1
|
Neuroinflammation Is Associated with GFAP and sTREM2 Levels in Multiple Sclerosis. Biomolecules 2022; 12:biom12020222. [PMID: 35204724 PMCID: PMC8961656 DOI: 10.3390/biom12020222] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 01/22/2023] Open
Abstract
Background: Astrocytes and microglia play an important role in the inflammatory process of multiple sclerosis (MS). We investigated the associations between the cerebrospinal fluid (CSF) levels of glial fibrillary acid protein (GFAP) and soluble triggering receptors expressed on myeloid cells-2 (sTREM-2), inflammatory molecules, and clinical characteristics in a group of patients with relapsing-remitting MS (RRMS). Methods: Fifty-one RRMS patients participated in the study. Clinical evaluation and CSF collection were performed at the time of diagnosis. The CSF levels of GFAP, sTREM-2, and of a large set of inflammatory and anti-inflammatory molecules were determined. MRI structural measures (cortical thickness, T2 lesion load, cerebellar volume) were examined. Results: The CSF levels of GFAP and sTREM-2 showed significant correlations with inflammatory cytokines IL-8, G-CSF, and IL-5. Both GFAP and sTREM-2 CSF levels positively correlated with age at diagnosis. GFAP was also higher in male MS patients, and was associated with an increased risk of MS progression, as evidenced by higher BREMS at the onset. Finally, a negative association was found between GFAP CSF levels and cerebellar volume in RRMS at diagnosis. Conclusions: GFAP and sTREM-2 represent suitable biomarkers of central inflammation in MS. Our results suggest that enhanced CSF expression of GFAP may characterize patients with a higher risk of progression.
Collapse
|
2
|
Smagina IV, Elchaninova SA, Palashchenko AS, Galaktionova LP. [Pathological and protective effects of tumor necrosis factor-alpha in multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 119:14-20. [PMID: 31934984 DOI: 10.17116/jnevro20191191014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The immunomodulatory cytokine tumor necrosis factor-alpha (TNF-α) is involved in the regulation of both physiological and pathological processes in the central nervous system (CNS). The effects of TNF-α on CNS reported in clinical trials and experimental studies, evidence of involvement of this cytokine in the pathogenesis of multiple sclerosis are analyzed. Possible causes of failures of non-selective pharmacological inhibition of TNF-α effects in MS are considered in view of current concepts on mechanisms of TNF-α action.
Collapse
Affiliation(s)
- I V Smagina
- Altai State Medical University, Barnaul, Russia; Regional Clinical Hospital, Barnaul, Russia
| | | | - A S Palashchenko
- Altai State Medical University, Barnaul, Russia; Regional Clinical Hospital, Barnaul, Russia
| | | |
Collapse
|
3
|
Rolf L, Damoiseaux J, Hupperts R, Huitinga I, Smolders J. Network of nuclear receptor ligands in multiple sclerosis: Common pathways and interactions of sex-steroids, corticosteroids and vitamin D3-derived molecules. Autoimmun Rev 2016; 15:900-10. [DOI: 10.1016/j.autrev.2016.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/08/2016] [Indexed: 01/12/2023]
|
4
|
Pennisi M, Russo G, Motta S, Pappalardo F. Agent based modeling of the effects of potential treatments over the blood-brain barrier in multiple sclerosis. J Immunol Methods 2015; 427:6-12. [PMID: 26343337 DOI: 10.1016/j.jim.2015.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/15/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis is a disease of the central nervous system that involves the destruction of the insulating sheath of axons, causing severe disabilities. Since the etiology of the disease is not yet fully understood, the use of novel techniques that may help to understand the disease, to suggest potential therapies and to test the effects of candidate treatments is highly advisable. To this end we developed an agent based model that demonstrated its ability to reproduce the typical oscillatory behavior observed in the most common form of multiple sclerosis, relapsing-remitting multiple sclerosis. The model has then been used to test the potential beneficial effects of vitamin D over the disease. Many scientific studies underlined the importance of the blood-brain barrier and of the mechanisms that influence its permeability on the development of the disease. In the present paper we further extend our previously developed model with a mechanism that mimics the blood-brain barrier behavior. The goal of our work is to suggest the best strategies to follow for developing new potential treatments that intervene in the blood-brain barrier. Results suggest that the best treatments should potentially prevent the opening of the blood-brain barrier, as treatments that help in recovering the blood-brain barrier functionality could be less effective.
Collapse
Affiliation(s)
- Marzio Pennisi
- Department of Mathematics and Computer Science, University of Catania, 95125 Catania, Italy
| | - Giulia Russo
- Department of Drug Science, University of Catania, 95125 Catania, Italy
| | - Santo Motta
- Department of Mathematics and Computer Science, University of Catania, 95125 Catania, Italy
| | | |
Collapse
|
5
|
Mancuso R, Hernis A, Agostini S, Rovaris M, Caputo D, Fuchs D, Clerici M. Indoleamine 2,3 Dioxygenase (IDO) Expression and Activity in Relapsing-Remitting Multiple Sclerosis. PLoS One 2015; 10:e0130715. [PMID: 26110930 PMCID: PMC4482492 DOI: 10.1371/journal.pone.0130715] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/23/2015] [Indexed: 01/09/2023] Open
Abstract
Background Interferon gamma (IFN-γ) production induces the transcription of indoleamine 2,3 dioxygenase (IDO) resulting in the reduction of T-cell activation and proliferation through the depletion of tryptophan and the elicitation of Treg lymphocytes. IDO was shown to be involved in the pathogenesis of autoimmune diseases; we investigated whether changes in IDO gene expression and activity could be indicative of onset of relapse in multiple sclerosis (MS) patients. Methods IDO and interferon-γ (IFN-γ) gene expression, serum IDO activity (Kynurenine/Tryptophan ratio) and serum neopterin concentration – a protein released by macrophages upon IFN-γ stimulation – were measured in 51 individuals: 36 relapsing remitting (RR)-MS patients (21 in acute phase -AMS, 15 in stable phase -SMS) and 15 healthy controls (HC). PBMCs samples in AMS patients were collected before (BT-AMS) and during glucocorticoids-based therapy (DT-AMS). Results IDO expression was increased and IFN-γ was decreased (p<0.001) in BT-AMS compared to SMS patients. Glucocorticoids-induced disease remission resulted in a significant reduction of IDO and IFN-γ gene expression, IDO catalytic activity (p<0.001). Serum neopterin concentration followed the same trend as IDO expression and activity. Conclusions Measurement of IDO gene expression and activity in blood could be a useful marker to monitor the clinical course of RR-MS. Therapeutic interventions modulating IDO activity may be beneficial in MS.
Collapse
Affiliation(s)
- Roberta Mancuso
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
- * E-mail:
| | - Ambra Hernis
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Simone Agostini
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Marco Rovaris
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Domenico Caputo
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innrain 80–82, A-6020, Innsbruck, Austria
| | - Mario Clerici
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
- Department of Physiopathology and Transplantation, University of Milano, Via Fratelli Cervi 93, 20090, Segrate, Milano, Italy
| |
Collapse
|
6
|
Jean-Gilles L, Braitch M, Latif ML, Aram J, Fahey AJ, Edwards LJ, Robins RA, Tanasescu R, Tighe PJ, Gran B, Showe LC, Alexander SP, Chapman V, Kendall DA, Constantinescu CS. Effects of pro-inflammatory cytokines on cannabinoid CB1 and CB2 receptors in immune cells. Acta Physiol (Oxf) 2015; 214:63-74. [PMID: 25704169 PMCID: PMC4669958 DOI: 10.1111/apha.12474] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 10/17/2014] [Accepted: 02/16/2015] [Indexed: 12/30/2022]
Abstract
Aims To investigate the regulation of cannabinoid receptors CB1 and CB2 on immune cells by pro‐inflammatory cytokines and its potential relevance to the inflammatory neurological disease, multiple sclerosis (MS). CB1 and CB2 signalling may be anti‐inflammatory and neuroprotective in neuroinflammatory diseases. Cannabinoids can suppress inflammatory cytokines but the effects of these cytokines on CB1 and CB2 expression and function are unknown. Methods Immune cells from peripheral blood were obtained from healthy volunteers and patients with MS. Expression of CB1 and CB2mRNA in whole blood cells, peripheral blood mononuclear cells (PBMC) and T cells was determined by quantitative real‐time polymerase chain reaction (qRT‐PCR). Expression of CB1 and CB2 protein was determined by flow cytometry. CB1 and CB2 signalling in PBMC was determined by Western blotting for Erk1/2. Results Pro‐inflammatory cytokines IL‐1β, IL‐6 and TNF‐α (the latter likely NF‐κB dependently) can upregulate CB1 and CB2 on human whole blood and peripheral blood mononuclear cells (PBMC). We also demonstrate upregulation of CB1 and CB2 and increased IL‐1β, IL‐6 and TNF‐α mRNA in blood of patients with MS compared with controls. Conclusion The levels of CB1 and CB2 can be upregulated by inflammatory cytokines, which can explain their increase in inflammatory conditions including MS.
Collapse
Affiliation(s)
- L. Jean-Gilles
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
| | - M. Braitch
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
| | - M. L. Latif
- Division of Neuroscience; School of Life Sciences; University of Nottingham; Nottingham UK
| | - J. Aram
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
| | - A. J. Fahey
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
| | - L. J. Edwards
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
| | - R. A. Robins
- Division of Immunity; School of Life Sciences; University of Nottingham; Nottingham UK
| | - R. Tanasescu
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
- Department of Neurology; Colentina Hospital; University of Medicine and Pharmacy Carol Davila; Bucharest Romania
| | - P. J. Tighe
- Division of Immunity; School of Life Sciences; University of Nottingham; Nottingham UK
| | - B. Gran
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
| | | | - S. P. Alexander
- Division of Neuroscience; School of Life Sciences; University of Nottingham; Nottingham UK
| | - V. Chapman
- Division of Neuroscience; School of Life Sciences; University of Nottingham; Nottingham UK
| | - D. A. Kendall
- Division of Neuroscience; School of Life Sciences; University of Nottingham; Nottingham UK
| | - C. S. Constantinescu
- Division of Clinical Neuroscience; School of Medicine; University of Nottingham; Nottingham UK
| |
Collapse
|
7
|
Siebert JR, Conta Steencken A, Osterhout DJ. Chondroitin sulfate proteoglycans in the nervous system: inhibitors to repair. BIOMED RESEARCH INTERNATIONAL 2014; 2014:845323. [PMID: 25309928 PMCID: PMC4182688 DOI: 10.1155/2014/845323] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 12/14/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are widely expressed in the normal central nervous system, serving as guidance cues during development and modulating synaptic connections in the adult. With injury or disease, an increase in CSPG expression is commonly observed close to lesioned areas. However, these CSPG deposits form a substantial barrier to regeneration and are largely responsible for the inability to repair damage in the brain and spinal cord. This review discusses the role of CSPGs as inhibitors, the role of inflammation in stimulating CSPG expression near site of injury, and therapeutic strategies for overcoming the inhibitory effects of CSPGs and creating an environment conducive to nerve regeneration.
Collapse
Affiliation(s)
- Justin R. Siebert
- Lake Erie College of Osteopathic Medicine at Seton Hill, 20 Seton Hill Drive, Greensburg, PA 15601, USA
| | - Amanda Conta Steencken
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Donna J. Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
8
|
Angiogenesis in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol Commun 2014; 2:84. [PMID: 25047180 PMCID: PMC4149233 DOI: 10.1186/s40478-014-0084-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/09/2014] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis, the formation of new vessels, is found in Multiple Sclerosis (MS) demyelinating lesions following Vascular Endothelial Growth Factor (VEGF) release and the production of several other angiogenic molecules. The increased energy demand of inflammatory cuffs and damaged neural cells explains the strong angiogenic response in plaques and surrounding white matter. An angiogenic response has also been documented in an experimental model of MS, experimental allergic encephalomyelitis (EAE), where blood–brain barrier disruption and vascular remodelling appeared in a pre-symptomatic disease phase. In both MS and EAE, VEGF acts as a pro-inflammatory factor in the early phase but its reduced responsivity in the late phase can disrupt neuroregenerative attempts, since VEGF naturally enhances neuron resistance to injury and regulates neural progenitor proliferation, migration, differentiation and oligodendrocyte precursor cell (OPC) survival and migration to demyelinated lesions. Angiogenesis, neurogenesis and oligodendroglia maturation are closely intertwined in the neurovascular niches of the subventricular zone, one of the preferential locations of inflammatory lesions in MS, and in all the other temporary vascular niches where the mutual fostering of angiogenesis and OPC maturation occurs. Angiogenesis, induced either by CNS inflammation or by hypoxic stimuli related to neurovascular uncoupling, appears to be ineffective in chronic MS due to a counterbalancing effect of vasoconstrictive mechanisms determined by the reduced axonal activity, astrocyte dysfunction, microglia secretion of free radical species and mitochondrial abnormalities. Thus, angiogenesis, that supplies several trophic factors, should be promoted in therapeutic neuroregeneration efforts to combat the progressive, degenerative phase of MS.
Collapse
|
9
|
A comparative study of experimental mouse models of central nervous system demyelination. Gene Ther 2014; 21:599-608. [PMID: 24718267 PMCID: PMC4047154 DOI: 10.1038/gt.2014.33] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/09/2014] [Accepted: 01/22/2014] [Indexed: 01/31/2023]
Abstract
Several mouse models of multiple sclerosis (MS) are now available. We have established a mouse model, in which ocular infection with a recombinant HSV-1 that expresses murine IL-2 constitutively (HSV-IL-2) causes CNS demyelination in different strains of mice. This model differs from most other models in that it represents a mixture of viral and immune triggers. In the present study, we directly compared MOG35–55, MBP35–47, and PLP190–209 models of EAE with our HSV-IL-2-induced MS model. Mice with HSV-IL-2-induced and MOG-induced demyelinating diseases demonstrated a similar pattern and distribution of demyelination in their brain, spinal cord, and optic nerves. In contrast, no demyelination was detected in the optic nerves of MBP- and PLP-injected mice. IFN-β injections significantly reduced demyelination in brains of all groups, in the spinal cords of the MOG and MBP groups, and completely blocked it in the spinal cords of the PLP and HSV-IL-2 groups as well as in optic nerves of MOG and HSV-IL-2 groups. In contrast to IFN-β treatment, IL-12p70 protected the HSV-IL-2 group from demyelination, while IL-4 was not effective at all in preventing demyelination. MOG-injected mice showed clinical signs of paralysis and disease-related mortality whereas mice in the other treatment groups did not. Collectively, the results indicate that the HSV-IL-2 model and the MOG model complement each other and, together, provide unique insights into the heterogeneity of human MS.
Collapse
|
10
|
Upregulation of IL-17, but not of IL-9, in circulating cells of CIS and relapsing MS patients. Impact of corticosteroid therapy on the cytokine network. J Neuroimmunol 2012; 243:73-80. [DOI: 10.1016/j.jneuroim.2011.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 12/07/2011] [Accepted: 12/12/2011] [Indexed: 12/18/2022]
|
11
|
Nyati KK, Prasad KN, Kharwar NK, Soni P, Husain N, Agrawal V, Jain AK. Immunopathology and Th1/Th2 immune response of Campylobacter jejuni-induced paralysis resembling Guillain-Barré syndrome in chicken. Med Microbiol Immunol 2011; 201:177-87. [PMID: 22102098 DOI: 10.1007/s00430-011-0220-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Indexed: 01/08/2023]
Abstract
Immunopathogenesis of Campylobacter jejuni-associated Guillain-Barré syndrome (GBS) is not yet well established probably due to lack of experimental model. Therefore, we studied the Th1/Th2 immune response and pathological changes in C. jejuni-induced chicken model for GBS. C. jejuni (5 × 10(9) CFU/ml) and placebo were fed to 30 chickens each. Stools of all birds were negative for C. jejuni by culture and PCR before experiment. The birds were regularly assessed for disease symptoms up to 30 days. Sciatic nerves from all chickens were examined at 5 days intervals by histopathology and immunohistochemistry, and also for the expression of Th1/Th2 cytokines. Twenty-two chickens (73.3%) developed diarrhea after C. jejuni infection; 18 (60.0%) experimental chickens developed GBS-like paralytic neuropathy. Pathology in the sciatic nerves of these chickens included perinodal and/or patchy demyelination, perivascular focal lymphocytic infiltration, myelin swelling and presence of macrophages within the nerve fibers on 10th-20th post-infection day (PID). Cytokines (IFN-γ, IL-1β, TNF-α, IL-6 and IL-2) were elevated in early phase (5th-15th PID) and TGF-β2, IL-10 and IL-4 in the recovery phase (25th-30th PID) of the disease. The study provides evidence that C. jejuni infection in the chicken can provide an experimental animal model of GBS.
Collapse
Affiliation(s)
- Kishan K Nyati
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India
| | | | | | | | | | | | | |
Collapse
|
12
|
Lindquist S, Hassinger S, Lindquist JA, Sailer M. The balance of pro-inflammatory and trophic factors in multiple sclerosis patients: effects of acute relapse and immunomodulatory treatment. Mult Scler 2011; 17:851-66. [DOI: 10.1177/1352458511399797] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: In multiple sclerosis inflammation is primarily injurious to the central nervous system, but its therapeutic suppression might inhibit repair-promoting factors. Objectives: We aimed at better describing the complexity of biological effects during an acute relapse and analysed the effects of intervention with high-dose i.v. glucocorticoids and immunomodulatory treatment with interferon-beta (IFNβ). Methods: We studied the intracellular expression levels of the pro-inflammatory mediators tumour necrosis factor alpha (TNFα) and inducible nitric oxide synthase (iNOS) together with the neurotrophins ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) in freshly isolated peripheral blood mononuclear cells of multiple sclerosis patients during an acute relapse, after intervention with i.v. methylprednisolone and at baseline, using a highly quantitative flow-cytometric approach. Results: We demonstrated the expression of CNTF in human leucocytes. We showed that CNTF levels differed in acutely relapsing multiple sclerosis patients compared with controls and increased after corticosteroid treatment. CNTF can counteract the toxicity of TNFα towards oligodendrocytes and we found TNFα increased during acute relapses. Following corticosteroids, neither TNFα nor iNOS expression was reduced. Levels of BDNF were not affected by glucocorticoids, but increased during IFNβ therapy. However, IFNβ also increased the expression of iNOS and major histocompatibility complex class I (MHC-I), underlining its immunomodulatory potential. Conclusions: Multiple sclerosis patients might benefit from reparative, and not solely from anti-inflammatory, effects of glucocorticoids. Interactive effects of glucocorticoid- and IFNβ-treatment need to be considered to improve neuroprotection and remyelination resulting from immunomodulatory treatment.
Collapse
Affiliation(s)
- Sabine Lindquist
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | - Sarah Hassinger
- University Clinic for Neurology, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jonathan A Lindquist
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke-University, Magdeburg, Germany
- Co-senior authors
| | - Michael Sailer
- University Clinic for Neurology, Otto-von-Guericke-University, Magdeburg, Germany
- Centre for Neurological Rehabilitation, Magdeburg, Germany
- Co-senior authors
| |
Collapse
|
13
|
Larochelle C, Alvarez JI, Prat A. How do immune cells overcome the blood-brain barrier in multiple sclerosis? FEBS Lett 2011; 585:3770-80. [PMID: 21550344 DOI: 10.1016/j.febslet.2011.04.066] [Citation(s) in RCA: 275] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 11/17/2022]
Abstract
The presence of the blood-brain barrier (BBB) restricts the movement of soluble mediators and leukocytes from the periphery to the central nervous system (CNS). Leukocyte entry into the CNS is nonetheless an early event in multiple sclerosis (MS), an inflammatory disorder of the CNS. Whether BBB dysfunction precedes immune cell infiltration or is the consequence of perivascular leukocyte accumulation remains enigmatic, but leukocyte migration modifies BBB permeability. Immune cells of MS subjects express inflammatory cytokines, reactive oxygen species (ROS) and enzymes that can facilitate their migration to the CNS by influencing BBB function, either directly or indirectly. In this review, we describe how immune cells from the peripheral blood overcome the BBB and promote CNS inflammation in MS through BBB disruption.
Collapse
Affiliation(s)
- Catherine Larochelle
- Neuroimmunology Research Laboratory, Center of Excellence in Neuromics, CRCHUM, Notre-Dame Hospital, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
14
|
Zandian M, Mott KR, Allen SJ, Dumitrascu O, Kuo JZ, Ghiasi H. Use of cytokine immunotherapy to block CNS demyelination induced by a recombinant HSV-1 expressing IL-2. Gene Ther 2011; 18:734-42. [PMID: 21412284 PMCID: PMC3118258 DOI: 10.1038/gt.2011.32] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We previously have described a model of multiple sclerosis (MS) in which constitutive expression of murine interleukin (IL)-2 by herpes simplex virus type 1 (HSV-1) (HSV-IL-2) causes central nervous system (CNS) demyelination in different strains of mice. In the current study, we investigated whether this HSV-IL-2-induced demyelination can be blocked using recombinant viruses expressing different cytokines or by injection of plasmid DNA. We have found that coinfection of HSV-IL-2-infected mice with recombinant viruses expressing IL-12p35, IL-12p40 or IL-12p35+IL-12p40 did not block the CNS demyelination, and that coinfection with a recombinant virus expressing interferon (IFN)-γ exacerbated it. In contrast, coinfection with a recombinant virus expressing IL-4 reduced demyelination, whereas coinfection of HSV-IL-2-infected mice with a recombinant HSV-1 expressing the IL-12 heterodimer (HSV-IL-12p70) blocked the CNS demyelination in a dose-dependent manner. Similarly, injection of IL-12p70 DNA blocked HSV-IL-2-induced CNS demyelination in a dose-dependent manner and injection of IL-35 DNA significantly reduced CNS demyelination. Injection of mice with IL-12p35 DNA, IL-12p40 DNA, IL-12p35+IL-12p40 DNA or IL-23 DNA did not have any effect on HSV-IL-2-induced demyelination, whereas injection of IL-27 DNA increased the severity of the CNS demyelination in the HSV-IL-2-infected mice. This study demonstrates for the first time that IL-12p70 can block HSV-IL-2-induced CNS demyelination and that IL-35 can also reduce this demyelination, whereas IFN-γ and IL-27 exacerbated the demyelination in the CNS of the HSV-IL-2-infected mice. Our results suggest a potential role for IL-12p70 and IL-35 signaling in the inhibition of HSV-IL-2-induced immunopathology by preventing development of autoaggressive T cells.
Collapse
Affiliation(s)
- M Zandian
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery and Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Madia F, Frisullo G, Nociti V, Conte A, Luigetti M, Del Grande A, Patanella AK, Iorio R, Tonali PA, Batocchi AP, Sabatelli M. pSTAT1, pSTAT3, and T-bet as markers of disease activity in chronic inflammatory demyelinating polyradiculoneuropathy. J Peripher Nerv Syst 2009; 14:107-17. [PMID: 19691533 DOI: 10.1111/j.1529-8027.2009.00220.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is considered an auto-immune disorder. We evaluated expression of pSTAT1, T-bet, and pSTAT3 in circulating T-cells, B-cells, and monocytes and spontaneous production of interleukin-17 (IL17), interferon-gamma (IFN gamma), and interleukin-10 (IL10) by peripheral blood mononuclear cells (PBMCs) from 14 active CIDP patients compared with 6 patients with long-lasting remission and 20 controls. Active disease patients showed higher pSTAT1, T-bet, and pSTAT3 in CD4(+) T-cells than controls (p < 0.001, p = 0.0002, p = 0.0097, respectively) and remission patients (p < 0.001, p = 0.0036, p = 0.0008, respectively). pSTAT1, T-bet, and pSTAT3 were also higher in monocytes from active CIDP patients than controls (p = 0.0011, p = 0.0041, p = 0.0413, respectively) and remission patients (p = 0.0073, p = 0.0274, p = 0.0251, respectively). Moreover in CD8(+) T-cells, pSTAT3 expression was higher in active CIDP patients than in remission patients (p = 0.0345) and in controls (p = 0.0023). IL17 and IFN gamma production were significantly higher in active CIDP patients than in controls (p < 0.0395, p = 0.0010, respectively); IFN gamma levels were higher also in remission CIDP patients (p = 0.0073). IL10 levels were higher in active phase patients than in controls (p = 0.0334). Our data suggest that pSTAT1, T-bet, and pSTAT3 can be considered putative markers of disease activity and potential targets for specific therapies.
Collapse
Affiliation(s)
- Francesca Madia
- Institute of Neurology, Department of Neuroscience, Catholic University, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Khademi M, Bornsen L, Rafatnia F, Andersson M, Brundin L, Piehl F, Sellebjerg F, Olsson T. The effects of natalizumab on inflammatory mediators in multiple sclerosis: prospects for treatment-sensitive biomarkers. Eur J Neurol 2009; 16:528-36. [PMID: 19220425 DOI: 10.1111/j.1468-1331.2009.02532.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Natalizumab affects systemic cytokine expressions and clinical course in relapsing-remitting multiple sclerosis (RRMS). We analyzed levels of inflammatory cytokines in cerebrospinal fluid (CSF) cells and peripheral blood mononuclear cells (PBMCs), levels of matrix metalloproteinase (MMP)-9 and osteopontin (OPN) in CSF, and clinical outcome measures in 22 natalizumab-treated RRMS patients. METHODS mRNA levels of cytokines in cells were detected with real-time RT-PCR. Protein levels of OPN and MMP-9 were measured by ELISA. RESULTS Natalizumab reduced CSF cell counts (P < 0.0001). Tumor necrosis factor (TNF) and interferon-gamma (IFN-gamma) mRNAs were significantly increased in PBMCs. In contrast, expressions of IFN-gamma and interleukin (IL)-23 were decreased but IL-10 increased in the CSF cells. OPN and MMP-9 were reduced in the CSF. Patients being in remission at baseline showed the same deviations of mediators as those in relapse after natalizumab treatment. The open label clinical outcome measures were either stable or improved during therapy. CONCLUSIONS Natalizumab attenuates pro-inflammatory mediators intrathecally and the reduced pro-inflammatory milieu may allow increased production of the anti-inflammatory mediator IL-10. The increased systemic cytokines may impede the improvement of certain clinical measures like fatigue. The affected mediators seem to be sensitive to an immune-modifying treatment which could be used as biomarkers for this therapy.
Collapse
Affiliation(s)
- M Khademi
- Neuroimmunology Unit, CMM, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
The persistency of high levels of pSTAT3 expression in circulating CD4+ T cells from CIS patients favors the early conversion to clinically defined multiple sclerosis. J Neuroimmunol 2008; 205:126-34. [DOI: 10.1016/j.jneuroim.2008.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 08/28/2008] [Accepted: 09/05/2008] [Indexed: 12/17/2022]
|
19
|
Núñez C, Dema B, Cénit MC, Polanco I, Maluenda C, Arroyo R, de las Heras V, Bartolomé M, de la Concha EG, Urcelay E, Martínez A. IL23R: a susceptibility locus for celiac disease and multiple sclerosis? Genes Immun 2008; 9:289-93. [PMID: 18368064 DOI: 10.1038/gene.2008.16] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent studies have shown association of the IL23R gene with inflammatory bowel disease, psoriasis and ankylosing spondylitis. We aimed at studying the involvement of IL23R in celiac disease (CD) and multiple sclerosis (MS). We performed a case-control study including 598 patients with CD, 414 with MS and 546 healthy controls, all of them white Spaniards. All samples were genotyped for two single nucleotide polymorphisms: rs7517847 and rs11209026 (Arg381Gln). Statistical analyses were performed using chi(2-)tests or the Fisher's exact test. The minor allele (Gln) of the coding variant Arg381Gln was significantly increased in CD and MS patients when compared to controls (8% in CD vs 6% in controls, P=0.02; 9% in MS, P=0.006). In MS, a stronger effect was observed in patients showing primary-progressive disease (16%, P=0.004). Moreover, heterozygotes for rs7517847 were significantly increased in this group of MS patients (81% in MS vs 48% in controls, P=0.0002). In conclusion, contrary to what has been described previously, the less frequent allele of the functional polymorphism Arg381Gln (rs11209026) seems to be increasing susceptibility to CD and MS, although in this last group of patients a stronger effect is observed in patients affected of a primary-progressive form.
Collapse
Affiliation(s)
- C Núñez
- Servicio de Inmunología Clínica, Hospital Clínico San Carlos, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lopatinskaya L, Zwemmer J, Uitdehaag B, Lucas K, Polman C, Nagelkerken L. Mediators of apoptosis Fas and FasL predict disability progression in multiple sclerosis over a period of 10 years. Mult Scler 2007; 12:704-9. [PMID: 17262997 DOI: 10.1177/1352458506070826] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TNF-alpha, IL-12p35, IL-12p40, IL-4, IL-10, TGF-beta1, CCR3, CXCR3, CCR5, Fas and FasL mRNA levels in PBMC of 25 multiple sclerosis (MS) patients were quantified at baseline by real-time PCR according to a post-hoc study design. The baseline values of the different markers were analysed with respect to their correlation with the increase in disability over a period of 10 years. High levels of Fas mRNA were associated with a favourable disease course in relapsing-remitting (RR) MS (R2 = 0.74, P = 0.0001, n = 13), as measured by the Expanded Disability Status Scale (EDSS); high levels of FasL mRNA were associated with relatively mild disease progression (R2 = 0.86, P = 0.0001, n =12) in secondary progressive (SP) MS. These findings suggest that Fas-mediated apoptosis plays a major role in the mechanism underlying long-term disease progression in MS.:
Collapse
Affiliation(s)
- L Lopatinskaya
- Division of Biomedical Research, TNO Quality of Life, P.O. Box 2215, 2301 CE, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
21
|
Gilli F, Marnetto F, Caldano M, Sala A, Malucchi S, Capobianco M, Bertolotto A. Biological markers of interferon-beta therapy: comparison among interferon-stimulated genes MxA, TRAIL and XAF-1. Mult Scler 2006; 12:47-57. [PMID: 16459719 DOI: 10.1191/135248506ms1245oa] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Biological activity of interferon-beta (IFNbeta) can be assessed by measuring IFN-stimulated genes (ISGs). Among them, myxovirus resistance protein A (MxA) appears to have the highest specificity, but it has no role in the pathogenesis of multiple sclerosis (MS). To investigate the reliability of MxA as a biomarker, we compared its expression to that of two other ISGs: TNF-related apoptosis-inducing ligand (TRAIL) and X-linked inhibitor of apoptosis factor-1 (XAF-1). Both were shown to be involved in immunoregulatory mechanisms and might play a role in MS. Quantitative-PCR measurements were performed in peripheral blood mononuclear cells from 73 MS patients after short-term and long-term treatment with IFNbeta. A time-dependent response for multiple ISGs was observed in all patients after short-term treatment. In contrast, long-term treatment induced concurrent inhibition of ISGs in 12.3% (9/73) of patients, in whom neutralizing antibodies (NAbs) were detectable. Besides, 22% (16/73) of chronically treated patients showed a non-NAbs-related abrogation of TRAIL expression. In summary, 1) MxA expression was significantly higher than both TRAIL and XAF-1, and 2) MxA was the most sensitive gene to detect decreased bioavailability due to NAbs. These findings identify MxA as an appropriate biomarker for IFNbeta, although there is no evidence for a functional role of it in MS.
Collapse
Affiliation(s)
- F Gilli
- Centro di Riferimento Regionale Sclerosi Multipla (CReSM) & Neurobiologia Clinica, ASO S. Luigi Gonzaga, Orbassano, Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Javed A, Reder AT. Therapeutic role of beta-interferons in multiple sclerosis. Pharmacol Ther 2006; 110:35-56. [PMID: 16229894 DOI: 10.1016/j.pharmthera.2005.08.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2005] [Accepted: 08/23/2005] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS). In the last 12 years, there has been a proliferation of studies elucidating the immune mechanisms that mediate tissue damage in MS. Interferons (IFNs) have an important role in regulating innate and adaptive immune responses. They decrease pro-inflammatory responses such as the autoimmunity in MS, but other autoimmune responses such as systemic lupus erythematosus (SLE) may be exacerbated. This review offers a general overview of the biological properties of IFNs, effects on immune cells, and clinical effectiveness in MS treatment. IFN signaling is complex, from receptor binding events to the generation of effector mechanisms that dampen inflammation. Immune cell function is altered in MS. IFN treatment of MS patients ameliorates immune dysfunction, but not completely. The incomplete resolution of immune dysfunction by IFNs partly explains their significant, but modest therapeutic effects. This observation also suggests that there are immune mechanisms in MS that are resistant to IFN therapy. In MS, abnormalities may exist at several points along the IFN signaling pathway, including molecular defects in the IFN second messenger system. Currently, several studies are ongoing evaluating ways of potentiating IFN effects. IFNs were the first agents to show clinical efficacy in treatment of MS. More than a decade of experience with IFNs has showed continued clinical efficacy over time. In the near future, IFNs will continue to play a major role in MS.
Collapse
Affiliation(s)
- Adil Javed
- Department of Neurology, MC-2030, University of Chicago, 5841 South Maryland Avenue, IL 60637, USA
| | | |
Collapse
|
23
|
Frisullo G, Angelucci F, Caggiula M, Nociti V, Iorio R, Patanella AK, Sancricca C, Mirabella M, Tonali PA, Batocchi AP. pSTAT1, pSTAT3, and T-bet expression in peripheral blood mononuclear cells from relapsing-remitting multiple sclerosis patients correlates with disease activity. J Neurosci Res 2006; 84:1027-36. [PMID: 16865709 DOI: 10.1002/jnr.20995] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system, and it is considered to be a T helper 1 (Th1) cell-mediated autoimmune disease. T-bet has been identified as a key transcription factor for the development of Th1 cells and the induction of interferon (IFN)-gamma production. T-bet is induced during T-cell activation by the IFN-gamma signal transducer and activator of transcription (STAT)-1 signalling pathway. In this study we found an up-regulation of T-bet and pSTAT1 in peripheral blood CD4+ and CD8+ T cells and monocytes from relapsing-remitting MS patients in relapse compared with patients in remission and with healthy subjects. The increased expression of pSTAT1 strongly correlated with T-bet expression in CD4+ and CD8+ cells and monocytes from patients in relapse and was associated with an increased production of IFN-gamma by peripheral blood mononuclear cells (PBMCs). pSTAT3 was also up-regulated in CD4+ and CD8+ cells and monocytes from patients in relapse and was associated with an increased production of interleukin (IL)-10 but not of IL-6. pSTAT1, pSTAT3, and T-bet expression strongly correlated with Gd-DTPA-enhanced lesions on brain and spinal cord magnetic resonance imaging. Our data show for the first time that there is an up-regulation of type 1 immunity-correlated transcription factors such as STAT1 and T-bet in peripheral blood subpopulations of MS patients in the active phase of disease. The evaluation of T-bet and pSTAT1 expression in peripheral blood CD4+, CD8+ T cells and monocytes could be used as a marker of disease activity in relapsing-remitting MS.
Collapse
Affiliation(s)
- Giovanni Frisullo
- Institute of Neurology, Department of Neuroscience, Catholic University, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Berghella AM, Totaro R, Pellegrini P, Contasta I, Russo T, Carolei A, Adorno D. Immunological study of IFNbeta-1a-treated and untreated multiple sclerosis patients: clarifying IFNbeta mechanisms and establishing specific dendritic cell immunotherapy. Neuroimmunomodulation 2005; 12:29-44. [PMID: 15756051 DOI: 10.1159/000082362] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Accepted: 02/17/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES A comparative immunological evaluation of multiple sclerosis (MS) patients receiving IFNbeta treatment and patients who are not receiving treatment may help clarify IFNbeta neurological mechanisms and lead the way to an effective dendritic cell (DC) immunotherapy. This type of study helps clarify the pathological function of T cells and DCs within the TH1/TH2/TH3 network as well as the specific interactions between TH1/TH2/TH3 cytokines implicated in MS pathological mechanisms and determine the best way of reestablishing the TH1/TH2/TH3 network equilibrium. METHODS We studied network interactions between TH1/TH2/TH3 cytokine levels in serum and supernatants of whole blood and CD14+ monocyte-derived DCs in the remission phase of the disease and in correlation to the Expanded Disability Status Scale (EDSS). RESULTS We found that TH1 dysregulation results in a disruption of the maturation and activation of dendritic and T cells, and a lack of T-regulating cells for the induction of self-tolerance; IFNbeta mechanisms restore regulation by reestablishing the network balance but fail to resolve the disease completely due to in vivo IL12p70 network interactions leading to the deletion of self-aggressive cells. CONCLUSIONS Our results indicate that a specific DC immunotherapy could cure rather than treat MS. The best point to reestablish the normal physiological cycle is at the immature DC stage which can be done in vitro with treated peripheral blood CD14+ cells and used in vivo to stimulate the expansion of specific regulatory T cells.
Collapse
Affiliation(s)
- Anna Maria Berghella
- Istituto CNR per i Trapianti d'Organo e l'Immunocitologia (ITOI), L'Aquila, Italia
| | | | | | | | | | | | | |
Collapse
|
25
|
Batocchi AP, Rotondi M, Caggiula M, Frisullo G, Odoardi F, Nociti V, Carella C, Tonali PA, Mirabella M. Leptin as a marker of multiple sclerosis activity in patients treated with interferon-beta. J Neuroimmunol 2003; 139:150-4. [PMID: 12799033 DOI: 10.1016/s0165-5728(03)00154-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The role of leptin was investigated in relapsing-remitting multiple sclerosis (MS). Control and MS patients showed comparable baseline serum leptin levels. During the first year of IFNbeta-1a treatment, leptin significantly decreased since 2 months after starting therapy in 11 patients who had no relapses. A significant decrease in IL12/IL10 ratio was observed in this group of patients only after 1 year of treatment. An increase of leptin was observed before the first clinical exacerbation in 13 relapsing patients. Leptin may play a pathogenic role in MS and can be a useful marker of disease activity and response to therapy.
Collapse
Affiliation(s)
- Anna Paola Batocchi
- Institute of Neurology, Catholic University, Largo Gemelli 8, 00168, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|