Next Article in Journal
Current Perspectives on the Auxin-Mediated Genetic Network that Controls the Induction of Somatic Embryogenesis in Plants
Next Article in Special Issue
Autoantibody Biomarkers in Rheumatic Diseases
Previous Article in Journal
Clinical Course and Electron Microscopic Findings in Lymphocytes of Patients with DRAM2-Associated Retinopathy
Previous Article in Special Issue
Natural Autoantibodies in Chronic Pulmonary Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ganglionic Acetylcholine Receptor Antibodies and Autonomic Dysfunction in Autoimmune Rheumatic Diseases

1
Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
2
Department of Molecular Neurology and Therapeutics, Kumamoto University Hospital, Kumamoto 860-8556, Japan
3
Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8576, Japan
4
Department of Clinical Research, National Hospital Organization Nagasaki Kawatana Medical Center, Nagasaki 859-3615, Japan
5
Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Medical Sciences, Nagasaki 852-8501, Japan
6
Department of Neurology, National Hospital Organization Nagasaki Kawatana Medical Center, Nagasaki 859-3615, Japan
7
Department of Rheumatology, Kumamoto Sakurajyuji Hospital, Kumamoto 861-4173, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2020, 21(4), 1332; https://doi.org/10.3390/ijms21041332
Submission received: 17 January 2020 / Revised: 13 February 2020 / Accepted: 13 February 2020 / Published: 16 February 2020
(This article belongs to the Special Issue Discovery of Antibody Biomarker)

Abstract

:
Autonomic neuropathy has been reported in autoimmune rheumatic diseases (ARD) including Sjögren’s syndrome, systemic sclerosis, rheumatoid arthritis, and systemic lupus erythematosus. However, the pathophysiological mechanism underlying autonomic dysfunction remains unknown to researchers. On the other hand, autoimmune autonomic ganglionopathy (AAG) is an acquired immune-mediated disorder, which causes dysautonomia that is mediated by autoantibodies against ganglionic acetylcholine receptors (gAChRs). The purpose of this review was to describe the characteristics of autonomic disturbance through previous case reports and the functional tests used in these studies and address the importance of anti-gAChR antibodies. We have established luciferase immunoprecipitation systems to detect antibodies against gAChR in the past and determined the prevalence of gAChR antibodies in various autoimmune diseases including AAG and rheumatic diseases. Autonomic dysfunction, which affects lower parasympathetic and higher sympathetic activity, is usually observed in ARD. The anti-gAChR antibodies may play a crucial role in autonomic dysfunction observed in ARD. Further studies are necessary to determine whether anti-gAChR antibody levels are correlated with the severity of autonomic dysfunction in ARD.

1. Introduction

Autonomic neuropathy has been reported in autoimmune rheumatic diseases including Sjögren’s syndrome (SS) [1,2], systemic sclerosis (SSc) [3], rheumatoid arthritis (RA) [4], and systemic lupus erythematosus (SLE) [5]. Several underlying mechanisms such as the immunological basis, which includes circulating autoantibodies, abnormalities of cellular immunity, vasculitis, and secondary amyloidosis have been proposed.
Autoimmune autonomic ganglionopathy (AAG) is an acquired immune-mediated disorder that causes widespread autonomic failure, which is mediated by autoantibodies against the ganglionic acetylcholine receptor (gAChR) [6,7,8]. Clinical manifestations result from the impairment of sympathetic [orthostatic hypotension (OH) and anhidrosis] and parasympathetic activity (abnormal pupillary response, sexual dysfunction, and a fixed heart rate) [7,9]. The gAChRs located in autonomic ganglia in the sympathetic and parasympathetic nervous system have a pentameric structure, consisting of two α3 and three β4 subunits [10]. The Mayo Clinic group was the first to report that autoantibodies targeting gAChRs detected in the sera of approximately 50% of patients with idiopathic autonomic neuropathy were proven to be pathogenic [7,11]. These autoantibodies induce the internalization of cell-surface nicotinic gAChRs and subsequent impairment in synaptic transmission within the autonomic nervous system [12,13]. Radio-immunoprecipitation assay with [125I]-labeled epibatidine, which was developed by the Mayo Clinic, has been used to detect gAChR antibodies [6,7,8]. We developed luciferase immunoprecipitation systems (LIPS) to detect antibodies that specifically bind to the α3 or β4 gAChR subunits with high sensitivity. We performed the LIPS analysis with the α3 or β4 subunit fused to Gaussia Luciferase8990 to measure the respective antibodies in human sera [9,14]. Recently, we determined the prevalence of anti-gAChR antibodies in autoimmune rheumatic diseases (ARD) including SS, SSc, RA, and SLE with this method [15,16,17]. In the present review, we aimed to critically examine the current literature on autonomic neuropathy and autonomic function tests and to propose that anti-gAChR antibodies provide a new perspective on the mechanism underlying autonomic dysfunction in ARD.
We searched for previous reports of autonomic dysfunction associated with ARD using a PubMed search. Search terms used were “Autonomic dysfunction”, “Autonomic neuropathy”, “Sjögren’s syndrome”, “Systemic sclerosis”, “Rheumatoid arthritis”, and “Systemic lupus erythematosus”. Results of the search were screened for related studies by applying inclusion and exclusion criteria to the full text of the related studies. Article type included research article, short communication, case series, case reports, literature review published between 1983 and 2019.

2. Autonomic Dysfunction in Sjögren’s Syndrome

SS is a systemic autoimmune disease characterized by exocrine impairment of the salivary and lacrimal glands, in addition to various extraglandular features. Exocrine glandular dysfunction is a pathognomonic feature of SS. Exocrine glandular function is highly regulated by the autonomic nervous system [18]. Cholinergic dysfunction may be independent of the inflammation and atrophy of the exocrine glands [19]. Newton et al. reported that autonomic symptoms, which were common among patients with primary SS, may contribute to the overall symptom burden and are linked with systemic disease activity [20]. Several studies have estimated dysfunction in both parasympathetic and sympathetic nerves or only parasympathetic nerves through cardiovascular autonomic reflex testing [21,22,23,24,25,26,27,28]. However, the use of heart rate variability (HRV) has resulted in contradictory findings [29,30,31,32,33,34]. The predominance of parasympathetic vagal modulation of cardiac function at rest, coupled with reduced cardiac baroreceptor control of heart rate and increased sympathetic vasoconstrictor activity in patients with primary SS has been reported using microneurography [35].
Autonomic neuropathy characterized by various autonomic features including orthostatic intolerance (OI), and gastrointestinal (GI), sudomotor, pupillary and genitourinal impairment in patients with SS may be the initial manifestation and before or after the appearance of the “sicca” symptoms, accompanied by symptoms other than dysautonomia including sensory disturbance [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55]. It could be localized [41,47,53] or systemic in distribution. Although autonomic neuropathy is potentially immunoresponsive, immunotherapy with intravenous immunoglobulin (IVIg) may require repetitive, continuous, or adjunctive therapy with rituximab for sustained improvement [56]. Pathological findings at autopsy revealed a decrease in the number of neurons within the thoracic sympathetic ganglia [50].
The possible pathogenesis of autonomic dysfunction in SS involves immunological factors including direct T-cell attack or ischemia caused by vasculitis in the autonomic ganglia and peripheral autonomic nerves [50,57,58], inhibition of neuropeptide secretion from nerve endings induced by cytokines [59], immune complex-mediated inflammation, and formation of pathogenic autoantibodies against receptors relative to autonomic function [60]. Type-3 muscarinic acetylcholine receptors (M3Rs) have been thought to be autoantigens in SS [61,62,63,64,65]. Importantly, Antibodies against M3Rs interfere with M3R-mediated parasympathetic neurotransmission and inhibit salivary secretion [66], GI motility [67], and bladder detrusor muscle contraction [68]. Passive transfer of SS immunoglobulins or rabbit antibodies to the second extracellular loop of the M3R in mice caused overactive bladder [69]. Moreover, neutralization of anti-M3R antibodies using IVIg improved the bladder and GI symptoms [70].

3. Autonomic Dysfunction in Systemic Scleroderma

SSc is an autoimmune rheumatic disease characterized by inflammation, vascular injury, autoantibody production, and fibrosis of the skin and internal organs [71]. Neurologic involvement including autonomic dysfunction has been recognized and reported in scleroderma [3,72,73,74,75,76]. Some manifestations of SSc including GI dysfunction [77] and impairment of microcirculation [78] are attributed to autonomic dysfunction [3]. Reduced vagal and increased sympathetic modulation at rest and deranged sympathetic response to orthostatic stress have been demonstrated by HRV analysis [79]. Cardiac autonomic dysfunction, which is related to right ventricular dysfunction [80], dysregulation of myocardial blood flow [81] and arrhythmic complications and mortality in patients with SSc [78] and precedes the development of fibrosis [82]. Autonomic dysfunction is also correlated with anorectal motility disorders [83], gastric compliance [84], and esophageal motor dysfunction [85,86]. Patients with more severe GI disease had significantly higher scores across several composite autonomic symptom scale-31 (COMPASS-31) subdomains, including OI and secretomotor dysfunction [87]. Pupillary autonomic dysfunction, including several patients with Adie’ tonic pupil [85,88,89,90], abnormal sympathetic skin response [91,92,93] and bladder dysfunction [94], was described. The pathogenesis of autonomic dysfunction in SSc remains elusive to researchers. However, it is thought to be caused by autoimmune damage to nerves, vascular disease, or direct nerve compression from tissue fibrosis [75]. Pulse cyclophosphamide therapy was reported to decrease sympathetic overdrive in a patient with early SSc [95].
It has been recognized that humoral immunity dysregulation plays an important role in SSc pathogenesis, and several antibodies can be detected in the sera of patients with SSc [96]. Three antibodies e.g., the anti-centromere antibody, the anti-scleroderma-70 antibody, and the anti- RNA polymerase III-70 antibody are specific for SSc and serve as specific markers [96]. Previous studies have demonstrated that GI dysmotility in SSc was associated with circulating autoantibodies against muscarinic AChRs and myenteric neurons [96,97,98,99,100,101,102,103]. Recently, McMahan and colleagues reported that patients with SSc and anti-RNPC3 antibodies had moderate-to-severe GI disease [104].

4. Autonomic Dysfunction in Rheumatoid Arthritis

RA is a chronic and systemic inflammatory condition that mainly affects the synovial joints but also presents with extra-articular manifestations [105]. Patients with RA present with autonomic symptoms including alterations of the skin, nails and hair, cyanotic extremities, peripheral vasospasm, tachycardia, and OH. Impaired heat rate responses to deep breathing, orthostasis and Valsalva maneuver (performed clinically for cardiovascular examination), abnormal HRV indices, impaired sympathetic skin response, and pupillary autonomic dysfunction have been demonstrated in a majority of patients with RA [106,107,108,109,110,111,112,113,114,115,116,117]. The prevalence of autonomic dysfunction detected by abnormal cardiovascular reflex tests varied from 33% to 86% (median prevalence: 60%) [4]. The characteristics of autonomic dysfunction (low HRV, reduced parasympathetic activity, elevated sympathetic activity and reduced cardiac baroreflex sensitivity [116]) are related to an increased risk of cardiovascular disease and mortality in patients with RA [4]. Autonomic dysfunction occurs in the early stage of RA and is not always a result of long-term RA and the inflammatory burden [117,118]. The association between the autonomic nervous system and elevated inflammation has been demonstrated using clinical cardiovascular tests and HRV [117,119,120,121]. Reduced parasympathetic and increased sympathetic activity on HRV were predictors of poor response to anti-tumor necrosis factor therapy [122]. The pathogenesis of autonomic dysfunction in patients with RA remains to be clearly elucidated in the literature. The presence of circulating autoantibodies against the nerve growth factor [123], cervical ganglia, and vagus nerve [124] and vasculitis of the vasa nervorum [125,126] and secondary amyloidosis have been proposed as the possible pathogenic mechanism. Proinflammatory cytokines have also been to linked to autonomic dysfunction. Cerebrospinal fluid interleukin-1β was inversely correlated with parasympathetic activity [127]. Interleukin-6 blockade with tocilizumab improved autonomic dysfunction in RA [125].

5. Autonomic Dysfunction in Systemic Lupus Erythematosus

SLE is an autoimmune disease that affects multiple organs and the central and peripheral nervous systems by the production of autoantibodies and immune-complex deposition [128]. The 1999 American College of Rheumatology provided definitions for seven peripheral manifestations (acute inflammatory demyelinating polyradiculopathy, autonomic disorder, mononeuropathy, myasthenia gravis, cranial neuropathy, plexopathy and polyneuropathy) related to SLE [129]. Abnormal HRV indices on the 24-h ambulatory electrocardiogram (ECG) and standard 12-lead ECG monitoring [130,131,132], abnormal sympathetic skin response [133], and pupillary autonomic disturbance [88] have been detected in patients with SLE. The prevalence of autonomic dysfunction ranged widely from 6 to 93% in patients with SLE [113,130]. Autonomic dysfunction in SLE is often asymptomatic and is not associated with disease activity, disease damage, and serological markers [131,134]. The findings of most of the studies reviewed supported sympathetic nervous system predominance or parasympathetic nervous system dysregulation, as reflected by decreased HRV in patients with SLE [135]. This autonomic imbalance is related to an increased risk of developing cardiovascular disease, which is a major cause of morbidity and mortality in patients with SLE [136,137]. Autonomic neuropathy can occur before or after the other clinical manifestations of SLE [138,139,140]. The precise pathogenic mechanism underlying autonomic dysfunction in SLE remains unclear. The positive response of several patients to immunotherapy for autonomic neuropathy characterized by severe sympathetic and parasympathetic impairment, supports the immune component in the pathogenesis of autonomic dysfunction in SLE [138,141,142]. Circulating autoantibodies against the nerve growth factor [123], cervical ganglia and vagus nerve (similar to rheumatoid arthritis) has been proposed [124].

6. Anti-Ganglionic Acetylcholine Receptor Antibody in Autoimmune Rheumatic Disease

6.1. Case Series and Prevalence of Anti-gAChR Antibodies in Sjögren’s Syndrome

Kondo et al. described two Japanese patients with SS, who presented with chronically progressive dysautonomia. One patient was seropositive for anti-gAChR antibodies and improved after oral intake of prednisolone. This case indicated that anti-gAChR antibodies are relevant to the pathophysiology of SS [143]. We also described a seropositive patient with SS, who presented with recurrent abdominal distention, constipation, weight loss, orthostatic dizziness, anhidrosis, and incomplete bladder emptying [144].
We determined the associations between autonomic dysfunction and anti-gAChR antibodies in SS by using the LIPS assay. We found that 9/39 (23.1%) patients with SS were seropositive and five of nine patients had autonomic symptoms. Moreover, we detected anti-gAChRα3 and anti-gAChRβ4 antibodies in 8/10 (80.0%) patients with SS with autonomic symptoms [15]. To identify the clinical characteristics of primary SS in patients with the gAChR antibodies, we obtained 22 serum samples (from 22 patients with primary SS) from teaching and general hospitals throughout Japan between January 2012 and March 2017 [145]. Clinical diagnoses were made in each hospital, and the diagnosis of SS was confirmed using the diagnostic criteria proposed by the American European Consensus Group and/or the Japanese Ministry of Health criteria for SS diagnostics. We measured levels of autoantibodies against gAChRα3 and gAChRβ4. A total of 11/22 patients tested positive for antibodies, including 9 gAChRα3-positive and 2 double antibody-positive patients. We reviewed clinical features and laboratory data (cerebrospinal fluid findings, other antibodies, etc.) for these 22 patients with primary SS (mean age: 53 years old; 6 men and 16 women). Chronic autonomic dysfunction was the predominant subtype in our investigation (6/16, 38%). No significant differences in clinical features involving autonomic dysfunction and laboratory data were noted, except for a higher frequency of OH/OI in the patients who were positive for the anti-gAChR antibodies compared to the antibody-negative patients (90% vs. 44%, p = 0.043).

6.2. Case Series and Prevalence of Anti-gAChR Antibodies in Systemic Sclerosis, Systemic Lupus Erythematosus, and Rheumatoid Arthritis

We described the cases of three patients with ARD and AAG. Patient 1 demonstrated an overlap between RA and SS, patient 2 demonstrated an overlap between RA and SSc, and patient 3 demonstrated an overlap between RA and SLE. Anti-gAChRα3 antibodies were detected in patients 1 and 2. A variety of autonomic nervous symptoms such as OI, early satiety, constipation, and diarrhea were observed. All patients received steroid pulse therapy, and their clinical symptoms improved [146].
We determined the associations between autonomic dysfunction and anti-gAChR antibodies in SSc, SLE, and RA using the LIPS assay. The LIPS assay detected anti-gAChRα3 and β4 antibodies in the sera of patients with SSc (13.2%, 5/38), SLE (12.5%, 4/32), and RA (18.6%, 8/43) [17].
We investigated human leukocyte antigen (HLA) alleles in patients with autoimmune hepatitis with or without anti-gAChR antibodies earlier. The frequency of the HLA-DRB1*0403 allele differed among Japanese patients with autoimmune hepatitis, based on the presence or absence of anti-gAChR antibodies. Thus, we should confirm the association of the HLA allele and each ARD [16].

6.3. Illustrative Cases That Tested Positive for Anti-gAChR Autoantibodies

6.3.1. Patient 1: SS with Widespread Neurological Symptoms Including Autonomic Dysfunction

A 58-year-old woman presented with a wide range of autonomic symptoms including sicca symptoms, constipation, OH and anhidrosis, pupillary abnormality two years ago. Neurological examination revealed “glove and stocking“ paresthesia, pyramidal tract sign, and ataxia. Although the patient tested negative for anti-SSA and SSB antibodies, lip biopsy demonstrated the infiltration of inflammatory cells around the ducts. She was diagnosed with primary SS, according to the American-European consensus criteria. She tested positive for anti-gAChRα3 and anti-M3R antibodies. The level of anti-gAChRα3 antibodies detected in serum was 1.343 antibody index (AI) (normal value < 1.000) using the LIPS assay. The serum levels of the second extracellular domain of M3R antibodies and the N-terminal of M3R antibodies detected by ELISA were 0.203 (normal value < 0.103) and 0.185 (normal value < 0.074), respectively. The coefficient of variation in R-R intervals (CVRR) was normal (4.15%, normal value > 1.4), but the coefficient of low frequency/high frequency was very low (0.35, normal range = 0.8–2.0) (Table 1). Sudomotor function testing revealed sympathetic dysfunction (Figure 1). She was treated with intravenous methylprednisolone (IVMP) (methylprednisolone 1 g per day for 3 days), which was followed by IVIg therapy (15 g per day for 5 days) twice in two months, based on the clinical diagnosis of SS with neurological symptoms including widespread autonomic dysfunction. She demonstrated improvement in OH and experienced recovery of sweating function with IVMP followed by IVIg. The levels of the anti-gAChR and anti-M3R antibodies returned to normal after immunotherapy, which resulted in improvement on autonomic function examination (Table 1 and Figure 1). Especially, we confirmed the denervation supersensitivity was improved in autonomic testing for pupil abnormality.
Anhidrosis was confirmed before immunotherapy, except on the axillary and palmar surfaces. Patient 1 was administered intravenous methylprednisolone, followed by monthly infusions of intravenous immunoglobulin for two months. Consequently, the anhidrosis improved with combination therapy, accompanied by a decrease in serum anti-gAChR and anti-M3R antibody levels.

6.3.2. Patient 2: Systemic Scleroderma and Autonomic Manifestations

A 76-year-old woman who had SSc for at least 9 years also experienced OH/OI and alternate bowel habits. She experienced recurrent light-headedness upon standing, severe constipation, and diarrhoea. Ocular examination confirmed a pupillary abnormality in the left eye. The serum level of anti-gAChRα3 antibodies was 3.607 AI. The CVRR was low (0.72%, normal value > 2.3). Moreover, the plasma norepinephrine (NE) level was also low in this patient (21, normal range = 90–420 pg/mL). Treatment with two doses of IVMP, followed by administration of oral prednisolone (1 mg/kg of body weight) based on the clinical diagnosis of SSc with AAG, led to sustained improvement in the autonomic symptoms. Subsequently, oral administration of tacrolimus was initiated at a dose of 3 mg once daily after obtaining informed consent. She maintained improvements in OI and experienced recovery of GI function with IVMP therapy, which was followed by oral prednisolone and tacrolimus treatment. The levels of anti-gAChR antibodies in this patient decreased after treatment (α3, 1.445 AI), which resulted in an improvement in CVRR (1.26%) and plasma NE level (679 pg/mL), respectively.

7. Limitations

This review has some limitations. First, the present study included a small study population. Further studies are necessary to determine the prevalence of autonomic disturbances in ARD. Second, we focused on only one aspect of the autonomic disturbances in of ARD, i.e., anti-gAChR antibodies. We should examine the immunological and autonomic biomarkers related to autonomic disturbance that could predict response to treatment. Third, a direct association between autonomic disturbance in ARD and gAChR antibodies has still not been proven. We should investigate the prevalence of autonomic disturbance and the immunopathogenesis of anti-gAChR antibodies for each rheumatic disease.

8. Conclusions

We performed a literature review of studies on autonomic dysfunction and determined that the prevalence of anti-gAChR antibodies was significant in ARD. Autonomic dysfunction, which is composed of parasympathetic underactivity and sympathetic overdrive is usually observed in ARD. Anti-gAChR antibodies may play a crucial role in autonomic dysfunction in ARD (Figure 2). Several phenomena other than anti-gAChR antibodies may play overlapping roles in the development of autonomic symptoms in ARD, and immune dysregulation of the autonomic nervous system could be responsible for the pathogenesis of ARD. Further studies are necessary to determine whether anti-gAChR antibody levels are correlated with the severity of autonomic dysfunction in ARD and to confirm the treatment strategy for countering autonomic dysfunction in ARD. Finally, rheumatologists and neurologists should foster cooperation among physicians, share clinical experiences and promote translational research. Moreover, we should expand on the concept and significance of our research, so that non-specialists can understand it and find it interesting.
Autonomic manifestations are frequently observed in patients with ARD, i.e., Sjögren’s syndrome (SS), systemic sclerosis (SSc), rheumatoid arthritis (RA), and systemic lupus erythematosus (SLE). It is well known that anti-M3R autoantibodies cause autonomic dysfunction, including the sicca complex and gastrointestinal dysmotility in ARD. On the other hand, anti-gAChR autoantibodies have the potential to physiologically block ganglionic synaptic transmission in patients with autoimmune autonomic ganglionopathy (AAG). Autoimmune diseases were observed in approximately 30% of AAGs in our previous study. We consider that AAG and other autoimmune diseases can coexist, owing to the same autoimmune basis. There is the possibility that the anti-gAChR autoantibodies contribute to the autonomic manifestations associated with ARD. We recommend that the highest priority for future experimental work in this field should be to analyze the autoantibodies against all the receptors in the autonomic nervous system such as M3R, gAChR, etc.

Author Contributions

Conceptualization: A.M. and S.N.; Methodology: A.M., H.T., O.H., H.N., and S.N.; Investigation and Data Curation: M.I., A.M., K.T., S.A. and S.N.; Writing—Original Draft Preparation M.I., A.M., and S.N.; Writing—Review and Editing: M.I., A.M., H.T., H.N., Y.A., H.M., T.N., T.S., A.K., and S.N.; Visualization: A.M. and S.N.; Supervision: S.N.; Project Administration: M.I., A.M. and S.N.; Funding Acquisition: A.M. and S.N. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Ministry of Health, Labor, and Welfare, Japan, and the Ministry of Education, Culture, Sports, Science, and Technology of Japan (JSPS KAKENHI Grant Numbers 16K19515, 19K17012, 16K09695, and 19H03549).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAGAutoimmune autonomic ganglionopathy
AIAntibody index
CVR-RCoefficient of variation of R-R intervals
ECGElectrocardiogram
gAChRGanglionic acetylcholine receptor
GIGastrointestinal
HRVHeart rate variability
IVMPIntravenous methylprednisolone
LIPSLuciferase immunoprecipitation systems
M3RType-3 muscarinic acetylcholine receptor
NENorepinephrine
OHOrthostatic hypotension
OIOrthostatic intolerance
RARheumatoid arthritis
SSSjögren’s syndrome
SLESystemic lupus erythematosus
SScSystemic sclerosis

References

  1. Goodman, B.P.; Crepeau, A.; Dhawan, P.S.; Khoury, J.A.; Harris, L.A. Spectrum of Autonomic Nervous System Impairment in Sjögren Syndrome. Neurologist 2017, 22, 127–130. [Google Scholar] [CrossRef] [PubMed]
  2. Perzyńska-Mazan, J.; Maślińska, M.; Gasik, R. Neurological manifestations of primary Sjögren’s syndrome. Reumatologia 2018, 56, 99–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Amaral, T.N.; Peres, F.A.; Lapa, A.T.; Marques-Neto, J.F.; Appenzeller, S. Neurologic involvement in scleroderma: A systematic review. Semin. Arthritis Rheum. 2013, 43, 335–347. [Google Scholar] [CrossRef]
  4. Adlan, A.M.; Lip, G.Y.; Paton, J.F.; Kitas, G.D.; Fisher, J.P. Autonomic function and rheumatoid arthritis: A systematic review. Semin. Arthritis Rheum. 2014, 44, 283–304. [Google Scholar] [CrossRef]
  5. Bortoluzzi, A.; Silvagni, E.; Furini, F.; Piga, M.; Govoni, M. Peripheral nervous system involvement in systemic lupus erythematosus: A review of the evidence. Clin. Exp. Rheumatol. 2019, 37, 146–155. [Google Scholar]
  6. Vernino, S.; Adamski, J.; Kryzer, T.J.; Fealey, R.D.; Lennon, V.A. Neuronal nicotinic ACh receptor antibody in subacute autonomic neuropathy and cancer-related syndromes. Neurology 1998, 50, 1806–1813. [Google Scholar] [CrossRef]
  7. Vernino, S.; Low, P.A.; Fealey, R.D.; Stewart, J.D.; Farrugia, G.; Lennon, V.A. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N. Engl. J. Med. 2000, 343, 847–855. [Google Scholar] [CrossRef]
  8. Vernino, S.; Hopkins, S.; Wang, Z. Autonomic ganglia, acetylcholine receptor antibodies, and autoimmune ganglionopathy. Auton. Neurosci. 2009, 146, 3–7. [Google Scholar] [CrossRef] [Green Version]
  9. Nakane, S.; Mukaino, A.; Higuchi, O.; Watari, M.; Maeda, Y.; Yamakawa, M.; Nakahara, K.; Takamatsu, K.; Matsuo, H.; Ando, Y. Autoimmune autonomic ganglionopathy: An update on diagnosis and treatment. Expert Rev. Neurother. 2018, 18, 953–965. [Google Scholar] [CrossRef]
  10. Vernino, S.; Sandroni, P.; Singer, W.; Low, P.A. Invited Article: Autonomic ganglia: Target and novel therapeutic tool. Neurology 2008, 70, 1926–1932. [Google Scholar] [CrossRef] [Green Version]
  11. Wang, Z.; Low, P.A.; Jordan, J.; Freeman, R.; Gibbons, C.H.; Schroeder, C.; Sandroni, P.; Vernino, S. Autoimmune autonomic ganglionopathy: IgG effects on ganglionic acetylcholine receptor current. Neurology 2007, 68, 1917–1921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Lennon, V.A.; Ermilov, L.G.; Szurszewski, J.H.; Vernino, S. Immunization with neuronal nicotinic acetylcholine receptor induces neurological autoimmune disease. J. Clin. Investig. 2003, 111, 907–913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kobayashi, S.; Yokoyama, S.; Maruta, T.; Negami, M.; Muroyama, A.; Mitsumoto, Y.; Iwasa, K.; Yamada, M.; Yoshikawa, H. Autoantibody-induced internalization of nicotinic acetylcholine receptor α3 subunit exogenously expressed in human embryonic kidney cells. J. Neuroimmunol. 2013, 257, 102–106. [Google Scholar] [CrossRef] [Green Version]
  14. Nakane, S.; Higuchi, O.; Koga, M.; Kanda, T.; Murata, K.; Suzuki, T.; Kurono, H.; Kunimoto, M.; Kaida, K.; Mukaino, A.; et al. Clinical features of autoimmune autonomic ganglionopathy and the detection of subunit-specific autoantibodies to the ganglionic acetylcholine receptor in Japanese patients. PLoS ONE 2015, 10, e0118312. [Google Scholar] [CrossRef]
  15. Mukaino, A.; Nakane, S.; Higuchi, O.; Nakamura, H.; Miyagi, T.; Shiroma, K.; Tokashiki, T.; Fuseya, Y.; Ochi, K.; Umeda, M.; et al. Insights from the ganglionic acetylcholine receptor autoantibodies in patients with Sjögren’s syndrome. Mod. Rheumatol. 2016, 26, 708–715. [Google Scholar] [CrossRef]
  16. Maeda, Y.; Migita, K.; Higuchi, O.; Mukaino, A.; Furukawa, H.; Komori, A.; Nakamura, M.; Hashimoto, S.; Nagaoka, S.; Abiru, S.; et al. Association between Anti-Ganglionic Nicotinic Acetylcholine Receptor (gAChR) Antibodies and HLA-DRB1 Alleles in the Japanese Population. PLoS ONE 2016, 11, e0146048. [Google Scholar] [CrossRef] [Green Version]
  17. Maeda, Y.; Nakane, S.; Higuchi, O.; Nakamura, H.; Komori, A.; Migita, K.; Mukaino, A.; Umeda, M.; Ichinose, K.; Tamai, M.; et al. Ganglionic acetylcholine receptor autoantibodies in patients with autoimmune diseases including primary biliary cirrhosis. Mod. Rheumatol. 2017, 27, 664–668. [Google Scholar] [CrossRef]
  18. Proctor, G.B.; Carpenter, G.H. Regulation of salivary gland function by autonomic nerves. Auton. Neurosci. 2007, 133, 3–18. [Google Scholar] [CrossRef]
  19. Imrich, R.; Alevizos, I.; Bebris, L.; Goldstein, D.S.; Holmes, C.S.; Illei, G.G.; Nikolov, N.P. Predominant Glandular Cholinergic Dysautonomia in Patients With Primary Sjögren’s Syndrome. Arthritis Rheumatol. 2015, 67, 1345–1352. [Google Scholar] [CrossRef] [Green Version]
  20. Newton, J.L.; Frith, J.; Powell, D.; Hackett, K.; Wilton, K.; Bowman, S.; Price, E.; Pease, C.; Andrews, J.; Emery, P.; et al. UK primary Sjögren’s syndrome registry. Autonomic symptoms are common and are associated with overall symptom burden and disease activity in primary Sjogren’s syndrome. Ann. Rheum. Dis. 2012, 71, 1973–1979. [Google Scholar] [CrossRef]
  21. Andonopoulos, A.P.; Christodoulou, J.; Ballas, C.; Bounas, A.; Alexopoulos, D. Autonomic cardiovascular neuropathy in Sjögren’s syndrome. A controlled study. J. Rheumatol. 1998, 25, 2385–2388. [Google Scholar]
  22. Barendregt, P.J.; van Den Meiracker, A.H.; Markusse, H.M.; Tulen, J.H.; Boomsma, F.; van Der Heijde, G.L.; Veld, A.J. Parasympathetic failure does not contribute to ocular dryness in primary Sjögren’s syndrome. Ann. Rheum. Dis. 1999, 58, 746–750. [Google Scholar] [CrossRef] [Green Version]
  23. Mandl, T.; Jacobsson, L.; Lilja, B.; Sundkvist, G.; Manthorpe, R. Disturbances of autonomic nervous function in primary Sjögren’s syndrome. Scand. J. Rheumatol. 1997, 26, 401–406. [Google Scholar] [CrossRef]
  24. Mandl, T.; Bornmyr, S.V.; Castenfors, J.; Jacobsson, L.T.; Manthorpe, R.; Wollmer, P. Sympathetic dysfunction in patients with primary Sjögren’s syndrome. J. Rheumatol. 2001, 28, 296–301. [Google Scholar]
  25. Mandl, T.; Wollmer, P.; Manthorpe, R.; Jacobsson, L.T. Autonomic and orthostatic dysfunction in primary Sjögren’s syndrome. J. Rheumatol. 2007, 34, 1869–1874. [Google Scholar]
  26. Mandl, T.; Granberg, V.; Apelqvist, J.; Wollmer, P.; Manthorpe, R.; Jacobsson, L.T. Autonomic nervous symptoms in primary Sjogren’s syndrome. Rheumatology 2008, 47, 914–919. [Google Scholar] [CrossRef] [Green Version]
  27. Mandl, T.; Hammar, O.; Theander, E.; Wollmer, P.; Ohlsson, B. Autonomic nervous dysfunction development in patients with primary Sjögren’s syndrome: A follow-up study. Rheumatology 2010, 49, 1101–1106. [Google Scholar] [CrossRef] [Green Version]
  28. Kovács, L.; Paprika, D.; Tákacs, R.; Kardos, A.; Várkonyi, T.T.; Lengyel, C.; Kovács, A.; Rudas, L.; Pokorny, G. Cardiovascular autonomic dysfunction in primary Sjögren’s syndrome. Rheumatology 2004, 43, 95–99. [Google Scholar] [CrossRef] [Green Version]
  29. Niemelä, R.K.; Pikkujämsä, S.M.; Hakala, M.; Huikuri, H.V.; Airaksinen, K.E. No signs of autonomic nervous system dysfunction in primary Sjörgen’s syndrome evaluated by 24 h heart rate variability. J. Rheumatol. 2000, 27, 2605–2610. [Google Scholar]
  30. Tumiati, B.; Perazzoli, F.; Negro, A.; Pantaleoni, M.; Regolisti, G. Heart rate variability in patients with Sjögren’s syndrome. Clin. Rheumatol. 2000, 19, 477–480. [Google Scholar] [CrossRef] [PubMed]
  31. Barendregt, P.J.; Tulen, J.H.; van den Meiracker, A.H.; Markusse, H.M. Spectral analysis of heart rate and blood pressure variability in primary Sjögren’s syndrome. Ann. Rheum. Dis. 2002, 61, 232–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Cai, F.Z.; Lester, S.; Lu, T.; Keen, H.; Boundy, K.; Proudman, S.M.; Tonkin, A.; Rischmueller, M. Mild autonomic dysfunction in primary Sjögren’s syndrome: A controlled study. Arthritis Res. Ther. 2008, 10, R31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ng, W.F.; Stangroom, A.J.; Davidson, A.; Wilton, K.; Mitchell, S.; Newton, J.L. Primary Sjogrens syndrome is associated with impaired autonomic response to orthostasis and sympathetic failure. QJM 2012, 105, 1191–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Koh, J.H.; Kwok, S.K.; Lee, J.; Park, S.H. Autonomic dysfunction in primary Sjögren’s syndrome: A prospective cohort analysis of 154 Korean patients. Korean J. Intern. Med. 2017, 32, 165–173. [Google Scholar] [CrossRef] [PubMed]
  35. Brunetta, E.; Shiffer, D.; Mandelli, P.; Achenza, S.; Folci, M.; Zumbo, A.; Minonzio, M.; Cairo, B.; Jacob, G.; Boccassini, L.; et al. Autonomic Abnormalities in Patients With Primary Sjögren’s Syndrome-Preliminary Results. Front. Physiol. 2019, 10, 1104. [Google Scholar] [CrossRef] [Green Version]
  36. Gemignani, F.; Manganelli, P.; Pavesi, G.; Marbini, A. Polyneuropathy in Sjögren’s syndrome. A case of prevalently autonomic neuropathy with tonic pupil and hypohidrosis. Funct. Neurol. 1988, 3, 337–348. [Google Scholar]
  37. McCombe, P.A.; Sheean, G.L.; McLaughlin, D.B.; Pender, M.P. Vestibular and ventilatory dysfunction in sensory and autonomic neuropathy associated with primary Sjörgren’s syndrome. J. Neurol. Neurosurg. Psychiatry 1992, 55, 1211–1212. [Google Scholar] [CrossRef] [Green Version]
  38. Kumazawa, K.; Sobue, G.; Yamamoto, K.; Mitsuma, T. Segmental anhidrosis in the spinal dermatomes in Sjögren’s syndrome-associated neuropathy. Neurology 1993, 43, 1820–1823. [Google Scholar] [CrossRef]
  39. Kumazawa, K.; Sobue, G.; Yamamoto, K.; Shimada, N.; Mitsuma, T. Autonomic dysfunction in sensory ataxic neuropathy with Sjögren’s syndrome. Rinsho Shinkeigaku 1993, 33, 1059–1065. (In Japanese) [Google Scholar]
  40. Gemignani, F.; Marbini, A.; Pavesi, G.; Di Vittorio, S.; Manganelli, P.; Cenacchi, G.; Mancia, D. Peripheral neuropathy associated with primary Sjögren’s syndrome. J. Neurol. Neurosurg. Psychiatry 1994, 57, 983–986. [Google Scholar] [CrossRef] [Green Version]
  41. Andonopoulos, A.P.; Ballas, C. Autonomic cardiovascular neuropathy in primary Sjögren’s syndrome. Rheumatol. Int. 1995, 15, 127–129. [Google Scholar] [CrossRef] [PubMed]
  42. Yasuda, T.; Kumazawa, K.; Sobue, G. Sensory ataxic neuropathy associated with Sjögren’s syndrome. Nihon Rinsho 1995, 53, 2568–2573. [Google Scholar] [PubMed]
  43. Barendregt, P.J.; Markusse, H.M.; Man In ‘t Veld, A.J. Primary Sjögren’s syndrome presenting as autonomic neuropathy. Case report. Neth. J. Med. 1998, 53, 196–200. [Google Scholar] [CrossRef]
  44. Chikazawa, H.; Nishiya, K.; Hashimoto, K. Peripheral neuropathy and dysautonomia in a patient with primary Sjögren’s syndrome. Nihon Rinsho Meneki Gakkai Kaishi 1998, 21, 57–63. (In Japanese) [Google Scholar] [CrossRef] [Green Version]
  45. Sorajja, P.; Poirier, M.K.; Bundrick, J.B.; Matteson, E.L. Autonomic failure and proximal skeletal myopathy in a patient with primary Sjögren syndrome. Mayo Clinic Proc. 1999, 74, 695–697. [Google Scholar] [CrossRef] [Green Version]
  46. Goto, H.; Matsuo, H.; Fukudome, T.; Shibuya, N.; Ohnishi, A.; Nakamura, H. Chronic autonomic neuropathy in a patient with primary Sjögren’s syndrome. J. Neurol. Neurosurg. Psychiatry 2000, 69, 135. [Google Scholar] [CrossRef] [Green Version]
  47. Shimoyama, M.; Ohtahara, A.; Okamura, T.; Watanabe, M.; Fujimoto, Y.; Teshima, S.; Takeda, S.; Hisatome, I.; Shigamasa, C. Isolated autonomic cardiovascular neuropathy in a patient with primary Sjögren syndrome: A case of successful treatment with glucocorticoid. Am. J. Med. Sci. 2002, 324, 170–172. [Google Scholar] [CrossRef]
  48. Nakazato, Y.; Tamura, N.; Yamamoto, T.; Abe, T.; Shimazu, K. A case of Sjögren’s syndrome with heat intolerance induced by generalized anhidrosis as its principal feature. Rinsho Shinkeigaku 2002, 42, 171–174. [Google Scholar]
  49. Sakakibara, R.; Hirano, S.; Asahina, M.; Sawai, S.; Nemoto, Y.; Hiraga, A.; Uchiyama, T.; Hattori, T. Primary Sjögren’s syndrome presenting with generalized autonomic failure. Eur. J. Neurol. 2004, 11, 635–638. [Google Scholar] [CrossRef]
  50. Mori, K.; Iijima, M.; Koike, H.; Hattori, N.; Tanaka, F.; Watanabe, H.; Katsuno, M.; Fujita, A.; Aiba, I.; Ogata, A.; et al. The wide spectrum of clinical manifestations in Sjögren’s syndrome-associated neuropathy. Brain 2005, 128, 2518–2534. [Google Scholar] [CrossRef] [Green Version]
  51. Luong, M.; Jomir, L.; Labauge, P.; Dandurand, M.; Meunier, L.; Stoebner, P.E. Ross syndrome with sweating anomaly associated with Sjögren syndrome: An infrared thermo-graphic case study. Acta Derm. Venereol. 2011, 91, 80–81. [Google Scholar] [CrossRef] [Green Version]
  52. Koike, H.; Sobue, G. Sjogren’s syndrome-associated neuropathy. Brain Nerve 2013, 65, 1333–1342. (In Japanese) [Google Scholar]
  53. Bhagwan, S.; Bhagwan, B.; Moodley, A. Bilateral Tonic Pupils as the Initial Manifestation of Sjögren’s Syndrome. Neuroophthalmology 2015, 39, 248–252. [Google Scholar] [CrossRef] [Green Version]
  54. Adamec, I.; Žarković, K.; Sentić, M.; Habek, M. Autonomic failure in Sjögren’s syndrome. Clin. Auton. Res. 2016, 26, 165–166. [Google Scholar] [CrossRef]
  55. Sivadasan, A.; Muthusamy, K.; Patel, B.; Benjamin, R.N.; Prabhakar, A.T.; Mathew, V.; Aaron, S.; Alexander, M. Clinical Spectrum, Therapeutic Outcomes, and Prognostic Predictors in Sjogren’s Syndrome-associated Neuropathy. Ann. Indian Acad. Neurol. 2017, 20, 278–283. [Google Scholar]
  56. Goodman, B.P. Immunoresponsive Autonomic Neuropathy in Sjögren Syndrome-Case Series and Literature Review. Am. J. Ther. 2019, 26, e66–e71. [Google Scholar] [CrossRef]
  57. Waterschoot, M.P.; Guerit, J.M.; Lambert, M.; de Barsy, T. Bilateral tonic pupils and polyneuropathy in Sjögren’s syndrome: A common pathophysiological mechanism? Eur. Neurol. 1991, 31, 114–116. [Google Scholar] [CrossRef]
  58. Pavlakis, P.P.; Alexopoulos, H.; Kosmidis, M.L.; Mamali, I.; Moutsopoulos, H.M.; Tzioufas, A.G.; Dalakas, M.C. Peripheral neuropathies in Sjögren’s syndrome: A critical update on clinical features and pathogenetic mechanisms. J. Autoimmun. 2012, 39, 27–33. [Google Scholar] [CrossRef]
  59. Elenkov, I.J.; Kovács, K.; Duda, E.; Stark, E.; Vizi, E.S. Presynaptic inhibitory effect of TNF-alpha on the release of noradrenaline in isolated median eminence. J. Neuroimmunol. 1992, 41, 117–120. [Google Scholar] [CrossRef]
  60. Straub, R.H.; Baerwald, C.G.; Wahle, M.; Jänig, W. Autonomic dysfunction in rheumatic diseases. Rheum. Dis. Clin. N. Am. 2005, 31, 61–75. [Google Scholar] [CrossRef]
  61. Bacman, S.; Sterin-Borda, L.; Camusso, J.J.; Arana, R.; Hubscher, O.; Borda, E. Circulating antibodies against rat parotid gland M3 muscarinic receptors in primary Sjögren’s syndrome. Clin. Exp. Immunol. 1996, 104, 454–459. [Google Scholar] [CrossRef] [PubMed]
  62. Bacman, S.; Berra, A.; Sterin-Borda, L.; Borda, E. Muscarinic acetylcholine receptor antibodies as a new marker of dry eye Sjögren syndrome. Investig. Ophthalmol. Vis. Sci. 2001, 42, 321–327. [Google Scholar]
  63. Dawson, L.; Tobin, A.; Smith, P.; Gordon, T. Antimuscarinic antibodies in Sjögren’s syndrome: Where are we, and where are we going? Arthritis Rheum. 2005, 52, 2984–2995. [Google Scholar] [CrossRef] [PubMed]
  64. Sumida, T.; Tsuboi, H.; Iizuka, M.; Asashima, H.; Matsumoto, I. Anti-M3 muscarinic acetylcholine receptor antibodies in patients with Sjögren’s syndrome. Mod. Rheumatol. 2013, 23, 841–845. [Google Scholar] [CrossRef] [PubMed]
  65. Yu, X.; Riemekasten, G.; Petersen, F. Autoantibodies against muscarinic acetylcholine receptor M3 in Sjögren’s syndrome and corresponding mouse models. Front. Biosci. (Landmark Ed.) 2018, 23, 2053–2064. [Google Scholar] [CrossRef]
  66. Dawson, L.J.; Stanbury, J.; Venn, N.; Hasdimir, B.; Rogers, S.N.; Smith, P.M. Antimuscarinic antibodies in primary Sjögren’s syndrome reversibly inhibit the mechanism of fluid secretion by human submandibular salivary acinar cells. Arthritis Rheum. 2006, 54, 1165–1173. [Google Scholar] [CrossRef]
  67. Park, K.; Haberberger, R.V.; Gordon, T.P.; Jackson, M.W. Antibodies interfering with the type 3 muscarinic receptor pathway inhibit gastrointestinal motility and cholinergic neurotransmission in Sjögren’s syndrome. Arthritis Rheum. 2011, 63, 1426–1434. [Google Scholar] [CrossRef]
  68. Waterman, S.A.; Gordon, T.P.; Rischmueller, M. Inhibitory effects of muscarinic receptor autoantibodies on parasympathetic neurotransmission in Sjögren’s syndrome. Arthritis Rheum. 2000, 43, 1647–1654. [Google Scholar] [CrossRef]
  69. Wang, F.; Jackson, M.W.; Maughan, V.; Cavill, D.; Smith, A.J.; Waterman, S.A.; Gordon, T.P. Passive transfer of Sjogren’s syndrome IgG produces the pathophysiology of overactive bladder. Arthritis Rheum. 2004, 50, 3637–3645. [Google Scholar] [CrossRef]
  70. Smith, A.J.; Jackson, M.W.; Wang, F.; Cavill, D.; Rischmueller, M.; Gordon, T.P. Neutralization of muscarinic receptor autoantibodies by intravenous immunoglobulin in Sjögren syndrome. Hum. Immunol. 2005, 66, 411–416. [Google Scholar] [CrossRef]
  71. Denton, C.P.; Khanna, D. Systemic sclerosis. Lancet 2017, 390, 1685–1699. [Google Scholar] [CrossRef]
  72. Sonnex, C.; Paice, E.; White, A.G. Autonomic neuropathy in systemic sclerosis: A case report and evaluation of six patients. Ann. Rheum. Dis. 1986, 45, 957–960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Dessein, P.H.; Gledhill, R.F. More on autonomic neuropathy in systemic sclerosis. Ann. Rheum. Dis. 1988, 47, 261–263. [Google Scholar] [CrossRef] [Green Version]
  74. Klimiuk, P.S.; Taylor, L.; Baker, R.D.; Jayson, M.I. Autonomic neuropathy in systemic sclerosis. Ann. Rheum. Dis. 1988, 47, 542–545. [Google Scholar] [CrossRef]
  75. Cerinic, M.M.; Generini, S.; Pignone, A.; Casale, R. The nervous system in systemic sclerosis (scleroderma). Clinical features and pathogenetic mechanisms. Rheum. Dis. Clin. N. Am. 1996, 22, 879–892. [Google Scholar] [CrossRef]
  76. Kister, I.; Inglese, M.; Laxer, R.M.; Herbert, J. Neurologic manifestations of localized scleroderma: A case report and literature review. Neurology 2008, 71, 1538–1545. [Google Scholar] [CrossRef]
  77. McMahan, Z.H. Gastrointestinal involvement in systemic sclerosis: An update. Curr. Opin. Rheumatol. 2019, 31, 561–568. [Google Scholar] [CrossRef]
  78. Di Franco, M.; Paradiso, M.; Riccieri, V.; Basili, S.; Mammarella, A.; Valesini, G. Autonomic dysfunction and microvascular damage in systemic sclerosis. Clin. Rheumatol. 2007, 26, 1278–1283. [Google Scholar] [CrossRef]
  79. Rodrigues, G.D.; Tobaldini, E.; Bellocchi, C.; Santaniello, A.; Caronni, M.; Severino, A.; Froldi, M.; Beretta, L.; da Silva Soares, P.P.; Montano, N. Cardiac autonomic modulation at rest and during orthostatic stress among different systemic sclerosis subsets. Eur. J. Intern. Med. 2019, 66, 75–80. [Google Scholar] [CrossRef]
  80. Tadic, M.; Zlatanovic, M.; Cuspidi, C.; Stevanovic, A.; Celic, V.; Damjanov, N.; Kocijancic, V.; Tadic, M.; Zlatanovic, M.; Cuspidi, C.; et al. Systemic sclerosis impacts right heart and cardiac autonomic nervous system. J. Clin. Ultrasound 2018, 46, 188–194. [Google Scholar] [CrossRef]
  81. Gigante, A.; Galea, N.; Borrazzo, C.; Tubani, L.; Liberatori, M.; Ciolina, F.; Fiorelli, A.; Romaniello, A.; Barbano, B.; Romaggioli, L.; et al. Role of autonomic dysfunction in the regulation of myocardial blood flow in systemic sclerosis evaluated by cardiac magnetic resonance. Int. J. Rheum. Dis. 2019, 22, 1029–1035. [Google Scholar] [CrossRef] [PubMed]
  82. Cozzolino, D.; Naclerio, C.; Iengo, R.; D’Angelo, S.; Cuomo, G.; Valentini, G. Cardiac autonomic dysfunction precedes the development of fibrosis in patients with systemic sclerosis. Rheumatology 2002, 41, 586–588. [Google Scholar] [CrossRef] [Green Version]
  83. Malandrini, A.; Selvi, E.; Villanova, M.; Berti, G.; Sabadini, L.; Salvadori, C.; Gambelli, S.; De Stefano, R.; Vernillo, R.; Marcolongo, R.; et al. Autonomic nervous system and smooth muscle cell involvement in systemic sclerosis: Ultrastructural study of 3 cases. J. Rheumatol. 2000, 27, 1203–1206. [Google Scholar] [PubMed]
  84. Iovino, P.; Valentini, G.; Ciacci, C.; De Luca, A.; Tremolaterra, F.; Sabbatini, F.; Tirri, E.; Mazzacca, G. Proximal stomach function in systemic sclerosis: Relationship with autonomic nerve function. Dig. Dis. Sci. 2001, 46, 723–730. [Google Scholar] [CrossRef]
  85. Lock, G.; Straub, R.H.; Zeuner, M.; Antoniou, E.; Holstege, A.; Schölmerich, J.; Lang, B. Association of autonomic nervous dysfunction and esophageal dysmotility in systemic sclerosis. J. Rheumatol. 1998, 25, 1330–1335. [Google Scholar] [PubMed]
  86. Stacher, G.; Merio, R.; Budka, C.; Schneider, C.; Smolen, J.; Tappeiner, G. Cardiovascular autonomic function, autoantibodies, and esophageal motor activity in patients with systemic sclerosis and mixed connective tissue disease. J. Rheumatol. 2000, 27, 692–697. [Google Scholar]
  87. Adler, B.L.; Russell, J.W.; Hummers, L.K.; McMahan, Z.H. Symptoms of Autonomic Dysfunction in Systemic Sclerosis Assessed by the COMPASS-31 Questionnaire. J. Rheumatol. 2018, 45, 1145–1152. [Google Scholar] [CrossRef]
  88. Straub, R.H.; Zeuner, M.; Lock, G.; Rath, H.; Hein, R.; Schölmerich, J.; Lang, B. Autonomic and sensorimotor neuropathy in patients with systemic lupus erythematosus and systemic sclerosis. J. Rheumatol. 1996, 23, 87–92. [Google Scholar]
  89. Del Rosso, A.; Bertinotti, L.; Pietrini, U.; Messori, A.; Fanciullacci, M.; Casale, R.; Giacomelli, R.; Generini, S.; Sicuteri, R.; Pignone, A.; et al. Pupillocynetic activity of substance P in systemic sclerosis. J. Rheumatol. 2003, 30, 1231–1237. [Google Scholar]
  90. Venkataraman, A.; Panda, B.B.; Sirka, C. Adie’s Tonic Pupil in Systemic Sclerosis: A Rare Association. Case Rep. Ophthalmol. Med. 2015, 2015, 491795. [Google Scholar] [CrossRef] [Green Version]
  91. Raszewa, M.; Hausmanowa-Petrusewicz, I.; Błaszczyk, M.; Jabłońska, S. Sympathetic skin response in scleroderma. Electromyogr. Clin. Neurophysiol. 1991, 31, 467–472. [Google Scholar] [PubMed]
  92. Zakrzewska-Pniewska, B.; Jabłońska, S.; Kowalska-Oledzka, E.; Błaszczyk, M.; Hausmanowa-Petrusewicz, I. Sympathetic skin response in scleroderma, scleroderma overlap syndromes and inflammatory myopathies. Clin. Rheumatol. 1999, 18, 473–480. [Google Scholar] [CrossRef] [PubMed]
  93. Badry, R.; Gamal, R.M.; Hassanien, M.M.; El Hamed, M.A.; Hammam, N.; El Fawal, B.M. Sympathetic skin response in patients with systemic sclerosis and rheumatoid arthritis. Egypt J. Neurol. Psychiatr. Neurosurg. 2018, 54, 38. [Google Scholar] [CrossRef] [Green Version]
  94. Minervini, R.; Morelli, G.; Minervini, A.; Pampaloni, S.; Tognetti, A.; Fiorentini, L.; Ciompi, M.L. Bladder involvement in systemic sclerosis: Urodynamic and histological evaluation in 23 patients. Eur. Urol. 1998, 34, 47–52. [Google Scholar] [CrossRef]
  95. Casale, R.; Generini, S.; Luppi, F.; Pignone, A.; Matucci-Cerinic, M. Pulse cyclophosphamide decreases sympathetic postganglionic activity, controls alveolitis, and normalizes vascular tone dysfunction (Raynaud’s phenomenon) in a case of early systemic sclerosis. Arthritis Rheum. 2004, 51, 665–669. [Google Scholar] [CrossRef] [Green Version]
  96. Berger, M.; Steen, V.D. Role of anti-receptor autoantibodies in pathophysiology of scleroderma. Autoimmun. Rev. 2017, 16, 1029–1035. [Google Scholar] [CrossRef]
  97. Kumar, S.; Singh, J.; Rattan, S.; DiMarino, A.J.; Cohen, S.; Jimenez, S.A. Review article: Pathogenesis and clinical manifestations of gastrointestinal involvement in systemic sclerosis. Aliment. Pharmacol. Ther. 2017, 45, 883–898. [Google Scholar] [CrossRef]
  98. Howe, S.; Eaker, E.Y.; Sallustio, J.E.; Peebles, C.; Tan, E.M.; Williams, R.C., Jr. Antimyenteric neuronal antibodies in scleroderma. J. Clin. Investig. 1994, 94, 761–770. [Google Scholar] [CrossRef] [Green Version]
  99. Eaker, E.Y.; Kuldau, J.G.; Verne, G.N.; Ross, S.O.; Sallustio, J.E. Myenteric neuronal antibodies in scleroderma: Passive transfer evokes alterations in intestinal myoelectric activity in a rat model. J. Lab. Clin. Med. 1999, 133, 551–556. [Google Scholar] [CrossRef]
  100. Goldblatt, F.; Gordon, T.P.; Waterman, S.A. Antibody-mediated gastrointestinal dysmotility in scleroderma. Gastroenterology 2002, 123, 1144–1150. [Google Scholar] [CrossRef]
  101. Kawaguchi, Y.; Nakamura, Y.; Matsumoto, I.; Nishimagi, E.; Satoh, T.; Kuwana, M.; Sumida, T.; Hara, M. Muscarinic-3 acetylcholine receptor autoantibody in patients with systemic sclerosis: Contribution to severe gastrointestinal tract dysmotility. Ann. Rheum. Dis. 2009, 68, 710–714. [Google Scholar] [CrossRef] [PubMed]
  102. Singh, J.; Mehendiratta, V.; Del Galdo, F.; Jimenez, S.A.; Cohen, S.; DiMarino, A.J.; Rattan, S. Immunoglobulins from scleroderma patients inhibit the muscarinic receptor activation in internal anal sphincter smooth muscle cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 297, G1206–G1213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kumar, S.; Singh, J.; Kedika, R.; Mendoza, F.; Jimenez, S.A.; Blomain, E.S.; DiMarino, A.J.; Cohen, S.; Rattan, S. Role of muscarinic-3 receptor antibody in systemic sclerosis: Correlation with disease duration and effects of IVIG. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G1052–G1060. [Google Scholar] [CrossRef] [PubMed]
  104. McMahan, Z.H.; Domsic, R.T.; Zhu, L.; Medsger, T.A.; Casciola-Rosen, L.; Shah, A.A. Anti-RNPC-3 (U11/U12) Antibodies in Systemic Sclerosis in Patients With Moderate-to-Severe Gastrointestinal Dysmotility. Arthritis Care Res. 2019, 71, 1164–1170. [Google Scholar] [CrossRef] [PubMed]
  105. Scott, D.L.; Wolfe, F.; Huizinga, T.W. Rheumatoid arthritis. Lancet 2010, 376, 1094–1108. [Google Scholar] [CrossRef]
  106. Edmonds, M.E.; Jones, T.C.; Saunders, W.A.; Sturrock, R.D. Autonomic neuropathy in rheumatoid arthritis. Br. Med. J. 1979, 2, 173–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Leden, I.; Eriksson, A.; Lilja, B.; Sturfelt, G.; Sundkvist, G. Autonomic nerve function in rheumatoid arthritis of varying severity. Scand. J. Rheumatol. 1983, 12, 166–170. [Google Scholar] [CrossRef]
  108. Tan, J.; Akin, S.; Beyazova, M.; Sepici, V.; Tan, E. Sympathetic skin response and R-R interval variation in rheumatoid arthritis. Two simple tests for the assessment of autonomic function. Am. J. Phys. Med. Rehabil. 1993, 72, 196–203. [Google Scholar] [CrossRef]
  109. Bekkelund, S.I.; Jorde, R.; Husby, G.; Mellgren, S.I. Autonomic nervous system function in rheumatoid arthritis. A controlled study. J. Rheumatol. 1996, 23, 1710–1714. [Google Scholar]
  110. Louthrenoo, W.; Ruttanaumpawan, P.; Aramrattana, A.; Sukitawut, W. Cardiovascular autonomic nervous system dysfunction in patients with rheumatoid arthritis and systemic lupus erythematosus. QJM 1999, 92, 97–102. [Google Scholar] [CrossRef] [Green Version]
  111. Gozke, E.; Erdogan, N.; Akyuz, G.; Turan, B.; Akyuz, E.; Us, O. Sympathetic skin response and R-R interval variation in cases with rheumatoid arthritis. Electromyogr. Clin. Neurophysiol. 2003, 43, 81–84. [Google Scholar] [PubMed]
  112. Schwemmer, S.; Beer, P.; Schölmerich, J.; Fleck, M.; Straub, R.H. Cardiovascular and pupillary autonomic nervous dysfunction in patients with rheumatoid arthritis-a cross-sectional and longitudinal study. Clin. Exp. Rheumatol. 2006, 24, 683–689. [Google Scholar] [PubMed]
  113. Stojanovich, L.; Milovanovich, B.; de Luka, S.R.; Popovich-Kuzmanovich, D.; Bisenich, V.; Djukanovich, B.; Randjelovich, T.; Krotin, M. Cardiovascular autonomic dysfunction in systemic lupus, rheumatoid arthritis, primary Sjögren syndrome and other autoimmune diseases. Lupus 2007, 16, 181–185. [Google Scholar] [CrossRef] [PubMed]
  114. Aydemir, M.; Yazisiz, V.; Basarici, I.; Avci, A.B.; Erbasan, F.; Belgi, A.; Terzioglu, E. Cardiac autonomic profile in rheumatoid arthritis and systemic lupus erythematosus. Lupus 2010, 19, 255–261. [Google Scholar] [CrossRef] [PubMed]
  115. Aggarwal, D.; Singla, S. Prevalence of Autonomic Neuropathy in Patients of Rheumatoid Arthritis and Its Correlation with Disease Severity. J. Clin. Diagn. Res. 2017, 11, OC09–OC13. [Google Scholar] [CrossRef]
  116. Adlan, A.M.; Paton, J.F.; Lip, G.Y.; Kitas, G.D.; Fisher, J.P. Increased sympathetic nerve activity and reduced cardiac baroreflex sensitivity in rheumatoid arthritis. J. Physiol. 2017, 595, 967–981. [Google Scholar] [CrossRef] [Green Version]
  117. Dekkers, J.C.; Geenen, R.; Godaert, G.L.; Bijlsma, J.W.; van Doornen, L.J. Elevated sympathetic nervous system activity in patients with recently diagnosed rheumatoid arthritis with active disease. Clin. Exp. Rheumatol. 2004, 22, 63–70. [Google Scholar]
  118. Geenen, R.; Godaert, G.L.; Jacobs, J.W.; Peters, M.L.; Bijlsma, J.W. Diminished autonomic nervous system responsiveness in rheumatoid arthritis of recent onset. J. Rheumatol. 1996, 23, 258–264. [Google Scholar]
  119. Anichkov, D.A.; Shostak, N.A.; Ivanov, D.S. Heart rate variability is related to disease activity and smoking in rheumatoid arthritis patients. Int. J. Clin. Pract. 2007, 61, 777–783. [Google Scholar] [CrossRef]
  120. Yadav, R.K.; Gupta, R.; Deepak, K.K. A pilot study on short term heart rate variability & its correlation with disease activity in Indian patients with rheumatoid arthritis. Indian J. Med. Res. 2012, 136, 593–598. [Google Scholar]
  121. Syngle, V.; Syngle, A.; Garg, N.; Krishan, P.; Verma, I. Predictors of autonomic neuropathy in rheumatoid arthritis. Auton. Neurosci. 2016, 201, 54–59. [Google Scholar] [CrossRef] [PubMed]
  122. Holman, A.J.; Ng, E. Heart rate variability predicts anti-tumor necrosis factor therapy response for inflammatory arthritis. Auton. Neurosci. 2008, 143, 58–67. [Google Scholar] [CrossRef] [PubMed]
  123. Dicou, E.; Hurez, D.; Nerrière, V. Natural autoantibodies against the nerve growth factor in autoimmune diseases. J. Neuroimmunol. 1993, 47, 159–167. [Google Scholar] [CrossRef]
  124. Maule, S.; Quadri, R.; Mirante, D.; Pellerito, R.A.; Marucco, E.; Marinone, C.; Vergani, D.; Chiandussi, L.; Zanone, M.M. Autonomic nervous dysfunction in systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA): Possible pathogenic role of autoantibodies to autonomic nervous structures. Clin. Exp. Immunol. 1997, 110, 423–427. [Google Scholar] [CrossRef]
  125. Syngle, A.; Verma, I.; Krishan, P. Interleukin-6 blockade improves autonomic dysfunction in rheumatoid arthritis. Acta Reumatol. Port. 2015, 40, 85–88. [Google Scholar]
  126. Saraswathi, P.V.; Neelambikai, N.; Mahesh, A.; Govindarajan, K. Cardiovascular parasympathetic nervous system dysfunction in female rheumatoid arthritis patients. Indian J. Physiol. Pharmacol. 2013, 57, 23–30. [Google Scholar]
  127. Kosek, E.; Altawil, R.; Kadetoff, D.; Finn, A.; Westman, M.; Le Maître, E.; Andersson, M.; Jensen-Urstad, M.; Lampa, J. Evidence of different mediators of central inflammation in dysfunctional and inflammatory pain—Interleukin-8 in fibromyalgia and interleukin-1 β in rheumatoid arthritis. J. Neuroimmunol. 2015, 280, 49–55. [Google Scholar] [CrossRef] [Green Version]
  128. Di Battista, M.; Marcucci, E.; Elefante, E.; Tripoli, A.; Governato, G.; Zucchi, D.; Tani, C.; Alunno, A. One year in review 2018: Systemic lupus erythematosus. Clin. Exp. Rheumatol. 2018, 36, 763–777. [Google Scholar]
  129. Liang, M.H.; Corzillius, M.; Bae, S.C.; Lew, R.A.; Fortin, P.R.; Gordon, C.; Isenberg, D.; Alarcón, G.S.; Straaton, K.V.; Denburg, J.; et al. The American College of Rheumatology nomenclature and case definitions for neuropsychiatric lupus syndromes. Arthritis Rheum. 1999, 42, 599–608. [Google Scholar]
  130. Milovanović, B.; Stojanović, L.; Milićevik, N.; Vasić, K.; Bjelaković, B.; Krotin, M. Cardiac autonomic dysfunction in patients with systemic lupus, rheumatoid arthritis and sudden death risk. Srp. Arh. Celok. Lek. 2010, 138, 26–32. [Google Scholar] [CrossRef]
  131. Yorgun, H.; Canpolat, U.; Aytemir, K.; Ateş, A.H.; Kaya, E.B.; Akdoğan, A.; Sunman, H.; Gökçay Canpolat, A.; Çalgüneri, M.; Kabakçi, G.; et al. Evaluation of cardiac autonomic functions in patients with systemic lupus erythematosus. Lupus 2012, 21, 373–379. [Google Scholar] [CrossRef] [PubMed]
  132. Poliwczak, A.R.; Waszczykowska, E.; Dziankowska-Bartkowiak, B.; Koziróg, M.; Dworniak, K. The use of heart rate turbulence and heart rate variability in the assessment of autonomic regulation and circadian rhythm in patients with systemic lupus erythematosus without apparent heart disease. Lupus 2018, 27, 436–444. [Google Scholar] [CrossRef] [PubMed]
  133. Haghighat, S.; Fatemi, A.; Andalib, S. The autonomic dysfunction in patients with lupus disease: An electrophysiological study. Adv. Biomed. Res. 2016, 5, 102. [Google Scholar] [CrossRef] [PubMed]
  134. Thanou, A.; Stavrakis, S.; Dyer, J.W.; Munroe, M.E.; James, J.A.; Merrill, J.T. Impact of heart rate variability, a marker for cardiac health, on lupus disease activity. Arthritis Res. Ther. 2016, 18, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Matusik, P.S.; Matusik, P.T.; Stein, P.K. Heart rate variability in patients with systemic lupus erythematosus: A systematic review and methodological considerations. Lupus 2018, 27, 1225–1239. [Google Scholar] [CrossRef] [PubMed]
  136. Malpas, S.C. Sympathetic nervous system overactivity and its role in the development of cardiovascular disease. Physiol. Rev. 2010, 90, 513–557. [Google Scholar] [CrossRef]
  137. Skamra, C.; Ramsey-Goldman, R. Management of cardiovascular complications in systemic lupus erythematosus. Int. J. Clin. Rheumtol. 2010, 5, 75–100. [Google Scholar] [CrossRef] [Green Version]
  138. Hoyle, C.; Ewing, D.J.; Parker, A.C. Acute autonomic neuropathy in association with systemic lupus erythematosus. Ann. Rheum. Dis. 1985, 44, 420–424. [Google Scholar] [CrossRef]
  139. McCombe, P.A.; McLeod, J.G.; Pollard, J.D.; Guo, Y.P.; Ingall, T.J. Peripheral sensorimotor and autonomic neuropathy associated with systemic lupus erythematosus. Clinical, pathological and immunological features. Brain 1987, 110, 533–549. [Google Scholar] [CrossRef]
  140. Jodo, S.; Sagawa, A.; Ogura, N.; Atsumi, T.; Amasaki, Y.; Nakabayashi, T.; Watanabe, I.; Mukai, M.; Fujisaku, A.; Nakagawa, S. A case of systemic lupus erythematosus (SLE) developing pan-dysautonomia. Ryumachi 1992, 32, 58–65. [Google Scholar]
  141. Gledhill, R.F.; Dessein, P.H. Autonomic neuropathy in systemic lupus erythematosus. J. Neurol. Neurosurg. Psychiatry 1988, 51, 1238–1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Arruda, W.O.; Teive, H.A.; Ramina, R.; Wunder, P.R.; Rocha, L.C. Autonomic neuropathy in systemic lupus erythematosus. J. Neurol. Neurosurg. Psychiatry 1989, 52, 539–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Kondo, T.; Inoue, H.; Usui, T.; Mimori, T.; Tomimoto, H.; Vernino, S.; Takahashi, R. Autoimmune autonomic ganglionopathy with Sjögren’s syndrome: Significance of ganglionic acetylcholine receptor antibody and therapeutic approach. Auton. Neurosci. 2009, 146, 33–35. [Google Scholar] [CrossRef] [PubMed]
  144. Yoshida, T.; Kinjo, M.; Nakane, S. Autoimmune autonomic ganglionopathy associated with Sjögren’s syndrome presenting with recurrent abdominal distension. BMJ Case Rep. 2018, 2018, Bcr-2017-223785. [Google Scholar] [CrossRef]
  145. Imamura, M.; Mukaino, A.; Nakane, S.; Takamatsu, K.; Watari, M.; Higuchi, O.; Maeda, Y.; Matsuo, H.; Ando, Y. Clinical characteristics of autonomic dysfunction in patients with Sjögren’s syndrome. Neuroimmunology 2017, 22, 90. [Google Scholar]
  146. Umeda, M.; Mitsuma, M.; Koga, T.; Ichinose, K.; Mukaino, A.; Kawano, H.; Higuchi, O.; Nakane, S.; Eguchi, K.; Ueki, T.; et al. Rheumatic diseases complicated with autoimmune autonomic ganglionopathy: Three cases. Kyushu J. Rheumatol. 2016, 36, 20–26. [Google Scholar]
Figure 1. Thermoregulatory sweat test in Patient 1 (AC): Pre-immunotherapy, (DF): Post-immunotherapy).
Figure 1. Thermoregulatory sweat test in Patient 1 (AC): Pre-immunotherapy, (DF): Post-immunotherapy).
Ijms 21 01332 g001
Figure 2. Anti-gAChR autoantibodies and autonomic dysfunction in autoimmune rheumatic diseases.
Figure 2. Anti-gAChR autoantibodies and autonomic dysfunction in autoimmune rheumatic diseases.
Ijms 21 01332 g002
Table 1. Autonomic function tests in Patient 1.
Table 1. Autonomic function tests in Patient 1.
Pre-ImmunotherapiesPost-ImmunotherapiesResponse to Immunotherapies
Pupillary responses to local instillation
Before local instillation (mm, R/L)5.0 */5.0 *4.0/4.0Improved
5% cocaine (mm, R/L)6.0 */6.0 *5.5/5.5
1% phenylephrine (mm, R/L)6.5 */5.5 *4.0/4.0
0.125% pilocarpine (mm, R/L)2.5 */3.53.5/3.5
Secretomotor function test
Gum test (mL/10 min)7.0 *12.0Improved
Schirmer tear test (mm/5 min)5 */2 *N.D. 7
Sudomotor and cutaneous vasomotor test
Thermoregulatory sweat test(Figure 1)Improved
Acetylcholine sweat testNo response *N.D.
Cardiovascular function test
BP1 response to postural change, BP (mmHg)171/101→143/87 *145/95→144/90Improved
HR2 response to postural change, HR (/min)65→9585→93
CVRR 3 (%)4.153.92(W.N.L. 8)
LF/HF 40.35 *0.20 *Not effective
Myocardial 123I-MIBG 5 scintigraphy, H/M ratio 6 (early)3.143.08(W.N.L.)
Myocardial 123I-MIBG scintigraphy, H/M ratio (delayed)3.824.04(W.N.L.)
* Mean abnormal value from autonomic function test data. 1 BP, blood pressure; 2 HR, heart rate; 3 CVRR, coefficient of variation in R-R intervals; 4 LF/HF, low frequency/high frequency; 5 MIBG, metaiodobenzylguanidine; 6 H/M ratio, heart-to-mediastinum ratio; 7 N.D., not done; 8 W.N.L., within normal limits.

Share and Cite

MDPI and ACS Style

Imamura, M.; Mukaino, A.; Takamatsu, K.; Tsuboi, H.; Higuchi, O.; Nakamura, H.; Abe, S.; Ando, Y.; Matsuo, H.; Nakamura, T.; et al. Ganglionic Acetylcholine Receptor Antibodies and Autonomic Dysfunction in Autoimmune Rheumatic Diseases. Int. J. Mol. Sci. 2020, 21, 1332. https://doi.org/10.3390/ijms21041332

AMA Style

Imamura M, Mukaino A, Takamatsu K, Tsuboi H, Higuchi O, Nakamura H, Abe S, Ando Y, Matsuo H, Nakamura T, et al. Ganglionic Acetylcholine Receptor Antibodies and Autonomic Dysfunction in Autoimmune Rheumatic Diseases. International Journal of Molecular Sciences. 2020; 21(4):1332. https://doi.org/10.3390/ijms21041332

Chicago/Turabian Style

Imamura, Michie, Akihiro Mukaino, Koutaro Takamatsu, Hiroto Tsuboi, Osamu Higuchi, Hideki Nakamura, Saori Abe, Yukio Ando, Hidenori Matsuo, Tadashi Nakamura, and et al. 2020. "Ganglionic Acetylcholine Receptor Antibodies and Autonomic Dysfunction in Autoimmune Rheumatic Diseases" International Journal of Molecular Sciences 21, no. 4: 1332. https://doi.org/10.3390/ijms21041332

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop