Xishuangbanna Dai Autonomous Prefecture, Kunming University of Science and Technology, Xishuangbanna, China
| | - Keqin Lin
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Xiaoqin Huang
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Zhaoqing Yang
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Jiayou Chu
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Lifan Huang
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Hao Sun
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| |
Collapse
|
6
|
de la Visitación N, Robles-Vera I, Toral M, Gómez-Guzmán M, Sánchez M, Moleón J, González-Correa C, Martín-Morales N, O'Valle F, Jiménez R, Romero M, Duarte J. Gut microbiota contributes to the development of hypertension in a genetic mouse model of systemic lupus erythematosus. Br J Pharmacol 2021; 178:3708-3729. [PMID: 33931880 DOI: 10.1111/bph.15512] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/15/2021] [Accepted: 04/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Hypertension is an important cardiovascular risk factor that is prevalent in the systemic lupus erythematosus patient population. Here, we have investigated whether intestinal microbiota is involved in hypertension in a genetic mouse model of systemic lupus erythematosus. EXPERIMENTAL APPROACH Twenty-six-week-old female NZW/LacJ (control) and NZBWF1 (F1 hybrid of New Zealand Black and New Zealand White strains; systemic lupus erythematosus) mice were treated for 6 weeks with a broad-spectrum antibiotic mixture or with vancomycin. Faecal microbiota transplantation was performed from donor systemic lupus erythematosus group to recipient to germ-depleted or germ-free mice. KEY RESULTS Antibiotic treatment inhibited the development of hypertension and renal injury, improved endothelial dysfunction and vascular oxidative stress, and decreased aortic Th17 infiltration in NZBWF1 mice. High BP and vascular complications found in systemic lupus erythematosus mice, but not autoimmunity, kidney inflammation and endotoxemia, were reproduced by the transfer of gut microbiota from systemic lupus erythematosus donors to germ-free or germ-depleted mice. Increased proportions of Bacteroides were linked with high BP in these mice. The reduced endothelium-dependent vasodilator responses to acetylcholine and the high BP induced by microbiota from hypertensive systemic lupus erythematosus mice were inhibited after IL-17 neutralization. CONCLUSION AND IMPLICATIONS Changes in T-cell populations, endothelial function, vascular inflammation and hypertension driven by a genetic systemic lupus erythematosus background can be modified by antibiotic-induced changes in gut microbiota. The vascular changes induced by hypertensive systemic lupus erythematosus microbiota were mediated by Th17 infiltration in the vasculature.
Collapse
Affiliation(s)
- Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - 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.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Manuel Gómez-Guzmán
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Manuel Sánchez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | | | - Francisco O'Valle
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.,Department of Pathology, School of Medicine, University of Granada, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| |
Collapse
|
7
|
Perez DM. Current Developments on the Role of α 1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism. Front Cell Dev Biol 2021; 9:652152. [PMID: 34113612 PMCID: PMC8185284 DOI: 10.3389/fcell.2021.652152] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
8
|
McClung DM, Kalusche WJ, Jones KE, Ryan MJ, Taylor EB. Hypertension and endothelial dysfunction in the pristane model of systemic lupus erythematosus. Physiol Rep 2021; 9:e14734. [PMID: 33527772 PMCID: PMC7851437 DOI: 10.14814/phy2.14734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/19/2020] [Indexed: 01/01/2023] Open
Abstract
Autoimmune diseases such as psoriasis, rheumatoid arthritis, and systemic lupus erythematosus (SLE) have high rates of hypertension and cardiovascular disease. Systemic lupus erythematosus is a prototypic autoimmune disorder that primarily affects women of childbearing age and is associated with a loss of self-tolerance, autoreactive B and T lymphocytes, and the production of autoantibodies, especially to nuclear components. In this study, we hypothesized that the pristane-inducible model of SLE would develop hypertension and vascular dysfunction as the disease progressed. To test this hypothesis, female C57BL/6 mice were administered PBS or pristane. Seven months after pristane administration, mice developed various autoantibodies, including anti-dsDNA IgG, anti-ssDNA IgG, and anti-nRNP IgG, as well as hypergammaglobulinemia. Several other immunological changes, including increased circulating neutrophils and increased CD4- CD8- (double negative) thymocytes were also detected. Mean arterial pressure (MAP) was elevated in pristane-treated mice when compared to PBS-treated mice. In addition, second-order mesenteric arteries from pristine-treated mice had impaired relaxation to the endothelium-dependent vasodilator acetylcholine compared to PBS-treated mice. These data suggest that the immune system dysfunction present in the pristane model of lupus contributes to the development of hypertension and vascular dysfunction.
Collapse
Affiliation(s)
- Daniel M. McClung
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - William J. Kalusche
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Katie E. Jones
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Michael J. Ryan
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
- G.V. (Sonny) Montgomery Veterans Affairs Medical CenterJacksonMSUSA
| | - Erin B. Taylor
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| |
Collapse
|
9
|
Stracke S, Lange S, Bornmann S, Kock H, Schulze L, Klinger-König J, Böhm S, Vogelgesang A, von Podewils F, Föel A, Gross S, Wenzel K, Wallukat G, Prüss H, Dressel A, Kunze R, Grabe HJ, Langner S, Dörr M. Immunoadsorption for Treatment of Patients with Suspected Alzheimer Dementia and Agonistic Autoantibodies against Alpha1a-Adrenoceptor-Rationale and Design of the IMAD Pilot Study. J Clin Med 2020; 9:jcm9061919. [PMID: 32575439 PMCID: PMC7356934 DOI: 10.3390/jcm9061919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND agonistic autoantibodies (agAABs) against G protein-coupled receptors (GPCR) have been linked to cardiovascular disease. In dementia patients, GPCR-agAABs against the α1- and ß2-adrenoceptors (α1AR- and ß2AR) were found at a prevalence of 50%. Elimination of agAABs by immunoadsorption (IA) was successfully applied in cardiovascular disease. The IMAD trial (Efficacy of immunoadsorption for treatment of persons with Alzheimer dementia and agonistic autoantibodies against alpha1A-adrenoceptor) investigates whether the removal of α1AR-AABs by a 5-day IA procedure has a positive effect (improvement or non-deterioration) on changes of hemodynamic, cognitive, vascular and metabolic parameters in patients with suspected Alzheimer's clinical syndrome within a one-year follow-up period. METHODS the IMAD trial is designed as an exploratory monocentric interventional trial corresponding to a proof-of-concept phase-IIa study. If cognition capacity of eligible patients scores 19-26 in the Mini Mental State Examination (MMSE), patients are tested for the presence of agAABs by an enzyme-linked immunosorbent assay (ELISA)-based method, followed by a bioassay-based confirmation test, further screening and treatment with IA and intravenous immunoglobulin G (IgG) replacement. We aim to include 15 patients with IA/IgG and to complete follow-up data from at least 12 patients. The primary outcome parameter of the study is uncorrected mean cerebral perfusion measured in mL/min/100 gr of brain tissue determined by magnetic resonance imaging with arterial spin labeling after 12 months. CONCLUSION IMAD is an important pilot study that will analyze whether the removal of α1AR-agAABs by immunoadsorption in α1AR-agAAB-positive patients with suspected Alzheimer's clinical syndrome may slow the progression of dementia and/or may improve vascular functional parameters.
Collapse
Affiliation(s)
- Sylvia Stracke
- Department for Internal Medicine A, Nephrology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
- Correspondence: (S.S.); (M.D.); Tel.: +49-(0)-3834-86-80752 (S.S.); +49-(0)-3834-86-80510 (M.D.); Fax: +49-(0)-3834-86-6662 (S.S.); +49-(0)-3834-86-80502 (M.D.)
| | - Sandra Lange
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.L.); (S.L.)
| | - Sarah Bornmann
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.B.); (A.V.); (F.v.P.); (A.F.)
| | - Holger Kock
- Strategic Research Management, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Lara Schulze
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany; (L.S.); (J.K.-K.); (H.J.G.)
| | - Johanna Klinger-König
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany; (L.S.); (J.K.-K.); (H.J.G.)
| | - Susanne Böhm
- Coordinating Centre for Clinical Trials, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Antje Vogelgesang
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.B.); (A.V.); (F.v.P.); (A.F.)
| | - Felix von Podewils
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.B.); (A.V.); (F.v.P.); (A.F.)
| | - Agnes Föel
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.B.); (A.V.); (F.v.P.); (A.F.)
- German Center for Neurodegenerative Diseases (DZNE), 17475 Rostock/Greifswald, partner site Greifswald, Germany
| | - Stefan Gross
- Department of Internal Medicine B, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany;
- German Centre for Cardiovascular Research (DZHK), 17475 Greifswald, Germany
| | - Katrin Wenzel
- Berlin Cures GmbH, 13125 Berlin, Germany; (K.W.); (G.W.)
| | - Gerd Wallukat
- Berlin Cures GmbH, 13125 Berlin, Germany; (K.W.); (G.W.)
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany;
- Department of Neurology and Experimental Neurology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Alexander Dressel
- Department of Neurology, Carl-Thiem-Klinikum, 03048 Cottbus, Germany;
| | - Rudolf Kunze
- Science Office, Hessenhagen 2, 17268 Flieth-Stegelitz, Germany;
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany; (L.S.); (J.K.-K.); (H.J.G.)
- German Center for Neurodegenerative Diseases (DZNE), 17475 Rostock/Greifswald, partner site Greifswald, Germany
| | - Sönke Langner
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, 17475 Greifswald, Germany; (S.L.); (S.L.)
- Institute of Diagnostic and Interventional Radiology, University Medicine Rostock, 18057 Rostock, Germany
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany;
- German Centre for Cardiovascular Research (DZHK), 17475 Greifswald, Germany
- Correspondence: (S.S.); (M.D.); Tel.: +49-(0)-3834-86-80752 (S.S.); +49-(0)-3834-86-80510 (M.D.); Fax: +49-(0)-3834-86-6662 (S.S.); +49-(0)-3834-86-80502 (M.D.)
| |
Collapse
|
10
|
Drummond GR, Vinh A, Guzik TJ, Sobey CG. Immune mechanisms of hypertension. Nat Rev Immunol 2020; 19:517-532. [PMID: 30992524 DOI: 10.1038/s41577-019-0160-5] [Citation(s) in RCA: 239] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension affects 30% of adults and is the leading risk factor for heart attack and stroke. Traditionally, hypertension has been regarded as a disorder of two systems that are involved in the regulation of salt-water balance and cardiovascular function: the renin-angiotensin-aldosterone system (RAAS) and the sympathetic nervous system (SNS). However, current treatments that aim to limit the influence of the RAAS or SNS on blood pressure fail in ~40% of cases, which suggests that other mechanisms must be involved. This Review summarizes the clinical and experimental evidence supporting a contribution of immune mechanisms to the development of hypertension. In this context, we highlight the immune cell subsets that are postulated to either promote or protect against hypertension through modulation of cardiac output and/or peripheral vascular resistance. We conclude with an appraisal of knowledge gaps still to be addressed before immunomodulatory therapies might be applied to at least a subset of patients with hypertension.
Collapse
Affiliation(s)
- Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia.
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Tomasz J Guzik
- Department of Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland.,BHF Centre of Research Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Hohberger B, Kunze R, Wallukat G, Kara K, Mardin CY, Lämmer R, Schlötzer-Schrehardt U, Hosari S, Horn F, Munoz L, Herrmann M. Autoantibodies Activating the β2-Adrenergic Receptor Characterize Patients With Primary and Secondary Glaucoma. Front Immunol 2019; 10:2112. [PMID: 31632387 PMCID: PMC6779694 DOI: 10.3389/fimmu.2019.02112] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/21/2019] [Indexed: 01/27/2023] Open
Abstract
Recently, agonistic autoantibodies (agAAb) activating the β2-adrenergic receptor were detected in primary open-angle glaucoma (POAG) or ocular hypertension (OHT) patients and were linked to intraocular pressure (IOP) (1). The aim of the present study was to quantify β2-agAAb in the sera of glaucoma suspects and patients with primary and secondary glaucoma. Patients with OHT (n = 33), pre-perimetric POAG (pre-POAG; n = 11), POAG (n = 28), and 11 secondary OAG (SOAG) underwent ophthalmological examinations including examinations with Octopus G1 perimetry and morphometry. Twenty-five healthy individuals served as controls. Serum-derived IgG samples were analyzed for β2-agAAb using a functional bioassay. The beat-rate-increase of spontaneously beating cultured neonatal rat cardiomyocytes was monitored with 1.6 beats/15 s as cut-off. None of the sera of normal subjects showed β2-agAAb. In POAG or OHT patients increased beating rates of 4.1 ± 2.2 beats/15 s, and 3.7 ± 2.8 beats/15 s were detected (p > 0.05). Glaucoma patients with (POAG) and without perimetric (pre-POAG) defects did not differ (pre-POAG 4.4 ± 2.6 beats/15 s, POAG 4.1 ± 2.0 beats/15 s, p > 0.05). Patients with SOAG yielded mean beating rates of 4.7 ± 1.7 beats/15 s (p > 0.05). β2-agAAb were seen in 73% of OHT, 82% of pre-POAG, 82% of POAG, and 91% SOAG patients (p < 0.001). Clinical data did not correlate with beating rate (p > 0.05). The robust β2-agAAb seropositivity in patients with OHT, pre-POAG, POAG, and SOAG suggest a primary common role for β2-agAAb starting early in glaucoma pathophysiology and turned out to be a novel marker identifying all patients with increased IOP independent of glaucoma stage and entity.
Collapse
Affiliation(s)
- Bettina Hohberger
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Rudolf Kunze
- Science Office, Berlin-Buch, Campus Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Gerd Wallukat
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Katja Kara
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Y Mardin
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Lämmer
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Sami Hosari
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Folkert Horn
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Luis Munoz
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Li H, Zhang G, Zhou L, Nuss Z, Beel M, Hines B, Murphy T, Liles J, Zhang L, Kem DC, Yu X. Adrenergic Autoantibody-Induced Postural Tachycardia Syndrome in Rabbits. J Am Heart Assoc 2019; 8:e013006. [PMID: 31547749 PMCID: PMC6806023 DOI: 10.1161/jaha.119.013006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Previous studies have demonstrated that functional autoantibodies to adrenergic receptors may be involved in the pathogenesis of postural tachycardia syndrome. The objective of this study was to examine the impact of these autoantibodies on cardiovascular responses to postural changes and adrenergic orthosteric ligand infusions in immunized rabbits. Methods and Results Eight New Zealand white rabbits were coimmunized with peptides from the α1-adrenergic receptor and β1-adrenergic receptor (β1AR). Tilt test and separate adrenergic agonist infusion studies were performed on conscious animals before and after immunization and subsequent treatment with epitope-mimetic peptide inhibitors. At 6 weeks after immunization, there was a greater percent increase in heart rate upon tilting compared with preimmune baseline. No significant difference in blood pressure response to tilting was observed. The heart rate response to infusion of the β-adrenoceptor agonist isoproterenol was significantly enhanced in immunized animals, suggesting a positive allosteric effect of β1AR antibodies. In contrast, the blood pressure response to infusion of the α1-adrenergic receptor agonist phenylephrine was attenuated in immunized animals, indicating a negative allosteric effect of α1-adrenergic receptor antibodies. Injections of antibody-neutralizing peptides suppressed the postural tachycardia and reversed the altered heart rate and blood pressure responses to orthosteric ligand infusions in immunized animals at 6 and 30 weeks. Antibody production and suppression were confirmed with in vitro bioassays. Conclusions The differential allosteric effect of α1-adrenergic receptor and β1AR autoantibodies would lead to a hyperadrenergic state and overstimulation of cardiac β1AR. These data support evidence for an autoimmune basis for postural tachycardia syndrome.
Collapse
Affiliation(s)
- Hongliang Li
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Gege Zhang
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Liping Zhou
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Zachary Nuss
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Marci Beel
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Brendon Hines
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Taylor Murphy
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Jonathan Liles
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Ling Zhang
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - David C Kem
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Xichun Yu
- Department of Medicine University of Oklahoma Health Sciences Center Oklahoma City OK
| |
Collapse
|
13
|
Williams TA, Jaquin D, Burrello J, Philippe A, Yang Y, Rank P, Nirschl N, Sturm L, Hübener C, Dragun D, Bidlingmaier M, Beuschlein F, Reincke M. Diverse Responses of Autoantibodies to the Angiotensin II Type 1 Receptor in Primary Aldosteronism. Hypertension 2019; 74:784-792. [PMID: 31476909 DOI: 10.1161/hypertensionaha.119.13156] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Primary aldosteronism is a common form of endocrine hypertension mainly caused by a unilateral aldosterone-producing adenoma (APA) or bilateral adrenal hyperplasia (BAH). AT1R-Abs (autoantibodies to the angiotensin II type 1 receptor) have been reported in patients with disorders associated with hypertension. Our objective was to assess AT1R-Ab levels in patients with primary aldosteronism (APA, n=40 and BAH, n=40) relative to patients with primary hypertension (n=40), preeclampsia (n=23), and normotensive individuals (n=25). AT1R-Abs in whole sera were measured using 2 different ELISAs which gave contrasting results. A functional cell-based assay was used to quantify activation of the AT1R (angiotensin II type 1 receptor) using whole sera or affinity-purified antibodies in the absence or presence of losartan (a specific AT1R antagonist). Serum samples from all groups displayed different levels of AT1R activation with different responses to losartan. Patients with BAH displayed higher losartan-independent affinity-isolated agonistic AT1R-Ab levels compared with patients with APA (P<0.01) and with normotensive individuals (P<0.0001). In patients with APA, BAH, and primary hypertension combined, higher aldosterone-to-renin ratios and lower plasma renin concentrations were associated with higher compared with lower agonistic AT1R-Ab levels. In patients with primary aldosteronism, higher AT1R-Ab activity was associated with an increased likelihood of a diagnosis of BAH compared with APA and with the presence of adrenal hyperplasia detected by computed tomography. Taken together, these data suggest that agonistic AT1R-Abs may have a functional role in a subgroup of patients with primary aldosteronism.
Collapse
Affiliation(s)
- Tracy Ann Williams
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.).,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (T.A.W., J.B.)
| | - Diana Jaquin
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.)
| | - Jacopo Burrello
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (T.A.W., J.B.)
| | - Aurélie Philippe
- Clinic for Nephrology and Critical Care Medicine, Campus Virchow-Klinikum and Center for Cardiovascular Research, Medical Faculty of the Charité Berlin, Berlin, Germany (A.P., D.D.)
| | - Yuhong Yang
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.)
| | - Petra Rank
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.)
| | - Nina Nirschl
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.)
| | - Lisa Sturm
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.)
| | - Christoph Hübener
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Universität München, Germany (C.H.)
| | - Duska Dragun
- Clinic for Nephrology and Critical Care Medicine, Campus Virchow-Klinikum and Center for Cardiovascular Research, Medical Faculty of the Charité Berlin, Berlin, Germany (A.P., D.D.).,Berlin Institute of Health, Anna-Luisa-Karsch Str 2 10178 Berlin, Germany (D.D.)
| | - Martin Bidlingmaier
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.)
| | - Felix Beuschlein
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.).,Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Switzerland (F.B.)
| | - Martin Reincke
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (T.A.W., D.J., Y.Y., P.R., N.N., L.S., M.B., F.B., M.R.)
| |
Collapse
|
14
|
Thyrian JR, Hertel J, Schulze LN, Dörr M, Prüss H, Hempel P, Bimmler M, Kunze R, Grabe HJ, Teipel S, Hoffmann W. Prevalence and Determinants of Agonistic Autoantibodies Against α1-Adrenergic Receptors in Patients Screened Positive for Dementia: Results from the Population-Based DelpHi-Study. J Alzheimers Dis 2019; 64:1091-1097. [PMID: 30010118 DOI: 10.3233/jad-171096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is a need to assess promising biomarkers for diagnosis and treatment response in real-life settings. Despite the important role of vascular risk factors, cardiovascular biomarkers have played a minor role in dementia research. Agonistic autoantibodies (agAAB) directed against G-protein-coupled receptors (GPCR) are discussed as modulators of pathology and clinical manifestation. OBJECTIVE 1) Describe prevalence of agAAB directed against GPCR, especially agABB against α1-adrenergic receptors (α1-AR-agAAB) and agABB directed against β2-adrenergic receptors (β2-AR-agAAB) and 2) identify factors associated with agAAB in people with dementia during routine care. METHODS Blood samples and data from 95 subjects who screened positive for dementia from a primary care cohort, analyzed using an enzyme-linked immunosorbent assay (ELISA) for detecting agAAB. Sociodemographic and clinical data were assessed, and medical records checked. RESULTS In 40 (42%) samples, agAAB was detected, with n = 29 (31%) representing α1-AR-agAAB and n = 21 (22%) β2-AR-agAAB. There was no association between the presence of any antibody and a formal diagnosis of dementia. However, patients with coronary heart disease were more likely (OR = 4.23) to have α1-AR-agAAB than those without coronary heart disease. There were no associations between agAAB and age, sex, education, or cognitive impairment. DISCUSSION For the first time, we show that autoantibodies have a significant prevalence in people with dementia in a routine care setting. Previous findings were restricted to highly selective samples. We replicated the association between α1-AR-agAAB in patients with coronary heart diseases but were not able to find other factors associated with the presence of agAAB.
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW In recent years, a vast body of evidence has accumulated indicating the role of the immune system in the regulation of blood pressure and modulation of hypertensive pathology. Numerous cells of the immune system, both innate and adaptive immunity, have been indicated to play an important role in the development and maintenance of hypertension. The purpose of this review was to summarize the role of adaptive immunity in experimental models of hypertension (genetic, salt-sensitive, and Angiotensin (Ang) II induced) and in human studies. In particular, the role of T and B cells is discussed. RECENT FINDINGS In response to hypertensive stimuli such as Ang II and high salt, T cells become pro-inflammatory and they infiltrate the brain, blood vessel adventitia and periadventitial fat, heart, and the kidney. Pro-inflammatory T cell-derived cytokines such as IFN-γ and TNF-α (from CD8+ and CD4+Th1) and IL-17A (from the γδ-T cell and CD4+Th17) exacerbate hypertensive responses mediating both endothelial dysfunction and cardiac, renal, and neurodegenerative injury. The modulation of adaptive immune activation in hypertension has been attributed to target organ oxidative stress that leads to the generation of neoantigens, including isolevuglandin-modified proteins. The role of adaptive immunity is sex-specific with much more pronounced mechanisms in males than that in females. Hypertension is also associated with B cell activation and production of autoantibodies (anti-Hsp70, anti-Hsp65, anti-Hsp60, anti-AT1R, anti-α1AR, and anti-β1AR). The hypertensive responses can be inhibited by T regulatory lymphocytes (Tregs) and their anti-inflammatory IL-10. Adaptive immunity and its interface with innate mechanisms may represent valuable targets in the modulation of blood pressure, as well as hypertension-related residual risk.
Collapse
Affiliation(s)
- Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland.
- BHF Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
16
|
Shapouri-Moghaddam A, Saeed Modaghegh MH, Rahimi HR, Ehteshamfar SM, Tavakol Afshari J. Molecular mechanisms regulating immune responses in thromboangiitis obliterans: A comprehensive review. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:215-224. [PMID: 31156780 PMCID: PMC6528722 DOI: 10.22038/ijbms.2019.31119.7513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thromboangiitis obliterans (TAO) is a thrombotic-occlusive as well as an inflammatory peripheral vascular disease with unknown etiology. Recent evidence has supported the immunopathogenesis of the disease, however, the factors contributing to the altered immune function and vascular tissue inflammation are still unclear. This review was intended to collate the more current knowledge on the regulatory molecules involved in TAO from an immunoreactive perspective. The homeostasis of the immune system as well as a variety of progenitor cell populations appear to be affected during TAO and these alterations are associated with intrinsic signaling defects that are directing to an improved understanding of the crosstalk between angiogenesis and the immune system, as well as the potential of new co-targeting strategies applying both immunotherapy and angiogenic therapy.
Collapse
Affiliation(s)
- Abbas Shapouri-Moghaddam
- Immunology Research Group, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hamid Reza Rahimi
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyyed-Morteza Ehteshamfar
- Immunology Research Group, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol Afshari
- Immunology Research Group, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Werner C, Müller N, Müller UA. Agonistic autoantibodies against B2-adrenergic receptors correlating with macrovascular disease in longstanding diabetes type 2. Acta Diabetol 2019; 56:659-665. [PMID: 30770998 DOI: 10.1007/s00592-019-01296-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/02/2019] [Indexed: 10/27/2022]
Abstract
AIMS Agonistic autoantibodies directed against adrenergic, endothelin, and angiotensin receptors are known as pathogenic factors in disease-causing vascular impairments such as Buergers' disease, dilatative cardiomyopathy, dementia, and preeclampsia. Diabetes mellitus also causes micro- and macrovascular damages, but pathogenesis is still not fully understood. Following indications for a pathogenic role of the mentioned antibodies from our preliminary investigations, we investigated the prevalence in a bigger cohort of patients with longstanding diabetes with or without diabetic complications. METHODS We included 200 patients in four groups (grouping due to duration of diabetes and presence of complications) from our university polyclinic with longstanding diabetes mellitus type 2 and evaluated the prevalence of the agonistic autoantibodies using ELISA technique. RESULTS Antibodies directed against the alpha1-(39%), the first extracellular loop of the beta2-(34,5%), and the first extracellular loop of the beta1-adrenergic receptor (29,0%) were the most often detectable. With progression of diabetes and its complications, we found a decrease in the prevalence of the antibodies. Regression analyses revealed a positive association of antibodies against the first loop of the beta2-receptor and the presence of macrovascular complications. CONCLUSIONS This investigation found mid frequent prevalence of agonistic autoantibodies in patients with longstanding diabetes mellitus type 2. The association between an antibody against one epitope and the presence of macrovascular complications may indicates a pathogenic linkage. This finding is inconsistent with our preliminary data and needs further evaluation, maybe by follow-up.
Collapse
Affiliation(s)
- Christoph Werner
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | - Nicolle Müller
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany.
| | | |
Collapse
|
18
|
Lethal immunoglobulins: Autoantibodies and sudden cardiac death. Autoimmun Rev 2019; 18:415-425. [DOI: 10.1016/j.autrev.2018.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/13/2018] [Indexed: 02/08/2023]
|
19
|
Taylor EB, Wolf VL, Dent E, Ryan MJ. Mechanisms of hypertension in autoimmune rheumatic diseases. Br J Pharmacol 2019; 176:1897-1913. [PMID: 30714094 PMCID: PMC6534791 DOI: 10.1111/bph.14604] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/12/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023] Open
Abstract
Patients with autoimmune rheumatic diseases including rheumatoid arthritis and systemic lupus erythematosus have an increased prevalence of hypertension. There is now a large body of evidence showing that the immune system is a key mediator in both human primary hypertension and experimental models. Many of the proposed immunological mechanisms leading to primary hypertension are paralleled in autoimmune rheumatic disorders. Therefore, examining the link between autoimmunity and hypertension can be informative for understanding primary hypertension. This review examines the prevalent hypertension, the immune mediators that contribute to the prevalent hypertension and their impact on renal function and how the risk of hypertension is potentially influenced by common hormonal changes that are associated with autoimmune rheumatic diseases. Linked Articles This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Victoria L Wolf
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Elena Dent
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA.,G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
20
|
Spradley FT. Sympathetic nervous system control of vascular function and blood pressure during pregnancy and preeclampsia. J Hypertens 2019; 37:476-487. [PMID: 30160658 PMCID: PMC6355368 DOI: 10.1097/hjh.0000000000001901] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
: Proper vascular tone and blood pressure regulation during pregnancy are important for immediate and long-term cardiovascular health of the mother and her offspring. Preeclampsia is clinically defined as new-onset maternal hypertension accompanied by cardiovascular, renal, and/or neural abnormalities presenting in the second half of pregnancy. There is strong evidence to support that preeclampsia is mediated by attenuations in uteroplacental vascular remodeling and increases in vasoconstriction with subsequent placental ischemia/reperfusion-induced release of hypertensive substances into the maternal circulation. These include antiangiogenic and pro-inflammatory factors. There is also evidence implicating increased sympathetic nervous system activity (SNA) in this maternal disorder, but this mostly includes data correlating severity of disease with catecholamine levels and elevated muscle SNA. These measurements have not confirmed a causative role for SNA in the pathogenesis of preeclampsia. Therefore, studies are needed to provide a comprehensive understanding of SNA and its control of vascular function and blood pressure regulation during normal pregnancy in order to set the stage for exploring the mechanisms mediating the exaggerated SNA and signaling during preeclampsia. This review examines the role of SNA in control of uteroplacental vascular tone and blood pressure regulation during normal pregnancy. Furthermore, it is proposed that over-activation of the SNA contributes to altered uteroplacental vascular tone and perfusion leading to placental ischemic events and modulates the systemic vasoconstriction and hypertensive responses to soluble placenta ischemic factors. Recognizing the integrative role and importance of SNA in the pathophysiology of preeclampsia will advance our understanding of this maternal disorder.
Collapse
Affiliation(s)
- Frank T Spradley
- Department of Surgery, The University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
21
|
Abstract
Dementia in general and Alzheimer’s disease in particular is increasingly seen in association with autoimmunity being causatively or supportively involved in the pathogenesis. Besides classic autoantibodies (AABs) present in dementia patients, there is the new autoantibody class called functional autoantibodies, which is directed against G-protein coupled receptors (GPCRs; GPCR-AABs) and are seen as pathogenic players. However, less is known about dementia patients’ burden with functional autoantibodies. We present here for the first time a study analyzing the prevalence of GPCR-AABs in patients with different dementia forms such as unclassified, Lewy body, vascular and Alzheimer’s dementia. We identified the GPCR-AABs’ specific targets on the receptors and introduced a neutralization strategy for GPCR-AABs. Patients with Alzheimer’s and vascular dementia carried GPCR-AABs targeting the first loop of the alpha1- and the second loop of the beta2-adrenergic receptors (α1-AABs; β2-AABs). Nearly all vascular dementia patients also carry autoantibodies targeting the endothelin A receptor (ETA-AABs). The majority of patients with Lewy body dementia lacked any of the GPCR-AABs. In vitro, the function of the dementia-associated GPCR-AABs could be neutralized by the aptamer BC007. Due to the presence of GPCR-AABs in dementia patients mainly in those suffering from Alzheimer’s and vascular dementia, the orchestra of immune players in these dementia forms, so far preferentially represented by the classic autoantibodies, should be supplemented by functional autoantibodies. As dementia-associated functional autoantibodies could be neutralized by the aptamer BC007, the first step was taken for a new in vivo treatment option in dementia patients who were positive for GPCR-AABs.
Collapse
Affiliation(s)
| | - Harald Prüss
- Klinik für Neurologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Berlin, Germany
| | | | | |
Collapse
|
22
|
Werner C, Müller N, Müller UA. Agonistic autoantibodies against adrenergic receptors correlating with antihypertensive therapy in long-standing diabetes type 2. Acta Diabetol 2018; 55:301-303. [PMID: 29349519 DOI: 10.1007/s00592-018-1100-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/03/2018] [Indexed: 10/18/2022]
Affiliation(s)
- Christoph Werner
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany.
| | - Nicolle Müller
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | | |
Collapse
|
23
|
Autoantibodies against angiotensin and adrenergic receptors: more than a biomarker? Clin Sci (Lond) 2018; 132:127-130. [PMID: 29326280 DOI: 10.1042/cs20171485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022]
Abstract
Agonistic autoantibodies (AAs) directed against receptors of the sympathetic nervous system and the renin-angiotensin system have been suggested to contribute to cardiovascular and renal disease, in particular hypertension, preeclampsia, and graft failure in kidney transplantation patients. Consequently, they are now also being studied as biomarker for these conditions. This commentary summarizes our current understanding of these AAs, critically discussing whether they truly act as agonist, and focusing on the wide array of assays that are currently used for their quantification.
Collapse
|
24
|
Autoantibodies against AT1 and α1-adrenergic receptors predict arterial stiffness progression in normotensive subjects over a 5-year period. Clin Sci (Lond) 2017; 131:2947-2957. [PMID: 29097625 DOI: 10.1042/cs20171305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 01/08/2023]
Abstract
Arterial stiffness is an independent indicator of cardiovascular risk. Autoantibodies (AAs) against angiotensin AT1 receptor (AT1-AAs) and α1-adrenergic receptor (α1-AAs) are important in the pathogenesis of hypertension. We identified the types of AT1-AAs and α1-AAs in normotensive subjects, with the aim of determining whether these antibodies predict aortic stiffness progression. Carotid–femoral pulse wave velocity (cf-PWV) was used to measure aortic stiffness. Overall, 816 subjects (71% of those invited) underwent a medical examination and evaluation of aortic stiffness. The types of AT1-AAs and α1-AAs were measured at baseline. Meanwhile, plasma renin, angiotensin II (Ang II), and norepinephrine (NE) concentrations were measured at baseline and follow-up. Baseline mean cf-PWV was 9.90 ± 0.84 m/s and follow-up was 10.51 ± 1.12 m/s. The annualized ΔPWV was 0.12 ± 0.08 m/s/year. At the end of follow-up, 129 normotensive subjects developed hypertension and 144 subjects had PWV progression. After adjustment for covariates, AA type was independently associated with ΔPWV, annualized ΔPWV, and abnormal PWV. In our study, the risk of developing hypertension (RR =2.028, 95% CI: 1.227–3.351, P=0.006) and PWV progression (RR =2.910, 95% CI: 1.612–5.253, P<0.001) in AA-positive subjects was significantly higher than that in AA-negative subjects. Receiver operating characteristic (ROC) curve showed AA had an identify power to discriminate subjects with or without PWV and hypertension progression. We have shown for the first time that the types of A1-AAs and α1-AAs are independent predictors for aortic stiffness progression in normotensive subjects. Our data collectively support the utility of these AAs as potential markers of aortic stiffness.
Collapse
|
25
|
Zhao LS, Lin YY, Liu Y, Xu CY, Liu Y, Bai WW, Tan XY, Li DZ, Xu JL. Doxazosin attenuates renal matrix remodeling mediated by anti-α1-adrenergic receptor antibody in a rat model of diabetes mellitus. Exp Ther Med 2017; 14:2543-2553. [PMID: 28962193 PMCID: PMC5609287 DOI: 10.3892/etm.2017.4827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 03/17/2017] [Indexed: 01/06/2023] Open
Abstract
Diabetic nephropathy is a major complication of diabetes mellitus (DM). Recent studies suggest that immunological mechanisms have a key role in the pathogenesis of DM, therefore these mechanisms may be important targets for diabetes therapy. The present study evaluated the effects of anti-α1-adrenergic receptor antibody (α1-R Ab) mediation and doxazosin treatment in a rat model of DM. It was observed that levels of 24-h urinary protein, serum creatinine and transforming growth factor-β1 in DM were significantly increased after α1-R Ab mediation (all P<0.05). In addition, electron microscopy identified severe damage in the renal tissue microstructures of DM rats following α1-R Ab mediation, while only mild abnormalities were observed in that of healthy rats mediated with α1-R Ab and of untreated DM rats. No marked abnormalities were observed in the renal tissue of healthy blank controls. Furthermore, in DM rats treated with α1-R Ab mediation + doxazosin intervention, the expression of TGF-β1 significantly decreased, and renal functions and renal matrix remodeling were significantly improved, relative to untreated DM controls (P<0.01). These results suggest that α1-R Ab may be involved in renal matrix remodeling during DM, and that kidney protection during DM may be achieved through treatment with corresponding receptor antagonists.
Collapse
|
26
|
Liu C, Kellems RE, Xia Y. Inflammation, Autoimmunity, and Hypertension: The Essential Role of Tissue Transglutaminase. Am J Hypertens 2017; 30:756-764. [PMID: 28338973 PMCID: PMC5861548 DOI: 10.1093/ajh/hpx027] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/09/2017] [Indexed: 12/19/2022] Open
Abstract
Inflammatory cytokines cause hypertension when introduced into animals. Additional evidence indicates that cytokines induce the production of autoantibodies that activate the AT1 angiotensin receptor (AT1R). Extensive evidence shows that these autoantibodies, termed AT1-AA, contribute to hypertension. We review here recent studies showing that cytokine-induced hypertension and AT1-AA production require the ubiquitous enzyme, tissue transglutaminase (TG2). We consider 3 mechanisms by which TG2 may contribute to hypertension. (i) One involves the posttranslational modification (PTM) of AT1Rs at a glutamine residue that is present in the epitope sequence (AFHYESQ) recognized by AT1-AA. (ii) Another mechanism by which TG2 may contribute to hypertension is by PTM of AT1Rs at glutamine 315. Modification at this glutamine prevents ubiquitination-dependent proteasome degradation and allows AT1Rs to accumulate. Increased AT1R abundance is likely to account for increased sensitivity to Ang II activation and in this way contribute to hypertension. (iii) The increased TG2 produced as a result of elevated inflammatory cytokines is likely to contribute to vascular stiffness by modification of intracellular contractile proteins or by crosslinking vascular proteins in the extracellular matrix. This process, termed inward remodeling, results in reduced vascular lumen, vascular stiffness, and increased blood pressure. Based on the literature reviewed here, we hypothesize that TG2 is an essential participant in cytokine-induced hypertension. From this perspective, selective TG2 inhibitors have the potential to be pharmacologic weapons in the fight against hypertension.
Collapse
Affiliation(s)
- Chen Liu
- Department of Biochemistry and Molecular Biology, McGovern Medical School of the University of Texas at Houston, Houston, Texas, USA
| | - Rodney E. Kellems
- Department of Biochemistry and Molecular Biology, McGovern Medical School of the University of Texas at Houston, Houston, Texas, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, McGovern Medical School of the University of Texas at Houston, Houston, Texas, USA
| |
Collapse
|
27
|
Ruzieh M, Batizy L, Dasa O, Oostra C, Grubb B. The role of autoantibodies in the syndromes of orthostatic intolerance: a systematic review. SCAND CARDIOVASC J 2017; 51:243-247. [DOI: 10.1080/14017431.2017.1355068] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Mohammed Ruzieh
- Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | - Lillian Batizy
- College of Medicine, University of Toledo, Toledo, OH, USA
| | - Osama Dasa
- Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | - Carson Oostra
- Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | - Blair Grubb
- Department of Cardiovascular Medicine, Syncope and Autonomic Dysfunction Center, University of Toledo, Toledo, OH, USA
| |
Collapse
|
28
|
|
29
|
Shpakov AO, Zharova OA, Derkach KV. Antibodies to extracellular regions of G protein-coupled receptors and receptor tyrosine kinases as one of the causes of autoimmune diseases. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s1234567817020021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Taylor EB, Ryan MJ. Immunosuppression With Mycophenolate Mofetil Attenuates Hypertension in an Experimental Model of Autoimmune Disease. J Am Heart Assoc 2017; 6:JAHA.116.005394. [PMID: 28242635 PMCID: PMC5524041 DOI: 10.1161/jaha.116.005394] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder that predominantly affects women and is associated with prevalent hypertension, renal injury, and cardiovascular disease. Immune system dysfunction is recognized as an important factor in the pathogenesis of hypertension. We recently showed that preventing autoimmunity prevents the development of hypertension in an experimental model of SLE (female NZBWF1 mice). The present study tests the hypothesis that mycophenolate mofetil (MMF), an immunosuppressive therapy used clinically to treat SLE by depleting proliferating B and T lymphocytes, can improve blood pressure control. Methods and Results Female SLE and control (NZW/LacJ) mice were treated daily for 8 weeks with 60 mg/kg MMF. Circulating CD45R+ B cells were lower in MMF‐treated SLE mice after 4 weeks of treatment, but neither CD4+ nor CD8+ T cells were reduced by MMF. Plasma anti–double‐stranded DNA IgG autoantibodies, a marker of SLE disease activity, were higher in SLE mice compared with controls and were lower in SLE mice after 8 weeks of MMF. Mean arterial pressure was elevated in SLE mice compared with controls and lower in SLE mice treated with MMF compared with vehicle‐treated SLE mice. MMF also reduced both renal injury (urinary albumin excretion and glomerulosclerosis) and the infiltration of CD45R+ B cells and CD3+CD4+ T cells in kidneys from mice with SLE. Conclusions These data suggest that MMF selectively depleted CD45R+ B cells and lowered subsequent autoantibody production, furthering the concept that autoantibodies mechanistically contribute to the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
31
|
Hu Z, Liu F, Li M, He J, Huang J, Rao DC, Hixson JE, Gu C, Kelly TN, Chen S, Gu D, Yang X. Associations of Variants in the CACNA1A and CACNA1C Genes With Longitudinal Blood Pressure Changes and Hypertension Incidence: The GenSalt Study. Am J Hypertens 2016; 29:1301-1306. [PMID: 27418245 DOI: 10.1093/ajh/hpw070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/13/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND We aimed to examine the associations of voltage-dependent calcium-channel genes CACNA1A and CACNA1C with blood pressure (BP) changes and hypertension incidence in a longitudinal family study. METHODS A total of 1,768 Han Chinese participants from the Genetic Epidemiology Network of Salt Sensitivity (GenSalt) follow-up study were eligible for the current study. Nine BP measurements were obtained at baseline and each follow-up visit using a random-zero sphygmomanometer. Mixed-effect models were used to assess additive associations of 176 tag single-nucleotide polymorphisms (SNPs) in CACNA1A and CACNA1C with longitudinal BP changes and hypertension incidence. The truncated product method was used for gene-based analysis. The Bonferroni correction was used for adjustment of multiple testing. RESULTS During an average of 7.2 years of follow-up, 512 (32.1%) participants developed hypertension. CACNA1A SNP rs8182538 was significantly associated with longitudinal diastolic BP (DBP) change after Bonferroni correction ( Pinteraction = 9.90×10 -5 ), with mean DBP increases of 0.85, 1.03, and 1.19mm Hg per year for participants with genotypes C/C , C/T , and T/T , respectively. A similar trend was observed for the association of rs8182538 with systolic BP (SBP) change. In the gene-based analysis, CACNA1A and CACNA1C were significantly associated with DBP change ( P = 2.0×10 -5 ) and SBP change ( P = 1.4×10 -4 ) after Bonferroni correction, respectively. The gene-based associations remained significant after removing rs8182538 within CACNA1A and rs758116 within CACNA1C in sensitivity analysis. CONCLUSIONS Our findings indicated that CACNA1A and CACNA1C might contribute to BP changes over time in Han Chinese population. Further replication of these findings is warranted.
Collapse
Affiliation(s)
- Zunsong Hu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Fangchao Liu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Mengting Li
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jianfeng Huang
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine , St. Louis, Missouri , USA
| | - James E Hixson
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health , Houston, Texas , USA
| | - Charles Gu
- Division of Biostatistics, Washington University School of Medicine , St. Louis, Missouri , USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine , New Orleans, Louisiana , USA
| | - Shufeng Chen
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Dongfeng Gu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Xueli Yang
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| |
Collapse
|
32
|
McCarthy CG, Wenceslau CF, Goulopoulou S, Ogbi S, Matsumoto T, Webb RC. Autoimmune therapeutic chloroquine lowers blood pressure and improves endothelial function in spontaneously hypertensive rats. Pharmacol Res 2016; 113:384-394. [PMID: 27639600 DOI: 10.1016/j.phrs.2016.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/22/2016] [Accepted: 09/08/2016] [Indexed: 12/18/2022]
Abstract
It has been suggested that hypertension results from a loss of immunological tolerance and the resulting autoimmunity may be an important underlying factor of its pathogenesis. This stems from the observations that many of the features involved in autoimmunity are also implicated in hypertension. Furthermore, the underlying presence of hypertension and cardiovascular disease are frequently observed in patients with autoimmune diseases. Antimalarial agents such as chloroquine are generally among the first line treatment options for patients with autoimmune diseases; however, whether they can improve a hypertensive phenotype in a genetic model of essential hypertension remains to be clarified. Therefore, we hypothesized that chloroquine treatment would improve endothelial function and lower blood pressure in spontaneously hypertensive rats (SHR). We treated adult SHR and Wistar-Kyoto rats (12 weeks old), as well as a group of young SHR (5 weeks old), with chloroquine (40mg/kg/day via intraperitoneal injection) for 21 days. Chloroquine lowered blood pressure in adult SHR, but did not impede the development of high blood pressure in young SHR. In isolated mesenteric resistance arteries from SHR of both ages, chloroquine treatment inhibited cyclooxygenase-dependent contraction to acetylcholine, lowered vascular and systemic generation of reactive oxygen species, and improved nitric oxide bioavailability. Overall, these data reveal the anti-hypertensive mechanisms of chloroquine in the vasculature, which may be important for lowering risk of cardiovascular disease in patients with autoimmune diseases. Furthermore, it adds to the growing body of evidence suggesting that autoimmunity underlies hypertension.
Collapse
Affiliation(s)
| | | | - Styliani Goulopoulou
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Safia Ogbi
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Tokyo, Japan
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, GA, USA
| |
Collapse
|
33
|
Oh YS, Appel LJ, Galis ZS, Hafler DA, He J, Hernandez AL, Joe B, Karumanchi SA, Maric-Bilkan C, Mattson D, Mehta NN, Randolph G, Ryan M, Sandberg K, Titze J, Tolunay E, Toney GM, Harrison DG. National Heart, Lung, and Blood Institute Working Group Report on Salt in Human Health and Sickness: Building on the Current Scientific Evidence. Hypertension 2016; 68:281-8. [PMID: 27324228 DOI: 10.1161/hypertensionaha.116.07415] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/27/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Young S Oh
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.).
| | - Lawrence J Appel
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Zorina S Galis
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - David A Hafler
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Jiang He
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Amanda L Hernandez
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Bina Joe
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - S Ananth Karumanchi
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Christine Maric-Bilkan
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - David Mattson
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Nehal N Mehta
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Gwendolyn Randolph
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Michael Ryan
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Kathryn Sandberg
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Jens Titze
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Eser Tolunay
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - Glenn M Toney
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| | - David G Harrison
- From the Division of Cardiovascular Sciences (Y.S.O, Z.S.G., C.M.-B., E.T.) and Division of Intramural Research (N.N.M.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Johns Hopkins University, Baltimore, MD (L.J.A.); Department of Neurology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT (A.L.H., D.A.H.); Department of Epidemiology, Tulane University, New Orleans, LA (J.H.); Department of Physiology and Pharmacology, University of Toledo, OH (B.J.); Department of Medicine and Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.A.K.); Department of Physiology, Medical College of Wisconsin, Milwaukee (D.M.); Department of Pathology and Immunology, Washington University in St. Louis, MO (G.R.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (M.R.); Department of Medicine, Georgetown University, Washington, DC (K.S.); Department of Medicine, Vanderbilt University, Nashville, TN (J.T., D.G.H.); and Department of Physiology, University of Texas Health Science Center at San Antonio (G.M.T.)
| |
Collapse
|
34
|
Hempel P, Heinig B, Jerosch C, Decius I, Karczewski P, Kassner U, Kunze R, Steinhagen-Thiessen E, Bimmler M. Immunoadsorption of Agonistic Autoantibodies Against α1-Adrenergic Receptors in Patients With Mild to Moderate Dementia. Ther Apher Dial 2016; 20:523-529. [PMID: 27096216 DOI: 10.1111/1744-9987.12415] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 12/21/2022]
Abstract
Dementia has been shown to be associated with agonistic autoantibodies. The deleterious action of autoantibodies on the α1-adrenergic receptor for brain vasculature has been demonstrated in animal studies. In the current study, 169 patients with dementia were screened for the presence of agonistic autoantibodies. 47% of patients suffering from mild to moderate Alzheimer's disease and/or vascular dementia carried these autoantibodies. Eight patients positive for autoantibodies underwent immunoadsorption. Patients treated on four consecutive days were subsequently negative for autoantibodies and displayed stabilization of cognitive and mental condition during 12-18 months' follow-up. In patients treated for 2-3 days, autoantibodies were reduced by only 78%. They suffered a rebound of autoantibodies during follow-up, benefited from immunoadsorption too, but their mental parameters worsened. We provide first data on the clinical relevance of agonistic autoantibodies in dementia and show that immunoadsorption is safe and efficient in removing autoantibodies with overall benefits for patients.
Collapse
Affiliation(s)
| | - Bente Heinig
- Charité - University Medicine Berlin, Berlin, Germany
| | | | - Imke Decius
- Charité - University Medicine Berlin, Berlin, Germany
| | | | | | | | | | - Marion Bimmler
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
35
|
Taylor EB, Ryan MJ. Understanding mechanisms of hypertension in systemic lupus erythematosus. Ther Adv Cardiovasc Dis 2016; 11:1753944716637807. [PMID: 26985016 PMCID: PMC5065379 DOI: 10.1177/1753944716637807] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder that predominately affects women of reproductive age. Hypertension is an important cardiovascular risk factor that is prevalent in this patient population. Despite the high incidence of hypertension in women with SLE, the pathophysiological mechanisms underlying the development of hypertension remain poorly understood. This review will focus on disease-related factors, including inflammation, autoantibodies, and sex hormones that may contribute to hypertension in patients with SLE. In addition, we will highlight studies performed by our laboratory using the female NZBWF1 (F1 hybrid of New Zealand Black and New Zealand White strains) mouse model, a spontaneous model of SLE that mimics human disease and develops hypertension and renal injury. Specifically, using female NZBWF1 mice, we have demonstrated that multiple factors contribute to the pathogenesis of hypertension, including the inflammatory cytokine, tumor necrosis factor (TNF)-α, oxidative stress, as well as B-cell hyperactivity and autoantibody production.
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| |
Collapse
|
36
|
Liu C, Luo R, Elliott SE, Wang W, Parchim NF, Iriyama T, Daugherty PS, Blackwell SC, Sibai BM, Kellems RE, Xia Y. Elevated Transglutaminase Activity Triggers Angiotensin Receptor Activating Autoantibody Production and Pathophysiology of Preeclampsia. J Am Heart Assoc 2015; 4:e002323. [PMID: 26675250 PMCID: PMC4845265 DOI: 10.1161/jaha.115.002323] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/07/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Preeclampsia (PE) is a life-threatening hypertensive disorder of pregnancy associated with autoantibodies, termed AT1-AA, that activate the AT1 angiotensin receptor. Although the pathogenic nature of these autoantibodies has been extensively studied, little is known about the molecular cause of their generation. METHODS AND RESULTS Here we show that tissue transglutaminase (TG2), an enzyme that conducts posttranslational modification of target proteins, directly modified the 7-amino acid (7-aa) epitope peptide that localizes to the second extracellular loop of the AT1 receptor. These findings led us to further discover that plasma transglutaminase activity was induced and contributed to the production of AT1-AA and disease development in an experimental model of PE induced by injection of LIGHT, a tumor necrosis factor superfamily member. Key features of PE were regenerated by adoptive transfer of purified IgG from LIGHT-injected pregnant mice and blocked by the 7-amino acid epitope peptide. Translating our mouse research to humans, we found that plasma transglutaminase activity was significantly elevated in PE patients and was positively correlated with AT1-AA levels and PE features. CONCLUSIONS Overall, we provide compelling mouse and human evidence that elevated transglutaminase underlies AT1-AA production in PE and highlight novel pathogenic biomarkers and innovative therapeutic possibilities for the disease.
Collapse
Affiliation(s)
- Chen Liu
- Departments of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonTX
| | - Renna Luo
- Departments of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonTX
- Nephrology DepartmentXiangya HospitalHunanChina
- Department of NephrologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Serra E. Elliott
- Department of Chemical EngineeringUniversity of CaliforniaSanta BarbaraCA
| | - Wei Wang
- Departments of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonTX
- Nephrology DepartmentXiangya HospitalHunanChina
| | - Nicholas F. Parchim
- Departments of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonTX
| | - Takayuki Iriyama
- Departments of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonTX
- Department of Obstetrics and GynecologyUniversity of TokyoJapan
| | | | - Sean C. Blackwell
- Department of Obstetrics, Gynecology and Reproductive SciencesThe University of Texas Health Science Center at HoustonTX
| | - Baha M. Sibai
- Department of Obstetrics, Gynecology and Reproductive SciencesThe University of Texas Health Science Center at HoustonTX
| | - Rodney E. Kellems
- Departments of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonTX
- The University of Texas Graduate School of Biomedical Sciences at HoustonTX
| | - Yang Xia
- Departments of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonTX
- The University of Texas Graduate School of Biomedical Sciences at HoustonTX
- Department of Chemical EngineeringUniversity of CaliforniaSanta BarbaraCA
| |
Collapse
|
37
|
Williams TA, Mulatero P, Bidlingmaier M, Beuschlein F, Reincke M. Genetic and potential autoimmune triggers of primary aldosteronism. Hypertension 2015; 66:248-53. [PMID: 26056334 DOI: 10.1161/hypertensionaha.115.05643] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/14/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Tracy Ann Williams
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany (T.A.W., M.B., F.B., M.R.); and Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy (T.A.W., P.M.).
| | - Paolo Mulatero
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany (T.A.W., M.B., F.B., M.R.); and Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy (T.A.W., P.M.)
| | - Martin Bidlingmaier
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany (T.A.W., M.B., F.B., M.R.); and Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy (T.A.W., P.M.)
| | - Felix Beuschlein
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany (T.A.W., M.B., F.B., M.R.); and Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy (T.A.W., P.M.)
| | - Martin Reincke
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany (T.A.W., M.B., F.B., M.R.); and Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy (T.A.W., P.M.).
| |
Collapse
|
38
|
Jahns R, Boege F. Questionable Validity of Peptide-Based ELISA Strategies in the Diagnostics of Cardiopathogenic Autoantibodies That Activate G-Protein-Coupled Receptors. Cardiology 2015; 131:149-50. [DOI: 10.1159/000376546] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 01/22/2015] [Indexed: 11/19/2022]
|
39
|
Li H, Kem DC, Zhang L, Huang B, Liles C, Benbrook A, Gali H, Veitla V, Scherlag BJ, Cunningham MW, Yu X. Novel retro-inverso peptide inhibitor reverses angiotensin receptor autoantibody-induced hypertension in the rabbit. Hypertension 2015; 65:793-9. [PMID: 25691619 DOI: 10.1161/hypertensionaha.114.05037] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activating autoantibodies to the angiotensin II type 1 receptor (AT1R) have been implicated in hypertensive disorders. We investigated whether AT1R antibodies produced in immunized rabbits will activate AT1R and contribute to hypertension by a direct contractile effect on the vasculature and whether they can be blocked by a novel decoy peptide. A multiple antigenic peptide containing the AT1R epitope AFHYESQ, which is the receptor-binding epitope of AT1R-activating autoantibodies, was used to immunize 6 rabbits. AT1R antibody activity was analyzed in AT1R-transfected cells, and their contractile effects were assayed using isolated perfused rat cremaster resistance arterioles. A retro-inverso D-amino acid epitope-mimetic peptide was tested for AT1R antibody inhibition in vitro and in vivo. All immunized animals produced high AT1R antibody titers and developed elevated blood pressure. No changes in measured blood chemistry values were observed after immunization. Rabbit anti-AT1R sera induced significant AT1R activation in transfected cells and vasoconstriction in the arteriole assay, both of which were blocked by losartan and the retro-inverso D-amino acid peptide. A single intravenous bolus injection of the retro-inverso d-amino acid peptide (1 mg/kg) into immunized rabbits dropped the mean arterial pressure from 122±11 to 82±6 mm Hg. Rabbit anti-AT1R sera partially suppressed angiotensin II-induced contraction of isolated rat cremaster arterioles, and the pressor response to angiotensin II infusion was attenuated in immunized animals. In conclusion, AT1R-activating autoantibodies and the retro-inverso d-amino acid peptide, respectively, have important etiologic and therapeutic implications in hypertensive subjects who harbor these autoantibodies.
Collapse
Affiliation(s)
- Hongliang Li
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - David C Kem
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Ling Zhang
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Bing Huang
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Campbell Liles
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Alexandria Benbrook
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Hariprasad Gali
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Vineet Veitla
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Benjamin J Scherlag
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Madeleine W Cunningham
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City
| | - Xichun Yu
- From the Department of Medicine and the Heart Rhythm Institute (H.L., D.C.K., L.Z., B.H., C.L., A.B., V.V., B.J.S., X.Y.), and Departments of Pharmaceutical Sciences (H.G.) and Microbiology and Immunology (M.W.C.), University of Oklahoma Health Sciences Center and Veterans Affairs Medical Center, Oklahoma City.
| |
Collapse
|
40
|
van der Westhuizen ET, Valant C, Sexton PM, Christopoulos A. Endogenous Allosteric Modulators of G Protein–Coupled Receptors. J Pharmacol Exp Ther 2015; 353:246-60. [DOI: 10.1124/jpet.114.221606] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
41
|
Michel MC, Seifert R. Selectivity of pharmacological tools: implications for use in cell physiology. A review in the theme: Cell signaling: proteins, pathways and mechanisms. Am J Physiol Cell Physiol 2015; 308:C505-20. [PMID: 25631871 DOI: 10.1152/ajpcell.00389.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/24/2015] [Indexed: 01/08/2023]
Abstract
Pharmacological inhibitors are frequently used to identify the receptors, receptor subtypes, and associated signaling pathways involved in physiological cell responses. Based on the effects of such inhibitors conclusions are drawn about the involvement of their assumed target or lack thereof. While such inhibitors can be useful tools for a better physiological understanding, their uncritical use can lead to incorrect conclusions. This article reviews the concept of inhibitor selectivity and its implication for cell physiology. Specifically, we discuss the implications of using inhibitor vs. activator approaches, issues of direct vs. indirect pathway modulation, implications of inverse agonism and biased signaling, and those of orthosteric vs. allosteric, competitive vs. noncompetitive, and reversible vs. irreversible inhibition. Additional problems can result from inconsistent estimates of inhibitor potency and differences in potency between cell-free systems and intact cells. These concepts are illustrated by several examples of inhibitors displaying affinity for related but distinct targets or even unrelated targets. Of note, many of the issues being addressed are also applicable to genetic inhibition strategies. The main practical conclusion following from these concepts is that investigators should be critical in the choice of inhibitor, its concentrations, and its mode of application. When this advice is adhered to, small-molecule pharmacological inhibitors can be important experimental tools in the hand of physiologists.
Collapse
Affiliation(s)
- Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany; and
| | - Roland Seifert
- Department of Pharmacology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
42
|
Autoantibodies targeting AT1 receptor from patients with acute coronary syndrome upregulate proinflammatory cytokines expression in endothelial cells involving NF-κB pathway. J Immunol Res 2014; 2014:342693. [PMID: 25762441 PMCID: PMC4266766 DOI: 10.1155/2014/342693] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/24/2014] [Accepted: 10/26/2014] [Indexed: 12/25/2022] Open
Abstract
Our study intended to prove whether agonistic autoantibodies to angiotensin II type 1 receptor (AT1-AAs) exist in patients with coronary heart disease (CHD) and affect the human endothelial cell (HEC) by upregulating proinflammatory cytokines expression involved in NF-κB pathway. Antibodies were determined by chronotropic responses of cultured neonatal rat cardiomyocytes coupled with receptor-specific antagonists (valsartan and AT1-EC2) as described previously. Interleukin-6 (IL-6), vascular cell adhesion molecule-1 (VCAM-1), and monocyte chemotactic protein-1 (MCP-1) expression were improved at both mRNA and protein levels in HEC, while NF-κB in the DNA level was improved detected by electrophoretic mobility shift assays (EMSA). These improvements could be inhibited by specific AT1 receptor blocker valsartan, NF-κB blocker pyrrolidine dithiocarbamate (PDTC), and specific short peptides from the second extracellular loop of AT1 receptor. These results suggested that AT1-AAs, via the AT1 receptor, induce expression of proinflammatory cytokines involved in the activation of NF-κB. AT1-AAs may play a great role in the pathogenesis of the acute coronary syndrome by mediating vascular inflammatory effects involved in the NF-κB pathway.
Collapse
|
43
|
Longstanding complex regional pain syndrome is associated with activating autoantibodies against alpha-1a adrenoceptors. Pain 2014; 155:2408-17. [DOI: 10.1016/j.pain.2014.09.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/09/2014] [Accepted: 09/16/2014] [Indexed: 11/22/2022]
|
44
|
Abstract
A large number of investigations have demonstrated the participation of the immune system in the pathogenesis of hypertension. Studies focusing on macrophages and Toll-like receptors have documented involvement of the innate immunity. The requirements of antigen presentation and co-stimulation, the critical importance of T cell-driven inflammation, and the demonstration, in specific conditions, of agonistic antibodies directed to angiotensin II type 1 receptors and adrenergic receptors support the role of acquired immunity. Experimental findings support the concept that the balance between T cell-induced inflammation and T cell suppressor responses is critical for the regulation of blood pressure levels. Expression of neoantigens in response to inflammation, as well as surfacing of intracellular immunogenic proteins, such as heat shock proteins, could be responsible for autoimmune reactivity in the kidney, arteries, and central nervous system. Persisting, low-grade inflammation in these target organs may lead to impaired pressure natriuresis, an increase in sympathetic activity, and vascular endothelial dysfunction that may be the cause of chronic elevation of blood pressure in essential hypertension.
Collapse
Affiliation(s)
- Bernardo Rodríguez-Iturbe
- Hospital Universitario y Universidad del Zulia, Maracaibo, Venezuela; Instituto Venezolano de Investigaciones Científicas-Zulia, Maracaibo, Venezuela;
| | - Héctor Pons
- Hospital Universitario y Universidad del Zulia, Maracaibo, Venezuela
| | - Yasmir Quiroz
- Instituto Venezolano de Investigaciones Científicas-Zulia, Maracaibo, Venezuela
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, Colorado
| |
Collapse
|
45
|
Bornholz B, Roggenbuck D, Jahns R, Boege F. Diagnostic and therapeutic aspects of β1-adrenergic receptor autoantibodies in human heart disease. Autoimmun Rev 2014; 13:954-62. [DOI: 10.1016/j.autrev.2014.08.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 06/16/2014] [Indexed: 01/19/2023]
|
46
|
Pohlmann A, Karczewski P, Ku MC, Dieringer B, Waiczies H, Wisbrun N, Kox S, Palatnik I, Reimann HM, Eichhorn C, Waiczies S, Hempel P, Lemke B, Niendorf T, Bimmler M. Cerebral blood volume estimation by ferumoxytol-enhanced steady-state MRI at 9.4 T reveals microvascular impact of α1 -adrenergic receptor antibodies. NMR IN BIOMEDICINE 2014; 27:1085-1093. [PMID: 25060359 DOI: 10.1002/nbm.3160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/11/2014] [Accepted: 06/05/2014] [Indexed: 06/03/2023]
Abstract
Cerebrovascular abnormality is frequently accompanied by cognitive dysfunctions, such as dementia. Antibodies against the α1 -adrenoceptor (α1 -AR) can be found in patients with Alzheimer's disease with cerebrovascular disease, and have been shown to affect the larger vessels of the brain in rodents. However, the impact of α1 -AR antibodies on the cerebral vasculature remains unclear. In the present study, we established a neuroimaging method to measure the relative cerebral blood volume (rCBV) in small rodents with the ultimate goal to detect changes in blood vessel density and/or vessel size induced by α1 -AR antibodies. For this purpose, mapping of R2 * and R2 was performed using MRI at 9.4 T, before and after the injection of intravascular iron oxide particles (ferumoxytol). The change in the transverse relaxation rates (ΔR2 *, ΔR2 ) showed a significant rCBV decrease in the cerebrum, cortex and hippocampus of rats (except hippocampal ΔR2 ), which was more pronounced for ΔR2 * than for ΔR2 . Immunohistological analyses confirmed that the α1 -AR antibody induced blood vessel deficiencies. Our findings support the hypothesis that α1 -AR antibodies lead to cerebral vessel damage throughout the brain, which can be monitored by MRI-derived rCBV, a non-invasive neuroimaging method. This demonstrates the value of rCBV estimation by ferumoxytol-enhanced MRI at 9.4 T, and further underlines the significance of this antibody in brain diseases involving vasculature impairments, such as dementia.
Collapse
Affiliation(s)
- Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mathis KW, Wallace K, Flynn ER, Maric-Bilkan C, LaMarca B, Ryan MJ. Preventing autoimmunity protects against the development of hypertension and renal injury. Hypertension 2014; 64:792-800. [PMID: 25024282 DOI: 10.1161/hypertensionaha.114.04006] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Several studies suggest a link between autoimmunity and essential hypertension in humans. However, whether autoimmunity can drive the development of hypertension remains unclear. The autoimmune disease systemic lupus erythematosus is characterized by autoantibody production, and the prevalence of hypertension is increased markedly in this patient population compared with normal healthy women. We hypothesized that preventing the development of autoimmunity would prevent the development of hypertension in a mouse model of lupus. Female lupus (NZBWF1) and control mice (NZW) were treated weekly with anti-CD20 or immunoglobulin G antibodies (both 10 mg/kg, IV) starting at 20 weeks of age for 14 weeks. Anti-CD20 therapy markedly attenuated lupus disease progression as evidenced by reduced CD45R+ B cells and lower double-stranded DNA autoantibody activity. In addition, renal injury in the form of urinary albumin, glomerulosclerosis, and tubulointerstitial fibrosis, as well as tubular injury (indicated by renal cortical expression of neutrophil gelatinase-associated lipocalin) was prevented by anti-CD20 therapy in lupus mice. Finally, lupus mice treated with anti-CD20 antibody did not develop hypertension. The protection against the development of hypertension was associated with lower renal cortical tumor necrosis factor-α expression, a cytokine that has been previously reported by us to contribute to the hypertension in this model, as well as renal cortical monocyte chemoattractant protein-1 expression and circulating T cells. These data suggest that the development of autoimmunity and the resultant increase in renal inflammation are an important underlying factor in the prevalent hypertension that occurs during systemic lupus erythematosus.
Collapse
Affiliation(s)
- Keisa W Mathis
- From the Departments of Physiology and Biophysics (K.W.M., E.R.F., C.M.-B., M.J.R.), Obstetrics and Gynecology (K.W.), and Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center, Jackson
| | - Kedra Wallace
- From the Departments of Physiology and Biophysics (K.W.M., E.R.F., C.M.-B., M.J.R.), Obstetrics and Gynecology (K.W.), and Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center, Jackson
| | - Elizabeth R Flynn
- From the Departments of Physiology and Biophysics (K.W.M., E.R.F., C.M.-B., M.J.R.), Obstetrics and Gynecology (K.W.), and Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center, Jackson
| | - Christine Maric-Bilkan
- From the Departments of Physiology and Biophysics (K.W.M., E.R.F., C.M.-B., M.J.R.), Obstetrics and Gynecology (K.W.), and Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center, Jackson
| | - Babbette LaMarca
- From the Departments of Physiology and Biophysics (K.W.M., E.R.F., C.M.-B., M.J.R.), Obstetrics and Gynecology (K.W.), and Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center, Jackson
| | - Michael J Ryan
- From the Departments of Physiology and Biophysics (K.W.M., E.R.F., C.M.-B., M.J.R.), Obstetrics and Gynecology (K.W.), and Pharmacology and Toxicology (B.L.), University of Mississippi Medical Center, Jackson.
| |
Collapse
|
48
|
Antibodies in the pathogenesis of hypertension. BIOMED RESEARCH INTERNATIONAL 2014; 2014:504045. [PMID: 25050352 PMCID: PMC4090532 DOI: 10.1155/2014/504045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/21/2014] [Accepted: 06/04/2014] [Indexed: 12/22/2022]
Abstract
It has long been known that circulating levels of IgG and IgM antibodies are elevated in patients with essential and pregnancy-related hypertension. Recent studies indicate these antibodies target, and in many cases activate, G-protein coupled receptors and ion channels. Prominent among these protein targets are AT1 receptors, α1-adrenoceptors, β1-adrenoceptors, and L-type voltage operated Ca2+ channels, all of which are known to play key roles in the regulation of blood pressure through modulation of vascular tone, cardiac output, and/or Na+/water reabsorption in the kidneys. This suggests that elevated antibody production may be a causal mechanism in at least some cases of hypertension. In this brief review, we will further describe the protein targets of the antibodies that are elevated in individuals with essential and pregnancy-related hypertension and the likely pathophysiological consequences of antibody binding to these targets. We will speculate on the potential mechanisms that underlie elevated antibody levels in hypertensive individuals and, finally, we will outline the therapeutic opportunities that could arise with a better understanding of how and why antibodies are produced in hypertension.
Collapse
|
49
|
Yan L, Tan X, Chen W, Zhu H, Cao J, Liu H. Enhanced vasoconstriction to α1 adrenoceptor autoantibody in spontaneously hypertensive rats. SCIENCE CHINA-LIFE SCIENCES 2014; 57:681-9. [PMID: 24950619 DOI: 10.1007/s11427-014-4672-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/02/2013] [Indexed: 11/29/2022]
Abstract
Autoimmune activities have been implicated in the pathogenesis of hypertension. High levels of autoantibodies against the second extracellular loop of α1-adrenoceptor (α1-AR autoantibody, α1-AA) are found in patients with hypertension, and α1-AA could exert a α1-AR agonist-like vasoconstrictive effect. However, whether the vasoconstrictive effect of α1-AA is enhanced in hypertension is unknown. Using aortic rings of spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto (WKY) rats, we observed the vasoconstrictive responses to α1-AA with phenylephrine (α1-AR agonist) as a positive control drug. Aortic nitrotyrosine levels were also measured by ELISA and immunohistochemistry. The results showed that the aortic constrictive responses to α1-AA and phenylephrine (both 1 nmol L(-1)-10 μmol L(-1)) were greater in SHR than in WKY rats. Endothelial denudation or L-NAME (a non-selective NOS inhibitor) (100 μmol L(-1)) increased α1-AA- or phenylephrine-induced vasoconstrictions both in SHR and WKY. However, selective iNOS inhibitor 1400 W (10 μmol L(-1)) enhanced the α1-AA-induced aortic constriction in WKY, but not in SHR. The aortic nitrotyrosine level was significantly higher in SHR than WKY, as shown by both ELISA and immunohistochemistry. These results indicate that the vasoconstrictive response to α1-AA is enhanced in SHR, and this altered responsiveness is due to endothelial dysfunction and decreased NO bioavailability. The study suggests an important role of α1-AR autoimmunity in the pathogenesis and management of hypertension especially in those harboring high α1-AA levels.
Collapse
Affiliation(s)
- Li Yan
- Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medical Sciences Peking Union Medical College, Beijing, 100005, China
| | | | | | | | | | | |
Collapse
|
50
|
Agonistic autoantibodies directed against G-protein-coupled receptors and their relationship to cardiovascular diseases. Semin Immunopathol 2014; 36:351-63. [PMID: 24777744 DOI: 10.1007/s00281-014-0425-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/01/2014] [Indexed: 02/06/2023]
Abstract
Agonistic autoantibodies (AABs) against G-protein-coupled receptor (GPCR) are present mainly in diseases of the cardiovascular system or in diseases associated with cardiovascular disturbances. The increasing knowledge about the role of autoantibodies against G-protein-coupled receptor (GPCR-AABs) as pathogenic drivers, the resulting development of strategies aimed at their removal or neutralization, and the evidenced patient benefit associated with such therapies have created the need for a summary of GPCR-AAB-associated diseases. Here, we summarize the present knowledge about GPCR-AABs in cardiovascular diseases. The identity of the GPCR-AABs and their prevalence in each of several specific cardiovascular diseases are documented. The structure of GPCR is also briefly discussed. Using this information, differences between classic agonists and GPCR-AABs in their GPCR binding and activation are presented and the resulting pathogenic consequences are discussed. Furthermore, treatment strategies that are currently under study, most of which are aimed at the removal and in vivo neutralization of GPCR-AABs, are indicated and their patient benefits discussed. In this context, immunoadsorption using peptides/proteins or aptamers as binders are introduced. The use of peptides or aptamers for in vivo neutralization of GPCR-AABs is also described. Particular attention is given to the GPCR-AABs directed against the adrenergic beta1-, beta2-, and α1-receptor as well as the muscarinic receptor M2, angiotensin II-angiotensin receptor type I, endothelin1 receptor type A, angiotensin (1-7) Mas-receptor, and 5-hydroxytryptamine receptor 4. Among the diseases associated with GPCR-AABs, special focus is given to idiopathic dilated cardiomyopathy, Chagas' cardiomyopathy, malignant and pulmonary hypertension, and kidney diseases. Relationships of GPCR-AABs are indicated to glaucoma, peripartum cardiomyopathy, myocarditis, pericarditis, preeclampsia, Alzheimer's disease, Sjörgren's syndrome, and metabolic syndrome after cancer chemotherapy.
Collapse
|