1
|
Dalakas MC. Stiff-person syndrome and related disorders - diagnosis, mechanisms and therapies. Nat Rev Neurol 2024; 20:587-601. [PMID: 39227464 DOI: 10.1038/s41582-024-01012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Stiff-person syndrome (SPS) is the prototypical and most common autoimmune neuronal hyperexcitability disorder. It presents with stiffness in the limbs and axial muscles, stiff gait with uncontrolled falls, and episodic painful muscle spasms triggered by anxiety, task-specific phobias and startle responses, collectively leading to disability. Increased awareness of SPS among patients and physicians has created concerns about diagnosis, misdiagnosis and treatment. This Review addresses the evolving diagnostic challenges in SPS and overlapping glutamic acid decarboxylase (GAD) antibody spectrum disorders, highlighting the growing number of overdiagnoses and focusing on the progress made in our understanding of SPS pathophysiology, antibodies against GAD and other inhibitory synaptic antigens, and the fundamentals of neuronal hyperexcitability. It considers the role of impaired GABAergic or glycinergic inhibition in the cortex and at multiple levels in the neuraxis; the underlying autoimmunity and involvement of GAD antibodies; immunopathogenic mechanisms beyond antibodies, including environmental triggers; familial and immunogenetic susceptibility; and potential T cell cytotoxicity. Finally, the mechanistic rationale for target-specific therapeutic interventions is presented along with the available therapeutic approaches, including enhancers of GABA signalling drugs and immunotherapies.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| |
Collapse
|
2
|
Brooks JK, Hanna YA, Al-Mefleh A. Stiff person spectrum disorder: overview with emphasis on head and neck comorbidities. Oral Surg Oral Med Oral Pathol Oral Radiol 2024:S2212-4403(24)00442-5. [PMID: 39317601 DOI: 10.1016/j.oooo.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 09/26/2024]
Abstract
Stiff person spectrum disorder (SPSD) is a rare progressive autoimmune neuromuscular syndrome, primarily resulting in severely painful spasms and rigidity of the axial and appendicular musculature. Affected individuals are predisposed to develop an array of other neuropathies, including cerebellar ataxia and seizure activity, ophthalmologic abnormalities, and other autoimmune-based systemic diseases, notably type 1 diabetes mellitus, thyroiditis, pernicious anemia, and malignancy. Limited information exists in the oral medicine literature regarding SPSD. Thus, the objective of this paper is to review the clinicopathologic features of SPSD, with particular emphasis on head and neck involvement. Additionally, clinical guidelines for dental management of affected individuals and a summary of surgical procedures and outcomes performed in the head and neck are provided. (Oral Surg Oral Med Oral Pathol Oral Radiol YEAR;VOL:page range).
Collapse
Affiliation(s)
- John K Brooks
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, USA.
| | | | - Amer Al-Mefleh
- University of Maryland School of Dentistry, Baltimore, MD, USA
| |
Collapse
|
3
|
Dalakas MC. Stiff Person Syndrome and GAD Antibody-Spectrum Disorders. Continuum (Minneap Minn) 2024; 30:1110-1135. [PMID: 39088290 DOI: 10.1212/con.0000000000001457] [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: 08/03/2024]
Abstract
OBJECTIVE Antibodies against glutamic acid decarboxylase (GAD), originally associated with stiff person syndrome (SPS), define the GAD antibody-spectrum disorders that also include cerebellar ataxia, autoimmune epilepsy, limbic encephalitis, progressive encephalomyelitis with rigidity and myoclonus (PERM), and eye movement disorders, all of which are characterized by autoimmune neuronal excitability. This article elaborates on the diagnostic criteria for SPS and SPS spectrum disorders, highlights disease mimics and misdiagnoses, describes the electrophysiologic mechanisms and underlying autoimmunity of stiffness and spasms, and provides a step-by-step therapeutic scheme. LATEST DEVELOPMENTS Very-high serum GAD antibody titers are diagnostic for GAD antibody-spectrum disorders and also predict the presence of GAD antibodies in the CSF, increased intrathecal synthesis, and reduced CSF γ-aminobutyric acid (GABA) levels. Low serum GAD antibody titers or the absence of antibodies generates diagnostic challenges that require careful distinction in patients with a variety of painful spasms and stiffness, including functional neurologic disorders. Antibodies against glycine receptors, first found in patients with PERM, are seen in 13% to 15% of patients with SPS, whereas amphiphysin and gephyrin antibodies, seen in 5% of patients with SPS spectrum disorders, predict a paraneoplastic association. GAD-IgG from different SPS spectrum disorders recognizes the same dominant GAD intracellular epitope and, although the pathogenicity is unclear, is an excellent diagnostic marker. The biological basis of muscle stiffness and spasms is related to autoimmune neuronal hyperexcitability caused by impaired reciprocal γ-aminobutyric acid-mediated (GABA-ergic) inhibition, which explains the therapeutic response to GABA-enhancing agents and immunotherapies. ESSENTIAL POINTS It is essential to distinguish SPS spectrum disorders from disease mimics to avoid both overdiagnoses and misdiagnoses, considering that SPS is treatable if managed correctly from the outset to prevent disease progression. A step-by-step, combination therapy of GABA-enhancing medications along with immunotherapies ensures prolonged clinical benefits.
Collapse
|
4
|
Patel J, Deschler E, Galang E. Spinal cord stimulation for the symptomatic treatment of rigidity and painful spasm in a case of stiff person syndrome. Pain Pract 2024; 24:798-804. [PMID: 38185725 DOI: 10.1111/papr.13340] [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: 01/09/2024]
Abstract
BACKGROUND Stiff person syndrome (SPS) is a rare neuroimmunological disorder characterized by rigidity and painful spasm primarily affecting the truncal and paraspinal musculature due to autoimmune-mediated neuronal hyperexcitability. Spinal cord stimulation (SCS) is an approved therapy for managing painful neuropathic conditions, including diabetic peripheral neuropathy and refractory angina pectoris. We describe the novel use of SCS for the treatment of spasm and rigidity in a 49-year-old man with seropositive stiff person syndrome (SPS). The patient was treated with intravenous immunoglobulin (IVIG) and oral medications over a 13-month period with minimal improvement, prompting consideration of SCS. To our knowledge, this is the first report of the successful use of SCS in SPS with the demonstration of multifaceted clinical improvement. METHODS Following a successful temporary SCS trial, permanent implantation was performed. Spasm/stiffness (Distribution of Stiffness Index; Heightened Sensitivity Scale; Penn Spasm Frequency Scale, PSFS), disability (Oswestry Disability Index, ODI; Pain Disability Index, PDI), depression (Patient Health Questionnaire-9, PHQ-9), sleep (Pittsburgh Sleep Quality Index, PSQI), fatigue (Fatigue Severity Scale, FSS), pain (Numerical Pain Rating Scale, NPRS), quality of life (EuroQoL 5 Dimension 5 Level, EQ-5D-5L), and medication usage were assessed at baseline, 6-month, and 10-month postimplantation. RESULTS ODI, PHQ-9, FSS, NPRS, PSQI, and EQ-5D-5L scores showed a notable change from baseline and surpassed the defined minimal clinically important difference (MCID) at 6-month and 10-month follow-up. Oral medication dosages were reduced. CONCLUSIONS The novel use of SCS therapy in seropositive SPS resulted in functional improvement and attenuation of symptoms. We present possible mechanisms by which SCS may produce clinical response in patients with SPS and aim to demonstrate proof-of-concept for a future comprehensive pilot study evaluating SCS-mediated response in SPS.
Collapse
Affiliation(s)
- Janus Patel
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Emily Deschler
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Enrique Galang
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
5
|
Celli SI, Nash R, Money KM, Garza M, Borko TL, Mizenko C, McMenamin C, Von Geldern G, Georges G, Piquet AL. Successful Autologous Hematopoietic Stem Cell Transplant in Glycine Receptor Antibody-Positive Stiff Person Syndrome: A Case Report. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200197. [PMID: 38170953 PMCID: PMC10766081 DOI: 10.1212/nxi.0000000000200197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND AND OBJECTIVES To describe a case of glycine receptor (GlyR) antibody-positive stiff person syndrome (SPS) treated with autologous hematopoietic stem cell transplant (aHSCT). METHODS This was a multicenter collaboration for the treatment of a single patient who underwent aHSCT as part of a clinical trial (NCT00716066). To objectively assess the response to transplantation, several clinical outcome measures were evaluated pretransplant and up to 18 months post-transplant, including modified Rankin Score (mRS), stiffness index, Hauser Ambulation Score (HAS), hypersensitivity index, timed 25-foot walk, and Montreal Cognitive Assessment. RESULTS After transplant, the patient achieved sustained clinical improvement evidenced across various clinical scales, including mRS, stiffness index, HAS, and 25-foot walk time. DISCUSSION aHSCT represents a promising treatment option for SPS, including for GlyR-positive patients. In addition, this case represents the need to validate and standardize best clinical outcome measures for patients with SPS. CLASSIFICATION OF EVIDENCE Class IV; this is a single observational study without controls.
Collapse
Affiliation(s)
- Sofia I Celli
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Richard Nash
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Kelli M Money
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Madeline Garza
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Tyler L Borko
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Christopher Mizenko
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Constance McMenamin
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Gloria Von Geldern
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - George Georges
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| | - Amanda L Piquet
- From the Department of Neurology (S.I.C., K.M.M., M.G., T.L.B., C. Mizenko, C. McMenamin, A.L.P.), University of Colorado, Aurora Colorado Blood Cancer Institute (R.N.), Presbyterian St. Luke's Medical Center, Denver, CO; Department of Neurology (G.V.G.); Fred Hutchinson Cancer Center (G.G.), University of Washington, Seattle, WA; and Northwestern University (G.G.), Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
6
|
McMahan ZH, Kulkarni S, Andrade F, Perin J, Zhang C, Hooper JE, Wigley FM, Rosen A, Pasricha PJ, Casciola-Rosen L. Anti-Gephyrin Antibodies: A Novel Specificity in Patients With Systemic Sclerosis and Lower Bowel Dysfunction. Arthritis Rheumatol 2024; 76:92-99. [PMID: 37530745 PMCID: PMC10834854 DOI: 10.1002/art.42667] [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: 09/01/2022] [Revised: 06/01/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE Autoantibodies are clinically useful in phenotyping patients with systemic sclerosis (SSc). Gastrointestinal (GI) function is regulated by the enteric nervous system (ENS) and commonly impaired in SSc, suggesting that the SSc autoimmune response may target ENS antigens. We sought to identify novel anti-ENS autoantibodies with an aim to clinically phenotype SSc GI dysfunction. METHODS Serum from a patient with SSc with GI dysfunction but without defined SSc-associated autoantibodies was used for autoantibody discovery. Immunoprecipitations performed with murine myenteric plexus lysates were on-bead digested, and autoantigens were identified by mass spectrometry. Prevalence was determined, and clinical features associated with novel autoantibodies were evaluated in a SSc cohort using regression analyses. The expression of gephyrin in human GI tract tissue was examined by immunohistochemistry. RESULTS We identified gephyrin as a novel SSc autoantigen. Anti-gephyrin antibodies were present in 9% of patients with SSc (16/188) and absent in healthy controls (0/46). Anti-gephyrin antibody-positive patients had higher constipation scores (1.00 vs 0.50, P = 0.02) and were more likely to have severe constipation and severe distention/bloating (46% vs 15%, P = 0.005; 54% vs 25%, P = 0.023, respectively). Anti-gephyrin antibody levels were significantly higher among patients with severe constipation (0.04 vs 0.00; P = 0.001) and severe distention and bloating (0.03 vs 0.004; P = 0.010). Severe constipation was associated with anti-gephyrin antibodies even in the adjusted model. Importantly, gephyrin was expressed in the ENS, which regulates gut motility. CONCLUSION Gephyrin is a novel ENS autoantigen that is expressed in human myenteric ganglia. Anti-gephyrin autoantibodies are associated with the presence and severity of constipation in patients with SSc.
Collapse
Affiliation(s)
| | - Subhash Kulkarni
- Beth Israel Deaconess Medical Center and Harvard Medical School, Massachusetts, Boston
| | | | - Jamie Perin
- Johns Hopkins University, Maryland, Baltimore
| | | | | | | | | | | | | |
Collapse
|
7
|
Matsui N, Tanaka K, Ishida M, Yamamoto Y, Matsubara Y, Saika R, Iizuka T, Nakamura K, Kuriyama N, Matsui M, Arisawa K, Nakamura Y, Kaji R, Kuwabara S, Izumi Y. Prevalence, Clinical Profiles, and Prognosis of Stiff-Person Syndrome in a Japanese Nationwide Survey. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200165. [PMID: 37739810 PMCID: PMC10519438 DOI: 10.1212/nxi.0000000000200165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/17/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND AND OBJECTIVES To elucidate current epidemiologic, clinical, and immunologic profiles and treatments of stiff-person syndrome (SPS) in Japan. METHODS A nationwide mail survey was conducted using an established method. Data processing sheets were sent to randomly selected departments of internal medicine, neurology, pediatrics, psychiatry, and neurosurgery in hospitals and clinics throughout Japan to identify patients with SPS who were seen between January 2015 and December 2017. RESULTS Thirty cases were identified as glutamic acid decarboxylase 65 (GAD65)-positive SPS cases on the basis of detailed clinical data of 55 cases. Four patients had α1 subunit of glycine receptor (GlyR) antibodies, and 1 patient had both GAD65 and GlyR antibodies. The total estimated number of patients with GAD65-positive SPS was 140, and the estimated prevalence was 0.11 per 100,000 population. The median age at onset was 51 years (range, 26-83 years), and 23 (76%) were female. Of these, 70% had classic SPS, and 30% had stiff-limb syndrome. The median time from symptom onset to diagnosis was significantly longer in the high-titer GAD65 antibody group than in the low-titer group (13 months vs 2.5 months, p = 0.01). The median modified Rankin Scale (mRS) at baseline was 4, and the median mRS at the last follow-up was 2. Among the 29 GAD65-positive patients with ≥1 year follow-up, 7 received only symptomatic treatment, 9 underwent immunotherapy without long-term immunotherapy, and 13 received long-term immunotherapy such as oral prednisolone. The coexistence of type 1 diabetes mellitus and the lack of long-term immunotherapy were independent risk factors for poor outcome (mRS ≥3) in the GAD65-positive patients (odds ratio, 15.0; 95% CI 2.6-131.6; p = 0.001; odds ratio, 19.8; 95% CI 3.2-191.5; p = 0.001, respectively). DISCUSSION This study provides the current epidemiologic and clinical status of SPS in Japan. The symptom onset to the diagnosis of SPS was longer in patients with high-titer GAD65 antibodies than in those with low-titer GAD65 antibodies. The outcome of patients with SPS was generally favorable, but more aggressive immunotherapies are necessary for GAD65-positive patients with SPS.
Collapse
Affiliation(s)
- Naoko Matsui
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Keiko Tanaka
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Mitsuyo Ishida
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Yohei Yamamoto
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Yuri Matsubara
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Reiko Saika
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Takahiro Iizuka
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Koshi Nakamura
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Nagato Kuriyama
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Makoto Matsui
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Kokichi Arisawa
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Yosikazu Nakamura
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Ryuji Kaji
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Satoshi Kuwabara
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| | - Yuishin Izumi
- From the Department of Neurology (N.M., M.I., Y.I.), Tokushima University Graduate School of Biomedical Sciences; Department of Animal Model Development (K.T.), Brain Research Institute, Niigata University; Department of Multiple Sclerosis Therapeutics (K.T.), Fukushima Medical University, School of Medicine; Department of Neurology (Y.Y.), Tokushima University Hospital; Department of Public Health (Y.M., Y.N.), Jichi Medical University, Shimotsuke; Department of Neurology (R.S.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara; Department of Public Health and Hygiene (K.N.), Graduate School of Medicine, University of the Ryukyus, Okinawa; Department of Epidemiology for Community Health and Medicine (N.K.), Kyoto Prefectural University of Medicine; Department of Social Health Medicine (N.K.), Shizuoka Graduate University of Public Health; Department of Neurology (M.M.), Kanazawa Medical University, Ishikawa; Department of Preventive Medicine (K.A.), Tokushima University Graduate School of Biomedical Sciences; National Hospital Organization Utano Hospital (R.K.), Kyoto; and Department of Neurology (S.K.), Graduate School of Medicine, Chiba University, Japan
| |
Collapse
|
8
|
Peng Y, Yang H, Xue YH, Chen Q, Jin H, Liu S, Yao SY, Du MQ. An update on malignant tumor-related stiff person syndrome spectrum disorders: clinical mechanism, treatment, and outcomes. Front Neurol 2023; 14:1209302. [PMID: 37859648 PMCID: PMC10582361 DOI: 10.3389/fneur.2023.1209302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/01/2023] [Indexed: 10/21/2023] Open
Abstract
Stiff person syndrome (SPS) is a rare central nervous system disorder associated with malignancies. In this review, we retrieved information from PubMed, up until August 2023, using various search terms and their combinations, including SPS, stiff person syndrome spectrum disorders (SPSSDs), paraneoplastic, cancer, and malignant tumor. Data from peer-reviewed journals printed in English were organized to explain the possible relationships between different carcinomas and SPSSD subtypes, as well as related autoantigens. From literature searching, it was revealed that breast cancer was the most prevalent carcinoma linked to SPSSDs, followed by lung cancer and lymphoma. Furthermore, classic SPS was the most common SPSSD subtype, followed by stiff limb syndrome and progressive encephalomyelitis with rigidity and myoclonus. GAD65 was the most common autoantigen in patients with cancer and SPSSDs, followed by amphiphysin and GlyR. Patients with cancer subtypes might have multiple SPSSD subtypes, and conversely, patients with SPSSD subtypes might have multiple carcinoma subtypes. The first aim of this review was to highlight the complex nature of the relationships among cancers, autoantigens, and SPSSDs as new information in this field continues to be generated globally. The adoption of an open-minded approach to updating information on new cancer subtypes, autoantigens, and SPSSDs is recommended to renew our database. The second aim of this review was to discuss SPS animal models, which will help us to understand the mechanisms underlying the pathogenesis of SPS. In future, elucidating the relationship among cancers, autoantigens, and SPSSDs is critical for the early prediction of cancer and discovery of new therapeutic modalities.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-hui Xue
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Quan Chen
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Hong Jin
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Shu Liu
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Shun-yu Yao
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Miao-qiao Du
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| |
Collapse
|
9
|
Papadopoulos VE, Papadimas GK, Androudi S, Anagnostouli M, Evangelopoulos ME. Stiff-Leg Syndrome Associated with Autoimmune Retinopathy and Its Treatment with IVIg-A Case Report and Review of the Literature. Brain Sci 2023; 13:1361. [PMID: 37891730 PMCID: PMC10605544 DOI: 10.3390/brainsci13101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Antibodies to glutamic acid decarboxylase (GAD) have been predominantly associated with stiff-person syndrome (SPS), which is often accompanied by organ-specific autoimmune diseases, such as late-onset type 1 diabetes. Autoimmune retinal pathology in SPS has recently been suggested to coexist in patients suffering from this disease; however, evidence reporting potential treatment options for the neurological and visual symptoms these patients experience remains scarce. We provide a review of the relevant literature, presenting a rare case of a middle-aged woman with autoimmune retinopathy (AIR) followed by stiff-leg syndrome who responded to intravenous immune globulin treatment (IVIg). Our report adds to previously reported data supporting the efficacy of IVIg in SPS spectrum disorders while also proposing the potential effect of IVIg in treating SPS spectrum patients with coexisting AIR.
Collapse
Affiliation(s)
- Vassilis E Papadopoulos
- First Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - George K Papadimas
- First Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Sofia Androudi
- Department of Ophthalmology, University of Thessaly, 41110 Larissa, Greece
| | - Maria Anagnostouli
- First Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Maria-Eleftheria Evangelopoulos
- First Department of Neurology, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
10
|
Aryal R, Shrestha S, Homagain S, Tiwari SB, Ojha R. Intravenous immunoglobulin in the management and outcome of Stiff‐Person syndrome: A systematic review. CLINICAL AND EXPERIMENTAL NEUROIMMUNOLOGY 2022; 13:302-315. [DOI: 10.1111/cen3.12713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/16/2022] [Indexed: 10/02/2024]
Abstract
AbstractBackgroundStiff Person Syndrome (SPS), a rare immune‐mediated neuromuscular disorder, is characterized by rigidity, stiffness, and intermittent spasms of axial and extremity muscles. Various immunotherapies including intravenous immunoglobulin (IVIG) have been used for this autoimmune condition. Here we aim to review the role of IVIG in the treatment of SPS along with its outcome.MethodsA systematic literature search of PubMed and Embase was conducted to identify the relevant published articles against the predefined criteria using suitable keywords combinations till September 20, 2021. Data were extracted to produce descriptive information of SPS patients on demographics, diagnostics, treatment with IVIG, and outcome.ResultsTwelve studies with 216 patients were included in the review and 63.89% of them had classical SPS. Glutamic acid decarboxylase (GAD) autoantibodies were present in 72.68% of the patients and 57.89% in whom electromyography (EMG) was performed had continuous motor activity. IVIG therapy was given to 95 patients in different regimens in various studies and varying scoring systems were used to assess the outcome, and 83.16% showed some form of improvement, 14.74% showed no improvement, while 2.10% worsened. None of the included studies mentioned an adverse effect of IVIG in the patients.ConclusionIVIG may benefit patients with SPS along with other medications. Owing to the rarity of the disease and insufficient studies on the assessment of immunotherapy in SPS, longitudinal studies with a sizable number of patients are required to clarify clinical course, treatment, and outcome in SPS with the use of IVIG.
Collapse
Affiliation(s)
- Roshan Aryal
- Maharajgunj Medical Campus Tribhuvan University Institute of Medicine Kathmandu Nepal
| | - Suraj Shrestha
- Maharajgunj Medical Campus Tribhuvan University Institute of Medicine Kathmandu Nepal
| | - Sushan Homagain
- Maharajgunj Medical Campus Tribhuvan University Institute of Medicine Kathmandu Nepal
| | - Sansar Babu Tiwari
- Department of Pathology Tribhuvan University Teaching Hospital Kathmandu Nepal
| | - Rajeev Ojha
- Department of Neurology Tribhuvan University Teaching Hospital Kathmandu Nepal
| |
Collapse
|
11
|
Newsome SD, Johnson T. Stiff person syndrome spectrum disorders; more than meets the eye. J Neuroimmunol 2022; 369:577915. [PMID: 35717735 PMCID: PMC9274902 DOI: 10.1016/j.jneuroim.2022.577915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 05/21/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
Stiff person syndrome spectrum disorders (SPSD) are a group of rare neuroimmunological disorders that often include painful spasms and rigidity. However, patients have highly heterogeneous signs and symptoms which may reflect different mechanistic disease processes. Understanding subsets of patients based on clinical phenotype may be important for prognosis and guiding treatment. The goal of this review is to provide updates on SPSD and its expanding clinical spectrum, prognostic markers, and treatment considerations. Further, we describe the current understanding in immunopathogenesis and highlight gaps in our knowledge appropriate for future research directions. Examples of revised diagnostic criteria for SPSD based on phenotype are also presented.
Collapse
Affiliation(s)
- Scott D Newsome
- Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Tory Johnson
- Johns Hopkins University School of Medicine, Baltimore, MD, USA; Section of Infections of the Nervous System, NINDS, NIH, Bethesda, MD, USA
| |
Collapse
|
12
|
Nie DA, Abud A, Serrano-Gonzalez M, Harrapannahally G, Patil R. Pediatric stiff limb syndrome with polyautoimmunity of anti-GAD-65, anti-islet cell, and thyroid peroxidase antibodies: A case report and review of literature. J Neuroimmunol 2022; 367:577865. [DOI: 10.1016/j.jneuroim.2022.577865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/27/2022] [Accepted: 04/08/2022] [Indexed: 12/26/2022]
|
13
|
Dalakas MC. Stiff-person Syndrome and GAD Antibody-spectrum Disorders: GABAergic Neuronal Excitability, Immunopathogenesis and Update on Antibody Therapies. Neurotherapeutics 2022; 19:832-847. [PMID: 35084720 PMCID: PMC9294130 DOI: 10.1007/s13311-022-01188-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2022] [Indexed: 01/10/2023] Open
Abstract
Although antibodies against Glutamic Acid Decarboxylase (GAD) were originally associated with Stiff Person Syndrome (SPS), they now denote the "GAD antibody-spectrum disorders (GAD-SD)" that include Cerebellar Ataxia, Autoimmune Epilepsy, Limbic Encephalitis, PERM and eye movement disorder. In spite of the unique clinical phenotype that each of these disorders has, there is significant overlapping symptomatology characterized by autoimmune neuronal excitability. In addition to GAD, three other autoantibodies, against glycine receptors, amphiphysin and gephyrin, are less frequently or rarely associated with SPS-SD. Very high serum anti-GAD antibody titers are a key diagnostic feature for all GAD-SD, commonly associated with the presence of GAD antibodies in the CSF, a reduced CSF GABA level and increased anti-GAD-specific IgG intrathecal synthesis denoting stimulation of B-cell clones in the CNS. Because anti-GAD antibodies from the various hyperexcitability syndromes recognize the same dominant GAD epitope, the clinical heterogeneity among GAD-SD patients remains unexplained. The paper highlights the biologic basis of autoimmune hyperexcitability connected with the phenomenon of reciprocal inhibition as the fundamental mechanism of the patients' muscle stiffness and spasms; addresses the importance of high-GAD antibody titers in diagnosis, pinpointing the diagnostic challenges in patients with low-GAD titers or their distinction from functional disorders; and discusses whether high GAD-antibodies are disease markers or pathogenic in the context of their association with reduced GABA level in the brain and CSF. Finally, it focuses on therapies providing details on symptomatic GABA-enhancing drugs and the currently available immunotherapies in a step-by-step approach. The prospects of future immunotherapeutic options with antibody therapies are also summarized.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit National and Kapodistrian University of Athens Medical School, Athens, Greece.
| |
Collapse
|
14
|
Treatment and Management of Disorders of Neuromuscular Hyperexcitability and Periodic Paralysis. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00018-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
15
|
Czempik PF, Gawryluk J, Wiórek A, Krzystanek E, Krzych ŁJ. Efficacy and safety of therapeutic plasma exchange in stiff person syndrome. Open Med (Wars) 2021; 16:526-531. [PMID: 33821220 PMCID: PMC8010154 DOI: 10.1515/med-2021-0220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/20/2020] [Accepted: 01/08/2021] [Indexed: 12/29/2022] Open
Abstract
The stiff person syndrome (SPS) is an extremely rare neurological disorder with primarily immune-mediated etiology. The cardinal symptoms are progressive, fluctuating axial/proximal limb muscle stiffness and spasms. The diagnosis is based on the clinical picture, electromyography examination and detection of antibodies to glutamic acid decarboxylase (anti-GAD). Adverse effects of medications might preclude its use or increase in dosing, therefore symptomatic and/or immunomodulatory medical therapy might be ineffective in acute exacerbation of the disease. We present a case of a 49-year-old female with exacerbation of SPS, in whom some standard pharmacotherapy could not be introduced (clonazepam, baclofen used in the past) and doses of existing standard medications could not be increased (diazepam, gabapentin, and levetiracetam) due to adverse effects. Moreover, a newly introduced medication (methylprednisolone) also led to a serious adverse effect (severe hyperglycemia). The patient underwent therapeutic plasma exchange (TPE) with good effect and no complications. We review the literature regarding the efficacy and safety profile of TPE in exacerbation of SPS unresponsive to medical therapy. The procedure seems to have a good safety profile as an adjunct therapy for exacerbation of SPS not responding to standard medical therapy in this patient population.
Collapse
Affiliation(s)
- Piotr F Czempik
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, Katowice, 40-752, Poland
| | - Justyna Gawryluk
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, Katowice, 40-752, Poland
| | - Agnieszka Wiórek
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, Katowice, 40-752, Poland
| | - Ewa Krzystanek
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, Katowice, 40-752, Poland
| | - Łukasz J Krzych
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, Katowice, 40-752, Poland
| |
Collapse
|
16
|
Tsiortou P, Alexopoulos H, Dalakas MC. GAD antibody-spectrum disorders: progress in clinical phenotypes, immunopathogenesis and therapeutic interventions. Ther Adv Neurol Disord 2021; 14:17562864211003486. [PMID: 33854562 PMCID: PMC8013924 DOI: 10.1177/17562864211003486] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 02/27/2021] [Indexed: 12/14/2022] Open
Abstract
Antibodies against glutamic acid decarboxylase (GAD), originally linked to stiff person syndrome (SPS), now denote the "GAD antibody-spectrum disorders" (GAD-SD) that also include autoimmune epilepsy, limbic encephalitis, cerebellar ataxia and nystagmus with overlapping symptomatology highlighting autoimmune neuronal excitability disorders. The reasons for the clinical heterogeneity among GAD-antibody associated syndromes remain still unsettled, implicating variable susceptibility of GABAergic neurons to anti-GAD or other still unidentified autoantibodies. Although anti-GAD antibody titers do not correlate with clinical severity, very high serum titers, often associated with intrathecal synthesis of anti-GAD-specific IgG, point to in-situ effects of GAD or related autoantibodies within the central nervous system. It remains, however, uncertain what drives these antibodies, why they persist and whether they are disease markers or have pathogenic potential. The review, focused on these concerns, describes the widened clinical manifestations and overlapping features of all GAD-SD; addresses the importance of GAD antibody titers and potential significance of GAD epitopes; summarizes the biologic basis of autoimmune hyperexcitability; highlights the electrophysiological basis of reciprocal inhibition in muscle stiffness; and provides practical guidelines on symptomatic therapies with gamma-aminobutyric acid-enhancing drugs or various immunotherapies.
Collapse
Affiliation(s)
- Popianna Tsiortou
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Harry Alexopoulos
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA; Neuroimmunology Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
17
|
Xie YY, Meng HM, Zhang FX, Maimaiti B, Jiang T, Yang Y. Involuntary movement in stiff-person syndrome with amphiphysin antibodies: A case report. Medicine (Baltimore) 2021; 100:e24312. [PMID: 33546061 PMCID: PMC7837982 DOI: 10.1097/md.0000000000024312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/24/2020] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Stiff-person syndrome (SPS) is a rare neurological immune disorder characterized by progressive axial and proximal limb muscle rigidity, stiffness, and painful muscle spasms. Amphiphysin antibodies are positive in approximately 5% of SPS patients. To date, there have been no relevant reports on involuntary movement in cases of SPS with amphiphysin antibodies. PATIENT CONCERNS We describe the case of a 69-year-old man with a 2-year history of progressive stiffness in the neck, bilateral shoulders, and chest muscles, and a more-than-a-year history of dyspnea accompanied by mandibular involuntary movement. The patient was a vegetarian and had good health in the past. The family's medical history was unremarkable. DIAGNOSES He was diagnosed with SPS based on the progressive muscle stiffness, the amphiphysin antibody seropositivity, the continuous motor activity on electromyography, and the effective treatment with benzodiazepines. INTERVENTIONS The patient was orally administered clonazepam and baclofen, and corticosteroid IV followed by prednisone orally. OUTCOMES In the hospital, after treatment with methylprednisolone, clonazepam, and baclofen, the patient's rigidity, stiffness, and dyspnea significantly improved. The involuntary movement of the mandible persisted throughout the treatment process. Currently, under oral treatment with baclofen and clonazepam, the patient's symptoms of muscle stiffness and dyspnea exist, and follow-up is continued. LESSONS We report a rare and novel case of involuntary movement in SPS with amphiphysin antibodies. The present report explores the relationship between SPS and involuntary movement and expands the spectrum of clinical manifestations of SPS.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Yang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
18
|
Hyperkinetic Movement Disorders. Neurology 2021. [DOI: 10.1007/978-3-030-55598-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
19
|
Benning L, Reinehr S, Grotegut P, Kuehn S, Stute G, Dick HB, Joachim SC. Synapse and Receptor Alterations in Two Different S100B-Induced Glaucoma-Like Models. Int J Mol Sci 2020; 21:ijms21196998. [PMID: 32977518 PMCID: PMC7583988 DOI: 10.3390/ijms21196998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/20/2020] [Indexed: 11/03/2022] Open
Abstract
Glaucoma is identified by an irreversible retinal ganglion cell (RGC) loss and optic nerve damage. Over the past few years, the immune system gained importance in its genesis. In a glaucoma-like animal model with intraocular S100B injection, RGC death occurs at 14 days. In an experimental autoimmune glaucoma model with systemic S100B immunization, a loss of RGCs is accompanied by a decreased synaptic signal at 28 days. Here, we aimed to study synaptic alterations in these two models. In one group, rats received a systemic S100B immunization (n = 7/group), while in the other group, S100B was injected intraocularly (n = 6–7/group). Both groups were compared to appropriate controls and investigated after 14 days. While inhibitory post-synapses remained unchanged in both models, excitatory post-synapses degenerated in animals with intraocular S100B injection (p = 0.03). Excitatory pre-synapses tendentially increased in animals with systemic S100B immunization (p = 0.08) and significantly decreased in intraocular ones (p = 0.04). Significantly more N-methyl-d-aspartate (NMDA) receptors (both p ≤ 0.04) as well as gamma-aminobutyric acid (GABA) receptors (both p < 0.03) were observed in S100B animals in both models. We assume that an upregulation of these receptors causes the interacting synapse types to degenerate. Heightened levels of excitatory pre-synapses could be explained by remodeling followed by degeneration.
Collapse
|
20
|
Xu M, Bennett DLH, Querol LA, Wu LJ, Irani SR, Watson JC, Pittock SJ, Klein CJ. Pain and the immune system: emerging concepts of IgG-mediated autoimmune pain and immunotherapies. J Neurol Neurosurg Psychiatry 2020; 91:177-188. [PMID: 30224548 DOI: 10.1136/jnnp-2018-318556] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/18/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022]
Abstract
The immune system has long been recognised important in pain regulation through inflammatory cytokine modulation of peripheral nociceptive fibres. Recently, cytokine interactions in brain and spinal cord glia as well as dorsal root ganglia satellite glia have been identified important- in pain modulation. The result of these interactions is central and peripheral sensitisation of nociceptive processing. Additionally, new insights and the term 'autoimmune pain' have emerged through discovery of specific IgGs targeting the extracellular domains of antigens at nodal and synaptic structures, causing pain directly without inflammation by enhancing neuronal excitability. Other discovered IgGs heighten pain indirectly by T-cell-mediated inflammation or destruction of targets within the nociceptive pathways. Notable identified IgGs in pain include those against the components of channels and receptors involved in inhibitory or excitatory somatosensory synapses or their pathways: nodal and paranodal proteins (LGI1, CASPR1, CASPR2); glutamate detection (AMPA-R); GABA regulation and release (GAD65, amphiphysin); glycine receptors (GLY-R); water channels (AQP4). These disorders have other neurological manifestations of central/peripheral hyperexcitabability including seizures, encephalopathy, myoclonus, tremor and spasticity, with immunotherapy responsiveness. Other pain disorders, like complex regional pain disorder, have been associated with IgGs against β2-adrenergic receptor, muscarinic-2 receptors, AChR-nicotinic ganglionic α-3 receptors and calcium channels (N and P/Q types), but less consistently with immune treatment response. Here, we outline how the immune system contributes to development and regulation of pain, review specific IgG-mediated pain disorders and summarise recent development in therapy approaches. Biological agents to treat pain (anti-calcitonin gene-related peptide and anti-nerve growth factor) are also discussed.
Collapse
Affiliation(s)
- Min Xu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Neurology, Xuan wu Hospital Capital Medical University, Beijing, China
| | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Luis Antonio Querol
- Neuromuscular Diseases Unit-Neuromuscular Lab Neurology Department, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarosh R Irani
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - James C Watson
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Pain Anesthesiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sean J Pittock
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher J Klein
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA .,Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
21
|
Piquet AL, Khan M, Warner JEA, Wicklund MP, Bennett JL, Leehey MA, Seeberger L, Schreiner TL, Paz Soldan MM, Clardy SL. Novel clinical features of glycine receptor antibody syndrome: A series of 17 cases. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e592. [PMID: 31355325 PMCID: PMC6624144 DOI: 10.1212/nxi.0000000000000592] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/16/2019] [Indexed: 12/30/2022]
Abstract
Objective To describe novel clinical features of GlyRα1-IgG-positive patients. Methods Patients with a positive serum GlyRα1-IgG were identified during a 2-year period from July 2016 to December 2018 at 2 academic centers and followed prospectively. All patients in this series were evaluated in the Neuroimmunology and Autoimmune Neurology clinics at the University of Utah or the University of Colorado. Results Thirteen of 17 patients had phenotypes more typically associated with glutamic acid decarboxylase (GAD65) antibody syndromes, consisting of stiff-person syndrome (SPS) with parkinsonism or cerebellar signs. One patient with parkinsonism had a presentation similar to rapidly progressive multiple system atrophy with severe dysautonomia. Ten of 17 patients had various visual symptoms including visual snow, spider web-like images forming shapes and 3-dimensional images, palinopsia, photophobia, visual hallucinations, synesthesia, and intermittent diplopia. Three of 17 patients presented with primarily autoimmune epilepsy accompanied by psychiatric symptoms. Conclusions Clinicians should consider testing for GlyR antibodies in GAD65 antibody-negative or low-positive GAD65 antibody patients with SPS-like presentations, especially in the setting of atypical features such as visual disturbances, parkinsonism, or epilepsy.
Collapse
Affiliation(s)
- Amanda L Piquet
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Murtaza Khan
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Judith E A Warner
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Matthew P Wicklund
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Jeffrey L Bennett
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Maureen A Leehey
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Lauren Seeberger
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Teri L Schreiner
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - M Mateo Paz Soldan
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Stacey L Clardy
- Department of Neurology (A.L.P., M.K., M.P.W., J.L.B., M.A.L., L.S., T.L.S.), University of Colorado, Aurora; Department of Neurology (A.L.P., J.E.A.W., M.M.P.S., S.L.C.), University of Utah; Department of Ophthalmology (J.E.A.W.), Moran Eye Center, University of Utah, Salt Lake City; Department of Ophthalmology and Program in Neuroscience (J.L.B.), University of Colorado; Department of Neurology (T.L.S.), Children's Hospital Colorado, Aurora; and Department of Veterans Affairs (M.M.P.S., S.L.C.), George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| |
Collapse
|
22
|
El-Abassi R, Soliman MY, Villemarette-Pittman N, England JD. SPS: Understanding the complexity. J Neurol Sci 2019; 404:137-149. [PMID: 31377632 DOI: 10.1016/j.jns.2019.06.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 05/31/2019] [Accepted: 06/17/2019] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Stiff-person syndrome (SPS), first described in 1956 by Moersch and Woltman, is a progressive autoimmune disorder with core features of chronic fluctuating progressive truncal and limb rigidity and painful muscle spasms leading to gait difficulties, falls and an appearance that resembles tin soldiers. The syndrome is a rare, highly disabling disorder of the central nervous and frequently results in significant disability. Understanding of the etiology, clinical spectrum, diagnostic workup and therapeutic modalities for this painful and disabling disorder has vastly evolved over the past few years with more confidence in classifying and treating the patients. The purpose of this review is to increase the awareness, early detection, and treatment of this disabling disease. METHOD PubMed was searched, all date inclusive, using the following phrases: stiff person syndrome,anti-Glutamic acid decarboxylase (Anti-GAD) antibody syndrome, Progressive encephalomyelitis with rigidity and myoclonus (PERM), and Paraneoplastic Stiff Person syndrome. No filters or restrictions were used. A total of 888 articles were identified. RESULTS The results were narrowed to 190 citations after excluding non-English and duplicate reports. Clinical presentation, laboratory testing, treatment, and prognosis were categorized and summarized. DISCUSSION In this article we will discuss the epidemiology, presentation and classification. Explain the pathophysiology of SPS and the autoimmune mechanisms involved. Discuss the diagnostic approach and treatments available, as well as, the prognosis and outcome.
Collapse
Affiliation(s)
- Rima El-Abassi
- Department of Neurology, Louisiana State University School of medicine, New Orleans, USA.
| | - Michael Y Soliman
- Department of Neurology, Louisiana State University School of medicine, New Orleans, USA
| | | | - John D England
- Department of Neurology, Louisiana State University School of medicine, New Orleans, USA
| |
Collapse
|
23
|
Baizabal-Carvallo JF. The neurological syndromes associated with glutamic acid decarboxylase antibodies. J Autoimmun 2019; 101:35-47. [DOI: 10.1016/j.jaut.2019.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/07/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
|
24
|
Zhang X, Cheng D, Liu Y, Wu Y, He Z. Gephyrin suppresses lung squamous cell carcinoma development by reducing mTOR pathway activation. Cancer Manag Res 2019; 11:5333-5341. [PMID: 31239782 PMCID: PMC6560210 DOI: 10.2147/cmar.s204358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/24/2019] [Indexed: 11/23/2022] Open
Abstract
Background: The mTOR pathway is altered in a multitude of cancers, including lung cancer; however, abnormal activation in this pathway is less common in lung adenocarcinoma (LUAD) than in lung squamous cell carcinoma (LUSC). Gephyrin is a highly conserved and widely expressed ancient protein in vertebrate tissues. Its role and molecular mechanism in lung cancer development are largely unknown. Method: We analyzed the expression profile of gephyrin and overall survival rates in LUAD and LUSC. The LUSC cells (H520 and SK-MES-1) were transfected with pLV-gephyrin to establish gephyrin stable overexpression cell lines. Real-time quantitative PCR and Western blot were performed to detect the mRNA and protein levels. The cell growth and cell cycle were detected by the MTT assay and flow cytometry. Finally, a xenograft tumor model was established to determine cell tumorigenesis in vivo. Results: Our results show that gephyrin was reduced in LUAD and LUSC, and its low expression in LUSC patients indicated poor prognosis. Gephyrin overexpression suppressed LUSC cell proliferation, arrested cell cycle progression, and decreased the expression of cell-cycle related proteins such as cyclin D1, cyclin-dependent kinase-2 (CDK2), and proliferation-related protein proliferating cell nuclear antigen (PCNA). Conversely, knockdown of gephyrin promoted LUSC cell growth. Moreover, gephyrin reduced mTOR pathway activation to inhibit cyclin D1 and CDK2 translation. Mechanistically, gephyrin suppressed mTOR pathway activation by promoting mTOR degradation. Furthermore, gephyrin overexpression suppressed LUSC tumorigenesis. Conclusion: Gephyrin suppressed LUSC development by reducing mTOR pathway activation, implicating gephyrin as a potential molecular target for LUSC management.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Dezhi Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yuanbo Wu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Zhifeng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| |
Collapse
|
25
|
Gata-Garcia A, Diamond B. Maternal Antibody and ASD: Clinical Data and Animal Models. Front Immunol 2019; 10:1129. [PMID: 31191521 PMCID: PMC6547809 DOI: 10.3389/fimmu.2019.01129] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/03/2019] [Indexed: 12/26/2022] Open
Abstract
Over the past several decades there has been an increasing interest in the role of environmental factors in the etiology of neuropsychiatric and neurodevelopmental disorders. Epidemiologic studies have shifted from an exclusive focus on the identification of genetic risk alleles for such disorders to recognizing and understanding the contribution of xenobiotic exposures, infections, and the maternal immune system during the prenatal and early post-natal periods. In this review we discuss the growing literature regarding the effects of maternal brain-reactive antibodies on fetal brain development and their contribution to the development of neuropsychiatric and neurodevelopmental disorders. Autoimmune diseases primarily affect women and are more prevalent in mothers of children with neurodevelopmental disorders. For example, mothers of children with Autism Spectrum Disorder (ASD) are significantly more likely to have an autoimmune disease than women of neurotypically developing children. Moreover, they are four to five times more likely to harbor brain-reactive antibodies than unselected women of childbearing age. Many of these women exhibit no apparent clinical consequence of harboring these antibodies, presumably because the antibodies never access brain tissue. Nevertheless, these maternal brain-reactive antibodies can access the fetal brain, and some may be capable of altering brain development when present during pregnancy. Several animal models have provided evidence that in utero exposure to maternal brain-reactive antibodies can permanently alter brain anatomy and cause persistent behavioral or cognitive phenotypes. Although this evidence supports a contribution of maternal brain-reactive antibodies to neurodevelopmental disorders, an interplay between antibodies, genetics, and other environmental factors is likely to determine the specific neurodevelopmental phenotypes and their severity. Additional modulating factors likely also include the microbiome, sex chromosomes, and gonadal hormones. These interactions may help to explain the sex-bias observed in neurodevelopmental disorders. Studies on this topic provide a unique opportunity to learn how to identify and protect at risk pregnancies while also deciphering critical pathways in neurodevelopment.
Collapse
Affiliation(s)
- Adriana Gata-Garcia
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
26
|
Transfer of the Experimental Autoimmune Glaucoma Model from Rats to Mice-New Options to Study Glaucoma Disease. Int J Mol Sci 2019; 20:ijms20102563. [PMID: 31137749 PMCID: PMC6566658 DOI: 10.3390/ijms20102563] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/13/2019] [Accepted: 05/21/2019] [Indexed: 12/25/2022] Open
Abstract
Studies have suggested an involvement of the immune system in glaucoma. Hence, a rat experimental autoimmune glaucoma model (EAG) was developed to investigate the role of the immune response. Here, we transferred this model into mice. Either 0.8 mg/mL of the optic nerve antigen homogenate (ONA; ONA 0.8) or 1.0 mg/mL ONA (ONA 1.0) were injected in 129/Sv mice. Controls received sodium chloride. Before and 6 weeks after immunization, the intraocular pressure (IOP) was measured. At 6 weeks, retinal neurons, glia cells, and synapses were analyzed via immunohistology and quantitative real-time PCR (RT-qPCR). Additionally, optic nerves were examined. The IOP stayed in the normal physiological range throughout the study (p > 0.05). A significant reduction of retinal ganglion cells (RGCs) was noted in both immunized groups (p < 0.001). Remodeling of glutamatergic and GABAergic synapses was seen in ONA 1.0 retinas. Furthermore, both ONA groups revealed optic nerve degeneration and macrogliosis (all: p < 0.001). An increase of activated microglia was noted in ONA retinas and optic nerves (p < 0.05). Both ONA concentrations led to RGC loss and optic nerve degeneration. Therefore, the EAG model was successfully transferred from rats to mice. In further studies, transgenic knockout mice can be used to investigate the pathomechanisms of glaucoma more precisely.
Collapse
|
27
|
Kasaragod VB, Hausrat TJ, Schaefer N, Kuhn M, Christensen NR, Tessmer I, Maric HM, Madsen KL, Sotriffer C, Villmann C, Kneussel M, Schindelin H. Elucidating the Molecular Basis for Inhibitory Neurotransmission Regulation by Artemisinins. Neuron 2019; 101:673-689.e11. [DOI: 10.1016/j.neuron.2019.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 10/11/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023]
|
28
|
Ozaki K, Ohkubo T, Yamada T, Yoshioka K, Ichijo M, Majima T, Kudo S, Akashi T, Honda K, Ito E, Watanabe M, Sekine M, Hamagaki M, Eishi Y, Sanjo N, Ishibashi S, Mizusawa H, Yokota T. Progressive Encephalomyelitis with Rigidity and Myoclonus Resolving after Thymectomy with Subsequent Anasarca: An Autopsy Case. Intern Med 2018; 57:3451-3458. [PMID: 29984771 PMCID: PMC6306531 DOI: 10.2169/internalmedicine.1238-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Progressive encephalomyelitis with rigidity and myoclonus (PERM) is an autoimmune disorder involving the brainstem and spinal cord and is sometimes associated with thymoma. We encountered a 75-year-old woman with typical PERM features, glycine receptor antibody, and thymoma. Her neurologic symptoms improved after thymectomy, but she unexpectedly developed anasarca with massive pleural effusions and hypoalbuminemia and finally succumbed to death. The autopsy showed edema and mononuclear infiltration in the pleura but no neuropathological findings typical of PERM. Effective treatment of PERM can reverse the neuropathological signs of encephalomyelitis. The autoimmune nature of anasarca is possible but not proven.
Collapse
Affiliation(s)
- Kokoro Ozaki
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Takuya Ohkubo
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Tetsuo Yamada
- Department of Clinical Laboratory Medicine, Graduate School of Health Care Science, Bunkyo Gakuin University, Japan
| | - Kotaro Yoshioka
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Masahiko Ichijo
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Takamasa Majima
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Shunsuke Kudo
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Takumi Akashi
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Keiji Honda
- Department of Otolaryngology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Eisaku Ito
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Mayumi Watanabe
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Masaki Sekine
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Miwako Hamagaki
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Yoshinobu Eishi
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Nobuo Sanjo
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Satoru Ishibashi
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | - Hidehiro Mizusawa
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
- The National Center Hospital, National Center of Neurology and Psychiatry, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| |
Collapse
|
29
|
Galli JR, Austin SD, Greenlee JE, Clardy SL. Stiff person syndrome with Anti-GAD65 antibodies within the national veterans affairs health administration. Muscle Nerve 2018; 58:801-804. [PMID: 30192027 DOI: 10.1002/mus.26338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/26/2018] [Accepted: 09/01/2018] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Stiff person syndrome (SPS) is a neurological disorder characterized by muscle rigidity primarily in the truncal muscles, commonly associated with autoantibodies to the glutamic acid-decarboxylase 65 kD receptor (GAD65). There is limited epidemiological information on patients with SPS. METHODS We performed a retrospective case review using the National United States Veterans Affairs Health Administration electronic medical record system. We analyzed prevalence, demographics, disease characteristics, and treatment outcomes in SPS patients who were anti-GAD65 antibody positive. RESULTS Fifteen patients met our criteria. Point prevalence was 2.06 per million, and period prevalence was 2.71 per million. Men to women ratio was 14:1. All patients benefitted from treatment with symptomatic antispasmodic agents. Ten of 15 patients received intravenous immunoglobulin, with a majority demonstrating stable or improved modified Rankin scores. DISCUSSION This investigation was a large North American epidemiological study of SPS with predominantly male patients. Symptomatic therapy was beneficial for most patients, with less clear sustained benefit of immunotherapy. Muscle Nerve 58:801-804, 2018.
Collapse
Affiliation(s)
- Jonathan R Galli
- Department of Neurology, University of Utah, Imaging and Neurosciences Center, 729 Arapeen Drive, Salt Lake City, Utah, 84108, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Sharon D Austin
- Department of Neurology, University of Utah, Imaging and Neurosciences Center, 729 Arapeen Drive, Salt Lake City, Utah, 84108, USA
| | - John E Greenlee
- Department of Neurology, University of Utah, Imaging and Neurosciences Center, 729 Arapeen Drive, Salt Lake City, Utah, 84108, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Stacey L Clardy
- Department of Neurology, University of Utah, Imaging and Neurosciences Center, 729 Arapeen Drive, Salt Lake City, Utah, 84108, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
30
|
Popławska-Domaszewicz K, Florczak-Wyspiańska J, Kozubski W, Michalak S. Paraneoplastic movement disorders. Rev Neurosci 2018; 29:745-755. [DOI: 10.1515/revneuro-2017-0081] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 01/26/2018] [Indexed: 12/22/2022]
Abstract
Abstract
Paraneoplastic movement disorders are rare, autoimmune-mediated, nonmetastatic complications of malignant neoplasms. Common paraneoplastic movement disorders include paraneoplastic chorea, dystonia, cerebellar degeneration, different types of encephalitis, opsoclonus-myoclonus syndrome, stiff person syndrome, and neuromyotonia. Syndromes usually develop before tumor diagnosis, have subacute onset, and are associated with serum or cerebrospinal fluid antibodies. Two types of antibodies can be distinguished: antibodies against nuclear and cytoplasmic neuronal antigens (anti-Hu, anti-Ri, anti-Yo, anti-Ma, anti-CV2/CRMP5, anti-Gephrin, and anti-GABATRAP) and antibodies recently identified against cell surface and synaptic proteins (anti-NMDAR, anti-LGI1, and anti-Caspr2). These two types differ from each other in a few important aspects. Antibodies against cell surface and synaptic protein disrupt cell-surface antigens. Clinical symptoms are related to the disruption of antigens and potentially can be reversed by immunotherapy. The association between these antibodies and malignancy is much less consistent. On the other hand, antibodies against nuclear and cytoplasmic neuronal antigens seem to be not pathogenic; however, they most likely indicate a T-cell-mediated immune response against neurons. Due to T-cell-mediated neuronal loss, response to immunotherapy is generally disappointing. Early recognition of all these diseases is crucial because it may lead to the disclosure of occult cancer. This review is focused on paraneoplastic movement disorders with emphasis on clinical presentations, investigational findings, and therapeutic results.
Collapse
Affiliation(s)
| | - Jolanta Florczak-Wyspiańska
- Department of Neurology , Poznan University of Medical Sciences , 49, Przybyszewskiego Str. , 60355 Poznan , Poland
| | - Wojciech Kozubski
- Department of Neurology , Poznan University of Medical Sciences , 49, Przybyszewskiego Str. , 60355 Poznan , Poland
| | - Sławomir Michalak
- Department of Neurochemistry and Neuropathology , Poznan University of Medical Sciences , 49, Przybyszewskiego Str. , 60355 Poznan , Poland
| |
Collapse
|
31
|
Yong SYS, Teo JY, Yong KP, Goh BKP. Paraneoplastic Stiff Person Syndrome Secondary to Pancreatic Adenocarcinoma. J Gastrointest Surg 2018; 22:172-174. [PMID: 29110193 DOI: 10.1007/s11605-017-3611-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/15/2017] [Indexed: 01/31/2023]
Affiliation(s)
- Stephanie Yu Shan Yong
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, 169856, Singapore.,National University Singapore, Yong Loo Lin School of Medicine, Singapore, 117597, Singapore
| | - Jin Yao Teo
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, 169856, Singapore
| | - Kok Pin Yong
- National Neuroscience Institute, Neurology, Singapore, 308433, Singapore
| | - Brian K P Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, 20 College Road, Singapore, 169856, Singapore. .,Duke NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
32
|
Martinez-Hernandez E, Ariño H, McKeon A, Iizuka T, Titulaer MJ, Simabukuro MM, Lancaster E, Petit-Pedrol M, Planagumà J, Blanco Y, Harvey RJ, Saiz A, Graus F, Dalmau J. Clinical and Immunologic Investigations in Patients With Stiff-Person Spectrum Disorder. JAMA Neurol 2017; 73:714-20. [PMID: 27065452 DOI: 10.1001/jamaneurol.2016.0133] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Symptoms of stiff-person syndrome (SPS), stiff-limb syndrome (SLS), or progressive encephalomyelitis with rigidity, myoclonus, or other symptoms (SPS-plus) can occur with several autoantibodies, but the relative frequency of each antibody, syndrome specificity, and prognostic implications are unclear. OBJECTIVE To report the clinical and immunologic findings of a large cohort of patients with stiff-person spectrum disorder (SPSD), including SPS, SLS, and SPS-plus. DESIGN, SETTING, AND PATIENTS This study retrospectively examined a case series (January 1, 1998, through December 31, 2014) of immunologic investigations performed in a neuroimmunology referral center. The study included 121 patients with clinical features of SPSD. Data analysis was performed from July 1, 2015, through November 1, 2015. MAIN OUTCOMES AND MEASURES Analysis of clinical-immunologic associations, including autoantibodies to 8 proteins expressed in inhibitory synapses. RESULTS The median age of the patients was 51 years (interquartile range, 40-61 years), and 75 (62.0%) were female. Fifty (41.3%) had SPS, 37 (30.6%) had SPS-plus, 24 (19.8%) had SLS, and 10 (8.3%) had SPS or SLS overlapping with ataxia, epilepsy, or encephalitis. Fifty-two patients (43.0%) had glutamic acid decarboxylase (GAD65) antibodies (2 with γ-aminobutyric acid-A [GABA-A] receptor antibodies), 24 (19.8%) had α1-subunit of the glycine receptor (GlyR) antibodies (2 with GAD65 antibodies), 5 (4.1%) had other antibodies, and 40 (33.1%) tested negative for antibodies. None had gephyrin or glycine transporter antibodies. Among the main immunologic groups (GAD65 antibodies, GlyR antibodies, and antibody negative), those with GAD65 antibodies were more likely to be female (45 [86.5%] of 52, 8 [36.4%] of 22, and 18 [45.0%] of 40, respectively; P < .001), have systemic autoimmunity (34 [65.4%] of 52, 7 [31.8%] of 22, and 13 [32.5%] of 40, respectively; P = .004), and have longer delays in being tested for antibodies (median, 3 vs 0.5 and 1 year; P < .001). Patients with GAD65 antibodies were more likely to develop SPS (27 [51.9%] of 52) or overlapping syndromes (8 [15.4%] of 52) than patients with GlyR antibodies (5 [22.7%] and 0 [0%] of 22, respectively), who more often developed SPS-plus (12 [54.5%] of 22 vs 7 [13.5%] in those with GAD65 antibodies); antibody-negative patients had an intermediate syndrome distribution. In multivariable analysis, symptom severity (P = .001) and immunologic group (P = .01) were independently associated with outcome. Compared with patients with GlyR antibodies, those with GAD65 antibodies (odds ratio, 11.1, 95% CI, 2.3-53.7; P = .003) had worse outcome. Patients without antibodies had similar outcome than patients with GlyR antibodies (odds ratio, 4.2, 95% CI, 0.9-20.0; P = .07). CONCLUSIONS AND RELEVANCE In SPSD, symptom severity and presence and type of antibodies are predictors of outcome.
Collapse
Affiliation(s)
- Eugenia Martinez-Hernandez
- Department of Neurology, Hospital Clínic, University of Barcelona, Barcelona, Spain2Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Helena Ariño
- Department of Neurology, Hospital Clínic, University of Barcelona, Barcelona, Spain2Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Andrew McKeon
- Departments of Neurology, Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Takahiro Iizuka
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Maarten J Titulaer
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mateus M Simabukuro
- Neurology Division, Hospital das Clinicas, São Paulo University, São Paulo, Brazil
| | - Eric Lancaster
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - Mar Petit-Pedrol
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Jesús Planagumà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Yolanda Blanco
- Department of Neurology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Robert J Harvey
- Department of Pharmacology, UCL School of Pharmacy, London, England
| | - Albert Saiz
- Department of Neurology, Hospital Clínic, University of Barcelona, Barcelona, Spain2Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Francesc Graus
- Department of Neurology, Hospital Clínic, University of Barcelona, Barcelona, Spain2Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Josep Dalmau
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain7Department of Neurology, University of Pennsylvania, Philadelphia9Institució Catalana de Recerca i Estudis Avançats (ICREA), Catalonia, Spain
| |
Collapse
|
33
|
McKeon A, Tracy JA. GAD65 neurological autoimmunity. Muscle Nerve 2017; 56:15-27. [PMID: 28063151 DOI: 10.1002/mus.25565] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/04/2017] [Indexed: 12/11/2022]
Abstract
The glutamic acid decarboxylase 65-kilodalton isoform (GAD65) antibody is a biomarker of autoimmune central nervous system (CNS) disorders and, more commonly, nonneurological autoimmune diseases. Type 1 diabetes, autoimmune thyroid disease, and pernicious anemia are the most frequent GAD65 autoimmune associations. One or more of these disorders coexists in approximately 70% of patients with GAD65 neurological autoimmunity. Neurological phenotypes have CNS localization and include limbic encephalitis, epilepsy, cerebellar ataxia, and stiff-person syndrome (SPS), among others. Classic SPS is a disorder on the spectrum of CNS hyperexcitability which also includes phenotypes that are either more restricted (stiff-limb syndrome) or more widespread (progressive encephalomyelitis with rigidity and myoclonus). GAD65 antibody is not highly predictive of a paraneoplastic cause for neurological disorders, but diverse cancer types have been occasionally reported. For all phenotypes, responses to immunotherapy are variable (approximately 50% improve). GAD65 autoimmunity is important to recognize for both coexisting nonneurological autoimmune associations and potential immunotherapy-response. Muscle Nerve 56: 15-27, 2017.
Collapse
Affiliation(s)
- Andrew McKeon
- Department of Neurology, College of Medicine, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA.,Department of Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jennifer A Tracy
- Department of Neurology, College of Medicine, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW This review highlights the recent developments in immune-mediated movement disorders and how they reflect on clinical practice and our understanding of the underlying pathophysiological mechanisms. RECENT FINDINGS The antibody spectrum associated with stiff person syndrome and related disorders (SPSD) has broadened and, apart from the classic glutamic acid decarboxylase (GAD)- and amphiphysin-antibodies, includes now also antibodies against dipeptidyl-peptidase-like protein-6 (DPPX), gamma-aminobutyric acid type A receptor (GABAAR), glycine receptor (GlyR) and glycine transporter 2 (GlyT2). The field of movement disorders with neuronal antibodies keeps expanding with the discovery for example of antibodies against leucine rich glioma inactivated protein 1 (LGI1) and contactin associated protein 2 (Caspr2) in chorea, or antibodies targeting ARHGAP26- or Na/K ATPase alpha 3 subunit (ATP1A3) in cerebellar ataxia. Moreover, neuronal antibodies may partly account for movement disorders attributed for example to Sydenham's chorea, coeliac disease, or steroid responsive encephalopathy with thyroid antibodies. Lastly, there is an interface of immunology, genetics and neurodegeneration, e.g. in Aicardi-Goutières syndrome or the tauopathy with IgLON5-antibodies. SUMMARY Clinicians should be aware of new antibodies such as dipeptidyl-peptidase-like protein-6, gamma-aminobutyric acid type A receptor and glycine transporter 2 in stiff person syndrome and related disorders, as well as of the expanding spectrum of immune-mediated movement disorders.
Collapse
|
35
|
Abstract
Autoimmune movement disorders encapsulate a large and diverse group of neurologic disorders occurring either in isolation or accompanying more diffuse autoimmune encephalitic illnesses. The full range of movement phenomena has been described and, as they often occur in adults, many of the presentations can mimic neurodegenerative disorders, such as Huntington disease. Disorders may be ataxic, hypokinetic (parkinsonism), or hyperkinetic (myoclonus, chorea, tics, and other dyskinetic disorders). The autoantibody targets are diverse and include neuronal surface proteins such as leucine-rich, glioma-inactivated 1 (LGI1) and glycine receptors, as well as antibodies (such as intracellular antigens) that are markers of a central nervous system process mediated by CD8+ cytotoxic T cells. However, there are two conditions, stiff-person syndrome (also known as stiff-man syndrome) and progressive encephalomyelitis with rigidity and myoclonus (PERM), that are always autoimmune movement disorders. In some instances (such as Purkinje cell cytoplasmic antibody-1 (PCA-1) autoimmunity), antibodies detected in serum and cerebrospinal fluid can be indicative of a paraneoplastic cause, and may direct the cancer search. In other instances (such as 65kDa isoform of glutamic acid decarboxylase (GAD65) autoimmunity), a paraneoplastic cause is very unlikely, and early treatment with immunotherapy may promote improvement or recovery. Here we describe the different types of movement disorder and the clinical features and antibodies associated with them, and discuss treatment.
Collapse
Affiliation(s)
- Andrew Mckeon
- Departments of Neurology and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
36
|
Tarulli A. Hyperkinetic Movement Disorders. Neurology 2016. [DOI: 10.1007/978-3-319-29632-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
37
|
Bhatti AB, Gazali ZA. Recent Advances and Review on Treatment of Stiff Person Syndrome in Adults and Pediatric Patients. Cureus 2015; 7:e427. [PMID: 26848416 PMCID: PMC4727915 DOI: 10.7759/cureus.427] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stiff Person Syndrome (SPS) is one of the rarest autoimmune neurological disorders, which is mostly reported in women. It is characterised by fluctuating muscle rigidity and spasms. There are many variants of SPS, these include the classical SPS, Stiff Leg Syndrome (SLS), paraneoplastic variant, gait ataxia, dysarthria, and abnormal eye movements. Studies have shown that the paraneoplastic variant of SPS is more common in patients with breast cancer who harbour amphiphysin antibodies, followed by colon cancer, lung cancer, Hodgkin's disease, and malignant thymoma. Currently, the treatment for SPS revolves around improving the quality of life by reducing the symptoms as far as possible with the use of GABAergic agonists, such as diazepam or other benzodiazepines, steroids, plasmapheresis, and intravenous immunoglobulin (IVIG). There have been random clinical trials with Rituximab, but nothing concrete has been suggested. A treatment approach with standard drugs and cognitive behavioral therapy (CBT) seems to be promising.
Collapse
Affiliation(s)
- Adnan Bashir Bhatti
- Department of Medicine, Capital Development Authority Hospital, Islamabad, Pakistan
| | | |
Collapse
|
38
|
Baizabal-Carvallo JF, Jankovic J. Stiff-person syndrome: insights into a complex autoimmune disorder. J Neurol Neurosurg Psychiatry 2015; 86:840-8. [PMID: 25511790 DOI: 10.1136/jnnp-2014-309201] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/21/2014] [Indexed: 02/06/2023]
Abstract
Stiff-person syndrome (SPS) is characterised by progressive rigidity and muscle spasms affecting the axial and limb muscles. Since its initial description in 1956, marked progress has been made in the clinical characterisation, understanding of pathogenesis and therapy of this disorder. SPS can be classified according to the clinical presentation into classic SPS and SPS variants: focal or segmental-SPS, jerking-SPS and progressive encephalomyelitis with rigidity and myoclonus. Most patients with SPS have antibodies directed against the glutamic acid decarboxylase, the rate-limiting enzyme for the production of the inhibitory neurotransmitter γ-aminobutyric acid (GABA). Antibodies directed against GABA(A) receptor-associated protein, and the glycine-α1 receptor can also be observed. Paraneoplastic SPS is commonly associated with antiamphiphysin antibodies and breast cancer. Treatment of SPS with drugs that increase the GABAergic tone combined with immunotherapy can improve the neurological manifestations of these patients. The prognosis, however, is unpredictable and spontaneous remissions are unlikely.
Collapse
Affiliation(s)
- José Fidel Baizabal-Carvallo
- Department of Neurology, Parkinson's Disease Center and Movement Disorders Clinic, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph Jankovic
- Department of Neurology, Parkinson's Disease Center and Movement Disorders Clinic, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
39
|
Choii G, Ko J. Gephyrin: a central GABAergic synapse organizer. Exp Mol Med 2015; 47:e158. [PMID: 25882190 DOI: 10.1038/emm.2015.5] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 12/18/2014] [Indexed: 01/22/2023] Open
Abstract
Gephyrin is a central element that anchors, clusters and stabilizes glycine and γ-aminobutyric acid type A receptors at inhibitory synapses of the mammalian brain. It self-assembles into a hexagonal lattice and interacts with various inhibitory synaptic proteins. Intriguingly, the clustering of gephyrin, which is regulated by multiple posttranslational modifications, is critical for inhibitory synapse formation and function. In this review, we summarize the basic properties of gephyrin and describe recent findings regarding its roles in inhibitory synapse formation, function and plasticity. We will also discuss the implications for the pathophysiology of brain disorders and raise the remaining open questions in this field.
Collapse
Affiliation(s)
- Gayoung Choii
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Jaewon Ko
- 1] Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea [2] Department of Psychiatry, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
40
|
Gresa-Arribas N, Ariño H, Martínez-Hernández E, Petit-Pedrol M, Sabater L, Saiz A, Dalmau J, Graus F. Antibodies to inhibitory synaptic proteins in neurological syndromes associated with glutamic acid decarboxylase autoimmunity. PLoS One 2015; 10:e0121364. [PMID: 25774787 PMCID: PMC4361655 DOI: 10.1371/journal.pone.0121364] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/31/2015] [Indexed: 12/12/2022] Open
Abstract
Antibodies to glutamic acid decarboxylase (GAD-ab) associate to different neurological syndromes. It is unknown if the diversity in syndrome association represents epitopes in different immunodominant domains or co-existence of antibodies to other proteins of the inhibitory synapsis. We examined the serum and CSF of 106 patients with anti-GAD related syndromes (39 cerebellar ataxia, 32 stiff-person syndrome [SPS], 18 epilepsy, and 17 limbic encephalitis [LE]). GAD65-ab titres were quantified by ELISA. Immunoblot was used to determine if the antibody-targeted epitopes of GAD65 and GAD67 were linear. A cell-based assay (CBA) with HEK293 cells expressing the GAD65 N-terminal, central catalytic domain, or C-terminal was used to investigate the immunodominant domains. Antibodies to GAD67, gamma-aminobutyric acid A receptor (GABAaR), glycine receptor (GlyR), GABAaR-associated protein (GABARAP), and gephyrin were determined with CBA. GAD-ab internalization was investigated using cultured rat hippocampal neurons. CSF GAD65-ab titres were higher in patients with cerebellar ataxia and LE compared to those with SPS (p = 0.02). GAD67-ab were identified in 81% of sera and 100% of CSF. GAD65-ab recognized linear epitopes in 98% of the patients and GAD67-ab in 42% (p<0.001). The GAD65 catalytic domain was recognized by 93% of sera, and the three domains by 22% of sera and 74% of CSF (p<0.001). Six patients had GABAaR-ab and another 6 had GlyR-ab without association to distinctive symptoms. None of the patients had gephyrin- or GABARAP-ab. GAD65-ab were not internalized by live neurons. Overall, these findings show that regardless of the neurological syndrome, the CSF immune response against GAD is more widespread than that of the serum and that there is no specific association between clinical phenotype and the presence of antibodies against other proteins of the inhibitory synapsis.
Collapse
Affiliation(s)
- Nuria Gresa-Arribas
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Helena Ariño
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Service of Neurology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Eugenia Martínez-Hernández
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Service of Neurology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Mar Petit-Pedrol
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lidia Sabater
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Albert Saiz
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Service of Neurology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Josep Dalmau
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Francesc Graus
- Neuroimmunology Program, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Service of Neurology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
41
|
Agca S, Houen G, Trier NH. Characterization of continuous B-cell epitopes in the N-terminus of glutamate decarboxylase67 using monoclonal antibodies. J Pept Sci 2014; 20:928-34. [DOI: 10.1002/psc.2703] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 08/27/2014] [Accepted: 09/01/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Selin Agca
- Department of Clinical Biochemistry, Immunology and Genetics; Statens Serum Institut; Artillerivej 5 2300 Copenhagen S Denmark
- Department of Molecular Biology and Genetics; Aarhus University; Forskerparken - Gustav Wieds Vej 10 8000 Aarhus C Denmark
| | - Gunnar Houen
- Department of Clinical Biochemistry, Immunology and Genetics; Statens Serum Institut; Artillerivej 5 2300 Copenhagen S Denmark
| | - Nicole Hartwig Trier
- Department of Clinical Biochemistry, Immunology and Genetics; Statens Serum Institut; Artillerivej 5 2300 Copenhagen S Denmark
| |
Collapse
|
42
|
Probst C, Blöcker IM, Mindorf S, Unger M, Fechner K, Meinck HM, Balint B, Stöcker W, Schlumberger W, Komorowski L. Autoantibodies against glycine-associated synaptic proteins in stiff-person syndrome. J Neuroimmunol 2014. [DOI: 10.1016/j.jneuroim.2014.08.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
43
|
Irani SR, Gelfand JM, Al-Diwani A, Vincent A. Cell-surface central nervous system autoantibodies: clinical relevance and emerging paradigms. Ann Neurol 2014; 76:168-84. [PMID: 24930434 PMCID: PMC4141019 DOI: 10.1002/ana.24200] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 06/13/2014] [Accepted: 06/13/2014] [Indexed: 01/17/2023]
Abstract
The recent discovery of several potentially pathogenic autoantibodies has helped identify patients with clinically distinctive central nervous system diseases that appear to benefit from immunotherapy. The associated autoantibodies are directed against the extracellular domains of cell-surface-expressed neuronal or glial proteins such as LGI1, N-methyl-D-aspartate receptor, and aquaporin-4. The original descriptions of the associated clinical syndromes were phenotypically well circumscribed. However, as availability of antibody testing has increased, the range of associated patient phenotypes and demographics has expanded. This in turn has led to the recognition of more immunotherapy-responsive syndromes in patients presenting with cognitive and behavioral problems, seizures, movement disorders, psychiatric features, and demyelinating disease. Although antibody detection remains diagnostically important, clinical recognition of these distinctive syndromes should ensure early and appropriate immunotherapy administration. We review the emerging paradigm of cell-surface-directed antibody-mediated neurological diseases, describe how the associated disease spectrums have broadened since the original descriptions, discuss some of the methodological issues regarding techniques for antibody detection and emphasize considerations surrounding immunotherapy administration. As these disorders continue to reach mainstream neurology and even psychiatry, more cell-surface-directed antibodies will be discovered, and their possible relevance to other more common disease presentations should become more clearly defined.
Collapse
Affiliation(s)
- Sarosh R Irani
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom; Multiple Sclerosis and Neuroinflammation Center, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | | | | | | |
Collapse
|
44
|
Stiff person-syndrome IgG affects presynaptic GABAergic release mechanisms. J Neural Transm (Vienna) 2014; 122:357-62. [PMID: 24990310 DOI: 10.1007/s00702-014-1268-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/25/2014] [Indexed: 10/25/2022]
Abstract
The majority of patients with stiff person-syndrome (SPS) are characterized by autoantibodies to glutamate decarboxylase 65 (GAD65). In previous passive-transfer studies, SPS immunoglobulin G (IgG) induced SPS core symptoms. We here provide evidence that SPS-IgG causes a higher frequency of spontaneous vesicle fusions. Sustained GABAergic transmission and presynaptic GABAergic vesicle pool size remained unchanged. Since these findings cannot be attributed to anti-GAD65 autoantibodies alone, we propose that additional autoantibodies with so far undefined antigen specificity might affect presynaptic release mechanisms.
Collapse
|
45
|
Martel S, De Angelis F, Lapointe E, Larue S, Speranza G. Paraneoplastic neurologic syndromes: Clinical presentation and management. Curr Probl Cancer 2014; 38:115-34. [DOI: 10.1016/j.currproblcancer.2014.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
Abstract
The neurotransmitters GABA and glycine mediate fast synaptic inhibition by activating ligand-gated chloride channels--namely, type A GABA (GABA(A)) and glycine receptors. Both types of receptors are anchored postsynaptically by gephyrin, which self-assembles into a scaffold and interacts with the cytoskeleton. Current research indicates that postsynaptic gephyrin clusters are dynamic assemblies that are held together and regulated by multiple protein-protein interactions. Moreover, post-translational modifications of gephyrin regulate the formation and plasticity of GABAergic synapses by altering the clustering properties of postsynaptic scaffolds and thereby the availability and function of receptors and other signalling molecules. Here, we discuss the formation and regulation of the gephyrin scaffold, its role in GABAergic and glycinergic synaptic function and the implications for the pathophysiology of brain disorders caused by abnormal inhibitory neurotransmission.
Collapse
|
47
|
Jung YJ, Jeong HG, Kim R, Kim HJ, Jeon BS. Stiff-person syndrome: case series. J Mov Disord 2014; 7:19-21. [PMID: 24926406 PMCID: PMC4051723 DOI: 10.14802/jmd.14004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 10/30/2013] [Accepted: 01/10/2014] [Indexed: 12/21/2022] Open
Abstract
Stiff-person syndrome (SPS) is a rare disorder, characterized by progressive fluctuating muscular rigidity and spasms. Glutamic acid decarboxylase (GAD) antibody is primarily involved in the pathogenesis of SPS and SPS is strongly associated with other autoimmune disease. Here we report three cases of patients with classical SPS finally confirmed by high serum level of GAD antibodies. All of our patients respond favorably to gamma amino butyric acid-enhancing drugs and immunotherapies.
Collapse
Affiliation(s)
- Yu Jin Jung
- Department of Neurology, Seoul National University Hospital, Seoul, Korea ; Movement Disorder Center, Parkinson Study Group, Seoul National University Hospital, Seoul, Korea
| | - Han G Jeong
- Movement Disorder Center, Parkinson Study Group, Seoul National University Hospital, Seoul, Korea
| | - Ryul Kim
- Movement Disorder Center, Parkinson Study Group, Seoul National University Hospital, Seoul, Korea
| | - Han-Joon Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Korea ; Movement Disorder Center, Parkinson Study Group, Seoul National University Hospital, Seoul, Korea
| | - Beom S Jeon
- Department of Neurology, Seoul National University Hospital, Seoul, Korea ; Movement Disorder Center, Parkinson Study Group, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
48
|
Koca I, Ucar M, Kalender ME, Alkan S. The horses are the first thought but one must not forget the zebras even if they are rare: Stiff person syndrome associated with malignant mesothelioma. BMJ Case Rep 2014; 2014:bcr-2013-203455. [PMID: 24711475 DOI: 10.1136/bcr-2013-203455] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Stiff person syndrome (SPS) is a rare condition that causes rigidity in the muscles of the body and extremities, difficulty in walking, episodic spasms and progressive disability. SPS is generally seen together with autoimmune disorders such as diabetes mellitus, thyroiditis, vitiligo and pernicious anaemia. Rarely, it may develop as a paraneoplastic condition. SPS cases associated with breast cancer, small cell lung carcinoma, thymoma, Hodgkin's lymphoma and colorectal cancer have been reported in the literature. We present a case of a 58-year-old female patient who had malignant mesothelioma-associated SPS. Patients who have muscle spasms and difficulty in movement of joints should be evaluated for SPS before diagnosis of Parkinson's or other neurological disorders, and possible underlying malignancies should be excluded.
Collapse
Affiliation(s)
- Irfan Koca
- Department of Physical Therapy and Rehabilitation, Gaziantep University, Faculty of Medicine, Gaziantep, Turkey
| | | | | | | |
Collapse
|
49
|
van Coevorden-Hameete MH, de Graaff E, Titulaer MJ, Hoogenraad CC, Sillevis Smitt PAE. Molecular and cellular mechanisms underlying anti-neuronal antibody mediated disorders of the central nervous system. Autoimmun Rev 2014; 13:299-312. [PMID: 24225076 DOI: 10.1016/j.autrev.2013.10.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 10/30/2013] [Indexed: 12/31/2022]
Abstract
Over the last decade multiple autoantigens located on the plasma membrane of neurons have been identified. Neuronal surface antigens include molecules directly involved in neurotransmission and excitability. Binding of the antibody to the antigen may directly alter the target protein's function, resulting in neurological disorders. The often striking reversibility of symptoms following early aggressive immunotherapy supports a pathogenic role for autoantibodies to neuronal surface antigens. In order to better understand and treat these neurologic disorders it is important to gain insight in the underlying mechanisms of antibody pathogenicity. In this review we discuss the clinical, circumstantial, in vitro and in vivo evidence for neuronal surface antibody pathogenicity and the possible underlying cellular and molecular mechanisms. This review shows that antibodies to neuronal surface antigens are often directed at conformational epitopes located in the extracellular domain of the antigen. The conformation of the epitope can be affected by specific posttranslational modifications. This may explain the distinct clinical phenotypes that are seen in patients with antibodies to antigens that are expressed throughout the brain. Furthermore, it is likely that there is a heterogeneous antibody population, consisting of different IgG subtypes and directed at multiple epitopes located in an immunogenic region. Binding of these antibodies may result in different pathophysiological mechanisms occurring in the same patient, together contributing to the clinical syndrome. Unraveling the predominant mechanism in each distinct antigen could provide clues for therapeutic interventions.
Collapse
Affiliation(s)
- M H van Coevorden-Hameete
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - E de Graaff
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - M J Titulaer
- Department of Neurology, Erasmus MC, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - C C Hoogenraad
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - P A E Sillevis Smitt
- Department of Neurology, Erasmus MC, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| |
Collapse
|
50
|
Alexopoulos H, Dalakas MC. Immunology of stiff person syndrome and other GAD-associated neurological disorders. Expert Rev Clin Immunol 2014; 9:1043-53. [DOI: 10.1586/1744666x.2013.845527] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|