51
|
Vollmer TL, Nair KV, Williams IM, Alvarez E. Multiple Sclerosis Phenotypes as a Continuum: The Role of Neurologic Reserve. Neurol Clin Pract 2021; 11:342-351. [PMID: 34476126 PMCID: PMC8382415 DOI: 10.1212/cpj.0000000000001045] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022]
Abstract
Purpose of Review This review presents the hypothesis that loss of neurologic reserve explains onset of progressive multiple sclerosis (PrMS). Recent Findings Evidence supporting the separate classification of PrMS and relapsing multiple sclerosis (RMS) is limited and does not explain PrMS or the response of these patients to therapy. Summary We argue that multiple sclerosis (MS) progresses along a continuum from RMS to PrMS, with differing levels of neurologic reserve accounting for phenotypic differences. In early MS, inflammation causes brain atrophy with symptoms buffered by neurologic reserve. As brain loss from normal aging and MS continues, reserve is depleted and effects of subclinical MS disease activity and aging are unmasked, manifesting as PrMS. Most therapies show limited benefit in PrMS; patients are older, have fewer inflammatory events, and the effects of aging cause continued loss of neurologic function, even if inflammation is terminated. Loss of neurologic reserve means patients with PrMS cannot recover function, unlike patients with RMS.
Collapse
Affiliation(s)
- Timothy L Vollmer
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| | - Kavita V Nair
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| | - Ian M Williams
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| | - Enrique Alvarez
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| |
Collapse
|
52
|
Mestre L, Alonso G, Feliú A, Mecha M, Martín C, Villar LM, Guaza C. Aging and neuroinflammation: Changes in immune cell responses, axon integrity, and motor function in a viral model of progressive multiple sclerosis. Aging Cell 2021; 20:e13440. [PMID: 34355492 PMCID: PMC8441417 DOI: 10.1111/acel.13440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/01/2021] [Accepted: 07/04/2021] [Indexed: 12/12/2022] Open
Abstract
Although aggravated multiple sclerosis (MS) disability has been reported in aged patients, the aging impact on immune cells remodeling within the CNS is not well understood. Here, we investigated the influence of aging on immune cells and the neuroinflammatory and neurodegenerative processes that occur in a well‐established viral model of progressive MS. We found an anomalous presence of CD4+ T, CD8+T, B cells, and cells of myeloid lineage in the CNS of old sham mice whereas a blunted cellular innate and adaptive immune response was observed in Theiler's murine encephalomyelitis virus (TMEV) infected old mice. Microglia and macrophages show opposite CNS viral responses regarding cell counts in the old mice. Furthermore, enhanced expression of Programmed Death‐ligand 1 (PD‐L1) was found in microglia isolated from old TMEV‐infected mice and not in isolated CNS macrophages. Immunocytochemical staining of microglial cells confirms the above differences between young and old mice. Age‐related axonal loss integrity in the mouse spinal cord was found in TMEV mice, but a less marked neurodegenerative process was present in old sham mice compared with young sham mice. TMEV and sham old mice also display alterations in innate and adaptive immunity in the spleen compared to the young mice. Our study supports the need of new or adapted pharmacological strategies for MS elderly patients.
Collapse
Affiliation(s)
- Leyre Mestre
- Neuroimmunology Group Functional and Systems Neurobiology Department Instituto CajalCSIC Madrid Spain
- Red Española de Esclerosis Múltiple (REEM) Barcelona Spain
| | - Graciela Alonso
- Neuroimmunology Group Functional and Systems Neurobiology Department Instituto CajalCSIC Madrid Spain
- Red Española de Esclerosis Múltiple (REEM) Barcelona Spain
| | - Ana Feliú
- Neuroimmunology Group Functional and Systems Neurobiology Department Instituto CajalCSIC Madrid Spain
- Red Española de Esclerosis Múltiple (REEM) Barcelona Spain
| | - Miriam Mecha
- Neuroimmunology Group Functional and Systems Neurobiology Department Instituto CajalCSIC Madrid Spain
- Red Española de Esclerosis Múltiple (REEM) Barcelona Spain
| | - Carolina Martín
- Neuroimmunology Group Functional and Systems Neurobiology Department Instituto CajalCSIC Madrid Spain
| | - Luisa M. Villar
- Red Española de Esclerosis Múltiple (REEM) Barcelona Spain
- Immunology Department Hospital Universitario Ramón y CajalInstituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Madrid Spain
| | - Carmen Guaza
- Neuroimmunology Group Functional and Systems Neurobiology Department Instituto CajalCSIC Madrid Spain
- Red Española de Esclerosis Múltiple (REEM) Barcelona Spain
| |
Collapse
|
53
|
Monahan RC, Inglese F, Middelkoop H, van Buchem M, Huizinga TW, Kloppenburg M, Ronen I, Steup-Beekman GM, de Bresser J. White matter hyperintensities associate with cognitive slowing in patients with systemic lupus erythematosus and neuropsychiatric symptoms. RMD Open 2021; 7:rmdopen-2021-001650. [PMID: 34321253 PMCID: PMC8320250 DOI: 10.1136/rmdopen-2021-001650] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/17/2021] [Indexed: 12/31/2022] Open
Abstract
Objective To compare cognitive function between patients with different phenotypes of neuropsychiatric systemic lupus erythematosus (NPSLE) and assess its association with brain and white matter hyperintensity (WMH) volumes. Methods Patients attending the Leiden University Medical Centre NPSLE clinic between 2007 and 2015 without large brain infarcts were included (n=151; 42±13 years, 91% women). In a multidisciplinary consensus meeting, neuropsychiatric symptoms were attributed to systemic lupus erythematosus (SLE) (NPSLE, inflammatory (n=24) or ischaemic (n=12)) or to minor/non-NPSLE (n=115). Multiple regression analyses were performed to compare cognitive function between NPSLE phenotypes and to assess associations between brain and WMH volumes and cognitive function cross-sectionally. Results Global cognitive function was impaired in 5%, learning and memory (LM) in 46%, executive function and complex attention (EFCA) in 39% and psychomotor speed (PS) in 46% of all patients. Patients with inflammatory NPSLE showed the most cognitive impairment in all domains (p≤0.05). Higher WMH volume associated with lower PS in the total group (B: −0.14 (95% CI −0.32 to −0.02)); especially in inflammatory NPSLE (B: −0.36 (95% CI −0.60 to −0.12). In the total group, lower total brain volume and grey matter volume associated with lower cognitive functioning in all domains (all: 0.00/0.01 (0.00;0.01)) and lower white matter volume associated with lower LM, EFCA and PS (all: 0.00/0.01 (0.00;0.01)). Conclusion We demonstrated that an association between brain and WMH volumes and cognitive function is present in patients with SLE, but differs between (NP)SLE phenotypes. WMHs associated with PS especially in inflammatory NPSLE, which suggests a different, potentially more severe underlying pathophysiological mechanism of cognitive impairment in this phenotype.
Collapse
Affiliation(s)
| | - Francesca Inglese
- Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Huub Middelkoop
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.,Institute of Psychology, Health, Medical and Neuropsychology Unit, Leiden University, Leiden, the Netherlands
| | - Mark van Buchem
- Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Tom Wj Huizinga
- Rheumatology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Margreet Kloppenburg
- Rheumatology, Leiden University Medical Centre, Leiden, the Netherlands.,Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Itamar Ronen
- Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gerda M Steup-Beekman
- Rheumatology, Leiden University Medical Centre, Leiden, the Netherlands.,Department of Rheumatology, Medisch Centrum Haaglanden, the Hague, the Netherlands
| | - Jeroen de Bresser
- Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
54
|
Epstein S, Fong KT, De Jager PL, Levine L, Riley C, Wesley S, Vargas WS, Farber R. Evaluation of ocrelizumab in older progressive multiple sclerosis patients. Mult Scler Relat Disord 2021; 55:103171. [PMID: 34329872 DOI: 10.1016/j.msard.2021.103171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/24/2021] [Accepted: 07/22/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Seminal trials evaluating anti-CD20 therapy in progressive MS primarily found benefit in younger, less-disabled patients with more inflammatory disease activity. The risks and benefits of ocrelizumab use in older patients with progressive froms of MS are not known. METHODS Retrospective chart review was performed for patients older than 55 with primary or secondary progressive MS at the time of ocrelizumab initiation. Clinical endpoints from 2 years prior to anti-CD20 therapy served as a within-subject control. RESULTS Data was reviewed for 56 patients older than the age of 55 at the time of ocrelizumab initiation. Of 37 patients with 2-years of follow up on ocrelizumab, 40%(n=15) experienced confirmed disability progression (CDP) while 60% (n=22) remained stable or improved. 24 patients had data available for the within-subject control; for these patients, median age was 67, baseline EDSS 6.3, and disease duration 20.5 years. Prior to anti-CD20 therapy, 58% (n=14) of patients remained stable and 42% (n=10) experienced CDP. After ocrelizumab initiation, 71% (n=17) remained stable and 29% (n=7) experienced CDP. There was no difference between CDP (p=0.54) or change in EDSS (p=0.09) between time periods. Ocrelizumab was well tolerated and no difference in infection rate was seen using the within-subject control. CONCLUSIONS We found no difference in clinical endpoints for patients on ocrelizumab compared to prior to anti-CD20 therapy; however, we could not exclude a modest effect given our sample size. Larger trials are needed to evaluate ocrelizumab use in this understudied MS subpopulation.
Collapse
Affiliation(s)
- Samantha Epstein
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Kathryn T Fong
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; Swedish Neuroscience Institute, Seattle, WA, USA
| | - Philip L De Jager
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Libby Levine
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Claire Riley
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah Wesley
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Wendy S Vargas
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Rebecca Farber
- Columbia Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
55
|
Dema M, Eixarch H, Villar LM, Montalban X, Espejo C. Immunosenescence in multiple sclerosis: the identification of new therapeutic targets. Autoimmun Rev 2021; 20:102893. [PMID: 34237417 DOI: 10.1016/j.autrev.2021.102893] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022]
Abstract
The number of elderly multiple sclerosis (MS) patients is growing, mainly due to the increase in the life expectancy of the general population and the availability of effective disease-modifying treatments. However, current treatments reduce the frequency of relapses and slow the progression of the disease, but they cannot stop the disability accumulation associated with disease progression. One possible explanation is the impact of immunosenescence, which is associated with the accumulation of unusual immune cell subsets that are thought to have a role in the development of an early ageing process in autoimmunity. Here, we provide a recent overview of how senescence affects immune cell function and how it is involved in the pathogenesis of autoimmune diseases, particularly MS. Numerous studies have demonstrated age-related immune changes in experimental autoimmune encephalomyelitis models, and the premature onset of immunosenescence has been demonstrated in MS patients. Therefore, potential therapeutic strategies based on rejuvenating the immune system have been proposed. Senolytics and regenerative strategies using haematopoietic stem cells, therapies based on rejuvenating oligodendrocyte precursor cells, microglia and monocytes, thymus cells and senescent B and T cells are capable of reversing the process of immunosenescence and could have a beneficial impact on the progression of MS.
Collapse
Affiliation(s)
- María Dema
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, 08035, Barcelona, Spain; Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, 08035, Barcelona, Spain; Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Luisa M Villar
- Red Española de Esclerosis Múltiple (REEM), Spain; Servicio de Inmunología, Hospital Universitario Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034, Madrid, Spain.
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, 08035, Barcelona, Spain; Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, 08035, Barcelona, Spain; Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| |
Collapse
|
56
|
Ribeiro AR, Barros C, Barateiro A, Howlett SE, Fernandes A. Improved assessment of overall health in variably aged murine models of Multiple Sclerosis with a novel frailty index tool. J Gerontol A Biol Sci Med Sci 2021; 77:1-9. [PMID: 34181005 DOI: 10.1093/gerona/glab185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Indexed: 11/12/2022] Open
Abstract
The experimental autoimmune encephalomyelitis (EAE) model is the most commonly used animal model of Multiple Sclerosis (MS). However, phenotypic characterization of mice based on the traditional 5-point clinical paralysis scale does not fully capture disease progression. The frailty index (FI) conceptualizes frailty as the accumulation of health deficits and it is widely used to assess overall health in aging humans and pre-clinical models. Here we adapted an established mouse FI tool for use in EAE mice and determined whether this could evaluate general signs of health in variably aged female EAE mice. The EAE-Clinical FI included 34 items related to clinical signs and deficits characteristic of aging and MS. This tool clearly showed more detailed EAE progression and severity at all ages, highlighting changes in systems other than motor paralysis measured with the traditional 5-point paralysis scale. When we induced disease at 3- and 6-months-of-age, mice showed typical EAE clinical manifestations with peak disease severity between 17-19 days post-induction and mean frailty scores of 0.36 ± 0.04 (3-month-old) and 0.43 ± 0.05 (6-month-old). By contrast, disease severity peaked after 14 days in 12-month-old mice. They showed atypical signs including wobbling, early belly drag, and splayed hindlegs that were better captured with the EAE-Clinical FI. Peak frailty scores also were higher than those of younger animals (0.54 ± 0.04). As MS most often develops in young to middle-aged people, this new tool may have significant value for use in EAE animal studies as first step towards translation to people with MS.
Collapse
Affiliation(s)
- Ana Rita Ribeiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Barros
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Andreia Barateiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Adelaide Fernandes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal.,Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
57
|
Miner AE, Graves JS. What telomeres teach us about MS. Mult Scler Relat Disord 2021; 54:103084. [PMID: 34371369 DOI: 10.1016/j.msard.2021.103084] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/14/2021] [Accepted: 06/09/2021] [Indexed: 02/03/2023]
Abstract
While the precise mechanisms driving progressive forms of MS are not fully understood, patient age has clear impact on disease phenotype. The very young with MS have high relapse rates and virtually no progressive disease, whereas older patients tend to experience more rapid disability accumulation with few relapses. Defining a patient's biological age may offer more precision in determining the role of aging processes in MS phenotype and pathophysiology than just working with an individual's birthdate. The most well recognized measurement of an individual's "biological clock" is telomere length (TL). While TL may differ across tissue types in an individual, most cells TL correlate well with leukocyte TL (LTL), which is the most common biomarker used for aging. LTL has been associated with risk for aging related diseases and most recently with higher levels of disability and brain atrophy in people living with MS. LTL explains 15% of the overall association of chronological age with MS disability level. While LTL may be used just as a biomarker of overall somatic aging processes, triggering of the DNA damage response by telomere attrition leads to senescence pathways that are likely highly relevant to a chronic autoimmune disease. Considering reproductive aging factors, particularly ovarian aging in women, which correlates with LTL and oocyte telomere length, may complement measurements of somatic aging in understanding MS progression. The key to stopping non-relapse related progression in MS might lie in targeting pathways related to biological aging effects on the immune and nervous systems.
Collapse
Affiliation(s)
- Annalise E Miner
- Department of Neurosciences, University of California, San Diego, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California, San Diego, USA.
| |
Collapse
|
58
|
Nayeri T, Sarvi S, Sharif M, Daryani A. Toxoplasma gondii: A possible etiologic agent for Alzheimer's disease. Heliyon 2021; 7:e07151. [PMID: 34141920 PMCID: PMC8187970 DOI: 10.1016/j.heliyon.2021.e07151] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/03/2021] [Accepted: 05/24/2021] [Indexed: 01/03/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is one of the most pervasive neurotropic pathogens causing different lesions in a wide variety of mammals as intermediate hosts, including humans. It is estimated that one-third of the world population is infected with T. gondii; however, for a long time, there has been much interest in the examination of the possible role of this parasite in the development of mental disorders, such as Alzheimer's disease (AD). T. gondii may play a role in the progression of AD using mechanisms, such as the induction of the host's immune responses, inflammation of the central nervous system (CNS), alteration in the levels of neurotransmitters, and activation of indoleamine-2,3-dyoxigenase. This paper presents an appraisal of the literature, reports, and studies that seek to the possible role of T. gondii in the development of AD. For achieving the purpose of the current study, a search of six English databases (PubMed, ScienceDirect, Web of Science, Scopus, ProQuest, and Google Scholar) was performed. The results support the involvement of T. gondii in the induction and development of AD. Indeed, T. gondii can be considered a risk factor for the development of AD and requires the special attention of specialists and patients. Furthermore, the results of this study may contribute to prevent or delay the progress of AD worldwide. Therefore, it is required to carry out further studies in order to better perceive the parasitic mechanisms in the progression of AD.
Collapse
Affiliation(s)
- Tooran Nayeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehdi Sharif
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
59
|
Papp V, Buron MD, Siersma V, Rasmussen PV, Illes Z, Kant M, Hilt C, Mezei Z, Roshanisefat H, Sejbæk T, Weglewski A, van Wingerden J, Geertsen SS, Bramow S, Sellebjerg F, Magyari M. Real-world outcomes for a complete nationwide cohort of more than 3200 teriflunomide-treated multiple sclerosis patients in The Danish Multiple Sclerosis Registry. PLoS One 2021; 16:e0250820. [PMID: 34003862 PMCID: PMC8130956 DOI: 10.1371/journal.pone.0250820] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/15/2021] [Indexed: 11/18/2022] Open
Abstract
Objective Teriflunomide is a once-daily, oral disease-modifying therapy (DMT) for relapsing forms of multiple sclerosis (MS). We studied clinical outcomes in a real-world setting involving a population-based large cohort of unselected patients enrolled in The Danish Multiple Sclerosis Registry (DMSR) who started teriflunomide treatment between 2013–2019. Methods This was a complete nationwide population-based cohort study with prospectively enrolled unselected cases. Demographic and disease-specific patient parameters related to treatment history, efficacy outcomes, and discontinuation and switching rates among other clinical variables were assessed at baseline and during follow-up visits. Results A total of 3239 patients (65.4% female) started treatment with teriflunomide during the study period, 56% of whom were treatment-naïve. Compared to previously treated patients, treatment-naïve patients were older on average at disease onset, had a shorter disease duration, a lower Expanded Disability Status Scale score at teriflunomide treatment start and more frequently experienced a relapse in the 12 months prior to teriflunomide initiation. In the 3001 patients initiating teriflunomide treatment at least 12 months before the cut-off date, 72.7% were still on treatment one year after treatment start. Discontinuations in the first year were due mainly to adverse events (15.6%). Over the full follow-up period, 47.5% of patients discontinued teriflunomide treatment. Sixty-three percent of the patients treated with teriflunomide for 5 years were relapse-free, while significantly more treatment-naïve versus previously treated patients experienced a relapse during the follow-up (p<0.0001). Furthermore, 85% of the patients with available data were free of disability worsening at the end of follow-up. Conclusions Solid efficacy and treatment persistence data consistent with other real-world studies were obtained over the treatment period. Treatment outcomes in this real-world scenario of the population-based cohort support previous findings that teriflunomide is an effective and generally well-tolerated DMT for relapsing MS patients with mild to moderate disease activity.
Collapse
Affiliation(s)
- Viktoria Papp
- Odense University Hospital, Odense, Denmark
- * E-mail:
| | - Mathias Due Buron
- The Danish Multiple Sclerosis Registry, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Danish Multiple Sclerosis Center, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Volkert Siersma
- The Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | - Zsolt Mezei
- Aalborg University Hospital, Aalborg, Denmark
| | | | - Tobias Sejbæk
- Department of Neurology, Hospital of South West Jutland, Esbjerg, Denmark
| | | | | | | | - Stephan Bramow
- Danish Multiple Sclerosis Center, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Melinda Magyari
- The Danish Multiple Sclerosis Registry, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Danish Multiple Sclerosis Center, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
60
|
Rebelo AL, Gubinelli F, Roost P, Jan C, Brouillet E, Van Camp N, Drake RR, Saldova R, Pandit A. Complete spatial characterisation of N-glycosylation upon striatal neuroinflammation in the rodent brain. J Neuroinflammation 2021; 18:116. [PMID: 33993882 PMCID: PMC8127229 DOI: 10.1186/s12974-021-02163-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neuroinflammation is an underlying pathology of all neurological conditions, the understanding of which is still being comprehended. A specific molecular pathway that has been overlooked in neuroinflammation is glycosylation (i.e., post-translational addition of glycans to the protein structure). N-glycosylation is a specific type of glycosylation with a cardinal role in the central nervous system (CNS), which is highlighted by congenital glycosylation diseases that result in neuropathological symptoms such as epilepsy and mental retardation. Changes in N-glycosylation can ultimately affect glycoproteins' functions, which will have an impact on cell machinery. Therefore, characterisation of N-glycosylation alterations in a neuroinflammatory scenario can provide a potential target for future therapies. METHODS With that aim, the unilateral intrastriatal injection of lipopolysaccharide (LPS) in the adult rat brain was used as a model of neuroinflammation. In vivo and post-mortem, quantitative and spatial characterisation of both neuroinflammation and N-glycome was performed at 1-week post-injection of LPS. These aspects were investigated through a multifaceted approach based on positron emission tomography (PET), quantitative histology, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), liquid chromatography and matrix-assisted laser desorption ionisation mass spectrometry imaging (MALDI-MSI). RESULTS In the brain region showing LPS-induced neuroinflammation, a significant decrease in the abundance of sialylated and core fucosylated structures was seen (approximately 7.5% and 8.5%, respectively), whereas oligomannose N-glycans were significantly increased (13.5%). This was confirmed by MALDI-MSI, which provided a high-resolution spatial distribution of N-glycans, allowing precise comparison between normal and diseased brain hemispheres. CONCLUSIONS Together, our data show for the first time the complete profiling of N-glycomic changes in a well-characterised animal model of neuroinflammation. These data represent a pioneering step to identify critical targets that may modulate neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ana Lúcia Rebelo
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Francesco Gubinelli
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Pauline Roost
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Caroline Jan
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Emmanuel Brouillet
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Nadja Van Camp
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, USA
| | - Radka Saldova
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
- National Institute for Bioprocessing Research and Training (NIBRT), University College Dublin, Dublin, Ireland
- UCD School of Medicine, UCD Conway Institute of Biomolecular and Biomedical, Dublin, Ireland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland.
| |
Collapse
|
61
|
Scalfari A. MS can be considered a primary progressive disease in all cases, but some patients have superimposed relapses - Yes. Mult Scler 2021; 27:1002-1004. [PMID: 33874813 PMCID: PMC8142117 DOI: 10.1177/13524585211001789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College London, Charing Cross Hospital, London, UK
| |
Collapse
|
62
|
Bove R, Okai A, Houtchens M, Elias-Hamp B, Lugaresi A, Hellwig K, Kubala Havrdová E. Effects of Menopause in Women With Multiple Sclerosis: An Evidence-Based Review. Front Neurol 2021; 12:554375. [PMID: 33815241 PMCID: PMC8017266 DOI: 10.3389/fneur.2021.554375] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 02/17/2021] [Indexed: 12/20/2022] Open
Abstract
Over two thirds of all individuals who develop multiple sclerosis (MS) will be women prior to the age of menopause. Further, an estimated 30% of the current MS population consists of peri- or postmenopausal women. The presence of MS does not appear to influence age of menopausal onset. In clinical practice, symptoms of MS and menopause can frequently overlap, including disturbances in cognition, mood, sleep, and bladder function, which can create challenges in ascertaining the likely cause of symptoms to be treated. A holistic and comprehensive approach to address these common physical and psychological changes is often suggested to patients during menopause. Although some studies have suggested that women with MS experience reduced relapse rates and increased disability progression post menopause, the data are not consistent enough for firm conclusions to be drawn. Mechanisms through which postmenopausal women with MS may experience disability progression include neuroinflammation and neurodegeneration from age-associated phenomena such as immunosenescence and inflammaging. Additional effects are likely to result from reduced levels of estrogen, which affects MS disease course. Following early retrospective studies of women with MS receiving steroid hormones, more recent interventional trials of exogenous hormone use, albeit as oral contraceptive, have provided some indications of potential benefit on MS outcomes. This review summarizes current research on the effects of menopause in women with MS, including the psychological impact and symptoms of menopause on disease worsening, and the treatment options. Finally, we highlight the need for more inclusion of MS patients from underrepresented racial and geographic groups in clinical trials, including among menopausal women.
Collapse
Affiliation(s)
- Riley Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Annette Okai
- Multiple Sclerosis Treatment Center of Dallas, Dallas, TX, United States
| | - Maria Houtchens
- Partners Multiple Sclerosis Center, Brigham and Women's Hospital, Boston, MA, United States
| | - Birte Elias-Hamp
- Neurological Private Practice, Institute of Neuroimmunology and Multiple Sclerosis, Hamburg, Germany
| | - Alessandra Lugaresi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Kerstin Hellwig
- Department of Neurology, Ruhr University Bochum and St. Josef-Hospital, Bochum, Germany
| | - Eva Kubala Havrdová
- Department of Neurology and Center of Clinical Neuroscience, First Medical Faculty, General University Hospital, Charles University, Prague, Czechia
| |
Collapse
|
63
|
Zhang B, Gems D. Gross ways to live long: Parasitic worms as an anti-inflammaging therapy? eLife 2021; 10:65180. [PMID: 33526169 PMCID: PMC7853715 DOI: 10.7554/elife.65180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Evolutionary medicine argues that disease can arise because modern conditions do not match those in which we evolved. For example, a decline in exposure to commensal microbes and gastrointestinal helminths in developed countries has been linked to increased prevalence of allergic and autoimmune inflammatory disorders (the hygiene hypothesis). Accordingly, probiotic therapies that restore ‘old friend’ microbes and helminths have been explored as Darwinian treatments for these disorders. A further possibility is that loss of old friend commensals also increases the sterile, aging-associated inflammation known as inflammaging, which contributes to a range of age-related diseases, including cardiovascular disease, dementia, and cancer. Interestingly, Crowe et al., 2020 recently reported that treatment with a secreted glycoprotein from a parasitic nematode can protect against murine aging by induction of anti-inflammatory mechanisms. Here, we explore the hypothesis that restorative helminth therapy would have anti-inflammaging effects. Could worm infections provide broad-spectrum protection against age-related disease?
Collapse
Affiliation(s)
- Bruce Zhang
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - David Gems
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| |
Collapse
|
64
|
Inojosa H, Proschmann U, Akgün K, Ziemssen T. Should We Use Clinical Tools to Identify Disease Progression? Front Neurol 2021; 11:628542. [PMID: 33551982 PMCID: PMC7859270 DOI: 10.3389/fneur.2020.628542] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023] Open
Abstract
The presence of disability progression in multiple sclerosis (MS) is an important hallmark for MS patients in the course of their disease. The transition from relapsing remitting (RRMS) to secondary progressive forms of the disease (SPMS) represents a significant change in their quality of life and perception of the disease. It could also be a therapeutic key for opportunities, where approaches different from those in the initial phases of the disease can be adopted. The characterization of structural biomarkers (e.g., magnetic resonance imaging or neurofilament light chain) has been proposed to differentiate between both phenotypes. However, there is no definite threshold between them. Whether the risk of clinical progression can be predicted by structural markers at early disease phases is still a focus of clinical research. However, several theories and pathological evidence suggest that both disease phenotypes are part of a continuum with common pathophysiological mechanisms. In this case, the clinical evaluation of the patients would play a preponderant role above destruction biomarkers for the early identification of disability progression and SPMS. For this purpose, the use of clinical tools beyond the Expanded Disability Status Scale (EDSS) should be considered. Besides established functional tests such as the Multiple Sclerosis Functional Composite (MSFC), patient's neurological history or digital resources may help neurologists in the decision-taking. In this article, we discuss arguments for the use of clinical markers in the detection of secondary progressive MS and the characterization of progressive disease activity.
Collapse
Affiliation(s)
- Hernan Inojosa
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Undine Proschmann
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Katja Akgün
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Tjalf Ziemssen
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| |
Collapse
|
65
|
Menascu S, Khavkin Y, Zilkha‐Falb R, Dolev M, Magalashvili D, Achiron A, Gurevich M. Clinical and transcriptional recovery profiles in pediatric and adult multiple sclerosis patients. Ann Clin Transl Neurol 2021; 8:81-94. [PMID: 33197148 PMCID: PMC7818128 DOI: 10.1002/acn3.51244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 09/07/2020] [Accepted: 10/18/2020] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE To determine whether pediatric-onset multiple sclerosis (POMS) and adults-onset multiple sclerosis (AOMS) patients are different in initial disease severity and recovery and to investigate the associations with peripheral blood mononuclear cells (PBMCs) transcriptional profiles. METHODS Clinical and radiological severity of first and second relapses and 6-month recovery were analyzed in 2153 multiple sclerosis (MS) patients and compared between POMS (onset at 8-18years old) and AOMS (onset at 19-40 years old) patients. PBMCs transcriptomes of 15 POMS and 15 gender-matched AOMS patients were analyzed 6 months after the first relapse and compared to 55 age-matched healthy controls. Differentially Expressed Genes (DEGs) with a false discovery rate ≤ 10% were evaluated using the Partek software. RESULTS POMS had increased Expanded Disability Status Scale (EDSS) score at first and second relapses, higher brain gadolinium-enhancing T1-lesions volume at first relapse, and more complete recovery after both relapses compared to AOMS. POMS patients, who recovered completely from the first relapse, were characterized by 19 DEGs that were mainly related to suppression of antigen presentation. Six upstream regulators of these genes were differentially expressed between pediatric and adult healthy controls. POMS patients, who showed no recovery from the first relapse, were characterized by 28 DEGs that were mainly associated with B-cell activation. Five upstream regulators of these genes were differentially expressed between pediatric and adult healthy controls. INTERPRETATION POMS patients may have more severe first and second relapses than AOMS. However, most often, POMS have better recovery that may be attributed to PBMCs age-related transcriptional profiles associated with antigen presentation and B-cell activation.
Collapse
Affiliation(s)
- Shay Menascu
- Multiple Sclerosis Center, Sheba Medical CenterRamat‐GanIsrael
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
| | - Yulia Khavkin
- Multiple Sclerosis Center, Sheba Medical CenterRamat‐GanIsrael
| | | | - Mark Dolev
- Multiple Sclerosis Center, Sheba Medical CenterRamat‐GanIsrael
| | | | - Anat Achiron
- Multiple Sclerosis Center, Sheba Medical CenterRamat‐GanIsrael
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
| | - Michael Gurevich
- Multiple Sclerosis Center, Sheba Medical CenterRamat‐GanIsrael
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
| |
Collapse
|
66
|
Krokidis MG. Biomarker-Driven Analysis Using High-Throughput Approaches in Neuroinflammation and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:51-58. [DOI: 10.1007/978-3-030-78787-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
67
|
|
68
|
Klistorner S, Barnett MH, Yiannikas C, Barton J, Parratt J, You Y, Graham SL, Klistorner A. Expansion of chronic lesions is linked to disease progression in relapsing-remitting multiple sclerosis patients. Mult Scler 2020; 27:1533-1542. [PMID: 33215557 DOI: 10.1177/1352458520974357] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Slow-burning inflammation is putatively associated with lesion expansion and leads to progressive loss of axons and disability worsening. OBJECTIVE To investigate the incidence and extent of chronic white matter lesion expansion in relapsing-remitting multiple sclerosis (RRMS) patients and to evaluate its relationship with biomarkers of disease progression. METHODS Pre- and post-gadolinium T1, fluid-attenuated inversion recovery (FLAIR) and diffusion tensor images were acquired from 33 patients. Lesional activity were analysed between baseline and 48 months using custom-designed software. RESULTS A total of 569 lesions were identified as chronic at baseline, of which 261 were expanding, 236 were stable and 72 were shrinking. In addition, 139 new lesions (both confluent and free-standing) were observed. Chronic lesion expansion was associated with patient's age and accounted for the bulk (67.3%) of total brain lesion volume increase, while only 32.7% was attributable to new lesion formation. Change in chronic lesion volume correlated with the rate of brain atrophy (r = -0.57, p = 0.001), change of Expanded Disability Status Scale (EDSS; r = 0.38, p = 0.03) and an increase of isotropic diffusivity inside the lesions (r = 0.75, p < 0.001). CONCLUSION Expansion of chronic lesions in RRMS patients is the primary determinant of increased T2 total lesion load. It significantly contributes to disease progression and partially driving axonal loss inside the lesions and brain damage outside of lesional tissue.
Collapse
Affiliation(s)
- Samuel Klistorner
- Sydney Medical School, Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Michael H Barnett
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia/Sydney Neuroimaging Analysis Centre, Sydney, NSW, Australia
| | | | - Joshua Barton
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - John Parratt
- Royal North Shore Hospital, Sydney, NSW, Australia
| | - Yuyi You
- Sydney Medical School, Save Sight Institute, The University of Sydney, Sydney, NSW, Australia/Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Alexander Klistorner
- Sydney Medical School, Save Sight Institute, The University of Sydney, Sydney, NSW, Australia/Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
69
|
Stojić-Vukanić Z, Hadžibegović S, Nicole O, Nacka-Aleksić M, Leštarević S, Leposavić G. CD8+ T Cell-Mediated Mechanisms Contribute to the Progression of Neurocognitive Impairment in Both Multiple Sclerosis and Alzheimer's Disease? Front Immunol 2020; 11:566225. [PMID: 33329528 PMCID: PMC7710704 DOI: 10.3389/fimmu.2020.566225] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Neurocognitive impairment (NCI) is one of the most relevant clinical manifestations of multiple sclerosis (MS). The profile of NCI and the structural and functional changes in the brain structures relevant for cognition in MS share some similarities to those in Alzheimer's disease (AD), the most common cause of neurocognitive disorders. Additionally, despite clear etiopathological differences between MS and AD, an accumulation of effector/memory CD8+ T cells and CD8+ tissue-resident memory T (Trm) cells in cognitively relevant brain structures of MS/AD patients, and higher frequency of effector/memory CD8+ T cells re-expressing CD45RA (TEMRA) with high capacity to secrete cytotoxic molecules and proinflammatory cytokines in their blood, were found. Thus, an active pathogenetic role of CD8+ T cells in the progression of MS and AD may be assumed. In this mini-review, findings supporting the putative role of CD8+ T cells in the pathogenesis of MS and AD are displayed, and putative mechanisms underlying their pathogenetic action are discussed. A special effort was made to identify the gaps in the current knowledge about the role of CD8+ T cells in the development of NCI to "catalyze" translational research leading to new feasible therapeutic interventions.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Senka Hadžibegović
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Olivier Nicole
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Mirjana Nacka-Aleksić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Sanja Leštarević
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
70
|
Potential anti-neuroinflammatory compounds from Australian plants - A review. Neurochem Int 2020; 142:104897. [PMID: 33186611 DOI: 10.1016/j.neuint.2020.104897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022]
Abstract
Neuroinflammation is a complex response to brain injury involving the activation of glia, release of inflammatory mediators, such as cytokines and chemokines, and generation of reactive oxygen and nitrogen species. Even though it is considered an event secondary to neuronal death or dysfunction, neuro-inflammation comprises a majority of the non-neuronal contributors to the cause and progression of neurodegenerative diseases like Alzheimer's Disease (AD), Parkinson's Disease (PD), Multiple Sclerosis (MS), Chronic Traumatic Encephalopathy (CTE) and others. As a result of the lack of effectiveness of current treatments for neurodegenerative diseases, neuroinflammation has become a legitimate therapeutic target for drug discovery, leading to the study of various in vivo and in vitro models of neuroinflammation. Several molecules sourced from plants have displayed anti-inflammatory properties in the study of neurodegenerative diseases. A group of these anti-inflammatory compounds has been classified as cytokine-suppressive anti-inflammatory drugs (CSAIDs), which target the pro-inflammatory AP1 and nuclear factor-κB signaling pathways and inhibit the expression of many pro-inflammatory cytokines, such as interleukin IL-1, IL-6, TNF-α, or nitric oxide. Australian plants, thriving amid the driest inhabited continent of the world, are an untapped source of chemical diversity in the form of secondary metabolites. These compounds are produced in response to biotic and abiotic stresses that the plants are exposed to in the highly biodiverse environment. This review is an attempt to highlight anti-inflammatory compounds isolated from Australian plants.
Collapse
|
71
|
Mésidor M, Sylvestre MP, Marrie RA, Rousseau MC. Does age at diagnosis influence the use of health services for multiple sclerosis? Mult Scler Relat Disord 2020; 46:102555. [DOI: 10.1016/j.msard.2020.102555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/17/2020] [Accepted: 09/30/2020] [Indexed: 11/25/2022]
|
72
|
Prosperini L, Haggiag S, Tortorella C, Galgani S, Gasperini C. Age-related adverse events of disease-modifying treatments for multiple sclerosis: A meta-regression. Mult Scler 2020; 27:1391-1402. [PMID: 33104449 DOI: 10.1177/1352458520964778] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To verify the hypothesis of an age-dependent increase of infections and neoplasms in patients with multiple sclerosis (MS) under disease-modifying treatments (DMTs) with different mechanisms of action. METHODS We extracted relevant data from 45 randomized clinical trials (RCTs) on currently licensed DMTs. We fitted inverse-variance weighted meta-regressions with random-effects models to estimate whether age and/or mechanism of action (immunomodulatory, sequestrating, and depletive) of currently licensed DMTs influenced the difference between experimental arm and control arm in the incidence of specific adverse events, namely, overall infections, opportunistic infections, and neoplasms. RESULTS A higher incidence of overall infections was observed in RCTs with depletive DMTs (event-rate ratio = 1.25, p < 0.001). Herpetic infections were more frequently observed in RCTs with both depletive (event-rate ratio = 3.51, p < 0.001) and, to a lesser extent, sequestrating DMTs (event-rate ratio = 1.52, p = 0.078). The interaction of age with depletive DMTs was associated with higher incidence of neoplasms (p = 0.017), especially above 45 years of age. DISCUSSION Our study supports a detrimental effect of age on the safety profile of depletive DMTs, with an increased incidence of neoplasms especially over 45 years of age. We failed to demonstrate an age-related increased incidence of infections, possibly due to latency in their occurrence.
Collapse
Affiliation(s)
- Luca Prosperini
- Department of Neurosciences, San Camillo-Forlanini Hospital, Rome, Italy
| | - Shalom Haggiag
- Department of Neurosciences, San Camillo-Forlanini Hospital, Rome, Italy
| | - Carla Tortorella
- Department of Neurosciences, San Camillo-Forlanini Hospital, Rome, Italy
| | - Simonetta Galgani
- Department of Neurosciences, San Camillo-Forlanini Hospital, Rome, Italy
| | - Claudio Gasperini
- Department of Neurosciences, San Camillo-Forlanini Hospital, Rome, Italy
| |
Collapse
|
73
|
Rahmani S, Noorolyai S, Ayromlou H, Khaze Shahgoli V, Shanehbandi D, Baghbani E, Abdoli Shadbad M, Jigari-Asl F, Khamiriyan M, Safaralizadeh R, Baradaran B. The expression analyses of RMRP, DDX5, and RORC in RRMS patients treated with different drugs versus naïve patients and healthy controls. Gene 2020; 769:145236. [PMID: 33068674 DOI: 10.1016/j.gene.2020.145236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 01/22/2023]
Abstract
Although T helper 17 (Th17) lymphocytes protect mucosal barriers against infections, they have been implicated in the development of multiple sclerosis (MS). RORC and DDX5 can regulate Th17 differentiation and the development of MS. Since RMRP, as a long non-coding RNA (lncRNA), can mediate the RORC-DDX5 complex, this lncRNA can be involved in developing MS. This study investigated the expression levels of RORC, DDX5, and RMRP in treatment-naïve relapsing-remitting multiple sclerosis (RRMS) patients, healthy controls, and RRMS patients treated with IFNβ-1α or fingolimod, or dimethyl fumarate (DMF), or glatiramer acetate (GA). There was substantial up-regulation in the expression of RORC, DDX5, and RMRP in treatment-naïve RRMS patients compared to healthy controls. Among the comparisons of their expressions in the different groups of treated patients with treatment-naïve patients, only the down-regulation of the RMRP expression level was significant in IFNβ-1α-treated patients. Also, these changes were more pronounced in female patient groups. Our analyses have highlighted the high diagnostic value of RORC, DDX5, and RMRP in treatment-naïve RRMS patients. Furthermore, RMRP has demonstrated moderate positive correlations with the expression of DDX5 and RORC in treated RRMS patients.
Collapse
Affiliation(s)
- Shima Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hormoz Ayromlou
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farinaz Jigari-Asl
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Khamiriyan
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
74
|
Gritsenko A, Green JP, Brough D, Lopez-Castejon G. Mechanisms of NLRP3 priming in inflammaging and age related diseases. Cytokine Growth Factor Rev 2020; 55:15-25. [PMID: 32883606 PMCID: PMC7571497 DOI: 10.1016/j.cytogfr.2020.08.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023]
Abstract
The NLRP3 inflammasome is a vital part of the innate immune response, whilst its aberrant activation drives the progression of a number of non-communicable diseases. Thus, NLRP3 inflammasome assembly must be tightly controlled at several checkpoints. The priming step of NLRP3 inflammasome activation is associated with increased NLRP3 gene expression, as well as post-translational modifications that control NLRP3 levels and licence the NLRP3 protein for inflammasome assembly. Increasing life expectancy in modern society is accompanied by a growing percentage of elderly individuals. The process of aging is associated with chronic inflammation that drives and/or worsens a range of age related non-communicable conditions. The NLRP3 inflammasome is known to contribute to pathological inflammation in many settings, but the mechanisms that prime NLRP3 for activation throughout aging and related co-morbidities have not been extensively reviewed. Here we dissect the biochemical changes that occur during aging and the pathogenesis of age related diseases and analyse the mechanisms by which they prime the NLRP3 inflammasome, thus exacerbating inflammation.
Collapse
Affiliation(s)
- Anna Gritsenko
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jack P Green
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - David Brough
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Gloria Lopez-Castejon
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
75
|
Amato MP, Fonderico M, Portaccio E, Pastò L, Razzolini L, Prestipino E, Bellinvia A, Tudisco L, Fratangelo R, Comi G, Patti F, De Luca G, Brescia Morra V, Cocco E, Pozzilli C, Sola P, Bergamaschi R, Salemi G, Inglese M, Millefiorini E, Galgani S, Zaffaroni M, Ghezzi A, Salvetti M, Lus G, Florio C, Totaro R, Granella F, Vianello M, Gatto M, Di Battista G, Aguglia U, Logullo FO, Simone M, Lucisano G, Iaffaldano P, Trojano M. Disease-modifying drugs can reduce disability progression in relapsing multiple sclerosis. Brain 2020; 143:3013-3024. [DOI: 10.1093/brain/awaa251] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/30/2020] [Accepted: 06/29/2020] [Indexed: 11/13/2022] Open
Abstract
Abstract
An ever-expanding number of disease-modifying drugs for multiple sclerosis have become available in recent years, after demonstrating efficacy in clinical trials. In the real-world setting, however, disease-modifying drugs are prescribed in patient populations that differ from those included in pivotal studies, where extreme age patients are usually excluded or under-represented. In this multicentre, observational, retrospective Italian cohort study, we evaluated treatment exposure in three cohorts of patients with relapsing-remitting multiple sclerosis defined by age at onset: paediatric-onset (≤18 years), adult-onset (18–49 years) and late-onset multiple sclerosis (≥50 years). We included patients with a relapsing-remitting phenotype, ≥5 years follow-up, ≥3 Expanded Disability Status Scale (EDSS) evaluations and a first neurological evaluation within 3 years from the first demyelinating event. Multivariate Cox regression models (adjusted hazard ratio with 95% confidence intervals) were used to assess the risk of reaching a first 12-month confirmed disability worsening and the risk of reaching a sustained EDSS of 4.0. The effect of disease-modifying drugs was assessed as quartiles of time exposure. We found that disease-modifying drugs reduced the risk of 12-month confirmed disability worsening, with a progressive risk reduction in different quartiles of exposure in paediatric-onset and adult-onset patients [adjusted hazard ratios in non-exposed versus exposed >62% of the follow-up time: 8.0 (3.5–17.9) for paediatric-onset and 6.3 (4.9–8.0) for adult-onset, P < 0.0001] showing a trend in late-onset patients [adjusted hazard ratio = 1.9 (0.9–4.1), P = 0.07]. These results were confirmed for a sustained EDSS score of 4.0. We also found that relapses were a risk factor for 12-month confirmed disability worsening in all three cohorts, and female sex exerted a protective role in the late-onset cohort. This study provides evidence that sustained exposure to disease-modifying drugs decreases the risk of disability accumulation, seemingly in a dose-dependent manner. It confirms that the effectiveness of disease-modifying drugs is lower in late-onset patients, although still detectable.
Collapse
Affiliation(s)
- Maria Pia Amato
- Department NEUROFARBA, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | | | - Emilio Portaccio
- SOC Neurologia, Ospedale San Giovanni di Dio, AUSL Toscana Centro1, Florence, Italy
| | - Luisa Pastò
- Department NEUROFARBA, University of Florence, Florence, Italy
| | | | - Elio Prestipino
- Department NEUROFARBA, University of Florence, Florence, Italy
| | | | - Laura Tudisco
- Department NEUROFARBA, University of Florence, Florence, Italy
| | | | - Giancarlo Comi
- San Raffaele Hospital - INSPE; Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Patti
- Dipartimento di Scienze Mediche e Chirurgiche e Tecnologie Avanzate, GF Ingrassia, Sez. Neuroscienze, Centro Sclerosi Multipla, University of Catania, Catania, Sicily, Italy
| | - Giovanna De Luca
- Centro Sclerosi Multipla, Clinica Neurologica, Policlinico SS Annunziata, Università ‘G. d'Annunzio’, Chieti-Pescara, Italy
| | - Vincenzo Brescia Morra
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University, Napoli, Italy
| | - Eleonora Cocco
- Centro Sclerosi Multipla, ASSL Cagliari (ATS Sardegna); Dipartimento di Scienze Mediche e Sanità Pubblica, University of Cagliari, Cagliari, Italy
| | - Carlo Pozzilli
- Multiple Sclerosis Center, S. Andrea Hospital, Dept. of Human Neuroscience, Sapienza University, Rome, Italy
| | - Patrizia Sola
- Centro Malattie Demielinizzanti - Dipartimento di Neuroscienze, Azienda Ospedaliero-Universitaria/OCSAE, UO Neurologia, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giuseppe Salemi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Sicily, Italy
| | - Matilde Inglese
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Enrico Millefiorini
- Multiple Sclerosis Center, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Simonetta Galgani
- multiple sclerosis Centre, Department of Neurosciences, S. Camillo - Forlanini Hospital, Rome, Italy
| | - Mauro Zaffaroni
- ASST della Valle Olona, Multiple Sclerosis Center, S. Antonio Abate Hospital of Gallarate, Gallarate, Italy
| | - Angelo Ghezzi
- ASST della Valle Olona, Multiple Sclerosis Center, S. Antonio Abate Hospital of Gallarate, Gallarate, Italy
| | - Marco Salvetti
- Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Centre for Experimental Neurological Therapies, S. Andrea Hospital/Sapienza University, Rome, Italy
- IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Rome, Italy
| | - Giacomo Lus
- Università della Campania Luigi Vanvitelli, Naples, Italy
| | - Ciro Florio
- Multiple Sclerosis Center, Cardarelli Hospital, Naples, Italy
| | - Rocco Totaro
- Demyelinating Diseases Center, Department of Neurology, San Salvatore Hospital, L'Aquila, Italy
| | - Franco Granella
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Italy
| | - Marika Vianello
- Centro Sclerosi Multipla - Ospedale Regionale ‘Ca’ Foncello', Neurology Unit, Treviso, Italy
| | - Maurizia Gatto
- Ospedale Generale Regionale ‘F. Miulli’, Neurology Unit, Acquaviva delle Fonti (BA), Italy
| | | | - Umberto Aguglia
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Neurology Unit, Catanzaro, Italy
| | | | - Marta Simone
- Child Neuropsychiatric Unit, Department of Biomedical Sciences and Human Oncology, University ‘Aldo Moro’ of Bari, Policlinico Piazza G. Cesare, 11, 70121, Bari, Italy
| | - Giuseppe Lucisano
- Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari ‘Aldo Moro’ Policlinico, Bari, Italy
| | - Pietro Iaffaldano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari ‘Aldo Moro’ Policlinico, Bari, Italy
| | - Maria Trojano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari ‘Aldo Moro’ Policlinico, Bari, Italy
| |
Collapse
|
76
|
Abstract
Multiple sclerosis (MS), a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system, is today a leading cause of unpredictable lifelong disability in young adults. The treatment of patients in progressive stages remains highly challenging, alluding to our limited understanding of the underlying pathological processes. In this review, we provide insights into the mechanisms underpinning MS progression from a perspective of epigenetics, that refers to stable and mitotically heritable, yet reversible, changes in the genome activity and gene expression. We first recapitulate findings from epigenetic studies examining the brain tissue of progressive MS patients, which support a contribution of DNA and histone modifications in impaired oligodendrocyte differentiation, defective myelination/remyelination and sustained neuro-axonal vulnerability. We next explore possibilities for identifying factors affecting progression using easily accessible tissues such as blood by comparing epigenetic signatures in peripheral immune cells and brain tissue. Despite minor overlap at individual methylation sites, nearly 30% of altered genes reported in peripheral immune cells of progressive MS patients were found in brain tissue, jointly converging on alterations of neuronal functions. We further speculate about the mechanisms underlying shared epigenetic patterns between blood and brain, which likely imply the influence of internal (genetic control) and/or external (e.g. smoking and ageing) factors imprinting a common signature in both compartments. Overall, we propose that epigenetics might shed light on clinically relevant mechanisms involved in disease progression and open new avenues for the treatment of progressive MS patients in the future.
Collapse
Affiliation(s)
- L Kular
- From the, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - M Jagodic
- From the, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
77
|
Stampanoni Bassi M, Iezzi E, Drulovic J, Pekmezovic T, Gilio L, Furlan R, Finardi A, Marfia GA, Sica F, Centonze D, Buttari F. IL-6 in the Cerebrospinal Fluid Signals Disease Activity in Multiple Sclerosis. Front Cell Neurosci 2020; 14:120. [PMID: 32655367 PMCID: PMC7324533 DOI: 10.3389/fncel.2020.00120] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/14/2020] [Indexed: 11/29/2022] Open
Abstract
Specific proinflammatory and anti-inflammatory molecules could represent useful cerebrospinal fluid (CSF) biomarkers to predict the clinical course of multiple sclerosis (MS). The proinflammatory molecule interleukin (IL)-6 has been investigated in the pathophysiology of MS and has been associated in previous smaller studies to increased disability and disease activity. Here, we wanted to further address IL-6 as a possible CSF biomarker of MS by investigating its detectability in a large cohort of 534 MS patients and in 103 individuals with other non-inflammatory neurological diseases. In these newly diagnosed patients, we also explored correlations between IL-6 detectability, MS phenotypes, and disease characteristics. We found that IL-6 was more frequently detectable in the CSF of MS patients compared with their control counterparts as significant differences emerged between patients with Clinically isolated syndrome (CIS), Relapsing–remitting (RR), and secondary progressive and primary progressive MS compared to non-inflammatory controls. IL-6 was equally present in the CSF of all MS phenotypes. In RR MS patients, IL-6 detectability was found to signal clinically and/or radiologically defined disease activity, among all other clinical characteristics. Our results add further evidence that CSF proinflammatory cytokines could be useful for the identification of those MS patients who are prone to increased disease activity. In particular, IL-6 could represent an interesting prognostic biomarker of MS, as also demonstrated in other diseases where CSF IL-6 was found to identify patients with worse disease severity.
Collapse
Affiliation(s)
| | - Ennio Iezzi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Jelena Drulovic
- Clinic of Neurology, Clinical Center of Serbia, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tatjana Pekmezovic
- Faculty of Medicine, Institute of Epidemiology, University of Belgrade, Belgrade, Serbia
| | - Luana Gilio
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Girolama Alessandra Marfia
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy.,Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Sica
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Diego Centonze
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy.,Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
78
|
Beers DR, Appel SH. Immune dysregulation in amyotrophic lateral sclerosis: mechanisms and emerging therapies. Lancet Neurol 2020; 18:211-220. [PMID: 30663610 DOI: 10.1016/s1474-4422(18)30394-6] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/13/2018] [Accepted: 10/15/2018] [Indexed: 01/04/2023]
Abstract
Neuroinflammation is a common pathological feature of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), and is characterised by activated CNS microglia and astroglia, proinflammatory peripheral lymphocytes, and macrophages. Data from clinical studies show that multiple genetic mutations linked to ALS (eg, mutations in SOD1, TARDBP, and C9orf72) enhance this neuroinflammation, which provides compelling evidence for immune dysregulation in the pathogenesis of ALS. Transgenic rodent models expressing these mutations induce an ALS-like disease with accompanying inflammatory responses, confirming the immune system's involvement in disease progression. Even in the absence of known genetic alterations, immune dysregulation has been shown to lead to dysfunctional regulatory T lymphocytes and increased proinflammatory macrophages in clinical studies. Therefore, an improved understanding of the biological processes that induce this immune dysregulation will help to identify therapeutic strategies that circumvent or ameliorate the pathogenesis of ALS. Emerging cell-based therapies hold the promise of accomplishing this goal and, therefore, improving quality of life and extending survival in patients with ALS.
Collapse
Affiliation(s)
- David R Beers
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Stanley H Appel
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
79
|
Pommerich UM, Nielsen RØ, Overvad K, Dahm CC, Tjønneland A, Olsen A, Dalgas U. Diet quality is not associated with late-onset multiple sclerosis risk– A Danish Cohort Study. Mult Scler Relat Disord 2020; 40:101968. [DOI: 10.1016/j.msard.2020.101968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/22/2020] [Accepted: 01/25/2020] [Indexed: 12/21/2022]
|
80
|
Marrodan M, Bensi C, Pappolla A, Rojas JI, Gaitán MI, Ysrraelit MC, Negrotto L, Fiol MP, Patrucco L, Cristiano E, Farez MF, Correale J. Disease activity impacts disability progression in primary progressive multiple sclerosis. Mult Scler Relat Disord 2020; 39:101892. [PMID: 31846866 DOI: 10.1016/j.msard.2019.101892] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/15/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although solid information on the natural history of primary progressive multiple sclerosis (PPMS) is available, evidence regarding impact of disease activity on PPMS progression remains controversial. OBJECTIVE To describe the clinical characteristics, presence or absence of MRI activity, and natural history of a PPMS cohort from two referral centers in Argentina and assess whether clinical and/or radiological disease activity correlated with disability worsening. METHODS Retrospective study conducted at two MS clinics in Buenos Aires, Argentina, through comparative analysis of patients with and without evidence of disease activity. RESULTS Clinical and/or radiologic activity was presented in 56 (31%) of 178 patients. When stratified by age at onset, we found that for every 10 years of increase in age at onset, risk of reaching EDSS scores of 4 and 6 increased by 26% and 31%, respectively (EDSS 4: HR 1.26, CI 95%: 1.06-1.50; EDSS 6: HR 1.31, CI 95%: 1.06-1.62). Patients who presented clinical exacerbations reached EDSS scores of 6, 7 and 8 faster than those without associated exacerbations (p = 0.009, p = 0.016 and p = 0.001, respectively). Likewise, patients who presented gadolinium-enhancing lesions during the course of disease reached EDSS scores of 7 earlier (p = 0.002). CONCLUSION Older age at onset and presence of clinical and/or radiological disease activity correlated with accelerated disability progression in this cohort of PPMS patients.
Collapse
Affiliation(s)
- M Marrodan
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - C Bensi
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - A Pappolla
- Department of Neurology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - J I Rojas
- Department of Neurology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina; Centro de Esclerosis Múltiple de Buenos Aires, Buenos Aires, Argentina
| | - M I Gaitán
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - M C Ysrraelit
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - L Negrotto
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - M P Fiol
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - L Patrucco
- Department of Neurology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina; Centro de Esclerosis Múltiple de Buenos Aires, Buenos Aires, Argentina
| | - E Cristiano
- Centro de Esclerosis Múltiple de Buenos Aires, Buenos Aires, Argentina
| | - M F Farez
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina; Center for Biostatistics, Epidemiology and Public Health (CEBES). Fleni, Buenos Aires, Argentina
| | - J Correale
- Department of Neurology, Fleni, Montañeses 2325 (1428), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
81
|
Vaughn CB, Jakimovski D, Kavak KS, Ramanathan M, Benedict RHB, Zivadinov R, Weinstock-Guttman B. Epidemiology and treatment of multiple sclerosis in elderly populations. Nat Rev Neurol 2020; 15:329-342. [PMID: 31000816 DOI: 10.1038/s41582-019-0183-3] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The prevalence of multiple sclerosis (MS) and the age of affected patients are increasing owing to increased longevity of the general population and the availability of effective disease-modifying therapies. However, ageing presents unique challenges in patients with MS largely as a result of their increased frequency of age-related and MS-related comorbidities as well as transition of the disease course from an inflammatory to a neurodegenerative phenotype. Immunosenescence (the weakening of the immune system associated with natural ageing) might be at least partly responsible for this transition, which further complicates disease management. Currently approved therapies for MS are effective in preventing relapse but are not as effective in preventing the accumulation of disability associated with ageing and disease progression. Thus, ageing patients with MS represent a uniquely challenging population that is currently underserved by existing therapeutic regimens. This Review focuses on the epidemiology of MS in ageing patients. Unique considerations relevant to this population are discussed, including the immunology and pathobiology of the complex relationship between ageing and MS, the safety and efficacy of disease-modifying therapies, when discontinuation of treatment might be appropriate and the important role of approaches to support wellness and cognition.
Collapse
Affiliation(s)
- Caila B Vaughn
- Jacobs Multiple Sclerosis Center for Treatment and Research, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY), Buffalo, NY, USA
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY), Buffalo, NY, USA
| | - Katelyn S Kavak
- Jacobs Multiple Sclerosis Center for Treatment and Research, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY), Buffalo, NY, USA
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY), Buffalo, NY, USA
| | - Ralph H B Benedict
- Jacobs Multiple Sclerosis Center for Treatment and Research, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY), Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY), Buffalo, NY, USA.,Center for Biomedical Imaging at the Clinical Translational Science Institute, State University of New York (SUNY), Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Jacobs Multiple Sclerosis Center for Treatment and Research, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY), Buffalo, NY, USA.
| |
Collapse
|
82
|
The effect of galanin gene polymorphism rs948854 on the severity of multiple sclerosis: A significant association with the age of onset. Mult Scler Relat Disord 2020; 37:101439. [DOI: 10.1016/j.msard.2019.101439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/04/2019] [Accepted: 10/08/2019] [Indexed: 01/15/2023]
|
83
|
Dunn-Pirio AM, Heyman BM, Kaufman DS, Kinkel RP. Outcomes and Cost-Effectiveness of Autologous Hematopoietic Cell Transplant for Multiple Sclerosis. Curr Treat Options Neurol 2019; 21:53. [PMID: 31624926 DOI: 10.1007/s11940-019-0588-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW This review presents a critical appraisal of the use of autologous hematopoietic cell transplant (AHCT) for the treatment of multiple sclerosis. We present the reader with a brief review on the AHCT procedure, its immunomodulatory mechanism of action in MS, the most recent evidence in support of its use in patients with relapsing-remitting multiple sclerosis (RRMS), as well as its cost considerations. RECENT FINDINGS The first meta-analysis of clinical trials of AHCT for patients with MS demonstrated durable 5-year progression-free survival rates and low treatment-related mortality. Recently, the first randomized controlled phase III clinical trial demonstrated AHCT to be superior to best available therapy for a subset of patients with RRMS. This led to the American society for transplant and cellular therapies (ASTCT) to recommend AHCT "for patients with relapsing forms of MS who have prognostic factors that indicate a high risk of future disability." AHCT should be considered for patients with RRMS with evidence of clinical activity who have failed 2 lines of therapy or at least one highly active disease-modifying therapy.
Collapse
Affiliation(s)
- Anastasie M Dunn-Pirio
- Division of Neuroimmunology, Department of Neurosciences, UC San Diego, La Jolla, CA, 92093, USA
| | - Benjamin M Heyman
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, UC San Diego, MC 0695, La Jolla, CA, 92093, USA.
| | - Dan S Kaufman
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, UC San Diego, MC 0695, La Jolla, CA, 92093, USA
| | - Revere P Kinkel
- Division of Neuroimmunology, Department of Neurosciences, UC San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
84
|
Krysko KM, Henry RG, Cree BAC, Lin J, Caillier S, Santaniello A, Zhao C, Gomez R, Bevan C, Smith DL, Stern W, Kirkish G, Hauser SL, Oksenberg JR, Graves JS. Telomere Length Is Associated with Disability Progression in Multiple Sclerosis. Ann Neurol 2019; 86:671-682. [PMID: 31486104 DOI: 10.1002/ana.25592] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/23/2019] [Accepted: 09/01/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To assess whether biological aging as measured by leukocyte telomere length (LTL) is associated with clinical disability and brain volume loss in multiple sclerosis (MS). METHODS Adults with MS/clinically isolated syndrome in the University of California, San Francisco EPIC cohort study were included. LTL was measured on DNA samples by quantitative polymerase chain reaction and expressed as telomere to somatic DNA (T/S) ratio. Expanded Disability Status Scale (EDSS) and 3-dimensional T1-weighted brain magnetic resonance imaging were performed at baseline and follow-up. Associations of baseline LTL with cross-sectional and longitudinal outcomes were assessed using simple and mixed effects linear regression models. A subset (n = 46) had LTL measured over time, and we assessed the association of LTL change with EDSS change with mixed effects models. RESULTS Included were 356 women and 160 men (mean age = 43 years, median disease duration = 6 years, median EDSS = 1.5 [range = 0-7], mean T/S ratio = 0.97 [standard deviation = 0.18]). In baseline analyses adjusted for age, disease duration, and sex, for every 0.2 lower LTL, EDSS was 0.27 higher (95% confidence interval [CI] = 0.13-0.42, p < 0.001) and brain volume was 7.4mm3 lower (95% CI = 0.10-14.7, p = 0.047). In longitudinal adjusted analyses, those with lower baseline LTL had higher EDSS and lower brain volumes over time. In adjusted analysis of the subset, LTL change was associated with EDSS change over 10 years; for every 0.2 LTL decrease, EDSS was 0.34 higher (95% CI = 0.08-0.61, p = 0.012). INTERPRETATION Shorter telomere length was associated with disability independent of chronological age, suggesting that biological aging may contribute to neurological injury in MS. Targeting aging-related mechanisms is a potential therapeutic strategy against MS progression. ANN NEUROL 2019;86:671-682.
Collapse
Affiliation(s)
- Kristen M Krysko
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Roland G Henry
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Bruce A C Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Jue Lin
- Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | -
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Stacy Caillier
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Adam Santaniello
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Chao Zhao
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Refujia Gomez
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Carolyn Bevan
- Department of Neurology, Northwestern University, Evanston, IL
| | - Dana L Smith
- Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | - William Stern
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Gina Kirkish
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Stephen L Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Jorge R Oksenberg
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Jennifer S Graves
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA.,Department of Neurosciences, University of California, San Diego, San Diego, CA
| |
Collapse
|
85
|
Giacco V, Panattoni G, Medelin M, Bonechi E, Aldinucci A, Ballerini C, Ballerini L. Cytokine inflammatory threat, but not LPS one, shortens GABAergic synaptic currents in the mouse spinal cord organotypic cultures. J Neuroinflammation 2019; 16:127. [PMID: 31238967 PMCID: PMC6593520 DOI: 10.1186/s12974-019-1519-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/11/2019] [Indexed: 01/02/2023] Open
Abstract
Background Synaptic dysfunction, named synaptopathy, due to inflammatory status of the central nervous system (CNS) is a recognized factor potentially underlying both motor and cognitive dysfunctions in neurodegenerative diseases. To gain knowledge on the mechanistic interplay between local inflammation and synapse changes, we compared two diverse inflammatory paradigms, a cytokine cocktail (CKs; IL-1β, TNF-α, and GM-CSF) and LPS, and their ability to tune GABAergic current duration in spinal cord cultured circuits. Methods We exploit spinal organotypic cultures, single-cell electrophysiology, immunocytochemistry, and confocal microscopy to explore synaptic currents and resident neuroglia reactivity upon CK or LPS incubation. Results Local inflammation in slice cultures induced by CK or LPS stimulations boosts network activity; however, only CKs specifically reduced GABAergic current duration. We pharmacologically investigated the contribution of GABAAR α-subunits and suggested that a switch of GABAAR α1-subunit might have induced faster GABAAR decay time, weakening the inhibitory transmission. Conclusions Lower GABAergic current duration could contribute to providing an aberrant excitatory transmission critical for pre-motor circuit tasks and represent a specific feature of a CK cocktail able to mimic an inflammatory reaction that spreads in the CNS. Our results describe a selective mechanism that could be triggered during specific inflammatory stress. Electronic supplementary material The online version of this article (10.1186/s12974-019-1519-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vincenzo Giacco
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.,Present address: Wolfson Centre for Age Related Disease, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Giulia Panattoni
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy
| | - Manuela Medelin
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Elena Bonechi
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy
| | | | - Clara Ballerini
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy.
| | - Laura Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
| |
Collapse
|
86
|
Tangestani Fard M, Stough C. A Review and Hypothesized Model of the Mechanisms That Underpin the Relationship Between Inflammation and Cognition in the Elderly. Front Aging Neurosci 2019; 11:56. [PMID: 30930767 PMCID: PMC6425084 DOI: 10.3389/fnagi.2019.00056] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/26/2019] [Indexed: 12/13/2022] Open
Abstract
Age is associated with increased risk for several disorders including dementias, cardiovascular disease, atherosclerosis, obesity, and diabetes. Age is also associated with cognitive decline particularly in cognitive domains associated with memory and processing speed. With increasing life expectancies in many countries, the number of people experiencing age-associated cognitive impairment is increasing and therefore from both economic and social terms the amelioration or slowing of cognitive aging is an important target for future research. However, the biological causes of age associated cognitive decline are not yet, well understood. In the current review, we outline the role of inflammation in cognitive aging and describe the role of several inflammatory processes, including inflamm-aging, vascular inflammation, and neuroinflammation which have both direct effect on brain function and indirect effects on brain function via changes in cardiovascular function.
Collapse
Affiliation(s)
| | - Con Stough
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|