1
|
Llansola M, Izquierdo-Altarejos P, Montoliu C, Mincheva G, Palomares-Rodriguez A, Pedrosa MA, Arenas YM, Felipo V. Role of peripheral inflammation in minimal hepatic encephalopathy. Metab Brain Dis 2024; 39:1667-1677. [PMID: 39177864 DOI: 10.1007/s11011-024-01417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
Many patients with liver cirrhosis show minimal hepatic encephalopathy (MHE) with mild cognitive impairment (MCI) and motor alterations that reduce their quality of life. Some patients with steatotic liver disease also suffer MCI. To design treatments to improve MHE/MCI it is necessary to understand the mechanisms by which liver disease induce them. This review summarizes studies showing that appearance of MHE/MCI is associated with a shift in the immunophenotype leading to an "autoimmune-like" form with increased pro-inflammatory monocytes, enhanced CD4 T and B lymphocytes activation and increased plasma levels of pro-inflammatory cytokines, including IL-17, IL-21, TNFα, IL-15 and CCL20. The contribution of peripheral inflammation to trigger MHE is supported by studies in animal models and by the fact that rifaximin treatment reverses MHE in around 60% of patients in parallel with reversal of the changes in peripheral inflammation. MHE does not improve in patients in which peripheral inflammation is not improved by rifaximin. The process by which peripheral inflammation induces MHE involves induction of neuroinflammation in brain, with activation of microglia and astrocytes and increased pro-inflammatory TNFα and IL-1β, which is observed in patients who died with steatotic liver disease (SLD) or liver cirrhosis and in animal models of MHE. Neuroinflammation alters glutamatergic and GABAergic neurotransmission, leading to cognitive and motor impairment. Transmission of peripheral alterations into the brain is mediated by infiltration in brain of extracellular vesicles from plasma and of cells from the peripheral immune system. Acting on any step of the process peripheral inflammation - neuroinflammation - altered neurotransmission may improve MHE.
Collapse
Affiliation(s)
- Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | - Carmina Montoliu
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain
- Fundación de Investigación Hospital Clínico Universitario de Valencia-INCLIVA, Valencia, Spain
| | - Gergana Mincheva
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | - María A Pedrosa
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Yaiza M Arenas
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| |
Collapse
|
2
|
Aharoni R, Milo R, Arnon R. Glatiramer Acetate for the Treatment of Multiple Sclerosis: From First-Generation Therapy to Elucidation of Immunomodulation and Repair. Pharmacol Rev 2024; 76:1133-1158. [PMID: 39406508 DOI: 10.1124/pharmrev.124.000927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 10/18/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), with a putative autoimmune origin and complex pathogenesis. Modification of the natural history of MS by reducing relapses and slowing disability accumulation was first attained in the 1990 s with the development of the first-generation disease-modifying therapies. Glatiramer acetate (GA), a copolymer of L-alanine, L-lysine, L-glutamic acid, and L-tyrosine, was discovered due to its ability to suppress the animal model of MS, experimental autoimmune encephalomyelitis. Extensive clinical trials and long-term assessments established the efficacy and the safety of GA. Furthermore, studies of the therapeutic processes induced by GA in animal models and in MS patients indicate that GA affects various levels of the innate and the adaptive immune response, generating deviation from proinflammatory to anti-inflammatory pathways. This includes competition for binding to antigen presenting cells; driving dendritic cells, monocytes, and B-cells toward anti-inflammatory responses; and stimulating T-helper 2 and T-regulatory cells. The immune cells stimulated by GA reach the CNS and secrete in situ anti-inflammatory cytokines alleviating the pathological processes. Furthermore, cumulative findings reveal that in addition to its immunomodulatory effect, GA promotes neuroprotective repair processes such as neurotrophic factors secretion, remyelination, and neurogenesis. This review aims to provide an overview of MS pathology diagnosis and treatment as well as the diverse mechanism of action of GA. SIGNIFICANCE STATEMENT: Understanding the complex MS immune pathogenesis provided multiple targets for therapeutic intervention, resulting in a plethora of agents, with various mechanisms of action, efficacy, and safety profiles. However, promoting repair beyond the body's limited spontaneous extent is still a major challenge. GA, one of the first approved disease-modifying therapies, induces diverse immunomodulatory effects. Furthermore, GA treatment results in elevated neurotrophic factors secretion, remyelination and neurogenesis, supporting the notion that immunomodulatory treatment can support in situ a growth-promoting and repair environment.
Collapse
Affiliation(s)
- Rina Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ron Milo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ruth Arnon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| |
Collapse
|
3
|
Nevalainen T, Autio-Kimura A, Hurme M. Human endogenous retrovirus W in multiple sclerosis: transcriptional activity is associated with decline in oligodendrocyte proportions in the white matter of the brain. J Neurovirol 2024; 30:393-405. [PMID: 38717678 PMCID: PMC11512866 DOI: 10.1007/s13365-024-01208-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/22/2024] [Accepted: 04/03/2024] [Indexed: 10/28/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease. One of the basic mechanisms in this disease is the autoimmune response against the myelin sheet leading to axonal damage. There is strong evidence showing that this response is regulated by both genetic and environmental factors. In addition, the role of viruses has been extensively studied, especially in the case of human endogenous retroviruses (HERVs). However, although several associations with MS susceptibility, especially in the case of HERV-W family have been observed, the pathogenic mechanisms have remained enigmatic. To clarify these HERV-mediated mechanisms as well as the responsible HERV-W loci, we utilized RNA sequencing data obtained from the white matter of the brain of individuals with and without MS. CIBERSORTx tool was applied to estimate the proportions of neuronal, glial, and endothelial cells in the brain. In addition, the transcriptional activity of 215 HERV-W loci were analyzed. The results indicated that 65 HERV-W loci had detectable expression, of which 14 were differentially expressed between MS and control samples. Of these, 12 HERV-W loci were upregulated in MS. Expression levels of the 8 upregulated HERV-W loci had significant negative correlation with estimated oligodendrocyte proportions, suggesting that they are associated with the dynamics of oligodendrocyte generation and/or maintenance. Furthermore, Gene Set Enrichment Analysis (GSEA) results indicated that expression levels of three upregulated HERV-W loci: 2p16.2, 2q13, and Xq13.3, are associated with suppression of oligodendrocyte development and myelination. Taken together, these data suggest new HERV-W loci candidates that might take part in MS pathogenesis.
Collapse
Affiliation(s)
- Tapio Nevalainen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
- Gerontology Research Center (GEREC), Tampere, Finland.
- Tampere University Hospital, Tampere, Finland.
| | - Arttu Autio-Kimura
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Gerontology Research Center (GEREC), Tampere, Finland
| | - Mikko Hurme
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
- Gerontology Research Center (GEREC), Tampere, Finland
- Tampere University Hospital, Tampere, Finland
| |
Collapse
|
4
|
Elendu C, Amaechi DC, Elendu TC, Ozigis MO, Adegbola MO, Adebayo MA, Afolabi OG. Renal disease associated with multiple sclerosis: A narrative review. Medicine (Baltimore) 2024; 103:e38222. [PMID: 38758897 PMCID: PMC11098179 DOI: 10.1097/md.0000000000038222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 03/14/2024] [Indexed: 05/19/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune neurological disorder characterized by central nervous system demyelination, leading to various neurological impairments. While the primary focus of research and clinical management has centered on the neurological aspects of MS, emerging evidence suggests a complex interplay between MS and renal disease. This narrative review endeavors to elucidate the intriguing association between MS and renal disease, providing a comprehensive overview of the current knowledge on this topic. Our review begins by outlining the pathophysiology of MS and the diverse mechanisms contributing to its progression. We then delve into renal disease, categorizing the various types and their clinical presentations. This review focuses on exploring the intricate relationship between these seemingly distinct conditions. We analyze existing literature to uncover shared risk factors, potential pathophysiological links, and the impact of MS on renal function. Furthermore, we discuss the clinical presentation and diagnostic challenges in identifying renal disease in MS patients. Importantly, we examine available treatment options and their efficacy in managing renal complications in this unique patient population. The consequences of renal disease on the overall quality of life (QOL) for individuals living with MS are also examined, shedding light on the multifaceted burden of these coexisting conditions.
Collapse
|
5
|
Imraish A, Abu Thiab T, Alsalem M, Dahbour S, khleif H, Abu-Irmaileh B, Qasem R, El-Salem K. The neuroprotective effect of human primary astrocytes in multiple sclerosis: In vitro model. PLoS One 2024; 19:e0300203. [PMID: 38564643 PMCID: PMC10987000 DOI: 10.1371/journal.pone.0300203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Recent studies highlighted the role of astrocytes in neuroinflammatory diseases, particularly multiple sclerosis, interacting closely with other CNS components but also with the immune cells. However, due to the difficulty in obtaining human astrocytes, their role in these pathologies is still unclear. In this study we develop an astrocyte in vitro model to evaluate their role in multiple sclerosis after being treated with CSF isolated from both healthy and MS diagnosed patients. Gene expression and ELISA assays reveal that several pro-inflammatory markers IL-1β, TNF-α and IL-6, were significantly downregulated in astrocytes treated with MS-CSF. In contrast, neurotrophic survival, and growth factors, and GFAP, BDNF, GDNF and VEGF, were markedly elevated upon the same treatment. In summary, this study supports the notion of the astrocyte involvement in MS. The results reveal the neuroprotective role of astrocyte in MS pathogenicity by suppressing excessive inflammation and increasing the expression of tropic factors.
Collapse
Affiliation(s)
- Amer Imraish
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Tuqa Abu Thiab
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Mohammad Alsalem
- Department of Anatomy and Histology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Saeed Dahbour
- Department of Neurology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Hiba khleif
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | | | - Raneen Qasem
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Khalid El-Salem
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
6
|
Slavov G. Changes in serum cytokine profile and deficit severity in patients with relapsing-remitting multiple sclerosis. Folia Med (Plovdiv) 2023; 65:625-630. [PMID: 37655382 DOI: 10.3897/folmed.65.e86576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 10/25/2022] [Indexed: 09/02/2023] Open
Abstract
INTRODUCTION In experimental autoimmune encephalomyelitis, neurological deficit correlates with axonal loss and the CD8+ T cells are a likely mediator of axonal damage. In relapsing-remitting multiple sclerosis, there is a correlation of the immune inflammatory activity in the lesion foci with the axon transection.
Collapse
|
7
|
Chaney AM, Cropper HC, Jain P, Wilson E, Simonetta F, Johnson EM, Alam IS, Patterson ITJ, Swarovski M, Stevens MY, Wang Q, Azevedo C, Nagy SC, Ramos Benitez J, Deal EM, Vogel H, Andreasson KI, James ML. PET imaging of TREM1 identifies CNS-infiltrating myeloid cells in a mouse model of multiple sclerosis. Sci Transl Med 2023; 15:eabm6267. [PMID: 37379371 DOI: 10.1126/scitranslmed.abm6267] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system (CNS) that causes substantial morbidity and diminished quality of life. Evidence highlights the central role of myeloid lineage cells in the initiation and progression of MS. However, existing imaging strategies for detecting myeloid cells in the CNS cannot distinguish between beneficial and harmful immune responses. Thus, imaging strategies that specifically identify myeloid cells and their activation states are critical for MS disease staging and monitoring of therapeutic responses. We hypothesized that positron emission tomography (PET) imaging of triggering receptor expressed on myeloid cells 1 (TREM1) could be used to monitor deleterious innate immune responses and disease progression in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. We first validated TREM1 as a specific marker of proinflammatory, CNS-infiltrating, peripheral myeloid cells in mice with EAE. We show that the 64Cu-radiolabeled TREM1 antibody-based PET tracer monitored active disease with 14- to 17-fold higher sensitivity than translocator protein 18 kDa (TSPO)-PET imaging, the established approach for detecting neuroinflammation in vivo. We illustrate the therapeutic potential of attenuating TREM1 signaling both genetically and pharmacologically in the EAE mice and show that TREM1-PET imaging detected responses to an FDA-approved MS therapy with siponimod (BAF312) in these animals. Last, we observed TREM1+ cells in clinical brain biopsy samples from two treatment-naïve patients with MS but not in healthy control brain tissue. Thus, TREM1-PET imaging has potential for aiding in the diagnosis of MS and monitoring of therapeutic responses to drug treatment.
Collapse
Affiliation(s)
- Aisling M Chaney
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Haley C Cropper
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Poorva Jain
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Edward Wilson
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Federico Simonetta
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Geneva 1205, Switzerland
- Translational Research Centre in Onco-Haematology, Faculty of Medicine, University of Geneva, Geneva 1205, Switzerland
| | - Emily M Johnson
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Israt S Alam
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Ian T J Patterson
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Michelle Swarovski
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Marc Y Stevens
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Qian Wang
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Carmen Azevedo
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Sydney C Nagy
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Javier Ramos Benitez
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Emily M Deal
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Michelle L James
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Sharifian A, Varshosaz J, Aliomrani M, Kazemi M. Nose to brain delivery of ibudilast micelles for treatment of multiple sclerosis in an experimental autoimmune encephalomyelitis animal model. Int J Pharm 2023; 638:122936. [PMID: 37030640 DOI: 10.1016/j.ijpharm.2023.122936] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Multiple sclerosis is a chronic inflammatory disease of the central nervous system ultimate to neurodegeneration and demyelination. Ibudilast is a phosphodiesterase inhibitor, effective on the function of glial cells and lymphocytes, and inhibits the release of TNF-α by inflammatory cells. Dysregulation of glia is one of the most important pathological causes of MS. Therefore, ibudilast as a glial attenuator can be a useful treatment. The objective of the present study was to investigate the effect of nasal spray of polydopamine coated micelles of surfactin, a biosurfactant, loaded with ibudilast on its brain targeted delivery and effectiveness in remylination and neuroprotection in animal model of MS. In animal studies the micelles were administrated intranasally in different doses of 10, 25 and 50 mg/kg/day in C57/BL6 mice immunized by experimental autoimmune encephalomyelitis (EAE) model. The results of Luxol fast blue staining indicated increment in myelin fiber percent more significantly (p<0.05) in the groups treated with the polydopamine coated micelles (PDAM) compared to nasal spray of free drug or oral administration. These formulations also increased expression of Mbp, Olig2 and Mog genes in the corpus callosum. These results suggest a positive outcome of polydopamine coated micelles loaded with ibudilast in active MS as an anti-inflammatory and neuroprotective agent.
Collapse
Affiliation(s)
- Akram Sharifian
- Department of Pharmaceutics, Novel Drug Delivery Systems Research Centre, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Jaleh Varshosaz
- Department of Pharmaceutics, Novel Drug Delivery Systems Research Centre, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mehdi Aliomrani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Kazemi
- Department of Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
9
|
Kumar N, Sahoo NK, Mehan S, Verma B. The importance of gut-brain axis and use of probiotics as a treatment strategy for multiple sclerosis. Mult Scler Relat Disord 2023; 71:104547. [PMID: 36805171 DOI: 10.1016/j.msard.2023.104547] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/16/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
It has been shown that the dysbiosis of the gut's microbes substantially impacts CNS illnesses, including Alzheimer's, Parkinson's, autism, and autoimmune diseases like multiple sclerosis (MS). MS is a CNS-affected autoimmune demyelination condition. Through a two-way communication pathway known as the gut-brain axis, gut microbes communicate with the CNS. When there is a disruption in the gut microbiome, cytokines and other immune cells are secreted, which affects the BBB and gastrointestinal permeability. Recent research using animal models has revealed that the gut microbiota may greatly influence the pathophysiology of EAE/MS. Any change in the gut might increase inflammatory cytokinesand affect the quantity of SCFAs, and other metabolites that cause neuroinflammation and demyelination. In- vivo and in-vitro studies have concluded that probiotics affect the immune system and can be utilized to treat gastrointestinal dysbiosis. Any alteration in the gut microbial composition caused by probiotic intake may serve as a preventive and treatment strategy for MS. The major goal of this review is to emphasize an overview of recent research on the function of gut microbiota in the onset of MS and how probiotics have a substantial impact on gastrointestinal disruption in MS and other neuro disorders. It will be easier to develop new therapeutic approaches, particularly probiotic-based supplements, for treating multiple sclerosis (MS) if we know the link between the gut and CNS.
Collapse
Affiliation(s)
- Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India.
| | - Nalini Kanta Sahoo
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, (An Autonomous College), Moga, Punjab 142001, India
| | - Bharti Verma
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| |
Collapse
|
10
|
Reaction-diffusion models in weighted and directed connectomes. PLoS Comput Biol 2022; 18:e1010507. [DOI: 10.1371/journal.pcbi.1010507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 11/23/2022] [Accepted: 08/22/2022] [Indexed: 11/07/2022] Open
Abstract
Connectomes represent comprehensive descriptions of neural connections in a nervous system to better understand and model central brain function and peripheral processing of afferent and efferent neural signals. Connectomes can be considered as a distinctive and necessary structural component alongside glial, vascular, neurochemical, and metabolic networks of the nervous systems of higher organisms that are required for the control of body functions and interaction with the environment. They are carriers of functional epiphenomena such as planning behavior and cognition, which are based on the processing of highly dynamic neural signaling patterns. In this study, we examine more detailed connectomes with edge weighting and orientation properties, in which reciprocal neuronal connections are also considered. Diffusion processes are a further necessary condition for generating dynamic bioelectric patterns in connectomes. Based on our high-precision connectome data, we investigate different diffusion-reaction models to study the propagation of dynamic concentration patterns in control and lesioned connectomes. Therefore, differential equations for modeling diffusion were combined with well-known reaction terms to allow the use of connection weights, connectivity orientation and spatial distances.
Three reaction-diffusion systems Gray-Scott, Gierer-Meinhardt and Mimura-Murray were investigated. For this purpose, implicit solvers were implemented in a numerically stable reaction-diffusion system within the framework of neuroVIISAS. The implemented reaction-diffusion systems were applied to a subconnectome which shapes the mechanosensitive pathway that is strongly affected in the multiple sclerosis demyelination disease. It was found that demyelination modeling by connectivity weight modulation changes the oscillations of the target region, i.e. the primary somatosensory cortex, of the mechanosensitive pathway.
In conclusion, a new application of reaction-diffusion systems to weighted and directed connectomes has been realized. Because the implementation were performed in the neuroVIISAS framework many possibilities for the study of dynamic reaction-diffusion processes in empirical connectomes as well as specific randomized network models are available now.
Collapse
|
11
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
12
|
Pearse DD, Hefley AB, Morales AA, Ghosh M. Comparative Profiling of TG2 and Its Effectors in Human Relapsing Remitting and Progressive Multiple Sclerosis. Biomedicines 2022; 10:biomedicines10061241. [PMID: 35740263 PMCID: PMC9220003 DOI: 10.3390/biomedicines10061241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple Sclerosis (MS) is a chronic CNS autoimmune disease characterized by immune-mediated demyelination, axon loss, and disability. Dysregulation of transglutaminase-2 (TG2) has been implicated in disease initiation and progression. Herein, TG2 expression in post-mortem human brain tissue from Relapsing Remitting MS (RRMS) or Progressive MS (PMS) individuals were examined and correlated with the presence of TG2 binding partners and effectors implicated in the processes of inflammation, scar formation, and the antagonism of repair. Tissues from Relapsing-Remitting Multiple Sclerosis (RRMS; n = 6), Progressive Multiple Sclerosis (PMS; n = 5), and non-MS control (n = 6) patients underwent immunohistochemistry for TG2, PLA2, COX-2, FN, CSPG, and HSPG. TG2 was strongly upregulated in active RRMS and PMS lesions, within blood vessels and the perivascular tissue of sclerotic plaques. TG2 colocalization was observed with GFAP+ astrocytes and ECM, including FN, HSPG, and CSPG, which also increased in either RRMS or PMS lesions. Although TG2 was not colocalized with inflammatory mediators COX-2 and PLA2, or the macrophage-microglia marker Iba1, its increased expression correlated with their elevation in active RRMS and PMS lesions. In summary, the correlation of strong TG2 induction in either RRMS or PMS with some of its binding partners but not others implicates potentially different roles for TG2 in disparate MS forms that may warrant further investigation.
Collapse
Affiliation(s)
- Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (D.D.P.); (A.B.H.); (A.A.M.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Andrew B. Hefley
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (D.D.P.); (A.B.H.); (A.A.M.)
| | - Alejo A. Morales
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (D.D.P.); (A.B.H.); (A.A.M.)
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (D.D.P.); (A.B.H.); (A.A.M.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- Correspondence: ; Tel.: +1-305-243-9968; Fax: +1-305-243-3923
| |
Collapse
|
13
|
The brainstem connectome database. Sci Data 2022; 9:168. [PMID: 35414055 PMCID: PMC9005652 DOI: 10.1038/s41597-022-01219-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/25/2022] [Indexed: 11/29/2022] Open
Abstract
Connectivity data of the nervous system and subdivisions, such as the brainstem, cerebral cortex and subcortical nuclei, are necessary to understand connectional structures, predict effects of connectional disorders and simulate network dynamics. For that purpose, a database was built and analyzed which comprises all known directed and weighted connections within the rat brainstem. A longterm metastudy of original research publications describing tract tracing results form the foundation of the brainstem connectome (BC) database which can be analyzed directly in the framework neuroVIISAS. The BC database can be accessed directly by connectivity tables, a web-based tool and the framework. Analysis of global and local network properties, a motif analysis, and a community analysis of the brainstem connectome provides insight into its network organization. For example, we found that BC is a scale-free network with a small-world connectivity. The Louvain modularity and weighted stochastic block matching resulted in partially matching of functions and connectivity. BC modeling was performed to demonstrate signal propagation through the somatosensory pathway which is affected in Multiple sclerosis. Measurement(s) | brainstem | Technology Type(s) | tract tracing metastudy | Factor Type(s) | brain region | Sample Characteristic - Organism | Rattus rattus | Sample Characteristic - Environment | Experimental setup | Sample Characteristic - Location | Germany |
Collapse
|
14
|
Maghrebi O, Belghith M, Jeridi C, Rachdi A, Fatnassi FN, Saied Z, Belal S, Ben Sassi S, Barbouche MR. B Cells Specific CpG Induces High IL-10 and IL-6 Expression In Vitro in Neuro-Behçet's Disease. Cells 2022; 11:cells11081306. [PMID: 35455984 PMCID: PMC9025002 DOI: 10.3390/cells11081306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/04/2022] [Accepted: 02/17/2022] [Indexed: 01/25/2023] Open
Abstract
Remitting-RelapsingMultiple Sclerosis (RRMS) and Neuro-Behçet Disease (NBD) are two chronic neuroinflammatory disorders leading to neurological damage. Herein, we investigated in these patients the IL-10-producing cells during the early stages of these disorders. Cellular and molecular investigations were carried out on treatment naive patients suffering from RRMS and NBD recruited at the first episode of clinical relapse. Our findings demonstrate that CSF-B cells from NBD patients, but not RRMS, are the major source of intrathecal IL-10 as compared to T-CD4 cells. Moreover, we showed a lower expression of TGF-β and IL35, in the CSF cells of NBD patients as compared to the control group. Specific in vitro CpG stimulation of peripheral blood B cells from NBD patients resulted in a concomitant early mRNA expression of IL6 and IL10 but was limited to IL10 for RRMS patients. Furthermore, mRNA expression of IL-6 and IL-10 receptors was assessed and intriguingly IL6ST receptor subunit was significantly lower in NBD CSF, but not RRMS while IL10RB was increased in both. Deciphering the role of increased IL-10-producing B cells and IL10RB despite relapsing disease as well as the discordant expression of IL6 and IL6ST may pave the way for a better understanding of the pathophysiology of these neuro-inflammatory disorders.
Collapse
Affiliation(s)
- Olfa Maghrebi
- Department of Biology, Tunis El Manar University, Tunis 1068, Tunisia;
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis 1002, Tunisia;
- Faculty of Medicine, Tunis El Manar University, Tunis 1007, Tunisia; (S.B.); (S.B.S.)
| | - Meriam Belghith
- Department of Biology, Tunis El Manar University, Tunis 1068, Tunisia;
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis 1002, Tunisia;
- Correspondence: ; Tel.: +216-718-43-755
| | - Cyrine Jeridi
- Neurology Department, Mongi Ben Hamida National Institute of Neurology, Tunis 1007, Tunisia; (C.J.); (A.R.); (F.N.F.); (Z.S.)
| | - Amine Rachdi
- Neurology Department, Mongi Ben Hamida National Institute of Neurology, Tunis 1007, Tunisia; (C.J.); (A.R.); (F.N.F.); (Z.S.)
| | - Fatma Nabli Fatnassi
- Neurology Department, Mongi Ben Hamida National Institute of Neurology, Tunis 1007, Tunisia; (C.J.); (A.R.); (F.N.F.); (Z.S.)
| | - Zakaria Saied
- Neurology Department, Mongi Ben Hamida National Institute of Neurology, Tunis 1007, Tunisia; (C.J.); (A.R.); (F.N.F.); (Z.S.)
| | - Samir Belal
- Faculty of Medicine, Tunis El Manar University, Tunis 1007, Tunisia; (S.B.); (S.B.S.)
- Neurology Department, Mongi Ben Hamida National Institute of Neurology, Tunis 1007, Tunisia; (C.J.); (A.R.); (F.N.F.); (Z.S.)
| | - Samia Ben Sassi
- Faculty of Medicine, Tunis El Manar University, Tunis 1007, Tunisia; (S.B.); (S.B.S.)
- Neurology Department, Mongi Ben Hamida National Institute of Neurology, Tunis 1007, Tunisia; (C.J.); (A.R.); (F.N.F.); (Z.S.)
| | - Mohamed-Ridha Barbouche
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis 1002, Tunisia;
- Faculty of Medicine, Tunis El Manar University, Tunis 1007, Tunisia; (S.B.); (S.B.S.)
| |
Collapse
|
15
|
Scalabrino G. Newly Identified Deficiencies in the Multiple Sclerosis Central Nervous System and Their Impact on the Remyelination Failure. Biomedicines 2022; 10:biomedicines10040815. [PMID: 35453565 PMCID: PMC9026986 DOI: 10.3390/biomedicines10040815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains enigmatic and controversial. Myelin sheaths in the central nervous system (CNS) insulate axons and allow saltatory nerve conduction. MS brings about the destruction of myelin sheaths and the myelin-producing oligodendrocytes (ODCs). The conundrum of remyelination failure is, therefore, crucial in MS. In this review, the roles of epidermal growth factor (EGF), normal prions, and cobalamin in CNS myelinogenesis are briefly summarized. Thereafter, some findings of other authors and ourselves on MS and MS-like models are recapitulated, because they have shown that: (a) EGF is significantly decreased in the CNS of living or deceased MS patients; (b) its repeated administration to mice in various MS-models prevents demyelination and inflammatory reaction; (c) as was the case for EGF, normal prion levels are decreased in the MS CNS, with a strong correspondence between liquid and tissue levels; and (d) MS cobalamin levels are increased in the cerebrospinal fluid, but decreased in the spinal cord. In fact, no remyelination can occur in MS if these molecules (essential for any form of CNS myelination) are lacking. Lastly, other non-immunological MS abnormalities are reviewed. Together, these results have led to a critical reassessment of MS pathogenesis, partly because EGF has little or no role in immunology.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
16
|
Perez GA, Villarraso JC. An Entropy Approach to Multiple Sclerosis Identification. J Pers Med 2022; 12:jpm12030398. [PMID: 35330398 PMCID: PMC8948909 DOI: 10.3390/jpm12030398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a relatively common neurodegenerative illness that frequently causes a large level of disability in patients. While its cause is not fully understood, it is likely due to a combination of genetic and environmental factors. Diagnosis of multiple sclerosis through a simple clinical examination might be challenging as the evolution of the illness varies significantly from patient to patient, with some patients experiencing long periods of remission. In this regard, having a quick and inexpensive tool to help identify the illness, such as DNA CpG (cytosine-phosphate-guanine) methylation, might be useful. In this paper, a technique is presented, based on the concept of Shannon Entropy, to select CpGs as inputs for non-linear classification algorithms. It will be shown that this approach generates accurate classifications that are a statistically significant improvement over using all the data available or randomly selecting the same number of CpGs. The analysis controlled for factors such as age, gender and smoking status of the patient. This approach managed to reduce the number of CpGs used while at the same time significantly increasing the accuracy.
Collapse
Affiliation(s)
- Gerardo Alfonso Perez
- Department of Biochemistry and Molecular Biology, University of Cordoba, 14071 Cordoba, Spain;
- Correspondence:
| | - Javier Caballero Villarraso
- Department of Biochemistry and Molecular Biology, University of Cordoba, 14071 Cordoba, Spain;
- Biochemical Laboratory, Reina Sofia University Hospital, 14004 Cordoba, Spain
| |
Collapse
|
17
|
Vollmer BL, Wolf AB, Sillau S, Corboy JR, Alvarez E. Evolution of Disease Modifying Therapy Benefits and Risks: An Argument for De-escalation as a Treatment Paradigm for Patients With Multiple Sclerosis. Front Neurol 2022; 12:799138. [PMID: 35145470 PMCID: PMC8821102 DOI: 10.3389/fneur.2021.799138] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/29/2021] [Indexed: 12/20/2022] Open
Abstract
BackgroundStrategies for sequencing disease modifying therapies (DMTs) in multiple sclerosis (MS) patients include escalation, high efficacy early, induction, and de-escalation.ObjectiveTo provide a perspective on de-escalation, which aims to match the ratio of DMT benefit/risk in aging patients.MethodsWe reanalyzed data from a retrospective, real-world cohort of MS patients to model disease activity for oral (dimethyl fumarate and fingolimod) and higher efficacy infusible (natalizumab and rituximab) DMTs by age. For patients with relapsing MS, we conducted a controlled, stratified analysis examining odds of disease activity for oral vs. infusible DMTs in patients <45 or ≥45 years. We reviewed the literature to identify DMT risks and predictors of safe discontinuation.ResultsYounger patients had lower probability of disease activity on infusible vs. oral DMTs. There was no statistical difference after age 54.2 years. When dichotomized, patients <45 years on oral DMTs had greater odds of disease activity compared to patients on infusible DMTs, while among those ≥45 years, there was no difference. Literature review noted that adverse events increase with aging, notably infections in patients with higher disability and longer DMT duration. Additionally, we identified factors predictive of disease reactivation including age, clinical stability, and MRI activity.ConclusionIn a real-world cohort of relapsing MS patients, high efficacy DMTs had less benefit with aging but were associated with increased risks. This cohort helps overcome some limitations of trials where older patients were excluded. To better balance benefits/risks, we propose a DMT de-escalation approach for aging MS patients.
Collapse
|
18
|
Role of IL-33/ST2 Axis in Chronic Inflammatory Neurological Disorderss. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2021. [DOI: 10.2478/sjecr-2020-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Interleukin-33 (IL-33) is a member of IL-1 family of cytokines, produced constitutively by fibroblasts, endothelial cells, and epithelial cells. IL-33 can be released passively from cells during tissue damage and cell necrosis, suggesting that it may act as an alarmin. Function of IL-33 is mediated by its interaction with ST2 molecule that is expressed on many immune cells: Th2 lymphocytes, NK, NKT and mast cells, monocytes, dendritic cells and granulocytes. IL-33/ST2 pathway plays, often dual, roles in different physiological and inflammatory processes, mediating both, pathological immune responses and tissue repair. Expression of IL-33 in the central nervous system (CNS) is significantly enhanced during various pathological processes, indicating its important role in the pathogenesis of neurological inflammatory and degenerative diseases. In this review the biological features, expression of IL-33 and its ligand ST2 in CNS, and the role of IL- 33/ST2 pathway in development of Alzheimer’s disease and multiple sclerosis are discussed.
Collapse
|
19
|
Assessing the differential sensitivities of wave-CAIPI ViSTa myelin water fraction and magnetization transfer saturation for efficiently quantifying tissue damage in MS. Mult Scler Relat Disord 2021; 56:103309. [PMID: 34688179 DOI: 10.1016/j.msard.2021.103309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/21/2021] [Accepted: 10/02/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Wave-CAIPI Visualization of Short Transverse relaxation time component (ViSTa) is a recently developed, short-T1-sensitized MRI method for fast quantification of myelin water fraction (MWF) in the human brain. It represents a promising technique for the evaluation of subtle, early signals of demyelination in the cerebral white matter of multiple sclerosis (MS) patients. Currently however, few studies exist that robustly assess the utility of ViSTa MWF measures of myelin compared to more conventional MRI measures of myelin in the brain of MS patients. Moreover, there are no previous studies evaluating the sensitivity of ViSTa MWF for the non-invasive detection of subtle tissue damage in both normal-appearing white matter (NAWM) and white matter lesions of MS patients. As a result, a central purpose of this study was to systematically evaluate the relationship between myelin sensitivity of T1-based ViSTa MWF mapping and a more generally recognized metric, Magnetization Transfer Saturation (MTsat), in healthy control and MS brain white matter. METHODS ViSTa MWF and MTsat values were evaluated in automatically-classified normal appearing white matter (NAWM), white matter (WM) lesion tissue, cortical gray matter, and deep gray matter of 29 MS patients and 10 healthy controls using 3T MRI. MWF and MT sat were also assessed in a tract-specific manner using the Johns Hopkins University WM atlas. MRI-derived measures of cerebral myelin content were uniquely compared by employing non-normal distribution-specific measures of median, interquartile range and skewness. Separate analyses of variance were applied to test tissue-specific differences in MTsat and ViSTa MWF distribution metrics. Non-parametric tests were utilized when appropriate. All tests were corrected for multiple comparisons using the False Discovery Rate method at the level, α=0.05. RESULTS Differences in whole NAWM MS tissue damage were detected with a higher effect size when using ViSTa MWF (q = 0.0008; ƞ2 = 0.34) compared to MTsat (q = 0.02; ƞ2= 0.24). We also observed that, as a possible measure of WM pathology, ViSTa-derived NAWM MWF voxel distributions of MS subjects were consistently skewed towards lower MWF values, while MTsat voxel distributions showed reduced skewness values. We further identified tract-specific reductions in mean ViSTa MWF of MS patients compared to controls that were not observed with MTsat. However, MTsat (q = 1.4 × 10-21; ƞ2 = 0.88) displayed higher effect sizes when differentiating NAWM and MS lesion tissue. Using regression analysis at the group level, we identified a linear relationship between MTsat and ViSTa MWF in NAWM (R2 = 0.46; p = 7.8 × 10-4) lesions (R2 = 0.30; p = 0.004), and with all tissue types combined (R2 = 0.71; p = 8.4 × 10-45). The linear relationship was also observed in most of the WM tracts we investigated. ViSTa MWF in NAWM of MS patients correlated with both disease duration (p = 0.02; R2 = 0.27) and WM lesion volume (p = 0.002; R2 = 0.34). CONCLUSION Because ViSTa MWF and MTsat metrics exhibit differential sensitivities to tissue damage in MS white matter, they can be collected in combination to provide an efficient, comprehensive measure of myelin water and macromolecular pool proton signals. These complementary measures may offer a more sensitive, non-invasive biopsy of early precursor signals in NAWM that occur prior to lesion formation. They may also aid in monitoring the efficacy of remyelination therapies.
Collapse
|
20
|
Manu MS, Hohjoh H, Yamamura T. Extracellular Vesicles as Pro- and Anti-inflammatory Mediators, Biomarkers and Potential Therapeutic Agents in Multiple Sclerosis. Aging Dis 2021; 12:1451-1461. [PMID: 34527421 PMCID: PMC8407883 DOI: 10.14336/ad.2021.0513] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune neurodegenerative disease of the central nervous system (CNS) characterized by multiple demyelinating lesions in the spinal cord and brain. Neuronal disruption caused by myelin loss or demyelination, which may accompany axonal changes, leads to multiple neurological symptoms. They may transiently appear for weeks during periods of disease worsening (relapse) in relapsing-remitting form of MS (RRMS). Although a number of genetic, metabolic and environmental factors influencing the development of MS have been identified, the precise mechanisms involved in the CNS tissue damage in MS are still poorly understood. Recent studies have revealed a significant role of circulating extracellular vesicles (EVs) in many diseases. EVs are known to serve as a cellular communication tool between two cell types either in close proximity or in different parts of the body. During the recent development in understanding of the pathogenesis of MS, studies have revealed the possible role of EVs in MS. Furthermore, circulating EVs can be used as a biomarker for monitoring disease progression and activity of MS, and they can also be therapeutic reagents or targets of therapy. In this review we overview and discuss in detail about generation of EVs and their diversified roles in MS.
Collapse
Affiliation(s)
- Mallahalli S Manu
- 1Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502 Japan
| | - Hirohiko Hohjoh
- 2Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502 Japan
| | - Takashi Yamamura
- 1Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502 Japan
| |
Collapse
|
21
|
The role of immune semaphorins in the pathogenesis of multiple sclerosis: Potential therapeutic targets. Int Immunopharmacol 2021; 95:107556. [PMID: 33756227 DOI: 10.1016/j.intimp.2021.107556] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/21/2021] [Accepted: 03/01/2021] [Indexed: 12/16/2022]
Abstract
The immune and nervous systems possess a highly intricate network of synaptic connections, shared messenger molecules, and exquisite communication ways, allowing intercellular signal transduction. The semaphorins (Semas) were initially identified as axonal guidance molecules in the development of the nervous system but later were found to be implicated also in regulating the immune system, known in this case as the "immune Semas" or "immunoregulatory Semas". Increasingly, these molecules are involved in multiple aspects of both physiological and pathological immune responses and were recently indicated to take part in various immunological disorders, encompassing allergy, cancer, and autoimmunity. Semas transduce signals by connecting to their cognate receptors, namely, plexins and neuropilins. Some of them, like Sema-3F, have been found to function as the inducer of the remyelination process whereas some others, like Sema-3A and Sema-4D, act to inhibit this process, either directly or indirectly. Besides, Sema-4A is crucial to the differentiation of T helper type 1 (Th1) and Th17 cells that are potentially involved in the pathogenesis of multiple sclerosis (MS), an autoimmune disease of the central nervous system. This review aims to reveal the role of immune Semas in the pathogenesis of MS and its animal model, experimental autoimmune encephalomyelitis, focusing on the therapeutic usages of these molecules to treat this neurodegenerative disease.
Collapse
|
22
|
Microglial Pruning: Relevance for Synaptic Dysfunction in Multiple Sclerosis and Related Experimental Models. Cells 2021; 10:cells10030686. [PMID: 33804596 PMCID: PMC8003660 DOI: 10.3390/cells10030686] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia, besides being able to react rapidly to a wide range of environmental changes, are also involved in shaping neuronal wiring. Indeed, they actively participate in the modulation of neuronal function by regulating the elimination (or “pruning”) of weaker synapses in both physiologic and pathologic processes. Mounting evidence supports their crucial role in early synaptic loss, which is emerging as a hallmark of several neurodegenerative diseases, including multiple sclerosis (MS) and its preclinical models. MS is an inflammatory, immune-mediated pathology of the white matter in which demyelinating lesions may cause secondary neuronal death. Nevertheless, primitive grey matter (GM) damage is emerging as an important contributor to patients’ long-term disability, since it has been associated with early and progressive cognitive decline (CD), which seriously worsens the quality of life of MS patients. Widespread synapse loss even in the absence of demyelination, axon degeneration and neuronal death has been demonstrated in different GM structures, thus raising the possibility that synaptic dysfunction could be an early and possibly independent event in the neurodegenerative process associated with MS. This review provides an overview of microglial-dependent synapse elimination in the neuroinflammatory process that underlies MS and its experimental models.
Collapse
|
23
|
Thadchanamoorthy V, Dayasiri K. Early-Onset Multiple Sclerosis With Frequent Relapses: A Challenging Diagnosis With a Less Favorable Prognosis. Cureus 2021; 13:e13963. [PMID: 33880297 PMCID: PMC8052520 DOI: 10.7759/cureus.13963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pediatric multiple sclerosis (MS) is a rare demyelinating disease of the brain, spinal cord, and optic nerve caused by immune modulators mediating against the neuronal axons of the central nervous system. MS is usually characterized by a series of neurological events, without any features of encephalopathy, separated in time and space. The complications arise from the permanent degeneration of the nerves. This condition can be diagnosed based on International Pediatric Multiple Sclerosis Study Group diagnostic criteria, and there is no definitive treatment for MS. We report the case of a male child who was diagnosed with MS at the age of six years when he presented with right hemiparesis and visual impairment. Subsequently, he had multiple relapses with varied neurological presentations, and each relapse was treated with methylprednisolone.
Collapse
|
24
|
Maghrebi O, Hanachi M, Bahrini K, Kchaou M, Jeridi C, Belal S, Ben Sassi S, Barbouche MR, Souiai O, Belghith M. Differential Gene Expression Patterns in Blood and Cerebrospinal Fluid of Multiple Sclerosis and Neuro-Behçet Disease. Front Genet 2021; 12:638236. [PMID: 33719347 PMCID: PMC7954360 DOI: 10.3389/fgene.2021.638236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/05/2021] [Indexed: 11/13/2022] Open
Abstract
Inflammatory demyelinating disorders of the central nervous system are debilitating conditions of the young adult, here we focus on multiple sclerosis (MS) and neuro-Behçet disease (NBD). MS is an autoimmune disorder of the central nervous system. NBD, a neurological manifestation of an idiopathic chronic relapsing multisystem inflammatory disease, the behçet disease. The diagnosis of MS and NBD relies on clinical symptoms, magnetic resonance imaging and laboratory tests. At first onset, clinical and imaging similarities between the two disorders may occur, making differential diagnosis challenging and delaying appropriate management. Aiming to identify additional discriminating biomarker patterns, we measured and compared gene expression of a broad panel of selected genes in blood and cerebrospinal fluid (CSF) cells of patients suffering from NBD, MS and non inflammatory neurological disorders (NIND). To reach this aim, bivariate and multivariate analysis were applied. The Principal Analysis Component (PCA) highlighted distinct profiles between NBD, MS, and controls. Transcription factors foxp3 in the blood along with IL-4, IL-10, and IL-17 expressions were the parameters that are the main contributor to the segregation between MS and NBD clustering. Moreover, parameters related to cellular activation and inflammatory cytokines within the CSF clearly differentiate between the two inflammatory diseases and the controls. We proceeded to ROC analysis in order to identify the most distinctive parameters between both inflammatory neurological disorders. The latter analysis suggested that IL-17, CD73 in the blood as well as IL-1β and IL-10 in the CSF were the most discriminating parameters between MS and NBD. We conclude that combined multi-dimensional analysis in blood and CSF suggests distinct mechanisms governing the pathophysiology of these two neuro-inflammatory disorders.
Collapse
Affiliation(s)
- Olfa Maghrebi
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Medicine, Tunis El Manar University, Tunis, Tunisia.,Tunis El Manar University, Tunis, Tunisia
| | - Mariem Hanachi
- Laboratory of Bioinformatics, Biomathematics and Biostatistics-LR16IPT09, Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Science of Bizerte, University of Carthage, Jarzouna, Tunisia
| | - Khadija Bahrini
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia.,Tunis El Manar University, Tunis, Tunisia
| | - Mariem Kchaou
- Faculty of Medicine, Tunis El Manar University, Tunis, Tunisia.,Charles Nicolle Hospital, Tunis, Tunisia
| | - Cyrine Jeridi
- Faculty of Medicine, Tunis El Manar University, Tunis, Tunisia.,Department of Neurology, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Samir Belal
- Faculty of Medicine, Tunis El Manar University, Tunis, Tunisia.,Department of Neurology, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Samia Ben Sassi
- Faculty of Medicine, Tunis El Manar University, Tunis, Tunisia.,Department of Neurology, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Mohamed-Ridha Barbouche
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Medicine, Tunis El Manar University, Tunis, Tunisia
| | - Oussama Souiai
- Tunis El Manar University, Tunis, Tunisia.,Laboratory of Bioinformatics, Biomathematics and Biostatistics-LR16IPT09, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Meriam Belghith
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia.,Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
25
|
Morelli N, Morelli H. Dual task training effects on gait and balance outcomes in multiple sclerosis: A systematic review. Mult Scler Relat Disord 2021; 49:102794. [PMID: 33540278 DOI: 10.1016/j.msard.2021.102794] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/30/2020] [Accepted: 01/21/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND People with Multiple Sclerosis (PwMS) exhibit deteriorated balance and gait performance during dual task (DT) conditions. Impaired dual tasking is related to increased fall risk and lower quality of life in PwMS. While dual task interventions have proven effective in various patient populations, evidence is lacking to support the use of DT interventions to improve clinical measures of balance and gait in PwMS. Therefore, the purpose of this systematic review was to synthesize the effectiveness of DT intervention at improving balance and gait in PwMS. METHODS A systematic search was completed using CINHAL, PubMed and MEDLINE. Methodological quality, level of evidence and recommendations for included studies was assessed by two reviewers. Effect sizes with 95% confidence intervals comparing single and DT outcomes were calculated for all balance and gait variables. RESULTS Five randomized control trials (RCTs) were included for review. Of the 23 effect sizes calculated, three had 95% confidence intervals which did not pass zero and were therefore interpreted as strong. Due to inconsistent level one evidence of DT interventions being superior to single task balance and gait interventions a grade B recommendation was given. CONCLUSION There is inconsistent evidence supporting the use of DT interventions to improve clinical balance measures in PwMS. There is supportive evidence for the use of DT interventions to improve both single and DT gait speed. However, there was heterogeneity between interventions and dosage among RCTs. Despite multiple promising findings, DT interventions appear to have minimal impact on clinical balance and gait measures in PwMS.
Collapse
Affiliation(s)
- Nathan Morelli
- Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S Limestone, Lexington, KY 40536, USA.
| | - Haley Morelli
- Rehabilitation Department, Chandler Medical Center at University of Kentucky, Lexington, KY, USA
| |
Collapse
|
26
|
De Masi R, Orlando S, De Donno A. The Age-Related Efficacy of Dimethyl Fumarate and Natalizumab in the Real-World Management of Multiple Sclerosis. Pharmaceuticals (Basel) 2021; 14:ph14020081. [PMID: 33499269 PMCID: PMC7911127 DOI: 10.3390/ph14020081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 11/25/2022] Open
Abstract
We investigated the comparative age-related efficacy of dimethyl fumarate (DMF) and natalizumab (NTZ) in clinical practice on multiple sclerosis (MS). Research in this area is lacking in the previous literature. In a three-year retrospective and clinical–paraclinical study, we compared 173 DMF patients and 94 NTZ patients with a similar average age (40 years) and disease duration (DD) (10 years). Expanded Disability Status Scale (EDSS) scores were higher in the NTZ group than in the DMF group at 3.5 vs. 2.5, respectively (p = 0.001). However, in both groups, age values correlated with DD (r = 0.42; p < 0.001), EDSS (r = 0.52; p < 0.001) and age at onset (r = 0.18; p < 0.001). Furthermore, age-adjusted Kaplan–Meier curves showed that NTZ-treated subjects maintained a 1.0–3.0 EDSS status score (p = 0.003) more frequently and a 3.5–7.0 score (p = 0.022) significantly less frequently compared with DMF-treated subjects. The EDSS percentage mean difference between NTZ and DMF groups was 81.6%, decreasing inversely with age (r = −0.34; p < 0.001). Finally, high EDSS score values were reached at the age of 39–40 years, regardless of their experimental group. We demonstrated age as a major contributor in disability and response to therapy in current management of MS. Thus, age should be considered in the risk/benefit evaluation in decision making for the disease modifying treatments in MS.
Collapse
Affiliation(s)
- Roberto De Masi
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, 73042 Casarano, Lecce, Italy;
- Complex Operative Unit of Neurology, “F. Ferrari” Hospital, 73042 Casarano, Lecce, Italy
| | - Stefania Orlando
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, 73042 Casarano, Lecce, Italy;
- Correspondence: ; Tel.: +39-0833-508-412
| | - Antonella De Donno
- Laboratory of Hygiene, Department of Biological and Environmental Sciences and Technologies, University of the Salento, 73100 Lecce, Italy;
| |
Collapse
|
27
|
CD146/sCD146 in the Pathogenesis and Monitoring of Angiogenic and Inflammatory Diseases. Biomedicines 2020; 8:biomedicines8120592. [PMID: 33321883 PMCID: PMC7764286 DOI: 10.3390/biomedicines8120592] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022] Open
Abstract
CD146 is a cell adhesion molecule expressed on endothelial cells, as well as on other cells such as mesenchymal stem cells and Th17 lymphocytes. This protein also exists in a soluble form, whereby it can be detected in biological fluids, including the serum or the cerebrospinal fluid (CSF). Some studies have highlighted the significance of CD146 and its soluble form in angiogenesis and inflammation, having been shown to contribute to the pathogenesis of many inflammatory autoimmune diseases, such as systemic sclerosis, mellitus diabetes, rheumatoid arthritis, inflammatory bowel diseases, and multiple sclerosis. In this review, we will focus on how CD146 and sCD146 contribute to the pathogenesis of the aforementioned autoimmune diseases and discuss the relevance of considering it as a biomarker in these pathologies.
Collapse
|
28
|
Piedra-Quintero ZL, Wilson Z, Nava P, Guerau-de-Arellano M. CD38: An Immunomodulatory Molecule in Inflammation and Autoimmunity. Front Immunol 2020; 11:597959. [PMID: 33329591 PMCID: PMC7734206 DOI: 10.3389/fimmu.2020.597959] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
CD38 is a molecule that can act as an enzyme, with NAD-depleting and intracellular signaling activity, or as a receptor with adhesive functions. CD38 can be found expressed either on the cell surface, where it may face the extracellular milieu or the cytosol, or in intracellular compartments, such as endoplasmic reticulum, nuclear membrane, and mitochondria. The main expression of CD38 is observed in hematopoietic cells, with some cell-type specific differences between mouse and human. The role of CD38 in immune cells ranges from modulating cell differentiation to effector functions during inflammation, where CD38 may regulate cell recruitment, cytokine release, and NAD availability. In line with a role in inflammation, CD38 appears to also play a critical role in inflammatory processes during autoimmunity, although whether CD38 has pathogenic or regulatory effects varies depending on the disease, immune cell, or animal model analyzed. Given the complexity of the physiology of CD38 it has been difficult to completely understand the biology of this molecule during autoimmune inflammation. In this review, we analyze current knowledge and controversies regarding the role of CD38 during inflammation and autoimmunity and novel molecular tools that may clarify current gaps in the field.
Collapse
Affiliation(s)
- Zayda L. Piedra-Quintero
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Zachary Wilson
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Biomedical Science Undergraduate Program, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Porfirio Nava
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados (CINVESTAV), México City, México
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
29
|
Neuronal and Endothelial Transglutaminase-2 Expression during Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Neuroscience 2020; 461:140-154. [PMID: 33253822 DOI: 10.1016/j.neuroscience.2020.11.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022]
Abstract
Transglutiminase-2 (TG2) is a multifunctional enzyme that has been implicated in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS) using global knockout mice and TG2 selective inhibitors. Previous studies have identified the expression of TG2 in subsets of macrophages-microglia and astrocytes after EAE. The aims of the current investigation were to examine neuronal expression of TG2 in rodent models of chronic-relapsing and non-relapsing EAE and through co-staining with intracellular and cell death markers, provide insight into the putative role of TG2 in neuronal pathology during disease progression. Here we report that under normal physiological conditions there is a low basal expression of TG2 in the nucleus of neurons, however following EAE or MS, robust induction of cytoplasmic TG2 occurs in most neurons surrounding perivascular lesion sites. Importantly, TG2-positive neurons also labeled for phosphorylated Extracellular signal-regulated kinase 1/2 (ERK1/2) and the apoptotic marker cleaved caspase-3. In white and gray matter lesions, high levels of TG2 were also found within the vasculature and endothelial cells as well as in tissue migrating pericytes or fibroblasts, though rarely did TG2 colocalize with cells identified with glial cell markers (astrocytes, oligodendrocytes and microglia). TG2 induction occurred concurrently with the upregulation of the blood vessel permeability factor and angiogenic molecule Vascular Endothelial Growth Factor (VEGF). Extracellular TG2 was found to juxtapose with fibronectin, within and surrounding blood vessels. Though molecular and pharmacological studies have implicated TG2 in the induction and severity of EAE, the cell autonomous functions of this multifunctional enzyme during disease progression remains to be elucidated.
Collapse
|
30
|
Lattanzi S, Rocchi C, Danni M, Taffi R, Cerqua R, Carletti S, Silvestrini M. Long-term outcome in multiple sclerosis patients treated with fingolimod. Mult Scler Relat Disord 2020; 45:102416. [DOI: 10.1016/j.msard.2020.102416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/22/2020] [Accepted: 07/20/2020] [Indexed: 11/16/2022]
|
31
|
Esmaeil Amini M, Shomali N, Bakhshi A, Rezaei S, Hemmatzadeh M, Hosseinzadeh R, Eslami S, Babaie F, Aslani S, Torkamandi S, Mohammadi H. Gut microbiome and multiple sclerosis: New insights and perspective. Int Immunopharmacol 2020; 88:107024. [PMID: 33182024 DOI: 10.1016/j.intimp.2020.107024] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
The human gastrointestinal microbiota, also known as the gut microbiota living in the human gastrointestinal tract, has been shown to have a significant impact on several human disorders including rheumatoid arthritis, diabetes, obesity, and multiple sclerosis (MS). MS is an inflammatory disease characterized by the destruction of the spinal cord and nerve cells in the brain due to an attack of immune cells, causing a wide range of harmful symptoms related to inflammation in the central nervous system (CNS). Despite extensive studies on MS that have shown that many external and genetic factors are involved in its pathogenesis, the exact role of external factors in the pathophysiology of MS is still unclear. Recent studies on MS and experimental autoimmune encephalomyelitis (EAE), an animal model of encephalitis, have shown that intestinal microbiota may play a key role in the pathogenesis of MS. Therefore, modification of the intestinal microbiome could be a promising strategy for the future treatment of MS. In this study, the characteristics of intestinal microbiota, the relationship between intestine and brain despite the blood-brain barrier, various factors involved in intestinal microbiota modification, changes in intestinal microbial composition in MS, intestinal microbiome modification strategies, and possible use of intestinal microbiome and factors affecting it have been discussed.
Collapse
Affiliation(s)
- Mohammad Esmaeil Amini
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Bakhshi
- Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Somaye Rezaei
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solat Eslami
- Dietary Supplements & Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farhad Babaie
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Torkamandi
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
32
|
Ershadinia N, Mortazavinia N, Babaniamansour S, Najafi-Nesheli M, Babaniamansour P, Aliniagerdroudbari E. The prevalence of autoimmune diseases in patients with multiple sclerosis: A cross-sectional study in Qom, Iran, in 2018. CURRENT JOURNAL OF NEUROLOGY 2020; 19:98-102. [PMID: 38011389 PMCID: PMC8185585 DOI: 10.18502/cjn.v19i3.5421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/18/2020] [Indexed: 07/15/2023]
Abstract
Background: Multiple sclerosis (MS) is one of the most common autoimmune diseases worldwide and various autoimmune comorbidities are reported with MS. The objective of this study is to estimate the prevalence of the autoimmune diseases' comorbidity in patients with MS. Methods: In this cross-sectional study, we investigated a group of patients with MS in terms of age, gender, duration of MS, presence of simultaneous autoimmune diseases, such as Graves' disease, Hashimoto's thyroiditis, type 1 diabetes mellitus (DM), and systemic lupus erythematous (SLE). Results: This study included 1215 patients with MS, of which 70.8% were women. The mean age of participants was 33.70 ± 27.63 years. 55 patients (4.5%) had at least one autoimmune disease. The most common comorbidity was for Hashimoto's thyroiditis (30 patients). The frequency of simultaneous autoimmune disease was higher in women. Mean age (P = 0.01), mean duration of MS (P = 0.03), and mean age on MS diagnosis (P = 0.02) were significantly higher in simultaneous MS and other autoimmune diseases. Conclusion: Our study revealed that the probability of autoimmune diseases co-occurrence in patients with MS could be higher in older patients, in longer duration of disease, and also in patients with higher age at time of MS diagnosis.
Collapse
Affiliation(s)
- Nazanin Ershadinia
- Department of Neurology, School of Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Nader Mortazavinia
- Department of Neurology, School of Medicine, Qom Branch, Islamic Azad University, Qom, Iran
| | - Sepideh Babaniamansour
- Department of Neurology, School of Medicine, Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mahdi Najafi-Nesheli
- Neuroscience Research Center, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parto Babaniamansour
- Department of Mechanical Engineering, College of Engineering, Temple University, Philadelphia, USA
| | - Ehsan Aliniagerdroudbari
- Department of Neurology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Sattar M, Poursadeghfard M. Concurrence of multiple sclerosis and primary biliary cholangitis: Report of 3 cases. CASPIAN JOURNAL OF INTERNAL MEDICINE 2020; 11:223-226. [PMID: 32509253 PMCID: PMC7265518 DOI: 10.22088/cjim.11.2.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system which damages the myelin and axon. Primary biliary cholangitis (PBC) is a slow progressive liver disease with autoimmune feature in which non-purulent destructive cholangitis and interlobular bile duct destruction occur. Involvement of each of PBC and MS is thought to be related to environmental exposure in genetically susceptible persons. Case Presentation: Here, we aim to report 3 women 52, 27 and 51 years old with MS and PBC. Conclusion: Although MS seems to have an association with some autoimmune gastrointestinal disorders such as ulcerative colitis, the concurrence of MS and PBC has been rarely reported.
Collapse
Affiliation(s)
- Mozhgan Sattar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Poursadeghfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
34
|
Milovanovic J, Arsenijevic A, Stojanovic B, Kanjevac T, Arsenijevic D, Radosavljevic G, Milovanovic M, Arsenijevic N. Interleukin-17 in Chronic Inflammatory Neurological Diseases. Front Immunol 2020; 11:947. [PMID: 32582147 PMCID: PMC7283538 DOI: 10.3389/fimmu.2020.00947] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
A critical role for IL-17, a cytokine produced by T helper 17 (Th17) cells, has been indicated in the pathogenesis of chronic inflammatory and autoimmune diseases. A positive effect of blockade of IL-17 secreted by autoreactive T cells has been shown in various inflammatory diseases. Several cytokines, whose production is affected by environmental factors, control Th17 differentiation and its maintenance in tissues during chronic inflammation. The roles of IL-17 in the pathogenesis of chronic neuroinflammatory conditions, multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), Alzheimer's disease, and ischemic brain injury are reviewed here. The role of environmental stimuli in Th17 differentiation is also summarized, highlighting the role of viral infection in the regulation of pathogenic T helper cells in EAE.
Collapse
Affiliation(s)
- Jelena Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Stojanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tatjana Kanjevac
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gordana Radosavljevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Marija Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
35
|
Pardridge WM. The Isolated Brain Microvessel: A Versatile Experimental Model of the Blood-Brain Barrier. Front Physiol 2020; 11:398. [PMID: 32457645 PMCID: PMC7221163 DOI: 10.3389/fphys.2020.00398] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022] Open
Abstract
A versatile experimental model for the investigation of the blood-brain barrier (BBB), including the neuro-vascular unit, is the isolated brain microvessel preparation. Brain microvessels are primarily comprised of endothelial cells, but also include pericytes, pre-capillary arteriolar smooth muscle cells, astrocyte foot processes, and occasional nerve endings. These microvessels can be isolated from brain with a 3 h procedure, and the microvessels are free of brain parenchyma. Brain microvessels have been isolated from fresh animal brain, fresh human brain obtained at neurosurgery, as well as fresh or frozen autopsy human brain. Brain microvessels are the starting point for isolation of brain microvessel RNA, which then enables the production of BBB cDNA libraries and a genomics analysis of the brain microvasculature. Brain microvessels, combined with quantitative targeted absolute proteomics, allow for the quantitation of specific transporters or receptors expressed at the brain microvasculature. Brain microvessels, combined with specific antibodies and immune labeling of isolated capillaries, allow for the cellular location of proteins expressed within the neuro-vascular unit. Isolated brain microvessels can be used as an “in vitro” preparation of the BBB for the study of the kinetic parameters of BBB carrier-mediated transport (CMT) systems, or for the determination of dissociation constants of peptide binding to BBB receptor-mediated transport (RMT) systems expressed at either the animal or the human BBB. This review will discuss how the isolated brain microvessel model system has advanced our understanding of the organization and functional properties of the BBB, and highlight recent renewed interest in this 50 year old model of the BBB.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
36
|
Solár P, Zamani A, Kubíčková L, Dubový P, Joukal M. Choroid plexus and the blood-cerebrospinal fluid barrier in disease. Fluids Barriers CNS 2020; 17:35. [PMID: 32375819 PMCID: PMC7201396 DOI: 10.1186/s12987-020-00196-2] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023] Open
Abstract
The choroid plexus (CP) forming the blood-cerebrospinal fluid (B-CSF) barrier is among the least studied structures of the central nervous system (CNS) despite its clinical importance. The CP is an epithelio-endothelial convolute comprising a highly vascularized stroma with fenestrated capillaries and a continuous lining of epithelial cells joined by apical tight junctions (TJs) that are crucial in forming the B-CSF barrier. Integrity of the CP is critical for maintaining brain homeostasis and B-CSF barrier permeability. Recent experimental and clinical research has uncovered the significance of the CP in the pathophysiology of various diseases affecting the CNS. The CP is involved in penetration of various pathogens into the CNS, as well as the development of neurodegenerative (e.g., Alzheimer´s disease) and autoimmune diseases (e.g., multiple sclerosis). Moreover, the CP was shown to be important for restoring brain homeostasis following stroke and trauma. In addition, new diagnostic methods and treatment of CP papilloma and carcinoma have recently been developed. This review describes and summarizes the current state of knowledge with regard to the roles of the CP and B-CSF barrier in the pathophysiology of various types of CNS diseases and sets up the foundation for further avenues of research.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne´s University Hospital Brno, Pekařská 53, CZ-656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
| | - Lucie Kubíčková
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
| | - Petr Dubový
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic.
| |
Collapse
|
37
|
Borhani-Haghighi M, Mohamadi Y. Intranasal administration of conditioned medium derived from mesenchymal stem cells-differentiated oligodendrocytes ameliorates experimental autoimmune encephalomyelitis. J Chem Neuroanat 2020; 106:101792. [PMID: 32353514 DOI: 10.1016/j.jchemneu.2020.101792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/05/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
In multiple sclerosis, myelin sheaths around the axons are degenerated due to uncontrolled inflammation in the central nervous system. Oligodendrocytes (OLs) are myelin-forming cells that secrete trophic factors necessary for myelin protection. Beneficial features of conditioned medium (CM) derived from different stem cells are nowadays under investigation in treating neurodegenerative diseases. Here, we used the differentiation capacity of Wharton's jelly mesenchymal stem cells (WJMSCs) to obtain OLs. Then, the study aimed to evaluate the status of inflammation and myelination in male experimental autoimmune encephalomyelitis (EAE) mice after intranasal administration of CM derived from OLs (OL-CM). Inflammation was studied by evaluating gliosis, inflammatory cell infiltration and expression of inflammation indicators including NLRP3 inflammasome, interleukin-1β, interleukin-18, glial fibrillary acidic protein, and ionized calcium binding adaptor molecule 1. Remyelination was studied by luxol fast blue staining and evaluating the expression of myelin indicators including myelin basic protein and oligodendrocyte transcription factor. In addition, we followed the trend of body weight and functional recovery during the 28-day study. ELISA assay revealed that OL-CM contained brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and ciliary neurotrophic factor. Data showed that OL-CM moderated inflammation, augmented remyelination, and gained normal body weight. Notably, these anti-inflammatory and regenerative effects of OL-CM improved neurological functions in EAE mice. In conclusion, the current study offered a new choice for treating multiple sclerosis using noninvasive intranasal administration of CM harvested from easily achievable WJMSCs-differentiated OLs.
Collapse
Affiliation(s)
- Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Mohamadi
- Department of Anatomy, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
38
|
Boyko AN, Khachanova NV, Melnikov MV, Sivertseva SA, Evdoshenko EP, Spirin NN, Vasilyev AV, Rozenson OL. [New directions of immunocorrection in multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:103-109. [PMID: 32307419 DOI: 10.17116/jnevro2020120021103] [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: 12/28/2022]
Abstract
Multiple sclerosis is a central nervous system disease with autoimmune and neurodegenerative mechanisms of development. This disease can lead to severe disability and neurological defects. Although its etiology and pathogenesis remain unclear, research data show that multiple sclerosis is a multifactorial disease, the development of which depends on environmental factors, as well as a genetic predisposition. The impact of these factors lead to the death of neural cells, accompanied by demyelination of nerves and neuronal dysfunction. Therapy of multiple sclerosis is based on the use of anti-inflammatory and immunomodulating substances, however, there are certain disadvantages associated with the constant use of these drugs and a possible change in dosage over time. This review discusses the pathogenesis of multiple sclerosis and the role of various subpopulations of immune cells in the development of diseases, as well as existing approaches to therapy. It is noted that immunoreconstitution therapy has advantages over immunomodulation and immunosuppression maintenance therapy for some patients. Thus, short courses of therapy provide more adequate treatment for patients and lower risks of adverse events associated with chronic immunosuppression. The review also discusses the data of clinical studies on the immunoreconstitution therapy drugs, such as alemtuzumab, ocrelizumab and cladribine. It is noted that nowadays the exact mechanisms underlying this type of therapy remain unclear. In this regard, further studies are needed to explain the therapeutic effects. It is assumed that patients with a high risk of multiple sclerosis progression are the optimal group of patients for the early use of selective immunoreconstitution therapy. Thus, immunoreconstitution therapy may be the treatment of choice for many patients with highle active multiple sclerosis.
Collapse
Affiliation(s)
- A N Boyko
- Pirogov National Research Medical University, Moscow, Russia
| | - N V Khachanova
- Pirogov National Research Medical University, Moscow, Russia
| | - M V Melnikov
- Pirogov National Research Medical University, Moscow, Russia
| | - S A Sivertseva
- Tyumen Regional Center for Multiple Sclerosis, Tyumen, Russia
| | - E P Evdoshenko
- Pavlov First State Medical University of St. Petersburg, St. Peterburg, Russia
| | - N N Spirin
- Yaroslavl' State Medical University, Yaroslavl, Russia
| | | | | |
Collapse
|
39
|
Ye Z, George A, Wu AT, Niu X, Lin J, Adusumilli G, Naismith RT, Cross AH, Sun P, Song SK. Deep learning with diffusion basis spectrum imaging for classification of multiple sclerosis lesions. Ann Clin Transl Neurol 2020; 7:695-706. [PMID: 32304291 PMCID: PMC7261762 DOI: 10.1002/acn3.51037] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/24/2020] [Accepted: 03/13/2020] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Multiple sclerosis (MS) lesions are heterogeneous with regard to inflammation, demyelination, axonal injury, and neuronal loss. We previously developed a diffusion basis spectrum imaging (DBSI) technique to better address MS lesion heterogeneity. We hypothesized that the profiles of multiple DBSI metrics can identify lesion-defining patterns. Here we test this hypothesis by combining a deep learning algorithm using deep neural network (DNN) with DBSI and other imaging methods. METHODS Thirty-eight MS patients were scanned with diffusion-weighted imaging, magnetization transfer imaging, and standard conventional MRI sequences (cMRI). A total of 499 regions of interest were identified on standard MRI and labeled as persistent black holes (PBH), persistent gray holes (PGH), acute black holes (ABH), acute gray holes (AGH), nonblack or gray holes (NBH), and normal appearing white matter (NAWM). DBSI, diffusion tensor imaging (DTI), and magnetization transfer ratio (MTR) were applied to the 43,261 imaging voxels extracted from these ROIs. The optimized DNN with 10 fully connected hidden layers was trained using the imaging metrics of the lesion subtypes and NAWM. RESULTS Concordance, sensitivity, specificity, and accuracy were determined for the different imaging methods. DBSI-DNN derived lesion classification achieved 93.4% overall concordance with predetermined lesion types, compared with 80.2% for DTI-DNN model, 78.3% for MTR-DNN model, and 74.2% for cMRI-DNN model. DBSI-DNN also produced the highest specificity, sensitivity, and accuracy. CONCLUSIONS DBSI-DNN improves the classification of different MS lesion subtypes, which could aid clinical decision making. The efficacy and efficiency of DBSI-DNN shows great promise for clinical applications in automatic MS lesion detection and classification.
Collapse
Affiliation(s)
- Zezhong Ye
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110
| | - Ajit George
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110
| | - Anthony T Wu
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, 63130
| | - Xuan Niu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110
| | - Joshua Lin
- Keck School of Medicine, University of Southern California, Los Angeles, California, 90033
| | - Gautam Adusumilli
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, 63110
| | - Robert T Naismith
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, 63110
| | - Anne H Cross
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, 63110
| | - Peng Sun
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110
| |
Collapse
|
40
|
Gökaslan S, Demirbaş H, Özer Gökaslan Ç. Evaluation of cardiovascular autonomic dysfunction according to heart rate turbulence and variability in patients with relapsing remitting multiple sclerosis. Turk J Med Sci 2020; 50:442-447. [PMID: 32222131 PMCID: PMC7164742 DOI: 10.3906/sag-1912-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/24/2020] [Indexed: 01/25/2023] Open
Abstract
Background/aim Multiple sclerosis (MS) is an autoimmune disease characterized by neurodegeneration or demyelination; the relapsing–remitting phase of MS is characterized by acute exacerbation of disease activity. The most commonly used noninvasive approach to assess autonomic function is the determination of heart rate turbulence (HRT) and heart rate variability (HRV). The aim of this study was to evaluate the presence of cardiovascular autonomic dysfunction using HRT and HRV parameters determined via 24-h Holter ECG monitoring in patients with relapsing–remitting MS without known heart disease. Materials and methods The study included 26 patients diagnosed with relapsing–remitting MS and 22 age- and sex-matched healthy controls. HRT and HRV parameters were analyzed via 24-h Holter ECG monitoring. Magnetic resonance imaging findings were reevaluated to identify any demyelinating lesions in the brain stem. Results The HRV parameters of SDNNI (mean of the standard deviations of all normal sinus RR intervals in all 5-min segments), rMSSD (root–mean–square successive difference), and sNN50 (percentage of successive normal sinus RR intervals >50 ms) were significantly lower in the MS group than in the control group (P < 0.05). Conclusion This study revealed that the patients with MS had reduced HRV; this was demonstrated by dysfunction with regard to parasympathetic and sympathetic parameters in HRV analysis.
Collapse
Affiliation(s)
- Serkan Gökaslan
- Department of Cardiology, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| | - Hayri Demirbaş
- Department of Neurology, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| | - Çiğdem Özer Gökaslan
- Department of Radiology, Faculty of Medicine, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| |
Collapse
|
41
|
Neuroinflammation in CNS diseases: Molecular mechanisms and the therapeutic potential of plant derived bioactive molecules. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100176] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
Sánchez-Fernández A, Skouras DB, Dinarello CA, López-Vales R. OLT1177 (Dapansutrile), a Selective NLRP3 Inflammasome Inhibitor, Ameliorates Experimental Autoimmune Encephalomyelitis Pathogenesis. Front Immunol 2019; 10:2578. [PMID: 31736980 PMCID: PMC6839275 DOI: 10.3389/fimmu.2019.02578] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/17/2019] [Indexed: 01/05/2023] Open
Abstract
IL-1β and IL-18 are pro-inflammatory cytokines that are linked to inflammation. Activation of the NOD-like receptor protein 3 (NLRP3) inflammasome is involved in the maturation and secretion of IL-1β and IL-18 and, thus, plays a key role in the pathogenesis of many inflammatory conditions, including multiple sclerosis (MS). OLT1177™ (Dapansutrile) is a newly developed drug that is safe in humans and inhibits specifically the NLRP3 inflammasome. In the present study, we investigated whether OLT1177 exerts therapeutic effects in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We found that EAE mice fed an OLT1177-enriched diet prophylactically were significantly protected against functional deficits and demyelination in the spinal cord. We also demonstrated that prophylactic oral administration of OLT1177 led to marked reduction (~2- to 3-fold) in the protein levels of IL-1β and IL-18, as well as, IL-6 and TNFα, in the spinal cord of EAE mice. Moreover, prophylactic oral administration of OLT1177 significantly attenuated the infiltration of CD4 T cells and macrophages in the spinal cord. We also demonstrated that oral administration of OLT1177, starting at disease onset, resulted in significant amelioration of the clinical signs of EAE. Overall, these first data suggest that OLT1177 could have clinical benefit for the treatment of MS in humans.
Collapse
Affiliation(s)
- Alba Sánchez-Fernández
- Institut de Neurociencies and Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autonoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Madrid, Spain
| | | | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rubèn López-Vales
- Institut de Neurociencies and Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autonoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Madrid, Spain
| |
Collapse
|
43
|
Hollinger KR, Smith MD, Kirby LA, Prchalova E, Alt J, Rais R, Calabresi PA, Slusher BS. Glutamine antagonism attenuates physical and cognitive deficits in a model of MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e609. [PMID: 31467038 PMCID: PMC6745721 DOI: 10.1212/nxi.0000000000000609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/09/2019] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To measure the impact of JHU-083, a novel prodrug of the glutamine antagonist 6-diazo-5-oxo-l-norleucine, on immune cell proliferation and activation, along with physical and cognitive impairments associated with the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS Splenic-derived T cells and bone marrow-derived dendritic cells (DCs) were cultured, activated, and treated daily with vehicle or JHU-083. Proliferation and activation were measured via flow cytometry and IncuCyte live cell analysis. C57BL/6 mice were immunized for EAE. Vehicle or JHU-083 was administered orally every other day either from the time of immunization in the prevention paradigm or from the time of disease onset in the treatment paradigm. Disease scores and body weight were monitored. In the treatment paradigm, cognition was evaluated using the Barnes maze test. RESULTS JHU-083 selectively inhibits T-cell proliferation and decreases T-cell activation, with no effect on DCs. In vivo, orally administered JHU-083 significantly decreases EAE severity in both prevention and treatment paradigms and reverses EAE-induced cognitive impairment. CONCLUSIONS JHU-083, a well-tolerated, brain penetrable glutamine antagonist, is a promising novel treatment for both the physical and cognitive deficits of MS.
Collapse
Affiliation(s)
- Kristen R Hollinger
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Matthew D Smith
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Leslie A Kirby
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Eva Prchalova
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Jesse Alt
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Rana Rais
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Peter A Calabresi
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD.
| | - Barbara S Slusher
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
44
|
Mansoor SR, Zabihi E, Ghasemi-Kasman M. The potential use of mesenchymal stem cells for the treatment of multiple sclerosis. Life Sci 2019; 235:116830. [PMID: 31487529 DOI: 10.1016/j.lfs.2019.116830] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/23/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory disease of the central nervous system (CNS). In attempt to identify an appropriate treatment for improving the neurological symptoms and remyelination process, autologous and allogenic transplantation of mesenchymal stem cells (MSCs) have been introduced as an effective therapeutic strategy in MS. MSCs are a heterogeneous subset of pluripotent non-hematopoietic stromal cells that are isolated from bone marrow, adipose tissue, placenta and other sources. MSCs have considerable therapeutic effects due to their ability in differentiation, migration, immune-modulation and neuroregeneration. To date, numerous experimental and clinical studies demonstrated that MSCs therapy improves the CNS repair and modulates functional neurological symptoms. Here, we provided an overview of the current knowledge about the clinical applications of MSCs in MS. Furthermore, the major challenges and risks of MSCs therapy in MS patients have been elucidated.
Collapse
Affiliation(s)
- Sahar Rostami Mansoor
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
45
|
Colamatteo A, Maggioli E, Azevedo Loiola R, Hamid Sheikh M, Calì G, Bruzzese D, Maniscalco GT, Centonze D, Buttari F, Lanzillo R, Perna F, Zuccarelli B, Mottola M, Cassano S, Galgani M, Solito E, De Rosa V. Reduced Annexin A1 Expression Associates with Disease Severity and Inflammation in Multiple Sclerosis Patients. THE JOURNAL OF IMMUNOLOGY 2019; 203:1753-1765. [PMID: 31462505 DOI: 10.4049/jimmunol.1801683] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
Chronic neuroinflammation is a key pathological hallmark of multiple sclerosis (MS) that suggests that resolution of inflammation by specialized proresolving molecules is dysregulated in the disease. Annexin A1 (ANXA1) is a protein induced by glucocorticoids that facilitates resolution of inflammation through several mechanisms that include an inhibition of leukocyte recruitment and activation. In this study, we investigated the ability of ANXA1 to influence T cell effector function in relapsing/remitting MS (RRMS), an autoimmune disease sustained by proinflammatory Th1/Th17 cells. Circulating expression levels of ANXA1 in naive-to-treatment RRMS subjects inversely correlated with disease score and progression. At the cellular level, there was an impaired ANXA1 production by CD4+CD25- conventional T and CD4+RORγt+ T (Th17) cells from RRMS subjects that associated with an increased migratory capacity in an in vitro model of blood brain barrier. Mechanistically, ANXA1 impaired monocyte maturation secondarily to STAT3 hyperactivation and potently reduced T cell activation, proliferation, and glycolysis. Together, these findings identify impaired disease resolution pathways in RRMS caused by dysregulated ANXA1 expression that could represent new potential therapeutic targets in RRMS.
Collapse
Affiliation(s)
- Alessandra Colamatteo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," 80131 Naples, Italy
| | - Elisa Maggioli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, United Kingdom
| | - Rodrigo Azevedo Loiola
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, United Kingdom
| | - Madeeha Hamid Sheikh
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, United Kingdom
| | - Gaetano Calì
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Dario Bruzzese
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli "Federico II," 80131 Naples, Italy
| | - Giorgia Teresa Maniscalco
- Dipartimento di Neurologia, Centro Regionale Sclerosi Multipla, Azienda Ospedaliera "A. Cardarelli," 80131 Naples, Italy
| | - Diego Centonze
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, 86077 Pozzilli, Italy.,Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Fabio Buttari
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, 86077 Pozzilli, Italy
| | - Roberta Lanzillo
- Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II," 80131 Naples, Italy
| | - Francesco Perna
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II," 80131 Naples, Italy
| | - Bruno Zuccarelli
- Unità Operativa Complessa di Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli Monaldi-Cotugno, Centro Traumatologico Ortopedico, 80131 Naples, Italy; and
| | - Maria Mottola
- Unità Operativa Complessa di Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli Monaldi-Cotugno, Centro Traumatologico Ortopedico, 80131 Naples, Italy; and
| | - Silvana Cassano
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Mario Galgani
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Egle Solito
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," 80131 Naples, Italy; .,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, United Kingdom
| | - Veronica De Rosa
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, 80131 Naples, Italy; .,Unità di NeuroImmunologia, Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
46
|
Tahamtan A, Rezaiy S, Samadizadeh S, Moradi A, Tabarraei A, Javid N, Oladnabi M, Naeimi MH. Cannabinoid CB2 Receptor Functional Variation (Q63R) Is Associated with Multiple Sclerosis in Iranian Subjects. J Mol Neurosci 2019; 70:26-31. [DOI: 10.1007/s12031-019-01395-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/01/2019] [Indexed: 02/02/2023]
|
47
|
Wooliscroft L, Silbermann E, Cameron M, Bourdette D. Approaches to Remyelination Therapies in Multiple Sclerosis. Curr Treat Options Neurol 2019; 21:34. [DOI: 10.1007/s11940-019-0574-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
48
|
Habek M, Crnošija L, Gabelić T, Barun B, Adamec I, Junaković A, Ruška B, Pavičić T, Krbot Skorić M. Longitudinal assessment of autonomic nervous system in patients with first demyelinating event suggestive of multiple sclerosis. Eur J Neurol 2019; 26:1377-1383. [PMID: 31099944 DOI: 10.1111/ene.13989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 05/13/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE As a high proportion of people with clinically isolated syndrome (pwCIS) exhibit sympathetic adrenergic and sudomotor dysfunction, the aim of this study was to investigate the evolution of autonomic nervous system (ANS) abnormalities in pwCIS over a 2-year follow-up. METHODS This was a prospective cohort study in which 121 pwCIS were enrolled and followed for 2 years. After 2-year follow-up, data were available for 84 pwCIS. ANS symptoms were evaluated with the Composite Autonomic System Score-31 (COMPASS-31) and results of the ANS tests were expressed using the Composite Autonomic Scoring Scale (CASS) at baseline and visit at month 24. Symptomatic dysautonomia was defined if the patient had a COMPASS-31 value above the median of the whole cohort at baseline evaluation (COMPASS-31 > 6.79) and CASS score >0. RESULTS Complete CASS data at baseline and month 24 were available for 62 patients; in 24 (38.7%) patients there was worsening, in 16 (25.8%) there was improvement and in 22 (35.5%) there was no change in CASS score. In 90% of pwCIS (72 of 80) there was no change in parasympathetic nervous system tests, whereas 47.3% (35 of 74) had either worsening or improvement in sympathetic adrenergic and 28.6% (20 of 70) had either worsening or improvement in sudomotor function. A multivariable regression model identified the total number of T2 lesions as an independent predictor for worsening of symptomatic dysautonomia. No predictors for worsening or improving of CASS score were identified. CONCLUSION A substantial proportion of pwCIS experienced worsening of ANS abnormalities during the 2-year follow-up and magnetic resonance imaging parameters seemed to predict these abnormalities.
Collapse
Affiliation(s)
- M Habek
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - L Crnošija
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia
| | - T Gabelić
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - B Barun
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - I Adamec
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia
| | - A Junaković
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia
| | - B Ruška
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - T Pavičić
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - M Krbot Skorić
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Zagreb, Croatia.,Faculty of Electrical Engineering and Computing, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
49
|
Jagannath VA, Pucci E, Asokan GV, Robak EW. Percutaneous transluminal angioplasty for treatment of chronic cerebrospinal venous insufficiency (CCSVI) in people with multiple sclerosis. Cochrane Database Syst Rev 2019; 5:CD009903. [PMID: 31150100 PMCID: PMC6543952 DOI: 10.1002/14651858.cd009903.pub3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a leading cause of neurological disability in young adults. The most widely accepted hypothesis regarding its pathogenesis is that it is an immune-mediated disease. It has been hypothesised that intraluminal defects, compression, or hypoplasia in the internal jugular or azygos veins may be important factors in the pathogenesis of MS. This condition has been named 'chronic cerebrospinal venous insufficiency' (CCSVI). It has been suggested that these intraluminal defects restrict the normal blood flow from the brain and spinal cord, causing the deposition of iron in the brain and the eventual triggering of an auto-immune response. The proposed treatment for CCSVI is venous percutaneous transluminal angioplasty (PTA), which is claimed to improve the blood flow in the brain thereby alleviating some of the symptoms of MS. This is an update of a review first published in 2012. OBJECTIVES To assess the benefit and safety of venous PTA in people with MS and CCSVI. SEARCH METHODS We searched the Cochrane Multiple Sclerosis and Rare Diseases of the Central Nervous System Group's Specialised Register up to 30 August 2018, CENTRAL (in the Cochrane Library 2018, issue 8), MEDLINE up to 30 August 2018, Embase up to 30 August 2018, metaRegister of Controlled Trials, ClinicalTrials.gov., the Australian New Zealand Clinical Trials Registry, and the World Health Organization (WHO) International Clinical Trials Registry platform. We examined the bibliographies of the included and excluded studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) in which PTA and sham interventions were compared in adults with MS and CCSVI. DATA COLLECTION AND ANALYSIS Two authors independently assessed study eligibility and risk of bias, and extracted data. We reported results as risk ratios (RR) with 95% confidence intervals (CI). We performed statistical analyses using the random-effects model; and we assessed the certainty of the evidence using GRADE. MAIN RESULTS We included three RCTs (238 participants) in this update. One hundred and thirty-four participants were randomised to PTA and 104 to sham treatment. We attributed low risk of bias to two (67%) studies for sequence generation and two (67%) studies for performance bias. All studies were at a low risk of detection bias, attrition bias, reporting bias and other potential sources of bias.There was moderate-quality evidence to suggest that venous PTA did not increase the proportion of patients who had operative or post-operative serious adverse events compared with the sham procedure (RR 3.33, 95% CI 0.36 to 30.44; 3 studies, 238 participants); nor did it increase the proportion of patients who improved on a functional composite measure including walking control, balance, manual dexterity, postvoid residual urine volume, and visual acuity over 12-month follow-up (RR 0.84, 95% CI 0.55 to 1.30; 1 study, 110 participants); nor did it reduce the proportion of patients who experienced new relapses at six- or 12-month follow-up (RR 0.87, 95% CI 0.51 to 1.49; 3 studies, 235 participants). There was no effect of venous PTA on disability worsening measured by the Expanded Disability Status Scale, which was reported at follow-up intervals of six months (one study), 11 months (one study) and 12 months (one study). Quality of life was reported in two studies with no difference between treatment groups. Moderate or severe pain during or post venography was reported in both PTA and sham-procedure participants in all included studies. Venous PTA was not effective in restoring blood flow assessed at one-month (one study) or 12-month follow-up (one study). AUTHORS' CONCLUSIONS This systematic review identified moderate-quality evidence that, compared with sham procedure, venous PTA intervention did not provide benefit on patient-centred outcomes (disability, physical or cognitive functions, relapses, quality of life) in people with MS. Venous PTA has proven to be a safe technique but in view of the available evidence of its ineffectiveness, this intervention cannot be recommended in people with MS. All ongoing trials were withdrawn or terminated and hence this updated review is conclusive. No further randomised clinical studies are needed.
Collapse
Affiliation(s)
- Vanitha A Jagannath
- American Mission HospitalDepartment of PaediatricsManamaManamaBahrainPO Box 1
| | - Eugenio Pucci
- ASUR Marche ‐ Zona Territoriale 9U.O. Neurologia ‐ Ospedale di MacerataVia Santa Lucia, 3MacerataItaly62100
| | - Govindaraj V Asokan
- University of BahrainCollege of Health SciencesSalmaniya Medical ComplexManamaBahrain
| | - Edward W Robak
- MS ConsumerApt 207, 825 McLeod AveFrederictonNBCanadaE3B 9V4
| | | |
Collapse
|
50
|
Habek M. Immune and autonomic nervous system interactions in multiple sclerosis: clinical implications. Clin Auton Res 2019; 29:267-275. [PMID: 30963343 DOI: 10.1007/s10286-019-00605-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/28/2019] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis is characterized by a wide spectrum of clinical manifestations, among which dysfunction of the autonomic nervous system represents an important cause of multiple sclerosis-related disability. The aim of this review is to provide an overview of autonomic dysfunction in people with multiple sclerosis, and to discuss the interactions between the immune and autonomic nervous systems and the effects of these interactions on various aspects of multiple sclerosis. Autonomic dysfunction in people with multiple sclerosis can be demonstrated clinically and on a molecular level. Clinically, it can be demonstrated by measuring autonomic symptoms with the Composite Autonomic Symptom Score (COMPASS-31), and neurophysiologically, with different autonomic nervous system tests. Both symptomatic and objectively determined autonomic dysfunction can be associated with increased risk of multiple sclerosis disease activity. Further supporting these clinical observations are molecular changes in immune cells. Changes in the sympathetic autonomic system, such as different expression of dopaminergic and adrenergic receptors on immune cells, or modulation of the cholinergic anti-inflammatory pathway over different subunits of the nicotinic acetylcholine receptor in the peripheral immune system, may mediate different effects on multiple sclerosis disease activity.
Collapse
Affiliation(s)
- Mario Habek
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Center Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia. .,School of Medicine, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|