1
|
Terracina S, Ferraguti G, Tarani L, Fanfarillo F, Tirassa P, Ralli M, Iannella G, Polimeni A, Lucarelli M, Greco A, Fiore M. Nerve Growth Factor and Autoimmune Diseases. Curr Issues Mol Biol 2023; 45:8950-8973. [PMID: 37998739 PMCID: PMC10670231 DOI: 10.3390/cimb45110562] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
NGF plays a crucial immunomodulatory role and increased levels are found in numerous tissues during autoimmune states. NGF directly modulates innate and adaptive immune responses of B and T cells and causes the release of neuropeptides and neurotransmitters controlling the immune system activation in inflamed tissues. Evidence suggests that NGF is involved in the pathogenesis of numerous immune diseases including autoimmune thyroiditis, chronic arthritis, multiple sclerosis, systemic lupus erythematosus, mastocytosis, and chronic granulomatous disease. Furthermore, as NGF levels have been linked to disease severity, it could be considered an optimal early biomarker to identify therapeutic approach efficacy. In conclusion, by gaining insights into how these molecules function and which cells they interact with, future studies can devise targeted therapies to address various neurological, immunological, and other disorders more effectively. This knowledge may pave the way for innovative treatments based on NGF manipulation aimed at improving the quality of life for individuals affected by diseases involving neurotrophins.
Collapse
Affiliation(s)
- Sergio Terracina
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesca Fanfarillo
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Paola Tirassa
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Massimo Ralli
- Department of Sensory Organs, Sapienza University of Rome, 00185 Roma, Italy
| | - Giannicola Iannella
- Department of Sensory Organs, Sapienza University of Rome, 00185 Roma, Italy
| | - Antonella Polimeni
- Department of Odontostomatological and Maxillofacial Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, 00185 Rome, Italy
| | - Antonio Greco
- Department of Sensory Organs, Sapienza University of Rome, 00185 Roma, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
2
|
Kania K, Ambrosius W, Kozubski W, Kalinowska-Łyszczarz A. The impact of disease modifying therapies on cognitive functions typically impaired in multiple sclerosis patients: a clinician's review. Front Neurol 2023; 14:1222574. [PMID: 37503514 PMCID: PMC10368887 DOI: 10.3389/fneur.2023.1222574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Objective Over the last few decades clinicians have become aware that cognitive impairment might be a major cause of disability, loss of employment and poor quality of life in patients suffering from multiple sclerosis [MS].The impact of disease modifying therapies [DMTs] on cognition is still a matter of debate. Theoretically, DMTs could exert a substantial beneficial effect by means of reducing neuroinflammation and brain atrophy, which are established correlates of cognitive dysfunction. The aim of the study was to review the evidence concerning the effect of DMTs on cognitive functions. Methods PubMed, Scopus, and the European Committee for Treatment and Research in Multiple Sclerosis [ECTRIMS] Library were searched for articles concerning the pediatric and adult populations of patients with multiple sclerosis, including clinical trials and RWD, where psychometric results were analyzed as secondary or exploratory endpoints. Results We reviewed a total of 44 studies that were found by our search strategy, analyzed the psychological tests that were applied, the length of the follow-up, and possible limitations. We pointed out the difficulties associated with assessing of DMTs' effects on cognitive functions, and pitfalls in cognitive tools used for evaluating of MS patients. Conclusion There is a need to highlight this aspect of MS therapies, and to collect adequate data to make informed therapeutic decisions, to improve our understanding of MS-related cognitive dysfunction and provide new therapeutic targets.
Collapse
Affiliation(s)
- Karolina Kania
- Department of Neurology, Poznan University of Medical Sciences, Poznań, Poland
| | - Wojciech Ambrosius
- Department of Neurology, Poznan University of Medical Sciences, Poznań, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, Poznań, Poland
| | - Alicja Kalinowska-Łyszczarz
- Department of Neurology, Division of Neurochemistry and Neuropathology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
3
|
Walsh J, Palandra J, Goihberg E, Deng S, Hurst S, Neubert H. Analysis of β-nerve growth factor and its precursor during human pregnancy by immunoaffinity-liquid chromatography tandem mass spectrometry. Sci Rep 2023; 13:9180. [PMID: 37280257 DOI: 10.1038/s41598-023-34695-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/05/2023] [Indexed: 06/08/2023] Open
Abstract
β-Nerve growth factor (NGF) is a neurotrophin that plays a critical role in fetal development during gestation. ProNGF is the precursor form of NGF with a distinct biological profile. In order to investigate the role of NGF and proNGF in pregnant human females, a sensitive and selective immunoaffinity liquid chromatography-tandem mass spectrometry assay was developed and qualified to simultaneously measure the levels of total NGF (tNGF; sum of mature and proNGF) and proNGF using full and relative quantification strategies, respectively. The assay was used to determine serum tNGF and proNGF levels in the three gestational trimesters of pregnancy and in non-pregnant female controls. Mean tNGF ± SD were 44.6 ± 12.3, 42.6 ± 9.3, 65.4 ± 17.6 and 77.0 ± 17.8 pg/mL for non-pregnant, first, second, and third trimesters, respectively, demonstrating no significant increase in circulating tNGF between the control and the first trimester, and a moderate yet significant 1.7-fold increase through gestation. proNGF levels during the first trimester were unchanged compared to control. In contrast to tNGF, however, proNGF levels during gestation remained stable without significant changes. The development of this sensitive, novel immunoaffinity duplexed assay for both tNGF and proNGF is expected to enable further elucidation of the roles these neurotrophins play in human pregnancy as well as other models.
Collapse
Affiliation(s)
- Jason Walsh
- Pfizer Inc., 1 Burtt Road, Andover, MA, 01810, USA.
| | - Joe Palandra
- Pfizer Inc., 1 Burtt Road, Andover, MA, 01810, USA
| | | | - Shibing Deng
- Pfizer Inc., 10777 Science Center Drive, San Diego, CA, 92121, USA
| | - Susan Hurst
- Pfizer Inc., 445 Eastern Point Road, Groton, CT, 06340, USA
| | | |
Collapse
|
4
|
Intranasal interferon-beta alleviates anxiety and depressive-like behaviors by modulating microglia polarization in an Alzheimer's disease model. Neurosci Lett 2023; 792:136968. [PMID: 36396023 DOI: 10.1016/j.neulet.2022.136968] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) patients frequently experience neuropsychiatric symptoms (NPS), which are linked to a lower quality of life and a faster rate of disease progression. A growing body of research indicates that several microglial phenotypes control the inflammatory response and are crucial in the pathophysiology of AD-related NPS. Given the crucial role played by inflammatory mediators produced by microglia in developing of NPS, interferon-beta (IFNβ), a cytokine with anti-inflammatory capabilities, maybe a successful treatment for NPS caused by AD. In this investigation, using a rat model of AD, we examined the impact of intranasal treatment of IFNβ on anxious/depressive-like behavior and microglial M1/M2 polarization. The rat hippocampus was bilaterally injected with lentiviruses harboring mutant human amyloid precursor protein. Rats were given recombinant IFNβ1a (68,000 IU/rat) via the intranasal route, starting on day 23 following viral infection and continuing until day 49. On days 47-49, the elevated plus maze, forced swim, and tail suspension tests were applied to measure anxiety- and depressive-like behavior. Additionally, qPCR was utilized to quantify the expression of M1 markers (CD68, CD86, and CD40) and M2 markers (Ym1, CD206, Arg1, GDNF, BDNF, and SOCS1). Our findings demonstrated that decreased M2 marker expression is accompanied by anxious/depressive-like behavior when the mutant human APP gene is overexpressed in the hippocampus. In the rat model of AD, IFNβ therapy reduces anxious/depressive-like behaviors, at least in part by polarizing microglia towards M2. Therefore, IFNβ may be a viable therapeutic drug for reducing NPS in the context of AD.
Collapse
|
5
|
Axonal Regeneration: Underlying Molecular Mechanisms and Potential Therapeutic Targets. Biomedicines 2022; 10:biomedicines10123186. [PMID: 36551942 PMCID: PMC9775075 DOI: 10.3390/biomedicines10123186] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Axons in the peripheral nervous system have the ability to repair themselves after damage, whereas axons in the central nervous system are unable to do so. A common and important characteristic of damage to the spinal cord, brain, and peripheral nerves is the disruption of axonal regrowth. Interestingly, intrinsic growth factors play a significant role in the axonal regeneration of injured nerves. Various factors such as proteomic profile, microtubule stability, ribosomal location, and signalling pathways mark a line between the central and peripheral axons' capacity for self-renewal. Unfortunately, glial scar development, myelin-associated inhibitor molecules, lack of neurotrophic factors, and inflammatory reactions are among the factors that restrict axonal regeneration. Molecular pathways such as cAMP, MAPK, JAK/STAT, ATF3/CREB, BMP/SMAD, AKT/mTORC1/p70S6K, PI3K/AKT, GSK-3β/CLASP, BDNF/Trk, Ras/ERK, integrin/FAK, RhoA/ROCK/LIMK, and POSTN/integrin are activated after nerve injury and are considered significant players in axonal regeneration. In addition to the aforementioned pathways, growth factors, microRNAs, and astrocytes are also commendable participants in regeneration. In this review, we discuss the detailed mechanism of each pathway along with key players that can be potentially valuable targets to help achieve quick axonal healing. We also identify the prospective targets that could help close knowledge gaps in the molecular pathways underlying regeneration and shed light on the creation of more powerful strategies to encourage axonal regeneration after nervous system injury.
Collapse
|
6
|
Intranasal interferon-beta as a promising alternative for the treatment of Alzheimer's disease. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
7
|
Tan PH, Ji J, Hsing CH, Tan R, Ji RR. Emerging Roles of Type-I Interferons in Neuroinflammation, Neurological Diseases, and Long-Haul COVID. Int J Mol Sci 2022; 23:ijms232214394. [PMID: 36430870 PMCID: PMC9696119 DOI: 10.3390/ijms232214394] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Interferons (IFNs) are pleiotropic cytokines originally identified for their antiviral activity. IFN-α and IFN-β are both type I IFNs that have been used to treat neurological diseases such as multiple sclerosis. Microglia, astrocytes, as well as neurons in the central and peripheral nervous systems, including spinal cord neurons and dorsal root ganglion neurons, express type I IFN receptors (IFNARs). Type I IFNs play an active role in regulating cognition, aging, depression, and neurodegenerative diseases. Notably, by suppressing neuronal activity and synaptic transmission, IFN-α and IFN-β produced potent analgesia. In this article, we discuss the role of type I IFNs in cognition, neurodegenerative diseases, and pain with a focus on neuroinflammation and neuro-glial interactions and their effects on cognition, neurodegenerative diseases, and pain. The role of type I IFNs in long-haul COVID-associated neurological disorders is also discussed. Insights into type I IFN signaling in neurons and non-neuronal cells will improve our treatments of neurological disorders in various disease conditions.
Collapse
Affiliation(s)
- Ping-Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Jasmine Ji
- Neuroscience Department, Wellesley College, Wellesley, MA 02482, USA
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Radika Tan
- Kaohsiung American School, Kaohsiung 81354, Taiwan
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
8
|
Raftopoulou S, Rapti A, Karathanasis D, Evangelopoulos ME, Mavragani CP. The role of type I IFN in autoimmune and autoinflammatory diseases with CNS involvement. Front Neurol 2022; 13:1026449. [PMID: 36438941 PMCID: PMC9685560 DOI: 10.3389/fneur.2022.1026449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Type I interferons (IFNs) are major mediators of innate immunity, with well-known antiviral, antiproliferative, and immunomodulatory properties. A growing body of evidence suggests the involvement of type I IFNs in the pathogenesis of central nervous system (CNS) manifestations in the setting of chronic autoimmune and autoinflammatory disorders, while IFN-β has been for years, a well-established therapeutic modality for multiple sclerosis (MS). In the present review, we summarize the current evidence on the mechanisms of type I IFN production by CNS cellular populations as well as its local effects on the CNS. Additionally, the beneficial effects of IFN-β in the pathophysiology of MS are discussed, along with the contributory role of type I IFNs in the pathogenesis of neuropsychiatric lupus erythematosus and type I interferonopathies.
Collapse
Affiliation(s)
- Sylvia Raftopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Rapti
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Karathanasis
- First Department of Neurology, National and Kapodistrian University of Athens, Aeginition Hospital, Athens, Greece
| | | | - Clio P. Mavragani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
9
|
Azarafrouz F, Farhangian M, Chavoshinezhad S, Dargahi S, Nassiri-Asl M, Dargahi L. Interferon beta attenuates recognition memory impairment and improves brain glucose uptake in a rat model of Alzheimer's disease: Involvement of mitochondrial biogenesis and PI3K pathway. Neuropeptides 2022; 95:102262. [PMID: 35709657 DOI: 10.1016/j.npep.2022.102262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/10/2022] [Accepted: 05/30/2022] [Indexed: 11/15/2022]
Abstract
Interferon beta (IFNβ) is a cytokine with anti-apoptotic and anti-inflammatory properties, and its beneficial effects on Alzheimer's disease (AD) have been recently shown. The alterations in cerebral glucose uptake are closely linked to memory deficit and AD progression. The current study was designed to determine if IFNβ can improve recognition memory and brain glucose uptake in a rat model of AD. The lentiviruses expressing mutant human amyloid precursor protein were injected bilaterally to the rat hippocampus. From day 23 after virus injection, rats were intranasally treated with recombinant IFNβ protein (68,000 IU/rat) every other day until day 50. Recognition memory performance was evaluated by novel object recognition test on days 46-49. The 18F-2- fluoro-deoxy-d-glucose positron emission tomography (18F-FDG-PET) was used to determine changes in brain glucose metabolism on day 50. The expression of the PI3K/Akt pathway components, neurotrophins and mitochondrial biogenesis factors were also measured by qPCR in the hippocampus. Our results showed that IFNβ treatment improves recognition memory performance in parallel with increased glucose uptake and neuronal survival in the hippocampus of the AD rats. The neuroprotective effect of IFNβ could be attributed, at least partly, to activation of PI3K-Akt-mTOR signaling pathway, increased expression of NGF, and mitochondrial biogenesis. Taken together, our findings suggest the therapeutic potential of IFNβ for AD.
Collapse
Affiliation(s)
- Forouzan Azarafrouz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Farhangian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Chavoshinezhad
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Saina Dargahi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Nassiri-Asl
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Type I IFN signaling in T regulatory cells modulates chemokine production and myeloid derived suppressor cells trafficking during EAE. J Autoimmun 2020; 115:102525. [PMID: 32709481 DOI: 10.1016/j.jaut.2020.102525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 01/18/2023]
Abstract
Interferon-β has therapeutic efficacy in Multiple Sclerosis by reducing disease exacerbations and delaying relapses. Previous studies have suggested that the effects of type I IFN in Experimental Autoimmune Encephalomyelitis (EAE) in mice were targeted to myeloid cells. We used mice with a conditional deletion (cKO) of the type I IFN receptor (IFNAR) in T regulatory (Treg) cells to dissect the role of IFN signaling on Tregs. cKO mice developed severe EAE with an earlier onset than control mice. Although Treg cells from cKO mice were more activated, the activation status and effector cytokine production of CD4+Foxp3- T cells in the draining lymph nodes (dLN) was similar in WT and cKO mice during the priming phase. Production of chemokines (CCL8, CCL9, CCL22) by CD4+Foxp3- T cells and LN resident cells from cKO mice was suppressed. Suppression of chemokine production was accompanied by a substantial reduction of myeloid derived suppressor cells (MDSCs) in the dLN of cKO mice, while generation of MDSCs and recruitment to peripheral organs was comparable. This study demonstrates that signaling by type I IFNs in Tregs reduces their capacity to suppress chemokine production, with resultant alteration of the entire microenvironment of draining lymph nodes leading to enhancement of MDSC homing, and beneficial effects on disease outcome.
Collapse
|
11
|
Chavoshinezhad S, Mohseni Kouchesfahani H, Ahmadiani A, Dargahi L. Interferon beta ameliorates cognitive dysfunction in a rat model of Alzheimer's disease: Modulation of hippocampal neurogenesis and apoptosis as underlying mechanism. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109661. [PMID: 31152860 DOI: 10.1016/j.pnpbp.2019.109661] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/08/2019] [Accepted: 05/28/2019] [Indexed: 12/21/2022]
Abstract
Neuronal apoptosis and impaired hippocampal neurogenesis are major players in cognitive/memory dysfunctions including Alzheimer's disease (AD). Interferon beta (IFNβ) is a cytokine with anti-apoptotic and neuroprotective properties on the central nervous system (CNS) cells which specifically affects neural progenitor cells (NPCs) even in the adult brain. In this study, we examined the effect of IFNβ on memory impairment as well as hippocampal neurogenesis and apoptosis in a rat model of AD. AD model was induced by lentiviral-mediated overexpression of mutant APP in the hippocampus of adult rats. Intranasal (IN) administration of IFNβ (0.5 μg/kg and 1 μg/kg doses) was started from day 23 after virus injection and continued every other day to the final day of experiments. The expression levels of APP, neurogenesis (Nestin, Ki67, DCX, and Reelin) and apoptosis (Bax/Bcl-2 ratio, cleaved-caspase-3 and seladin-1) markers were evaluated by immunohistochemistry, real-time PCR, immunofluorescence and western blotting. Moreover, thioflavin T and Nissl stainings were used to assess Aβ plaque levels and neuronal degeneration in the hippocampus, respectively. Our results showed that IFNβ treatment reduced APP expression and Aβ plaque formation, and concomitantly ameliorated spatial learning and memory deficits examined in Y-maze and Morris water maze tests. Moreover, in parallel with reducing apoptosis and neural loss in the hippocampal subfields, IFNβ decreased ectopic neurogenesis in the CA1 and CA3 regions of the AD rat hippocampus. However, IFNβ increased neurogenesis in the dentate gyrus neurogenic niche. Our findings suggest that IFNβ exerts neuroprotective effects at least partly by inhibition of apoptosis and modulation of neurogenesis. Taken together, IFNβ can be a promising therapeutic approach to improve cognitive performance in AD-like neurodegenerative context.
Collapse
Affiliation(s)
- Sara Chavoshinezhad
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Tsareva EY, Favorova OO, Boyko AN, Kulakova OG. Genetic Markers for Personalized Therapy of Polygenic Diseases: Pharmacogenetics of Multiple Sclerosis. Mol Biol 2019. [DOI: 10.1134/s0026893319040149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Chavoshinezhad S, Mohseni Kouchesfahani H, Salehi MS, Pandamooz S, Ahmadiani A, Dargahi L. Intranasal interferon beta improves memory and modulates inflammatory responses in a mutant APP-overexpressing rat model of Alzheimer’s disease. Brain Res Bull 2019; 150:297-306. [DOI: 10.1016/j.brainresbull.2019.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 06/01/2019] [Accepted: 06/19/2019] [Indexed: 02/09/2023]
|
14
|
Fodelianaki G, Lansing F, Bhattarai P, Troullinaki M, Zeballos MA, Charalampopoulos I, Gravanis A, Mirtschink P, Chavakis T, Alexaki VI. Nerve Growth Factor modulates LPS - induced microglial glycolysis and inflammatory responses. Exp Cell Res 2019; 377:10-16. [PMID: 30817930 DOI: 10.1016/j.yexcr.2019.02.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/22/2019] [Accepted: 02/24/2019] [Indexed: 01/09/2023]
Abstract
Microglia, the parenchymal immune cells of the central nervous system, orchestrate neuroinflammation in response to infection or damage, and promote tissue repair. However, aberrant microglial responses are integral to neurodegenerative diseases and critically contribute to disease progression. Thus, it is important to elucidate how microglia - mediated neuroinflammation is regulated by endogenous factors. Here, we explored the effect of Nerve Growth Factor (NGF), an abundant neurotrophin, on microglial inflammatory responses. NGF, via its high affinity receptor TrkA, downregulated LPS - induced production of pro-inflammatory cytokines and NO in primary mouse microglia and inhibited TLR4 - mediated activation of the NF-κB and JNK pathways. Furthermore, NGF attenuated the LPS - enhanced glycolytic activity in microglia, as suggested by reduced glucose uptake and decreased expression of the glycolytic enzymes Pfkβ3 and Ldhα. Consistently, 2DG - mediated glycolysis inhibition strongly downregulated LPS - induced cytokine production in microglial cells. Our findings demonstrate that NGF attenuates pro-inflammatory responses in microglia and may thereby contribute to regulation of microglia - mediated neuroinflammation.
Collapse
Affiliation(s)
- Georgia Fodelianaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Felix Lansing
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Prabesh Bhattarai
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Maria Troullinaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Maria Alejandra Zeballos
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | | | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
15
|
Abstract
Infection with Human Immunodeficiency Virus (HIV)-1 continues to cause HIV-associated neurocognitive disorders despite combined antiretroviral therapy. Interferons (IFNs) are important for any antiviral immune response, but the lasting production of IFNα causes exhaustive activation leading eventually to progression to AIDS. Expression of IFNα in the HIV-exposed central nervous system has been linked to cognitive impairment and inflammatory neuropathology. In contrast, IFNβ exerts anti-inflammatory effects, appears to control, at least temporarily, lentiviral infection in the brain and provides neuroprotection. The dichotomy of type I IFN effects on HIV-1 infection and the associated brain injury will be discussed in this review, because the underlying mechanisms require further investigation to allow harnessing these innate immune factors for therapeutic purposes.
Collapse
Affiliation(s)
- Victoria E Thaney
- 1 Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute , La Jolla, California
| | - Marcus Kaul
- 1 Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute , La Jolla, California.,2 Division of Biomedical Sciences, School of Medicine, University of California , Riverside, Riverside, California
| |
Collapse
|
16
|
Kalinowska-Łyszczarz A, Pawlak MA, Wyciszkiewicz A, Osztynowicz K, Kozubski W, Michalak S. Immune-cell BDNF expression in treatment-naïve relapsing-remitting multiple sclerosis patients and following one year of immunomodulation therapy. Neurol Neurochir Pol 2018; 52:483-489. [PMID: 29643001 DOI: 10.1016/j.pjnns.2018.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/20/2018] [Indexed: 11/30/2022]
Abstract
Although neurons are the main source of neurotrophins in the healthy brain, neurotrophins can also be expressed in the immune system. We have previously shown that in relapsing-remitting multiple sclerosis (RRMS) lower immune-cell neurotrophin levels are associated with brain atrophy and cognitive impairment. The aim of the present study was to assess if immune-cell neurotrophin expression is impaired in MS as compared with the healthy controls, and to describe if these levels change in treatment-naïve RRMS patients, following one year of immunomodulation. Fifty treatment-naïve RRMS patients were assessed at baseline and after one year of immunomodulation (beta-interferons/glatiramer acetate). The control group included 39 healthy subjects matched according to age and gender. Peripheral blood mononuclear cells (PBMCs) were isolated from heparinized blood using Ficoll-Histopaque gradient. The levels of brain-derived-neurotrophic-factor (BDNF), beta-nerve-growth-factor (beta-NGF), neurotrophin-3 (NT-3) and neurotrophin-4/5 (NT-4/5) were measured in PBMC lysates with ELISA. BDNF levels were significantly lower in MS than in the healthy controls (median 613 vs. 1657pg/mg protein, p<0.001). After one year of immunomodulation, BDNF expression did not change significantly (p=0.06) on the group level. In 70% of patients there was no increase in BDNF level, and in 30% it increased. We observed no differences between treatment groups. Other neurotrophins were detected in a minority of MS samples (as opposed to the controls). To conclude, we have shown that immune-cell production of neurotrophins is impaired in MS patients. In our MS cohort standard immunomodulation failed to restore normal BDNF levels in PBMCs within one year of therapy.
Collapse
Affiliation(s)
- Alicja Kalinowska-Łyszczarz
- Division of Neurochemistry and Neuropathology, Department of Neurology, Poznan University of Medical Sciences (PUMS), 49 Przybyszewskiego Street, 60-355 Poznan, Poland.
| | - Mikołaj A Pawlak
- Department of Neurology and Cerebrovascular Disorders, Poznan University of Medical Sciences, 34 Dojazd Street, 60-631 Poznan, Poland.
| | - Aleksandra Wyciszkiewicz
- Division of Neurochemistry and Neuropathology, Department of Neurology, Poznan University of Medical Sciences (PUMS), 49 Przybyszewskiego Street, 60-355 Poznan, Poland.
| | - Krystyna Osztynowicz
- Division of Neurochemistry and Neuropathology, Department of Neurology, Poznan University of Medical Sciences (PUMS), 49 Przybyszewskiego Street, 60-355 Poznan, Poland.
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, 49 Przybyszewskiego Street, 60-355 Poznan, Poland.
| | - Sławomir Michalak
- Division of Neurochemistry and Neuropathology, Department of Neurology, Poznan University of Medical Sciences (PUMS), 49 Przybyszewskiego Street, 60-355 Poznan, Poland.
| |
Collapse
|
17
|
Sayad A, Salmani T, Hemmesi MK, Ganji M, Ghafouri-Fard S, Hatami M, Soudyab M, Taheri M. Down-regulation of RORA gene expression in the blood of multiple sclerosis patients. Hum Antibodies 2018; 26:219-224. [PMID: 29889063 DOI: 10.3233/hab-180341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by recurrent episodes of demyelination and loss of oligodendrocytes. The demyelination process is caused by various subsets of CD4+ T cells with a Th1 and Th17 phenotype. The retinoid acid-related orphan receptor A (RORA) is expressed in Th17 cells and promote Th17 differentiation. In this study, we compared the expression level of RORA gene in the blood of 50 relapsing-remitting MS (RRMS) patients who were treated with IFN-β and 50 healthy controls by TaqMan Quantitative Real-Time PCR.We found that RORA expression was significantly down-regulated in MS patients compared with controls (P= 0.006). However, there was no significant correlation between RORA gene expression and Kurtzke Expanded Disability Status Scale (EDSS). Our findings suggest a possible contribution of IFN-β in the downregulation of RORA. In addition, RORA downregulation may be a potential indicator of positive response to interferon beta treatment of multiple sclerosis patients.
Collapse
Affiliation(s)
- Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebali Salmani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Maziar Ganji
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Hatami
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Soudyab
- Department of Medical Genetics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Taheri
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Furber KL, Van Agten M, Evans C, Haddadi A, Doucette JR, Nazarali AJ. Advances in the treatment of relapsing-remitting multiple sclerosis: the role of pegylated interferon β-1a. Degener Neurol Neuromuscul Dis 2017; 7:47-60. [PMID: 30050377 PMCID: PMC6053102 DOI: 10.2147/dnnd.s71986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a progressive, neurodegenerative disease with unpredictable phases of relapse and remission. The cause of MS is unknown, but the pathology is characterized by infiltration of auto-reactive immune cells into the central nervous system (CNS) resulting in widespread neuroinflammation and neurodegeneration. Immunomodulatory-based therapies emerged in the 1990s and have been a cornerstone of disease management ever since. Interferon β (IFNβ) was the first biologic approved after demonstrating decreased relapse rates, disease activity and progression of disability in clinical trials. However, frequent dosing schedules have limited patient acceptance for long-term therapy. Pegylation, the process by which molecules of polyethylene glycol are covalently linked to a compound, has been utilized to increase the half-life of IFNβ and decrease the frequency of administration required. To date, there has been one clinical trial evaluating the efficacy of pegylated IFN. The purpose of this article is to provide an overview of the role of IFN in the treatment of MS and evaluate the available evidence for pegylated IFN therapy in MS.
Collapse
Affiliation(s)
- Kendra L Furber
- Laboratory of Molecular Cell Biology, .,College of Pharmacy and Nutrition, .,Neuroscience Research Cluster, University of Saskatchewan,
| | - Marina Van Agten
- Laboratory of Molecular Cell Biology, .,College of Pharmacy and Nutrition, .,Neuroscience Research Cluster, University of Saskatchewan,
| | - Charity Evans
- College of Pharmacy and Nutrition, .,Cameco Multiple Sclerosis Neuroscience Research Center, City Hospital,
| | | | - J Ronald Doucette
- Neuroscience Research Cluster, University of Saskatchewan, .,Cameco Multiple Sclerosis Neuroscience Research Center, City Hospital, .,Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Adil J Nazarali
- Laboratory of Molecular Cell Biology, .,College of Pharmacy and Nutrition, .,Neuroscience Research Cluster, University of Saskatchewan, .,Cameco Multiple Sclerosis Neuroscience Research Center, City Hospital,
| |
Collapse
|
19
|
Buzzard K, Chan WH, Kilpatrick T, Murray S. Multiple Sclerosis: Basic and Clinical. ADVANCES IN NEUROBIOLOGY 2017; 15:211-252. [DOI: 10.1007/978-3-319-57193-5_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Pandini G, Satriano C, Pietropaolo A, Gianì F, Travaglia A, La Mendola D, Nicoletti VG, Rizzarelli E. The Inorganic Side of NGF: Copper(II) and Zinc(II) Affect the NGF Mimicking Signaling of the N-Terminus Peptides Encompassing the Recognition Domain of TrkA Receptor. Front Neurosci 2016; 10:569. [PMID: 28090201 PMCID: PMC5201159 DOI: 10.3389/fnins.2016.00569] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/25/2016] [Indexed: 12/31/2022] Open
Abstract
The nerve growth factor (NGF) N-terminus peptide, NGF(1–14), and its acetylated form, Ac-NGF(1–14), were investigated to scrutinize the ability of this neurotrophin domain to mimic the whole protein. Theoretical calculations demonstrated that non-covalent forces assist the molecular recognition of TrkA receptor by both peptides. Combined parallel tempering/docking simulations discriminated the effect of the N-terminal acetylation on the recognition of NGF(1–14) by the domain 5 of TrkA (TrkA-D5). Experimental findings demonstrated that both NGF(1–14) and Ac-NGF(1–14) activate TrkA signaling pathways essential for neuronal survival. The NGF-induced TrkA internalization was slightly inhibited in the presence of Cu2+ and Zn2+ ions, whereas the metal ions elicited the NGF(1–14)-induced internalization of TrkA and no significant differences were found in the weak Ac-NGF(1–14)-induced receptor internalization. The crucial role of the metals was confirmed by experiments with the metal-chelator bathocuproine disulfonic acid, which showed different inhibitory effects in the signaling cascade, due to different metal affinity of NGF, NGF(1–14) and Ac-NGF(1–14). The NGF signaling cascade, activated by the two peptides, induced CREB phosphorylation, but the copper addition further stimulated the Akt, ERK and CREB phosphorylation in the presence of NGF and NGF(1–14) only. A dynamic and quick influx of both peptides into PC12 cells was tracked by live cell imaging with confocal microscopy. A significant role of copper ions was found in the modulation of peptide sub-cellular localization, especially at the nuclear level. Furthermore, a strong copper ionophoric ability of NGF(1–14) was measured. The Ac-NGF(1–14) peptide, which binds copper ions with a lower stability constant than NGF(1–14), exhibited a lower nuclear localization with respect to the total cellular uptake. These findings were correlated to the metal-induced increase of CREB and BDNF expression caused by NGF(1–14) stimulation. In summary, we here validated NGF(1–14) and Ac-NGF(1–14) as first examples of monomer and linear peptides able to activate the NGF-TrkA signaling cascade. Metal ions modulated the activity of both NGF protein and the NGF-mimicking peptides. Such findings demonstrated that NGF(1–14) sequence can reproduce the signal transduction of whole protein, therefore representing a very promising drug candidate for further pre-clinical studies.
Collapse
Affiliation(s)
- Giuseppe Pandini
- Endocrinology, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Medical Center, University of CataniaCatania, Italy; Institute of Biostructures and Bioimages - Catania, National Research CouncilCatania, Italy
| | - Cristina Satriano
- Department of Chemical Sciences, University of CataniaCatania, Italy; Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi BiologiciBari, Italy
| | | | - Fiorenza Gianì
- Endocrinology, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Medical Center, University of CataniaCatania, Italy; Institute of Biostructures and Bioimages - Catania, National Research CouncilCatania, Italy
| | | | - Diego La Mendola
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi BiologiciBari, Italy; Department of Pharmacy, University of PisaPisa, Italy
| | - Vincenzo G Nicoletti
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi BiologiciBari, Italy; Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of CataniaCatania, Italy
| | - Enrico Rizzarelli
- Institute of Biostructures and Bioimages - Catania, National Research CouncilCatania, Italy; Department of Chemical Sciences, University of CataniaCatania, Italy; Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi BiologiciBari, Italy
| |
Collapse
|
21
|
Pul R, Saadat M, Morbiducci F, Skripuletz T, Pul Ü, Brockmann D, Sühs KW, Schwenkenbecher P, Kahl KG, Pars K, Stangel M, Trebst C. Longitudinal time-domain optic coherence study of retinal nerve fiber layer in IFNβ-treated and untreated multiple sclerosis patients. Exp Ther Med 2016; 12:190-200. [PMID: 27347038 PMCID: PMC4906774 DOI: 10.3892/etm.2016.3300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
Quantification of the retinal nerve fiber layer (RNFL) by optical coherence tomography (OCT) has been proposed to provide an indirect measure for retinal axonal loss. The aim of the present study was to determine whether interferon beta (IFNβ) treatment impedes retinal axonal loss in multiple sclerosis (MS) patients. A total of 48 patients with MS (24 IFNβ-1b-treated and 24 untreated subjects) and 12 healthy controls were enrolled in a prospective longitudinal OCT study. OCT measurements were performed for both eyes of each subject at baseline, and at 3-, 6-, and 12-month follow-up examinations using a time-domain OCT. At each visit, we additionally recorded full-field visual evoked potential (VEP) responses and performed the paced auditory serial addition test (PASAT), in addition to expanded disability status scale (EDSS) scoring. Generalized estimation equation (GEE) was used to account for repeated measurements and paired-data. The model-based approach predicted a monthly reduction in the RNFL thickness by 0.19 µm in the eyes of the MS subjects. The reduction was estimated to be 0.17 µm in case of IFNβ-treatment and 0.16 µm in case of no treatment. Treatment duration and group allocation were not significantly associated with the RNFL thickness. Inclusion of further longitudinal data (EDSS, two and three second PASAT) in each of our models did not result in any significant association. In summary, over a period of one year no significant association between IFNβ-1b treatment and RNFL thinning was identified in patients with MS.
Collapse
Affiliation(s)
- Refik Pul
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Mehdi Saadat
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Franco Morbiducci
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Ünsal Pul
- Department of Thoracic and Cardiovascular Surgery, University Hospital Essen, 45147 Essen, Germany
| | - Dorothee Brockmann
- Department of Ophthalmology, Hannover Medical School, 30625 Hannover, Germany
| | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Kai Günter Kahl
- Department of Psychiatry, Hannover Medical School, 30625 Hannover, Germany
| | - Kaweh Pars
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Martin Stangel
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Corinna Trebst
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
22
|
Interferon-β Inhibits Neurotrophin 3 Signalling and Pro-Survival Activity by Upregulating the Expression of Truncated TrkC-T1 Receptor. Mol Neurobiol 2016; 54:1825-1843. [PMID: 26887385 DOI: 10.1007/s12035-016-9789-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/11/2016] [Indexed: 12/17/2022]
Abstract
Although clinically useful for the treatment of various diseases, type I interferons (IFNs) have been implicated as causative factors of a number of neuroinflammatory disorders characterized by neuronal damage and altered CNS functions. As neurotrophin 3 (NT3) plays a critical role in neuroprotection, we examined the effects of IFN-β on the signalling and functional activity of the NT3/TrkC system. We found that prolonged exposure of differentiated human SH-SY5Y neuroblastoma cells to IFN-β impaired the ability of NT3 to induce transphosphorylation of the full-length TrkC receptor (TrkC-FL) and the phosphorylation of downstream signalling molecules, including PLCγ1, Akt, GSK-3β and ERK1/2. NT3 was effective in protecting the cells against apoptosis triggered by serum withdrawal or thapsigargin but not IFN-β. Prolonged exposure to the cytokine had little effects on TrkC-FL levels but markedly enhanced the messenger RNA (mRNA) and protein levels of the truncated isoform TrkC-T1, a dominant-negative receptor that inhibits TrkC-FL activity. Cell depletion of TrkC-T1 by small interfering RNA (siRNA) treatment enhanced NT3 signalling through TrkC-FL and allowed the neurotrophin to counteract IFN-β-induced apoptosis. Furthermore, the upregulation of TrkC-T1 by IFN-β was associated with the inhibition of NT3-induced recruitment of the scaffold protein tamalin to TrkC-T1 and tamalin tyrosine phosphorylation. These data indicate that IFN-β exerts a negative control on NT3 pro-survival signalling through a novel mechanism involving the upregulation of TrkC-T1.
Collapse
|
23
|
Increased Serum Brain-derived Neurotrophic Factor in Multiple Sclerosis Patients on Interferon-β and Its Impact on Functional Abilities. Neurologist 2015; 20:57-60. [DOI: 10.1097/nrl.0000000000000053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
24
|
Severa M, Rizzo F, Giacomini E, Salvetti M, Coccia EM. IFN-β and multiple sclerosis: cross-talking of immune cells and integration of immunoregulatory networks. Cytokine Growth Factor Rev 2014; 26:229-39. [PMID: 25498525 DOI: 10.1016/j.cytogfr.2014.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/05/2014] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is characterized by autoimmune inflammation affecting the central nervous system and subsequent neurodegeneration. Historically, damage was thought to be mediated exclusively by auto-antigen-activated pro-inflammatory T cells. However, more recently, we are gaining increasing knowledge on the pathogenic role played in MS by B cells, dendritic cells and monocytes. IFN-β therapy was one the first approved therapy for MS for its ability to reduce relapse rate and MRI lesion activity and to significantly decrease risk of disability progression. IFN-β-mediated mechanisms of action, even if not completely understood, mainly rely on its multifaceted pleiotropic effects resulting in sustained anti-inflammatory properties directed toward almost every immune cell type. Here, we will discuss in detail literature data characterizing the pathogenic activity of the different immune cell subsets involved in MS pathogenesis and how IFN-β therapy regulates their function by modulating bystander responses. We believe that the effectiveness of this drug in MS treatment, even if in use for a long time, can unveil new insights on this disease and still teach a lesson to researchers in the MS field.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Fabiana Rizzo
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Giacomini
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Salvetti
- Centre for Experimental Neurological Therapies (CENTERS) - Neurology and Department of Neurosciences, Mental Health and Sensory Organs; Sapienza, University of Rome, S. Andrea Hospital Site, Italy
| | - Eliana M Coccia
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
25
|
Adipose tissue-derived mesenchymal stem cells cultured at high cell density express brain-derived neurotrophic factor and exert neuroprotective effects in a 6-hydroxydopamine rat model of Parkinson’s disease. Genes Genomics 2014. [DOI: 10.1007/s13258-014-0239-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
26
|
The nerve growth factor signaling and its potential as therapeutic target for glaucoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:759473. [PMID: 25250333 PMCID: PMC4164261 DOI: 10.1155/2014/759473] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 08/12/2014] [Indexed: 12/25/2022]
Abstract
Neuroprotective therapies which focus on factors leading to retinal ganglion cells (RGCs) degeneration have been drawing more and more attention. The beneficial effects of nerve growth factor (NGF) on the glaucoma have been recently suggested, but its effects on eye tissue are complex and controversial in various studies. Recent clinical trials of systemically and topically administrated NGF demonstrate that NGF is effective in treating several ocular diseases, including glaucoma. NGF has two receptors named high affinity NGF tyrosine kinase receptor TrkA and low affinity receptor p75NTR. Both receptors exist in cells in retina like RGC (expressing TrkA) and glia cells (expressing p75NTR). NGF functions by binding to TrkA or p75NTR alone or both together. The binding of NGF to TrkA alone in RGC promotes RGC's survival and proliferation through activation of TrkA and several prosurvival pathways. In contrast, the binding of NGF to p75NTR leads to apoptosis although it also promotes survival in some cases. Binding of NGF to both TrkA and p75NTR at the same time leads to survival in which p75NTR functions as a TrkA helping receptor. This review discusses the current understanding of the NGF signaling in retina and the therapeutic implications in the treatment of glaucoma.
Collapse
|
27
|
Antimisiaris S, Mourtas S, Markoutsa E, Skouras A, Papadia K. Nanoparticles for Diagnosis and/or Treatment of Alzheimer's Disease. Adv Healthc Mater 2014. [DOI: 10.1002/9781118774205.ch4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
28
|
Affiliation(s)
- Eleonora Tavazzi
- Multiple Sclerosis Center - Unit of Motor Neurorehabilitation, IRCCS Santa Maria Nascente, Fondazione Don Gnocchi, Milan, Italy
| | - Marco Rovaris
- Multiple Sclerosis Center - Unit of Motor Neurorehabilitation, IRCCS Santa Maria Nascente, Fondazione Don Gnocchi, Milan, Italy
| | - Loredana La Mantia
- Multiple Sclerosis Center - Unit of Motor Neurorehabilitation, IRCCS Santa Maria Nascente, Fondazione Don Gnocchi, Milan, Italy
| |
Collapse
|
29
|
Dedoni S, Olianas MC, Ingianni A, Onali P. Type I interferons up-regulate the expression and signalling of p75 NTR/TrkA receptor complex in differentiated human SH-SY5Y neuroblastoma cells. Neuropharmacology 2014; 79:321-34. [PMID: 24333329 DOI: 10.1016/j.neuropharm.2013.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 10/25/2013] [Accepted: 12/02/2013] [Indexed: 01/10/2023]
Abstract
Both type I interferons (IFNs) and neurotrophins regulate neuroadaptive responses, but relatively little is known on the interaction between these two classes of regulatory proteins. Here we investigated the effect of IFN-β on the expression and functional activity of the common neurotrophin receptor p75NTR and the nerve growth factor (NGF) receptor TrkA. In differentiated human SH-SY5Y neuroblastoma cells prolonged exposure to IFN-β up-regulated p75NTR and TrkA levels, failed to affect the content of sortilin, a p75NTR co-receptor, and, consistent with our previous finding, down-regulated the brain-derived neurotrophic factor receptor TrkB. Quantitative real time RT-PCR indicated that IFN-β increased p75NTR and TrkA mRNA levels. In control and IFN-β treated cells proNGF failed to induce c-Jun N-terminal kinase and nuclear factor/kB activation, two p75NTR/sortilin signalling pathways mediating neuronal death. On the other hand, IFN-β treatment enhanced TrkA autophosphorylation and signalling induced by NGF and proNGF. Knockdown of p75NTR by siRNA reduced TrkA activation by proNGF and a subnanomolar concentration of NGF, whereas co-immunoprecipitation indicated close association of p75NTR and TrkA. Co-treatment with either NGF or proNGF reduced IFN-β pro-apoptotic and anti-neurotrophic effects. Similarly, in primary mouse hippocampal neurons IFN-β increased p75NTR and TrkA expression, down-regulated TrkB and enhanced NGF-induced phosphorylation of the pro-survival protein kinase Akt. The data demonstrate that in neuronal cells IFN-β differentially affects the expression and signalling of neurotrophin receptors and suggest that the up-regulation of the p75NTR/TrkA signalling complex may constitute a novel mechanism by which this cytokine selectively attenuates its pro-apoptotic effect in NGF-responsive cells.
Collapse
Affiliation(s)
- Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Angela Ingianni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
30
|
Grimaldi LME, Zappalà G, Iemolo F, Castellano AE, Ruggieri S, Bruno G, Paolillo A. A pilot study on the use of interferon beta-1a in early Alzheimer's disease subjects. J Neuroinflammation 2014; 11:30. [PMID: 24524367 PMCID: PMC3931325 DOI: 10.1186/1742-2094-11-30] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/26/2014] [Indexed: 11/10/2022] Open
Abstract
Despite the fact that multiple sclerosis (MS) and Alzheimer’s disease (AD) share common neuroimmunological features, interferon beta 1a (IFNβ1a), the well-established treatment for the prevention of disease progression and cognitive decline in MS patients, has never been used in AD. We evaluated the safety and efficacy of IFNβ1a in subjects affected by mild-to-moderate AD in a double-blind, randomized, placebo-controlled, multicenter pilot study. Forty-two early Alzheimer’s patients were randomized to receive either a 22 mcg subcutaneous injection of IFNβ1a or placebo three times per week. A treatment period of 28 weeks was followed by 24 weeks of observation. IFNβ1a was well tolerated and adverse events were infrequent and mild to moderate. Although not statistically significant, a reduction in disease progression during follow-up was measured in IFNβ1a-treated patients by the Alzheimer’s Disease Assessment Scale cognitive subscale. Interestingly, the treatment group showed significant improvements in the Instrumental Activities of Daily Living and Physical Self-maintenance Scale. This study suggests that IFNβ1a is safe and well tolerated in early AD patients, and its possible beneficial role should be further investigated in larger studies.
Collapse
Affiliation(s)
- Luigi Maria Edoardo Grimaldi
- U,O, di Neurologia, Fondazione Istituto San Raffaele "G, Giglio" di Cefalù, Contrada Pietrapollastra, 90015 Cefalù, PA, Italy.
| | | | | | | | | | | | | |
Collapse
|
31
|
Aharoni R. New findings and old controversies in the research of multiple sclerosis and its model experimental autoimmune encephalomyelitis. Expert Rev Clin Immunol 2014; 9:423-40. [DOI: 10.1586/eci.13.21] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
32
|
Sühs KW, Hein K, Pehlke JR, Käsmann-Kellner B, Diem R. Retinal nerve fibre layer thinning in patients with clinically isolated optic neuritis and early treatment with interferon-beta. PLoS One 2012; 7:e51645. [PMID: 23272128 PMCID: PMC3521715 DOI: 10.1371/journal.pone.0051645] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 11/02/2012] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Optic neuritis is associated with neurodegeneration leading to chronic impairment of visual functions. OBJECTIVE This study investigated whether early treatment with interferon beta (IFN-β) slows retinal nerve fibre layer (RNFL) thinning in clinically isolated optic neuritis. METHODS Twenty patients with optic neuritis and visual acuity decreased to ≤0.5 (decimal system) were included into this prospective, open-label, parallel group 4-month observation. After methylprednisolone pulse therapy, 10 patients received IFN-β from week 2 onwards. This group was compared to 10 patients free of any disease modifying treatment (DMT). The parameter of interest was change in RNFL thickness assessed at baseline and at weeks 4, 8, and 16. Changes in visual acuity, visual field, and visual evoked potentials (VEPs) served as additional outcome parameters. RESULTS RNFL thinning did not differ between the groups with a mean reduction of 9.80±2.80 µm in IFN-β-treated patients (±SD) vs. 12.44±5.79 µm in patients who did not receive DMT (baseline non-affected eye minus affected eye at week 16; p = 0.67, t-test, 95% confidence interval: -15.77 to 10.48). Parameters of visual function did not show any differences between the groups either. CONCLUSIONS In isolated optic neuritis, early IFN-β treatment did not influence RNFL thinning nor had it any effect on recovery of visual functions.
Collapse
Affiliation(s)
- Kurt-Wolfram Sühs
- Department of Neurology, Saarland University, Homburg, Germany
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Katharina Hein
- Department of Neurology, Georg-August University, Göttingen, Germany
| | - Jens R. Pehlke
- Department of Neurology, Saarland University, Homburg, Germany
- Department of Addiction Disorders, LWL Clinic Münster, Münster, Germany
| | | | - Ricarda Diem
- Department of Neurology, Saarland University, Homburg, Germany
- Department of Neuro-oncology, University Clinic Heidelberg, Heidelberg, Germany
| |
Collapse
|
33
|
Interferon-β induces hepatocyte growth factor in monocytes of multiple sclerosis patients. PLoS One 2012; 7:e49882. [PMID: 23166786 PMCID: PMC3498184 DOI: 10.1371/journal.pone.0049882] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 10/15/2012] [Indexed: 12/24/2022] Open
Abstract
Interferon-β is a first-line therapy used to prevent relapses in relapsing-remitting multiple sclerosis. The clinical benefit of interferon-β in relapsing-remitting multiple sclerosis is attributed to its immunomodulatory effects on inflammatory mediators and T cell reactivity. Here, we evaluated the production of hepatocyte growth factor, a neuroprotective and neuroinflammation-suppressive mediator, by peripheral blood mononuclear cells collected from interferon-β−treated relapsing-remitting multiple sclerosis patients, relapsing remitting multiple sclerosis patients not treated with interferon-β, and healthy volunteers. Using intracellular flow cytometry analysis, increased production of hepatocyte growth factor was observed in circulating CD14+ monocytes from patients undergoing long-term treatment with interferon-β versus untreated patients. Complementary in vitro studies confirmed that treatment with interferon-β induced rapid and transient transcription of the hepatocyte growth factor gene in CD14+ monocytes and that intracellular and secreted monocytic hepatocyte growth factor protein levels were markedly stimulated by interferon-β treatment. Additional exploration revealed that “pro-inflammatory” (CD14+CD16+) monocytes produced similar levels of hepatocyte growth factor in response to interferon-β as “classical” (CD14+CD16−) monocytes, and that CD14+ monocytes but not CD4+ T cells express the hepatocyte growth factor receptor c-Met. Our findings suggest that interferon-β may mediate some of its therapeutic effects in relapsing-remitting multiple sclerosis through the induction of hepatocyte growth factor by blood monocytes by coupling immune regulation and neuroprotection.
Collapse
|
34
|
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS) with unknown etiology. It was recently suggested that autoimmunity, which had long been considered to be destructive in MS, might also play a protective role in the CNS of MS patients. Neurotrophins are polypeptides belonging to the neurotrophic factor family. While neurotrophins mediate cell survival and proliferation in the nervous system, they are also expressed within peripheral blood mononuclear cells fraction (PBMCs) of immunological system. In MS additional neurotrophic support from PBMCs might compensate relative neurotrophins deficiency in the damaged CNS tissue that needs to be repaired. Failure to produce the adequate neurotrophins concentrations might result in decreased protection of the CNS, consequently leading to increased atrophy, which is the main determinant of MS patients' end-point disability. There are several lines of evidence, both from clinical research and animal models, suggesting that neurotrophins play a pivotal role in neuroprotective and neuroregenerative processes that are often defective in the course of MS. It seems that neuroprotective strategies might be used as potentially valuable add-on therapies, alongside traditional immunomodulatory treatment in multiple sclerosis.
Collapse
|
35
|
Buzzard KA, Broadley SA, Butzkueven H. What do effective treatments for multiple sclerosis tell us about the molecular mechanisms involved in pathogenesis? Int J Mol Sci 2012. [PMID: 23202920 PMCID: PMC3497294 DOI: 10.3390/ijms131012665] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Multiple sclerosis is a potentially debilitating disease of the central nervous system. A concerted program of research by many centers around the world has consistently demonstrated the importance of the immune system in its pathogenesis. This knowledge has led to the formal testing of a number of therapeutic agents in both animal models and humans. These clinical trials have shed yet further light on the pathogenesis of MS through their sometimes unexpected effects and by their differential effects in terms of impact on relapses, progression of the disease, paraclinical parameters (MRI) and the adverse events that are experienced. Here we review the currently approved medications for the commonest form of multiple sclerosis (relapsing-remitting) and the emerging therapies for which preliminary results from phase II/III clinical trials are available. A detailed analysis of the molecular mechanisms responsible for the efficacy of these medications in multiple sclerosis indicates that blockade or modulation of both T- and B-cell activation and migration pathways in the periphery or CNS can lead to amelioration of the disease. It is hoped that further therapeutic trials will better delineate the pathogenesis of MS, ultimately leading to even better treatments with fewer adverse effects.
Collapse
Affiliation(s)
- Katherine A. Buzzard
- Department of Neurology, Royal Melbourne Hospital, Royal Parade, Parkville VIC 3050, Australia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +61-3-8344-1802; Fax: +61-3-9348-1707
| | - Simon A. Broadley
- School of Medicine, Griffith University, Gold Coast Campus, QLD 4222, Australia; E-Mail:
- Department of Neurology, Gold Coast Hospital, 108 Nerang Street, Southport QLD 4215, Australia
| | - Helmut Butzkueven
- Melbourne Brain Centre at the Royal Melbourne Hospital, Department of Medicine, University of Melbourne, Royal Parade, Parkville VIC 3010, Australia; E-Mail:
| |
Collapse
|
36
|
Patti F. Treatment of cognitive impairment in patients with multiple sclerosis. Expert Opin Investig Drugs 2012; 21:1679-99. [DOI: 10.1517/13543784.2012.716036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
Kalinowska-Łyszczarz A, Pawlak MA, Michalak S, Losy J. Cognitive deficit is related to immune-cell beta-NGF in multiple sclerosis patients. J Neurol Sci 2012; 321:43-8. [PMID: 22877509 DOI: 10.1016/j.jns.2012.07.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/29/2012] [Accepted: 07/19/2012] [Indexed: 01/17/2023]
Abstract
OBJECTIVES Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), causing cognitive impairment in 45-65% of patients. Beta-NGF facilitates proper cholinergic transmission in the healthy CNS. In MS-damaged tissue there is a relative deficit of neurotrophins that might be compensated by peripheral blood mononuclear cells (PBMCs) synthesis. Our aim was to evaluate the relationship between PBMCs neurotrophins' expression and cognitive performance in relapsing-remitting MS (RRMS) patients. PATIENTS AND METHODS Beta-NGF, NT-3 and NT-4/5 levels were measured in sera and in PBMCs by ELISA method in 41 RRMS patients in remission. All patients underwent neuropsychological assessment with a battery of 10 tests evaluating a wide range of cognitive functions. RESULTS PBMCs beta-NGF concentration correlated significantly with spontaneous word list generation test (Pearson R=0.37, p=0.02) and 15-Word List Recall Test results (Pearson R=0.40, p<0.00). Both tests assessing semantic memory correlated significantly with the cognitive composite score, defined as a number of tests in which patients performed below group median for the given test. CONCLUSIONS In RRMS beta-NGF is strongly linked to cognitive performance, which makes it an attractive therapeutic target. It might play a neuroprotective role in MS, especially in the cognitive domain.
Collapse
Affiliation(s)
- Alicja Kalinowska-Łyszczarz
- Department of Neurochemistry and Neuropathology, Chair of Neurology, Poznan University of Medical Sciences, Poland.
| | | | | | | |
Collapse
|
38
|
Bar-Or A, Rieckmann P, Traboulsee A, Yong VW. Targeting progressive neuroaxonal injury: lessons from multiple sclerosis. CNS Drugs 2011; 25:783-99. [PMID: 21870889 DOI: 10.2165/11587820-000000000-00000] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), are characterized by progressive neuroaxonal injury, suggesting a common pathophysiological pathway. Identification and development of neuroprotective therapies for such diseases has proven a major challenge, particularly because of an already substantial neuroaxonal compromise at the time of initial onset of clinical symptoms. Methods for early identification of neurodegeneration are therefore vital to ensure that neuroprotective therapies are applied as early as possible. Recent investigations have enhanced our understanding of the role of neuroaxonal injury in multiple sclerosis (MS). As MS generally manifests earlier in life and can be diagnosed much earlier in the course of the disease than the above-mentioned 'classic' neurodegenerative diseases, it is possible that MS could be used as a model disease to study degeneration and regeneration of the CNS. The mechanism of neuroaxonal injury in MS is believed to be inflammation-led neurodegeneration; however, the reverse may also be true (i.e. neuroaxonal degeneration may precede inflammation). Animal models of PD, AD and ALS have shown that it is likely that most cases of disease are due to initial inflammation, followed by a degenerative process, providing a parallel between MS and the classic neurodegenerative diseases. Other common factors between MS and the neurodegenerative diseases include iron and mitochondrial dysregulation, abnormalities in α-synuclein and tau protein, and a number of immune mediators. Conventional MRI techniques, using markers such as T2-weighted lesions, gadolinium-enhancing lesions and T1-weighted hypointensities, are readily available and routinely used in clinical practice; however, the utility of these MRI measures to predict disease progression in MS is limited. More recently, MRI techniques that provide more pathology-specific data have been applied in MS studies, including magnetic resonance spectroscopy, magnetization transfer ratio and myelin water imaging. Optical coherence tomography (OCT) is a non-MRI technique that quantifies optic nerve integrity and retinal ganglion cell loss as markers of neuroaxonal injury; more research is needed to evaluate whether information obtained from OCT is a reliable marker of axonal injury and long-term disability in MS. Using these advanced techniques, it may become possible to follow degeneration and regeneration longitudinally in patients with MS and to better differentiate the effects of drugs under investigation. Currently available immune-directed therapies that are approved by the US FDA for the first-line treatment of MS (interferon-β and glatiramer acetate) have been shown to decelerate the inflammatory process in MS; however, such therapy is less effective in preventing the progression of the disease and neuroaxonal injury. The use of MS as a clinical model to study modulation of neuroaxonal injury in the brain could have direct implications for the development of treatment strategies in neurodegenerative diseases such as AD, PD and ALS.
Collapse
Affiliation(s)
- Amit Bar-Or
- Department of Neurology and Neurosurgery and Microbiology and Immunology, McGill University, Neuroimmunology Unit, Montreal, QC, Canada
| | | | | | | |
Collapse
|
39
|
Gold R, Wolinsky JS, Amato MP, Comi G. Evolving expectations around early management of multiple sclerosis. Ther Adv Neurol Disord 2011; 3:351-67. [PMID: 21179596 DOI: 10.1177/1756285610385608] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Multiple sclerosis is a progressive inflammatory disease of the central nervous system. With prevention or at least delay of disease progression as a key target in the management of multiple sclerosis, current opinion on treatment encourages early intervention with well-tolerated disease-modifying treatments in order to optimize long-term clinical outcomes. Patients presenting with a clinically isolated syndrome (CIS) may progress to clinically definite multiple sclerosis, and clinical trials have demonstrated that early treatment with interferon beta can reduce the conversion rate. Cognitive impairment may already be present in patients with CISs. Today there is evolving evidence that cognitive impairment may be relevant for prognosis and that early treatment with interferon beta may also have a protective effect on the cognitive function. As an accumulation of neuronal loss is now considered to underlie the development of persistent disability in multiple sclerosis, it is crucial that treatment can protect against neuronal damage. In addition to its anti-inflammatory activity, interferon beta may have direct and indirect neuroprotective effects, and several studies have explored the role of interferon beta in regulating neuroprotective factors. With over 15 years of clinical experience as evidence, the long-term safety and efficacy of interferon beta treatment is unquestionable. Results from the CIS studies have demonstrated the high percentage of patients converting to clinically definite multiple sclerosis without treatment and the short- and long-term benefits of an early use of disease-modifying treatments. These findings support starting disease-modifying treatment as soon as the diagnosis of MS is reasonably formulated.
Collapse
Affiliation(s)
- Ralf Gold
- Department of Neurology, St. Josef-Hospital/Ruhr-University Bochum, Gudrunstrasse 56, 44791 Bochum, Germany
| | | | | | | |
Collapse
|
40
|
Abstract
Multiple sclerosis (MS) is characterized by autoimmune inflammation and subsequent neurodegeneration. It is believed that early in the disease course, proinflammatory T cells that are activated in the periphery by antigen presentation cross the blood-brain barrier (BBB) into the CNS directed by various chemotaxic agents. However, to date, there has been no formal demonstration of a specific precipitating antigen. Once inside the CNS, activated T cells including T helper-1 (T(h)1), T(h)17, γδ and CD8+ types are believed to secrete proinflammatory cytokines. Decreased levels of T(h)2 cells also correlate with relapses and disease progression in MS, since T(h)2-derived cytokines are predominantly anti-inflammatory. In healthy tissue, inflammatory effects are opposed by specific subsets of regulatory T cells (T(regs)) including CD4+, CD25+ and FoxP3+ cells that have the ability to downregulate the activity of proinflammatory T cells, allowing repair and recovery to generally follow inflammatory insult. Given their function, the pathogenesis of MS most likely involves deficits of T(reg) function, which allow autoimmune inflammation and resultant neurodegeneration to proceed relatively unchecked. Interferons (IFNs) are naturally occurring cytokines possessing a wide range of anti-inflammatory properties. Recombinant forms of IFNβ are widely used as first-line treatment in relapsing forms of MS. The mechanism of action of IFNβ is complex, involving effects at multiple levels of cellular function. IFNβ appears to directly increase expression and concentration of anti-inflammatory agents while downregulating the expression of proinflammatory cytokines. IFNβ treatment may reduce the trafficking of inflammatory cells across the BBB and increase nerve growth factor production, leading to a potential increase in neuronal survival and repair. IFNβ can also increase the number of CD56bright natural killer cells in the peripheral blood. These cells are efficient producers of anti-inflammatory mediators, and may have the ability to curb neuron inflammation. The mechanistic effects of IFNβ manifest clinically as reduced MRI lesion activity, reduced brain atrophy, increased time to reach clinically definite MS after the onset of neurological symptoms, decreased relapse rate and reduced risk of sustained disability progression. The mechanism of action of IFNβ in MS is multifactorial and incompletely understood. Ongoing and future studies will increase our understanding of the actions of IFNβ on the immune system and the CNS, which will in turn aid advances in the management of MS.
Collapse
Affiliation(s)
- Bernd C Kieseier
- Department of Neurology, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
41
|
Filippi M, Rocca MA, Camesasca F, Cook S, O'Connor P, Arnason BGW, Kappos L, Goodin D, Jeffery D, Hartung HP, Comi G, Wolinsky JS, Bogumil T, Pohl C, Beckmann K, Sandbrink R, Croze E, Brown C, Desimone TM, Arnold DL, Cutter G, Knappertz V. Interferon β-1b and glatiramer acetate effects on permanent black hole evolution. Neurology 2011; 76:1222-8. [PMID: 21464426 DOI: 10.1212/wnl.0b013e3182143577] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To compare interferon β-1b (IFNβ-1b) and glatiramer acetate (GA) on new lesion (NL) (gadolinium-enhancing, new T2) evolution into permanent black holes (PBH)--a marker of irreversible tissue damage--in patients with relapsing-remitting multiple sclerosis (RRMS). METHODS BEYOND was a large, phase III, clinical trial comparing IFNβ-1b 250 μg, IFNβ-1b 500 μg, and GA (2:2:1). Patient scans were reexamined post hoc for PBH in a rater-blinded manner. Two predefined coprimary endpoints compared IFNβ-1b 250 μg with GA: first, number of PBH per patient at year 2 evolving from year 1 NL, then proportion of year 1 NL evolving into PBH at year 2. IFNβ-1b 500 μg and GA were compared in an exploratory fashion. RESULTS Approximately 90% (1,957/2,244) of patients had NL at year 1 with follow-up at year 2. Mean numbers of PBH per patient at year 2 evolving from year 1 NL were lower for IFNβ-1b 250 μg than GA (0.30 vs 0.43; p = 0.0451). The proportion of NL evolving into PBH was similar (IFNβ-1b 250 μg vs GA: 21.6% vs 23.5%; p > 0.20). For IFNβ-1b 500 μg, both the mean PBH number per patient at year 2 evolving from year 1 NL (0.26 vs 0.43; p = 0.0037) and proportion of NL evolving into PBH (16.3% vs 23.5%; p = 0.0409) were lower relative to GA. CONCLUSION IFNβ-1b affected PBH development to a similar or better extent than GA. IFNβ-1b favorably influences an MRI outcome indicative of permanent tissue destruction in the brains of patients with multiple sclerosis. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that IFNβ-1b is associated with a reduction in MRI PBH formation and evolution compared with GA between years 1 and 2 of treatment.
Collapse
Affiliation(s)
- M Filippi
- University Hospital San Raffaele, Via Olgettina, 20132 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The fundamental role of inflammatory immune processes in the pathology of multiple sclerosis (MS) provides the rationale for immunomodulatory therapies that attempt to shift the immune system from pro-inflammatory to anti-inflammatory pathways and induce regulatory mechanisms. Growing understanding of immune cellular and molecular mechanisms together with modern biotechnology engendered promising immunomodulatory treatment strategies, with novel mechanisms of actions and different levels of specificity. These include inhibitory molecules, monoclonal antibodies, cell therapies and agents that are administered orally or by infrequent infusions. Several of these treatments have demonstrated impressive efficacy in Phase II and III clinical trials by reducing disease activity and accumulation of disability. However, with the advent of potent therapies, rare but severe adverse effects, such as CNS infections and malignancies, have occurred. This article describes current and upcoming immunomodulatory strategies for MS therapy. The potential of immunomodulatory treatments to counteract the inflammatory characteristics of MS and support neuroprotective processes is discussed.
Collapse
Affiliation(s)
- Rina Aharoni
- The Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
43
|
Interferon-beta treatment normalises the inhibitory effect of serum from multiple sclerosis patients on oligodendrocyte progenitor proliferation. Neurosci Lett 2010; 485:107-11. [DOI: 10.1016/j.neulet.2010.08.075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Revised: 08/25/2010] [Accepted: 08/25/2010] [Indexed: 11/21/2022]
|
44
|
Sättler MB, Bähr M. Future neuroprotective strategies. Exp Neurol 2010; 225:40-7. [DOI: 10.1016/j.expneurol.2009.08.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 08/18/2009] [Accepted: 08/19/2009] [Indexed: 12/27/2022]
|
45
|
Lecht S, Arien-Zakay H, Wagenstein Y, Inoue S, Marcinkiewicz C, Lelkes PI, Lazarovici P. Transient signaling of Erk1/2, Akt and PLCgamma induced by nerve growth factor in brain capillary endothelial cells. Vascul Pharmacol 2010; 53:107-14. [PMID: 20434587 DOI: 10.1016/j.vph.2010.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 03/15/2010] [Accepted: 04/22/2010] [Indexed: 11/26/2022]
Abstract
Cumulative evidences suggest that nerve growth factor (NGF) promotes angiogenic effects such as proliferation and migration of endothelial cells (ECs) from different vascular beds, induces capillary sprouting in chorioallantoic membrane and improves in vivo vascularization in a hind-limb ischemic model. In the present study, we sought to investigate the signaling properties of NGF in a microcapillary ECs model compared to those of a neuronal model. NGF-induced phosphorylation of signaling molecules Erk1/2, Akt and PLCgamma were measured using Western blotting and compared between mouse NGF (mNGF) and snake venom NGF analogues. NGFs-induced signaling was TrkA mediated as evident by inhibition with the TrkA antagonist K252a. NGF and its analogues-induced signaling in ECs were characterized by a transient effect in contrast to a prolonged stimulation in neuronal cells. The potency of mouse, cobra and viper NGFs to induce Erk1/2 phosphorylation in ECs was higher than in neurons. In ECs, mNGF exhibited the highest efficacy of stimulation of Erk1/2 phosphorylation, followed by viper and cobra NGFs. The efficacy of stimulation of Erk1/2 phosphorylation measured with neurons was opposite from that in ECs. NGF-induced temporal signaling differences between ECs and neurons may explain the dual vascular and neurotrophic effects of this growth factor.
Collapse
Affiliation(s)
- Shimon Lecht
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
46
|
Patti F, Amato MP, Bastianello S, Caniatti L, Di Monte E, Ferrazza P, Goretti B, Gallo P, Brescia Morra V, Lo Fermo S, Picconi O, Tola MR, Trojano M. Effects of immunomodulatory treatment with subcutaneous interferon beta-1a oncognitive decline in mildly disabled patients with relapsing—remitting multiple sclerosis. Mult Scler 2009; 16:68-77. [DOI: 10.1177/1352458509350309] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The objective of this study was to assess the effects of subcutaneous (sc) interferon beta-1a (IFNβ-1a) on cognition in mildly disabled patients with relapsing—remitting multiple sclerosis (RRMS). Patients aged 18—50 years with RRMS (McDonald criteria; Expanded Disability Status Scale score ≤4.0) were assigned IFNβ therapy at the physician’s discretion and underwent standardized magnetic resonance imaging, neurological examination and neuropsychological testing at the baseline and regular intervals for up to three years. This analysis included 459 patients who received sc IFNβ-1a (44 mcg: n = 236; 22 mcg: n = 223; three-year follow up was available for 318 patients). The hazard ratio for cognitive impairment over three years (44 mcg versus 22 mcg) was 0.68 (95% confidence interval [CI]: 0.480—0.972), suggesting a 32% lower risk with the higher dose treatment. At year 3, the proportion of patients who were cognitively impaired increased slightly from 23.5% at the baseline to 24.8% in the IFNβ-1a 22 mcg treatment group, but remained stable at 15.2% in the IFNβ-1a 44 mcg treatment group. The proportion of patients with cognitive impairment at year 3 was significantly higher in the 22 mcg group than in the 44 mcg group (P = 0.03), although a trend was also seen at the baseline (P = 0.058). Multivariate logistic regression (corrected for baseline cognitive deficits) indicated that treatment with the higher dose of IFNβ-1a was predictive of lower cognitive impairment at three years (odds ratio: 0.51, 95% CI: 0.26—0.99) compared with the lower dose of IFNβ-1a. These findings suggest that sc IFNβ-1a may have dose-dependent cognitive benefits in mildly disabled patients with RRMS, and may support early initiation of high-dose IFNβ-1a treatment.
Collapse
Affiliation(s)
- F. Patti
- Multiple Sclerosis Centre Sicilia Region, First Neurology Clinic, University Hospital Catania, Catania, Italy,
| | - MP Amato
- Department of Neurology, University of Florence, Florence, Italy
| | - S. Bastianello
- Neurological Institute, IRCCS Fondazione C. Mondino, Pavia, Italy
| | - L. Caniatti
- U.O. Neurology, Department of Neuroscience and Rehabilitation, Azienda Universita-Ospedale, S. Anna, Ferrara, Italy
| | - E. Di Monte
- Department of Neurological and Psychiatric Sciences, University of Bari, Bari, Italy
| | - P. Ferrazza
- Opera CRO Scientific Advisor Board, Genoa, Italy/Neuromed Clinical Department, Pozzilli, Italy
| | - B. Goretti
- Department of Neurology, University of Florence, Florence, Italy
| | - P. Gallo
- Multiple Sclerosis Centre Veneto Region, First Neurology Clinic, University Hospital, Padova, Padova, Italy
| | | | - S. Lo Fermo
- Multiple Sclerosis Centre Sicilia Region, First Neurology Clinic, University Hospital Catania, Catania, Italy
| | - O. Picconi
- Public Health Agency of Regione Lazio, Rome, Italy
| | - MR Tola
- U.O. Neurology, Department of Neuroscience and Rehabilitation, Azienda Universita-Ospedale, S. Anna, Ferrara, Italy
| | - M. Trojano
- Department of Neurological and Psychiatric Sciences, University of Bari, Bari, Italy
| | | |
Collapse
|
47
|
De Santi L, Annunziata P, Sessa E, Bramanti P. Brain-derived neurotrophic factor and TrkB receptor in experimental autoimmune encephalomyelitis and multiple sclerosis. J Neurol Sci 2009; 287:17-26. [DOI: 10.1016/j.jns.2009.08.057] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 08/24/2009] [Accepted: 08/27/2009] [Indexed: 01/31/2023]
|
48
|
Havrdova E, Zivadinov R, Krasensky J, Dwyer MG, Novakova I, Dolezal O, Ticha V, Dusek L, Houzvickova E, Cox JL, Bergsland N, Hussein S, Svobodnik A, Seidl Z, Vaneckova M, Horakova D. Randomized study of interferon beta-1a, low-dose azathioprine, and low-dose corticosteroids in multiple sclerosis. Mult Scler 2009; 15:965-76. [DOI: 10.1177/1352458509105229] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Studies evaluating interferon beta (IFNβ) for multiple sclerosis (MS) showed only partial efficacy. In many patients, IFNβ does not halt relapses or disability progression. One strategy to potentially enhance efficacy is to combine IFNβ with classical immunosuppressive agents, such as azathioprine (AZA) or corticosteroids, commonly used for other autoimmune disorders. Objective The Avonex–Steroids–Azathioprine study was placebo-controlled trial and evaluated efficacy of IFNβ-1a alone and combined with low-dose AZA alone or low-dose AZA and low-dose corticosteroids as initial therapy. Methods A total of 181 patients with relapsing–remitting MS (RRMS) were randomized to receive IFNβ-1a 30 μg intramuscularly (IM) once weekly, IFNβ-1a 30 μg IM once weekly plus AZA 50 mg orally once daily, or IFNβ-1a 30 μg IM once weekly plus AZA 50 mg orally once daily plus prednisone 10 mg orally every other day. The primary end point was annualized relapse rate (ARR) at 2 years. Patients were eligible for enrollment in a 3-year extension. Results At 2 years, adjusted ARR was 1.05 for IFNβ-1a, 0.91 for IFNβ-1a plus AZA, and 0.73 for combination. The cumulative probability of sustained disability progression was 16.8% for IFNβ-1a, 20.7% for IFNβ-1a plus AZA, and 17.5% for combination. There were no statistically significant differences among groups for either measure at 2 and 5 years. Percent T2 lesion volume change at 2 years was significantly lower for combination (+14.5%) versus IFNβ-1a alone (+30.3%, P < 0.05). Groups had similar safety profiles. Conclusion In IFNβ-naïve patients with early active RRMS, combination treatment did not show superiority over IFNβ-1a monotherapy.
Collapse
Affiliation(s)
- E Havrdova
- Department of Neurology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| | - R Zivadinov
- Department of Neurology, Buffalo Neuroimaging Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - J Krasensky
- Department of Radiology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| | - MG Dwyer
- Department of Neurology, Buffalo Neuroimaging Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - I Novakova
- Department of Neurology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| | - O Dolezal
- Department of Neurology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| | - V Ticha
- Department of Neurology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| | - L Dusek
- Center of Biostatistics and Analyses, Faculty of Medicine and Faculty of Sciences, Masaryk University, Brno, Czech Republic
| | - E Houzvickova
- Department of Neurology, Charles University in Prague, Second Faculty of Medicine, Prague, Czech Republic
| | - JL Cox
- Department of Neurology, Buffalo Neuroimaging Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - N Bergsland
- Department of Neurology, Buffalo Neuroimaging Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - S Hussein
- Department of Neurology, Buffalo Neuroimaging Analysis Center, State University of New York at Buffalo, Buffalo, NY, USA
| | - A Svobodnik
- Center of Biostatistics and Analyses, Faculty of Medicine and Faculty of Sciences, Masaryk University, Brno, Czech Republic
| | - Z Seidl
- Department of Radiology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| | - M Vaneckova
- Department of Radiology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| | - D Horakova
- Department of Neurology, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| |
Collapse
|
49
|
Mirowska-Guzel D. The role of neurotrophic factors in the pathology and treatment of multiple sclerosis. Immunopharmacol Immunotoxicol 2009; 31:32-8. [PMID: 18792835 DOI: 10.1080/08923970802379819] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of primary autoimmune origin with essential component of subsequent axonal degeneration. It has been found that inflammatory cells crucial for MS pathogenesis are able to release neurotrophic factors (NFs). Thus the concept of neuroprotective effect of inflammation has arisen. Over recent years, increasing number of studies has revealed that NFs play an important role in MS and its animal model - experimental autoimmune encephalomyelitis (EAE). In the current review the evidence for the role of NFs in MS pathogenesis the same as their remarkable potential in MS therapy is presented.
Collapse
|
50
|
Vercellino M, Romagnolo A, Mattioda A, Masera S, Piacentino C, Merola A, Chiò A, Mutani R, Cavalla P. Multiple sclerosis relapses: a multivariable analysis of residual disability determinants. Acta Neurol Scand 2009; 119:126-30. [PMID: 18684216 DOI: 10.1111/j.1600-0404.2008.01076.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Recovery from multiple sclerosis (MS) relapses is variable. The factors influencing persistence of residual disability (RD) after a relapse are still to be thoroughly elucidated. AIMS OF STUDY To assess RD after MS relapses and to define the factors associated with persistence of RD. METHODS Data were retrospectively collected for all relapses in a population of relapsing-remitting MS patients during 3 years. Relapse severity and RD after 1 year were calculated on Expanded Disability Status Scale basis. A multivariable analysis for factors influencing RD and relapse severity was performed (variables: age, gender, disease duration, oligoclonal bands, relapse severity, monosymptomatic/polysymptomatic relapse, immunomodulating treatment, incomplete recovery at 1 month). RESULTS A total of 174 relapses were assessed. RD after 1 year was observed in 54.5% of the relapses. Higher risk of RD was associated with occurrence of a severe relapse (P = 0.024). Incomplete recovery at 1 month was highly predictive of RD at 1 year (P < 0.0001). Risk of a severe relapse was associated with age <or= 30 years (P = 0.025) and inversely associated with the use of immunomodulating treatment (P = 0.006). CONCLUSIONS Incomplete recovery at 1 month is a predictor of long-term persistence of RD. Higher relapse severity is associated with higher risk of RD. Risk of severe relapses is lower in patients treated with immunomodulating drugs.
Collapse
Affiliation(s)
- M Vercellino
- Department of Neuroscience, University of Turin, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|