1
|
Shastri A, Al Aiyan A, Kishore U, Farrugia ME. Immune-Mediated Neuropathies: Pathophysiology and Management. Int J Mol Sci 2023; 24:7288. [PMID: 37108447 PMCID: PMC10139406 DOI: 10.3390/ijms24087288] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Dysfunction of the immune system can result in damage of the peripheral nervous system. The immunological mechanisms, which include macrophage infiltration, inflammation and proliferation of Schwann cells, result in variable degrees of demyelination and axonal degeneration. Aetiology is diverse and, in some cases, may be precipitated by infection. Various animal models have contributed and helped to elucidate the pathophysiological mechanisms in acute and chronic inflammatory polyradiculoneuropathies (Guillain-Barre Syndrome and chronic inflammatory demyelinating polyradiculoneuropathy, respectively). The presence of specific anti-glycoconjugate antibodies indicates an underlying process of molecular mimicry and sometimes assists in the classification of these disorders, which often merely supports the clinical diagnosis. Now, the electrophysiological presence of conduction blocks is another important factor in characterizing another subgroup of treatable motor neuropathies (multifocal motor neuropathy with conduction block), which is distinct from Lewis-Sumner syndrome (multifocal acquired demyelinating sensory and motor neuropathy) in its response to treatment modalities as well as electrophysiological features. Furthermore, paraneoplastic neuropathies are also immune-mediated and are the result of an immune reaction to tumour cells that express onconeural antigens and mimic molecules expressed on the surface of neurons. The detection of specific paraneoplastic antibodies often assists the clinician in the investigation of an underlying, sometimes specific, malignancy. This review aims to discuss the immunological and pathophysiological mechanisms that are thought to be crucial in the aetiology of dysimmune neuropathies as well as their individual electrophysiological characteristics, their laboratory features and existing treatment options. Here, we aim to present a balance of discussion from these diverse angles that may be helpful in categorizing disease and establishing prognosis.
Collapse
Affiliation(s)
- Abhishek Shastri
- Central and North West London NHS Foundation Trust, London NW1 3AX, UK
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine, UAE University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine, UAE University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Maria Elena Farrugia
- Department of Neurology, Institute of Neurological Sciences, Southern General Hospital, Glasgow G51 4TF, UK
| |
Collapse
|
2
|
Serum C3 complement levels predict prognosis and monitor disease activity in Guillain-Barré syndrome. J Neurol Sci 2023; 444:120512. [PMID: 36462224 DOI: 10.1016/j.jns.2022.120512] [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: 07/20/2022] [Revised: 10/27/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Biomarkers are needed to predict prognosis and disease activity in patients with Guillain-Barré syndrome (GBS). The complement system is a key player in the pathogenesis of GBS. This study aimed to assess the potential utility of serum complement proteins as novel biomarkers in GBS. METHODS We reviewed the medical records of 76 GBS patients with C3 and C4 measurements during hospitalization between 2010 and 2021. Clinical outcomes were correlated with baseline serum C3, C4, and seven additional predictors: four existing biomarkers (GM1, albumin, immunoglobulin G, neutrophil-lymphocyte ratio) and three clinical factors from the modified Erasmus GBS outcome score model. Five complement activation products (C3a, C4a, C5a, soluble C5b-9, factor Bb) were measured in 35 patients and were compared with C3 and C4 levels. Longitudinal changes in C3 and C4 levels were compared with the disease course in 12 patients. RESULTS Higher C3, but not C4, was associated with poorer outcomes: lower Medical Research Council sum scores (MRCSS), higher GBS disability score (GBSDS), longer hospitalization, and more frequent treatment-related fluctuations. Age, MRCSS at admission, and baseline serum C3 were significant independent indicators of 1- and 3-month GBSDS. We found that C3 was positively correlated with C3a (r = 0.32) and C5a (r = 0.37), which indicates an activated complement cascade with high C3. Longitudinal change of C3 coincided with clinical severity of the disease course. INTERPRETATION This study highlights the use of serum C3 as a novel mechanistic biomarker in GBS. Larger prospective studies are needed to validate our findings.
Collapse
|
3
|
Reed CB, Feltri ML, Wilson ER. Peripheral glia diversity. J Anat 2022; 241:1219-1234. [PMID: 34131911 PMCID: PMC8671569 DOI: 10.1111/joa.13484] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Recent years have seen an evolving appreciation for the role of glial cells in the nervous system. As we move away from the typical neurocentric view of neuroscience, the complexity and variability of central nervous system glia is emerging, far beyond the three main subtypes: astrocytes, oligodendrocytes, and microglia. Yet the diversity of the glia found in the peripheral nervous system remains rarely discussed. In this review, we discuss the developmental origin, morphology, and function of the different populations of glia found in the peripheral nervous system, including: myelinating Schwann cells, Remak Schwann cells, repair Schwann cells, satellite glia, boundary cap-derived glia, perineurial glia, terminal Schwann cells, glia found in the skin, olfactory ensheathing cells, and enteric glia. The morphological and functional heterogeneity of glia found in the periphery reflects the diverse roles the nervous system performs throughout the body. Further, it highlights a complexity that should be appreciated and considered when it comes to a complete understanding of the peripheral nervous system in health and disease.
Collapse
Affiliation(s)
- Chelsey B. Reed
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - M. Laura Feltri
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - Emma R. Wilson
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| |
Collapse
|
4
|
Meidaninikjeh S, Sabouni N, Taheri M, Borjkhani M, Bengar S, Majidi Zolbanin N, Khalili A, Jafari R. SARS-CoV-2 and Guillain-Barré Syndrome: Lessons from Viral Infections. Viral Immunol 2022; 35:404-417. [PMID: 35766944 DOI: 10.1089/vim.2021.0187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for the COVID-19 pandemic. COVID-19 has a broad clinical spectrum from asymptomatic patients to multiorgan dysfunction and septic shock. Most of the common symptoms of COVID-19 are classified as respiratory disorders, but some reports show neurological involvements. During the COVID-19 pandemic, a case series of neurological complications, such as Guillain-Barré syndrome (GBS), were reported. GBS is a neuroimmune disorder with acute inflammatory radicular polyneuropathy in different parts of the peripheral nerve. Some studies have reported GBS as an inflammatory neuropathy related to various viral infections, such as cytomegalovirus (CMV), Epstein-Barr Virus (EBV), herpes simplex virus (HSV), human immunodeficiency virus (HIV), influenza, and Zika virus. There are some immunomodulation approaches for the management of GBS. Studies have evaluated the effects of the various therapeutic approaches, including intravenous immunoglobulin (IVIG), plasma exchange (PE), complement inhibitors, and corticosteroids to regulate overactivation of immune responses during GBS in experimental and clinical studies. In this regard, the possible association between GBS and SARS-CoV-2 infection during the outbreak of the current pandemic and also the mentioned therapeutic approaches were reviewed.
Collapse
Affiliation(s)
- Sepideh Meidaninikjeh
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran.,Cancer Biomedical Center (CBC) Research Institute, Tehran, Iran
| | - Nasim Sabouni
- Department of Immunology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdie Taheri
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahdis Borjkhani
- Bioprocess Engineering Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Sajad Bengar
- Department of Microbiology, Faculty of Science, Shahre Ghods Branch, Islamic Azad University, Shahre Ghods, Tehran, Iran
| | - Naime Majidi Zolbanin
- Experimental and Applied Pharmaceutical Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Ahmad Khalili
- Cancer Biomedical Center (CBC) Research Institute, Tehran, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
5
|
Halstead SK, Gourlay DS, Penderis J, Bianchi E, Dondi M, Wessmann A, Musteata M, Le Chevoir M, Martinez-Anton L, Bhatti SFM, Volk H, Mateo I, Tipold A, Ives E, Pakozdy A, Gutierrez-Quintana R, Brocal J, Whitehead Z, Granger N, Pazzi P, Harcourt-Brown T, José-López R, Rupp S, Schenk HC, Smith P, Gandini G, Menchetti M, Mortera-Balsa V, Rusbridge C, Tauro A, Cozzi F, Deutschland M, Tirrito F, Freeman P, Lowrie M, Jackson MR, Willison HJ, Rupp A. Serum anti-GM2 and anti-GalNAc-GD1a IgG antibodies are biomarkers for acute canine polyradiculoneuritis. J Small Anim Pract 2022; 63:104-112. [PMID: 34791652 DOI: 10.1111/jsap.13439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 08/13/2021] [Accepted: 09/19/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES A previous single-country pilot study indicated serum anti-GM2 and anti-GA1 anti-glycolipid antibodies as potential biomarkers for acute canine polyradiculoneuritis. This study aims to validate these findings in a large geographically heterogenous cohort. MATERIALS AND METHODS Sera from 175 dogs clinically diagnosed with acute canine polyradiculoneuritis, 112 dogs with other peripheral nerve, cranial nerve or neuromuscular disorders and 226 neurologically normal dogs were screened for anti-glycolipid antibodies against 11 common glycolipid targets to determine the immunoglobulin G anti-glycolipid antibodies with the highest combined sensitivity and specificity for acute canine polyradiculoneuritis. RESULTS Anti-GM2 anti-glycolipid antibodies reached the highest combined sensitivity and specificity (sensitivity: 65.1%, 95% confidence interval 57.6 to 72.2%; specificity: 90.2%, 95% confidence interval 83.1 to 95.0%), followed by anti-GalNAc-GD1a anti-glycolipid antibodies (sensitivity: 61.7%, 95% confidence interval 54.1 to 68.9%; specificity: 89.3%, 95% confidence interval 82.0 to 94.3%) and these anti-glycolipid antibodies were frequently present concomitantly. Anti-GA1 anti-glycolipid antibodies were detected in both acute canine polyradiculoneuritis and control animals. Both for anti-GM2 and anti-GalNAc-GD1a anti-glycolipid antibodies, sex was found a significantly associated factor with a female to male odds ratio of 2.55 (1.27 to 5.31) and 3.00 (1.22 to 7.89), respectively. Anti-GalNAc-GD1a anti-glycolipid antibodies were more commonly observed in dogs unable to walk (OR 4.56, 1.56 to 14.87). CLINICAL SIGNIFICANCE Anti-GM2 and anti-GalNAc-GD1a immunoglobulin G anti-glycolipid antibodies represent serum biomarkers for acute canine polyradiculoneuritis.
Collapse
Affiliation(s)
- S K Halstead
- Neuroimmunology Laboratory, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - D S Gourlay
- Neuroimmunology Laboratory, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - J Penderis
- Vet Extra Neurology, Broadleys Veterinary Hospital, Stirling, FK7 7LE, UK
| | - E Bianchi
- Department of Veterinary Science, University of Parma, 43126, Parma, Italy
| | - M Dondi
- Department of Veterinary Science, University of Parma, 43126, Parma, Italy
| | - A Wessmann
- Neurology and Neurosurgery Service, Pride Veterinary Centre, Pride Park, Derby, DE24 8HX, UK
| | - M Musteata
- Neurology Service, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Iași, 700489, Romania
| | - M Le Chevoir
- Department of Neurology and Neurosurgery, University of Melbourne, Werribee, Victoria, 3030, Australia
| | - L Martinez-Anton
- Chestergates Veterinary Specialists, Telford Court, Chestergates, CH1 6LT, UK
| | - S F M Bhatti
- Small Animal Department, Small Animal Teaching Hospital, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - H Volk
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - I Mateo
- Servicio de Neurología, Hospital Clínico Veterinario - Universidad Alfonso X el Sabio, Madrid, Spain
| | - A Tipold
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - E Ives
- Anderson Moores Veterinary Specialists, Hursley, Winchester, SO21 2LL, UK
| | - A Pakozdy
- University Hospital for Small Animals, University of Veterinary Medicine, Vienna, Austria
| | | | - J Brocal
- Wear Referrals Veterinary Hospital, Bradbury, Stockton-on-Tees, TS21 2ES, UK
| | - Z Whitehead
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, 0110, South Africa
| | - N Granger
- The Royal Veterinary College, University of London, Hatfield, Hertfordshire, UK.,CVS Referrals, Bristol Veterinary Specialists at Highcroft, Bristol, UK
| | - P Pazzi
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, 0110, South Africa
| | - T Harcourt-Brown
- Langford Veterinary Services, School of Veterinary Sciences, University of Bristol, Lower Langford, BS40 5DU, UK
| | - R José-López
- School of Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK
| | - S Rupp
- Tierklinik Hofheim, 65719, Hofheim, Germany
| | - H C Schenk
- Tierklinik Lüneburg, 21337, Lüneburg, Germany
| | - P Smith
- Hamilton Specialist Referrals, Cressex Business Park, High Wycombe, HP12 3SD, UK
| | - G Gandini
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia, Italy
| | - M Menchetti
- Neurology and Neurosurgery Division, San Marco Veterinary Clinic, Veggiano, Italy
| | - V Mortera-Balsa
- North Downs Specialist Referrals, 3&4 The Brewerstreet Dairy Business Park, Bletchingley, Surrey, RH1 4QP, UK
| | - C Rusbridge
- Neurology Section, Fitzpatrick Referrals, Godalming, Surrey, GU2 7AL, UK.,School of Veterinary Medicine, Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - A Tauro
- Chestergates Veterinary Specialists, Telford Court, Chestergates, CH1 6LT, UK
| | - F Cozzi
- Clinica Neurologica Veterinaria, 20148, Milan, Italy
| | | | - F Tirrito
- Clinica Neurologica Veterinaria, 20148, Milan, Italy
| | - P Freeman
- The Queen's Veterinary School Hospital, Cambridge, CB3 0ES, UK
| | - M Lowrie
- Dovecote Veterinary Hospital, Castle Donington, Derby, DE74 2LJ, UK
| | - M R Jackson
- Institute of Cancer Sciences, University of Glasgow, Bearsden, G61 1QH, UK
| | - H J Willison
- Neuroimmunology Laboratory, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - A Rupp
- School of Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK
| |
Collapse
|
6
|
Emerging Role of C5 Complement Pathway in Peripheral Neuropathies: Current Treatments and Future Perspectives. Biomedicines 2021; 9:biomedicines9040399. [PMID: 33917266 PMCID: PMC8067968 DOI: 10.3390/biomedicines9040399] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
The complement system is a key component of innate immunity since it plays a critical role in inflammation and defense against common pathogens. However, an inappropriate activation of the complement system is involved in numerous disorders, including peripheral neuropathies. Current strategies for neuropathy-related pain fail to achieve adequate pain relief, and although several therapies are used to alleviate symptoms, approved disease-modifying treatments are unavailable. This urgent medical need is driving the development of therapeutic agents for this condition, and special emphasis is given to complement-targeting approaches. Recent evidence has underscored the importance of complement component C5a and its receptor C5aR1 in inflammatory and neuropathic pain, indicating that C5a/C5aR1 axis activation triggers a cascade of events involved in pathophysiology of peripheral neuropathy and painful neuro-inflammatory states. However, the underlying pathophysiological mechanisms of this signaling in peripheral neuropathy are not fully known. Here, we provide an overview of complement pathways and major components associated with dysregulated complement activation in peripheral neuropathy, and of drugs under development targeting the C5 system. C5/C5aR1 axis modulators could represent a new strategy to treat complement-related peripheral neuropathies. Specifically, we describe novel C5aR allosteric modulators, which may potentially become new tools in the therapeutic armory against neuropathic pain.
Collapse
|
7
|
Kassimatis T, Greenlaw R, Hunter JP, Douiri A, Flach C, Rebollo-Mesa I, Nichols LL, Qasem A, Danzi G, Olsburgh J, Drage M, Friend PJ, Neri F, Karegli J, Horsfield C, Smith RA, Sacks SH. Ex vivo delivery of Mirococept: A dose-finding study in pig kidney after showing a low dose is insufficient to reduce delayed graft function in human kidney. Am J Transplant 2021; 21:1012-1026. [PMID: 33225626 DOI: 10.1111/ajt.16265] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/17/2020] [Accepted: 08/06/2020] [Indexed: 01/25/2023]
Abstract
The complement system plays a pivotal role in the pathogenesis of ischemia-reperfusion injury in solid organ transplantation. Mirococept is a potent membrane-localizing complement inhibitor that can be administered ex vivo to the donor kidney prior to transplantation. To evaluate the efficacy of Mirococept in reducing delayed graft function (DGF) in deceased donor renal transplantation, we undertook the efficacy of mirococept (APT070) for preventing ischaemia-reperfusion injury in the kidney allograft (EMPIRIKAL) trial (ISRCTN49958194). A dose range of 5-25 mg would be tested, starting with 10 mg in cohort 1. No significant difference between Mirococept at 10 mg and control was detected; hence the study was stopped to enable a further dose saturation study in a porcine kidney model. The optimal dose of Mirococept in pig kidney was 80 mg. This dose did not induce any additional histological damage compared to controls or after a subsequent 3 hours of normothermic machine perfusion. The amount of unbound Mirococept postperfusion was found to be within the systemic dose range considered safe in the Phase I trial. The ex vivo administration of Mirococept is a safe and feasible approach to treat DGF in deceased donor kidney transplantation. The porcine kidney study identified an optimal dose of 80 mg (equivalent to 120 mg in human kidney) that provides a basis for further clinical development.
Collapse
Affiliation(s)
- Theodoros Kassimatis
- Renal Unit, Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK.,School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Roseanna Greenlaw
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - James P Hunter
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Abdel Douiri
- School of Population Health and Environmental Studies, King's College London, London, UK
| | - Clare Flach
- School of Population Health and Environmental Studies, King's College London, London, UK
| | - Irene Rebollo-Mesa
- School of Immunology and Microbial Sciences, King's College London, London, UK.,UCB Biopharma, Brussels, Belgium
| | - Laura L Nichols
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Anass Qasem
- Renal Unit, Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK.,Department of Internal Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Guilherme Danzi
- School of Immunology and Microbial Sciences, King's College London, London, UK.,Department of Nephrology, Clinic Hospital, Federal University of Pernambuco, Recife, Brazil
| | - Jonathon Olsburgh
- Department of Transplantation, Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Martin Drage
- Department of Transplantation, Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Peter J Friend
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Flavia Neri
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Julieta Karegli
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Catherine Horsfield
- Department of Histopathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Richard A Smith
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Steven H Sacks
- School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
8
|
Hagen KM, Ousman SS. The Neuroimmunology of Guillain-Barré Syndrome and the Potential Role of an Aging Immune System. Front Aging Neurosci 2021; 12:613628. [PMID: 33584245 PMCID: PMC7873882 DOI: 10.3389/fnagi.2020.613628] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Guillain-Barré syndrome (GBS) is a paralyzing autoimmune condition affecting the peripheral nervous system (PNS). Within GBS there are several variants affecting different aspects of the peripheral nerve. In general, there appears to be a role for T cells, macrophages, B cells, and complement in initiating and perpetuating attacks on gangliosides of Schwann cells and axons. Of note, GBS has an increased prevalence and severity with increasing age. In addition, there are alterations in immune cell functioning that may play a role in differences in GBS with age alongside general age-related declines in reparative processes (e.g., delayed de-differentiation of Schwann cells and decline in phagocytic ability of macrophages). The present review will explore the immune response in GBS as well as in animal models of several variants of the disorder. In addition, the potential involvement of an aging immune system in contributing to the increased prevalence and severity of GBS with age will be theorized.
Collapse
Affiliation(s)
- Kathleen M. Hagen
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Shalina S. Ousman
- Departments of Clinical Neurosciences and Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
9
|
Agostinis C, Balduit A, Mangogna A, Zito G, Romano F, Ricci G, Kishore U, Bulla R. Immunological Basis of the Endometriosis: The Complement System as a Potential Therapeutic Target. Front Immunol 2021; 11:599117. [PMID: 33505394 PMCID: PMC7829336 DOI: 10.3389/fimmu.2020.599117] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Endometriosis (EM) is a chronic disease characterized by the presence and proliferation of functional endometrial glands and stroma outside the uterine cavity. Ovaries and pelvic peritoneum are the most common locations for endometrial ectopic tissue, followed by deep infiltrating EM sites. The cyclic and recurrent bleeding, the progressive fibrosis and the peritoneal adhesions of ectopic endometrial glands, may cause different symptoms depending on the origin involved. EM is a frequent clinical condition affecting around 10% of women of mainly reproductive age, as well as in post-menopausal women and adolescents, especially with uterine anomalies. The risk of developing EM depends on a complex interaction between genetic, immunological, hormonal, and environmental factors. It is largely considered to arise due to a dysfunction of immunological surveillance. In fact, women with EM exhibit altered functions of peritoneal macrophages, lymphocytes and natural killer cells, as well as levels of inflammatory mediators and growth factors in the peritoneal fluid. In EM patients, peritoneal macrophages are preponderant and highly active compared to healthy women. Peritoneal macrophages are able to regulate the events that determine the production of cytokines, prostaglandins, growth factors and complement components. Several studies have shown alteration in the regulation of the complement activation, leading to chronic inflammation characteristic of EM. Aberrant regulation/activation of the complement system has been observed in the peritoneal cavity of women affected by EM. Thus, complement inhibition may represent a new approach for the treatment of EM, given that a number of complement inhibitors are under pre-clinical and clinical development. Such an intervention may provide a broader therapeutic control of complement-mediated inflammatory damage in EM patients. This review will focus on our current understanding of the role of complement activation in EM and possible modalities available for complement-based therapy.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Andrea Balduit
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Federico Romano
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy.,Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
10
|
Berciano J. Axonal degeneration in Guillain-Barré syndrome: a reappraisal. J Neurol 2020; 268:3728-3743. [PMID: 32607643 DOI: 10.1007/s00415-020-10034-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 11/28/2022]
Abstract
The aim of this review was to analyse the pathophysiology of axonal degeneration in Guillain-Barré syndrome (GBS) with emphasis on early stages (≤ 10 days after onset). An overview of experimental autoimmune neuritis (EAN) models is provided. Originally GBS and acute inflammatory demyelinating polyneuropathy were equated, presence of axonal degeneration being attributed to a "bystander" effect. Afterwards, primary axonal GBS forms were reported, designated as acute motor axonal neuropathy/acute motor-sensory axonal neuropathy. Revision of the first pathological description of axonal GBS indicates the coexistence of active axonal degeneration and demyelination in spinal roots, and pure Wallerian-like degeneration in peripheral nerve trunks. Nerve conduction studies are essential for syndrome subtyping, though their sensitivity is scanty in early GBS. Serum markers of axonal degeneration include increased levels of neurofilament light chain and presence of anti-ganglioside reactivity. According to nerve ultrasonographic features and autopsy studies, ventral rami of spinal nerves are a hotspot in early GBS. In P2-induced EAN models, the initial pathogenic change is inflammatory oedema of spinal roots and sciatic nerve, which is followed by demyelination, and Wallerian-like degeneration in nerve trunks possessing epi-perineurium; a critical elevation of endoneurial fluid pressure is a pre-requisite for inducing ischemic axonal degeneration. Similar lesion topography may occur in GBS. The repairing role of adaxonal Schwann cytoplasm in axonal degeneration is analysed. A novel pathophysiological mechanism for nerve trunk pain in GBS, including pure motor forms, is provided. The potential therapeutic role of intravenous boluses of methylprednisolone for early severe GBS and intractable pain is argued.
Collapse
Affiliation(s)
- José Berciano
- Professor Emeritus of Neurology, Service of Neurology, University Hospital "Marqués de Valdecilla (IDIVAL)", "Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)", University of Cantabria, Santander, Spain.
| |
Collapse
|
11
|
Doets AY, Hughes RAC, Brassington R, Hadden RDM, Pritchard J. Pharmacological treatment other than corticosteroids, intravenous immunoglobulin and plasma exchange for Guillain-Barré syndrome. Cochrane Database Syst Rev 2020; 1:CD008630. [PMID: 31981368 PMCID: PMC6984651 DOI: 10.1002/14651858.cd008630.pub5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Plasma exchange and intravenous immunoglobulin, but not corticosteroids, are beneficial in Guillain-Barré syndrome (GBS). The efficacy of other pharmacological agents is unknown. This review was first published in 2011 and previously updated in 2013, and 2016. OBJECTIVES To assess the effects of pharmacological agents other than plasma exchange, intravenous immunoglobulin and corticosteroids for GBS. SEARCH METHODS On 28 October 2019, we searched the Cochrane Neuromuscular Specialised Register, CENTRAL, MEDLINE, and Embase for treatments for GBS. We also searched clinical trials registries. SELECTION CRITERIA We included all randomised controlled trials (RCTs) or quasi-RCTs of acute GBS (within four weeks from onset) of all types and degrees of severity, and in individuals of all ages. We discarded trials that investigated only corticosteroids, intravenous immunoglobulin or plasma exchange. We included other pharmacological treatments or combinations of treatments compared with no treatment, placebo or another treatment. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. MAIN RESULTS We found six trials of five different interventions eligible for inclusion in this review. The trials were conducted in hospitals in Canada, China, Germany, Japan and the UK, and included 151 participants in total. All trials randomised participants aged 16 years and older (mean or median age in the trials ranged from 36 to 57 years in the intervention groups and 34 to 60 years in the control groups) with severe GBS, defined by the inability to walk unaided. One trial also randomised patients with mild GBS who were still able to walk unaided. We identified two new trials at this update.The primary outcome measure for this review was improvement in disability grade four weeks after randomisation. Four of six trials had a high risk of bias in at least one respect. We assessed all evidence for the outcome mean improvement in disability grade as very low certainty, which means that we were unable to draw any conclusions from the data. One RCT with 19 participants compared interferon beta-1a (IFNb-1a) and placebo. It is uncertain whether IFNb-1a improves disability after four weeks (mean difference (MD) -0.1; 95% CI -1.58 to 1.38; very low-certainty evidence). A trial with 10 participants compared brain-derived neurotrophic factor (BNDF) and placebo. It is uncertain whether BDNF improves disability after four weeks (MD 0.75; 95% CI -1.14 to 2.64; very low-certainty evidence). A trial with 37 participants compared cerebrospinal fluid (CSF) filtration and plasma exchange. It is uncertain whether CSF filtration improves disability after four weeks (MD 0.02; 95% CI -0.62 to 0.66; very low-certainty evidence). One trial that compared the Chinese herbal medicine tripterygium polyglycoside with corticosteroids with 43 participants did not report the risk ratio (RR) for an improvement by one or more disability grade after four weeks, but did report improvement after eight weeks. It is uncertain whether tripterygium polyglycoside improves disability after eight weeks (RR 1.47; 95% CI 1.02 to 2.11; very low-certainty evidence). We performed a meta-analysis of two trials comparing eculizumab and placebo with 41 participants. It is uncertain whether eculizumab improves disability after four weeks (MD -0.23; 95% CI -1.79 to 1.34; very low-certainty evidence). Serious adverse events were uncommon in each of the trials and evidence was graded as either low or very low. It is uncertain whether serious adverse events were more common with IFNb-1a versus placebo (RR 0.92, 95% CI 0.23 to 3.72; 19 participants), BNDF versus placebo (RR 1.00, 95% CI 0.28 to 3.54; 10 participants) or CSF filtration versus plasma exchange (RR 0.13, 95% CI 0.01 to 2.25; 37 participants). The trial of tripterygium polyglycoside did not report serious adverse events. There may be no clear difference in the number of serious adverse events after eculizumab compared to placebo (RR 1.90, 0.34 to 10.50; 41 participants). We found no clinically important differences in any of the outcome measures selected for this review in any of the six trials. However, sample sizes were small and therefore clinically important benefit or harm cannot be excluded. AUTHORS' CONCLUSIONS All six RCTs were too small to exclude clinically important benefit or harm from the assessed interventions. The certainty of the evidence was low or very low for all interventions and outcomes.
Collapse
Affiliation(s)
- Alex Y Doets
- Erasmus University Medical CentreP.O. Box 2040University Medical Centre RotterdamRotterdamNetherlands3000 CA
| | - Richard AC Hughes
- National Hospital for Neurology and NeurosurgeryMRC Centre for Neuromuscular DiseasesPO Box 114Queen SquareLondonUKWC1N 3BG
| | - Ruth Brassington
- National Hospital for Neurology and NeurosurgeryQueen Square Centre for Neuromuscular DiseasesPO Box 114LondonUKWC1N 3BG
| | - Robert DM Hadden
- King's College HospitalDepartment of NeurologyDenmark HillLondonUKSE5 9RS
| | - Jane Pritchard
- Charing Cross HospitalNeuromuscular Unit 3 NorthFulham Palace RoadLondonUKW6 8RF
| | | |
Collapse
|
12
|
Pellegrini D, Del Grosso A, Angella L, Giordano N, Dilillo M, Tonazzini I, Caleo M, Cecchini M, McDonnell LA. Quantitative Microproteomics Based Characterization of the Central and Peripheral Nervous System of a Mouse Model of Krabbe Disease. Mol Cell Proteomics 2019; 18:1227-1241. [PMID: 30926673 PMCID: PMC6553931 DOI: 10.1074/mcp.ra118.001267] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/15/2019] [Indexed: 11/06/2022] Open
Abstract
Krabbe disease is a rare, childhood lysosomal storage disorder caused by a deficiency of galactosylceramide beta-galactosidase (GALC). The major effect of GALC deficiency is the accumulation of psychosine in the nervous system and widespread degeneration of oligodendrocytes and Schwann cells, causing rapid demyelination. The molecular mechanisms of Krabbe disease are not yet fully elucidated and a definite cure is still missing. Here we report the first in-depth characterization of the proteome of the Twitcher mouse, a spontaneous mouse model of Krabbe disease, to investigate the proteome changes in the Central and Peripheral Nervous System. We applied a TMT-based workflow to compare the proteomes of the corpus callosum, motor cortex and sciatic nerves of littermate homozygous Twitcher and wild-type mice. More than 400 protein groups exhibited differences in expression and included proteins involved in pathways that can be linked to Krabbe disease, such as inflammatory and defense response, lysosomal proteins accumulation, demyelination, reduced nervous system development and cell adhesion. These findings provide new insights on the molecular mechanisms of Krabbe disease, representing a starting point for future functional experiments to study the molecular pathogenesis of Krabbe disease. Data are available via ProteomeXchange with identifier PXD010594.
Collapse
Affiliation(s)
- Davide Pellegrini
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy
| | - Ambra Del Grosso
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- ¶NEST, Istituto Nanoscienze-CNR, Pisa, Italy
| | | | | | - Marialaura Dilillo
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy
| | | | | | - Marco Cecchini
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- ¶NEST, Istituto Nanoscienze-CNR, Pisa, Italy
| | - Liam A McDonnell
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy;
- **Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Tissue-targeted complement therapeutics. Mol Immunol 2018; 102:120-128. [PMID: 30220307 DOI: 10.1016/j.molimm.2018.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023]
Abstract
Complement activation contributes to the pathogenesis of numerous inflammatory and autoimmune diseases. Therapeutic complement inhibitors have proven effective in several of these diseases and have now entered clinical use. Complement activation has multiple different biologic effects, however, and the currently available drugs can have undesirable side-effects, such as an increased risk of infection. Several different complement inhibitors have been developed that bind to target molecules, thereby concentrating the drug at a specific anatomic site. This approach appears to be both more effective than untargeted drugs and to have fewer side effects. In this article we review different targeting strategies that have been developed and the evidence supporting the use and benefits of targeted drugs.
Collapse
|
14
|
Santosa KB, Keane AM, Jablonka-Shariff A, Vannucci B, Snyder-Warwick AK. Clinical relevance of terminal Schwann cells: An overlooked component of the neuromuscular junction. J Neurosci Res 2018; 96:1125-1135. [PMID: 29536564 PMCID: PMC6292684 DOI: 10.1002/jnr.24231] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/30/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022]
Abstract
The terminal Schwann cell (tSC), a type of nonmyelinating Schwann cell, is a significant yet relatively understudied component of the neuromuscular junction. In addition to reviewing the role tSCs play on formation, maintenance, and remodeling of the synapse, we review studies that implicate tSCs in neuromuscular diseases including spinal muscular atrophy, Miller-Fisher syndrome, and amyotrophic lateral sclerosis, among others. We also discuss the importance of these cells on degeneration and regeneration after nerve injury. Knowledge of tSC biology may improve our understanding of disease pathogenesis and help us identify new and innovative therapeutic strategies for the many patients who suffer from neuromuscular disorders and nerve injuries.
Collapse
Affiliation(s)
- Katherine B. Santosa
- Postdoctoral Research Fellow, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Alexandra M. Keane
- Medical Student, Washington University School of Medicine, St. Louis, MO
| | - Albina Jablonka-Shariff
- Research Scientist, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Bianca Vannucci
- Medical Student, Washington University School of Medicine, St. Louis, MO
| | - Alison K. Snyder-Warwick
- Assistant Professor, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
15
|
Motamed-Gorji N, Matin N, Tabatabaie O, Pavone P, Romano C, Falsaperla R, Vitaliti G. Biological Drugs in Guillain-Barré Syndrome: An Update. Curr Neuropharmacol 2018; 15:938-950. [PMID: 27964705 PMCID: PMC5652014 DOI: 10.2174/1570159x14666161213114904] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022] Open
Abstract
Background: Guillain-Barré Syndrome (GBS) is currently considered the most common global cause of acute flaccid paralysis. Currently, standard therapy for Guillain-Barré Syndrome includes intravenous immunoglobulin or plasma exchange. Despite medical advances regarding these treatments, many treated patients do not reach full recovery. Therefore several biological agents have attracted the attentions from researchers during the last decades, and various studies have investigated their role in Guillain-Barré Syndrome. Objective: The present study aims to address emerging biological approaches to GBS while considering their efficiency and safety in treating the disease. Materials and Methods: An extensive electronic literature search was conducted by two researchers from April 2016 to July 2016. Original articles, clinical trials, systematic reviews (with or without meta-analysis) and case reports were selected. Titles and abstracts of papers were screened by reviewers to determine whether they met the eligibility criteria, and full texts of the selected articles were retrieved. Results: Herein authors focused on the literature data concerning emerging biological therapeutic agents, namely anti-C5 monoclonal antibody (Eculizumab), anti-C1q monoclonal antibody, anti-T cell monoclonal antibody, anti-CD2 monoclonal antibody, anti L-selectin monoclonal antibody, anti-CD20 monoclonal antibody (Rituximab), anti-CD52 monoclonal antibody (Alemtuzumab) and cytokine targets. By far, none of these agents have been approved for the treatment of GBS by FDA. Conclusion: Literature findings represented in current review herald promising results for using these biological targets. Current review represents a summary of what is already in regards and what progress is required to improve the immunotherapeutic approach of treating GBS via future studies.
Collapse
Affiliation(s)
| | - Nassim Matin
- Department of Neurology, Massachusetts General Hospital, Boston, MA. United States
| | - Omidreza Tabatabaie
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. United States
| | - Piero Pavone
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Catia Romano
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Raffaele Falsaperla
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Giovanna Vitaliti
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| |
Collapse
|
16
|
Restrepo-Jiménez P, Rodríguez Y, González P, Chang C, Gershwin ME, Anaya JM. The immunotherapy of Guillain-Barré syndrome. Expert Opin Biol Ther 2018; 18:619-631. [DOI: 10.1080/14712598.2018.1468885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Paula Restrepo-Jiménez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Paulina González
- Neurology Service, Clínica Universitaria Bolivariana, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, CA, USA
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, CA, USA
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
17
|
Schafflick D, Kieseier BC, Wiendl H, Meyer Zu Horste G. Novel pathomechanisms in inflammatory neuropathies. J Neuroinflammation 2017; 14:232. [PMID: 29179723 PMCID: PMC5704548 DOI: 10.1186/s12974-017-1001-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/13/2017] [Indexed: 12/19/2022] Open
Abstract
Inflammatory neuropathies are rare autoimmune-mediated disorders affecting the peripheral nervous system. Considerable progress has recently been made in understanding pathomechanisms of these disorders which will be essential for developing novel diagnostic and therapeutic strategies in the future. Here, we summarize our current understanding of antigenic targets and the relevance of new immunological concepts for inflammatory neuropathies. In addition, we provide an overview of available animal models of acute and chronic variants and how new diagnostic tools such as magnetic resonance imaging and novel therapeutic candidates will benefit patients with inflammatory neuropathies in the future. This review thus illustrates the gap between pre-clinical and clinical findings and aims to outline future directions of development.
Collapse
Affiliation(s)
- David Schafflick
- Department of Neurology, Westfälische Wilhems-University, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Bernd C Kieseier
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Heinz Wiendl
- Department of Neurology, Westfälische Wilhems-University, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Gerd Meyer Zu Horste
- Department of Neurology, Westfälische Wilhems-University, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
18
|
Kassimatis T, Qasem A, Douiri A, Ryan EG, Rebollo-Mesa I, Nichols LL, Greenlaw R, Olsburgh J, Smith RA, Sacks SH, Drage M. A double-blind randomised controlled investigation into the efficacy of Mirococept (APT070) for preventing ischaemia reperfusion injury in the kidney allograft (EMPIRIKAL): study protocol for a randomised controlled trial. Trials 2017; 18:255. [PMID: 28587616 PMCID: PMC5461672 DOI: 10.1186/s13063-017-1972-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/05/2017] [Indexed: 02/06/2023] Open
Abstract
Background Delayed graft function (DGF) is traditionally defined as the requirement for dialysis during the first week after transplantation. DGF is a common complication of renal transplantation, and it negatively affects short- and long-term graft outcomes. Ischaemia reperfusion injury (IRI) is a prime contributor to the development of DGF. It is well established that complement system activation plays a pivotal role in the pathogenesis of IRI. Mirococept is a highly effective complement inhibitor that can be administered ex vivo to the donor kidney just before transplantation. Preclinical and clinical evidence suggests that Mirococept inhibits inflammatory responses that follow IRI. The EMPIRIKAL trial (REC 12/LO/1334) aims to evaluate the efficacy of Mirococept in reducing the incidence of DGF in cadaveric renal transplantation. Methods/design EMPIRIKAL is a multicentre double-blind randomised case-control trial designed to test the superiority of Mirococept in the prevention of DGF in cadaveric renal allografts, as compared to standard cold perfusion fluid (Soltran®). Patients will be randomised to Mirococept or placebo (Pbo) and will be enrolled in cohorts of N = 80 with a maximum number of 7 cohorts. The first cohort will be randomised to 10 mg of Mirococept or Pbo. After the completion of each cohort, an interim analysis will be carried out in order to evaluate the dose allocation for the next cohort (possible doses: 5–25 mg). Immunosuppression therapy, antibiotic and antiviral prophylaxis will be administered as per local centre protocols. The enrolment will take approximately 24 months, and patients will be followed for 12 months. The primary endpoint is DGF, defined as the requirement for dialysis during the first week after transplantation. Secondary endpoints include duration of DGF, functional DGF, renal function at 12 months, acute rejection episodes at 6 and 12 months, primary non-function and time of hospital stay on first admission and in the first year following transplant. Safety evaluation will include the monitoring of laboratory data and the recording of all adverse events. Discussion The EMPIRIKAL trial is the first study to evaluate the efficacy of an ex vivo administered complement inhibitor (Mirococept) in preventing DGF in cadaveric human renal transplantation. Mirococept has a unique ‘cytotopic’ property that permits its retention in the organ microvasculature. Trial registration ISRCTN registry, ISRCTN49958194. Registered on 3 August 2012. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-1972-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Theodoros Kassimatis
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Anass Qasem
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Abdel Douiri
- Department of Primary Care and Public Health Sciences, King's College London, London, UK
| | - Elizabeth G Ryan
- Biostatistics and Health Informatics Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Irene Rebollo-Mesa
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,UCB Biopharma, Berkshire, UK
| | - Laura L Nichols
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Roseanna Greenlaw
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Jonathon Olsburgh
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Richard A Smith
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Steven H Sacks
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Martin Drage
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK. .,Department of Transplantation, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
19
|
Abstract
In 1916, Guillain, Barré and Strohl reported on two cases of acute flaccid paralysis with high cerebrospinal fluid protein levels and normal cell counts - novel findings that identified the disease we now know as Guillain-Barré syndrome (GBS). 100 years on, we have made great progress with the clinical and pathological characterization of GBS. Early clinicopathological and animal studies indicated that GBS was an immune-mediated demyelinating disorder, and that severe GBS could result in secondary axonal injury; the current treatments of plasma exchange and intravenous immunoglobulin, which were developed in the 1980s, are based on this premise. Subsequent work has, however, shown that primary axonal injury can be the underlying disease. The association of Campylobacter jejuni strains has led to confirmation that anti-ganglioside antibodies are pathogenic and that axonal GBS involves an antibody and complement-mediated disruption of nodes of Ranvier, neuromuscular junctions and other neuronal and glial membranes. Now, ongoing clinical trials of the complement inhibitor eculizumab are the first targeted immunotherapy in GBS.
Collapse
Affiliation(s)
- John A Goodfellow
- Neuroimmunology Laboratory, Laboratory Building, Queen Elizabeth University Hospital, Glasgow, G51 4TF, Scotland, UK
| | - Hugh J Willison
- Institute of Infection, Immunity and Inflammation, University of Glasgow, University Place, Glasgow, G12 8TA, Scotland, UK
| |
Collapse
|
20
|
Holers VM, Tomlinson S, Kulik L, Atkinson C, Rohrer B, Banda N, Thurman JM. New therapeutic and diagnostic opportunities for injured tissue-specific targeting of complement inhibitors and imaging modalities. Semin Immunol 2016; 28:260-7. [PMID: 27282113 DOI: 10.1016/j.smim.2016.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 01/27/2023]
Abstract
Despite substantial opportunity and commercial interest in developing drugs that modulate the complement system in a broad range of non-orphan indications, several obstacles remain to be overcome. Among these issues is the biophysical nature of complement proteins, whose circulating levels are typically very high and whose turnover rates are relatively rapid, especially in the setting of chronic inflammatory conditions. This situation necessitates the use of very high levels of therapeutic compounds in order to achieve both multi-pathway and multiple effector mechanism inhibition. In addition, one must avoid infectious complications or the systemic impairment of the other important physiological functions of complement. Herein we focus on the development of a novel therapeutic strategy based on injured tissue-specific targeting of complement inhibitors using the antigen-combining domains of a small subset of natural IgM antibodies, which as endogenous antibodies specifically recognize sites of local damage across a broad range of tissues and locally activate complement C3, resulting in C3 fragment covalent fixation. Because the use of such recombinant tissue-targeting inhibitors precludes the utility of measuring systemic levels of complement biomarkers or function, since a goal of this targeting strategy is to leave those processes intact and unimpeded, we also briefly describe a new method designed to quantitatively measure using imaging modalities the inhibition of generation of fixed C3 fragments at sites of inflammation/injury. In addition to the ability to determine whether complement activation is locally constrained with the use of inhibitors, there is also a broader application of this imaging approach to inflammatory and autoimmune diseases characterized by local complement activation.
Collapse
Affiliation(s)
- V Michael Holers
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Aurora, CO, United States.
| | - Stephen Tomlinson
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, United States; Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Liudmila Kulik
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States; Department of Surgery, Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC, United States
| | - Bärbel Rohrer
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, United States; Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, United States
| | - Nirmal Banda
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
21
|
McGonigal R, Cunningham ME, Yao D, Barrie JA, Sankaranarayanan S, Fewou SN, Furukawa K, Yednock TA, Willison HJ. C1q-targeted inhibition of the classical complement pathway prevents injury in a novel mouse model of acute motor axonal neuropathy. Acta Neuropathol Commun 2016; 4:23. [PMID: 26936605 PMCID: PMC4776408 DOI: 10.1186/s40478-016-0291-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/18/2023] Open
Abstract
Introduction Guillain-Barré syndrome (GBS) is an autoimmune disease that results in acute paralysis through inflammatory attack on peripheral nerves, and currently has limited, non-specific treatment options. The pathogenesis of the acute motor axonal neuropathy (AMAN) variant is mediated by complement-fixing anti-ganglioside antibodies that directly bind and injure the axon at sites of vulnerability such as nodes of Ranvier and nerve terminals. Consequently, the complement cascade is an attractive target to reduce disease severity. Recently, C5 complement component inhibitors that block the formation of the membrane attack complex and subsequent downstream injury have been shown to be efficacious in an in vivo anti-GQ1b antibody-mediated mouse model of the GBS variant Miller Fisher syndrome (MFS). However, since gangliosides are widely expressed in neurons and glial cells, injury in this model was not targeted exclusively to the axon and there are currently no pure mouse models for AMAN. Additionally, C5 inhibition does not prevent the production of early complement fragments such as C3a and C3b that can be deleterious via their known role in immune cell and macrophage recruitment to sites of neuronal damage. Results and Conclusions In this study, we first developed a new in vivo transgenic mouse model of AMAN using mice that express complex gangliosides exclusively in neurons, thereby enabling specific targeting of axons with anti-ganglioside antibodies. Secondly, we have evaluated the efficacy of a novel anti-C1q antibody (M1) that blocks initiation of the classical complement cascade, in both the newly developed anti-GM1 antibody-mediated AMAN model and our established MFS model in vivo. Anti-C1q monoclonal antibody treatment attenuated complement cascade activation and deposition, reduced immune cell recruitment and axonal injury, in both mouse models of GBS, along with improvement in respiratory function. These results demonstrate that neutralising C1q function attenuates injury with a consequent neuroprotective effect in acute GBS models and promises to be a useful new target for human therapy.
Collapse
|
22
|
Xiao F, Ma L, Zhao M, Smith RA, Huang G, Jones PM, Persaud S, Pingitore A, Dorling A, Lechler R, Lombardi G. APT070 (mirococept), a membrane-localizing C3 convertase inhibitor, attenuates early human islet allograft damage in vitro and in vivo in a humanized mouse model. Br J Pharmacol 2016; 173:575-87. [PMID: 26565566 PMCID: PMC4728428 DOI: 10.1111/bph.13388] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/03/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022] Open
Abstract
Background and Purpose A major obstacle to islet cell transplantation is the early loss of transplanted islets resulting from the instant blood‐mediated inflammation reaction (IBMIR). The activation of complement pathways plays a central role in IBMIR. The aim of this study was to test the inhibitory effect of “painting” human islets with APT070, a membrane‐localizing C3 convertase inhibitor, on inflammation evoked by exposure to human serum in vitro and by transplantation in vivo in a humanized diabetic mouse model. Experimental Approach In vitro, human islets pre‐incubated with APT070 were exposed to allogeneic whole blood. In vivo, similarly treated islets were transplanted underneath the kidney capsule of streptozotocin‐induced diabetic NOD‐SCID IL2rγ−/− mice that had been reconstituted with human CD34+ stem cells. Complement activation and islet hormone content were assayed using enzyme‐linked immunosorbent assays. Supernatants and sera were assayed for cytokines using cytometric beads array. Morphology of the islets incubated with human serum in vitro and in graft‐bearing kidney were evaluated using immunofluorescence staining. Key Results Pre‐incubation with APT070 decreased C‐peptide release and iC3b production in vitro, with diminished deposition of C4d and C5b‐9 in islets embedded in blood clots. In vivo, the APT070‐treated islets maintained intact structure and showed less infiltration of inflammatory cells than untreated islets. The pretreatments also significantly reduced pro‐inflammatory cytokines in supernatants and sera. Conclusions and Implications Pre‐treatment of islets with APT070 could reduce intra‐islet inflammation with accompanying preservation of insulin secretion by beta cells. APT070 could be as a potential therapeutic tool in islet transplantation.
Collapse
Affiliation(s)
- Fang Xiao
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, 5th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Liang Ma
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, 5th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Min Zhao
- Department of Diabetes & Endocrinology, King's College London, 2nd Floor, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU, UK
| | - Richard A Smith
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, 5th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Guocai Huang
- Department of Diabetes & Endocrinology, King's College London, 2nd Floor, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU, UK
| | - Peter M Jones
- Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Shanta Persaud
- Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Attilio Pingitore
- Division of Diabetes & Nutritional Sciences, School of Medicine, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Anthony Dorling
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, 5th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Robert Lechler
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, 5th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Giovanna Lombardi
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, 5th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| |
Collapse
|
23
|
Abstract
The severe clinical symptoms of inherited CD59 deficiency confirm the importance of CD59 as essential complement regulatory protein for protection of cells against complement attack, in particular protection of hematopoietic cells and human neuronal tissue. Targeted complement inhibition might become a treatment option as suggested by a case report. The easy diagnostic approach by flow cytometry and the advent of a new treatment option should increase the awareness of this rare differential diagnosis and lead to further studies on their pathophysiology.
Collapse
|
24
|
Abstract
The neuroglia of the peripheral nervous system (PNS) are derived from the neural crest and are a diverse family of cells. They consist of myelinating Schwann cells, non-myelinating Schwann cells, satellite cells, and perisynaptic Schwann cells. Due to their prominent role in the formation of myelin, myelinating Schwann cells are the best recognised of these cells. However, Schwann cells and the other neuroglia of the PNS have many functions that are independent of myelination and contribute significantly to the functioning of the peripheral nerve in both health and disease. Here we discuss the contribution of PNS neuroglial cells to clinical deficit in neurodegenerative disease, peripheral neuropathy, and pain.
Collapse
Affiliation(s)
- Patricia J Armati
- Neuroinflammation Group, Brain & Mind Research Institute, University of Sydney, Sydney, Australia
| | | |
Collapse
|
25
|
Harschnitz O, Jongbloed BA, Franssen H, Straver DCG, van der Pol WL, van den Berg LH. MMN: from immunological cross-talk to conduction block. J Clin Immunol 2014; 34 Suppl 1:S112-9. [PMID: 24728842 PMCID: PMC4050293 DOI: 10.1007/s10875-014-0026-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 03/19/2014] [Indexed: 12/11/2022]
Abstract
Multifocal motor neuropathy (MMN) is a rare inflammatory neuropathy characterized by progressive, asymmetric distal limb weakness and conduction block (CB). Clinically MMN is a pure motor neuropathy, which as such can mimic motor neuron disease. GM1-specific IgM antibodies are present in the serum of approximately half of all MMN patients, and are thought to play a key role in the immune pathophysiology. Intravenous immunoglobulin (IVIg) treatment has been shown to be effective in MMN in five randomized placebo-controlled trials. Despite long-term treatment with intravenous immunoglobulin (IVIg), which is efficient in the majority of patients, slowly progressive axonal degeneration and subsequent muscle weakness cannot be fully prevented. In this review, we will discuss the current understanding of the immune pathogenesis underlying MMN and how this may cause CB, available treatment strategies and future therapeutic targets.
Collapse
Affiliation(s)
- Oliver Harschnitz
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, 3584 CG The Netherlands
- Department of Translational Neuroscience, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, 3584 CG The Netherlands
| | - Bas A. Jongbloed
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, 3584 CG The Netherlands
- Department of Neurology, St. Elisabeth Hospital, Tilburg, 5000 LC The Netherlands
| | - Hessel Franssen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, 3584 CG The Netherlands
| | - Dirk C. G Straver
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, 3584 CG The Netherlands
| | - W. Ludo van der Pol
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, 3584 CG The Netherlands
| | - Leonard H. van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, 3584 CG The Netherlands
| |
Collapse
|
26
|
|
27
|
Schnaar RL, Gerardy-Schahn R, Hildebrandt H. Sialic acids in the brain: gangliosides and polysialic acid in nervous system development, stability, disease, and regeneration. Physiol Rev 2014; 94:461-518. [PMID: 24692354 DOI: 10.1152/physrev.00033.2013] [Citation(s) in RCA: 525] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Every cell in nature carries a rich surface coat of glycans, its glycocalyx, which constitutes the cell's interface with its environment. In eukaryotes, the glycocalyx is composed of glycolipids, glycoproteins, and proteoglycans, the compositions of which vary among different tissues and cell types. Many of the linear and branched glycans on cell surface glycoproteins and glycolipids of vertebrates are terminated with sialic acids, nine-carbon sugars with a carboxylic acid, a glycerol side-chain, and an N-acyl group that, along with their display at the outmost end of cell surface glycans, provide for varied molecular interactions. Among their functions, sialic acids regulate cell-cell interactions, modulate the activities of their glycoprotein and glycolipid scaffolds as well as other cell surface molecules, and are receptors for pathogens and toxins. In the brain, two families of sialoglycans are of particular interest: gangliosides and polysialic acid. Gangliosides, sialylated glycosphingolipids, are the most abundant sialoglycans of nerve cells. Mouse genetic studies and human disorders of ganglioside metabolism implicate gangliosides in axon-myelin interactions, axon stability, axon regeneration, and the modulation of nerve cell excitability. Polysialic acid is a unique homopolymer that reaches >90 sialic acid residues attached to select glycoproteins, especially the neural cell adhesion molecule in the brain. Molecular, cellular, and genetic studies implicate polysialic acid in the control of cell-cell and cell-matrix interactions, intermolecular interactions at cell surfaces, and interactions with other molecules in the cellular environment. Polysialic acid is essential for appropriate brain development, and polymorphisms in the human genes responsible for polysialic acid biosynthesis are associated with psychiatric disorders including schizophrenia, autism, and bipolar disorder. Polysialic acid also appears to play a role in adult brain plasticity, including regeneration. Together, vertebrate brain sialoglycans are key regulatory components that contribute to proper development, maintenance, and health of the nervous system.
Collapse
|
28
|
Tzekova N, Heinen A, Küry P. Molecules involved in the crosstalk between immune- and peripheral nerve Schwann cells. J Clin Immunol 2014; 34 Suppl 1:S86-104. [PMID: 24740512 DOI: 10.1007/s10875-014-0015-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022]
Abstract
Schwann cells are the myelinating glial cells of the peripheral nervous system and establish myelin sheaths on large caliber axons in order to accelerate their electrical signal propagation. Apart from this well described function, these cells revealed to exhibit a high degree of differentiation plasticity as they were shown to re- and dedifferentiate upon injury and disease as well as to actively participate in regenerative- and inflammatory processes. This review focuses on the crosstalk between glial- and immune cells observed in many peripheral nerve pathologies and summarizes functional evidences of molecules, regulators and factors involved in this process. We summarize data on Schwann cell's role presenting antigens, on interactions with the complement system, on Schwann cell surface molecules/receptors and on secreted factors involved in immune cell interactions or para-/autocrine signaling events, thus strengthening the view for a broader (patho) physiological role of this cell lineage.
Collapse
Affiliation(s)
- Nevena Tzekova
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, D-40225, Düsseldorf, Germany
| | | | | |
Collapse
|
29
|
Zhang G, Bogdanova N, Gao T, Song JJ, Cragg MS, Glennie MJ, Sheikh KA. Fcγ receptor-mediated inflammation inhibits axon regeneration. PLoS One 2014; 9:e88703. [PMID: 24523933 PMCID: PMC3921223 DOI: 10.1371/journal.pone.0088703] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 01/10/2014] [Indexed: 01/03/2023] Open
Abstract
Anti-glycan/ganglioside antibodies are the most common immune effectors found in patients with Guillain-Barré Syndrome, which is a peripheral autoimmune neuropathy. We previously reported that disease-relevant anti-glycan autoantibodies inhibited axon regeneration, which echo the clinical association of these antibodies and poor recovery in Guillain-Barré Syndrome. However, the specific molecular and cellular elements involved in this antibody-mediated inhibition of axon regeneration are not previously defined. This study examined the role of Fcγ receptors and macrophages in the antibody-mediated inhibition of axon regeneration. A well characterized antibody passive transfer sciatic nerve crush and transplant models were used to study the anti-ganglioside antibody-mediated inhibition of axon regeneration in wild type and various mutant and transgenic mice with altered expression of specific Fcγ receptors and macrophage/microglia populations. Outcome measures included behavior, electrophysiology, morphometry, immunocytochemistry, quantitative real-time PCR, and western blotting. We demonstrate that the presence of autoantibodies, directed against neuronal/axonal cell surface gangliosides, in the injured mammalian peripheral nerves switch the proregenerative inflammatory environment to growth inhibitory milieu by engaging specific activating Fcγ receptors on recruited monocyte-derived macrophages to cause severe inhibition of axon regeneration. Our data demonstrate that the antibody orchestrated Fcγ receptor-mediated switch in inflammation is one mechanism underlying inhibition of axon regeneration. These findings have clinical implications for nerve repair and recovery in antibody-mediated immune neuropathies. Our results add to the complexity of axon regeneration in injured peripheral and central nervous systems as adverse effects of B cells and autoantibodies on neural injury and repair are increasingly recognized.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Neurology, University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Nataliia Bogdanova
- Department of Neurology, University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Tong Gao
- Department of Neurology, University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Julia J. Song
- Department of Neurology, University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Mark S. Cragg
- Antibody and Vaccine Group, Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Martin J. Glennie
- Antibody and Vaccine Group, Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Kazim A. Sheikh
- Department of Neurology, University of Texas Medical School at Houston, Houston, Texas, United States of America
| |
Collapse
|
30
|
Abstract
Understanding of Guillain-Barré syndrome (GBS) has progressed substantially since the seminal 1916 report by Guillain et al. Although Guillain, Barré, and Strohl summarised the syndrome based on observations of two French infantrymen, 2012 saw the beginning of an ambitious collaborative study designed to collect detailed data from at least 1,000 patients worldwide (IGOS, www.gbsstudies.org/about-igos). Progress has been made in many areas even since GBS was last reviewed in this journal in 2009. GBS subsequently received prominent attention in light of concerns regarding H1N1 influenza vaccinations, and several large-scale surveillance studies resulted. Despite these developments, and promising pre-clinical studies, disease-modifying therapies for GBS have not substantially altered since intravenous immunoglobulin was introduced over 20 years ago. In other areas, management has improved. Antibiotic prophylaxis in ventilated patients reduces respiratory tract infection, thromboprophylaxis has reduced the risk of venous thromboembolism, and there is increasing awareness of the benefit of high-intensity rehabilitation. This article highlights some of the interesting and thought-provoking developments of the last 3 years, and is based on a plenary lecture given at the 2012 Peripheral Nerve Society (PNS) meeting.
Collapse
Affiliation(s)
- Simon Rinaldi
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
31
|
Kaida K, Kusunoki S. Guillain–Barré syndrome: update on immunobiology and treatment. Expert Rev Neurother 2014; 9:1307-19. [DOI: 10.1586/ern.09.77] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Uncini A, Yuki N. Electrophysiologic and immunopathologic correlates in Guillain–Barré syndrome subtypes. Expert Rev Neurother 2014; 9:869-84. [DOI: 10.1586/ern.09.43] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
33
|
Lehmann HC, Hughes RAC, Kieseier BC, Hartung HP. Recent developments and future directions in Guillain-Barré syndrome. J Peripher Nerv Syst 2013; 17 Suppl 3:57-70. [PMID: 23279434 DOI: 10.1111/j.1529-8027.2012.00433.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Guillain-Barré syndrome (GBS) encompasses a spectrum of acquired neuropathic conditions characterized by inflammatory demyelinating or axonal peripheral neuropathy with acute onset. Clinical and experimental studies in the past years have led to substantial progress in epidemiology, pathogenesis of GBS variants, and identification of prognostic factors relevant to treatment. In this review we provide an overview and critical assessment of the most recent developments and future directions in GBS research.
Collapse
Affiliation(s)
- Helmar C Lehmann
- Department of Neurology, Heinrich-Heine-University, Medical School, Moorenstrasse 5, Düsseldorf, Germany
| | | | | | | |
Collapse
|
34
|
Kaida K. Pathogenic roles of antiganglioside antibodies in immune-mediated neuropathies. ACTA ACUST UNITED AC 2013. [DOI: 10.1111/cen3.12007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kenichi Kaida
- Division of Neurology; Department of Internal Medicine 3; National Defense Medical College; Saitama; Japan
| |
Collapse
|
35
|
Rupp A, Cunningham ME, Yao D, Furukawa K, Willison HJ. The effects of age and ganglioside composition on the rate of motor nerve terminal regeneration following antibody-mediated injury in mice. Synapse 2013; 67:382-9. [PMID: 23401234 PMCID: PMC4495252 DOI: 10.1002/syn.21648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 02/01/2013] [Indexed: 01/08/2023]
Abstract
Gangliosides are glycosphingolipids highly enriched in neural plasma membranes, where they mediate a diverse range of functions and can act as targets for auto-antibodies present in human immune-mediated neuropathy sera. The ensuing autoimmune injury results in axonal and motor nerve terminal (mNT) degeneration. Both aging and ganglioside-deficiency have been linked to impaired axonal regeneration. To assess the effects of age and ganglioside expression on mNT regeneration in an autoimmune injury paradigm, anti-ganglioside antibodies and complement were applied to young adult and aged mice wildtype (WT) mice, mice deficient in either b- and c-series (GD3sKO) or mice deficient in all complex gangliosides (GM2sKO). The extent of mNT injury and regeneration was assessed immediately or after 5 days, respectively. Depending on ganglioside expression and antibody-specificity, either a selective mNT injury or a combined injury of mNTs and neuromuscular glial cells was elicited. Immediately after induction of the injury, between 1.5% and 11.8% of neuromuscular junctions (NMJs) in the young adult groups exhibited healthy mNTs. Five days later, most NMJs, regardless of age and strain, had recovered their mNTs. No significant differences could be observed between young and aged WT and GM2sKO mice; aged GD3sKO showed a mildly impaired rate of mNT regeneration when compared with their younger counterparts. Comparable rates were observed between all strains in the young and the aged mice. In summary, the rate of mNT regeneration following anti-ganglioside antibody and complement-mediated injury does not differ majorly between young adult and aged mice irrespective of the expression of particular gangliosides.
Collapse
Affiliation(s)
- Angie Rupp
- Neuroimmunology Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | | | | | | | | |
Collapse
|
36
|
Rupp A, Galban-Horcajo F, Bianchi E, Dondi M, Penderis J, Cappell J, Burgess K, Matiasek K, McGonigal R, Willison HJ. Anti-GM2 ganglioside antibodies are a biomarker for acute canine polyradiculoneuritis. J Peripher Nerv Syst 2013; 18:75-88. [PMID: 23521648 PMCID: PMC4854321 DOI: 10.1111/jns5.12011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acute canine polyradiculoneuritis (ACP) is considered to be the canine equivalent of the human peripheral nerve disorder Guillain-Barré syndrome (GBS); an aetiological relationship, however, remains to be demonstrated. In GBS, anti-glycolipid antibodies (Abs) are considered as important disease mediators. To address the possibility of common Ab biomarkers, the sera of 25 ACP dogs, 19 non-neurological, and 15 epileptic control dogs were screened for IgG Abs to 10 glycolipids and their 1 : 1 heteromeric complexes using combinatorial glycoarrays. Anti-GM2 ganglioside Abs were detected in 14/25 ACP dogs, and anti-GA1 Abs in one further dog. All controls except for one were negative for anti-glycolipid Abs. In this cohort of cases and controls, the glycoarray screen reached a diagnostic sensitivity of 60% and a specificity of 97%; a lower sensitivity (32%) was reported using a conventional glycolipid ELISA. To address the possible pathogenic role for anti-GM2 Abs in ACP, we identified GM2 in canine sciatic nerve by both mass spectrometry and thin layer chromatography overlay. In immunohistological studies, GM2 was localized predominantly to the abaxonal Schwann cell membrane. The presence of anti-GM2 Abs in ACP suggests that it may share a similar pathophysiology with GBS, for which it could thus be considered a naturally occurring animal model.
Collapse
Affiliation(s)
- Angie Rupp
- Neuroimmunology Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Francesc Galban-Horcajo
- Neuroimmunology Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ezio Bianchi
- Veterinary Teaching Hospital, University of Parma, Parma, Italy
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Maurizio Dondi
- Veterinary Teaching Hospital, University of Parma, Parma, Italy
| | - Jacques Penderis
- School of Veterinary Medicine, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Joanna Cappell
- Neuroimmunology Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Glasgow Polyomics, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Karl Burgess
- Glasgow Polyomics, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Kaspar Matiasek
- Section of Clinical and Comparative Neuropathology, Department of Veterinary Clinical Sciences, Ludwig-Maximilians-University, Munich, Germany
| | - Rhona McGonigal
- Neuroimmunology Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Hugh J. Willison
- Neuroimmunology Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
37
|
Stübgen JP. A review of the use of biological agents for chronic inflammatory demyelinating polyradiculoneuropathy. J Neurol Sci 2013; 326:1-9. [PMID: 23337197 DOI: 10.1016/j.jns.2013.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 12/24/2012] [Accepted: 01/03/2013] [Indexed: 12/26/2022]
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is a group of idiopathic, acquired, immune-mediated inflammatory demyelinating diseases of the peripheral nervous system. A majority of patients with CIDP respond to "first-line" treatment with IVIG, plasmapheresis and/or corticosteroids. There exists insufficient evidence to ascertain the benefit of treatment with "conventional" immunosuppressive drugs. The inconsistent efficacy, long-term financial burden and health risks of non-specific immune altering therapy have drawn recurrent attention to the possible usefulness of a variety of biological agents that target key aspects in the CIDP immunopathogenic pathways. This review aims to give an updated account of the scientific rationale and potential use of biological therapeutics in patients with CIDP. No specific treatment recommendations are given. The discovery, development and application of biological markers by modern molecular diagnostic techniques may help identify drug-naïve or treatment-resistant CIDP patients most likely to respond to targeted immunotherapy.
Collapse
Affiliation(s)
- Joerg-Patrick Stübgen
- Department of Neurology and Neuroscience, Weill Cornell Medical College/New York Presbyterian Hospital, NY 10065-4885, USA.
| |
Collapse
|
38
|
CR2-mediated targeting of complement inhibitors: bench-to-bedside using a novel strategy for site-specific complement modulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:137-54. [PMID: 23402024 DOI: 10.1007/978-1-4614-4118-2_9] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent approval of the first human complement pathway-directed therapeutics, along with high-profile genetic association studies, has catalyzed renewed biopharmaceutical interest in developing drugs that modulate the complement system. Substantial challenges remain, however, that must be overcome before widespread application of complement inhibitors in inflammatory and autoimmune diseases becomes possible. Among these challenges are the following: (1) defining the complement pathways and effector mechanisms that cause tissue injury in humans and determining whether the relative importance of each varies by disease, (2) blocking or modulating, using traditional small molecule or biologic approaches, the function of complement proteins whose circulating levels are very high and whose turnover rates are relatively rapid, especially in the setting of acute and chronic autoimmune diseases, and (3) avoiding infectious complications or impairment of other important physiological functions of complement when using systemically active complement-blocking agents. This chapter will review data that address these challenges to therapeutic development, with a focus on the development of a novel strategy of blocking specific complement pathways by targeting inhibitors using a recombinant portion of the human complement receptor type 2 (CR2/CD21) which specifically targets to sites of local complement C3 activation where C3 fragments are covalently fixed. Recently, the first of these CR2-targeted proteins has entered human phase I studies in the human disease paroxysmal nocturnal hemoglobinuria. The results of murine translational studies using CR2-targeted inhibitors strongly suggest that a guiding principle going forward in complement therapeutic development may well be to focus on developing strategies to modulate the pathway as precisely as possible by physically localizing therapeutic inhibitory effects.
Collapse
|
39
|
Hartung HP, Keller-Stanislawski B, Hughes RA, Lehmann HC. [Guillain-Barré syndrome after exposure to influenza]. DER NERVENARZT 2012; 83:714-30. [PMID: 22528062 DOI: 10.1007/s00115-012-3479-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Guillain-Barré Syndrome (GBS) is an acquired, monophasic inflammatory polyradiculoneuritis of autoimmune origin, which occurs after infection and occasionally also after vaccination. Seasonal and pandemic influenza vaccines have in particular been implicated as triggers for GBS. However, a number of recent studies indicate that infection with influenza virus may also cause GBS. This review summarizes the epidemiological and experimental data of the association of GBS with exposure to influenza antigens by immunization (including vaccines against A/H1N1/2009) and infection. Vaccination against influenza is associated with a very low risk for the occurrence of GBS. In contrast infection with influenza may play a more important role as a triggering factor for GBS than previously assumed.
Collapse
Affiliation(s)
- H-P Hartung
- Neurologische Klinik, Heinrich-Heine-Universität, Moorenstr. 5, 40225 Düsseldorf, Deutschland.
| | | | | | | |
Collapse
|
40
|
|
41
|
Fewou SN, Rupp A, Nickolay LE, Carrick K, Greenshields KN, Pediani J, Plomp JJ, Willison HJ. Anti-ganglioside antibody internalization attenuates motor nerve terminal injury in a mouse model of acute motor axonal neuropathy. J Clin Invest 2012; 122:1037-51. [PMID: 22307327 PMCID: PMC3287221 DOI: 10.1172/jci59110] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 12/21/2011] [Indexed: 01/17/2023] Open
Abstract
In the Guillain-Barré syndrome subform acute motor axonal neuropathy (AMAN), Campylobacter jejuni enteritis triggers the production of anti-ganglioside Abs (AGAbs), leading to immune-mediated injury of distal motor nerves. An important question has been whether injury to the presynaptic neuron at the neuromuscular junction is a major factor in AMAN. Although disease modeling in mice exposed to AGAbs indicates that complement-mediated necrosis occurs extensively in the presynaptic axons, evidence in humans is more limited, in comparison to the extensive injury seen at nodes of Ranvier. We considered that rapid AGAb uptake at the motor nerve terminal membrane might attenuate complement-mediated injury. We found that PC12 rat neuronal cells rapidly internalized AGAb, which were trafficked to recycling endosomes and lysosomes. Consequently, complement-mediated cytotoxicity was attenuated. Importantly, we observed the same AGAb endocytosis and protection from cytotoxicity in live mouse nerve terminals. AGAb uptake was attenuated following membrane cholesterol depletion in vitro and ex vivo, indicating that this process may be dependent upon cholesterol-enriched microdomains. In contrast, we observed minimal AGAb uptake at nodes of Ranvier, and this structure thus remained vulnerable to complement-mediated injury. These results indicate that differential endocytic processing of AGAbs by different neuronal and glial membranes might be an important modulator of site-specific injury in acute AGAb-mediated Guillain-Barré syndrome subforms and their chronic counterparts.
Collapse
Affiliation(s)
- Simon N. Fewou
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Angie Rupp
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Lauren E. Nickolay
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kathryn Carrick
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kay N. Greenshields
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - John Pediani
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jaap J. Plomp
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hugh J. Willison
- Institute of Infection, Immunity and Inflammation and
Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
Department of Neurology and
Department of Molecular Cell Biology — Group Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
42
|
Motor nerve terminal destruction and regeneration following anti-ganglioside antibody and complement-mediated injury: An in and ex vivo imaging study in the mouse. Exp Neurol 2012; 233:836-48. [DOI: 10.1016/j.expneurol.2011.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 10/27/2011] [Accepted: 12/07/2011] [Indexed: 01/10/2023]
|
43
|
Lehmann HC, Meyer Zu Horste G, Kieseier BC, Hartung HP. Pathogenesis and treatment of immune-mediated neuropathies. Ther Adv Neurol Disord 2011; 2:261-81. [PMID: 21179533 DOI: 10.1177/1756285609104792] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Immune-mediated neuropathies represent a heterogeneous spectrum of peripheral nerve disorders that can be classified according to time course, predominant involvement of motor/sensory fibers, distribution of deficits and paraclinical parameters such as electrophysiology and serum antibodies. In the last few years, significant advances have been achieved in elucidating underlying pathomechanisms, which made it possible to identify potential therapeutic targets. In this review, we discuss the latest development in pathogenesis and treatment of immune-mediated neuropathies.
Collapse
|
44
|
Kaida K, Kusunoki S. [Antiganglioside antibodies--their pathophysiological effects on Guillain-Barré syndrome and variants]. NIHON RINSHO MEN'EKI GAKKAI KAISHI = JAPANESE JOURNAL OF CLINICAL IMMUNOLOGY 2011; 34:29-39. [PMID: 21372511 DOI: 10.2177/jsci.34.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Gangliosides, N-acetylneuraminic acid (sialic acid)-bearing glycosphingolipids, are believed to reside in clusters within membrane microdomains, called lipid rafts or glycosynapse. Recent studies demonstrated that antiganglioside antibodies play an important role in the pathogenesis of Guillain-Barré syndrome (GBS) and Fisher syndrome (FS). The anti-GM1 antibodies are likely to damage peripheral nerves through complement activation with dysfunction of voltage-gated sodium channels. Some antiganglioside antibodies may cause dysfunction of voltage-gated calcium channels without complement activation. Clustered epitopes of ganglioside complexes (GSCs) consisting of two gangliosides can be targeted by serum antibodies in GBS and FS. Anti-GD1a/GD1b complex antibodies are associated with severe GBS. Approximately 50% of FS patients have antibodies to GSCs containing GQ1b or GT1a. Various glycolipids including GSCs may form complex glycolipid environment in the cell membrane, regulating the accessibility and the avidity of antiganglioside antibodies. In addition to antibody specificity, the glycolipid environment or specific distribution of target gangliosides in peripheral nervous system can influence pathogenic effects of antiganglioside antibodies in GBS and FS. Conformational and functional analyses of glycoepitopes of GSCs in the biological membrane will provide new vistas to research on antibody-antigen interaction in GBS, and shed light on microdomain function mediated by carbohydrate-to-carbohydrate interaction.
Collapse
Affiliation(s)
- Kenichi Kaida
- Division of Neurology, Department of Internal Medicine 3, National Defense Medical College, Japan
| | | |
Collapse
|
45
|
Walgaard C, Jacobs BC, van Doorn PA. Emerging drugs for Guillain-Barré syndrome. Expert Opin Emerg Drugs 2011; 16:105-20. [DOI: 10.1517/14728214.2011.531699] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
46
|
Lehmann HC, Hartung HP. Plasma exchange and intravenous immunoglobulins: mechanism of action in immune-mediated neuropathies. J Neuroimmunol 2010; 231:61-9. [PMID: 21056913 DOI: 10.1016/j.jneuroim.2010.09.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Immune-mediated neuropathies are a heterogeneous group of peripheral nerve disorders, which are classified by time course, clinical pattern, affected nerves and pathological features. Plasma exchange (PE) and intravenous immunoglobulins (IVIg) are mainstays in the treatment of immune-mediated neuropathies. Of all treatments currently used, IVIg has probably the widest application range in immune-mediated neuropathies and efficacy has been well documented in several randomized controlled trials for Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy (CIDP). Beneficial effects of IVIg have also been proven for multifocal motor neuropathy (MMN). Likewise, PE is an established treatment for GBS and CIDP, whereas it is considered to be ineffective in MMN. Different mechanisms of action are sought to be responsible for the immunemodulatory effect of PE and IVIg in autoimmune disorders. Some of those might be important for immune-mediated neuropathies, while others are probably negligible. The aim of this review is to summarize the recent advances in elucidating disease-specific mechanisms of actions of PE and IVIg in the treatment of immune-mediated neuropathies.
Collapse
Affiliation(s)
- Helmar C Lehmann
- Medical Faculty, Department of Neurology, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany.
| | | |
Collapse
|
47
|
Guillain-Barré syndrome after exposure to influenza virus. THE LANCET. INFECTIOUS DISEASES 2010; 10:643-51. [PMID: 20797646 DOI: 10.1016/s1473-3099(10)70140-7] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Guillain-Barré syndrome (GBS) is an acute, acquired, monophasic autoimmune disorder of peripheral nerves that develops in susceptible individuals after infection and, in rare cases, after immunisation. Exposure to influenza via infection or vaccination has been associated with GBS. We review the relation between GBS and these routes of exposure. Epidemiological studies have shown that, except for the 1976 US national immunisation programme against swine-origin influenza A H1N1 subtype A/NJ/76, influenza vaccine has probably not caused GBS or, if it has, rates have been extremely low (less than one case per million vaccine recipients). By contrast, influenza-like illnesses seem to be relevant triggering events for GBS. The concerns about the risk of inducing GBS in mass immunisation programmes against H1N1 2009 do not, therefore, seem justified by the available epidemiological data. However, the experiences from the 1976 swine flu vaccination programme emphasise the importance for active and passive surveillance to monitor vaccine safety.
Collapse
|
48
|
McGonigal R, Rowan EG, Greenshields KN, Halstead SK, Humphreys PD, Rother RP, Furukawa K, Willison HJ. Anti-GD1a antibodies activate complement and calpain to injure distal motor nodes of Ranvier in mice. ACTA ACUST UNITED AC 2010; 133:1944-60. [PMID: 20513658 DOI: 10.1093/brain/awq119] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The motor axonal variant of Guillain-Barré syndrome is associated with anti-GD1a immunoglobulin antibodies, which are believed to be the pathogenic factor. In previous studies we have demonstrated the motor terminal to be a vulnerable site. Here we show both in vivo and ex vivo, that nodes of Ranvier in intramuscular motor nerve bundles are also targeted by anti-GD1a antibody in a gradient-dependent manner, with greatest vulnerability at distal nodes. Complement deposition is associated with prominent nodal injury as monitored with electrophysiological recordings and fluorescence microscopy. Complete loss of nodal protein staining, including voltage-gated sodium channels and ankyrin G, occurs and is completely protected by both complement and calpain inhibition, although the latter provides no protection against electrophysiological dysfunction. In ex vivo motor and sensory nerve trunk preparations, antibody deposits are only observed in experimentally desheathed nerves, which are thereby rendered susceptible to complement-dependent morphological disruption, nodal protein loss and reduced electrical activity of the axon. These studies provide a detailed mechanism by which loss of axonal conduction can occur in a distal dominant pattern as observed in a proportion of patients with motor axonal Guillain-Barré syndrome, and also provide an explanation for the occurrence of rapid recovery from complete paralysis and electrophysiological in-excitability. The study also identifies therapeutic approaches in which nodal architecture can be preserved.
Collapse
Affiliation(s)
- Rhona McGonigal
- University of Glasgow Division of Clinical Neurosciences, Glasgow Biomedical Research Centre, Room B330, 120 University Place, Glasgow G12 8TA, UK
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Sheikh KA, Zhang G. An update on pathobiologic roles of anti-glycan antibodies in Guillain-Barré syndrome. F1000 BIOLOGY REPORTS 2010; 2. [PMID: 20948812 PMCID: PMC2948347 DOI: 10.3410/b2-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Anti-glycan antibodies directed against gangliosides are now considered the major immune effectors that induce damage to intact nerve fibers in some variants of the monophasic neuropathic disorders that comprise Guillain-Barré syndrome. Recent experimental studies elucidating the complexity of anti-glycan antibody-mediated pathobiologic effects on intact and injured nerves undergoing repair are discussed.
Collapse
Affiliation(s)
- Kazim A Sheikh
- Department of Neurology, University of Texas Medical School at Houston 6431 Fannin Street, Houston, TX 77030 USA
| | | |
Collapse
|
50
|
Kaida K, Kusunoki S. Antibodies to gangliosides and ganglioside complexes in Guillain-Barré syndrome and Fisher syndrome: mini-review. J Neuroimmunol 2010; 223:5-12. [PMID: 20172612 DOI: 10.1016/j.jneuroim.2010.02.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/01/2010] [Accepted: 02/02/2010] [Indexed: 01/05/2023]
Abstract
Antiganglioside antibodies play a pathogenic role in the pathophysiology of Guillain-Barré syndrome (GBS) and Fisher syndrome (FS). Antiganglioside antibody-mediated nerve injury is likely to result from nerve damage through complement activation or dysfunction of molecules such as voltage-gated sodium and calcium channels. Clustered epitopes of complexes of two gangliosides in the cell membrane can be targeted by serum antibodies in GBS and FS and may regulate the accessibility and avidity of antiganglioside antibodies. The glycolipid environment or the specific distribution of target gangliosides in the peripheral nervous system may also influence the pathogenic effect of antiganglioside antibodies in GBS and FS. Structural and functional analyses of glycoepitopes of ganglioside complexes in membranes will provide new vistas on antibody-antigen interaction in GBS and shed light on microdomain function mediated by carbohydrate-carbohydrate interactions, which may lead to novel treatments for GBS and FS.
Collapse
Affiliation(s)
- Kenichi Kaida
- Division of Neurology, Department of Internal Medicine 3, National Defense Medical College, Tokorozawa, Saitama-ken, Japan
| | | |
Collapse
|