1
|
Neuropatie sensitive. Neurologia 2022. [DOI: 10.1016/s1634-7072(21)46002-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
2
|
Long-term efficacy of immunoglobulins in small fiber neuropathy related to Sjögren’s syndrome. J Neurol 2020; 267:3499-3507. [DOI: 10.1007/s00415-020-10033-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/28/2022]
|
3
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
4
|
Corneal nerves in health and disease. Prog Retin Eye Res 2019; 73:100762. [DOI: 10.1016/j.preteyeres.2019.05.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
|
5
|
Ginsberg MR, Morren JA. Utility of electrodiagnostic studies in patients referred with a diagnosis of polyneuropathy. Muscle Nerve 2019; 61:288-292. [PMID: 31650552 DOI: 10.1002/mus.26746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/13/2019] [Accepted: 10/19/2019] [Indexed: 01/25/2023]
Abstract
BACKGROUND Peripheral polyneuropathies (PN) are common neuromuscular conditions. The role of electrodiagnostic study (EDX) in diagnosis of PN is not well-defined. METHODS We performed a retrospective chart review of patients referred for EDX evaluation of PN. RESULTS Of 162 patients analyzed, 23 had pure peripheral neuropathy (pPN; 14.2%), 29 had peripheral neuropathy and another diagnosis (PN+; 17.9%), 51 had an alternative diagnosis (nonPN; 31.5%), and 59 had normal studies (36.4%). In univariable analysis, age (P < .001) and gender (P = .004) were weakly associated with final diagnosis. In multinomial logistic regression analysis, significant predictors included age (odds ratio [OR] for nonPN/PN+:1.07 per year; 95% confidence interval [CI], 1.03-1.11), gender (OR for PN+:0.2, 95% CI, 0.07-0.61), and diabetes/prediabetes (OR for pPN:3.29; 95% CI, 1.17-9.27). CONCLUSIONS These data suggest that EDX commonly yields additional or nonPNs in patients referred with a diagnosis of PN, and although some variables predict electrodiagnosis, none have a large enough effect to suggest poor utility in any subpopulation.
Collapse
Affiliation(s)
- Matthew R Ginsberg
- Neuromuscular Center, Neurological Institute-Cleveland Clinic, Cleveland, Ohio
| | - John A Morren
- Neuromuscular Center, Neurological Institute-Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
6
|
Chen Y, Wang C, Xu F, Ming F, Zhang H. Efficacy and Tolerability of Intravenous Immunoglobulin and Subcutaneous Immunoglobulin in Neurologic Diseases. Clin Ther 2019; 41:2112-2136. [PMID: 31445679 DOI: 10.1016/j.clinthera.2019.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 06/01/2019] [Accepted: 07/10/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE IV immunoglobulin (Ig) therapy has been widely used for the treatment of neurologic disorders, autoimmune diseases, immunodeficiency-related diseases, blood system diseases, and cancers. In this review, we summarize the efficacy and tolerability of IVIg and SCIg therapy in neurologic diseases. METHODS We summarized and analyzed the efficacy and tolerability of IVIg and SCIg in neurologic diseases, by analyzing the literature pertaining to the use of IVIg and SCIg to treat nervous system diseases. FINDINGS In clinical neurology practice, IVIg has been shown to be useful for the treatment of new-onset or recurrent immune diseases and for long-term maintenance treatment of chronic diseases. Moreover, IVIg may have applications in the management of intractable autoimmune epilepsy, paraneoplastic syndrome, autoimmune encephalitis, and neuromyelitis optica. SCIg is emerging as an alternative to IVIg treatment. Although SCIg has a composition similar to that of IVIg, the applications of this therapy are different. Notably, the bioavailability of SCIg is lower than that of IVIg, but the homeostasis level is more stable. Current studies have shown that these 2 therapies have pharmacodynamic equivalence. IMPLICATIONS In this review, we explored the efficacy of IVIg in the treatment of various neurologic disorders. IVIg administration still faces many challenges. Thus, it will be necessary to standardize the use of IVIg in the clinical setting. SCIg administration is a novel and feasible treatment option for neurologic and immune-related diseases, such as chronic inflammatory demyelinating polyradiculoneuropathy and idiopathic inflammatory myopathies. As our understanding of the mechanisms of action of IVIg improve, potential next-generation biologics can being developed.
Collapse
Affiliation(s)
- Yun Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunyu Wang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fanxi Xu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fengyu Ming
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hainan Zhang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
7
|
Abstract
Sensory polyneuropathies, which are caused by dysfunction of peripheral sensory nerve fibers, are a heterogeneous group of disorders that range from the common diabetic neuropathy to the rare sensory neuronopathies. The presenting symptoms, acuity, time course, severity, and subsequent morbidity vary and depend on the type of fiber that is affected and the underlying cause. Damage to small thinly myelinated and unmyelinated nerve fibers results in neuropathic pain, whereas damage to large myelinated sensory afferents results in proprioceptive deficits and ataxia. The causes of these disorders are diverse and include metabolic, toxic, infectious, inflammatory, autoimmune, and genetic conditions. Idiopathic sensory polyneuropathies are common although they should be considered a diagnosis of exclusion. The diagnostic evaluation involves electrophysiologic testing including nerve conduction studies, histopathologic analysis of nerve tissue, serum studies, and sometimes autonomic testing and cerebrospinal fluid analysis. The treatment of these diseases depends on the underlying cause and may include immunotherapy, mitigation of risk factors, symptomatic treatment, and gene therapy, such as the recently developed RNA interference and antisense oligonucleotide therapies for transthyretin familial amyloid polyneuropathy. Many of these disorders have no directed treatment, in which case management remains symptomatic and supportive. More research is needed into the underlying pathophysiology of nerve damage in these polyneuropathies to guide advances in treatment.
Collapse
Affiliation(s)
- Kelly Graham Gwathmey
- Virginia Commonwealth University, Department of Neurology, 1101 E. Marshall Street, PO Box 980599, Richmond, VA 23298, USA
| | - Kathleen T Pearson
- Virginia Commonwealth University, Department of Neurology, 1101 E. Marshall Street, PO Box 980599, Richmond, VA 23298, USA
| |
Collapse
|
8
|
Liu X, Treister R, Lang M, Oaklander AL. IVIg for apparently autoimmune small-fiber polyneuropathy: first analysis of efficacy and safety. Ther Adv Neurol Disord 2018; 11:1756285617744484. [PMID: 29403541 PMCID: PMC5791555 DOI: 10.1177/1756285617744484] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 11/03/2017] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Small-fiber polyneuropathy (SFPN) has various underlying causes, including associations with systemic autoimmune conditions. We have proposed a new cause; small-fiber-targeting autoimmune diseases akin to Guillain-Barré and chronic inflammatory demyelinating polyneuropathy (CIDP). There are no treatment studies yet for this 'apparently autoimmune SFPN' (aaSFPN), but intravenous immunoglobulin (IVIg), first-line for Guillain-Barré and CIDP, is prescribed off-label for aaSFPN despite very high cost. This project aimed to conduct the first systematic evaluation of IVIg's effectiveness for aaSFPN. METHODS With IRB approval, we extracted all available paper and electronic medical records of qualifying patients. Inclusion required having objectively confirmed SFPN, autoimmune attribution and other potential causes excluded. IVIg needed to have been dosed at ⩾1 g/kg/4 weeks for ⩾3 months. We chose two primary outcomes - changes in composite autonomic function testing (AFT) reports of SFPN and in ratings of pain severity - to capture objective as well as patient-prioritized outcomes. RESULTS Among all 55 eligible patients, SFPN had been confirmed by 3/3 nerve biopsies, 62% of skin biopsies, and 89% of composite AFT. Evidence of autoimmunity included 27% of patients having systemic autoimmune disorders, 20% having prior organ-specific autoimmune illnesses and 80% having ⩾1/5 abnormal blood-test markers associated with autoimmunity. A total of 73% had apparent small-fiber-restricted autoimmunity. IVIg treatment duration averaged 28 ± 25 months. The proportion of AFTs interpreted as indicating SFPN dropped from 89% at baseline to 55% (p ⩽ 0.001). Sweat production normalized (p = 0.039) and the other four domains all trended toward improvement. Among patients with pre-treatment pain ⩾3/10, severity averaging 6.3 ± 1.7 dropped to 5.2 ± 2.1 (p = 0.007). Overall, 74% of patients rated themselves 'improved' and their neurologists labeled 77% as 'IVIg responders'; 16% entered remissions that were sustained after IVIg withdrawal. All adverse events were expected; most were typical infusion reactions. The two moderate complications (3.6%) were vein thromboses not requiring discontinuation. The one severe event (1.8%), hemolytic anemia, remitted after IVIg discontinuation. CONCLUSION These results provide Class IV, real-world, proof-of-concept evidence suggesting that IVIg is safe and effective for rigorously selected SFPN patients with apparent autoimmune causality. They provide rationale for prospective trials, inform trial design and indirectly support the discovery of small-fiber-targeting autoimmune/inflammatory illnesses.
Collapse
Affiliation(s)
- Xiaolei Liu
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology, Dayi Hospital of Shanxi Medical University, China
| | - Roi Treister
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA; Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Magdalena Lang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Anne Louise Oaklander
- Department of Neurology, Massachusetts General Hospital, 275 Charles Street/Warren Building 310, Boston, MA 02114, USA
| |
Collapse
|
9
|
Abstract
Small fiber neuropathy, which affects the sensory Aδ and C fibers, is now a major diagnostic and therapeutic challenge. Nearly 7% of the general population have chronic neuropathic pain responsible for severe quality-of-life impairments. Awareness must therefore be raised among clinicians of the somatosensory and autonomic symptoms that can reveal small fiber neuropathy, appropriate diagnostic investigations, most common causes, and best treatment options for each patient profile. To help achieve this goal, the present review article discusses the clinical presentation of neuropathic pain and paresthesia and/or autonomic dysfunction due to involvement of nerves supplying exocrine glands and smooth muscle; normal findings from standard electrophysiological investigations; most informative diagnostic tests (epidermal nerve fiber density in a skin biopsy, laser-evoked potentials, heat- and cold-detection thresholds, electrochemical skin conductance); main causes, which consist chiefly of metabolic diseases (diabetes mellitus, glucose intolerance), dysimmunity syndromes (Sjögren's syndrome, sarcoidosis, monoclonal gammopathy), and genetic abnormalities (familial amyloidosis due to a transthyretin mutation, Fabry disease, sodium channel diseases); and the available symptomatic and etiological treatments.
Collapse
|
10
|
Rolyan H, Liu S, Hoeijmakers JG, Faber CG, Merkies IS, Lauria G, Black JA, Waxman SG. A painful neuropathy-associated Nav1.7 mutant leads to time-dependent degeneration of small-diameter axons associated with intracellular Ca2+ dysregulation and decrease in ATP levels. Mol Pain 2016; 12:1744806916674472. [PMID: 27821467 PMCID: PMC5102167 DOI: 10.1177/1744806916674472] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 11/25/2022] Open
Abstract
Small fiber neuropathy is a painful sensory nervous system disorder characterized by damage to unmyelinated C- and thinly myelinated Aδ- nerve fibers, clinically manifested by burning pain in the distal extremities and dysautonomia. The clinical onset in adulthood suggests a time-dependent process. The mechanisms that underlie nerve fiber injury in small fiber neuropathy are incompletely understood, although roles for energetic stress have been suggested. In the present study, we report time-dependent degeneration of neurites from dorsal root ganglia neurons in culture expressing small fiber neuropathy-associated G856D mutant Nav1.7 channels and demonstrate a time-dependent increase in intracellular calcium levels [Ca2+]i and reactive oxygen species, together with a decrease in ATP levels. Together with a previous clinical report of burning pain in the feet and hands associated with reduced levels of Na+/K+-ATPase in humans with high altitude sickness, the present results link energetic stress and reactive oxygen species production with the development of a painful neuropathy that preferentially affects small-diameter axons.
Collapse
Affiliation(s)
- Harshvardhan Rolyan
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Neuroscience and Regeneration Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Shujun Liu
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Neuroscience and Regeneration Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Janneke Gj Hoeijmakers
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ingemar Sj Merkies
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS, Carlo Besta Neurological Institute, Milan, Italy
| | - Joel A Black
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Neuroscience and Regeneration Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Neuroscience and Regeneration Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
11
|
de Greef BTA, Geerts M, Hoeijmakers JGJ, Faber CG, Merkies ISJ. Intravenous immunoglobulin therapy for small fiber neuropathy: study protocol for a randomized controlled trial. Trials 2016; 17:330. [PMID: 27439408 PMCID: PMC4955261 DOI: 10.1186/s13063-016-1450-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/15/2016] [Indexed: 01/02/2023] Open
Abstract
Background Small fiber neuropathy is the most common cause of neuropathic pain in peripheral neuropathies, with a minimum prevalence of 53/100,000. Patients experience excruciating pain, and currently available anti-neuropathic and other pain drugs do not relieve the pain substantially. Several open-label studies have suggested an immunological basis in small fiber neuropathy and have reported efficacy of treatment with intravenous immunoglobulin. Therefore, immunological mechanisms conceivably may play a role in small fiber neuropathy. To date, no randomized controlled study with intravenous immunoglobulin in patients with small fiber neuropathy has been performed. Methods/design This study is a randomized, double-blind, placebo-controlled, clinical trial in patients with idiopathic small fiber neuropathy. The primary objective is to investigate the efficacy of intravenous immunoglobulin versus placebo on pain alleviation. A 1-point change in the PI-NRS compared to baseline is considered the minimum clinically important difference. In the IVIg-treated group, we assume a response rate of approximately 60 % based on the criteria composed by the IMMPACT group for measurement of pain. Based on this, a sample size of 60 patients is needed. Eligible patients fulfilling the inclusion/exclusion criteria will be randomized to receive either intravenous immunoglobulin or placebo (0.9 % saline). The treatment regimen will start with a loading dose of 2 g/kg body weight over 2–4 consecutive days, followed by a maintenance dose of 1 g/kg body weight over 1–2 consecutive days given three times at a 3-week interval. The primary endpoint is the comparison of the percentage of responder subjects between the two treatment groups from the first randomization during the 12 weeks of treatment. A responder is defined as ≥ 1-point Pain Intensity Numerical Rating Scale improvement on the mean weekly peak pain relative to baseline. The secondary outcomes are pain intensity, pain qualities, other small fiber neuropathy-related complaints, daily and social functioning, as well as quality of life. In addition, safety assessments will be performed for adverse events, vital signs, and laboratory values outside the normal range. Responders during the 12-week treatment period will be followed during a 3-month extension phase. Discussion This is the first randomized, double-blind, placebo-controlled clinical trial with intravenous immunoglobulin in patients with idiopathic small fiber neuropathy. Positive findings will result in a new treatment option for small fiber neuropathy and support an immunological role in this condition. Trial registration ClinicalTrials.gov, NCT02637700. Registered on 16 December 2015. Electronic supplementary material The online version of this article (doi:10.1186/s13063-016-1450-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - Margot Geerts
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.,Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| |
Collapse
|
12
|
Efficacy, safety, and tolerability of lacosamide in patients with gain-of-function Nav1.7 mutation-related small fiber neuropathy: study protocol of a randomized controlled trial-the LENSS study. Trials 2016; 17:306. [PMID: 27363506 PMCID: PMC4929773 DOI: 10.1186/s13063-016-1430-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 05/18/2016] [Indexed: 12/02/2022] Open
Abstract
Background Small fiber neuropathy generally leads to considerable pain and autonomic symptoms. Gain-of-function mutations in the SCN9A- gene, which codes for the Nav1.7 voltage-gated sodium channel, have been reported in small fiber neuropathy, suggesting an underlying genetic basis in a subset of patients. Currently available sodium channel blockers lack selectivity, leading to cardiac and central nervous system side effects. Lacosamide is an anticonvulsant, which blocks Nav1.3, Nav1.7, and Nav1.8, and stabilizes channels in the slow-inactivation state. Since multiple Nav1.7 mutations in small fiber neuropathy showed impaired slow-inactivation, lacosamide might be effective. Methods/design The Lacosamide-Efficacy-‘N’-Safety in Small fiber neuropathy (LENSS) study is a randomized, double-blind, placebo-controlled, crossover trial in patients with SCN9A-associated small fiber neuropathy, with the primary objective to evaluate the efficacy of lacosamide versus placebo. Eligible patients (the aim is to recruit 25) fulfilling the inclusion and exclusion criteria will be randomized to receive lacosamide (200 mg b.i.d.) or placebo during the first double-blinded treatment period (8 weeks), which is preceded by a titration period (3 weeks). The first treatment period will be followed by a tapering period (2 weeks). After a 2-week washout period, patients will crossover to the alternate arm for the second period consisting of an equal titration phase, treatment period, and tapering period. The primary efficacy endpoint will be the proportion of patients demonstrating a 1-point average pain score reduction compared to baseline using the Pain Intensity Numerical Rating Scale. We assume a response rate of approximately 60 % based on the criteria composed by the Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (IMMPACT) group for measurement of pain. Patients withdrawing from the study will be considered non- responders. Secondary outcomes will include changes in maximum pain score, the Small Fiber Neuropathy Symptoms Inventory Questionnaire, sleep quality and the quality of life assessment, patients’ global impressions of change, and safety and tolerability measurements. Sensitivity analyses will include assessing the proportion of patients having ≥ 2 points average pain improvement compared to the baseline Pain Intensity Numerical Rating Scale scores. Discussion This is the first study that will be evaluating the efficacy, safety, and tolerability of lacosamide versus placebo in patients with SCN9A-associated small fiber neuropathy. The findings may increase the knowledge on lacosamide as a potential treatment option in patients with painful neuropathies, considering the central role of Nav1.7 in pain. Trial registration ClinicalTrials.gov, NCT01911975. Registered on 13 July 2013. Electronic supplementary material The online version of this article (doi:10.1186/s13063-016-1430-1) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Dabby R, Sadeh M, Broitman Y, Yosovich K, Dickman R, Leshinsky-Silver E. Painful small fiber neuropathy with gastroparesis: A new phenotype with a novel mutation in the SCN10A gene. J Clin Neurosci 2016; 26:84-8. [DOI: 10.1016/j.jocn.2015.05.071] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 05/16/2015] [Accepted: 05/17/2015] [Indexed: 01/27/2023]
|
14
|
de Greef BTA, Hoeijmakers JGJ, Wolters EE, Smeets HJM, van den Wijngaard A, Merkies ISJ, Faber CG, Gerrits MM. No Fabry Disease in Patients Presenting with Isolated Small Fiber Neuropathy. PLoS One 2016; 11:e0148316. [PMID: 26866599 PMCID: PMC4750945 DOI: 10.1371/journal.pone.0148316] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/07/2015] [Indexed: 12/31/2022] Open
Abstract
Objective Screening for Fabry disease in patients with small fiber neuropathy has been suggested, especially since Fabry disease is potentially treatable. However, the diagnostic yield of testing for Fabry disease in isolated small fiber neuropathy patients has never been systematically investigated. Our aim is to determine the presence of Fabry disease in patients with small fiber neuropathy. Methods Patients referred to our institute, who met the criteria for isolated small fiber neuropathy were tested for Fabry disease by measurement of alpha-Galactosidase A activity in blood, lysosomal globotriaosylsphingosine in urine and analysis on possible GLA gene mutations. Results 725 patients diagnosed with small fiber neuropathy were screened for Fabry disease. No skin abnormalities were seen except for redness of the hands or feet in 30.9% of the patients. Alfa-Galactosidase A activity was tested in all 725 patients and showed diminished activity in eight patients. Lysosomal globotriaosylsphingosine was examined in 509 patients and was normal in all tested individuals. Screening of GLA for mutations was performed for 440 patients, including those with diminished α-Galactosidase A activity. Thirteen patients showed a GLA gene variant. One likely pathogenic variant was found in a female patient. The diagnosis Fabry disease could not be confirmed over time in this patient. Eventually none of the patients were diagnosed with Fabry disease. Conclusions In patients with isolated small fiber neuropathy, and no other signs compatible with Fabry disease, the diagnostic yield of testing for Fabry disease is extremely low. Testing for Fabry disease should be considered only in cases with additional characteristics, such as childhood onset, cardiovascular disease, renal failure, or typical skin lesions.
Collapse
Affiliation(s)
- Bianca T. A. de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
- * E-mail:
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Emma E. Wolters
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hubertus J. M. Smeets
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Arthur van den Wijngaard
- Department of Clinical Genetics, 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, Spaarne hospital, Hoofddorp, The Netherlands
| | - Catharina G. Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | | |
Collapse
|
15
|
Abstract
EMG, which consists of nerve conduction studies and needle electromyography, is an essential diagnostic tool in the evaluation of patients with suspected peripheral neuropathy. Many neurologists order an EMG for all patients with suspected peripheral neuropathy. Not surprisingly, evidence now exists that shows EMG is a major driver of health care costs associated with neuropathy diagnoses. As neurologic practice evolves from fee for service to value-based compensation, neurologists will need to justify the diagnostic utility of EMG (outcome) relative to its cost. While carefully performed studies of diagnostic utility in many patient populations are lacking, a robust literature provides guidance regarding the potential role and limitations of EMG in neuropathy diagnosis as well as the pitfalls referring providers and electrodiagnostic consultants must consider. Do all neuropathy patients need an EMG at least once? This article attempts to answer this question using an illustrative case to highlight critical factors every neurologist must consider before ordering an EMG for neuropathy diagnosis.
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW This article discusses the clinical features, pathophysiology, and management of primary and secondary acquired immune axonal neuropathies. RECENT FINDINGS Although there are many collagen vascular disorders associated with vasculitic neuropathy, a quarter of cases have been described to be due to nonsystemic vasculitis of the peripheral nervous system. Enhanced surveillance and aggressive treatment of conditions such as cryoglobulin-related vasculitic neuropathy with cyclophosphamide, rituximab, and alfa interferons has led to improved morbidity and mortality, however, many cases of immune axonal acquired neuropathy are still associated with poor outcomes. Acute motor axonal neuropathy (AMAN) and acute motor sensory axonal neuropathy (AMSAN) are well-characterized variants of Guillain-Barré syndrome. SUMMARY Characterizing the clinical and electrophysiologic phenotype can help diagnose conditions such as nonsystemic vasculitic neuropathy, AMAN, AMSAN, and immune small fiber neuropathy, while careful evaluation of systemic features is key to identifying secondary immune axonal neuropathies such as vasculitic neuropathy related to collagen vascular disease. Additional research is needed to determine the exact immune pathogenesis and optimized treatment regimens for all acquired immune axonal neuropathies.
Collapse
|
17
|
Camdessanché JP, Antoine JC. Neuropatie sensitive. Neurologia 2015. [DOI: 10.1016/s1634-7072(15)70522-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
18
|
Lagerburg V, Bakkers M, Bouwhuis A, Hoeijmakers JG, Smit AM, Van Den Berg SJ, Hordijk-De Boer I, Brouwer-Van Der Lee MD, Kranendonk D, Reulen JP, Faber CG, Merkies IS. Contact heat evoked potentials: Normal values and use in small-fiber neuropathy. Muscle Nerve 2015; 51:743-9. [DOI: 10.1002/mus.24465] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 09/06/2014] [Accepted: 09/22/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Vera Lagerburg
- Department of Clinical Physics; Catharina Hospital; Eindhoven The Netherlands
| | - Mayienne Bakkers
- Department of Neurology Maastricht University Medical Centre; P.O. Box 5800 6202 AZ Maastricht The Netherlands
| | - Anne Bouwhuis
- Department of Neurology Maastricht University Medical Centre; P.O. Box 5800 6202 AZ Maastricht The Netherlands
| | - Janneke G.J. Hoeijmakers
- Department of Neurology Maastricht University Medical Centre; P.O. Box 5800 6202 AZ Maastricht The Netherlands
| | - Arjen M. Smit
- Department of Clinical Neurophysiology; Spaarne Hospital; Hoofddorp The Netherlands
| | | | | | | | - Douwe Kranendonk
- Department of Neurology Spaarne Hospital Hoofddorp; The Netherlands
| | - Jos P.H. Reulen
- Department of Clinical Neurophysiology Maastricht University Medical Centre; Maastricht The Netherlands
| | - Catharina G. Faber
- Department of Neurology Maastricht University Medical Centre; P.O. Box 5800 6202 AZ Maastricht The Netherlands
| | | |
Collapse
|
19
|
Brouwer BA, Merkies ISJ, Gerrits MM, Waxman SG, Hoeijmakers JGJ, Faber CG. Painful neuropathies: the emerging role of sodium channelopathies. J Peripher Nerv Syst 2014; 19:53-65. [PMID: 25250524 DOI: 10.1111/jns5.12071] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pain is a frequent debilitating feature reported in peripheral neuropathies with involvement of small nerve (Aδ and C) fibers. Voltage-gated sodium channels are responsible for the generation and conduction of action potentials in the peripheral nociceptive neuronal pathway where NaV 1.7, NaV 1.8, and NaV 1.9 sodium channels (encoded by SCN9A, SCN10A, and SCN11A) are preferentially expressed. The human genetic pain conditions inherited erythromelalgia and paroxysmal extreme pain disorder were the first to be linked to gain-of-function SCN9A mutations. Recent studies have expanded this spectrum with gain-of-function SCN9A mutations in patients with small fiber neuropathy and in a new syndrome of pain, dysautonomia, and small hands and small feet (acromesomelia). In addition, painful neuropathies have been recently linked to SCN10A mutations. Patch-clamp studies have shown that the effect of SCN9A mutations is dependent upon the cell-type background. The functional effects of a mutation in dorsal root ganglion (DRG) neurons and sympathetic neuron cells may differ per mutation, reflecting the pattern of expression of autonomic symptoms in patients with painful neuropathies who carry the mutation in question. Peripheral neuropathies may not always be length-dependent, as demonstrated in patients with initial facial and scalp pain symptoms with SCN9A mutations showing hyperexcitability in both trigeminal ganglion and DRG neurons. There is some evidence suggesting that gain-of-function SCN9A mutations can lead to degeneration of peripheral axons. This review will focus on the emerging role of sodium channelopathies in painful peripheral neuropathies, which could serve as a basis for novel therapeutic strategies.
Collapse
Affiliation(s)
- Brigitte A Brouwer
- Department of Anesthesiology and Pain Management, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
20
|
Rezaei N, Abolhassani H, Aghamohammadi A, Ochs HD. Indications and safety of intravenous and subcutaneous immunoglobulin therapy. Expert Rev Clin Immunol 2014; 7:301-16. [DOI: 10.1586/eci.10.104] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
Sachedina S, Toth C. Progression in idiopathic, diabetic, paraproteinemic, alcoholic, and B12 deficiency neuropathy. J Peripher Nerv Syst 2013; 18:247-55. [DOI: 10.1111/jns5.12042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 08/03/2013] [Accepted: 08/05/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Shafina Sachedina
- Royal College of Surgeons Ireland, School of Medicine; Dublin Ireland
| | - Cory Toth
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute; University of Calgary; Calgary AB Canada
| |
Collapse
|
22
|
Abstract
Chronic sensory or sensorimotor polyneuropathy is a common cause for referral to neurologists. Despite extensive diagnostic testing, up to one-third of these patients remain without a known cause, and are referred to as having cryptogenic sensory peripheral neuropathy. Symptoms progress slowly. On examination, there may be additional mild toe flexion and extension weakness. Electrophysiologic testing and histology reveals axonal neuropathy. Prognosis is usually favorable, as most patients maintain independent ambulation. Besides patient education and reassurance, management is focused on pharmacotherapy for neuropathic pain and physical therapy for balance training, and, occasionally, assistive devices.
Collapse
Affiliation(s)
- Mamatha Pasnoor
- Department of Neurology, University of Kansas Medical Center, 3599 Rainbow Boulevard, Kansas City, KS 66160, USA.
| | | | | |
Collapse
|
23
|
Sarcoidosis and pain caused by small-fiber neuropathy. PAIN RESEARCH AND TREATMENT 2012; 2012:256024. [PMID: 23304492 PMCID: PMC3523152 DOI: 10.1155/2012/256024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 10/01/2012] [Indexed: 12/20/2022]
Abstract
Sarcoidosis is a chronic inflammatory illness and small-fiber neuropathy (SFN) is one of the disabling and often chronic manifestations of the disease. SFN presents with peripheral pain and symptoms of autonomic dysfunction. The character of the pain can be burning or shooting. Besides, allodynia and hyperesthesia can exist. Diagnosis is usually made on the basis of clinical features, in combination with abnormal specialized tests. The aim of treatment is often to reduce pain; however, total pain relieve is seldom achieved. The role of TNF-α in the pathogenesis of SFN in sarcoidosis appears interesting to explore. Novel therapeutic agents such as ARA 290, a nonhematopoietic erythropoietin analogue with potent anti-inflammatory and tissue protective properties, are interesting to explore in the treatment of SFN in sarcoidosis.
Collapse
|
24
|
Abstract
Peripheral neuropathies are among the most common disorders encountered by neuromuscular specialists and their evaluation can be challenging. The first part of this review outlined an algorithm based on anatomy, pathology, electrodiagnosis, and clinical localization that leads to a full characterization of the peripheral neuropathy. In the second part, we apply this approach, emphasizing recognition of atypical features and formulation of a focused differential diagnosis, thus reducing the number of uninformative tests. We review evidence supporting the routine use of commonly ordered laboratory tests and recommend a panel of tests that should be performed in patients with symmetric, distal, sensory-predominant peripheral neuropathy. Using this diagnostic approach, a diagnosis could be made in two thirds of patients.
Collapse
|
25
|
Hoeijmakers JGJ, Merkies ISJ, Gerrits MM, Waxman SG, Faber CG. Genetic aspects of sodium channelopathy in small fiber neuropathy. Clin Genet 2012; 82:351-8. [PMID: 22803682 DOI: 10.1111/j.1399-0004.2012.01937.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/11/2012] [Accepted: 07/11/2012] [Indexed: 12/12/2022]
Abstract
Small fiber neuropathy (SFN) is a disorder typically dominated by neuropathic pain and autonomic dysfunction, in which the thinly myelinated Aδ-fibers and unmyelinated C-fibers are selectively injured. The diagnosis SFN is based on a reduced intraepidermal nerve fiber density and/or abnormal thermal thresholds in quantitative sensory testing. The etiologies of SFN are diverse, although no apparent cause is frequently seen. Recently, SCN9A-gene variants (single amino acid substitutions) have been found in ∼30% of a cohort of idiopathic SFN patients, producing gain-of-function changes in sodium channel Na(V)1.7, which is preferentially expressed in small diameter peripheral axons. Functional testing showed that these variants altered fast inactivation, slow inactivation or resurgent current and rendered dorsal root ganglion neurons hyperexcitable. In this review, we discuss the role of Na(V)1.7 in pain and highlight the molecular genetics and pathophysiology of SCN9A-gene variants in SFN. With increasing knowledge regarding the underlying pathophysiology in SFN, the development of specific treatment in these patients seems a logical target for future studies.
Collapse
Affiliation(s)
- J G J Hoeijmakers
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
26
|
Han C, Hoeijmakers JGJ, Liu S, Gerrits MM, te Morsche RHM, Lauria G, Dib-Hajj SD, Drenth JPH, Faber CG, Merkies ISJ, Waxman SG. Functional profiles of SCN9A variants in dorsal root ganglion neurons and superior cervical ganglion neurons correlate with autonomic symptoms in small fibre neuropathy. Brain 2012; 135:2613-28. [DOI: 10.1093/brain/aws187] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
27
|
Hoeijmakers JG, Faber CG, Lauria G, Merkies IS, Waxman SG. Small-fibre neuropathies—advances in diagnosis, pathophysiology and management. Nat Rev Neurol 2012; 8:369-79. [DOI: 10.1038/nrneurol.2012.97] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Singer MA, Vernino SA, Wolfe GI. Idiopathic neuropathy: new paradigms, new promise. J Peripher Nerv Syst 2012; 17 Suppl 2:43-9. [DOI: 10.1111/j.1529-8027.2012.00395.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Neurological Disorders in Primary Sjögren's Syndrome. Autoimmune Dis 2012; 2012:645967. [PMID: 22474573 PMCID: PMC3303537 DOI: 10.1155/2012/645967] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 12/15/2011] [Indexed: 02/07/2023] Open
Abstract
Sjögren's syndrome is an autoimmune disease characterized by an autoimmune exocrinopathy involving mainly salivary and lacrimal glands. The histopathological hallmark is periductal lymphocytic infiltration of the exocrine glands, resulting in loss of their secretory function. Several systemic manifestations may be found in patients with Sjögren's syndrome including neurological disorders. Neurological involvement ranges from 0 to 70% among various series and may present with central nervous system and/or peripheral nervous system involvement. This paper endeavors to review the main clinical neurological manifestations in Sjögren syndrome, the physiopathology, and their therapeutic response.
Collapse
|
30
|
Hoeijmakers JGJ, Han C, Merkies ISJ, Macala LJ, Lauria G, Gerrits MM, Dib-Hajj SD, Faber CG, Waxman SG. Small nerve fibres, small hands and small feet: a new syndrome of pain, dysautonomia and acromesomelia in a kindred with a novel NaV1.7 mutation. ACTA ACUST UNITED AC 2012; 135:345-58. [PMID: 22286749 DOI: 10.1093/brain/awr349] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Na(V)1.7 sodium channel is preferentially expressed within dorsal root ganglion and sympathetic ganglion neurons and their small-diameter peripheral axons. Gain-of-function variants of Na(V)1.7 have recently been described in patients with painful small fibre neuropathy and no other apparent cause. Here, we describe a novel syndrome of pain, dysautonomia, small hands and small feet in a kindred carrying a novel Na(V)1.7 mutation. A 35-year-old male presented with erythema and burning pain in the hands since early childhood, later disseminating to the feet, cheeks and ears. He also experienced progressive muscle cramps, profound sweating, bowel disturbances (diarrhoea or constipation), episodic dry eyes and mouth, hot flashes, and erectile dysfunction. Neurological examination was normal. Physical examination was remarkable in revealing small hands and feet (acromesomelia). Blood examination and nerve conduction studies were unremarkable. Intra-epidermal nerve fibre density was significantly reduced compared to age- and sex-matched normative values. The patient's brother and father reported similar complaints including distal extremity redness and pain, and demonstrated comparable distal limb under-development. Quantitative sensory testing revealed impaired warmth sensation in the proband, father and brother. Genetic analysis revealed a novel missense mutation in the SCN9A gene encoding sodium channel Na(V)1.7 (G856D; c.2567G > A) in all three affected subjects, but not in unaffected family members. Functional analysis demonstrated that the mutation hyperpolarizes (-9.3 mV) channel activation, depolarizes (+6.2 mV) steady-state fast-inactivation, slows deactivation and enhances persistent current and the response to slow ramp stimuli by 10- to 11-fold compared with wild-type Na(V)1.7 channels. Current-clamp analysis of dorsal root ganglion neurons transfected with G856D mutant channels demonstrated depolarized resting potential, reduced current threshold, increased repetitive firing in response to suprathreshold stimulation and increased spontaneous firing. Our results demonstrate that the G856D mutation produces DRG neuron hyperexcitability which underlies pain in this kindred, and suggest that small peripheral nerve fibre dysfunction due to this mutation may have contributed to distal limb under-development in this novel syndrome.
Collapse
Affiliation(s)
- Janneke G J Hoeijmakers
- Neuroscience and Regeneration Research Centre, VA Connecticut Healthcare System, 950 Campbell Avenue, Building 34, West Haven, CT 06516, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Faber CG, Hoeijmakers JGJ, Ahn HS, Cheng X, Han C, Choi JS, Estacion M, Lauria G, Vanhoutte EK, Gerrits MM, Dib-Hajj S, Drenth JPH, Waxman SG, Merkies ISJ. Gain of function NaV1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol 2011; 71:26-39. [DOI: 10.1002/ana.22485] [Citation(s) in RCA: 394] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 05/10/2011] [Accepted: 05/13/2011] [Indexed: 11/10/2022]
|
32
|
Nardin RA, Fogerson PM, Nie R, Rutkove SB. Foot temperature in healthy individuals: effects of ambient temperature and age. J Am Podiatr Med Assoc 2010; 100:258-64. [PMID: 20660876 DOI: 10.7547/1000258] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Patient complaints of excessively warm or cold feet are common in medical practice. Such symptoms can be caused by underlying vascular or neurologic disease, and measurement of foot temperature during daily activity and sleep could provide a deeper understanding of their actual thermal basis. METHODS We used a Thermochron iButton to assess surface foot temperature variation and its relationship to ambient temperature during the day with activity and at night during sleep in 39 healthy individuals aged 18 to 65 years in a temperate region of the United States. We simultaneously used actigraphy to record leg movement. RESULTS We identified a mean +/- SD awake temperature of 30.6 degrees +/- 2.6 degrees C and asleep temperature of 34.0 degrees +/- 1.8 degrees C, with values reaching as low as 15.9 degrees C in the winter and as high as 37.5 degrees C in the summer. Foot temperature was found to be independent of foot movement or sex; however, there was, as expected, a strong association between foot temperature and ambient temperature (r = .59, P < .001). Several measures of foot temperature variation demonstrated a significant or near-significant reduction with increasing age, including the Euclidean distance (r = -.38, P = .02) for awake periods and the variance (r = -.30, P = .06) during sleep. CONCLUSIONS These results provide data on the normal variation of foot temperature in individuals living in a temperate climate and demonstrate the potential use of Thermochron iButton technology in clinical contexts, including the evaluation of patients with excessively warm or cold feet.
Collapse
Affiliation(s)
- Rachel A Nardin
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
33
|
Fauchais AL, Richard L, Gondran G, Ghorab K, Palat S, Bezanahary H, Loustaud-Ratti V, Ly K, Jauberteau MO, Vallat JM, Vidal E, Magy L. [Small fibre neuropathy in primary Sjögren syndrome]. Rev Med Interne 2010; 32:142-8. [PMID: 20943291 DOI: 10.1016/j.revmed.2010.08.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 07/25/2010] [Accepted: 08/23/2010] [Indexed: 12/16/2022]
Abstract
PURPOSE About forty percent of the patients with primary Sjögren's syndrome (pSS) experience chronic neuropathic pain with normal electrodiagnostic studies. Two previous studies suggest that chronic neuropathic pain in pSS is due to small fiber neuropathy (SFN). Quantification of epidermal nerve fiber density after skin biopsy has been validated to diagnose small fiber neuropathy. METHODS Skin biopsy was performed in 14 consecutive pSS patients (satisfying the american-european classification criteria) with chronic neuropathic pain and normal electrodiagnostic studies suggesting SFN. RESULTS Fourteen female pSS patients exhibited chronic neuropathic pain [burning sensation (n=14), prickling (n=4), dysesthesia (n=8)] with paroxystic exacerbations (n=10) and allodynia (n=13), for a mean period of 18.4±12.4 months. Neuropathic pain involved mostly hands and feet (n=13), with a distal (n=9) and leg (n=4) predominant distribution. Neurological examination disclosed normal deep tendon responses and absence of motor weakness (n=14). Small fiber neuropathy was confirmed by skin biopsy in 13 cases. Epidermal nerve fiber density was decreased in distal [(n=12), mean 3.5±1.7 fibers/mm (N>6.9)] and proximal site of biopsy [(n=9), mean 7.04±2.63 fibers/mm (N>9.3)]. CONCLUSION Small fiber neuropathy is commonly responsible of chronic neuropathic pain in pSS. Prevalence, physiopathology and neurological evolution of such neuropathies still remain unknown.
Collapse
Affiliation(s)
- A-L Fauchais
- EA3842, service de médecine interne, homéostasie cellulaire et pathologies, CHU Dupuytren, faculté de médecine, 2 avenue Martin-Luther-King, Limoges, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sène D, Authier FJ, Amoura Z, Cacoub P, Lefaucheur JP. Neuropathie des petites fibres : approche diagnostique et traitement, et place de son association au syndrome de Gougerot-Sjögren primaire. Rev Med Interne 2010; 31:677-84. [DOI: 10.1016/j.revmed.2010.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 06/28/2010] [Accepted: 07/28/2010] [Indexed: 12/18/2022]
|
35
|
Gemignani F, Giovanelli M, Vitetta F, Santilli D, Bellanova MF, Brindani F, Marbini A. Non-length dependent small fiber neuropathy. A prospective case series. J Peripher Nerv Syst 2010; 15:57-62. [DOI: 10.1111/j.1529-8027.2010.00252.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Donofrio PD, Berger A, Brannagan TH, Bromberg MB, Howard JF, Latov N, Quick A, Tandan R. Consensus statement: The use of intravenous immunoglobulin in the treatment of neuromuscular conditions report of the aanem AD HOC committee. Muscle Nerve 2009; 40:890-900. [DOI: 10.1002/mus.21433] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
37
|
|
38
|
Waldinger MD, Venema PL, Van Gils AP, Schweitzer DH. New Insights into Restless Genital Syndrome: Static Mechanical Hyperesthesia and Neuropathy of the Nervus Dorsalis Clitoridis. J Sex Med 2009; 6:2778-87. [DOI: 10.1111/j.1743-6109.2009.01435.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
39
|
Singleton JR, Bixby B, Russell JW, Feldman EL, Peltier A, Goldstein J, Howard J, Smith AG. The Utah Early Neuropathy Scale: a sensitive clinical scale for early sensory predominant neuropathy. J Peripher Nerv Syst 2008; 13:218-27. [PMID: 18844788 DOI: 10.1111/j.1529-8027.2008.00180.x] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Early neuropathy is often sensory predominant and prominently involves small-diameter nerve fibers. Established neuropathy examination scales such as the Michigan Diabetic Neuropathy Scale (MDNS) and the Neuropathy Impairment Score-Lower Leg (NIS-LL) focus primarily on large-fiber sensory and motor function. Here, we validate the Utah Early Neuropathy Scale (UENS), a physical examination scale specific to early sensory predominant polyneuropathy. Compared with other scales, the UENS emphasizes severity and spatial distribution of pin (sharp) sensation loss in the foot and leg and focuses less on motor weakness. UENS, MDNS, and NIS-LL were compared in 215 diabetic or prediabetic subjects, with (129) or without neuropathy (86), and repeated in 114 neuropathy subjects after 1 year of follow-up. Neuropathy severity was also evaluated with nerve conduction studies, quantitative sensory testing, quantitative sudomotor axonal reflex testing, and intraepidermal nerve fiber density determination. The UENS had a high degree of interrater reliability (interclass correlation of 94%). UENS correlated significantly to MDNS and NIS-LL (p < 0.01), and more significantly than MDNS or NIS-LL to confirmatory tests. In this cohort, UENS had a superior profile to receiver operating characteristic analysis across a range of scores, with a sensitivity (92%) higher than MDNS (67%) or NIS-LL (81%), without sacrificing specificity. UENS more closely correlated with change in ancillary and small-fiber neuropathy measures over 1 year follow-up than did MDNS or NIS-LL. UENS is a sensitive and reproducible clinical measure of sensory and small-fiber nerve injury and may be useful in trials of early neuropathy.
Collapse
Affiliation(s)
- J Robinson Singleton
- Department of Neurology, University of Utah School of Medicine, SOM 3R-152, Salt Lake City, UT 84132, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Obermann M, Katsarava Z, Esser S, Sommer C, He L, Selter L, Yoon MS, Kaube H, Diener HC, Maschke M. Correlation of epidermal nerve fiber density with pain-related evoked potentials in HIV neuropathy. Pain 2008; 138:79-86. [DOI: 10.1016/j.pain.2007.11.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 11/09/2007] [Accepted: 11/15/2007] [Indexed: 11/28/2022]
|
41
|
Abstract
Length-dependent polyneuropathy has long been recognized as a frequent complication of diabetes. A decade of observational and epidemiologic studies support the concept that intermittent hyperglycemia or insulin resistance associated with prediabetes may be sufficient to damage distal nerves. Features of the metabolic syndrome have been implicated as independent neuropathy risk factors in large population-based studies of diabetic patients. Preferential injury to small unmyelinated nerves is suggested by prominent neuropathic pain, predominant sensory injury, and early autonomic dysfunction. Small uncontrolled trials suggest that diet and exercise may transiently improve distal nerve function and neuropathy symptoms in these patients. Patients with prediabetes neuropathy may permit greater insight into the balance between distal axonal injury and nerve regenerative capacity that determines neuropathy progression, and will be good candidates for evaluation of rational therapy based on known pathophysiology of hyperglycemic neuropathy.
Collapse
Affiliation(s)
- J Rob Singleton
- Department of Neurology, University of Utah, 1900 East, 30 North, Salt Lake City, UT 84132, USA.
| | | |
Collapse
|
42
|
Abstract
The occurrence of diabetic peripheral neuropathy (DPN) is linked to poor glycemic control over time. While most people never develop diabetic peripheral neuropathic pain (DPNP) as a consequence of DPN, enough of them do that we must have effective options for the management of this disabling condition. Two years ago there were no formally approved medications for the treatment of DPNP, and now there are two medications with Food and Drug Administration approval for DPNP. One of these medications, duloxetine has been established to significantly improve pain and to address depression by its reuptake inhibition of norepinephrine and serotonin. This article examines the epidemiology of DPNP, its underlying pathogenesis, necessary evaluation methods, and treatment options available with a focus on the role of duloxetine.
Collapse
Affiliation(s)
- B Eliot Cole
- American Society of Pain Educators, Montclair, New Jersey 07042, USA.
| |
Collapse
|
43
|
Rudolph T, Farbu E. Hospital-referred polyneuropathies-causes, prevalences, clinical- and neurophysiological findings. Eur J Neurol 2007; 14:603-8. [PMID: 17539935 DOI: 10.1111/j.1468-1331.2007.01758.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The aim of this study was to evaluate the causes, prevalences, clinical manifestations of hospital-referred polyneuropathies, and evaluate neurophysiological findings in idiopathic polyneuropathy. From 2000 to 2005, 226 patients with polyneuropathy were examined. Polyneuropathy was diagnosed when symptoms, clinical- and neurophysiological findings were compatible with affection of at least two peripheral nerves. They were classified in symptomatic and idiopathic polyneuropathy after investigation. Clinical manifestations were evaluated for diabetes- (DPN), inflammatory- (INPN), hereditary- (HPN) and idiopathic polyneuropathy (IDPN). Neurophysiological findings were investigated in IDPN. 72% had a symptomatic polyneuropathy. Most frequent causes were diabetes mellitus (18%), inflammation, (16%) and hereditary (14%). Most common prevalences per 100,000 were as follows: IDPN, 21; DPN, 13 and HPN, 11. Predominating clinical manifestations were: sensory and motor in INPN, HPN and IPN; sensory in DPN. Pain was more present in IDPN and DPN than in others. In IDPN axonal demyelinating affection was present in 20%. Symptomatic polyneuropathy was common and diabetes mellitus, inflammation and hereditary were frequent causes. In IDPN, DPN, HPN and INPN different clinical patterns were found. Additionally, in IDPN axonal demyelinating affection was more frequent than previously reported.
Collapse
Affiliation(s)
- T Rudolph
- Department of Neurology, Stavanger University Hospital, Stavanger, Norway.
| | | |
Collapse
|
44
|
Oh SJ. Neuropathies of the foot. Clin Neurophysiol 2007; 118:954-80. [PMID: 17336147 DOI: 10.1016/j.clinph.2006.12.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2006] [Revised: 12/21/2006] [Accepted: 12/23/2006] [Indexed: 11/17/2022]
Abstract
Compared with the common neuropathies affecting the hands (carpal tunnel syndrome and ulnar neuropathy), neuropathies of the feet have received less attention in the past. This is partly because of the rarity of these disorders as well as the lack of reliable electrophysiological tests for them. Over the years, nerve conduction tests for various nerves of the feet have been reported, and at this time techniques for all the nerves of the feet are available to the electromyographer. This review will provide up-to-date information on the current status of the research and issues relating to the neuropathies of the foot, with an emphasis on the most useful tests and the caveats for clinical neurophysiologists.
Collapse
Affiliation(s)
- Shin J Oh
- Department of Neurology, University of Alabama at Birmingham, Veterans Affairs Medical Center, Birmingham, AL 35294-0017, USA.
| |
Collapse
|
45
|
Argoff CE, Cole BE, Fishbain DA, Irving GA. Diabetic peripheral neuropathic pain: clinical and quality-of-life issues. Mayo Clin Proc 2006; 81:S3-11. [PMID: 16608048 DOI: 10.1016/s0025-6196(11)61474-2] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Diabetic peripheral neuropathy (DPN) is estimated to be present in 50% of people living with diabetes mellitus (DM). Comorbidities of DM, such as macrovascular and microvascular changes, also Interact with DPN and affect its course. In patients with DM, DPN Is the leading cause of foot ulcers, which in turn are a major cause of amputation in the United States. Although most patients with DPN do not have pain, approximately 11% of patients with DPN have chronic, painful symptoms that diminish quality of life, disrupt sleep, and can lead to depression. Despite the number of patients affected by DPN pain, little consensus exists about the pathophysiology, best diagnostic tools, and primary treatment choices. This article reviews the current knowledge about and presents recommendations for diagnostic assessment of DPN pain based on a review of the literature.
Collapse
Affiliation(s)
- Charles E Argoff
- New York University School of Medicine and Cohn Pain Management Center, Northshore University Hospital, Manhasset, USA
| | | | | | | |
Collapse
|
46
|
Latov N, Gorson KC, Brannagan TH, Freeman RL, Apostolski S, Berger AR, Bradley WG, Briani C, Bril V, Busis NA, Cros DP, Dalakas MC, Donofrio PD, Dyck PJB, England JD, Fisher MA, Herrmann DN, Menkes DL, Sahenk Z, Sander HW, Triggs WJ, Vallat JM. Diagnosis and Treatment of Chronic Immune-mediated Neuropathies. J Clin Neuromuscul Dis 2006; 7:141-157. [PMID: 19078800 DOI: 10.1097/01.cnd.0000205575.26451.e4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The chronic autoimmune neuropathies are a diverse group of disorders, whose diagnosis and classification is based on the clinical presentations and results of ancillary tests. In chronic inflammatory demyelinating polyneuropathy, controlled therapeutic trials demonstrated efficacy for intravenous gamma-globulins, corticosteroids, and plasmaphereis. In multifocal motor neuropathy, intravenous gamma-globulins have been shown to be effective. In the other immune-mediated neuropathies, there are no reported controlled therapeutic trials, but efficacy has been reported for some treatments in non-controlled trials on case studies. Choice of therapy in individual cases is based on reported efficacy, as well as severity, progression, coexisting illness, predisposition to developing complications, and potential drug interactions.
Collapse
|
47
|
Dabby R, Gilad R, Sadeh M, Lampl Y, Watemberg N. Acute steroid responsive small-fiber sensory neuropathy: a new entity? J Peripher Nerv Syst 2006; 11:47-52. [PMID: 16519781 DOI: 10.1111/j.1085-9489.2006.00062.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Small-fiber neuropathy is often idiopathic and commonly follows a chronic course. Treatment is often effective in treating the core symptom of pain, but it has no effect on the pathologic process. We describe four patients with acute small-fiber neuropathy who responded dramatically to steroid therapy. All patients had acute onset neuropathic pain, normal nerve conduction studies, and evidence of small-fiber dysfunction in quantitative sensory testing and skin biopsy. Symptoms were distal and symmetrical in three patients and generalized in one patient. In two cases, the neuropathy presented as an erythromelalgia-like syndrome. Marked clinical improvement occurred 1-2 weeks after oral prednisone therapy was initiated. Three patients remained symptom free, and one patient experienced recurrence of neuropathy after prednisone was tapered.
Collapse
Affiliation(s)
- Ron Dabby
- Department of Neurology, Wolfson Medical Center, Holon, Israel. @netvision.net.il
| | | | | | | | | |
Collapse
|
48
|
Ginsberg L. Chapter 42 Specific painful neuropathies. HANDBOOK OF CLINICAL NEUROLOGY 2006; 81:635-652. [PMID: 18808864 DOI: 10.1016/s0072-9752(06)80046-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
49
|
Sommer C, Lauria G. Chapter 41 Painful small-fiber neuropathies. HANDBOOK OF CLINICAL NEUROLOGY 2006; 81:621-633. [PMID: 18808863 DOI: 10.1016/s0072-9752(06)80045-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
50
|
Lindh J, Tondel M, Osterberg A, Vrethem M. Cryptogenic polyneuropathy: clinical and neurophysiological findings. J Peripher Nerv Syst 2005; 10:31-7. [PMID: 15703016 DOI: 10.1111/j.1085-9489.2005.10106.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The purpose of this study was to describe the clinical and neurophysiological features of cryptogenic polyneuropathy in 168 patients in the neurological departments at three Swedish hospitals. The medical records of all patients aged 40-79 years with the diagnosis of cryptogenic polyneuropathy from 1993 to 2000 were analysed. One hundred and fourteen patients (68%) were men. The mean age at first symptom was 61 years and at diagnosis it was 64 years. Distal numbness (n = 115, 68%) was the most common symptom, but some patients complained of pain, pedal paresthesiae, and impairment of balance. The most common clinical findings were decreased or lost proprioception or sense of vibration (n = 135, 80%) and loss of ankle jerks (n = 131, 78%). Neurography in 139 patients showed mixed sensorimotor polyneuropathy of axonal or mixed axonal and demyelinating type in 97 (70%). Cryptogenic polyneuropathy is a slowly progressive sensorimotor nerve lesion of mainly axonal type. Men are more often affected than women. Most patients have a minor or moderate severe polyneuropathy.
Collapse
Affiliation(s)
- Jonas Lindh
- Section of Neurology, Department of Internal Medicine, Ryhov County Hospital, Jönköping, Sweden.
| | | | | | | |
Collapse
|