Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Front Immunol ; 12: 644548, 2021.
Article in English | MEDLINE | ID: mdl-33889154

ABSTRACT

Soluble cluster of differentiation 26 (sCD26) has a wide range of enzymatic functions affecting immunological, metabolic and vascular regulation. Diminished sCD26 concentrations have been reported in various autoimmune diseases and also in Myalgic Encephalomyelitis/Chronic fatigue syndrome (ME/CFS). Here we re-evaluate sCD26 as a diagnostic marker and perform a comprehensive correlation analysis of sCD26 concentrations with clinical and paraclinical parameters in ME/CFS patients. Though this study did find significantly lower concentrations of sCD26 only in the female cohort and could not confirm diagnostic suitability, results from correlation analyses provide striking pathomechanistic insights. In patients with infection-triggered onset, the associations of low sCD26 with elevated autoantibodies (AAB) against alpha1 adrenergic (AR) and M3 muscarinic acetylcholine receptors (mAChR) point to a pathomechanism of infection-triggered autoimmune-mediated vascular and immunological dysregulation. sCD26 concentrations in infection-triggered ME/CFS were found to be associated with activated T cells, liver enzymes, creatin kinase (CK) and lactate dehydrogenase (LDH) and inversely with Interleukin-1 beta (IL-1b). Most associations are in line with the known effects of sCD26/DPP-4 inhibition. Remarkably, in non-infection-triggered ME/CFS lower sCD26 in patients with higher heart rate after orthostatic challenge and postural orthostatic tachycardia syndrome (POTS) suggest an association with orthostatic regulation. These findings provide evidence that the key enzyme sCD26 is linked to immunological alterations in infection-triggered ME/CFS and delineate a different pathomechanism in the non-infectious ME/CFS subset.


Subject(s)
Autoantibodies/immunology , Cardiovascular System/immunology , Dipeptidyl Peptidase 4/immunology , Fatigue Syndrome, Chronic/immunology , Infections/immunology , Receptor, Muscarinic M3/immunology , Receptors, Adrenergic, alpha-1/immunology , Adult , Female , Humans , Male
2.
Curr HIV Res ; 18(3): 154-164, 2020.
Article in English | MEDLINE | ID: mdl-32539678

ABSTRACT

BACKGROUND: Emergence of Kaposi's Sarcoma in the cases other than HIV, following the use of immunosuppressant drugs, demonstrates that it is related to weak immunity. The fact that this malignancy does not occur in every HIV-positive patient suggests that genetic predisposition may also be effective. Replacement of one of the base pairs of adenine, guanine, cytosine, and thymine that constitute the DNA sequence in the human genome with another base pair can affect susceptibility to disease, response to treatment, and immunity. OBJECTIVE: The purpose of this study is to analyze the Single Nucleotide Polymorphism that could predispose to Kaposi's sarcoma of an HIV-infected patient and to identify which nucleotides such SNPs correspond to, using the microarray technology. MATERIALS AND METHODS: The blood samples of individuals, one of whom was diagnosed with Kaposi's Sarcoma HIV (+) visiting the outpatient clinic of infectious diseases polyclinic of Harran University Research and Practice Hospital and of a healthy individual with no Kaposi's Sarcoma, were used in the study. Following the DNA isolation of the blood samples taken from the respective individuals, a SNP analysis was conducted on the microarray device. 204,000 SNPs obtained were scanned later on in the databases in an attempt to identify the SNPs related to Kaposi's Sarcoma. RESULTS: In the 204,000 SNP screenings, we scrutinized the SNPs that differ in the case of Kaposi's Sarcoma [KS (+) and HIV (+)] on the basis of Control [KS(-) and HIV(-)] and HIV+ [KS(-)], and two SNPs of the ENDRA gene, three SNPs of the ADRA1A gene, six SNPs of the STIM1 gene, four SNPs of the EFNB2 gene, and one SNP of the CD209 gene were found to be different. However, when it comes to all SNPs (all the 204.000 SNPs) screened in terms of allele, it was observed that the AA and BB alleles were lower in the patient with Kaposi's Sarcoma [KS (+) and HIV (+)] compared to other groups and AB alleles were found to be higher than others in the patient with Kaposi's sarcoma [KS] (+) and HIV (+)]. CONCLUSION: In the microarray study we have conducted, 204,000 SNPs were screened for Control (HIV-) HIV (+) and HIV (+) patient with Kaposi's Sarcoma. It was found that 32,362 of those SNPs had different alleles in the Kaposi's Sarcoma [KS + HIV (+)] patient, while they had the same ones in the control [KS (-) and HIV (-)] and HIV + [KS (-)] group. 16 of the 32,362 SNPs took place among the genes related to Kaposi's Sarcoma. In the cases of Kaposi's Sarcoma with suspected diagnosis, it can be used as a beneficial laboratory test.


Subject(s)
Cell Adhesion Molecules/genetics , Ephrin-B2/genetics , HIV Infections/genetics , Lectins, C-Type/genetics , Neoplasm Proteins/genetics , Receptor, Endothelin A/genetics , Receptors, Adrenergic, alpha-1/genetics , Receptors, Cell Surface/genetics , Sarcoma, Kaposi/genetics , Stromal Interaction Molecule 1/genetics , Adult , Alleles , Case-Control Studies , Cell Adhesion Molecules/immunology , Ephrin-B2/immunology , Gene Expression , Genetic Predisposition to Disease , HIV/growth & development , HIV/pathogenicity , HIV Infections/complications , HIV Infections/immunology , HIV Infections/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Lectins, C-Type/immunology , Male , Microarray Analysis , Middle Aged , Neoplasm Proteins/immunology , Polymorphism, Single Nucleotide , Receptor, Endothelin A/immunology , Receptors, Adrenergic, alpha-1/immunology , Receptors, Cell Surface/immunology , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/virology , Stromal Interaction Molecule 1/immunology
3.
J Am Heart Assoc ; 8(18): e013602, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31495251

ABSTRACT

Background The etiology of postural orthostatic tachycardia syndrome (POTS) is yet to be established. The disorder is often misdiagnosed as chronic anxiety or a panic disorder because the autonomic failure in these patients is not severe. A growing body of evidence suggests that POTS may be an autoimmune disorder. Antinuclear antibodies and elevations of ganglionic, adrenergic, and muscarinic acetylcholine receptor antibodies have all been reported. Methods and Results We collected detailed clinical symptoms of 55 patients diagnosed with POTS. We also evaluated serum levels of autoantibodies against 4 subtypes of G-protein coupled adrenergic receptors and 5 subtypes of G-protein coupled muscarinic acetylcholine receptors by ELISA. Our patients had a multitude of comorbidities, were predominantly young females, and reported viral-like symptoms preceding episodes of syncope. We detected a significant number of patients with elevated levels of autoantibodies against the adrenergic alpha 1 receptor (89%) and against the muscarinic acetylcholine M4 receptor (53%). Surprisingly, elevations of muscarinic receptor autoantibodies appeared to be dependent upon elevation of autoantibodies against the A1 adrenergic receptor! Four patients had elevations of G-protein coupled autoantibodies against all 9 receptor subtypes measured in our study. Five POTS patients had no elevation of any autoantibody; similarly, controls were also negative for autoantibody elevations. There was a weak correlation of clinical symptom severity with G-protein coupled autoantibodies. Conclusions Our observations provide further evidence that, in most cases, POTS patients have at least 1 elevated G-protein coupled adrenergic autoantibody and, in some instances, both adrenergic and muscarinic autoantibodies, supporting the hypothesis that POTS may be an autoimmune disorder.


Subject(s)
Autoantibodies/immunology , Postural Orthostatic Tachycardia Syndrome/immunology , Receptor, Muscarinic M4/immunology , Receptors, Adrenergic, alpha-1/immunology , Adolescent , Adult , Case-Control Studies , Dyspnea , Fatigue , Female , Headache , Humans , Joint Instability , Male , Migraine Disorders , Myalgia , Postural Orthostatic Tachycardia Syndrome/physiopathology , Receptors, Adrenergic, alpha-2 , Receptors, Adrenergic, beta/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Muscarinic/immunology , Young Adult
4.
J Alzheimers Dis ; 64(4): 1091-1097, 2018.
Article in English | MEDLINE | ID: mdl-30010118

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 ß2-adrenergic receptors (ß2-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%) ß2-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.


Subject(s)
Autoantibodies/blood , Dementia/blood , Dementia/epidemiology , Receptors, Adrenergic, alpha-1/immunology , Aged , Aged, 80 and over , Cohort Studies , Community Health Planning , Female , Humans , Logistic Models , Male , Psychiatric Status Rating Scales , Psychometrics , Receptors, Adrenergic, beta-2/immunology , Severity of Illness Index
5.
Front Biosci (Landmark Ed) ; 23(11): 2082-2089, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29772547

ABSTRACT

Agonistic autoantibodies (agAAB) for alpha-1 adrenoceptor were found in approx. 50% of patients with Alzheimer's disease. These antibodies activate the receptor and trigger the signal cascades similarly to how natural agonists do. The agAAB bond to the receptor is persistent and prolonged. This results in a non-physiological elevation of intracellular calcium. An animal model has shown that agAAB causes macrovascular and microvascular impairment in the vessels of the brain. Reduction in blood flow and the density of intact vessels was significantly demonstrated. The agAAB was removed through immunoadsorption in a small cohort of patients with Alzheimer's disease. Subsequent follow-up observations over 12-18 months noted stabilization of cognition levels.


Subject(s)
Alzheimer Disease/immunology , Autoantibodies/immunology , Dementia/immunology , Receptors, Adrenergic, alpha-1/immunology , Alzheimer Disease/blood , Alzheimer Disease/metabolism , Animals , Autoantibodies/blood , Brain/blood supply , Brain/immunology , Brain/metabolism , Dementia/blood , Dementia/metabolism , Disease Models, Animal , Humans
6.
Clin Sci (Lond) ; 131(24): 2947-2957, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29097625

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.


Subject(s)
Autoantibodies/blood , Hypertension/immunology , Receptor, Angiotensin, Type 1/immunology , Receptors, Adrenergic, alpha-1/immunology , Vascular Stiffness , Adult , Area Under Curve , Biomarkers/blood , Female , Humans , Hypertension/blood , Hypertension/diagnosis , Hypertension/physiopathology , Male , Middle Aged , Predictive Value of Tests , Prognosis , Pulse Wave Analysis , ROC Curve , Risk Assessment , Risk Factors , Time Factors
7.
Europace ; 19(7): 1211-1219, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-27702852

ABSTRACT

AIMS: Postural tachycardia syndrome (POTS), a common and debilitating cardiovascular disorder, is characterized by an exaggerated heart rate increase during orthostasis and a wide spectrum of adrenergic-related symptoms. To determine the aetiology of POTS, we examined a possible pathophysiological role for autoantibodies against α1-adrenergic (α1AR) and ß1/2-adrenergic receptors (ß1/2AR). METHODS AND RESULTS: Immunoglobulin G (IgG) derived from 17 POTS patients, 7 with recurrent vasovagal syncope (VVS), and 11 normal controls was analysed for its ability to modulate activity and ligand responsiveness of α1AR and ß1/2AR in transfected cells and to alter contractility of isolated rat cremaster arterioles in vitro. Immunoglobulin G activation of α1AR and ß1/2AR was significantly higher in POTS compared with VVS and controls in cell-based assays. Eight, 11, and 12 of the 17 POTS patients possessed autoantibodies that activated α1AR, ß1AR and ß2AR, respectively. Pharmacological blockade suppressed IgG-induced activation of α1AR and ß1/2AR. Eight of 17 POTS IgG decreased the α1AR responsiveness to phenylephrine and 13 of 17 POTS IgG increased the ß1AR responsiveness to isoproterenol irrespective of their ability to directly activate their receptors. Postural tachycardia syndrome IgG contracted rat cremaster arterioles, which was reversed by α1AR blockade. The upright heart rate correlated with IgG-mediated ß1AR and α1AR activity but not with ß2AR activity. CONCLUSION: These data confirm a strong relationship between adrenergic autoantibodies and POTS. They support the concept that allosteric-mediated shifts in the α1AR and ß1AR responsiveness are important in the pathophysiology of postural tachycardia.


Subject(s)
Abdominal Muscles/blood supply , Autoantibodies/blood , Autoimmunity , Immunoglobulin G/blood , Postural Orthostatic Tachycardia Syndrome/immunology , Receptors, Adrenergic, alpha-1/immunology , Receptors, Adrenergic, beta-1/immunology , Receptors, Adrenergic, beta-2/immunology , Adolescent , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Adult , Animals , Arterioles/drug effects , Arterioles/metabolism , CHO Cells , Case-Control Studies , Cricetulus , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Male , Postural Orthostatic Tachycardia Syndrome/blood , Postural Orthostatic Tachycardia Syndrome/diagnosis , Rats , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Transfection , Vasoconstriction/drug effects , Young Adult
8.
Ther Apher Dial ; 20(5): 523-529, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27096216

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.


Subject(s)
Autoantibodies/immunology , Dementia/therapy , Immunosorbent Techniques , Receptors, Adrenergic, alpha-1/immunology , Aged , Dementia/immunology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Time Factors , Treatment Outcome
9.
Exp Mol Med ; 46: e118, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25323788

ABSTRACT

This study assessed the roles of chronic stress (CS) in the stimulation of the sympathetic nervous system and explored the underlying mechanisms of periodontitis. Using an animal model of periodontitis and CS, the expression of tyrosine hydroxylase (TH) and the protein levels of the α1-adrenergic receptor (α1-AR) and ß2-adrenergic receptor (ß2-AR) were assessed. Furthermore, human periodontal ligament fibroblasts (HPDLFs) were stimulated with lipopolysaccharide (LPS) to mimic the process of inflammation. The proliferation of the HPDLFs and the expression of α1-AR and ß2-AR were assessed. The inflammatory-related cytokines interleukin (IL)-1ß, IL-6 and IL-8 were detected after pretreatment with the α1/ß2-AR blockers phentolamine/propranolol, both in vitro and in vivo. Results show that periodontitis under CS conditions enhanced the expression of TH, α1-AR and ß2-AR. Phentolamine significantly reduced the inflammatory cytokine levels. Furthermore, we observed a marked decrease in HPDLF proliferation and the increased expression of α1-ARfollowing LPS pretreatment. Pretreatment with phentolamine dramatically ameliorated LPS-inhibited cell proliferation. In addition, the blocking of α1-ARsignaling also hindered the upregulation of the inflammatory-related cytokines IL-1ß, IL-6 and IL-8. These results suggest that CS can significantly enhance the pathological progression of periodontitis by an α1-adrenergic signaling-mediated inflammatory response. We have identified a potential therapeutic target for the treatment of periodontal disease, particularly in those patients suffering from concurrent CS.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/therapeutic use , Periodontitis/drug therapy , Periodontitis/etiology , Phentolamine/therapeutic use , Receptors, Adrenergic, alpha-1/immunology , Stress, Physiological , Animals , Cells, Cultured , Cytokines/immunology , Fibroblasts/immunology , Fibroblasts/pathology , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Male , Periodontal Ligament/cytology , Periodontal Ligament/immunology , Periodontal Ligament/pathology , Periodontitis/immunology , Periodontitis/pathology , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/analysis , Signal Transduction/drug effects , Stress, Physiological/drug effects , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/immunology
10.
Pain ; 155(11): 2408-17, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25250722

ABSTRACT

Complex regional pain syndrome (CRPS) is a limb-confined posttraumatic pain syndrome with sympathetic features. The cause is unknown, but the results of a randomized crossover trial on low-dose intravenous immunoglobulins (IVIG) treatment point to a possible autoimmune mechanism. We tested purified serum immunoglobulin G (IgG) from patients with longstanding CRPS for evidence of antibodies interacting with autonomic receptors on adult primary cardiomyocytes, comparing with control IgG from healthy and diseased controls, and related the results to the clinical response to treatment with low-dose IVIG. We simultaneously recorded both single-cell contractions and intracellular calcium handling in an electrical field. Ten of 18 CRPS preparations and only 1/57 control preparations (P<0.0001) increased the sensitivity of the myocytes to the electric field, and this effect was abrogated by preincubation with α-1a receptor blockers. By contrast, effects on baseline calcium were blocked by preincubation with atropine. Interestingly, serum-IgG preparations from all 4 CRPS patients who had responded to low-dose IVIG with meaningful pain relief were effective in these assays, although 4/8 of the nonresponders were also active. To see if there were antibodies to the α-1a receptor, CRPS-IgG was applied to α-1a receptor-transfected rat-1 fibroblast cells. The CRPS serum IgG induced calcium flux, and fluorescence-activated cell sorting showed that there was serum IgG binding to the cells. The results suggest that patients with longstanding CRPS have serum antibodies to α-1a receptors, and that measurement of these antibodies may be useful in the diagnosis and management of the patients.


Subject(s)
Autoantibodies/blood , Complex Regional Pain Syndromes/blood , Complex Regional Pain Syndromes/immunology , Receptors, Adrenergic, alpha-1/immunology , Adult , Animals , Atropine/pharmacology , Calcium/metabolism , Cells, Cultured , Complex Regional Pain Syndromes/therapy , Cross-Over Studies , Dioxanes/pharmacology , Female , Humans , Immunoglobulin G/pharmacology , Immunoglobulins, Intravenous/therapeutic use , Male , Membrane Potentials/drug effects , Middle Aged , Muscarinic Antagonists/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Protein Binding/drug effects , Rats , Transfection , Young Adult
11.
NMR Biomed ; 27(9): 1085-93, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25060359

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.


Subject(s)
Autoantibodies/immunology , Blood Volume/immunology , Cerebrovascular Circulation/immunology , Ferrosoferric Oxide , Magnetic Resonance Angiography/methods , Receptors, Adrenergic, alpha-1/immunology , Animals , Blood Flow Velocity/immunology , Blood Volume Determination/methods , Contrast Media , Male , Microvessels/immunology , Microvessels/pathology , Rats , Rats, Wistar
12.
Sci China Life Sci ; 57(7): 681-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24950619

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.


Subject(s)
Autoantibodies/immunology , Hypertension/immunology , Receptors, Adrenergic, alpha-1/immunology , Vasoconstriction/immunology , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Aorta/immunology , Aorta/metabolism , Aorta/physiopathology , Autoantibodies/metabolism , Blotting, Western , Dose-Response Relationship, Drug , Endothelium, Vascular/immunology , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Humans , Hypertension/metabolism , Hypertension/physiopathology , In Vitro Techniques , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Phenylephrine/pharmacology , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar , Receptors, Adrenergic, alpha-1/physiology , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
13.
Naunyn Schmiedebergs Arch Pharmacol ; 387(7): 703-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24866500

ABSTRACT

We tested the specificity of three commercially available antibodies (AB) against individual α1-adrenergic receptor subtypes (α1-ARST). We used these ABs to localize the α1-ARST proteins by immunohistochemistry in paraffin-embedded murine tissues. The specificity of the ABs was tested by comparing staining patterns in tissues from wild-type mice with those in corresponding tissues from mice with gene-targeted disruption of the respective α1-ARST, one of the most rigorous negative controls. None of the tested ABs proved to be specific for the indicated target antigen. We conclude that the tested ABs are unsuitable for immunohistochemistry in paraffin-embedded murine tissues.


Subject(s)
Antibodies/immunology , Antibody Specificity/immunology , Immunohistochemistry/methods , Receptors, Adrenergic, alpha-1/immunology , Animals , Coronary Vessels/metabolism , Male , Mesenteric Arteries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Paraffin Embedding , Protein Subunits/immunology , Receptors, Adrenergic, alpha-1/genetics
14.
J Am Heart Assoc ; 3(1): e000755, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24572257

ABSTRACT

BACKGROUND: Patients with postural tachycardia syndrome (POTS) have exaggerated orthostatic tachycardia often following a viral illness, suggesting autoimmunity may play a pathophysiological role in POTS. We tested the hypothesis that they harbor functional autoantibodies to adrenergic receptors (AR). METHODS AND RESULTS: Fourteen POTS patients (7 each from 2 institutions) and 10 healthy subjects were examined for α1AR autoantibody-mediated contractility using a perfused rat cremaster arteriole assay. A receptor-transfected cell-based assay was used to detect the presence of ß1AR and ß2AR autoantibodies. Data were normalized and expressed as a percentage of baseline. The sera of all 14 POTS patients demonstrated significant arteriolar contractile activity (69±3% compared to 91±1% of baseline for healthy controls, P<0.001) when coexisting ß2AR dilative activity was blocked; and this was suppressed by α1AR blockade with prazosin. POTS sera acted as a partial α1AR antagonist significantly shifting phenylephrine contractility curves to the right. All POTS sera increased ß1AR activation (130±3% of baseline, P<0.01) and a subset had increased ß2AR activity versus healthy subjects. POTS sera shifted isoproterenol cAMP response curves to the left, consistent with enhanced ß1AR and ß2AR agonist activity. Autoantibody-positive POTS sera demonstrated specific binding to ß1AR, ß2AR, and α1AR in transfected cells. CONCLUSIONS: POTS patients have elevated α1AR autoantibodies exerting a partial peripheral antagonist effect resulting in a compensatory sympathoneural activation of α1AR for vasoconstriction and concurrent ßAR-mediated tachycardia. Coexisting ß1AR and ß2AR agonistic autoantibodies facilitate this tachycardia. These findings may explain the increased standing plasma norepinephrine and excessive tachycardia observed in many POTS patients.


Subject(s)
Autoantibodies/blood , Autoimmune Diseases/immunology , Autoimmunity , Hemodynamics , Postural Orthostatic Tachycardia Syndrome/immunology , Receptors, Adrenergic/immunology , Adrenergic Agonists/pharmacology , Adult , Animals , Autoimmune Diseases/blood , Autoimmune Diseases/diagnosis , Autoimmune Diseases/physiopathology , Biological Assay , Biomarkers/blood , CHO Cells , Case-Control Studies , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , Heart Rate , Hemodynamics/drug effects , Humans , Male , Middle Aged , Oklahoma , Postural Orthostatic Tachycardia Syndrome/blood , Postural Orthostatic Tachycardia Syndrome/diagnosis , Postural Orthostatic Tachycardia Syndrome/physiopathology , Rats , Receptors, Adrenergic/drug effects , Receptors, Adrenergic/genetics , Receptors, Adrenergic, alpha-1/immunology , Receptors, Adrenergic, beta-1/immunology , Receptors, Adrenergic, beta-2/immunology , Tennessee , Transfection , Vasoconstriction , Vasodilation , Young Adult
15.
Int Immunopharmacol ; 17(3): 774-84, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24055020

ABSTRACT

UNLABELLED: The mechanistic implications of the presence of sympathetic noradrenergic innervation in lymphoid organs in synaptic association with lymphocytes open to the influence of hormonal fluctuations throughout reproductive age in females has not been investigated yet. OBJECTIVES: The aim of the present study is to investigate the role of alpha-adrenoceptors (α-ARs) and estrogen in modulating immune responses in the spleen through intracellular signaling targets such as ERK 1/2, CREB, Akt, NF-κB. METHODS: Splenocytes from young Sprague-Dawley rats were incubated with α1- and α2- AR specific agonists, phenylephrine and clonidine, without and with 17b-estradiol or specific antagonists prazosin and idazoxan to examine their effects on proliferation, cytokine production, nitric oxide production, and intracellular signaling molecules. RESULTS: α1-AR stimulation inhibited lymphocyte proliferation and IFN-g production and enhanced IL-2, p-ERK and p-CREB expression. Co-stimulation using estrogen enhanced cytokine production and suppressed p-Akt expression. α1-AR blockade reversed agonist-induced IL-2 production alone. α2-AR stimulation inhibited lymphocyte proliferation, p-ERK and p-CREB expression, and increased p-NF-kB and p-Akt expression. Co-stimulation with estrogen increased IL-2 and suppressed p-CREB expression. α2-AR Idazoxan prevented IL-2 production in the absence and presence of estrogen, and reversed clonidine-induced increase in NO production and p-ERK and p-Akt expression in the presence of estrogen. CONCLUSIONS: These results suggest that the cell-mediated immune responses are selectively modulated depending upon the subtypes of α-AR and further, these effects are differentially regulated in the presence of estrogen mediated through selective alteration in the intracellular signaling pathways involving ERK, CREB, Akt, and NF-κB.


Subject(s)
Receptors, Adrenergic, alpha-1/immunology , Receptors, Adrenergic, alpha-2/immunology , Spleen/immunology , T-Lymphocytes/immunology , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Clonidine/pharmacology , Cyclic AMP Response Element-Binding Protein/immunology , Estradiol/pharmacology , Estrogens/pharmacology , Extracellular Signal-Regulated MAP Kinases/immunology , Idazoxan/pharmacology , Interferon-gamma/immunology , Interleukin-2/immunology , Male , NF-kappa B/immunology , Phenylephrine/pharmacology , Proto-Oncogene Proteins c-akt/immunology , Rats , Rats, Sprague-Dawley , Spleen/cytology , T-Lymphocytes/drug effects
16.
Atheroscler Suppl ; 14(1): 203-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23357166

ABSTRACT

Dilated cardiomyopathy (DCM) which is a common cause of heart failure is often related to elevated levels of autoantibodies (AABs) against cardiac structural or functional proteins. Among several AABs which react against cardiac cellular proteins that have been detected in sera from DCM patients, those against ß(1)-adreno-receptors (ß(1)-ARs) appeared particularly relevant from a pathophysiological point of view. During the last 15 years several studies evaluating the short-term efficacy of immunoadsorption (IA) in idiopathic DCM have shown improvement in cardiac function and patient outcome. However, the invasive and complicated aspects of the IA, which is also costly, have limited its broad clinical application as long as only its short-term efficacy has been definitely proved. Autoimmunity is also suspected to play a key role in the pathogenesis of pulmonary arterial hypertension (PAH). Recently we identified functional AABs against the α(1)-AR and/or the endothelin-A-receptor (ETA) in sera of patients with PAH. These AABs activate the receptors like corresponding agonists but, unlike the agonists, the AABs induce long-lasting stimulatory effects and do not desensitize the receptors. The AABs against the α(1)-AR and the ETA-receptor belong to IgG3 and IGg2 subclass, respectively, and can be removed by IA. The first 5 potential transplant candidates with idiopathic PAH who underwent IA showed good results after this therapy. This update aims to summarize the present knowledge about the role of AABs in the etiopathogenesis of DCM and PAH and the potential therapeutic benefits of AAB removal by IA. Special attention is focused on the therapeutic benefits of IA for patients with life-threatening end-stage disease where all pharmacological therapeutic options are exhausted.


Subject(s)
Autoantibodies/blood , Autoimmunity , Blood Component Removal/methods , Cardiomyopathy, Dilated/therapy , Hypertension, Pulmonary/therapy , Immunosorbent Techniques , Immunosorbents/therapeutic use , Absorption , Animals , Biomarkers/blood , Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/immunology , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/immunology , Receptor, Endothelin A/immunology , Receptors, Adrenergic, alpha-1/immunology , Receptors, Adrenergic, beta-1/immunology , Treatment Outcome
17.
PLoS One ; 7(7): e41602, 2012.
Article in English | MEDLINE | ID: mdl-22860001

ABSTRACT

BACKGROUND: Circulating agonistic autoantibodies acting at G protein-coupled receptors have been associated with numerous sever pathologies in humans. Antibodies directed predominantly against the α(1)-adrenergig receptor were detected in patients suffering from widespread diseases such as hypertension and type 2 diabetes. Their deleterious action has been demonstrated for peripheral organs. We postulate that antibodies to the α(1)-adrenergig receptor are relevant pathomolecules in diseases of the central nervous system associated with vascular impairments. METHODOLOGY/PRINCIPAL FINDINGS: Using a rat model we studied the long-term action of antibodies against the α(1)-adrenergig receptor either induced by immunization with a receptor peptide or applied by intravenous injection. The vasculature in the rat brains was investigated by time-of-flight magnetic resonance angiography using a 9.4 Tesla small animal MR imaging system. Visual examination of maximum-intensity-projections (MIPs) of brain angiographs revealed the development of vascular defects in antibody- exposed animals between three and eight months of treatment. Relative vascular areas were derived from representative MIP image sections by grayscale analysis and used to form an index of vascular circulation. Animals exposed to the action of α(1)-adrenergig receptor antibodies showed significantly reduced vascular areas (p<0.05). Calculated index values indicated attenuated blood flow in both antibody-treated cohorts compared to their respective controls reaching with (relative units ± standard error, n = 10) 0.839 ± 0.026 versus 0.919 ± 0.026 statistical significance (p<0.05) for peptide-immunized rats. CONCLUSION/SIGNIFICANCE: We present evidence that antibodies to the α(1)-adrenergig receptor cause cerebrovascular impairments in the rat. Our findings suggest the pathological significance of these antibodies in pathologies of the human central nervous system linked to impairments of brain vasculature such as stroke and dementia.


Subject(s)
Autoantibodies/immunology , Brain/blood supply , Receptors, Adrenergic, alpha-1/immunology , Animals , Autoantibodies/blood , Brain/immunology , Brain/metabolism , Cerebrovascular Circulation/immunology , Magnetic Resonance Angiography , Male , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/metabolism
18.
Scand J Immunol ; 75(5): 524-30, 2012 May.
Article in English | MEDLINE | ID: mdl-22260197

ABSTRACT

Although primary causes of Alzheimer's and vascular dementia are unknown, the importance of preceding vascular lesions is widely accepted. Furthermore, there is strong evidence for the involvement of autoimmune mechanisms. Here, we report the presence of agonistic autoantibodies directed at adrenergic receptors in the circulation of patients with mild to moderate Alzheimer's and vascular dementia. In 59% of these patients, agonistic autoantibodies against the α(1) -adrenergic receptor and the ß(2) -adrenergic receptor were identified. The majority of positive patients (66%) contained both types of autoantibodies in combination. In a control group of patients with neurological impairments others than Alzheimer's and vascular dementia, only 17% were found to harbour these autoantibodies. The autoantibodies to the α(1) -adrenergic receptor interacted preferably with the extracellular loop1 of the receptor. They were further studied in IgG preparations from the column regenerate of a patient who underwent immunoadsorption. The α(1) -adrenergic receptor autoantibodies specifically bound to the extracellular loop1 peptide of the receptor with an apparent EC(50) value of 30 nm. They mobilized intracellular calcium in a clonal cell line expressing the human form of the α(1) -adrenergic receptor. Our data support the notion that autoimmune mechanisms play a significant role in the pathogenesis of Alzheimer's and vascular dementia. We suggest that agonistic autoantibodies to the α(1) -adrenergic and the ß(2) -adrenergic receptor may contribute to vascular lesions and increased plaque formation.


Subject(s)
Alzheimer Disease/immunology , Autoantibodies/immunology , Dementia, Vascular/immunology , Receptors, Adrenergic, alpha-1/immunology , Receptors, Adrenergic, beta-2/immunology , Aged , Aged, 80 and over , Animals , Calcium/blood , Calcium/metabolism , Cells, Cultured , Female , Humans , Immunoglobulin G/analysis , Immunoglobulin G/blood , Male , Middle Aged , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Rats
19.
Cardiovasc Res ; 93(2): 280-90, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22131354

ABSTRACT

AIMS: The alpha1A-adrenergic receptor (α1A-AR) regulates various vascular functions and participates in the pathogenesis of primary hypertension. However, highly specific and subtype-selective antagonists of α1A-AR have not been developed. METHODS AND RESULTS: Two novel antibodies against the short peptides CP-7, which is located in the second extracellular loop of α1A-AR, and CPE-8, which is located in the third extracellular loop of α1A-AR, were prepared. The two antibodies specifically bound to α1A-AR. However, neither antibody prevented [(3)H]-epinephrine or [(3)H]-prazosin from binding to the receptor. In vitro, the anti-CP-7 antibody inhibited Ca(2+)-dependent signal transduction processes including protein kinase C translocation and extracellular signal-regulated kinase (ERK1/2) phosphorylation induced by phenylephrine (PHE). The anti-CP-7 antibody decreased the beating rates of neonatal rat cardiomyocytes with or without PHE stimulation and reduced the blood pressure of spontaneously hypertensive rats that were immunized with CP-7-keyhole limpet haemocyanin. CONCLUSION: The anti-CP-7 antibody specifically inhibited the activation of α1A-AR both in vitro and in vivo. No competition binding was found between anti-CP-7 and [(3)H]-epinephrine or [(3)H]-prazosin. An antibody that specifically inhibits a receptor could be useful in research on G-protein-coupled receptors that lack specific antagonists. The antibody against the epitope CP-7 might have potential in a therapeutic application for treating primary hypertension.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Antibodies/pharmacology , Epitopes/immunology , Receptors, Adrenergic, alpha-1/immunology , Animals , Calcium/metabolism , Cells, Cultured , Epinephrine/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunization , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Phenylephrine/pharmacology , Phosphorylation , Prazosin/metabolism , Protein Kinase C-alpha/metabolism , Rabbits , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...