1
|
Cantrell C, Rilinger R, Stallkamp Tidd SJ, Wilson R. Corneal Confocal Microscopy in Postural Orthostatic Tachycardia Syndrome (POTS) as a Diagnostic Tool for Small Fiber Neuropathy. Cureus 2025; 17:e82781. [PMID: 40271232 PMCID: PMC12015762 DOI: 10.7759/cureus.82781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE Postural orthostatic tachycardia syndrome (POTS) is a debilitating condition characterized by autonomic dysregulation. Patients with this disorder may experience orthostatic intolerance, palpitations, fatigue, and a wide variety of other symptoms. The neuropathic symptoms of POTS may be caused by small fiber neuropathy (SFN), which is currently diagnosed using skin nerve biopsy. Corneal confocal microscopy (CCM) is an imaging modality that allows visualization of the corneal nerve layer. Our study aimed to determine whether CCM could detect differences in small nerve fiber parameters between POTS patients with and without signs or symptoms of SFN. MATERIALS AND METHODS CCM was performed on nine patients, along with a neurological examination. Participants were also asked about neuropathic symptoms by a researcher. Based on examination findings and/or reported symptoms, patients were categorized into SFN+ and SFN- groups for comparison. A chart review was conducted to gather demographic data, medications, autonomic testing results, and medical history, including common POTS comorbidities. RESULTS Comparison of nerve fiber parameters using CCM did not reveal a statistically significant difference between the groups. However, valuable insights were gained regarding the logistics of conducting this type of study in POTS patients, including adapting to challenges and improving coordination between the neurology and ophthalmology departments. CONCLUSIONS CCM may one day replace skin nerve biopsy as a diagnostic tool for SFN in POTS patients. Although this preliminary analysis did not demonstrate significant findings, likely due to the small sample size, we believe CCM may still have a role in POTS research and could eventually become a diagnostic tool used in autonomic clinics.
Collapse
Affiliation(s)
| | - Ryan Rilinger
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, USA
| | | | | |
Collapse
|
2
|
Murin PJ, Gao VD, Geisler S. Beyond pain: Using Unsupervised Machine Learning to Identify Phenotypic Clusters of Small Fiber Neuropathy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.09.24313341. [PMID: 39314965 PMCID: PMC11419207 DOI: 10.1101/2024.09.09.24313341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background and Objectives Small fiber neuropathy (SFN) is characterized by dysfunction and loss of peripheral unmyelinated and thinly myelinated nerve fibers, resulting in a phenotype that includes varying combinations of somatosensory and dysautonomia symptoms, which can be profoundly disabling and lead to decreased quality of life. Treatment aimed mainly at pain reduction, which may not target the underlying pathophysiology, is frequently ineffective. Another impediment to the effective management of SFN may be the significant between-patient heterogeneity. Accordingly, we launched this study to gain insights into the symptomatic variability of SFN and determine if SFN patients can be sub-grouped based on clinical characteristics. Methods To characterize the phenotype and investigate how patients with SFN differ from those with large fiber involvement, 105 patients with skin-biopsy proven SFN and 45 with mixed fiber neuropathy (MFN) were recruited. Using unsupervised machine learning, SFN patients were clustered based upon symptom concurrence and severity. Demographics, clinical data, symptoms, and skin biopsy- and laboratory findings were compared between the groups. Results MFN- as compared to SFN patients, were more likely to be male, older, had a lower intraepidermal nerve fiber density at the ankle and more frequent abnormal immunofixation. Beyond these differences, symptom prevalence and intensities were similar in the two cohorts. SFN patients comprised three distinct phenotypic clusters, which differed significantly in symptom severity, co-occurrence, localization, and skin biopsy findings. Only one subgroup, constituting about 20% of the patient population, was characterized by intense neuropathic pain, which was always associated with several other SFN symptoms of similarly high intensities. A pauci-symptomatic cluster comprised patients who experienced few SFN symptoms, generally of low to moderate intensity. The largest cluster was characterized by intense fatigue, myalgias and subjective weakness, but lower intensities of burning pain and paresthesia. Discussion This data-driven study introduces a new approach to subgrouping patients with SFN. Considering both neuropathic pain and pernicious symptoms beyond pain, we identified three clusters, which may be related to distinct pathophysiological mechanisms. Although additional validation will be required, our findings represent a step towards stratified treatment approaches and, ultimately, personalized treatment.
Collapse
|
3
|
Raasing LRM, Vogels OJM, Datema M, Ambarus CA, Tannemaat MR, Grutters JC, Veltkamp M. New phenotyping questionnaire for diagnosing sarcoidosis-associated small fiber neuropathy. Brain Commun 2024; 6:fcae289. [PMID: 39291161 PMCID: PMC11406462 DOI: 10.1093/braincomms/fcae289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/01/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Small fiber neuropathy is a common complication in patients with sarcoidosis and its prevalence is estimated at 40-86%. The underlying mechanism influences the presentation of small fiber neuropathy. For example, patients with metabolic diseases are often associated with a classic length-dependent small fiber neuropathy pattern, while patients with inflammatory diseases are more often present with a non-length-dependent small fiber neuropathy. Detailed phenotyping may be useful to improve diagnostic efficiency, as a clue to underlying mechanisms and as a precondition for personalized medicine. This study examined four phenotypes distinguishing between length-dependent and non-length-dependent presentation with a new subdivision for continuous and intermittent presentation. Forty-eight sarcoid patients with symptoms and at least two clinical signs of small fiber neuropathy and normal nerve conduction studies were classified as having probable small fiber neuropathy. A new small fiber neuropathy phenotyping questionnaire has been developed that allows patients to mark the anatomical locations of pain at three different levels: the skin, muscles, and joints. The location of symptoms was used to define length dependence, and two colors were used to distinguish continuous (red) from intermittent (blue) symptoms. In addition, skin biopsy, corneal confocal microscopy, Sudoscan and water immersion skin wrinkling were used to investigate a correlation between the four phenotypes, sensory function, nerve fiber density, and autonomic nerve function. Overall, 35% of patients with probable small fiber neuropathy showed length-dependent symptoms and 44% showed non-length-dependent symptoms while 21% suffered from non-neuropathic musculoskeletal pain. The distinction between intermittent and continuous symptoms showed significantly less continuous than intermittent non-length-dependent symptoms (odds ratio = 0.3, P = 0.01). Moreover, continuous length-dependent symptoms were the only phenotype that correlated with thermal threshold testing (R = 0.3; P = 0.02) and the small fiber neuropathy screening list (R = 0.3; P = 0.03). In addition, thermal threshold testing (TTT) also correlated with the small fiber neuropathy (SFN) screening list (R = 0.3; P = 0.03). Other diagnostic methods showed no correlation with any of the four defined phenotypes. A novel finding is that TTT is only associated with continuous length-dependent pain, suggesting that TTT could result in more false negatives in patients with other pain phenotypes. Determining the pathophysiologic mechanisms could help develop new diagnostic methods. If patients suspected of SFN show symptoms without a length-dependent continuous presentation, the diagnosis should focus less on the diagnostic methods used.
Collapse
Affiliation(s)
- Lisette R M Raasing
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Oscar J M Vogels
- ILD Center of Excellence, Department of Neurology, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Mirjam Datema
- ILD Center of Excellence, Department of Clinical Neurophysiology, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Carmen A Ambarus
- ILD Center of Excellence, Department of Pathology, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Martijn R Tannemaat
- Department of Clinical Neurophysiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jan C Grutters
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Marcel Veltkamp
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
4
|
Moak JP, Ramwell CB, Gordish-Dressman H, Sule SD, Bettini E. Small fiber neuropathy in children, adolescents, and young adults with chronic orthostatic intolerance and postural orthostatic tachycardia syndrome: A retrospective study. Auton Neurosci 2024; 253:103163. [PMID: 38537312 DOI: 10.1016/j.autneu.2024.103163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 02/11/2024] [Accepted: 03/07/2024] [Indexed: 05/28/2024]
Abstract
PURPOSE To determine in children, adolescent and young adult (CAYA) patients presenting with Orthostatic Intolerance (OI) or Postural Orthostatic Tachycardia Syndrome (POTS) associated with the additional symptoms of neuropathic discomfort (pain, paresthesia and/or allodynia): 1) the incidence of small fiber neuropathy, and 2) assess if there was serologic evidence for an underlying inflammatory or autoimmune state. METHODS A cohort of 109 CAYA patients with the above symptoms underwent epidermal skin biopsy for nerve fiber density. Blood biomarkers for inflammation were tested (CRP, ESR, ANA, complement (C3), thyroid function testing with antibodies (thyroid peroxidase antibody and thyroglobulin antibody), and cytokine panel 13). Patients completed a Quality of Health questionnaire. Statistical analysis was performed using Wilcoxon rank sum tests. RESULTS In CAYA patients with OI or POTS and neuropathic symptoms, skin biopsy for small fiber neuropathy was abnormal in 53 %. The sample population was predominantly female and Caucasian with moderately decreased perceived quality of health. OI /POTS patients with small fiber neuropathy had a 3-fold probability of having a positive ANA or anti-thyroid antibody, suggesting an underlying autoimmune or inflammatory process. CONCLUSION Our data suggest a link between OI and POTS and small fiber neuropathy. Small fiber neuropathy was found by skin biopsy in over half of the patients tested. OI and Postural orthostatic tachycardia patients with small fiber neuropathy expressed multiple markers suggesting an underlying autoimmune or inflammatory process. Future research will be done to evaluate the symptomatic implication of SFN and whether immune or pharmacologic manipulation can alter patient symptoms.
Collapse
Affiliation(s)
- Jeffrey P Moak
- Division of Cardiology, Children's National Hospital, Washington, DC, United States of America.
| | - Carolyn B Ramwell
- Division of Cardiology, Children's National Hospital, Washington, DC, United States of America
| | - Heather Gordish-Dressman
- Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, United States of America
| | - Sangeeta D Sule
- Division of Rheumatology, Children's National Hospital, Washington, DC, United States of America
| | - Elizabeth Bettini
- Division of Cardiology, Children's National Hospital, Washington, DC, United States of America
| |
Collapse
|
5
|
Aspegren O, Pourhamidi K. Reliable Method for Estimating Nerve Fiber Density in Epidermis Using Routine Histopathologic Tissue Preparation: A Promising Diagnostic Tool for Small Fiber Neuropathy. Appl Immunohistochem Mol Morphol 2024; 32:215-221. [PMID: 38650330 DOI: 10.1097/pai.0000000000001193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 02/27/2024] [Indexed: 04/25/2024]
Abstract
Practical yet reliable diagnostic tools for small-fiber neuropathy are needed. We aimed to establish a histopathologic protocol for estimating intraepidermal nerve fiber density (eIENFD) on formalin-fixed, paraffin-embedded tissue (FFPE), evaluate its reliability through intraobserver and interobserver analyses, and provide normative reference values for clinical use. Sixty-eight healthy participants underwent nerve conduction studies and quantitative sensory testing. Skin biopsies from the distal and proximal leg were taken and processed using routine immunohistochemistry (anti-PGP9.5 antibodies) on thin 5 µm sections. eIENFD was assessed with a modified counting protocol. Interobserver and intraobserver reliabilities were excellent (ICC=0.9). eIENFD was higher in females than males (fibers/mm, 14.3±4.4 vs. 11.6±5.8, P <0.05), decreased with age ( r s =-0.47, P <0.001), and was higher proximally than distally (15.0±5.5 vs. 13.0±5.3, P =0.002). Quantile regression equations for the fifth percentile of distal and proximal eIENFD were presented: 13.125-0.161×age (y)-0.932×sex (male=1; female=0) and 17.204-0.192×age (y)-3.313×sex (male=1; female=0), respectively. This study introduces a reliable and reproducible method for estimating epidermal nerve fiber density through immunostaining on 5-µm thin FFPE tissue samples. Normative data on eIENFD is provided. Regression equations help identify abnormal decreases in small nerve fiber density.
Collapse
Affiliation(s)
- Oskar Aspegren
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital
| | - Kaveh Pourhamidi
- Department of Clinical Neurophysiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Shadiack EC, Osinubi O, Gruber-Fox A, Bhate C, Patrick-DeLuca L, Cohen P, Helmer DA. Small Fiber Neuropathy in Veterans With Gulf War Illness. Fed Pract 2024; 41:136-141. [PMID: 39398967 PMCID: PMC11468626 DOI: 10.12788/fp.0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Background Gulf War veterans deployed to operations Desert Shield and Desert Storm returned with chronic multisystemic symptoms. This Gulf War Illness (GWI) has defied attempts to identify an underlying etiology. Pain and other symptoms attributable to autonomic nervous system (ANS) dysfunction are common, which may suggest a pathophysiologic underpinning. Small fiber neuropathy (SFN) presents with similar symptoms. Toxic exposures have been implicated in both SFN and GWI. Methods A retrospective chart review of clinical data at the New Jersey War Related Illness and Injury Study Center addressed the following questions: (1) how common was biopsy-confirmed SFN in veterans with GWI; (2) do veterans with GWI and SFN report more symptoms attributable to ANS dysfunction as compared to veterans with GWI and no SFN; and (3) can SFN in veterans with GWI and SFN be explained by conditions commonly associated with SFN? Chart review abstracted GWI status, skin biopsy results, and ANS symptom burden. For veterans with GWI and SFN, additional chart abstraction was explored for commonly reported contributing conditions. Results From March 1, 2015, to January 31, 2019, 51 Gulf War veterans evaluated at the War Related Illness and Injury Study center had a skin biopsy. Of these, 42 (83%) were diagnosed with GWI and 24 of 42 (57%) also had SFN. No differences were observed in ANS symptoms when compared with veterans with GWI and no SFN. A potential etiology for SFN was identified in 16 of 24 (67%) veterans with GWI and SFN, increasing to 19 (79%) when hyperlipidemia was included. Our analysis did not identify an explanation in 5 of 24 (21%) veterans with GWI and SFN. Conclusions SFN was common in this clinical sample of veterans diagnosed with GWI. A well-established potential etiology was identified in most cases of SFN. About 20% of veterans with GWI in our clinical sample had idiopathic SFN, and it is plausible that deployment-related exposures could have contributed to this condition. Symptoms of ANS are prevalent in GWI, though SFN cannot solely account for this. Our study does not generally support SFN as etiologic for GWI, though this may still be relevant for some. Additional research is required to explore relationships between Gulf War exposures and SFN.
Collapse
Affiliation(s)
- Edward C Shadiack
- Veterans Affairs New Jersey Health Care Systems, East Orange
- War Related Illness and Injury Study Center, East Orange, New Jersey
| | - Omowunmi Osinubi
- Veterans Affairs New Jersey Health Care Systems, East Orange
- War Related Illness and Injury Study Center, East Orange, New Jersey
| | | | - Chinmoy Bhate
- Veterans Affairs New Jersey Health Care Systems, East Orange
| | - Lydia Patrick-DeLuca
- Veterans Affairs New Jersey Health Care Systems, East Orange
- War Related Illness and Injury Study Center, East Orange, New Jersey
| | - Philip Cohen
- Veterans Affairs New Jersey Health Care Systems, East Orange
| | - Drew A Helmer
- War Related Illness and Injury Study Center, East Orange, New Jersey
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
7
|
Bon M, Dardis A, Scarpa M, Sechi A. Relief of nocturnal neuropathic pain with the use of cannabis in a patient with Fabry disease. Mol Genet Metab Rep 2023; 37:101010. [PMID: 38053923 PMCID: PMC10694749 DOI: 10.1016/j.ymgmr.2023.101010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 12/07/2023] Open
Abstract
Neuropathic pain is one of the most invalidating symptoms in patients with Fabry disease (FD), affecting their quality of life, it is linked to small fiber neuropathy and it may not respond to available disease specific treatments. We report the case of a 32 years old man with classic FD and severe neuropathic pain who, after the failure of several standard pharmaceutical approaches, was treated with medical cannabis with relief of nocturnal pain and sleep improvement.
Collapse
Affiliation(s)
- Martina Bon
- Regional Coordinating Center for Rare Diseases, University Hospital of Udine, Udine, Italy
| | - Andrea Dardis
- Regional Coordinating Center for Rare Diseases, University Hospital of Udine, Udine, Italy
| | - Maurizio Scarpa
- Regional Coordinating Center for Rare Diseases, University Hospital of Udine, Udine, Italy
| | - Annalisa Sechi
- Regional Coordinating Center for Rare Diseases, University Hospital of Udine, Udine, Italy
| |
Collapse
|
8
|
Krasselt M, Baerwald C. [Fibromyalgia as a Rheumatic Pain Syndrome]. Dtsch Med Wochenschr 2023; 148:1467-1472. [PMID: 37918433 DOI: 10.1055/a-1965-6870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Epidemiology and pathophysiology: Fibromyalgia is significantly more common in people with rheumatic diseases than in the general population. Nevertheless, it can occur independent of other diseases. Physical and psychosocial factors are responsible for the genesis for fibromyalgia making it a multifactorial disease. Most importantly, central pain processing seems to be abnormal. The relevance of a small fibre neuropathy is yet to be determined. For the very first time, a study was able to demonstrate that fibromyalgia might be passively transferred from one organism to another in an experimental setting.Diagnosis: Fibromyalgia is a clinical diagnose. Besides generalized pain, sleep disturbances and fatigue are common features. Furthermore, there can be an association with depressive disorders. Determining the Widespread Pain Index (WPI) and the Symptom Severity Score (SSS) can help in diagnosing Fibromyalgia and to determine severity of the disease.Therapy: Cornerstones of the treatment are patient education, physical exercise, physical therapy, and cognitive behavioural therapy. In therapy-resistant cases, a multimodal approach might be considered. Analgesic drugs, particularly opioids, should basically be avoided or only be used for a short period of time. Naltrexone, an opioid antagonist, is a promising treatment candidate. Another possible approach might be the use of TENS. While there are positive observational studies on the therapeutic use of cannabinoids, evidence from controlled trials is still missing.
Collapse
Affiliation(s)
- Marco Krasselt
- Endokrinologie, Nephrologie und Rheumatologie, Department für Innere Medizin, Neurologie und Dermatologie, Medizinische Klinik III, Universitätsklinikum Leipzig AöR, Leipzig
| | - Christoph Baerwald
- Universitäres Zentrum für seltene Erkrankungen, Universitätsklinikum Leizpig AöR, Leipzig
| |
Collapse
|
9
|
Loose S, Lischka A, Kuehs S, Nau C, Heinemann SH, Kurth I, Leipold E. Peripheral temperature dysregulation associated with functionally altered Na V1.8 channels. Pflugers Arch 2023; 475:1343-1355. [PMID: 37695396 PMCID: PMC10567936 DOI: 10.1007/s00424-023-02856-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/23/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
The voltage-gated sodium channel NaV1.8 is prominently expressed in the soma and axons of small-caliber sensory neurons, and pathogenic variants of the corresponding gene SCN10A are associated with peripheral pain and autonomic dysfunction. While most disease-associated SCN10A variants confer gain-of-function properties to NaV1.8, resulting in hyperexcitability of sensory neurons, a few affect afferent excitability through a loss-of-function mechanism. Using whole-exome sequencing, we here identify a rare heterozygous SCN10A missense variant resulting in alteration p.V1287I in NaV1.8 in a patient with a 15-year history of progressively worsening temperature dysregulation in the distal extremities, particularly in the feet. Further symptoms include increasingly intensifying tingling and numbness in the fingers and increased sweating. To assess the impact of p.V1287I on channel function, we performed voltage-clamp recordings demonstrating that the alteration confers loss- and gain-of-function characteristics to NaV1.8 characterized by a right-shifted voltage dependence of channel activation and inactivation. Current-clamp recordings from transfected mouse dorsal root ganglion neurons further revealed that NaV1.8-V1287I channels broaden the action potentials of sensory neurons and increase their firing rates in response to depolarizing current stimulations, indicating a gain-of-function mechanism of the variant at the cellular level in a heterozygous setting. The data support the hypothesis that the properties of NaV1.8 p.V1287I are causative for the patient's symptoms and that nonpainful peripheral paresthesias should be considered part of the clinical spectrum of NaV1.8-associated disorders.
Collapse
Affiliation(s)
- Simon Loose
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany
| | - Annette Lischka
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Samuel Kuehs
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany
| | - Carla Nau
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany
| | - Stefan H Heinemann
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Enrico Leipold
- Department of Anesthesiology and Intensive Care & CBBM - Center of Brain, Behavior and Metabolism, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany.
| |
Collapse
|
10
|
Krämer HH, Bücker P, Jeibmann A, Richter H, Rosenbohm A, Jeske J, Baka P, Geber C, Wassenberg M, Fangerau T, Karst U, Schänzer A, van Thriel C. Gadolinium contrast agents: dermal deposits and potential effects on epidermal small nerve fibers. J Neurol 2023:10.1007/s00415-023-11740-z. [PMID: 37138180 DOI: 10.1007/s00415-023-11740-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/05/2023]
Abstract
Small fiber neuropathy (SFN) affects unmyelinated and thinly myelinated nerve fibers causing neuropathic pain with distal distribution and autonomic symptoms. In idiopathic SFN (iSFN), 30% of the cases, the underlying aetiology remains unknown. Gadolinium (Gd)-based contrast agents (GBCA) are widely used in magnetic resonance imaging (MRI). However, side-effects including musculoskeletal disorders and burning skin sensations were reported. We investigated if dermal Gd deposits are more prevalent in iSFN patients exposed to GBCAs, and if dermal nerve fiber density and clinical parameters are likewise affected. 28 patients (19 females) with confirmed or no GBCA exposure were recruited in three German neuromuscular centers. ISFN was confirmed by clinical, neurophysiological, laboratory and genetic investigations. Six volunteers (two females) served as controls. Distal leg skin biopsies were obtained according to European recommendations. In these samples Gd was quantified by elemental bioimaging and intraepidermal nerve fibers (IENF) density via immunofluorescence analysis. Pain phenotyping was performed in all patients, quantitative sensory testing (QST) only in a subset (15 patients; 54%). All patients reported neuropathic pain, described as burning (n = 17), jabbing (n = 16) and hot (n = 11) and five QST scores were significantly altered. Compared to an equal distribution significantly more patients reported GBCA exposures (82%), while 18% confirmed no exposures. Compared to unexposed patients/controls significantly increased Gd deposits and lower z-scores of the IENF density were confirmed in exposed patients. QST scores and pain characteristics were not affected. This study suggests that GBCA exposure might alter IENF density in iSFN patients. Our results pave the road for further studies investigating the possible role of GBCA in small fiber damage, but more investigations and larger samples are needed to draw firm conclusions.
Collapse
Affiliation(s)
- Heidrun H Krämer
- Department of Neurology, Justus Liebig University of Giessen, 35392, Giessen, Germany
| | - Patrick Bücker
- Institute of Inorganic and Analytical Chemistry, University of Münster, 48149, Münster, Germany
| | - Astrid Jeibmann
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany
| | - Henning Richter
- Clinic for Diagnostic Imaging, Diagnostic Imaging Research Unit (DIRU),Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | | | - Johanna Jeske
- Department of Neurology, Justus Liebig University of Giessen, 35392, Giessen, Germany
| | - Panoraia Baka
- Department of Neurology, University Medical Center, 55101, Mainz, Germany
| | - Christian Geber
- Department of Neurology, University Medical Center, 55101, Mainz, Germany
- DRK Pain Center Mainz, 55131, Mainz, Germany
| | - Matthias Wassenberg
- Department of Neurology, Justus Liebig University of Giessen, 35392, Giessen, Germany
| | - Tanja Fangerau
- Department of Neurology, University of Ulm, 89081, Ulm, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, 48149, Münster, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus Liebig University Giessen, 35392, Giessen, Germany.
| | - Christoph van Thriel
- IfADo-Leibniz Research Centre for Working Environment and Human Factors, 44139, Dortmund, Germany.
| |
Collapse
|
11
|
Starshinova A, Zinchenko Y, Malkova A, Kudlay D, Kudryavtsev I, Yablonskiy P. Sarcoidosis and Autoimmune Inflammatory Syndrome Induced by Adjuvants. Life (Basel) 2023; 13:1047. [PMID: 37109576 PMCID: PMC10145559 DOI: 10.3390/life13041047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Currently, sarcoidosis remains one of the diseases with unknown etiology, which significantly complicates its diagnosis and treatment. Various causes of sarcoidosis have been studied for many years. Both organic and inorganic trigger factors, provoking the development of granulomatous inflammation are considered. However, the most promising and evidence-based hypothesis is the development of sarcoidosis as an autoimmune disease, provoked by various adjuvants in genetic predisposed individuals. This concept fits into the structure of the autoimmune/inflammatory syndrome, induced by adjuvants (ASIA) that was proposed in 2011 by Professor Shoenfeld Y. In this paper, the authors reveal the presence of major and minor ASIA criteria for sarcoidosis, propose a new concept of the course of sarcoidosis within the framework of ASIA, and point out the difficulties in creating a model of the disease and the selection of therapy. It is obvious that the data obtained not only bring us closer to understanding the nature of sarcoidosis, but also potentiate new studies confirming this hypothesis by obtaining a model of the disease.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia;
| | - Yulia Zinchenko
- Saint-Petersburg Research Institute of Phthisiopulmonology, 194064 Saint-Petersburg, Russia (P.Y.)
| | - Anna Malkova
- Laboratory of the Mosaic of Autoimmunity, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia;
| | - Dmitriy Kudlay
- Medical Department, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
- Institute of Immunology, 115478 Moscow, Russia
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia;
- Department of Immunology, Institution of Experimental Medicine, 197022 Saint-Petersburg, Russia
| | - Piotr Yablonskiy
- Saint-Petersburg Research Institute of Phthisiopulmonology, 194064 Saint-Petersburg, Russia (P.Y.)
- Laboratory of the Mosaic of Autoimmunity, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia;
| |
Collapse
|
12
|
Chan ACY, Kumar S, Tan G, Wong HY, Ong JJY, Chandra B, Huang H, Sharma VK, Lai PS. Expanding the genetic causes of small-fiber neuropathy: SCN genes and beyond. Muscle Nerve 2023; 67:259-271. [PMID: 36448457 DOI: 10.1002/mus.27752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/31/2022] [Accepted: 11/06/2022] [Indexed: 12/05/2022]
Abstract
Small-fiber neuropathy (SFN) is a disorder that exclusively affects the small nerve fibers, sparing the large nerve fibers. Thinly myelinated Aδ-fibers and unmyelinated C-fibers are damaged, leading to development of neuropathic pain, thermal dysfunction, sensory symptoms, and autonomic disturbances. Although many SFNs are secondary and due to immunological causes or metabolic disturbances, the etiology is unknown in up to half of the patients. Over the years, this proportion of "idiopathic SFN" has decreased, as familial and genetic causes have been discovered, thus shifting a proportion of once "idiopathic" cases to the genetic category. After the discovery of SCN9A-gene variants in 2012, SCN10A and SCN11A variants have been found to be pathogenic in SFN. With improved accessibility of SFN diagnostic tools and genetic tests, many non-SCN variants and genetically inherited systemic diseases involving the small nerve fibers have also been described, but only scattered throughout the literature. There are 80 SCN variants described as causing SFN, 8 genes causing hereditary sensory autonomic neuropathies (HSAN) described with pure SFN, and at least 7 genes involved in genetically inherited systemic diseases associated with SFN. This systematic review aims to consolidate and provide an updated overview on the genetic variants of SFN to date---SCN genes and beyond. Awareness of these genetic causes of SFN is imperative for providing treatment directions, prognostication, and management of expectations for patients and their health-care providers.
Collapse
Affiliation(s)
- Amanda C Y Chan
- Division of Neurology, Department of Medicine, National University Hospital, Singapore, Singapore
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shivaram Kumar
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Grace Tan
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hiu Yi Wong
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Jonathan J Y Ong
- Division of Neurology, Department of Medicine, National University Hospital, Singapore, Singapore
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bharatendu Chandra
- Division of Neurology, Department of Medicine, National University Hospital, Singapore, Singapore
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Medical Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Hua Huang
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vijay Kumar Sharma
- Division of Neurology, Department of Medicine, National University Hospital, Singapore, Singapore
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Poh San Lai
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
- Adjunct Faculty, Genome Institute of Singapore, Singapore, Singapore
| |
Collapse
|
13
|
Panagiotides NG, Zimprich F, Machold K, Schlager O, Müller M, Ertl S, Löffler-Stastka H, Koppensteiner R, Wadowski PP. A Case of Autoimmune Small Fiber Neuropathy as Possible Post COVID Sequelae. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4918. [PMID: 36981826 PMCID: PMC10049708 DOI: 10.3390/ijerph20064918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is reported to induce and augment autoimmune processes. Moreover, postinfectious effects of coronavirus disease 2019 (COVID-19) are still poorly understood and often resemble symptoms of the acute infection phase. A patient with swollen extremities was presented to the Department of Angiology at the Medical University of Vienna with complaints of muscle and joint pain, paresthesia, and arterial hypertension with intense headache. Prior to these complaints, she had been suffering from various symptoms since November 2020, following a SARS-CoV-2 infection in the same month. These included recurrent sore throat, heartburn, dizziness, and headache. Paresthesia and muscle and joint pain started in temporal relation to a human papillomavirus (HPV) vaccination. Since the patient was suffering from severe pain, intensive pain management was performed. Skin and nerve biopsies revealed autoimmune small fiber neuropathy. The patient's condition could be related to COVID-19, as her first symptoms began in temporal relation to the SARS-CoV-2 infection. Furthermore, in the disease course, antinuclear (ANA) and anti-Ro antibodies, as well as anti-cyclic citrullinated peptide (anti-CCP) antibodies, could be detected. Together with the symptoms of xerophthalmia and pharyngeal dryness, primary Sjögren's syndrome was diagnosed. In conclusion, though biopsy results could not distinguish a cause of the disease, SARS-CoV-2 infection can be discussed as a likely trigger for the patient's autoimmune reactions.
Collapse
Affiliation(s)
- Noel G. Panagiotides
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (O.S.); (M.M.); (S.E.); (R.K.)
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Fritz Zimprich
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Klaus Machold
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria;
| | - Oliver Schlager
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (O.S.); (M.M.); (S.E.); (R.K.)
| | - Markus Müller
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (O.S.); (M.M.); (S.E.); (R.K.)
| | - Sebastian Ertl
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (O.S.); (M.M.); (S.E.); (R.K.)
- Division of Internal Medicine II, Klinikum Wels-Grieskirchen, 4600 Wels-Grieskirchen, Austria
| | | | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (O.S.); (M.M.); (S.E.); (R.K.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (O.S.); (M.M.); (S.E.); (R.K.)
| |
Collapse
|
14
|
Rasmussen TK, Karlsson P, Finnerup NB, Jensen TS, Nyengaard JR, Terkelsen AJ. Functional and structural markers of peripheral microvascular autonomic neuropathy. Muscle Nerve 2023; 67:146-153. [PMID: 36504143 PMCID: PMC10108116 DOI: 10.1002/mus.27770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION/AIMS Autonomic dysfunction is a common complication of small-fiber neuropathy (SFN). In this study we aimed to assess the applicability of autonomic microvascular indices as a potential marker for SFN assessment. METHODS Fifteen patients with confirmed SFN (idiopathic neuropathy [n = 10], chemotherapy-induced peripheral neuropathy [n = 2], impaired glucose tolerance [n = 1], hereditary transthyretin amyloidosis (hATTR) [n = 1], pulmonary sarcoidosis [n = 1]) and 15 matched control subjects underwent assessment of vascular skin responses assessed through laser Doppler flowmetry and evaluation of microvascular vessel and nerve density in skin biopsies. All participants underwent peripheral autonomic evaluation by quantitative sudomotor axon reflex testing (QSART). RESULTS We found no significant differences in vascular skin responses, or in any microvascular skin biopsy markers, when comparing SFN with control subjects. We found no correlation between vascular skin responses and skin biopsy indices. We saw no significant difference in any microvascular indices when comparing subjects with and without impaired sudomotor function. DISCUSSION Our findings suggest markers of peripheral microvascular innervation and function are not associated with the diagnosis of SFN. Furthermore, we saw no association between microvascular markers and sudomotor function, suggesting that these are independent and unrelated components of the autonomic nervous system.
Collapse
Affiliation(s)
- Thorsten K Rasmussen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Páll Karlsson
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Troels S Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Astrid J Terkelsen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Autoimmune autonomic nervous system imbalance and conditions: Chronic fatigue syndrome, fibromyalgia, silicone breast implants, COVID and post-COVID syndrome, sick building syndrome, post-orthostatic tachycardia syndrome, autoimmune diseases and autoimmune/inflammatory syndrome induced by adjuvants. Clin Exp Rheumatol 2023; 22:103230. [PMID: 36347462 DOI: 10.1016/j.autrev.2022.103230] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Chronic fatigue syndrome (CFS), fibromyalgia, silicone breast implants syndrome (SBIs), COVID and post-COVID syndrome (PCS), sick building syndrome (SBS), post-orthostatic tachycardia syndrome (POTS), autoimmune diseases and autoimmune/inflammatory syndrome induced by adjuvants (ASIA) are frequently accompanied by clinical symptoms characteristic for dysautonomia: severe fatigue, dizziness, fogginess, memory loss, dry mouth and eyes, hearing dysfunction, tachycardia etc. The recent discovery of an imbalance of autoantibodies against G protein-coupled receptors (GPCR) in some autoimmune diseases, post-COVID syndrome, SBIs allowed researchers to assume the novel mechanism in these conditions - autoimmune autonomic nervous system imbalance. In this review, all data published on an imbalance of autoantibodies against GPCR, clinical symptoms and pathogenic mechanisms in CFS, Fibromyalgia, SBIs, COVID and PCS, SBS, POTS, and some autoimmune diseases were analyzed. Possible criteria to diagnose the autoimmune autonomic nervous system imbalance were created.
Collapse
|
16
|
Dormer A, Narayanan M, Schentag J, Achinko D, Norman E, Kerrigan J, Jay G, Heydorn W. A Review of the Therapeutic Targeting of SCN9A and Nav1.7 for Pain Relief in Current Human Clinical Trials. J Pain Res 2023; 16:1487-1498. [PMID: 37168847 PMCID: PMC10166096 DOI: 10.2147/jpr.s388896] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/14/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction There is a great need to find alternative treatments for chronic pain which have become a healthcare problem. We discuss current therapeutic targeting Nav1.7. Areas Covered Nav1.7 is a sodium ion channel protein that is associated with several human pain genetic syndromes. It has been found that mutations associated with Nav1.7 lead to the loss of the ability to perceive pain in individuals that are otherwise normal. Several therapeutic interventions are presently undergoing preclinical and research using the methodology of damping Nav1.7 expressions as a methodology to decrease the sensation of pain leading to analgesia. Expert Opinion It is our strong belief that there is a viable future in the targeting of protein of Nav1.7 for the relief of chronic pain in humans. The review will look at the genomics associated with SCN1A and proteomic of Nav1.7 as a foundation to explain the mechanism of the therapeutic interventions targeting Nav1.7, the human disease that are associated with Nav1.7, and the current development of treatment for chronic pain whether in preclinical or clinical trials targeting Nav1.7 expressions. The development of therapeutic antagonists targeting Nav1.7 could be a viable alternative to the current treatments which have led to the opioid crisis. Therefore, Nav1.7 targeted treatment has a major clinical significance that will have positive consequences as it relates to chronic pain interventions.
Collapse
Affiliation(s)
- Anton Dormer
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
- Correspondence: Anton Dormer, Research and Development, PepVax, Inc, 8720 Georgia Ave #1000, Silver Spring, MD, 20910, USA, Email
| | | | - Jerome Schentag
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Daniel Achinko
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Elton Norman
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - James Kerrigan
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | - Gary Jay
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | | |
Collapse
|
17
|
Malkova AM, Shoenfeld Y. WITHDRAWN: Autoimmune autonomic nervous system imbalance and conditions: Chronic fatigue syndrome, fibromyalgia, silicone breast implants, COVID and post-COVID syndrome, sick building syndrome, post-orthostatic tachycardia syndrome, autoimmune diseases and autoimmune/inflammatory syndrome induced by adjuvants. Autoimmun Rev 2022:103231. [PMID: 36356798 DOI: 10.1016/j.autrev.2022.103231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, https://doi.org/10.1016/j.autrev.2022.103230. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- A M Malkova
- Zabludowicz Center of autoimmunity, Sheba Medical Center, Tel Hashomer, Israel.
| | - Y Shoenfeld
- Zabludowicz Center of autoimmunity, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
18
|
Barroso FA, Coelho T, Dispenzieri A, Conceição I, Waddington-Cruz M, Wixner J, Maurer MS, Rapezzi C, Planté-Bordeneuve V, Kristen AV, González-Duarte A, Chapman D, Stewart M, Amass L. Characteristics of patients with autonomic dysfunction in the Transthyretin Amyloidosis Outcomes Survey (THAOS). Amyloid 2022; 29:175-183. [PMID: 35451899 DOI: 10.1080/13506129.2022.2043270] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Autonomic dysfunction is common in transthyretin amyloidosis (ATTR amyloidosis), but its frequency, characteristics, and quality-of-life (QoL) impact are not well understood. METHODS The Transthyretin Amyloidosis Outcomes Survey (THAOS) is an ongoing, global, longitudinal survey of patients with ATTR amyloidosis, including patients with inherited (ATTRv) and wild-type (ATTRwt) disease and asymptomatic patients with TTR mutations (ClinicalTrials.gov: NCT00628745). In a descriptive analysis, characteristics and Norfolk QoL-DN total (TQoL) scores at enrolment were compared in patients with vs without autonomic dysfunction (analysis cut-off: 1 August 2020). RESULTS Autonomic dysfunction occurred in 1181/2922 (40.4%) symptomatic patients, and more commonly in ATTRv (1107/1181 [93.7%]) than ATTRwt (74/1181 [6.3%]) amyloidosis. Time (mean [SD]) from ATTR amyloidosis symptom onset to first autonomic dysfunction symptom was shorter in ATTRv (3.4 [5.7] years) than ATTRwt disease (9.7 [10.4]). In ATTRv disease, patients with vs without autonomic dysfunction had worse QoL (TQoL, 47.3 [33.2] vs 16.1 [18.1]); in ATTRwt disease, those with vs without autonomic dysfunction had similar QoL (23.0 [18.2] vs 19.9 [20.5]). CONCLUSIONS Autonomic dysfunction was more common and presented earlier in symptomatic ATTRv than ATTRwt amyloidosis and adversely affected QoL in ATTRv disease. These THAOS findings may aid clinicians in diagnosing and treating patients with ATTR amyloidosis. Trial registration: ClinicalTrials.gov: NCT00628745.
Collapse
Affiliation(s)
- Fabio A Barroso
- Institute for Neurological Research, FLENI, Buenos Aires, Argentina
| | - Teresa Coelho
- Unidade Corino Andrade, Hospital Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | | | - Isabel Conceição
- Hospital de Santa Maria-CHULN, FML Universidade de Lisboa, Lisbon, Portugal
| | - Marcia Waddington-Cruz
- CEPARM, University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonas Wixner
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Mathew S Maurer
- Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Claudio Rapezzi
- Cardiological Center, University of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, Cotignola (RA), Italy
| | - Violaine Planté-Bordeneuve
- Department of Neurology, East-Paris University, Hospital Henri Mondor, Assistance Publique Hopitaux de Paris, INSERM U955 Team 10 "Biology of the Neuro-Muscular System", Crétei, France
| | - Arnt V Kristen
- Department of Cardiology, Angiology, and Respiratory Medicine, Medical University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Thimm A, Carpinteiro A, Oubari S, Papathanasiou M, Kessler L, Rischpler C, Malik RA, Reinhardt HC, Rassaf T, Herrmann K, Kleinschnitz C, Stettner M, Hagenacker T. Corneal confocal microscopy to detect early immune-mediated small nerve fibre loss in AL amyloidosis. Ann Clin Transl Neurol 2022; 9:853-863. [PMID: 35488792 PMCID: PMC9186132 DOI: 10.1002/acn3.51565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Light chain (AL) amyloidosis is a life‐threatening disorder characterised by extracellular deposition of amyloid leading to dysfunction of multiple organs. Peripheral nerve involvement, particularly small fibre neuropathy, may be associated with poorer survival. Corneal confocal microscopy (CCM) is a rapid and non‐invasive imaging technique to quantify corneal small nerve fibres and immune cells in vivo. We aimed to evaluate CCM as a tool for early diagnosis of peripheral nerve involvement in AL amyloidosis. Methods CCM and nerve conduction studies (NCS) were undertaken in 21 newly diagnosed, treatment‐naïve AL amyloidosis patients and 21 age‐ and sex‐matched healthy controls. Corneal nerve fibre density (CNFD), corneal nerve branch density and fibre length, and cell infiltrates were quantified in the sub‐basal layer of the cornea. Results There was a significant reduction in CNFD and nerve fibre length, even without large fibre affection and an increase in cell density, particularly around corneal nerve fibres in patients with AL amyloidosis compared to controls. Additionally, cell infiltration correlated with reduced nerve fibre density in patients with AL amyloidosis, but reduced CNFD did not correlate with laboratory parameters of organ dysfunction. Interpretation Our study is the first to show that CCM allows rapid non‐invasive identification of early small nerve fibre damage associated with immune cell infiltration in patients with AL amyloidosis. CCM detects peripheral nerve involvement more sensitively than NCS.
Collapse
Affiliation(s)
- Andreas Thimm
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Alexander Carpinteiro
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany.,Institute of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Sara Oubari
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Maria Papathanasiou
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| | - Lukas Kessler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | | | - Rayaz Ahmed Malik
- Institute of Cardiovascular Science, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK.,Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Mark Stettner
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Tim Hagenacker
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| |
Collapse
|
20
|
Strand N, Wie C, Peck J, Maita M, Singh N, Dumbroff J, Tieppo Francio V, Murphy M, Chang K, Dickerson DM, Maloney J. Small Fiber Neuropathy. Curr Pain Headache Rep 2022; 26:429-438. [PMID: 35384587 DOI: 10.1007/s11916-022-01044-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This narrative review aims to summarize advances in the field of small fiber neuropathy made over the last decade, with emphasis on novel research highlighting the distinctive features of SFN. RECENT FINDINGS While the management of SFNs is ideally aimed at treating the underlying cause, most patients will require pain control via multiple, concurrent therapies. Herein, we highlight the most up-to-date information for diagnosis, medication management, interventional management, and novel therapies on the horizon. Despite the prevalence of small fiber neuropathies, there is no clear consensus on guidelines specific for the treatment of SFN. Despite the lack of specific guidelines for SFN treatment, the most recent general neuropathic pain guidelines are based on Cochrane studies and randomized controlled trials (RCTs) which have individually examined therapies used for the more commonly studied SFNs, such as painful diabetic neuropathy and HIV neuropathy. The recommendations from current guidelines are based on variables such as number needed to treat (NNT), safety, ease of use, and effect on quality of life.
Collapse
Affiliation(s)
- N Strand
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA.
| | - C Wie
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - J Peck
- Performing Arts Medicine Department, Shenandoah University, Winchester, USA
| | - M Maita
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - N Singh
- OrthoAlabama Spine and Sports, Birmingham, AL, USA
| | - J Dumbroff
- Mount Sinai Morningside and West Department of Anesthesiology, New York, NY, USA
| | - V Tieppo Francio
- Department of Rehabilitation on Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - M Murphy
- Department of Rehabilitation on Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - K Chang
- Department of Anesthesiology and Critical Care, Emory University, Atlanta, GA, USA
| | - D M Dickerson
- NorthShore University HealthSystem, Evanston, IL, USA
- University of Chicago Medicine, Chicago,, IL, USA
| | - J Maloney
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| |
Collapse
|
21
|
The Role of Neuropathy Screening Tools in Patients Affected by Fibromyalgia. J Clin Med 2022; 11:jcm11061533. [PMID: 35329860 PMCID: PMC8953231 DOI: 10.3390/jcm11061533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/01/2023] Open
Abstract
Fibromyalgia syndrome (sFM) is one of the most common causes of chronic pain. This study aimed to assess the presence of small and large fiber impairment in fibromyalgic patients by applying validated scores used in the screening for diabetic neuropathy. The endpoints for the study were the assessment of neuropathy prevalence in sFM patients using the NerveCheck Master (NCM), the Michigan Neuropathy Screening Instrument (MNSI), the Diabetic Neuropathy Symptom (DNS) and the Douleur Neuropathique 4 Questions (DN4). The sample was composed of 46 subjects: subjects with sFM (n = 23) and healthy controls (HC) (n = 23). The positivity rates in each group for DN4 were significantly different (p < 0.001), with a prevalence in symptomatic subjects of 56.3% (n = 9) among sFM individuals. A similar difference was also observed with the DNS total score (p < 0.001). NCM and MNSI did not disclose significant differences between the two groups. This finding seems to confirm the data regarding the prevalence of a neuropathic pain in sFM patients.
Collapse
|
22
|
Reduced Gut Microbiome Diversity in People With HIV Who Have Distal Neuropathic Pain. THE JOURNAL OF PAIN 2022; 23:318-325. [PMID: 34530155 PMCID: PMC9854399 DOI: 10.1016/j.jpain.2021.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/04/2021] [Accepted: 08/17/2021] [Indexed: 02/03/2023]
Abstract
Gut dysbiosis, defined as pathogenic alterations in the distribution and abundance of different microbial species, is associated with neuropathic pain in a variety of clinical conditions, but this has not been explored in the context of neuropathy in people with HIV (PWH). We assessed gut microbial diversity and dysbiosis in PWH and people without HIV (PWoH), some of whom reported distal neuropathic pain (DNP). DNP was graded on a standardized, validated severity scale. The gut microbiome was characterized using 16S rRNA sequencing and diversity was assessed using phylogenetic tree construction. Songbird analysis (https://github.com/mortonjt/songbird) was used to produce a multinomial regression model predicting counts of specific microbial taxa through metadata covariate columns. Participants were 226 PWH and 101 PWoH, mean (SD) age 52.0 (13.5), 21.1% female, 54.7% men who have sex with men, 44.7% non-white. Among PWH, median (interquartile range, IQR) nadir and current CD4 were 174 (21, 302) and 618 (448, 822), respectively; 90% were virally suppressed on antiretroviral therapy. PWH and PWoH did not differ with respect to microbiome diversity as indexed by Faith's phylogenetic diversity (PD). More severe DNP was associated with lower alpha diversity as indexed by Faith's phylogenetic diversity in PWH (Spearman's ρ = .224, P = 0.0007), but not in PWoH (Spearman's ρ = .032, P = .748). These relationships were not confounded by demographics or disease factors. In addition, the log-ratio of features identified at the genus level as Blautia to Lachnospira was statistically significantly higher in PWH with DNP than in PWH without DNP (t-test, P = 1.01e-3). Furthermore, the log-ratio of Clostridium features to Lachnospira features also was higher in PWH with DNP than in those without (t-test, P = 6.24e-5). Our results, in combination with previous findings in other neuropathic pain conditions, suggest that gut dysbiosis, particularly reductions in diversity and relative increases in the ratios of Blautia and Clostridium to Lachnospira, may contribute to prevalent DNP in PWH. Two candidate pathways for these associations, involving microbial pro-inflammatory components and microbially-produced anti-inflammatory short chain fatty acids, are discussed. Future studies might test interventions to re-establish a healthy gut microbiota and determine if this prevents or improves DNP. PERSPECTIVE: The association of neuropathic pain in people with HIV with reduced gut microbial diversity and dysbiosis raises the possibility that re-establishing a healthy gut microbiota might ameliorate neuropathic pain in HIV by reducing proinflammatory and increasing anti-inflammatory microbial products.
Collapse
|
23
|
Small-Fiber-Neuropathien. DGNEUROLOGIE 2022; 5. [PMCID: PMC9559077 DOI: 10.1007/s42451-022-00488-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Die Small-Fiber-Neuropathie (SFN) ist eine Erkrankung des peripheren Nervensystems aufgrund einer isolierten oder vorwiegenden Schädigung dünn myelinisierter Aδ-Fasern und/oder unmyelinisierter C‑Fasern. Für die sichere Diagnose einer SFN ist neben der klinischen Symptomatik mit Schmerzen und Sensibilitätsstörungen, typischerweise mit distal betonter Ausbreitung, der apparative Nachweis einer Rarefizierung oder einer Funktionsstörung der dünn myelinisierten Aδ-Fasern und/oder der unmyelinisierten C‑Fasern gefordert. Im vorliegenden Beitrag wird eine Übersicht über die diagnostischen Verfahren zum Nachweis einer SFN sowie über mögliche Ursachen und Behandlungsoptionen gegeben.
Collapse
|
24
|
Burand AJ, Stucky CL. Fabry disease pain: patient and preclinical parallels. Pain 2021; 162:1305-1321. [PMID: 33259456 PMCID: PMC8054551 DOI: 10.1097/j.pain.0000000000002152] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/31/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
ABSTRACT Severe neuropathic pain is a hallmark of Fabry disease, a genetic disorder caused by a deficiency in lysosomal α-galactosidase A. Pain experienced by these patients significantly impacts their quality of life and ability to perform everyday tasks. Patients with Fabry disease suffer from peripheral neuropathy, sensory abnormalities, acute pain crises, and lifelong ongoing pain. Although treatment of pain through medication and enzyme replacement therapy exists, pain persists in many of these patients. Some has been learned in the past decades regarding clinical manifestations of pain in Fabry disease and the pathological effects of α-galactosidase A insufficiency in neurons. Still, it is unclear how pain and sensory abnormalities arise in patients with Fabry disease and how these can be targeted with therapeutics. Our knowledge is limited in part due to the lack of adequate preclinical models to study the disease. This review will detail the types of pain, sensory abnormalities, influence of demographics on pain, and current strategies to treat pain experienced by patients with Fabry disease. In addition, we discuss the current knowledge of Fabry pain pathogenesis and which aspects of the disease preclinical models accurately recapitulate. Understanding the commonalities and divergences between humans and preclinical models can be used to further interrogate mechanisms causing the pain and sensory abnormalities as well as advance development of the next generation of therapeutics to treat pain in patients with Fabry disease.
Collapse
Affiliation(s)
- Anthony J. Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
25
|
Tulbă D, Popescu BO, Manole E, Băicuș C. Immune Axonal Neuropathies Associated With Systemic Autoimmune Rheumatic Diseases. Front Pharmacol 2021; 12:610585. [PMID: 33935704 PMCID: PMC8079948 DOI: 10.3389/fphar.2021.610585] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/10/2021] [Indexed: 01/19/2023] Open
Abstract
Immune axonal neuropathies are a particular group of immune-mediated neuropathies that occasionally accompany systemic autoimmune rheumatic diseases such as connective tissue dissorders and primary systemic vasculitides. Apart from vasculitis of vasa nervorum, various other mechanisms are involved in their pathogenesis, with possible therapeutic implications. Immune axonal neuropathies have highly heterogeneous clinical presentation and course, ranging from mild chronic distal sensorimotor polyneuropathy to severe subacute mononeuritis multiplex with rapid progression and constitutional symptoms such as fever, malaise, weight loss and night sweats, underpinning a vasculitic process. Sensory neuronopathy (ganglionopathy), small fiber neuropathy (sensory and/or autonomic), axonal variants of Guillain-Barré syndrome and cranial neuropathies have also been reported. In contrast to demyelinating neuropathies, immune axonal neuropathies show absent or reduced nerve amplitudes with normal latencies and conduction velocities on nerve conduction studies. Diagnosis and initiation of treatment are often delayed, leading to accumulating disability. Considering the lack of validated diagnostic criteria and evidence-based treatment protocols for immune axonal neuropathies, this review offers a comprehensive perspective on etiopathogenesis, clinical and paraclinical findings as well as therapy guidance for assisting the clinician in approaching these patients. High quality clinical research is required in order to provide indications and follow up rules for treatment in immune axonal neuropathies related to systemic autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Delia Tulbă
- Department of Neurology, Colentina Clinical Hospital, Bucharest, Romania.,Colentina-Research and Development Center, Colentina Clinical Hospital, Bucharest, Romania.,"Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Bogdan Ovidiu Popescu
- Department of Neurology, Colentina Clinical Hospital, Bucharest, Romania.,"Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Laboratory of Cell Biology, Neurosciences and Experimental Myology, "Victor Babeș" National Institute of Pathology, Bucharest, Romania
| | - Emilia Manole
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, "Victor Babeș" National Institute of Pathology, Bucharest, Romania
| | - Cristian Băicuș
- Colentina-Research and Development Center, Colentina Clinical Hospital, Bucharest, Romania.,"Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Department of Internal Medicine, Colentina Clinical Hospital, Bucharest, Romania
| |
Collapse
|
26
|
Ghosh S, Tourtellotte WG. The Complex Clinical and Genetic Landscape of Hereditary Peripheral Neuropathy. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:487-509. [PMID: 33497257 DOI: 10.1146/annurev-pathol-030320-100822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hereditary peripheral neuropathy (HPN) is a complex group of neurological disorders caused by mutations in genes expressed by neurons and Schwann cells. The inheritance of a single mutation or multiple mutations in several genes leads to disease phenotype. Patients exhibit symptoms during development, at an early age or later in adulthood. Most of the mechanistic understanding about these neuropathies comes from animal models and histopathological analyses of postmortem human tissues. Diagnosis is often very complex due to the heterogeneity and overlap in symptoms and the frequent overlap between various genes and different mutations they possess. Some symptoms in HPN are common through different subtypes such as axonal degeneration, demyelination, and loss of motor and sensory neurons, leading to similar physiologic abnormalities. Recent advances in gene-targeted therapies, genetic engineering, and next-generation sequencing have augmented our understanding of the underlying pathogenetic mechanisms of HPN.
Collapse
Affiliation(s)
- Soumitra Ghosh
- Department of Pathology and Laboratory Medicine, Neurology, and Neurological Surgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA;
| | - Warren G Tourtellotte
- Department of Pathology and Laboratory Medicine, Neurology, and Neurological Surgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA;
| |
Collapse
|
27
|
Dunker Ø, Lie M, Nilsen K. Can within-subject comparisons of thermal thresholds be used for diagnostic purposes? Clin Neurophysiol Pract 2021; 6:63-71. [PMID: 33665518 PMCID: PMC7905396 DOI: 10.1016/j.cnp.2021.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/23/2020] [Accepted: 01/04/2021] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Quantitative thermal testing (QTT) is a psychophysical assessment method of small nerve fibers that relies on reference material to assess function. Normal limits for within-subject comparisons of thermal thresholds are scarce, and their association with age, height and sex is not fully elucidated. The aim of this study was to investigate the normal limits for distal-proximal- and contralateral homologous comparisons of thermal thresholds with QTT, and their association with age, sex or height. METHODS Fifty healthy volunteers ages 20-79 participated in the experiment. Cold detection thresholds (CDT), warm detection thresholds (WDT), heat pain thresholds (HPT), and cold pain thresholds (CPT) were measured bilaterally at the thenar eminence, anterior thigh, distal medial leg and foot dorsum. Sample normal limits were calculated as (mean) ± 2 SD. RESULTS Forty-eight subjects were included in the analysis. CPT was excluded from all analyses due to a large floor-effect. Sample normal limits for side-differences ranged from 1.8 to 7.2 °C for CDT, 2.4-6.8 °C for WDT and 3.2-4.0 °C for HPT, depending on anatomical site. For distal-proximal comparisons, sample normal limits ranged from 4.0 to 8.7 °C for CDT, 6.0-14.0 °C for WDT and 4.2-9.0 °C for HPT, depending on the pairs compared. Age was associated with side-differences for CDT in the thenar eminences (p < 0.001) and distal medial legs (p < 0.002), and with 11 of 18 distal-proximal comparisons (p < 0.01). CONCLUSIONS The normal limits for distal-proximal- and contralateral homologous thermal thresholds were wide, and thus of limited use in a clinical setting, although the reported values may be somewhat inflated by low sample-size and consequent age-pooling. Age, but not sex or height, was associated with contralateral differences in CDT in the thenar eminences and distal medial legs, and with most distal-proximal differences. SIGNIFICANCE Due to wide normal limits, we advise caution when utilizing relative comparisons of thermal thresholds for diagnostic purposes.
Collapse
Affiliation(s)
- Ø. Dunker
- Research and Communication Unit for Musculoskeletal Health (FORMI), Oslo University Hospital, Oslo, Norway
- Oslo Metropolitan University, Oslo, Norway
| | - M.U. Lie
- Research and Communication Unit for Musculoskeletal Health (FORMI), Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - K.B. Nilsen
- Research and Communication Unit for Musculoskeletal Health (FORMI), Oslo University Hospital, Oslo, Norway
- Department of Neurology and Clinical Neurophysiology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
28
|
Raasing LR, Vogels OJ, Veltkamp M, van Swol CF, Grutters JC. Current View of Diagnosing Small Fiber Neuropathy. J Neuromuscul Dis 2021; 8:185-207. [PMID: 33337383 PMCID: PMC8075405 DOI: 10.3233/jnd-200490] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Small fiber neuropathy (SFN) is a disorder of the small myelinated Aδ-fibers and unmyelinated C-fibers [5, 6]. SFN might affect small sensory fibers, autonomic fibers or both, resulting in sensory changes, autonomic dysfunction or combined symptoms [7]. As a consequence, the symptoms are potentially numerous and have a large impact on quality of life [8]. Since diagnostic methods for SFN are numerous and its pathophysiology complex, this extensive review focusses on categorizing all aspects of SFN as disease and its diagnosis. In this review, sensitivity in combination with specificity of different diagnostic methods are described using the areas under the curve. In the end, a diagnostic work-flow is suggested based on different phenotypes of SFN.
Collapse
Affiliation(s)
- Lisette R.M. Raasing
- ILD Center of Excellence, Department of Pulmonology,St Antonius Hospital, CM, Nieuwegein, The Netherlands
| | - Oscar J.M. Vogels
- Department of Neurology, St Antonius Hospital, CM, Nieuwegein, The Netherlands
| | - Marcel Veltkamp
- ILD Center of Excellence, Department of Pulmonology,St Antonius Hospital, CM, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Center Utrecht, CX, Utrecht, The Netherlands
| | | | - Jan C. Grutters
- ILD Center of Excellence, Department of Pulmonology,St Antonius Hospital, CM, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Center Utrecht, CX, Utrecht, The Netherlands
| |
Collapse
|
29
|
Denes E. Temporal and spatial concomitance of exanthema and dysesthesia in a patient with SARS-cov-2 infection. Brain Behav Immun Health 2020; 9:100165. [PMID: 33111131 PMCID: PMC7582041 DOI: 10.1016/j.bbih.2020.100165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/11/2020] [Accepted: 10/21/2020] [Indexed: 11/26/2022] Open
Affiliation(s)
- Eric Denes
- Infectious Diseases Department, ELSAN Polyclinique de Limoges, 18 Rue Du Général Catroux, 87000, Limoges, France
| |
Collapse
|
30
|
The Roles of Autoimmunity and Biotoxicosis in Sick Building Syndrome as a "Starting Point" for Irreversible Dampness and Mold Hypersensitivity Syndrome. Antibodies (Basel) 2020; 9:antib9020026. [PMID: 32580407 PMCID: PMC7345570 DOI: 10.3390/antib9020026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/13/2020] [Accepted: 06/19/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The terminology of "sick building syndrome" (SBS), meaning that a person may feel sick in a certain building, but when leaving the building, the symptoms will reverse, is imprecise. Many different environmental hazards may cause the feeling of sickness, such as high indoor air velocity, elevated noise, low or high humidity, vapors or dust. The Aim: To describe SBS in connection with exposure to indoor air dampness microbiota (DM). Methods: A search through Medline/Pubmed. Results and Conclusions: Chronic course of SBS may be avoided. By contrast, persistent or cumulative exposure to DM may make SBS potentially life-threatening and lead to irreversible dampness and mold hypersensitivity syndrome (DMHS). The corner feature of DMHS is acquired by dysregulation of the immune system in the direction of hypersensitivities (types I-IV) and simultaneous deprivation of immunity that manifests as increased susceptibility to infections. DMHS is a systemic low-grade inflammation and a biotoxicosis. There is already some evidence that DMHS may be linked to autoimmunity. Autoantibodies towards, e.g., myelin basic protein, myelin-associated glycoprotein, ganglioside GM1, smooth muscle cells and antinuclear autoantibodies were reported in mold-related illness. DMHS is also a mitochondropathy and endocrinopathy. The association of autoimmunity with DMHS should be confirmed through cohort studies preferably using chip-based technology.
Collapse
|
31
|
Bonacin YS, Marques ICS, Garcia SB, Silva SBG, Canola PA, Marques JA. The role of vanilloid receptor type 1 (TRPV1) in hyperalgesia related to bovine digital dermatitis. J Dairy Sci 2020; 103:7315-7321. [PMID: 32505399 DOI: 10.3168/jds.2019-17035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 03/31/2020] [Indexed: 12/22/2022]
Abstract
Bovine digital dermatitis is a contagious and chronic disease affecting the digits of dairy cattle worldwide. Tissue degradation may alter ionic channels and further activate vanilloid channels, more specifically the vanilloid receptor type 1 (TRPV1) that can generate and modulate hyperalgesia in cows affected with bovine digital dermatitis. The aim of this pilot study was to identify and quantify TRPV1 channels in dairy cows presenting with different stages of bovine digital dermatitis and compare these data according to the disease evolution and degree of hyperalgesia described in previous studies. Biopsies were taken from 15 lactating Holstein cows (23 lesions), and immunochemistry was performed to identify the number of TRPV1 fibers in the 4 M-stages of digital dermatitis and the control group. This pilot study had 5 experimental groups, M1 (5 samples), M2 (5 samples), M3 (4 samples), M4 (4 samples), and the control group (5 samples), with inclusion criteria was the presence of a bovine digital dermatitis lesion in at least one digit. The pilot results demonstrate an increase in expression of TRPV1 receptors in group M4 in comparison with the other groups. Bovine digital dermatitis may cause an increase in expression of TRPV1 receptors in the chronic stages of the disease, possibly contributing to the hyperalgesia described in affected animals; nevertheless, further research is needed to define this relation.
Collapse
Affiliation(s)
- Yuri S Bonacin
- Department of Clinical and Surgery, São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, São Paulo, Brazil 14884-900.
| | - Isabela C S Marques
- Department of Pathology and Legal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, São Paulo, Brazil 14049-900
| | - Sérgio B Garcia
- Department of Pathology and Legal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, São Paulo, Brazil 14049-900
| | - Samara B G Silva
- Department of Clinical and Surgery, São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, São Paulo, Brazil 14884-900
| | - Paulo A Canola
- Department of Clinical and Surgery, São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, São Paulo, Brazil 14884-900
| | - José A Marques
- Department of Clinical and Surgery, São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, São Paulo, Brazil 14884-900
| |
Collapse
|
32
|
Shoenfeld Y, Ryabkova VA, Scheibenbogen C, Brinth L, Martinez-Lavin M, Ikeda S, Heidecke H, Watad A, Bragazzi NL, Chapman J, Churilov LP, Amital H. Complex syndromes of chronic pain, fatigue and cognitive impairment linked to autoimmune dysautonomia and small fiber neuropathy. Clin Immunol 2020; 214:108384. [PMID: 32171889 DOI: 10.1016/j.clim.2020.108384] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/21/2022]
Abstract
Chronic fatigue syndrome, postural orthostatic tachycardia syndrome, complex regional pain syndrome and silicone implant incompatibility syndrome are a subject of debate among clinicians and researchers. Both the pathogenesis and treatment of these disorders require further study. In this paper we summarize the evidence regarding the role of autoimmunity in these four syndromes with respect to immunogenetics, autoimmune co-morbidities, alteration in immune cell subsets, production of autoantibodies and presentation in animal models. These syndromes could be incorporated in a new concept of autoimmune neurosensory dysautonomia with the common denominators of autoantibodies against G-protein coupled receptors and small fiber neuropathy. Sjogren's syndrome, which is a classical autoimmune disease, could serve as a disease model, illustrating the concept. Development of this concept aims to identify an apparently autoimmune subgroup of the disputable disorders, addressed in the review, which may most benefit from the immunotherapy.
Collapse
Affiliation(s)
- Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel; Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Russia.
| | - Varvara A Ryabkova
- Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Russia
| | - Carmen Scheibenbogen
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Louise Brinth
- Department of Nuclear Medicine, Herlev Gentofte Hospital, Gentofte Hospitalsvej 1, 2900 Hellerup, Denmark
| | - Manuel Martinez-Lavin
- Rheumatology Department, National Institute of Cardiology, Juan Badiano 1, 14080 Mexico City, Mexico
| | - Shuichi Ikeda
- Intractable Disease Care Center, Shinshu University Hospital, Matsumoto 390-0802, Japan
| | | | - Abdulla Watad
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel; Section of Musculoskeletal Disease, Leeds Institute of Molecular Medicine, University of Leeds, NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| | - Nicola L Bragazzi
- Postgraduate School of Public Health, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Joab Chapman
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel
| | - Leonid P Churilov
- Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Russia
| | - Howard Amital
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Aviv, Israel
| |
Collapse
|
33
|
Evidence for Dietary Agmatine Sulfate Effectiveness in Neuropathies Associated with Painful Small Fiber Neuropathy. A Pilot Open-Label Consecutive Case Series Study. Nutrients 2020; 12:nu12020576. [PMID: 32102167 PMCID: PMC7071502 DOI: 10.3390/nu12020576] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathies associated with painful small fiber neuropathy (SFN) are complex conditions, resistant to treatment with conventional medications. Previous clinical studies strongly support the use of dietary agmatine as a safe and effective treatment for neuropathic pain. Based on this evidence, we conducted an open-label consecutive case series study to evaluate the effectiveness of agmatine in neuropathies associated with painful SFN (Study Registry: ClinicalTrials.gov, System Identifier: NCT01524666). Participants diagnosed with painful SFN and autonomic dysfunctions were treated with 2.67 g/day agmatine sulfate (AgmaSet® capsules containing G-Agmatine® brand of agmatine sulfate) for a period of 2 months. Before the beginning (baseline) and at the end of the treatment period, participants answered the established 12-item neuropathic pain questionnaire specifically developed to distinguish symptoms associated with neuropathy and to quantify their severity. Secondary outcomes included other treatment options and a safety assessment. Twelve patients were recruited, and 11 patients—8 diagnosed with diabetic neuropathy, two with idiopathic neuropathy and one with inflammatory neuropathy—completed the study. All patients showed improvement in neuropathic pain to a varied extent. The average decrease in pain intensity was 26.0 rating points, corresponding to a 46.4% reduction in overall pain (p < 0.00001). The results suggest that dietary agmatine sulfate has a significant effect in reducing neuropathic pain intensity associated with painful SFN resistant to treatment with conventional neuropathic pain medications. Larger randomized placebo-controlled studies are expected to establish agmatine sulfate as a preferred treatment.
Collapse
|
34
|
Pál E, Fülöp K, Tóth P, Deli G, Pfund Z, Janszky J, Komoly S. Small Fiber Neuropathy: Clinicopathological Correlations. Behav Neurol 2020; 2020:8796519. [PMID: 32399084 PMCID: PMC7199601 DOI: 10.1155/2020/8796519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/13/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022] Open
Abstract
Small fiber neuropathy develops due to the selective damage of the thin fibers of peripheral nerves. Many common diseases can cause this condition, including diabetes, infections, autoimmune and endocrine disorders, but it can occur due to genetic alterations, as well. Eighty-five skin biopsy-proven small-fiber neuropathy cases were analyzed. Forty-one (48%) cases were idiopathic; among secondary types, hypothyreosis (9.4%), diabetes mellitus (7%), cryoglobulinemia (7%), monoclonal gammopathy with unproved significance (4.7%), Sjögren's disease (3%), and paraneoplastic neuropathy (3%) were the most common causes. Two-thirds (68%) of the patients were female, and the secondary type started 8 years later than the idiopathic one. In a vast majority of the cases (85%), the distribution followed a length-dependent pattern. Intraepidermal fiber density was comparable in idiopathic and secondary forms. Of note, we found significantly more severe pathology in men and in diabetes. Weak correlation was found between patient-reported measures and pathology, as well as with neuropathic pain-related scores. Our study confirmed the significance of small fiber damage-caused neuropathic symptoms in many clinical conditions, the gender differences in clinical settings, and pathological alterations, as well as the presence of severe small fiber pathology in diabetes mellitus, one of the most common causes of peripheral neuropathy.
Collapse
Affiliation(s)
- Endre Pál
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
- Department of Pathology, Neuropathology Unit, University of Pécs, Medical School, Pécs, Hungary
| | - Krisztina Fülöp
- Department of Pathology, Neuropathology Unit, University of Pécs, Medical School, Pécs, Hungary
| | - Péter Tóth
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
| | - Gabriella Deli
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
| | - Zoltán Pfund
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
| | - József Janszky
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
| | - Sámuel Komoly
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
| |
Collapse
|
35
|
Görlach J, Amsel D, Kölbel H, Grzybowsky M, Rutsch F, Schlierbach H, Vanlander A, Pogatzki‐Zahn E, Habig K, Garkisch S, Müller V, Fritz T, Ziegler A, Hahn A, Krämer HH, Van Coster R, Schänzer A. Diagnostic utility of small fiber analysis in skin biopsies from children with chronic pain. Muscle Nerve 2019; 61:173-181. [DOI: 10.1002/mus.26766] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Jonas Görlach
- Institute of NeuropathologyJustus‐Liebig‐University Giessen Giessen Germany
| | - Daniel Amsel
- Institute of NeuropathologyJustus‐Liebig‐University Giessen Giessen Germany
| | - Heike Kölbel
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, Children's HospitalUniversity Duisburg‐Essen Essen Germany
| | - Michelle Grzybowsky
- Department of Child NeurologyJustus‐Liebig‐University Giessen Giessen Germany
| | - Frank Rutsch
- Department of General Pediatrics, Children's HospitalUniversity of Muenster Muenster Germany
| | - Hannah Schlierbach
- Institute of NeuropathologyJustus‐Liebig‐University Giessen Giessen Germany
| | - Arnaud Vanlander
- Division of Child Neurology, Department of PediatricsUniversity Hospital Gent Gent Belgium
| | - Esther Pogatzki‐Zahn
- Department of Anesthesiology, Intensive Care and Pain MedicineUniversity Hospital Muenster Muenster Germany
| | - Kathrin Habig
- Department of NeurologyJustus‐Liebig‐University Giessen Giessen Germany
| | - Stefanie Garkisch
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, Children's HospitalUniversity Duisburg‐Essen Essen Germany
| | | | - Thorsten Fritz
- Centre for Pain Therapy and Anaesthesiology at Schloss Butzbach Butzbach Germany
| | - Andreas Ziegler
- Department of General Pediatrics and Neuropediatrics, HeidelbergUniversity Hospital Heidelberg Germany
| | - Andreas Hahn
- Department of Child NeurologyJustus‐Liebig‐University Giessen Giessen Germany
| | - Heidrun H. Krämer
- Department of NeurologyJustus‐Liebig‐University Giessen Giessen Germany
| | - Rudy Van Coster
- Division of Child Neurology, Department of PediatricsUniversity Hospital Gent Gent Belgium
| | - Anne Schänzer
- Institute of NeuropathologyJustus‐Liebig‐University Giessen Giessen Germany
| |
Collapse
|
36
|
|
37
|
Matthews CA, Deveshwar SP, Evans RJ, Badlani G, Walker SJ. Small fiber polyneuropathy as a potential therapeutic target in interstitial cystitis/bladder pain syndrome. Int Urogynecol J 2019; 30:1817-1820. [PMID: 31240362 DOI: 10.1007/s00192-019-04011-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/30/2019] [Indexed: 01/18/2023]
Abstract
INTRODUCTION AND HYPOTHESIS Interstitial cystitis/bladder pain syndrome (IC/BPS) and fibromyalgia (FM) are frequently co-occurring medical diagnoses in patients referred to the urology clinic for secondary and tertiary treatment options. METHODS Abundant literature has shown that many patients with FM have small fiber polyneuropathy (SFPN) that can be confirmed via skin punch biopsy and immunological staining to measure nerve density. RESULTS AND CONCLUSIONS This finding of SFPN provides a therapeutic target for FM and in this article we hypothesize and provide rationale for the idea that this same phenomenon (SFPN) might explain, in some IC/BPS patients, the finding of widespread pain and likewise provide a therapeutic target for these patients.
Collapse
Affiliation(s)
- Catherine A Matthews
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Shaun P Deveshwar
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, 391 Technology Way, Winston Salem, NC, 27101, USA
| | - Robert J Evans
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Gopal Badlani
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Stephen J Walker
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA.
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, 391 Technology Way, Winston Salem, NC, 27101, USA.
| |
Collapse
|
38
|
Abstract
Sensory polyneuropathies, which are caused by dysfunction of peripheral sensory nerve fibers, are a heterogeneous group of disorders that range from the common diabetic neuropathy to the rare sensory neuronopathies. The presenting symptoms, acuity, time course, severity, and subsequent morbidity vary and depend on the type of fiber that is affected and the underlying cause. Damage to small thinly myelinated and unmyelinated nerve fibers results in neuropathic pain, whereas damage to large myelinated sensory afferents results in proprioceptive deficits and ataxia. The causes of these disorders are diverse and include metabolic, toxic, infectious, inflammatory, autoimmune, and genetic conditions. Idiopathic sensory polyneuropathies are common although they should be considered a diagnosis of exclusion. The diagnostic evaluation involves electrophysiologic testing including nerve conduction studies, histopathologic analysis of nerve tissue, serum studies, and sometimes autonomic testing and cerebrospinal fluid analysis. The treatment of these diseases depends on the underlying cause and may include immunotherapy, mitigation of risk factors, symptomatic treatment, and gene therapy, such as the recently developed RNA interference and antisense oligonucleotide therapies for transthyretin familial amyloid polyneuropathy. Many of these disorders have no directed treatment, in which case management remains symptomatic and supportive. More research is needed into the underlying pathophysiology of nerve damage in these polyneuropathies to guide advances in treatment.
Collapse
Affiliation(s)
- Kelly Graham Gwathmey
- Virginia Commonwealth University, Department of Neurology, 1101 E. Marshall Street, PO Box 980599, Richmond, VA 23298, USA
| | - Kathleen T Pearson
- Virginia Commonwealth University, Department of Neurology, 1101 E. Marshall Street, PO Box 980599, Richmond, VA 23298, USA
| |
Collapse
|
39
|
Small-fiber neuropathy definition, diagnosis, and treatment. Neurol Sci 2019; 40:1343-1350. [PMID: 30968230 DOI: 10.1007/s10072-019-03871-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 03/23/2019] [Indexed: 12/13/2022]
Abstract
In the last 30 years, improvement of diagnostic methods enabled routine evaluation of small A-delta and C nerve fibers impairment, which results with the clinical condition known as a small-fiber neuropathy (SFN). This syndrome develops as a result of metabolic, toxic, immune-mediated, or genetic factors. The main clinical features include neuropathic pain and autonomic disturbance, which are occasionally disclaimed due to outstanding fatigue, daily performance decline, anxiety, and depression. As clinical, neurological, nerve conduction, and electromyography studies are commonly normal, diagnosis often depends on the finding of decreased intra-epidermal density of nerve fibers, per skin biopsy. This review highlights the etiology, clinical, diagnostic aspects, and SFN treatment.
Collapse
|