1
|
Jacoby N, Anziska Y. A Practical Approach to Diagnosing Peripheral Neuropathies. Semin Neurol 2025; 45:4-12. [PMID: 39433285 DOI: 10.1055/s-0044-1791721] [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: 10/23/2024]
Abstract
Polyneuropathies are common, with the incidence increasing with older age. The causes of polyneuropathies are diverse and numerous, and it can be challenging for clinicians to determine the etiology of a particular patient's neuropathy. In this article, we systematically detail a practical approach to polyneuropathies, beginning with the most important aspects of the workup, the history and physical. We then discuss the limited diagnostic approach required for patients who present with a distal symmetric polyneuropathy and the more comprehensive approach for patients who present with other neuropathy subtypes.
Collapse
Affiliation(s)
- Nuri Jacoby
- Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, New York
- Maimonides Medical Center, Brooklyn, New York
| | - Yaacov Anziska
- Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, New York
| |
Collapse
|
2
|
Zohar DN, Keren D, Qassim L, Eltity M, Shavit-Stein E, Chapman J, Dori A. Serum neurofilament light chain levels in patients with small-fiber neuropathy. J Neuromuscul Dis 2024:22143602241284130. [PMID: 39973399 DOI: 10.1177/22143602241284130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND Serum neurofilament light chain (sNfL) levels are an increasingly employed tool for the assessment of active axonal injury in a variety of neurological disorders including polyneuropathy. Injury to the small nerve fibers can lead to small fiber neuropathy (SFN), a neurological condition which is clinically manifested by combination of burning pain, various sensory disturbances and symptoms of autonomic dysfunction. SFN is evident by identifying reduced epidermal nerve fiber density (ENFD) in skin biopsy. OBJECTIVE To assess the utility of sNfL measurement as a marker for active axonal injury in a population of patients who were referred for skin biopsy as part of the evaluation of chronic sensory symptoms. METHODS sNfL levels were measured in 94 patients at the time of skin biopsy. Clinical, electrodiagnostic, and imaging data were collected and neurological comorbidities including central nervous system disorders, large-fiber polyneuropathy, and radiculopathy which may increase sNfL levels were reviewed. RESULTS Eighty-six patients had pathological skin biopsy result supporting the diagnosis of SFN. sNfL was increased in 9 (10%) SFN patients. Seven of them had neurological comorbidities which may explain the elevated sNfL. The other two patients had history of hypothyroidism and endometriosis. CONCLUSIONS sNfL levels are typically normal in patients with SFN. Furthermore, when sNfL are elevated, evaluation for another neurological disorder should be considered.
Collapse
Affiliation(s)
- Daniela Noa Zohar
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daria Keren
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Lamis Qassim
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mailam Eltity
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The TELEM Rubin Excellence in Biomedical Research Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Robert and Martha Harden Chair in Mental and Neurological Diseases, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Dori
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Burgess J, Marshall A, Rapteas L, Riley D, Matsumoto K, Boon C, Alchawaf A, Ferdousi M, Malik RA, Marshall A, Kaye S, Gosal D, Frank B, Alam U. Idiopathic Distal Sensory Polyneuropathy and Fibromyalgia Syndrome: A Comparative Phenotyping Study. Pain Ther 2024; 13:1541-1558. [PMID: 39264538 PMCID: PMC11543958 DOI: 10.1007/s40122-024-00646-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
INTRODUCTION Painful idiopathic distal sensory polyneuropathy (IDSP) and fibromyalgia syndrome (FMS) are cryptogenic chronic pain syndromes. The contribution of small fibre pathology (SFP) in FMS remains controversial. This study aims to quantify small nerve pathology in participants with IDSP and FMS and identify relationships of SFP with sensory phenotypes. METHODS In this study, 73 individuals (FMS: 25, IDSP: 23, healthy volunteers: 25) underwent comprehensive assessment, including neurological exams, questionnaires, sensory tests, and corneal confocal microscopy. RESULTS IDSP participants displayed lower wind-up ratio (WUR) relative to FMS (p < 0.001), loss of function to thermal and mechanical stimuli and elevated neuropathy disability scores compared to FMS and healthy volunteers (all p < 0.001). FMS participants demonstrated gain of function to heat and blunt pressure pain responses relative to IDSP, and healthy volunteers (heat: p = 0.002 and p = 0.003; pressure: both p < 0.001) and WUR (both p < 0.001). FMS participants exhibited reduced corneal nerve fibre density (p = 0.02), while IDSP participants had lower global corneal nerve measures (density, branch density, and length) relative to healthy volunteers (all p < 0.001). Utilising corneal nerve fibre length, SFP was demonstrated in 66.6% of participants (FMS: 13/25; IDSP: 22/23). CONCLUSION Participants with SFP, in both FMS and IDSP, reported symptoms indicative of small nerve fibre disease. Although distinctions in pain distributions are evident between individuals with FMS and IDSP, over 50% of participants between the two conditions displayed both a loss and gain of thermal and mechanical function suggestive of shared mechanisms. However, sensory phenotypes were associated with the presence of SFP in IDSP but not in FMS.
Collapse
Affiliation(s)
- Jamie Burgess
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Clinical Sciences Centre, Aintree University Hospital, Longmoor Lane, Liverpool, L9 7AL, UK.
| | - Anne Marshall
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Leandros Rapteas
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - David Riley
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Liverpool University Hospitals NHS Foundation Trust, Aintree Hospital, Liverpool, UK
| | - Kohei Matsumoto
- Liverpool University Hospitals NHS Foundation Trust, Aintree Hospital, Liverpool, UK
| | - Cheng Boon
- Department of Clinical Oncology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | | | - Maryam Ferdousi
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rayaz A Malik
- Division of Medicine, Qatar Foundation, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Andrew Marshall
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Clinical Neurophysiology, The Walton Centre, Liverpool, UK
- Pain Research Institute, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Stephen Kaye
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - David Gosal
- Department of Neurology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Bernhard Frank
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Pain Medicine, The Walton Centre, Liverpool, UK
- Pain Research Institute, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Uazman Alam
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Pain Research Institute, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
- Department of Diabetes and Endocrinology, Liverpool University Hospital NHS Foundation Trust, Liverpool, UK
- Centre for Biomechanics and Rehabilitation Technologies, Staffordshire University, Stoke-on-Trent, UK
| |
Collapse
|
4
|
Kool D, Hoeijmakers JG, Waxman SG, Faber CG. Small fiber neuropathy. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:181-231. [PMID: 39580213 DOI: 10.1016/bs.irn.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Small fiber neuropathy (SFN) is a condition involving the small nerve fibers of the peripheral nervous system, specifically the thinly myelinated Aδ and unmyelinated C fibers. It is an increasingly acknowledged condition within the spectrum of neuropathic pain disorders, leading to a rise in diagnosed patients. SFN is characterized by neuropathic pain, that is often described as burning, and typically presents in the hands and feet ascending proximally. Since small nerve fibers are involved in the autonomic nervous system, SFN can also lead to autonomic dysfunction. In the clinical setting, SFN diagnosis is frequently based on the Besta Criteria, which include skin biopsy and quantitative sensory testing. For clinical trials, the ACTTION criteria are also recommended. However, the diagnostic process is often complex, prompting research towards more accessible diagnostic methods. The pathophysiology of SFN remains unclear, thereby challenging therapeutic strategies. A large variety of underlying conditions has been associated with SFN, including metabolic, immune-mediated, infectious, toxic and hereditary conditions. The discovery of genetic sodium channelopathies in SFN provides insight into its underlying mechanisms. Newly discovered mutations within these genes reveal that SFN often shows overlapping clinical presentations with other sodium channelopathies. This chapter provides an in-depth look at SFN, including its clinical features, diagnostic methods, underlying conditions and possible therapeutic strategies.
Collapse
Affiliation(s)
- Dennis Kool
- Department of Neurology, Mental Health and Neuroscience Research Institute, Maastricht University Medical Center+, Maastricht, Netherlands.
| | - Janneke Gj Hoeijmakers
- Department of Neurology, Mental Health and Neuroscience Research Institute, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, United States; Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Catharina G Faber
- Department of Neurology, Mental Health and Neuroscience Research Institute, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
5
|
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
|
6
|
Røikjer J, Croosu SS, Sejergaard BF, Hansen TM, Frøkjær JB, Søndergaard CB, Petropoulos IN, Malik RA, Nielsen E, Mørch CD, Ejskjaer N. Diagnostic Accuracy of Perception Threshold Tracking in the Detection of Small Fiber Damage in Type 1 Diabetes. J Diabetes Sci Technol 2024; 18:1157-1164. [PMID: 36825610 PMCID: PMC11418516 DOI: 10.1177/19322968231157431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
AIM An objective assessment of small nerve fibers is key to the early detection of diabetic peripheral neuropathy (DPN). This study investigates the diagnostic accuracy of a novel perception threshold tracking technique in detecting small nerve fiber damage. METHODS Participants with type 1 diabetes (T1DM) without DPN (n = 20), with DPN (n = 20), with painful DPN (n = 20) and 20 healthy controls (HCs) underwent perception threshold tracking on the foot and corneal confocal microscopy. Diagnostic accuracy of perception threshold tracking compared to corneal confocal microscopy was analyzed using logistic regression. RESULTS The rheobase, corneal nerve fiber density (CNFD), corneal nerve branch density (CNBD), and corneal nerve fiber length (CNFL) (all P < .001) differed between groups. The diagnostic accuracy of perception threshold tracking (rheobase) was excellent for identifying small nerve fiber damage, especially for CNFL with a sensitivity of 94%, specificity 94%, positive predictive value 97%, and negative predictive value 89%. There was a significant correlation between rheobase with CNFD, CNBD, CNFL, and Michigan Neuropathy Screening Instrument (all P < .001). CONCLUSION Perception threshold tracking had a very high diagnostic agreement with corneal confocal microscopy for detecting small nerve fiber loss and may have clinical utility for assessing small nerve fiber damage and hence early DPN. CLINICAL TRIALS NCT04078516.
Collapse
Affiliation(s)
- Johan Røikjer
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Suganthiya Santhiapillai Croosu
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Tine Maria Hansen
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jens Brøndum Frøkjær
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | | | | | - Esben Nielsen
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | - Carsten Dahl Mørch
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Center for Neuroplasticity and Pain, Center for Sensory-Motor Interaction, Aalborg, Denmark
| | - Niels Ejskjaer
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Clinical Medicine and Endocrinology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
7
|
Oaklander AL. Increasing associations of long-COVID with small-fiber neuropathy. Pain 2024; 165:e93-e95. [PMID: 39159474 PMCID: PMC11343176 DOI: 10.1097/j.pain.0000000000003260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 08/21/2024]
Affiliation(s)
- Anne Louise Oaklander
- Dept. Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- Dept. Pathology (Neuropathology), Massachusetts General Hospital, Boston, USA
| |
Collapse
|
8
|
Burgess J, Marshall A, Rapteas L, Hamill KJ, Marshall A, Malik RA, Frank B, Alam U. Automated immunohistochemistry of intra-epidermal nerve fibres in skin biopsies: A proof-of-concept study. J Peripher Nerv Syst 2024; 29:329-338. [PMID: 39164223 DOI: 10.1111/jns.12650] [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: 05/16/2024] [Revised: 07/15/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
AIMS To develop a standardised, automated protocol for detecting protein gene product 9.5 (PGP9.5) positive intra-epidermal nerve fibres (IENFs) in skin biopsies, transitioning from the established manual technique to an automated platform. This automated method, although currently intended for research applications, may improve the accessibility of this diagnostic test for small fibre neuropathy in clinical settings. METHODS Skin biopsies (n = 274) from 100 participants (fibromyalgia syndrome n = 62; idiopathic small fibre neuropathy: n = 16; healthy volunteers: n = 22) were processed using an automated immunohistochemistry platform. IENF quantification was performed by blinded examiners, with reliability assessed via a two-way mixed-effects model to evaluate inter- and intra-observer variability. RESULTS The automated staining system reproduced intra-epidermal nerve fibre density (IENFD) counts consistent with free-floating sections (mean ± standard deviation: free-floating: 5.6 ± 3.4 fibres/mm; automated: 5.9 ± 3.2 fibres/mm). A median difference of 0.3 with a lower bound 95% Confidence Interval (CI) at -0.00005 established non-inferiority against a margin of -0.4 (p = .08). Specifically, the inter-class correlation coefficient (class denotes consistency in measured observations) was 99% (95% CI: 0.9-1), indicating excellent agreement between free-floating and automated methods. The inter- and intra-class coefficient between examiners were both 99% (95% CI: 0.9-0.1) for IENFD, demonstrating high reliability using sections stained using the automated method. INTERPRETATION Automated immunohistochemistry provides high-throughput reliable and reproducible intra-epidermal nerve fibre quantification. This method, although currently proof-of-concept, for research use only, may be more widely deployed in histopathology laboratories to increase the adoption of IENFD assessment for the diagnosis of peripheral neuropathies.
Collapse
Affiliation(s)
- Jamie Burgess
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Anne Marshall
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Leandros Rapteas
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Kevin J Hamill
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Andrew Marshall
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- The Walton Centre NHS Foundation Trust, Liverpool, UK
| | | | | | - Uazman Alam
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Cardiovascular and Metabolic Medicine, University of Liverpool, Liverpool, UK
- Centre for Biomechanics and Rehabilitation Technologies, Staffordshire University, Stoke-on-Trent, UK
| |
Collapse
|
9
|
Currò R, Dominik N, Facchini S, Vegezzi E, Sullivan R, Galassi Deforie V, Fernández-Eulate G, Traschütz A, Rossi S, Garibaldi M, Kwarciany M, Taroni F, Brusco A, Good JM, Cavalcanti F, Hammans S, Ravenscroft G, Roxburgh RH, Parolin Schnekenberg R, Rugginini B, Abati E, Manini A, Quartesan I, Ghia A, Lòpez de Munaìn A, Manganelli F, Kennerson M, Santorelli FM, Infante J, Marques W, Jokela M, Murphy SM, Mandich P, Fabrizi GM, Briani C, Gosal D, Pareyson D, Ferrari A, Prados F, Yousry T, Khurana V, Kuo SH, Miller J, Troakes C, Jaunmuktane Z, Giunti P, Hartmann A, Basak N, Synofzik M, Stojkovic T, Hadjivassiliou M, Reilly MM, Houlden H, Cortese A. Role of the repeat expansion size in predicting age of onset and severity in RFC1 disease. Brain 2024; 147:1887-1898. [PMID: 38193360 PMCID: PMC11068103 DOI: 10.1093/brain/awad436] [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: 10/06/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 01/10/2024] Open
Abstract
RFC1 disease, caused by biallelic repeat expansion in RFC1, is clinically heterogeneous in terms of age of onset, disease progression and phenotype. We investigated the role of the repeat size in influencing clinical variables in RFC1 disease. We also assessed the presence and role of meiotic and somatic instability of the repeat. In this study, we identified 553 patients carrying biallelic RFC1 expansions and measured the repeat expansion size in 392 cases. Pearson's coefficient was calculated to assess the correlation between the repeat size and age at disease onset. A Cox model with robust cluster standard errors was adopted to describe the effect of repeat size on age at disease onset, on age at onset of each individual symptoms, and on disease progression. A quasi-Poisson regression model was used to analyse the relationship between phenotype and repeat size. We performed multivariate linear regression to assess the association of the repeat size with the degree of cerebellar atrophy. Meiotic stability was assessed by Southern blotting on first-degree relatives of 27 probands. Finally, somatic instability was investigated by optical genome mapping on cerebellar and frontal cortex and unaffected peripheral tissue from four post-mortem cases. A larger repeat size of both smaller and larger allele was associated with an earlier age at neurological onset [smaller allele hazard ratio (HR) = 2.06, P < 0.001; larger allele HR = 1.53, P < 0.001] and with a higher hazard of developing disabling symptoms, such as dysarthria or dysphagia (smaller allele HR = 3.40, P < 0.001; larger allele HR = 1.71, P = 0.002) or loss of independent walking (smaller allele HR = 2.78, P < 0.001; larger allele HR = 1.60; P < 0.001) earlier in disease course. Patients with more complex phenotypes carried larger expansions [smaller allele: complex neuropathy rate ratio (RR) = 1.30, P = 0.003; cerebellar ataxia, neuropathy and vestibular areflexia syndrome (CANVAS) RR = 1.34, P < 0.001; larger allele: complex neuropathy RR = 1.33, P = 0.008; CANVAS RR = 1.31, P = 0.009]. Furthermore, larger repeat expansions in the smaller allele were associated with more pronounced cerebellar vermis atrophy (lobules I-V β = -1.06, P < 0.001; lobules VI-VII β = -0.34, P = 0.005). The repeat did not show significant instability during vertical transmission and across different tissues and brain regions. RFC1 repeat size, particularly of the smaller allele, is one of the determinants of variability in RFC1 disease and represents a key prognostic factor to predict disease onset, phenotype and severity. Assessing the repeat size is warranted as part of the diagnostic test for RFC1 expansion.
Collapse
Affiliation(s)
- Riccardo Currò
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Natalia Dominik
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Stefano Facchini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | | | - Roisin Sullivan
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | | | - Gorka Fernández-Eulate
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, 75013 Paris, France
| | - Andreas Traschütz
- Research Division ‘Translational Genomics of Neurodegenerative Diseases’, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, 72076 Tübingen, Germany
| | - Salvatore Rossi
- Dipartimento di Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, UOC Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Matteo Garibaldi
- Neuromuscular and Rare Disease Center, Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Sant'Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | - Mariusz Kwarciany
- Department of Adult Neurology, Medical University of Gdańsk, 80-952 Gdańsk, Poland
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, 10124 Turin, Italy
| | - Jean-Marc Good
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Francesca Cavalcanti
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), 87050 Mangone, Italy
| | - Simon Hammans
- Wessex Neurological Centre, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Gianina Ravenscroft
- Neurogenetic Diseases Group, Centre for Medical Research, QEII Medical Centre, University of Western Australia, Nedland, WA 6009, Australia
| | - Richard H Roxburgh
- Neurology Department, Auckland City Hospital, New Zealand and the Centre for Brain Research, University of Auckland, Auckland 1142, New Zealand
| | | | - Bianca Rugginini
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Elena Abati
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Arianna Manini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Ilaria Quartesan
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Arianna Ghia
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Adolfo Lòpez de Munaìn
- Neurology Department, Donostia University Hospital, University of the Basque Country-Osakidetza-CIBERNED-Biodonostia, 20014 Donostia-San Sebastián, Spain
| | - Fiore Manganelli
- Department of Neuroscience and Reproductive and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Marina Kennerson
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
| | - Filippo Maria Santorelli
- IRCCS Stella Maris Foundation, Molecular Medicine for Neurodegenerative and Neuromuscular Disease Unit, 56128 Pisa, Italy
| | - Jon Infante
- University Hospital Marquès de Valdecilla-IDIVAL, University of Cantabria, 39008 Santander, Spain
| | - Wilson Marques
- Department of Neurology, School of Medicine of Ribeirão Preto, University of São Paulo, 2650 Ribeirão Preto, Brazil
| | - Manu Jokela
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, 33520 Tampere, Finland
- Neurocenter, Department of Neurology, Clinical Neurosciences, Turku University Hospital and University of Turku, 20014 Turku, Finland
| | - Sinéad M Murphy
- Department of Neurology, Tallaght University Hospital, D24 NR0A Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Paola Mandich
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino-UOC Genetica Medica, 16132 Genova, Italy
| | - Gian Maria Fabrizi
- Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Chiara Briani
- Department of Neurosciences, ERN Neuromuscular Unit, University of Padova, 35100 Padova, Italy
| | - David Gosal
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Greater Manchester, M6 8HD, UK
| | - Davide Pareyson
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | | | - Ferran Prados
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College London, London, WC1V 6LJ, UK
- NMR Research Unit, Institute of Neurology, University College London (UCL), London, WC1N 3BG, UK
- e-Health Centre, Universitat Oberta de Catalunya, 08018 Barcelona, Spain
| | - Tarek Yousry
- Neuroradiological Academic Unit, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Vikram Khurana
- Division of Movement Disorders and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sheng-Han Kuo
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - James Miller
- Department of Neurology, Royal Victoria Hospitals, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, NE1 4LP, UK
| | - Claire Troakes
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE21 8EA, UK
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Paola Giunti
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Annette Hartmann
- Division of General Psychiatry, Medical University of Vienna, 1090 Vienna, Austria
| | - Nazli Basak
- Koç University, School of Medicine, Suna and İnan Kıraç Foundation, Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine, 34010 Istanbul, Turkey
| | - Matthis Synofzik
- Research Division ‘Translational Genomics of Neurodegenerative Diseases’, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, 72076 Tübingen, Germany
| | - Tanya Stojkovic
- Nord/Est/Ile-de-France Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital, APHP, 75013 Paris, France
| | - Marios Hadjivassiliou
- Academic Department of Neurosciences, Sheffield Teaching Hospitals NHS Trust and University of Sheffield, Sheffield, S10 2JF, UK
| | - Mary M Reilly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Andrea Cortese
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
10
|
Geerts M, Hoeijmakers JGJ, Essers BAB, Merkies ISJ, Faber CG, Goossens MEJB. Patient satisfaction and patient accessibility in a small fiber neuropathy diagnostic service in the Netherlands: A single-center, prospective, survey-based cohort study. PLoS One 2024; 19:e0298881. [PMID: 38626240 PMCID: PMC11020963 DOI: 10.1371/journal.pone.0298881] [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: 12/22/2023] [Accepted: 01/27/2024] [Indexed: 04/18/2024] Open
Abstract
INTRODUCTION Small fiber neuropathy (SFN) is a common cause of neuropathic pain in peripheral neuropathies. Good accessibility of diagnostics and treatment is necessary for an accurate diagnosis and treatment of SFN. Evidence is lacking on the quality performance of the diagnostic SFN service in the Netherlands. Our aim was to determine the patient satisfaction and -accessibility of the diagnostic SFN service, and to identify areas for improvement. METHODS In a single-center, prospective, survey-based cohort study, 100 visiting patients were asked to fill in the SFN patient satisfaction questionnaire (SFN-PSQ), with 10 domains and 51 items. Cut-off point for improvement was defined as ≥ 25% dissatisfaction on an item. A chi-square test and linear regression analyses was used for significant differences and associations of patient satisfaction. RESULTS From November 2020 to May 2021, 98 patients with SFN-related complaints filled in the online SFN-PSQ within 20 minutes. In 84% of the patients SFN was confirmed, average age was 55.1 (52.5-57.8) years and 67% was female. High satisfaction was seen in the domains 'Waiting List Period', Chest X-ray', 'Consultation with the Doctor or Nurse Practitioner (NP)', 'Separate Consultation with the Doctor or NP about Psychological Symptoms', and 'General' of the SFN service. Overall average patient satisfaction score was 8.7 (IQR 8-10) on a 1-to-10 rating scale. Main area for improvement was shortening the 8-week period for receiving the results of the diagnostic testing (p < 0.05). General health status was statistically significant associated with patient satisfaction (p < 0.05). CONCLUSION A good reflection of the high patient satisfaction and -accessibility of the SFN-service is shown, with important points for improvement. These results could help hospitals widely to optimize the logistic and diagnostic pathway of SFN analysis, benchmarking patient satisfaction results among the hospitals, and to improve the quality of care of comparable SFN services.
Collapse
Affiliation(s)
- Margot Geerts
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Brigitte A. B. Essers
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ingemar S. J. Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Neurology, Curacao Medical Center, Willemstad, Curacao
| | - Catharina G. Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Mariëlle E. J. B. Goossens
- Department of Rehabilitation Research & Department of Clinical Psychological Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
11
|
Gad H, Kalra S, Pinzon R, Garcia RAN, Yotsombut K, Coetzee A, Nafach J, Lim LL, Fletcher PE, Lim V, Malik RA. Earlier diagnosis of peripheral neuropathy in primary care: A call to action. J Peripher Nerv Syst 2024; 29:28-37. [PMID: 38268316 DOI: 10.1111/jns.12613] [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: 10/16/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Peripheral neuropathy (PN) often remains undiagnosed (~80%). Earlier diagnosis of PN may reduce morbidity and enable earlier risk factor reduction to limit disease progression. Diabetic peripheral neuropathy (DPN) is the most common PN and the 10 g monofilament is endorsed as an inexpensive and easily performed test for DPN. However, it only detects patients with advanced neuropathy at high risk of foot ulceration. There are many validated questionnaires to diagnose PN, but they can be time-consuming and have complex scoring systems. Primary care physicians (PCPs) have busy clinics and lack access to a readily available screening method to diagnose PN. They would prefer a short, simple, and accurate tool to screen for PN. Involving the patient in the screening process would not only reduce the time a physician requires to make a diagnosis but would also empower the patient. Following an expert meeting of diabetologists and neurologists from the Middle East, South East Asia and Latin America, a consensus was formulated to help improve the diagnosis of PN in primary care using a simple tool for patients to screen themselves for PN followed by a consultation with the physician to confirm the diagnosis.
Collapse
Affiliation(s)
- Hoda Gad
- Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
| | - Rizaldy Pinzon
- Neurology Department of the Bethesda, General Hospital Yogyakarta, Yogyakarta, Indonesia
| | - Rey-An Nino Garcia
- College of Medicine, De LA Salle, Health Medical and Science Institute College of Medicine, Manila, Philippines
| | - Kitiyot Yotsombut
- Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Ankia Coetzee
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jalal Nafach
- Dubai Diabetes Center, Dubai Academic Health Corporation, Dubai, UAE
| | - Lee-Ling Lim
- Department of Medicine, Diabetes Care Unit, University of Malaya, Kuala Lumpur, Malaysia
| | - Pablo E Fletcher
- Endocrinology Department, Medical School, University of Panama, Panama, Panama
| | - Vivien Lim
- Endocrinology Department, Gleneagles Hospital, Singapore, Singapore
| | - Rayaz A Malik
- Research Department, Weill Cornell Medicine-Qatar, Doha, Qatar
- Institute of Cardiovascular Medicine, University of Manchester, Manchester, UK
| |
Collapse
|
12
|
Gendre T, Lefaucheur JP, Nordine T, Baba-Amer Y, Authier FJ, Devaux J, Créange A. Characterizing Acute-Onset Small Fiber Neuropathy. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200195. [PMID: 38170952 PMCID: PMC10766082 DOI: 10.1212/nxi.0000000000200195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/06/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND AND OBJECTIVES Immune-mediated small fiber neuropathy (SFN) is increasingly recognized. Acute-onset SFN (AOSFN) remains poorly described. Herein, we report a series of AOSFN cases in which immune origins are debatable. METHODS We included consecutive patients with probable or definite AOSFN. Diagnosis of SFN was based on the NEURODIAB criteria. Acute onset was considered when the maximum intensity and extension of both symptoms and signs were reached within 28 days. We performed the following investigations: clinical examination, neurophysiologic assessment encompassing a nerve conduction study to rule out large fiber neuropathy, laser-evoked potentials (LEPs), warm detection thresholds (WDTs), electrochemical skin conductance (ESC), epidermal nerve fiber density (ENF), and patient serum reactivity against mouse sciatic nerve teased fibers, mouse dorsal root ganglion (DRG) sections, and cultured DRG. The serum reactivity of healthy subjects (n = 10) and diseased controls (n = 12) was also analyzed. Data on baseline characteristics, biological investigations, and disease course were collected. RESULTS Twenty patients presenting AOSFN were identified (60% women; median age: 44.2 years [interquartile range: 35.7-56.2]). SFN was definite in 18 patients (90%) and probable in 2 patients. A precipitating event was present in 16 patients (80%). The median duration of the progression phase was 14 days [5-28]. Pain was present in 17 patients (85%). Twelve patients (60%) reported autonomic involvement. The clinical pattern was predominantly non-length-dependent (85%). Diagnosis was confirmed by abnormal LEPs (60%), ENF (55%), WDT (39%), or ESC (31%). CSF analysis was normal in 5 of 5 patients. Antifibroblast growth factor 3 antibodies were positive in 4 of 18 patients (22%) and anticontactin-associated protein-2 antibodies in one patient. In vitro studies showed IgG immunoreactivity against nerve tissue in 14 patients (70%), but not in healthy subjects or diseased controls. Patient serum antibodies bound to unmyelinated fibers, Schwann cells, juxtaparanodes, paranodes, or DRG. Patients' condition improved after a short course of oral corticosteroids (3/3). Thirteen patients (65%) showed partial or complete recovery. Others displayed relapses or a chronic course. DISCUSSION AOSFN primarily presents as an acute, non-length-dependent, symmetric painful neuropathy with a variable disease course. An immune-mediated origin has been suggested based on in vitro immunohistochemical studies.
Collapse
Affiliation(s)
- Thierry Gendre
- From the Service de Neurologie (T.G., A.C.), CHU Henri Mondor APHP; Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France (T.G., J.-P.L., T.N., F.-J.A., A.C.); Unité de Neurophysiologie Clinique (J.-P.L., T.N.), CHU Henri Mondor APHP; Unité de Recherche EA 4391 (J.-P.L., T.N., A.C.), Faculté de Santé, UniversitéParis Est Créteil; IMRB INSERM U955-Equipe 10 (Y.B.-A., F.-J.A.), UniversitéParis Est Créteil; Service d'Anatomo-Pathologie (F.-J.A.), CHU Henri Mondor APHP, Créteil; and Institut de Génomique Fonctionnelle (J.D.), Universitéde Montpellier, CNRS, INSERM, France
| | - Jean-Pascal Lefaucheur
- From the Service de Neurologie (T.G., A.C.), CHU Henri Mondor APHP; Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France (T.G., J.-P.L., T.N., F.-J.A., A.C.); Unité de Neurophysiologie Clinique (J.-P.L., T.N.), CHU Henri Mondor APHP; Unité de Recherche EA 4391 (J.-P.L., T.N., A.C.), Faculté de Santé, UniversitéParis Est Créteil; IMRB INSERM U955-Equipe 10 (Y.B.-A., F.-J.A.), UniversitéParis Est Créteil; Service d'Anatomo-Pathologie (F.-J.A.), CHU Henri Mondor APHP, Créteil; and Institut de Génomique Fonctionnelle (J.D.), Universitéde Montpellier, CNRS, INSERM, France
| | - Tarik Nordine
- From the Service de Neurologie (T.G., A.C.), CHU Henri Mondor APHP; Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France (T.G., J.-P.L., T.N., F.-J.A., A.C.); Unité de Neurophysiologie Clinique (J.-P.L., T.N.), CHU Henri Mondor APHP; Unité de Recherche EA 4391 (J.-P.L., T.N., A.C.), Faculté de Santé, UniversitéParis Est Créteil; IMRB INSERM U955-Equipe 10 (Y.B.-A., F.-J.A.), UniversitéParis Est Créteil; Service d'Anatomo-Pathologie (F.-J.A.), CHU Henri Mondor APHP, Créteil; and Institut de Génomique Fonctionnelle (J.D.), Universitéde Montpellier, CNRS, INSERM, France
| | - Yasmine Baba-Amer
- From the Service de Neurologie (T.G., A.C.), CHU Henri Mondor APHP; Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France (T.G., J.-P.L., T.N., F.-J.A., A.C.); Unité de Neurophysiologie Clinique (J.-P.L., T.N.), CHU Henri Mondor APHP; Unité de Recherche EA 4391 (J.-P.L., T.N., A.C.), Faculté de Santé, UniversitéParis Est Créteil; IMRB INSERM U955-Equipe 10 (Y.B.-A., F.-J.A.), UniversitéParis Est Créteil; Service d'Anatomo-Pathologie (F.-J.A.), CHU Henri Mondor APHP, Créteil; and Institut de Génomique Fonctionnelle (J.D.), Universitéde Montpellier, CNRS, INSERM, France
| | - François-Jérôme Authier
- From the Service de Neurologie (T.G., A.C.), CHU Henri Mondor APHP; Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France (T.G., J.-P.L., T.N., F.-J.A., A.C.); Unité de Neurophysiologie Clinique (J.-P.L., T.N.), CHU Henri Mondor APHP; Unité de Recherche EA 4391 (J.-P.L., T.N., A.C.), Faculté de Santé, UniversitéParis Est Créteil; IMRB INSERM U955-Equipe 10 (Y.B.-A., F.-J.A.), UniversitéParis Est Créteil; Service d'Anatomo-Pathologie (F.-J.A.), CHU Henri Mondor APHP, Créteil; and Institut de Génomique Fonctionnelle (J.D.), Universitéde Montpellier, CNRS, INSERM, France
| | - Jérôme Devaux
- From the Service de Neurologie (T.G., A.C.), CHU Henri Mondor APHP; Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France (T.G., J.-P.L., T.N., F.-J.A., A.C.); Unité de Neurophysiologie Clinique (J.-P.L., T.N.), CHU Henri Mondor APHP; Unité de Recherche EA 4391 (J.-P.L., T.N., A.C.), Faculté de Santé, UniversitéParis Est Créteil; IMRB INSERM U955-Equipe 10 (Y.B.-A., F.-J.A.), UniversitéParis Est Créteil; Service d'Anatomo-Pathologie (F.-J.A.), CHU Henri Mondor APHP, Créteil; and Institut de Génomique Fonctionnelle (J.D.), Universitéde Montpellier, CNRS, INSERM, France
| | - Alain Créange
- From the Service de Neurologie (T.G., A.C.), CHU Henri Mondor APHP; Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France (T.G., J.-P.L., T.N., F.-J.A., A.C.); Unité de Neurophysiologie Clinique (J.-P.L., T.N.), CHU Henri Mondor APHP; Unité de Recherche EA 4391 (J.-P.L., T.N., A.C.), Faculté de Santé, UniversitéParis Est Créteil; IMRB INSERM U955-Equipe 10 (Y.B.-A., F.-J.A.), UniversitéParis Est Créteil; Service d'Anatomo-Pathologie (F.-J.A.), CHU Henri Mondor APHP, Créteil; and Institut de Génomique Fonctionnelle (J.D.), Universitéde Montpellier, CNRS, INSERM, France
| |
Collapse
|
13
|
Berry D, Ene J, Nathani A, Singh M, Li Y, Zeng C. Effects of Physical Cues on Stem Cell-Derived Extracellular Vesicles toward Neuropathy Applications. Biomedicines 2024; 12:489. [PMID: 38540102 PMCID: PMC10968089 DOI: 10.3390/biomedicines12030489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 11/28/2024] Open
Abstract
The peripheral nervous system undergoes sufficient stress when affected by diabetic conditions, chemotherapeutic drugs, and personal injury. Consequently, peripheral neuropathy arises as the most common complication, leading to debilitating symptoms that significantly alter the quality and way of life. The resulting chronic pain requires a treatment approach that does not simply mask the accompanying symptoms but provides the necessary external environment and neurotrophic factors that will effectively facilitate nerve regeneration. Under normal conditions, the peripheral nervous system self-regenerates very slowly. The rate of progression is further hindered by the development of fibrosis and scar tissue formation, which does not allow sufficient neurite outgrowth to the target site. By incorporating scaffolding supplemented with secretome derived from human mesenchymal stem cells, it is hypothesized that neurotrophic factors and cellular signaling can facilitate the optimal microenvironment for nerve reinnervation. However, conventional methods of secretory vesicle production are low yield, thus requiring improved methods to enhance paracrine secretions. This report highlights the state-of-the-art methods of neuropathy treatment as well as methods to optimize the clinical application of stem cells and derived secretory vesicles for nerve regeneration.
Collapse
Affiliation(s)
- Danyale Berry
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida Agricultural and Mechanical University, Tallahassee, FL 32310, USA;
- High Performance Materials Institute, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 23210, USA
| | - Justice Ene
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida Agricultural and Mechanical University, Tallahassee, FL 32307, USA; (A.N.); (M.S.)
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida Agricultural and Mechanical University, Tallahassee, FL 32307, USA; (A.N.); (M.S.)
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
| | - Changchun Zeng
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida Agricultural and Mechanical University, Tallahassee, FL 32310, USA;
- High Performance Materials Institute, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 23210, USA
| |
Collapse
|
14
|
Fischer F, Dohrn MF, Kapfenberger R, Igharo D, Seeber D, de Moya Rubio E, Pitarokoili K, Börsch N, Mücke M, Rolke R, Schulz JB, Maier A. [Neuropathic pain as a symptom in autonomic neuropathies and other rare diseases : Small fiber neuropathy: its recognition, diagnosis, and treatment]. Schmerz 2024; 38:33-40. [PMID: 38197939 DOI: 10.1007/s00482-023-00783-w] [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] [Accepted: 12/18/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND Neuropathic pain is difficult to diagnose and treat. Small fiber neuropathy (SFN) flies under the radar of nerve conduction studies. OBJECTIVES The importance of a structured patient history and physical examination in the context of neuropathic pain is emphasized. Describing SFN as an important cause, the authors consider rare but partially treatable differential diagnoses. They conclude that autonomic symptoms are frequently associated, often presenting with diverse symptoms. METHODS A selective literature research to present SFN symptoms as well as differential diagnostic and therapeutic steps in the context of SFN and rare diseases focusing on the autonomic nervous system. RESULTS Neuropathic pain significantly reduces quality of life. To shorten the time until diagnosis and to initiate therapy, the authors recommend a structured patient history including sensory plus and minus symptoms and non-specific autonomic signs. If the initial search for the cause is not successful, rare causes such as treatable transthyretin (ATTR) amyloidosis and Fabry's disease or autoimmune causes should be considered, particularly in the case of progressive and/or autonomic symptoms. CONCLUSION The diagnosis and therapy of rare SFN requires interdisciplinary collaboration and, in many cases, a referral to specialized centers to achieve the best patient care.
Collapse
Affiliation(s)
- Fiona Fischer
- Klinik für Neurologie, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
| | - Maike F Dohrn
- Klinik für Neurologie, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
| | - Romina Kapfenberger
- Klinik für Neurologie, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
| | - Denver Igharo
- Klinik für Neurologie, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
- Hals-Nasen-Ohren-Klinik, Helios Klinikum Krefeld, Krefeld, Deutschland
| | - Diana Seeber
- Klinik für Neurologie, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
| | - Elena de Moya Rubio
- POTS und andere Dysautonomien e. V., Bochum, Deutschland
- Marfan Hilfe (Deutschland) e. V., Olpe, Deutschland
| | - Kalliopi Pitarokoili
- Neurologische Universitätsklinik am St. Josef Hospital Katholisches Klinikum Bochum, Bochum, Deutschland
| | - Natalie Börsch
- Institut für Digitale Allgemeinmedizin, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
- Zentrum für Seltene Erkrankungen Aachen (ZSEA), Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
| | - Martin Mücke
- Institut für Digitale Allgemeinmedizin, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
- Zentrum für Seltene Erkrankungen Aachen (ZSEA), Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
| | - Roman Rolke
- Klinik für Palliativmedizin, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
| | - Jörg B Schulz
- Klinik für Neurologie, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland
- JARA-BRAIN Institut II, Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich GmbH und RWTH Aachen, Jülich, Deutschland
| | - Andrea Maier
- Klinik für Neurologie, Medizinische Fakultät, RWTH Aachen, Aachen, Deutschland.
- POTS und andere Dysautonomien e. V., Bochum, Deutschland.
| |
Collapse
|
15
|
Zirpoli GR, Farhad K, Klein MC, Downs S, Klein MM, Oaklander AL. Initial validation of the Mass. General Neuropathy Exam Tool (MAGNET) for evaluation of distal small-fiber neuropathy. Muscle Nerve 2024; 69:185-198. [PMID: 38112169 PMCID: PMC10842781 DOI: 10.1002/mus.28013] [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: 06/28/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 12/20/2023]
Abstract
INTRODUCTION/AIMS Diagnosis of small-fiber neuropathy (SFN) is hampered by its subjective symptoms and signs. Confirmatory testing is insufficiently available and expensive, so predictive examinations have value. However, few support the 2020 SFN consensus-case-definition requirements or were validated for non-diabetes neuropathies. Thus we developed the Massachusetts General Hospital Neuropathy Exam Tool (MAGNET) and measured diagnostic performance in 160 symptomatic patients evaluated for length-dependent SFN from any cause and 37 healthy volunteers. METHODS We compared prevalences of abnormalities (vital signs, pupil responses, lower-limb appearance, pin, light touch, vibration and position sensitivity, great-toe strength, muscle stretch reflexes), and validated diagnostic performance against objective SFN tests: lower-leg skin-biopsy epidermal neurite densities and autonomic function testing (AFT). Sensitivity/specificity, feasibility, test-retest and inter-rater reliability, and convergence with the Utah Early Neuropathy Scale were calculated. RESULTS Patients' ages averaged 48.5 ± 14.7 years and 70.6% were female. Causes of neuropathy varied, remaining unknown in 59.5%. Among the 46 with abnormal skin biopsies, the most prevalent abnormality was reduced pin sharpness at the toes (71.7%). Inter-rater reliability, test-retest reliability, and convergent validity excelled (range = 91.3-95.6%). Receiver operating characteristics comparing all symptomatic patients versus healthy controls indicated that a MAGNET threshold score of 14 maximized predictive accuracy for skin biopsies (0.74) and a 30 cut-off maximized accuracy for predicting AFT (0.60). Analyzing patients with any abnormal neuropathy-test results identified areas-under-the-curves of 0.87-0.89 for predicting a diagnostic result, accuracy = 0.80-0.89, and Youden's index = 0.62. Overall, MAGNET was 80%-85% accurate for stratifying patients with abnormal versus normal neuropathy test results. DISCUSSION MAGNET quickly generates research-quality metrics during clinical examinations.
Collapse
Affiliation(s)
- Gary R. Zirpoli
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Khosro Farhad
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Madeleine C. Klein
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean Downs
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Max M. Klein
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anne Louise Oaklander
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology (Neuropathology), Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
16
|
Geerts M, Hoeijmakers JG, van Eijk-Hustings Y, Brandts L, Gorissen-Brouwers CM, Merkies IS, Joore MA, Faber CG. Cost of illness of patients with small fiber neuropathy in the Netherlands. Pain 2024; 165:153-163. [PMID: 37556388 PMCID: PMC10723644 DOI: 10.1097/j.pain.0000000000003008] [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: 02/09/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 08/11/2023]
Abstract
ABSTRACT Neuropathic pain is associated with substantial healthcare costs. However, cost-of-illness studies of small fiber neuropathy (SFN) are scarce. Our aim was to estimate the healthcare, patient and family, and productivity costs of patients with SFN in the Netherlands from a healthcare and societal perspective. In addition, the association of costs with age, pain impact on daily life, anxiety, depression, and quality of life (Qol) were examined. Cost questionnaires were completed by 156 patients with confirmed SFN. The average annual total health care and societal cost (€, 2020) was calculated at patient, SFN adult population, and societal level. The average annual healthcare, patient and family, and productivity costs per patient with a Pain Impact Numerical Rating Scale of 0 to 3 (mild), 4 to 6 (moderate), and 7 to 10 (severe) were calculated by using the cost questionnaire data. Quality of life was determined by the EuroQol 5D utility scores. Anxiety and depression were assessed using the Hospital Anxiety and Depression Scale. Associations of all costs were analyzed using linear regression analyses. At the patient level, the average annual SFN healthcare and societal cost of SFN was €3614 (95% confidence interval [CI] €3171-€4072) and €17,871 (95% CI €14,395-€21,480). At the SFN population level, the average healthcare costs were €29.8 (CI €26.4-€34.2) million, and on a societal level, these were €147.7 (CI 120.5-176.3) million. Severe pain was associated with significant lower Qol and higher depression scores, higher healthcare, patient and family, and productivity costs ( P < 0.001).
Collapse
Affiliation(s)
- Margot Geerts
- Department of Neurology, Maastricht University Medical Center+, School of Mental Health and Neuroscience, Maastricht, the Netherlands
| | - Janneke G.J. Hoeijmakers
- Department of Neurology, Maastricht University Medical Center+, School of Mental Health and Neuroscience, Maastricht, the Netherlands
| | - Yvonne van Eijk-Hustings
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, CAPHRI Care and Public Health Research Institute, Maastricht, the Netherlands
| | - Lloyd Brandts
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, CAPHRI Care and Public Health Research Institute, Maastricht, the Netherlands
| | - Carla M.L. Gorissen-Brouwers
- Department of Neurology, Maastricht University Medical Center+, School of Mental Health and Neuroscience, Maastricht, the Netherlands
| | - Ingemar S.J. Merkies
- Department of Neurology, Maastricht University Medical Center+, School of Mental Health and Neuroscience, Maastricht, the Netherlands
- Department of Neurology, Curacao Medical Center, J. H. J. Hamelbergweg, Willemstad, Curacao
| | - Manuela A. Joore
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, CAPHRI Care and Public Health Research Institute, Maastricht, the Netherlands
| | - Catharina G. Faber
- Department of Neurology, Maastricht University Medical Center+, School of Mental Health and Neuroscience, Maastricht, the Netherlands
| |
Collapse
|
17
|
Silsby M, Feldman EL, Dortch RD, Roth A, Haroutounian S, Rajabally YA, Vucic S, Shy ME, Oaklander AL, Simon NG. Advances in diagnosis and management of distal sensory polyneuropathies. J Neurol Neurosurg Psychiatry 2023; 94:1025-1039. [PMID: 36997315 PMCID: PMC10544692 DOI: 10.1136/jnnp-2021-328489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/23/2023] [Indexed: 04/01/2023]
Abstract
Distal sensory polyneuropathy (DSP) is characterised by length-dependent, sensory-predominant symptoms and signs, including potentially disabling symmetric chronic pain, tingling and poor balance. Some patients also have or develop dysautonomia or motor involvement depending on whether large myelinated or small fibres are predominantly affected. Although highly prevalent, diagnosis and management can be challenging. While classic diabetes and toxic causes are well-recognised, there are increasingly diverse associations, including with dysimmune, rheumatological and neurodegenerative conditions. Approximately half of cases are initially considered idiopathic despite thorough evaluation, but often, the causes emerge later as new symptoms develop or testing advances, for instance with genetic approaches. Improving and standardising DSP metrics, as already accomplished for motor neuropathies, would permit in-clinic longitudinal tracking of natural history and treatment responses. Standardising phenotyping could advance research and facilitate trials of potential therapies, which lag so far. This review updates on recent advances and summarises current evidence for specific treatments.
Collapse
Affiliation(s)
- Matthew Silsby
- Neurology, Westmead Hospital, Westmead, New South Wales, Australia
- Brain and Nerve Research Centre, Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard D Dortch
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Institute of Imaging Science, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University Institute of Imaging Science, Nashville, Tennessee, USA
| | - Alison Roth
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University School of Medicine in Saint Louis, St Louis, Missouri, USA
| | - Yusuf A Rajabally
- Inflammatory Neuropathy Clinic, Department of Neurology, University Hospitals Birmingham, Aston Medical School, Aston University, Birmingham, UK
| | - Steve Vucic
- Brain and Nerve Research Centre, Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Anne Louise Oaklander
- Nerve Unit, Departments of Neurology and Pathology (Neuropathology), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Neil G Simon
- Northern Beaches Clinical School, Macquarie University, Frenchs Forest, New South Wales, Australia
| |
Collapse
|
18
|
Kyle K, Hutto SK, Reda H, Zonozi R, Farhad K, Jeyabalan A, Chwalisz BK. Small fiber neuropathy associated with ANCA positivity: a case series and brief literature review. Neurol Sci 2023; 44:4473-4479. [PMID: 37453951 DOI: 10.1007/s10072-023-06954-y] [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: 03/02/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Small fiber neuropathy [SFN] is a common peripheral neurologic disorder with a vast array of implicated etiologies. It has previously been proposed that some forms of immune-mediated small fiber neuropathy are driven by vasculitis, though antinuclear cytoplasmic antibodies [ANCA] antibodies have not commonly been reported in association with SFN, thus far. We present this case series to discuss the observation of a possible novel association between ANCA and SFN. METHODS This is a retrospective case series of 6 patients with SFN and ANCA positivity, with and without systemic manifestations. Patients included were diagnosed with SFN by skin biopsy or autonomic function testing and were seropositive for ANCA by ELISA. RESULTS Six patients are outlined, including 4 females and 2 males. Antigen specific antibodies were MPO alone in 4 cases, PR3 alone in 1 case and both MPO and PR3 in 1 case. Systemic vasculitis was noted in 2 patients. Five patients received immunosuppression. Three patients experienced partial improvement, while symptoms stabilized in 3 patients. DISCUSSION This is the first series of patients with suspected immune-mediated SFN and ANCA antibody positivity, raising the possibility of ANCA mediated isolated SFN. This is in contradistinction to the more typical ANCA-mediated peripheral neuropathy manifestations of mononeuropathy multiplex or axonal sensorimotor neuropathy. We cannot unequivocally prove ANCA-associated vasculitis [AAV] causality in these cases; however, the stabilization in SFN symptomatology and associated improvement in ANCA antibody titer, after AAV treatment, may be indicative of an association.
Collapse
Affiliation(s)
- Kevin Kyle
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
| | - Spencer K Hutto
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Haatem Reda
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Reza Zonozi
- Vasculitis and Glomerulonephritis Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Khosro Farhad
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Anushya Jeyabalan
- Vasculitis and Glomerulonephritis Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Bart K Chwalisz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| |
Collapse
|
19
|
Faber CG, Attal N, Lauria G, Dworkin RH, Freeman R, Dawson KT, Finnigan H, Hajihosseini A, Naik H, Serenko M, Morris CJ, Kotecha M. Efficacy and safety of vixotrigine in idiopathic or diabetes-associated painful small fibre neuropathy (CONVEY): a phase 2 placebo-controlled enriched-enrolment randomised withdrawal study. EClinicalMedicine 2023; 59:101971. [PMID: 37152360 PMCID: PMC10154969 DOI: 10.1016/j.eclinm.2023.101971] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Background No pharmacological treatments are specifically indicated for painful small fibre neuropathy (SFN). CONVEY, a phase 2 enriched-enrolment study, evaluated the efficacy and safety of vixotrigine, a voltage- and use-dependent sodium channel blocker, in participants with idiopathic or diabetes-associated painful SFN. Methods CONVEY was a phase 2, multicentre, placebo-controlled, double-blind (DB), enriched-enrolment, randomised withdrawal study. The study was conducted at 68 sites in 13 countries (Europe and Canada) between May 17, 2018, and April 12, 2021. Following a 4-week open-label period in which 265 adults with painful SFN (a mixture of large and small fibre neuropathy was not exclusionary) received oral vixotrigine 350 mg twice daily (BID), 123 participants (with a ≥30% reduction from baseline in average daily pain [ADP] score during the open-label period) were randomised 1:1:1 to receive 200 mg BID, 350 mg BID or placebo for a 12-week double-blind (DB) period. Primary endpoint was change from baseline in ADP at DB Week 12. Secondary endpoints included the proportion of participants with a ≥30% reduction from baseline in ADP and the proportion of Patient Global Impression of Pain (PGIC) responders at DB Week 12. Treatment-emergent adverse events (AEs) were monitored. Statistical significance was set at 0.10 (2-sided). The trial was registered on ClinicalTrials.gov (NCT03339336) and on ClinicalTrialsregister.eu (2017-000991-27). Findings A statistically significant difference from placebo in least squares mean reduction in ADP score from baseline to DB Week 12 was observed with vixotrigine 200 mg BID (-0.85; SE, 0.43; 95% CI, -1.71 to 0.00; p = 0.050) but not 350 mg BID (-0.17; SE, 0.43; 95% CI, -1.01 to 0.68; p = 0.70). Numerically, but not statistically significantly, more participants who received vixotrigine vs placebo experienced a ≥30% ADP reduction from baseline (68.3-72.5% vs 52.5%), and only the 350 mg BID group had significantly more PGIC responders vs placebo (48.8% vs 30.0%; odds ratio = 2.60; 95% CI, 0.97-6.99; p = 0.058) at DB Week 12. AEs were mostly mild to moderate in the vixotrigine groups. The most common AEs (≥5% of vixotrigine-treated participants) in the DB 200 mg BID and 350 mg BID vixotrigine groups were falls, nasopharyngitis, muscle spasm, and urinary tract infection. Interpretation In our study, vixotrigine 200 mg BID, but not 350 mg BID, met the primary endpoint; more vixotrigine-treated participants experienced a ≥30% reduction from baseline in ADP at DB Week 12. Vixotrigine (at both dosages) was well tolerated in participants with SFN. Funding Biogen, Inc.
Collapse
Affiliation(s)
- Catharina G. Faber
- Department of Neurology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, P. Debeyelaan 25, Maastricht 6229 HX, the Netherlands
| | - Nadine Attal
- INSERM U987, Ambroise Paré Hospital, APHP, Boulogne-Billancourt F-92100, France
- Université UVSQ Paris-Saclay, Versailles 78000, France
| | - Giuseppe Lauria
- Third Neurology Unit, Fondazione IRCCS Istituto Neurologico “Carlo Besta”, Via Celoria 11, Milan 20133, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Festa del Perdono 7, Milan 20122, Italy
| | - Robert H. Dworkin
- Department of Anesthesiology and Perioperative Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Roy Freeman
- Department of Neurology, Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | - Mona Kotecha
- Biogen, Cambridge, MA 02142, USA
- Corresponding author.
| |
Collapse
|
20
|
Geerts M, Hoeijmakers JG, Gorissen-Brouwers CM, Faber CG, Merkies IS. Small Fiber Neuropathy: A Clinical and Practical Approach. J Nurse Pract 2023; 19:104547. [DOI: 10.1016/j.nurpra.2023.104547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
21
|
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
|
22
|
Seeliger T, Dreyer HN, Siemer JM, Bönig L, Gingele S, Dohrn MF, Prenzler N, Ernst D, Witte T, Skripuletz T. Clinical and paraclinical features of small fiber neuropathy in Sjögren's syndrome. J Neurol 2023; 270:1004-1010. [PMID: 36331613 PMCID: PMC9886580 DOI: 10.1007/s00415-022-11431-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Sjögren's syndrome is a potentially treatable cause of Small Fiber Neuropathy (SFN)-a condition that severely affects patients' quality of life. We therefore aimed to characterize patients with SFN and Sjögren's syndrome to raise awareness of this disease and facilitate its early recognition as an essential step for appropriate treatment. In 97 SFN patients (median age 48 years, 77% female), we studied the clinical features associated with Sjögren's syndrome compared to the idiopathic SFN subtype. According to the current ACR/EULAR classification criteria (Shiboski et al., Ann Rheum Dis 76:9-16, 2017), 24/97 individuals (25%, median age 48.5 years, 75% female) were diagnosed with Sjögren's syndrome. We did not observe any differences in SFN-defining sensory plus symptoms. Furthermore, intraepidermal nerve fiber densities (IENFD) were significantly lower in patients with SFN and Sjögren's syndrome (mean 2.6 ± 1.2/mm) compared to patients with idiopathic SFN (mean 3.2 ± 1.5/mm; p = 0.048). There were no significant group differences when analyzing cerebrospinal fluid (CSF) parameters. We conclude that Sjögren's syndrome-associated SFN is difficult to distinguish from idiopathic forms based on initial clinical symptoms and CSF results. However, lower IENFD values in patients with Sjögren's syndrome-associated SFN might indicate a distinct different pathomechanism in this entity compared to idiopathic SFN.
Collapse
Affiliation(s)
- Tabea Seeliger
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Henrike Neelke Dreyer
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Janna Margaretha Siemer
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Lena Bönig
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Nils Prenzler
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Diana Ernst
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
23
|
Metwally NAE, Hasan MM, Abd Elaziz AES, Elhadad AF, Ibraheem KS, Ali MAA, Elsalam HAA. Assessment of intraepidermal nerve fiber density and neurophysiological studies in patients with idiopathic polyneuropathy. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2022. [DOI: 10.1186/s41983-022-00548-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Abstract
Background
Idiopathic polyneuropathy is an asymmetrical, length-dependent neuropathy in which neurophysiology demonstrates axonal damage involving large fibers, along with insidious onset and slow progression over 6 months, with no identified etiology in spite of thorough investigations. This study aimed to evaluate the diagnostic role of clinical, electrophysiological, and histopathological studies in patients with idiopathic polyneuropathy.
Methods
Case–control study included 20 patients with clinical and neurophysiological evidence of sensory or sensory–motor neuropathy with no apparent etiology after laboratory investigation were recruited from 127 patients with sensory–motor neuropathy of unknown etiology (the patients group). Twenty apparently healthy individuals, age- and sex-matched, with no neuropathy symptoms (the control group), were recruited from the Neurology Clinic of Al-Azhar University, Assuit.
Results
Age of onset of patients with idiopathic polyneuropathy (44–70) years, duration of illness (1–6) years, 60% had painful neuropathy, diagnostic neuropathic pain questioner (DN4 score) (5–7), abnormal pin brick (80%), abnormal vibration (90%), abnormal fine touch (75%), distal weakness (70%), and lost ankle reflex (90%). In the control group, there were substantial differences with respect to prolonged latency, diminished sympathetic skin response amplitude, and significant intraepidermal nerve fiber density reduction in skin biopsy cases. In diagnosing idiopathic polyneuropathy, the specificity and sensitivity of sympathetic skin response were (80–86)% and (81–89.5)%, respectively, whereas those of diminished intraepidermal nerve fiber density were (92.5%) and (97.5%), respectively.
Conclusion
The assessment of intraepidermal nerve fiber density had an important good diagnostic role in cases presented with polyneuropathy.
Collapse
|
24
|
Tsai NW, Lin CC, Yeh TY, Chiu YA, Chiu HH, Huang HP, Hsieh ST. An induced pluripotent stem cell-based model identifies molecular targets of vincristine neurotoxicity. Dis Model Mech 2022; 15:dmm049471. [PMID: 36518084 PMCID: PMC10655812 DOI: 10.1242/dmm.049471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2023] Open
Abstract
To model peripheral nerve degeneration and investigate molecular mechanisms of neurodegeneration, we established a cell system of induced pluripotent stem cell (iPSC)-derived sensory neurons exposed to vincristine, a drug that frequently causes chemotherapy-induced peripheral neuropathy. Sensory neurons differentiated from iPSCs exhibit distinct neurochemical patterns according to the immunocytochemical phenotypes, and gene expression of peripherin (PRPH, hereafter referred to as Peri) and neurofilament heavy chain (NEFH, hereafter referred to as NF). The majority of iPSC-derived sensory neurons were PRPH positive/NEFH negative, i.e. Peri(+)/NF(-) neurons, whose somata were smaller than those of Peri(+)/NF(+) neurons. On exposure to vincristine, projections from the cell body of a neuron, i.e. neurites, were degenerated quicker than somata, the lethal concentration to kill 50% (LC50) of neurites being below the LC50 for somata, consistent with the clinical pattern of length-dependent neuropathy. We then examined the molecular expression in the MAP kinase signaling pathways of, extracellular signal-regulated kinases 1/2 (MAPK1/3, hereafter referred to as ERK), p38 mitogen-activated protein kinases (MAPK11/12/13/14, hereafter referred to as p38) and c-Jun N-terminal kinases (MAPK8/9/10, hereafter referred to as JNK). Regarding these three cascades, only phosphorylation of JNK was upregulated but not that of p38 or ERK1/2. Furthermore, vincristine-treatment resulted in impaired autophagy and reduced autophagic flux. Rapamycin-treatment reversed the effect of impaired autophagy and JNK activation. These results not only established a platform to study peripheral degeneration of human neurons but also provide molecular mechanisms for neurodegeneration with the potential for therapeutic targets.
Collapse
Affiliation(s)
- Neng-Wei Tsai
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Cheng-Chen Lin
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Ti-Yen Yeh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Yu-An Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsin-Hui Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsiang-Po Huang
- Department of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei 100, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
25
|
Abstract
Distal symmetric diabetic peripheral polyneuropathy (DPN) is the most common form of neuropathy in the world, affecting 30 to 50% of diabetic individuals and resulting in significant morbidity and socioeconomic costs. This review summarizes updates in the diagnosis and management of DPN. Recently updated clinical criteria facilitate bedside diagnosis, and a number of new technologies are being explored for diagnostic confirmation in specific settings and for use as surrogate measures in clinical trials. Evolving literature indicates that distinct but overlapping mechanisms underlie neuropathy in type 1 versus type 2 diabetes, and there is a growing focus on the role of metabolic factors in the development and progression of DPN. Exercise-based lifestyle interventions have shown therapeutic promise. A variety of potential disease-modifying and symptomatic therapies are in development. Innovations in clinical trial design include the incorporation of detailed pain phenotyping and biomarkers for central sensitization.
Collapse
Affiliation(s)
- Qihua Fan
- Department of Neurology, Division of Neuromuscular Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - A Gordon Smith
- Department of Neurology, Division of Neuromuscular Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
26
|
Hoeijmakers JGJ, Merkies ISJ, Faber CG. Small fiber neuropathies: expanding their etiologies. Curr Opin Neurol 2022; 35:545-552. [PMID: 35950732 DOI: 10.1097/wco.0000000000001103] [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: 11/25/2022]
Abstract
PURPOSE OF REVIEW Several conditions have been associated with the development of small fiber neuropathy (SFN). The list of metabolic, immune-mediated, infectious, toxic, drugs-related, and hereditary conditions is still growing and various hypotheses are made about the underlying pathophysiological mechanisms. Understanding these processes is important to provide new targets for treatment. In addition, the specific SFN phenotype can provide direction for the underlying etiology. This review discusses the latest developments concerning the expanding etiologies in SFN. RECENT FINDINGS In the past 18 months, special attention has been paid to immunological etiologies, partly due to the coronavirus disease 2019 pandemic, but also new auto-antibodies in SFN have been demonstrated. Identifying patients with immune-mediated SFN can be challenging, since contrary to the classical distal sensory phenotype, a nonlength-dependent pattern is more common.Besides the etiologies of classical SFN, small fiber pathology is increasingly described in diseases without the typical neuropathic pain features of SFN, sometimes called syndromic SFN. However, the clinical relevance is not yet fully understood. SUMMARY The expansion of the etiologies of SFN continues and brings more insight in possible targets for treatment. The clinical presentation may vary as a result of the underlying condition.
Collapse
Affiliation(s)
- Janneke G J Hoeijmakers
- Department of Neurology, Maastricht University Medical Center+, Maastricht
- MHeNS, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Ingemar S J Merkies
- Department of Neurology, Maastricht University Medical Center+, Maastricht
- MHeNS, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
- Curaçao Medical Center, Willemstad, Curaçao
| | - Catharina G Faber
- Department of Neurology, Maastricht University Medical Center+, Maastricht
- MHeNS, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| |
Collapse
|
27
|
Dohrn MF, Dumke C, Hornemann T, Nikolin S, Lampert A, Espenkott V, Vollert J, Ouwenbroek A, Zanella M, Schulz JB, Gess B, Rolke R. Deoxy-sphingolipids, oxidative stress, and vitamin C correlate with qualitative and quantitative patterns of small fiber dysfunction and degeneration. Pain 2022; 163:1800-1811. [PMID: 35239546 PMCID: PMC9393801 DOI: 10.1097/j.pain.0000000000002580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/05/2022]
Abstract
ABSTRACT Defined by dysfunction or degeneration of Aδ and C fibers, small fiber neuropathies (SFNs) entail a relevant health burden. In 50% of cases, the underlying cause cannot be identified or treated. In 100 individuals (70% female individuals; mean age: 44.8 years) with an idiopathic, skin biopsy-confirmed SFN, we characterized the symptomatic spectrum and measured markers of oxidative stress (vitamin C, selenium, and glutathione) and inflammation (transforming growth factor beta, tumor necrosis factor alpha), as well as neurotoxic 1-deoxy-sphingolipids. Neuropathic pain was the most abundant symptom (95%) and cause of daily life impairment (72%). Despite the common use of pain killers (64%), the painDETECT questionnaire revealed scores above 13 points in 80% of patients. In the quantitative sensory testing (QST), a dysfunction of Aδ fibers was observed in 70% and of C fibers in 44%, affecting the face, hands, or feet. Despite normal nerve conduction studies, QST revealed Aβ fiber involvement in 46% of patients' test areas. Despite absence of diabetes mellitus or mutations in SPTLC1 or SPTLC2 , plasma 1-deoxy-sphingolipids were significantly higher in the sensory loss patient cluster when compared with those in patients with thermal hyperalgesia ( P < 0.01) or those in the healthy category ( P < 0.1), correlating inversely with the intraepidermal nerve fiber density (1-deoxy-SA: P < 0.05, 1-deoxy-SO: P < 0.01). Patients with arterial hypertension, overweight (body mass index > 25 kg/m 2 ), or hyperlipidemia showed significantly lower L-serine (arterial hypertension: P < 0.01) and higher 1-deoxy-sphingolipid levels (arterial hypertension: P < 0.001, overweight: P < 0.001, hyperlipidemia: P < 0.01). Lower vitamin C levels correlated with functional Aβ involvement ( P < 0.05). Reduced glutathione was lower in patients with Aδ dysfunction ( P < 0.05). Idiopathic SFNs are heterogeneous. As a new pathomechanism, plasma 1-deoxy-sphingolipids might link the metabolic syndrome with small fiber degeneration.
Collapse
Affiliation(s)
- Maike F. Dohrn
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Christina Dumke
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zürich, Zurich, Switzerland
| | - Stefan Nikolin
- Institute of Neuropathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Volker Espenkott
- Department of Palliative Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jan Vollert
- Pain Research, Department of Surgery and Cancer (MSK), Imperial College London, London, United Kingdom
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
- Neurophysiology, Mannheim Center of Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Annabelle Ouwenbroek
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Martina Zanella
- Institute of Clinical Chemistry, University Hospital Zürich, Zurich, Switzerland
| | - Jörg B. Schulz
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Burkhard Gess
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Roman Rolke
- Department of Palliative Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
28
|
Safavi F, Gustafson L, Walitt B, Lehky T, Dehbashi S, Wiebold A, Mina Y, Shin S, Pan B, Polydefkis M, Oaklander AL, Nath A. Neuropathic symptoms with SARS-CoV-2 vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.05.16.22274439. [PMID: 35611338 PMCID: PMC9128783 DOI: 10.1101/2022.05.16.22274439] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Background and Objectives Various peripheral neuropathies, particularly those with sensory and autonomic dysfunction may occur during or shortly after acute COVID-19 illnesses. These appear most likely to reflect immune dysregulation. If similar manifestations can occur with the vaccination remains unknown. Results In an observational study, we studied 23 patients (92% female; median age 40years) reporting new neuropathic symptoms beginning within 1 month after SARS-CoV-2 vaccination. 100% reported sensory symptoms comprising severe face and/or limb paresthesias, and 61% had orthostasis, heat intolerance and palpitations. Autonomic testing in 12 identified seven with reduced distal sweat production and six with positional orthostatic tachycardia syndrome. Among 16 with lower-leg skin biopsies, 31% had diagnostic/subthreshold epidermal neurite densities (≤5%), 13% were borderline (5.01-10%) and 19% showed abnormal axonal swelling. Biopsies from randomly selected five patients that were evaluated for immune complexes showed deposition of complement C4d in endothelial cells. Electrodiagnostic test results were normal in 94% (16/17). Together, 52% (12/23) of patients had objective evidence of small-fiber peripheral neuropathy. 58% patients (7/12) treated with oral corticosteroids had complete or near-complete improvement after two weeks as compared to 9% (1/11) of patients who did not receive immunotherapy having full recovery at 12 weeks. At 5-9 months post-symptom onset, 3 non-recovering patients received intravenous immunoglobulin with symptom resolution within two weeks. Conclusions This observational study suggests that a variety of neuropathic symptoms may manifest after SARS-CoV-2 vaccinations and in some patients might be an immune-mediated process.
Collapse
Affiliation(s)
- Farinaz Safavi
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Lindsey Gustafson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Brian Walitt
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Tanya Lehky
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Sara Dehbashi
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA
| | - Amanda Wiebold
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Yair Mina
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Susan Shin
- Department of Neurology, Icahn School of Medicine at Mt Sinai, New York, NY
| | - Baohan Pan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michael Polydefkis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anne Louise Oaklander
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA
- Department of Pathology (Neuropathology), Massachusetts General Hospital, Boston, MA
| | - Avindra Nath
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| |
Collapse
|
29
|
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
|
30
|
Petropoulos IN, Bitirgen G, Ferdousi M, Kalteniece A, Azmi S, D'Onofrio L, Lim SH, Ponirakis G, Khan A, Gad H, Mohammed I, Mohammadi YE, Malik A, Gosal D, Kobylecki C, Silverdale M, Soran H, Alam U, Malik RA. Corneal Confocal Microscopy to Image Small Nerve Fiber Degeneration: Ophthalmology Meets Neurology. FRONTIERS IN PAIN RESEARCH 2022; 2:725363. [PMID: 35295436 PMCID: PMC8915697 DOI: 10.3389/fpain.2021.725363] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
Neuropathic pain has multiple etiologies, but a major feature is small fiber dysfunction or damage. Corneal confocal microscopy (CCM) is a rapid non-invasive ophthalmic imaging technique that can image small nerve fibers in the cornea and has been utilized to show small nerve fiber loss in patients with diabetic and other neuropathies. CCM has comparable diagnostic utility to intraepidermal nerve fiber density for diabetic neuropathy, fibromyalgia and amyloid neuropathy and predicts the development of diabetic neuropathy. Moreover, in clinical intervention trials of patients with diabetic and sarcoid neuropathy, corneal nerve regeneration occurs early and precedes an improvement in symptoms and neurophysiology. Corneal nerve fiber loss also occurs and is associated with disease progression in multiple sclerosis, Parkinson's disease and dementia. We conclude that corneal confocal microscopy has good diagnostic and prognostic capability and fulfills the FDA criteria as a surrogate end point for clinical trials in peripheral and central neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Gulfidan Bitirgen
- Department of Ophthalmology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Maryam Ferdousi
- Faculty of Biology, Medicine and Health, University of Manchester, Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Alise Kalteniece
- Faculty of Biology, Medicine and Health, University of Manchester, Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Shazli Azmi
- Faculty of Biology, Medicine and Health, University of Manchester, Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom.,Centre for Diabetes, Endocrinology and Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Luca D'Onofrio
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Sze Hway Lim
- Department of Neurology, Salford Royal National Health System (NHS) Foundation Trust, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | | | - Adnan Khan
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Hoda Gad
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ibrahim Mohammed
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | - Ayesha Malik
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - David Gosal
- Department of Neurology, Salford Royal National Health System (NHS) Foundation Trust, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Christopher Kobylecki
- Department of Neurology, Salford Royal National Health System (NHS) Foundation Trust, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Monty Silverdale
- Department of Neurology, Salford Royal National Health System (NHS) Foundation Trust, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Handrean Soran
- Faculty of Biology, Medicine and Health, University of Manchester, Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Uazman Alam
- Department of Cardiovascular and Metabolic Medicine, Clinical Sciences Centre, Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool University Hospital National Health System (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Rayaz A Malik
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar.,Faculty of Biology, Medicine and Health, University of Manchester, Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
31
|
Campolo M, Correa L, Gabarrón E, Albayrak M, Quintero-Diaz C, Castellote JM, Casanova-Molla J, Valls-Sole J. Adaptation to tonic heat in healthy subjects and patients with sensory polyneuropathy. Eur J Pain 2022; 26:1056-1068. [PMID: 35263818 DOI: 10.1002/ejp.1930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 02/10/2022] [Accepted: 02/27/2022] [Indexed: 12/11/2022]
Abstract
Adaptation to a constant sensory stimulus involves many sites along the path of sensory volleys towards perception. The evaluation of such phenomenon may be of clinical interest. We studied adaptation to a constant temperature stimulus in healthy subjects to set normative data, and in patients with sensory polyneuropathy (SPN), as proof of concept. Twenty-six healthy subjects and 26 patients with SPN in the context of chemotherapy treatment with oxaliplatin for colon cancer were instructed to express through an electronic VAS system (eVAS) the level of sensation felt when a thermode set at either 39º, 41º, 43º, 45º or 47º was applied to their ventral forearm. The eVAS recordings showed typically an abrupt onset that slowed to approach maximum sensation and continued with a slow decrease indicating adaptation. The time to respond (TR), the velocity of the initial response (VR), the maximum sensation (MA), the time to reach MA (MAt), the onset of adaptation (AO), and the decrease in the sensation level with respect to MA at 30 s after stimulus application (SL30), were dependent on the temperature level in all subjects. However, patients showed significantly delayed TR, slowed VR, decreased MA, delayed AO, and reduced SL30, with respect to healthy subjects. Differences were more pronounced at low temperature levels, with absent AO in 25 patients vs. 2 healthy subjects at temperatures of 39º and 41ºC. The study of adaptation to a constant temperature stimulus can furnish valuable data for the assessment of SPN patients.
Collapse
Affiliation(s)
- Michela Campolo
- EMG and Neuropathic Pain Unit, Department of Neurology. Hospital Clínic, Barcelona
| | - Lilia Correa
- Department of Neurology and Neurophysiology. Hospital del Mar, Barcelona
| | - Eva Gabarrón
- EMG and Neuropathic Pain Unit, Department of Neurology. Hospital Clínic, Barcelona
| | - Merve Albayrak
- EMG and Neuropathic Pain Unit, Department of Neurology. Hospital Clínic, Barcelona
| | | | - Juan M Castellote
- Department of Radiology, Rehabilitation and Physiotherapy, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Jordi Casanova-Molla
- EMG and Neuropathic Pain Unit, Department of Neurology. Hospital Clínic, Barcelona.,Institut d'Investigació Biomedica August Pi Sunyer, IDIBAPS. Barcelona.,Department of Medicine, School of Medicine and Health Sciences, University of Barcelona
| | - Josep Valls-Sole
- Institut d'Investigació Biomedica August Pi Sunyer, IDIBAPS. Barcelona.,Department of Medicine, School of Medicine and Health Sciences, University of Barcelona
| |
Collapse
|
32
|
Malik RA, Efron N. Corneal Confocal Microscopy and the Nervous System: Introduction to the Special Issue. J Clin Med 2022; 11:jcm11061475. [PMID: 35329801 PMCID: PMC8953792 DOI: 10.3390/jcm11061475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 12/07/2022] Open
Affiliation(s)
- Rayaz A. Malik
- Weill Cornell Medicine-Qatar, Research Division, Qatar Foundation, Education City, Doha 24144, Qatar;
| | - Nathan Efron
- School of Optometry and Vision Science, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
- Correspondence:
| |
Collapse
|
33
|
Sopacua M, Gorissen-Brouwers CM, de Greef BT, Joosten IB, Faber CG, Merkies IS, Hoeijmakers JG. The applicability of the digit wrinkle scan to quantify sympathetic nerve function. Clin Neurophysiol Pract 2022; 7:115-119. [PMID: 35434427 PMCID: PMC9006743 DOI: 10.1016/j.cnp.2022.03.005] [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: 01/21/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 11/23/2022] Open
Abstract
Normative values for stimulated skin wrinkling are age-dependent. Stimulated skin wrinkling has never been evaluated quantitatively. The clinical application of the stimulated skin wrinkling in an ordinal fashion is doubtful.
Objective Stimulated skin wrinkling test (SSW) has been launched as a non-invasive diagnostic procedure. However, no normative age dependent values have been reported that can be applied in clinical practice. The objectives of the study were to (1) collect age-dependent normative values according to the 5-point scale assessment for the SSW, to (2) determine reliability scores for the obtained norm values, and to (3) introduce a new digital method for SSW assessment, the Digit Wrinkle Scan© (DWS©) for detection of wrinkles in a more quantitative manner. Methods Firstly, 82 healthy participants were included, divided in 5 age groups. The participants underwent SSW using lidocaine and prilocaine topical cream. Secondly, 35 healthy participants were included to test whether the DWS© could be a novel manner to assess the grade of wrinkling quantitatively. We determined the inter-observer reliability of both methods. Also, the intra-observer reliability was calculated for the DWS©. Results We found a decrease in normative values over age. The inter-observer reliability of assessment by the 5-point scale method was moderate after SSW (Cohen’s k: 0.53). Results of the DWS© indicate that total wrinkle length per mm2 showed moderate to good agreement for the 4th and 5th digits after SSW, and a low agreement for the other digits. Conclusions Age-dependent normative values were obtained according to the 5-point scale, but its clinical application is doubtful since we found a moderate inter-observer reliability. We introduced the DWS© as a possible new method in order to quantify the grade of wrinkling. Significance We found unsatisfactory reliability scores, which hampers its usefulness for clinical practice.
Collapse
|
34
|
Ahmed N, Vigouroux M, Ingelmo P. Implications of Nerve Fiber Density on the Diagnosis and Treatment of Juvenile Fibromyalgia. J Pain Res 2022; 15:513-520. [PMID: 35210850 PMCID: PMC8860391 DOI: 10.2147/jpr.s340038] [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: 09/25/2021] [Accepted: 01/28/2022] [Indexed: 11/23/2022] Open
Abstract
Juvenile fibromyalgia (JFM) is a condition that presents as chronic widespread musculoskeletal pain and affects children and adolescents. JFM remains a challenging diagnosis, as it is both based on subjective criteria and the pathogenesis is poorly understood. Small fiber neuropathy (SFN) is a distinct condition, which is characterized by pathology of small A-delta and C fibers, and can present similarly to JFM. Small fiber pathology is characterized by reduced intraepidermal nerve fiber density (IENFD) on skin biopsy. Recent studies have found that as many as half of patients with JFM can demonstrate decreased IENFD, in pattern similar to SFN. This phenomenon has been referred to as small fiber pathology. The meaning of these findings was disputed; however, the current consensus remains that fibromyalgia and SFN are distinct conditions. Additionally, among patients with fibromyalgia, there are two phenotypes: those with small fiber pathology and those without. The purpose of this review was to characterize the role assessment of IENFD plays in the clinical context. We conducted a narrative review of pertinent articles pertaining to JFM, SFN and small fiber pathology in fibromyalgia. We concluded that assessment of IENFD should be completed if SFN is suspected either when a patient first presents or in patients who were previously diagnosed with fibromyalgia and SFN is later suspected. Distinguishing between JFM and SFN is important because recommended therapies differ between the two conditions. However, there is no evidence to support the use of skin biopsy to distinguish between the two discussed fibromyalgia phenotypes. More studies are needed to elucidate whether IENFD varies with morbidity and if both fibromyalgia phenotypes vary in their response to different therapeutic regimens.
Collapse
Affiliation(s)
- Nabeel Ahmed
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Marie Vigouroux
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
- Edwards Family Interdisciplinary Center for Complex Pain, Montreal Children’s Hospital, Montreal, QC, Canada
- Correspondence: Marie Vigouroux, Family Interdisciplinary Center for Complex Pain, Montreal Children’s Hospital, 1001 boul. Décarie A02.3523, Montreal, QC, H4A 3J1, Canada, Tel +1 514 412 4448, Fax +1 514 412 4341, Email
| | - Pablo Ingelmo
- Edwards Family Interdisciplinary Center for Complex Pain, Montreal Children’s Hospital, Montreal, QC, Canada
- Research Institute, McGill University Health Centre, Montreal, QC, Canada
- Alan Edwards Research Center for Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
35
|
Özdağ Acarli AN, Klein T, Egenolf N, Sommer C, Üçeyler N. Subepidermal Schwann cell counts correlate with skin innervation - an exploratory study. Muscle Nerve 2022; 65:471-479. [PMID: 35020203 DOI: 10.1002/mus.27496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION/AIMS Schwann cell clusters have been described at the murine dermis-epidermis border. We quantified dermal Schwann cells in the skin of patients with small fiber neuropathy (SFN) compared to healthy controls to correlate with the clinical phenotype. METHODS Skin punch biopsies from the lower legs of 28 patients with SFN (eleven men, 17 women, median age 54 years [19-73]) and 9 healthy controls (five men, four women, median age 34 years [25-69]) were immunoreacted for S100 calcium-binding protein B as a Schwann cell marker, protein-gene product 9.5 as a pan-neuronal marker, and CD207 as a Langerhans cell marker. Intraepidermal nerve fiber density (IENFD) and subepidermal Schwann cell counts were determined. RESULTS Skin samples of patients with SFN showed lower IENFD (p<0.05), fewer Schwann cells/mm (p<0.01), and fewer Schwann cell clusters/mm (p<0.05) than controls. When comparing SFN patients with reduced (n=13, median age 53 years, 19-73 years) and normal distal (n=15, median age 54 years, 43-68 years) IENFD, the number of solitary Schwann cells/mm (p<0.01) and subepidermal nerve fibers associated with Schwann cell branches (p<0.05) were lower in patients with reduced IENFD. All three parameters positively correlated with distal IENFD (p<0.05 to p<0.01), while no correlation was found between Schwann cell counts and clinical pain characteristics. DISCUSSION Our data raise questions about the mechanisms underlying the interdependence of dermal Schwann cells and skin innervation in SFN. The temporal course and functional impact of Schwann cell presence and kinetics need further investigation.
Collapse
Affiliation(s)
| | - Thomas Klein
- Department of Neurology, University of Würzburg, Germany
| | - Nadine Egenolf
- Department of Neurology, University of Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University of Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University of Würzburg, Germany
| |
Collapse
|
36
|
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
|
37
|
Fan Q, Gordon Smith A. Recent updates in the treatment of diabetic polyneuropathy. Fac Rev 2022. [PMID: 36311537 DOI: 10.1270/r/11-30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
Distal symmetric diabetic peripheral polyneuropathy (DPN) is the most common form of neuropathy in the world, affecting 30 to 50% of diabetic individuals and resulting in significant morbidity and socioeconomic costs. This review summarizes updates in the diagnosis and management of DPN. Recently updated clinical criteria facilitate bedside diagnosis, and a number of new technologies are being explored for diagnostic confirmation in specific settings and for use as surrogate measures in clinical trials. Evolving literature indicates that distinct but overlapping mechanisms underlie neuropathy in type 1 versus type 2 diabetes, and there is a growing focus on the role of metabolic factors in the development and progression of DPN. Exercise-based lifestyle interventions have shown therapeutic promise. A variety of potential disease-modifying and symptomatic therapies are in development. Innovations in clinical trial design include the incorporation of detailed pain phenotyping and biomarkers for central sensitization.
Collapse
Affiliation(s)
- Qihua Fan
- Department of Neurology, Division of Neuromuscular Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - A Gordon Smith
- Department of Neurology, Division of Neuromuscular Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
38
|
Corneal Confocal Microscopy in the Diagnosis of Small Fiber Neuropathy: Faster, Easier, and More Efficient Than Skin Biopsy? PATHOPHYSIOLOGY 2021; 29:1-8. [PMID: 35366285 PMCID: PMC8954271 DOI: 10.3390/pathophysiology29010001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic pain may affect 30–50% of the world’s population and an important cause is small fiber neuropathy (SFN). Recent research suggests that autoimmune diseases may be one of the most common causes of small nerve fiber damage. There is low awareness of SFN among patients and clinicians and it is difficult to diagnose as routine electrophysiological methods only detect large fiber abnormalities, and specialized small fiber tests, like skin biopsy and quantitative sensory testing, are not routinely available. Corneal confocal microscopy (CCM) is a rapid, non-invasive, reproducible method for quantifying small nerve fiber degeneration and regeneration, and could be an important tool for diagnosing SFN. This review considers the advantages and disadvantages of CCM and highlights the evolution of this technique from a research tool to a diagnostic test for small fiber damage, which can be a valuable contribution to the study and management of autoimmune disease.
Collapse
|
39
|
Bitirgen G, Kucuk A, Ergun MC, Baloglu R, Gharib MH, Al Emadi S, Ponirakis G, Malik RA. Subclinical Corneal Nerve Fiber Damage and Immune Cell Activation in Systemic Lupus Erythematosus: A Corneal Confocal Microscopy Study. Transl Vis Sci Technol 2021; 10:10. [PMID: 34905000 PMCID: PMC8684301 DOI: 10.1167/tvst.10.14.10] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose The purpose of this study was to evaluate the utility of corneal confocal microscopy (CCM) in identifying small nerve fiber damage and immune cell activation in patients with systemic lupus erythematosus (SLE). Methods This cross-sectional comparative study included 39 consecutive patients with SLE and 30 healthy control participants. Central corneal sensitivity was assessed using a Cochet-Bonnet contact corneal esthesiometer and a laser scanning CCM (Heidelberg, Germany) was used to quantify corneal nerve fiber density (CNFD), nerve branch density (CNBD), nerve fiber length (CNFL), and Langerhans cell (LC) density. Results Age was comparable among patients with SLE (33.7 ± 12.7) and controls (35.0 ± 13.7 years, P = 0.670) and the median duration of disease was 3.0 years (2.0–10.0 years). CNBD (P = 0.003) and CNFL (P = 0.019) were lower and mature LC density (P = 0.002) was higher, but corneal sensitivity (P = 0.178) and CNFD (P = 0.198) were comparable in patients with SLE compared with controls. The SELENA-SLEDAI score correlated with CNFD (ρ = −0.319, P = 0.048) and CNFL (ρ = −0.373, P = 0.019), and the total and immature LC densities correlated with CNBD (ρ = −0.319. P = 0.048, and ρ = −0.328, P = 0.041, respectively). Immature LC density was higher (P = 0.025), but corneal sensitivity and nerve fiber parameters were comparable between patients with (33%) and without neuropsychiatric symptoms and SLE. Conclusions Corneal confocal microscopy identifies distal corneal nerve fiber loss and increased immune cell density in patients with SLE and corneal nerve loss was associated with disease activity. Translational Relevance Corneal confocal microscopy may enable the detection of subclinical corneal nerve loss and immune cell activation in SLE.
Collapse
Affiliation(s)
- Gulfidan Bitirgen
- Department of Ophthalmology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Adem Kucuk
- Division of Rheumatology, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Mustafa Cagri Ergun
- Division of Rheumatology, Department of Internal Medicine, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Ruveyda Baloglu
- Department of Ophthalmology, Necmettin Erbakan University Meram Faculty of Medicine, Konya, Turkey
| | - Miral H Gharib
- Rheumatology Department, Hamad Medical Corporation, Doha, Qatar
| | - Samar Al Emadi
- Rheumatology Department, Hamad Medical Corporation, Doha, Qatar
| | - Georgios Ponirakis
- Weill Cornell Medicine-Qatar, Research Division, Qatar Foundation, Doha, Qatar
| | - Rayaz A Malik
- Weill Cornell Medicine-Qatar, Research Division, Qatar Foundation, Doha, Qatar.,Institute of Cardiovascular Sciences, Cardiac Centre, Faculty of Medical and Human Sciences, University of Manchester and NIHR Clinical Research Facility, Manchester, UK
| |
Collapse
|
40
|
Johnson SA, Shouman K, Shelly S, Sandroni P, Berini SE, Dyck PJB, Hoffman EM, Mandrekar J, Niu Z, Lamb CJ, Low PA, Singer W, Mauermann ML, Mills J, Dubey D, Staff NP, Klein CJ. Small Fiber Neuropathy Incidence, Prevalence, Longitudinal Impairments, and Disability. Neurology 2021; 97:e2236-e2247. [PMID: 34706972 DOI: 10.1212/wnl.0000000000012894] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/24/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES There are limited population-based data on small fiber neuropathy (SFN). We wished to determine SFN incidence, prevalence, comorbid conditions, longitudinal impairments, and disabilities. METHODS Test-confirmed patients with SFN in Olmsted, Minnesota, and adjacent counties were compared 3:1 to matched controls (January 1, 1998-December 31, 2017). RESULTS Ninety-four patients with SFN were identified, with an incidence of 1.3/100,000/y that increased over the study period and a prevalence of 13.3 per 100,000. Average follow-up was 6.1 years (0.7-43 years), and mean onset age was 54 years (range 14-83 years). Female sex (67%), obesity (body mass index mean 30.4 vs 28.5 kg/m2), insomnia (86% vs 54%), analgesic-opioid prescriptions (72% vs 46%), hypertriglyceridemia (180 mg/dL mean vs 147 mg/dL), and diabetes (51% vs 22%, p < 0.001) were more common (odds ratio 3.8-9.0, all p < 0.03). Patients with SFN did not self-identify as disabled with a median modified Rankin Scale score of 1.0 (range 0-6) vs 0.0 (0-6) for controls (p = 0.04). Higher Charlson comorbid conditions (median 6, range 3-9) occurred vs controls (median 3, range 1-9, p < 0.001). Myocardial infarctions occurred in 46% vs 27% of controls (p < 0.0001). Classifications included idiopathic (70%); diabetes (15%); Sjögren disease (2%); AL-amyloid (1%); transthyretin-amyloid (1%); Fabry disease (1%); lupus (1%); postviral (1%); Lewy body (1%), and multifactorial (5%). Foot ulcers occurred in 17, with 71% having diabetes. Large fiber neuropathy developed in 36%, on average 5.3 years (range 0.2-14.3 years) from SFN onset. Median onset Composite Autonomic Severity Score (CASS) was 3 (change per year 0.08, range 0-2.0). Median Neuropathy Impairment Scale (NIS) score was 2 at onset (range 0-8, change per year 1.0, range -7.9 to +23.3). NIS score and CASS change >1 point per year occurred in only AL-amyloid, hereditary transthyretin-amyloid, Fabry, uncontrolled diabetes, and Lewy body. Death after symptom onset was higher in patients with SFN (19%) vs controls (12%, p < 0.001), 50% secondary to diabetes complications. DISCUSSION Isolated SFN is uncommon but increasing in incidence. Most patients do not develop major neurologic impairments and disability but have multiple comorbid conditions, including cardiovascular ischemic events, and increased mortality from SFN onsets. Development of large fiber involvements and diabetes are common over time. Targeted testing facilitates interventional therapies for diabetes but also rheumatologic and rare genetic forms.
Collapse
|
41
|
Abstract
This edition of What is in the Literature focuses on chronic immune neuropathies as they represent treatable conditions. There are formal criteria to solidify the diagnosis of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), but patients are encountered who have clinical and electrodiagnostic features of CIDP but do not fulfill diagnostic criteria. These patients are addressed in recent publications. CIDP (and variants) and other forms of immune-mediated neuropathies (multifocal motor neuropathy) are responsive early on to treatment, but long-term factors are less well described, and a number of publications focus on extended consequences. Acute immune neuropathies have been described in the setting of viral illness, and recent publications look at the question as to whether they are associated with the COVID-19 pandemic. Finally, idiopathic sensory neuropathies are the most common polyneuropathy, and consensus efforts to codify features into subtypes can be used clinically for a more precise diagnosis.
Collapse
Affiliation(s)
- Mark B Bromberg
- Department of Neurology, University of Utah, Salt Lake City, UT
| |
Collapse
|
42
|
Egenolf N, Zu Altenschildesche CM, Kreß L, Eggermann K, Namer B, Gross F, Klitsch A, Malzacher T, Kampik D, Malik RA, Kurth I, Sommer C, Üçeyler N. Diagnosing small fiber neuropathy in clinical practice: a deep phenotyping study. Ther Adv Neurol Disord 2021; 14:17562864211004318. [PMID: 34335876 PMCID: PMC8283814 DOI: 10.1177/17562864211004318] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/02/2021] [Indexed: 11/16/2022] Open
Abstract
Background and aims Small fiber neuropathy (SFN) is increasingly suspected in patients with pain of uncertain origin, and making the diagnosis remains a challenge lacking a diagnostic gold standard. Methods In this case-control study, we prospectively recruited 86 patients with a medical history and clinical phenotype suggestive of SFN. Patients underwent neurological examination, quantitative sensory testing (QST), and distal and proximal skin punch biopsy, and were tested for pain-associated gene loci. Fifty-five of these patients additionally underwent pain-related evoked potentials (PREP), corneal confocal microscopy (CCM), and a quantitative sudomotor axon reflex test (QSART). Results Abnormal distal intraepidermal nerve fiber density (IENFD) (60/86, 70%) and neurological examination (53/86, 62%) most frequently reflected small fiber disease. Adding CCM and/or PREP further increased the number of patients with small fiber impairment to 47/55 (85%). Genetic testing revealed potentially pathogenic gene variants in 14/86 (16%) index patients. QST, QSART, and proximal IENFD were of lower impact. Conclusion We propose to diagnose SFN primarily based on the results of neurological examination and distal IENFD, with more detailed phenotyping in specialized centers.
Collapse
Affiliation(s)
- Nadine Egenolf
- Department of Neurology, University of Würzburg, Germany
| | | | - Luisa Kreß
- Department of Neurology, University of Würzburg, Germany
| | - Katja Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Barbara Namer
- Institute of Physiology, University of Erlangen, Bayern, Germany
| | | | | | | | - Daniel Kampik
- Department of Ophthalmology, University of Würzburg, Bayern, Germany
| | - Rayaz A Malik
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Claudia Sommer
- Department of Neurology, University of Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, Würzburg, 97080, Germany
| |
Collapse
|
43
|
Joseph P, Arevalo C, Oliveira RKF, Faria-Urbina M, Felsenstein D, Oaklander AL, Systrom DM. Insights From Invasive Cardiopulmonary Exercise Testing of Patients With Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Chest 2021; 160:642-651. [PMID: 33577778 DOI: 10.1016/j.chest.2021.01.082] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) affects tens of millions worldwide; the causes of exertional intolerance are poorly understood. The ME/CFS label overlaps with postural orthostatic tachycardia (POTS) and fibromyalgia, and objective evidence of small fiber neuropathy (SFN) is reported in approximately 50% of POTS and fibromyalgia patients. RESEARCH QUESTION Can invasive cardiopulmonary exercise testing (iCPET) and PGP9.5-immunolabeled lower-leg skin biopsies inform the pathophysiology of ME/CFS exertional intolerance and potential relationships with SFN? STUDY DESIGN AND METHODS We analyzed 1,516 upright invasive iCPETs performed to investigate exertional intolerance. After excluding patients with intrinsic heart or lung disease and selecting those with right atrial pressures (RAP) <6.5 mm Hg, results from 160 patients meeting ME/CFS criteria who had skin biopsy test results were compared with 36 control subjects. Rest-to-peak changes in cardiac output (Qc) were compared with oxygen uptake (Qc/VO2 slope) to identify participants with low, normal, or high pulmonary blood flow by Qc/VO2 tertiles. RESULTS During exercise, the 160 ME/CFS patients averaged lower RAP (1.9 ± 2 vs 8.3 ± 1.5; P < .0001) and peak VO2 (80% ± 21% vs 101.4% ± 17%; P < .0001) than control subjects. The low-flow tertile had lower peak Qc than the normal and high-flow tertiles (88.4% ± 19% vs 99.5% ± 23.8% vs 99.9% ± 19.5% predicted; P < .01). In contrast, systemic oxygen extraction was impaired in high-flow vs low- and normal-flow participants (0.74% ± 0.1% vs 0.88 ± 0.11 vs 0.86 ± 0.1; P < .0001) in association with peripheral left-to-right shunting. Among the 160 ME/CFS patient biopsies, 31% were consistent with SFN (epidermal innervation ≤5.0% of predicted; P < .0001). Denervation severity did not correlate with exertional measures. INTERPRETATION These results identify two types of peripheral neurovascular dysregulation that are biologically plausible contributors to ME/CFS exertional intolerance-depressed Qc from impaired venous return, and impaired peripheral oxygen extraction. In patients with small-fiber pathology, neuropathic dysregulation causing microvascular dilation may limit exertion by shunting oxygenated blood from capillary beds and reducing cardiac return.
Collapse
Affiliation(s)
- Phillip Joseph
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale-New Haven Hospital, Yale University, New Haven, CT.
| | - Carlo Arevalo
- Department of Medicine, University of Rochester Medical Center, Rochester, NY
| | - Rudolf K F Oliveira
- Division of Respiratory Diseases, Department of Medicine, Federal University of Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Mariana Faria-Urbina
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Donna Felsenstein
- Infectious Diseases Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Anne Louise Oaklander
- Department of Neurology, Massachusetts General Hospital, Boston, MA; Department of Pathology (Neuropathology), Massachusetts General Hospital, Boston, MA
| | - David M Systrom
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
44
|
Felicetti G, Thoumie P, Do MC, Schieppati M. Cutaneous and muscular afferents from the foot and sensory fusion processing: Physiology and pathology in neuropathies. J Peripher Nerv Syst 2021; 26:17-34. [PMID: 33426723 DOI: 10.1111/jns.12429] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/30/2020] [Accepted: 12/30/2020] [Indexed: 12/16/2022]
Abstract
The foot-sole cutaneous receptors (section 2), their function in stance control (sway minimisation, exploratory role) (2.1), and the modulation of their effects by gait pattern and intended behaviour (2.2) are reviewed. Experimental manipulations (anaesthesia, temperature) (2.3 and 2.4) have shown that information from foot sole has widespread influence on balance. Foot-sole stimulation (2.5) appears to be a promising approach for rehabilitation. Proprioceptive information (3) has a pre-eminent role in balance and gait. Reflex responses to balance perturbations are produced by both leg and foot muscle stretch (3.1) and show complex interactions with skin input at both spinal and supra-spinal levels (3.2), where sensory feedback is modulated by posture, locomotion and vision. Other muscles, notably of neck and trunk, contribute to kinaesthesia and sense of orientation in space (3.3). The effects of age-related decline of afferent input are variable under different foot-contact and visual conditions (3.4). Muscle force diminishes with age and sarcopenia, affecting intrinsic foot muscles relaying relevant feedback (3.5). In neuropathy (4), reduction in cutaneous sensation accompanies the diminished density of viable receptors (4.1). Loss of foot-sole input goes along with large-fibre dysfunction in intrinsic foot muscles. Diabetic patients have an elevated risk of falling, and vision and vestibular compensation strategies may be inadequate (4.2). From Charcot-Marie-Tooth 1A disease (4.3) we have become aware of the role of spindle group II fibres and of the anatomical feet conditions in balance control. Lastly (5) we touch on the effects of nerve stimulation onto cortical and spinal excitability, which may participate in plasticity processes, and on exercise interventions to reduce the impact of neuropathy.
Collapse
Affiliation(s)
- Guido Felicetti
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Neuromotor Rehabilitation, Institute of Montescano, Pavia, Italy
| | - Philippe Thoumie
- Service de rééducation neuro-orthopédique, Hôpital Rothschild APHP, Université Sorbonne, Paris, France.,Agathe Lab ERL Inserm U-1150, Paris, France
| | - Manh-Cuong Do
- Université Paris-Saclay, CIAMS, Orsay, France.,Université d'Orléans, CIAMS, Orléans, France
| | | |
Collapse
|
45
|
Itani M, Gylfadottir SS, Krøigård T, Kristensen AG, Christensen DH, Karlsson P, Möller S, Andersen H, Tankisi H, Nielsen JS, Jensen TS, Thomsen RW, Finnerup NB, Sindrup SH. Small and large fiber sensory polyneuropathy in type 2 diabetes: Influence of diagnostic criteria on neuropathy subtypes. J Peripher Nerv Syst 2020; 26:55-65. [PMID: 33295647 DOI: 10.1111/jns.12424] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/16/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022]
Abstract
Diabetic polyneuropathy (DPN) can be classified based on fiber diameter into three subtypes: small fiber neuropathy (SFN), large fiber neuropathy (LFN), and mixed fiber neuropathy (MFN). We examined the effect of different diagnostic models on the frequency of polyneuropathy subtypes in type 2 diabetes patients with DPN. This study was based on patients from the Danish Center for Strategic Research in Type 2 Diabetes cohort. We defined DPN as probable or definite DPN according to the Toronto Consensus Criteria. DPN was then subtyped according to four distinct diagnostic models. A total of 277 diabetes patients (214 with DPN and 63 with no DPN) were included in the study. We found a considerable variation in polyneuropathy subtypes by applying different diagnostic models independent of the degree of certainty of DPN diagnosis. For probable and definite DPN, the frequency of subtypes across diagnostic models varied from: 1.4% to 13.1% for SFN, 9.3% to 21.5% for LFN, 51.4% to 83.2% for MFN, and 0.5% to 14.5% for non-classifiable neuropathy (NCN). For the definite DPN group, the frequency of subtypes varied from: 1.6% to 13.5% for SFN, 5.6% to 20.6% for LFN, 61.9% to 89.7% for MFN, and 0.0% to 6.3% for NCN. The frequency of polyneuropathy subtypes depends on the type and number of criteria applied in a diagnostic model. Future consensus criteria should clearly define sensory functions to be tested, methods of testing, and how findings should be interpreted for both clinical practice and research purpose.
Collapse
Affiliation(s)
- Mustapha Itani
- Research Unit for Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sandra Sif Gylfadottir
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Krøigård
- Research Unit for Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | | | - Pall Karlsson
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, Denmark
| | - Sören Möller
- OPEN-Open Patient data Explorative Network, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Steen Nielsen
- Danish Center for Strategic Research in Type 2 Diabetes, Steno Diabetes Center Odense, Odense, Denmark
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Hein Sindrup
- Research Unit for Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
46
|
Oaklander AL. Clinical significance of angiotensin-converting enzyme 2 receptors for severe acute respiratory syndrome coronavirus 2 (COVID-19) on peripheral small-fiber sensory neurons is unknown today. Pain 2020; 161:2431-2433. [PMID: 32826753 PMCID: PMC10034827 DOI: 10.1097/j.pain.0000000000002050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Anne Louise Oaklander
- Nerve Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Pathology (Neuropathology), Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|