1
|
Giovenzana A, Codazzi V, Pandolfo M, Petrelli A. T cell trafficking in human chronic inflammatory diseases. iScience 2024; 27:110528. [PMID: 39171290 PMCID: PMC11338127 DOI: 10.1016/j.isci.2024.110528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
Circulating T cells, which migrate from the periphery to sites of tissue inflammation, play a crucial role in the development of various chronic inflammatory conditions. Recent research has highlighted subsets of tissue-resident T cells that acquire migratory capabilities and re-enter circulation, referred to here as "recirculating T cells." In this review, we examine recent advancements in understanding the biology of T cell trafficking in diseases where T cell infiltration is pivotal, such as multiple sclerosis and inflammatory bowel diseases, as well as in metabolic disorders where the role of T cell migration is less understood. Additionally, we discuss current insights into therapeutic strategies aimed at modulating T cell circulation across tissues and the application of state-of-the-art technologies for studying recirculation in humans. This review underscores the significance of investigating T trafficking as a novel potential target for therapeutic interventions across a spectrum of human chronic inflammatory diseases.
Collapse
Affiliation(s)
- Anna Giovenzana
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valentina Codazzi
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Michele Pandolfo
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
2
|
Košćak Lukač J, Baronica KB, Šućur A, Sremec J, Tomasović S, Baronica R, Kelava T, Grčević D, Kovačić N. Serum Concentrations of Chemokines CCL20, CXCL8 and CXCL10 in Relapsing-Remitting Multiple Sclerosis and Their Association with Presence of Antibodies against Epstein-Barr Virus. Int J Mol Sci 2024; 25:8064. [PMID: 39125633 PMCID: PMC11311471 DOI: 10.3390/ijms25158064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Epstein-Barr virus (EBV) infection and various chemokines, including CCL20, CXCL8 and CXCL10 are considered to participate in the pathogenesis of multiple sclerosis (MS), and several studies point to a direct regulatory effect of EBV on the expression of these chemokines. In our study we hypothesized that serum concentrations of CCL20, CXCL8 and CXCL0 are induced in patients with relapsing-remitting MS (RRMS) in comparison to healthy individuals, and that they are associated with EBV infection. Serum concentrations of CXCL8 and CXCL10 were lower in RRMS patients in relapse in comparison to healthy controls. Although potential effects of corticosteroid therapy introduced in a subgroup of RRMS patients prior to sampling were excluded by subgroup comparison, this possibility has to be considered while interpreting the results. We found an inverse association between serum concentrations of CXCL8 and anti-Epstein-Barr Virus Nuclear Antigen (EBNA) IgG and decreased expression of CXCL8 in peripheral blood mononuclear cells (PBMC) in relapse compared to remission. Lower serum concentrations of CXCL8 and CXCL10 in RRMS patients and decreased peripheral production of CXCL8 in relapse may indicate compensatory anti-inflammatory counter-regulation in MS.
Collapse
Affiliation(s)
- Jelena Košćak Lukač
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
| | - Koraljka Bačić Baronica
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
- Neurology Clinic, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31 000 Osijek, Croatia
| | - Alan Šućur
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Šalata 3, 10 000 Zagreb, Croatia
| | - Josip Sremec
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
| | - Sanja Tomasović
- Department of Neurology, Clinical Hospital “Sveti Duh”, Sveti Duh 64, 10 000 Zagreb, Croatia; (K.B.B.); (J.S.); (S.T.)
- Neurology Clinic, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31 000 Osijek, Croatia
| | - Robert Baronica
- Department of Anesthesiology, Reanimatology, Intensive Medicine and Pain Therapy, University Hospital Centre Zagreb, Kišpatićeva 12, 10 000 Zagreb, Croatia;
| | - Tomislav Kelava
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Šalata 3, 10 000 Zagreb, Croatia
| | - Danka Grčević
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Šalata 3, 10 000 Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, Šalata 12, 10 000 Zagreb, Croatia; (A.Š.); (T.K.); (D.G.); (N.K.)
- Department of Anatomy, University of Zagreb School of Medicine, Šalata 11, 10 000 Zagreb, Croatia
| |
Collapse
|
3
|
Brignone MS, Lanciotti A, Molinari P, Mallozzi C, De Nuccio C, Caprini ES, Petrucci TC, Visentin S, Ambrosini E. Megalencephalic leukoencephalopathy with subcortical cysts protein-1: A new calcium-sensitive protein functionally activated by endoplasmic reticulum calcium release and calmodulin binding in astrocytes. Neurobiol Dis 2024; 190:106388. [PMID: 38141856 DOI: 10.1016/j.nbd.2023.106388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/04/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND MLC1 is a membrane protein highly expressed in brain perivascular astrocytes and whose mutations account for the rare leukodystrophy (LD) megalencephalic leukoencephalopathy with subcortical cysts disease (MLC). MLC is characterized by macrocephaly, brain edema and cysts, myelin vacuolation and astrocyte swelling which cause cognitive and motor dysfunctions and epilepsy. In cultured astrocytes, lack of functional MLC1 disturbs cell volume regulation by affecting anion channel (VRAC) currents and the consequent regulatory volume decrease (RVD) occurring in response to osmotic changes. Moreover, MLC1 represses intracellular signaling molecules (EGFR, ERK1/2, NF-kB) inducing astrocyte activation and swelling following brain insults. Nevertheless, to date, MLC1 proper function and MLC molecular pathogenesis are still elusive. We recently reported that in astrocytes MLC1 phosphorylation by the Ca2+/Calmodulin-dependent kinase II (CaMKII) in response to intracellular Ca2+ release potentiates MLC1 activation of VRAC. These results highlighted the importance of Ca2+ signaling in the regulation of MLC1 functions, prompting us to further investigate the relationships between intracellular Ca2+ and MLC1 properties. METHODS We used U251 astrocytoma cells stably expressing wild-type (WT) or mutated MLC1, primary mouse astrocytes and mouse brain tissue, and applied biochemistry, molecular biology, video imaging and electrophysiology techniques. RESULTS We revealed that WT but not mutant MLC1 oligomerization and trafficking to the astrocyte plasma membrane is favored by Ca2+ release from endoplasmic reticulum (ER) but not by capacitive Ca2+ entry in response to ER depletion. We also clarified the molecular events underlining MLC1 response to cytoplasmic Ca2+ increase, demonstrating that, following Ca2+ release, MLC1 binds the Ca2+ effector protein calmodulin (CaM) at the carboxyl terminal where a CaM binding sequence was identified. Using a CaM inhibitor and generating U251 cells expressing MLC1 with CaM binding site mutations, we found that CaM regulates MLC1 assembly, trafficking and function, being RVD and MLC-linked signaling molecules abnormally regulated in these latter cells. CONCLUSION Overall, we qualified MLC1 as a Ca2+ sensitive protein involved in the control of volume changes in response to ER Ca2+ release and astrocyte activation. These findings provide new insights for the comprehension of the molecular mechanisms responsible for the myelin degeneration occurring in MLC and other LD where astrocytes have a primary role in the pathological process.
Collapse
Affiliation(s)
- M S Brignone
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - A Lanciotti
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - P Molinari
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - C Mallozzi
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - C De Nuccio
- Istituto Superiore di Sanità, Research Coordination and Support Service, Viale Regina Elena 299, 00161 Rome, Italy
| | - E S Caprini
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - T C Petrucci
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - S Visentin
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - E Ambrosini
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
4
|
Mora P, Chapouly C. Astrogliosis in multiple sclerosis and neuro-inflammation: what role for the notch pathway? Front Immunol 2023; 14:1254586. [PMID: 37936690 PMCID: PMC10627009 DOI: 10.3389/fimmu.2023.1254586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Multiple sclerosis is an autoimmune inflammatory disease of the central nervous system leading to neurodegeneration. It affects 2.3 million people worldwide, generally younger than 50. There is no known cure for the disease, and current treatment options - mainly immunotherapies to limit disease progression - are few and associated with serious side effects. In multiple sclerosis, disruption of the blood-brain barrier is an early event in the pathogenesis of lesions, predisposing to edema, excito-toxicity and inflammatory infiltration into the central nervous system. Recently, the vision of the blood brain barrier structure and integrity has changed and include contributions from all components of the neurovascular unit, among which astrocytes. During neuro-inflammation, astrocytes become reactive. They undergo morphological and molecular changes named "astrogliosis" driving the conversion from acute inflammatory injury to a chronic neurodegenerative state. Astrogliosis mechanisms are minimally explored despite their significance in regulating the autoimmune response during multiple sclerosis. Therefore, in this review, we take stock of the state of knowledge regarding astrogliosis in neuro-inflammation and highlight the central role of NOTCH signaling in the process of astrocyte reactivity. Indeed, a very detailed nomenclature published in nature neurosciences in 2021, listing all the reactive astrocyte markers fully identified in the literature, doesn't cover the NOTCH signaling. Hence, we discuss evidence supporting NOTCH1 receptor as a central regulator of astrogliosis in the pathophysiology of neuro-inflammation, notably multiple sclerosis, in human and experimental models.
Collapse
Affiliation(s)
- Pierre Mora
- Université de Bordeaux, Institut national de la santé et de la recherche médicale (INSERM), Biology of Cardiovascular Diseases, Pessac, France
| | | |
Collapse
|
5
|
Iro MA, Rollier CS, Irani SR, Sadarangani M, Al-Diwani A, Pollard AJ, Clutterbuck EA. Regulatory T cell profiles in patients with N-methyl-ᴅ-aspartate receptor-antibody encephalitis. ENCEPHALITIS 2023; 3:15-23. [PMID: 37469716 PMCID: PMC10295821 DOI: 10.47936/encephalitis.2022.00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/24/2022] [Accepted: 09/30/2022] [Indexed: 07/21/2023] Open
Abstract
Purpose Purpose Regulatory T cells (Tregs) have been implicated in the pathogenesis of several autoimmune disorders and used in adoptive cell transfer therapies. Neither have been explored in patients with autoimmune encephalitis where treated patient outcomes remain suboptimal with frequent relapses. Here, to identify new treatment strategies for autoimmune encephalitis, we sought to evaluate the proportion of circulating Tregs and Treg subpopulations in peripheral blood of patients with N-methyl-ᴅ-aspartate receptor-antibody encephalitis (NMDAR-Ab-E) and compared this with healthy controls. Methods We compared the phenotype of peripheral blood Tregs in four adult NMDAR-Ab-E patients and four age- and sex-matched healthy controls using an 11-color flow cytometry assay panel for characterization of Tregs (CD4+ CD25+ FoxP3+) cells into naïve (chemokine receptor [CCR] 7+ CD45RA+), central memory (CCR7+ CD45RA-), and effector memory (CCR7- CD45RA-) cells. We also examined and compared the expression of the CCR6 by circulating Tregs and the respective Treg subpopulations between the study groups. Results The proportion of circulating Tregs was similar between patients with NMDAR-Ab-E and healthy controls but the proportion of naïve Tregs was lower in NMDAR-Ab-E patients (p = 0.0026). Additionally, the frequency of circulating effector memory Tregs was higher, and the proportion of circulating effector memory Tregs expressing CCR6 was lower, in NMDAR-Ab-E patients compared with healthy controls (p = 0.0026). Conclusion Altered Treg homeostasis may be a feature of patients with NMDAR-Ab-E. Future studies with larger samples are warranted to validate these findings.
Collapse
Affiliation(s)
- Mildred A. Iro
- Faculty of Medicine and Institute of Life Sciences, University of Southampton, United Kingdom
- Department of Paediatric Infectious Diseases, Southampton Children’s Hospital, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Christine S. Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Sarosh R. Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation, Trust, Oxford, United Kingdom
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Adam Al-Diwani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- University Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Elizabeth A. Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
6
|
Implications of fractalkine on glial function, ablation and glial proteins/receptors/markers—understanding its therapeutic usefulness in neurological settings: a narrative review. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-022-00446-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Abstract
Background
Fractalkine (CX3CL1) is a chemokine predominantly released by neurons. As a signaling molecule, CX3CL1 facilitates talk between neurons and glia. CX3CL1 is considered as a potential target which could alleviate neuroinflammation. However, certain controversial results and ambiguous role of CX3CL1 make it inexorable to decipher the overall effects of CX3CL1 on the physiopathology of glial cells.
Main body of the abstract
Implications of cross-talk between CX3CL1 and different glial proteins/receptors/markers will give a bird eye view of the therapeutic significance of CX3CL1. Keeping with the need, this review identifies the effects of CX3CL1 on glial physiopathology, glial ablation, and gives a wide coverage on the effects of CX3CL1 on certain glial proteins/receptors/markers.
Short conclusion
Pinpoint prediction of the therapeutic effect of CX3CL1 on neuroinflammation needs further research. This is owing to certain obscure roles and implications of CX3CL1 on different glial proteins/receptors/markers, which are crucial under neurological settings. Further challenges are imposed due to the dichotomous roles played by CX3CL1. The age-old chemokine shows many newer scopes of research in near future. Thus, overall assessment of the effect of CX3CL1 becomes crucial prior to its administration in neuroinflammation.
Collapse
|
7
|
Heng AHS, Han CW, Abbott C, McColl SR, Comerford I. Chemokine-Driven Migration of Pro-Inflammatory CD4 + T Cells in CNS Autoimmune Disease. Front Immunol 2022; 13:817473. [PMID: 35250997 PMCID: PMC8889115 DOI: 10.3389/fimmu.2022.817473] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
Pro-inflammatory CD4+ T helper (Th) cells drive the pathogenesis of many autoimmune conditions. Recent advances have modified views of the phenotype of pro-inflammatory Th cells in autoimmunity, extending the breadth of known Th cell subsets that operate as drivers of these responses. Heterogeneity and plasticity within Th1 and Th17 cells, and the discovery of subsets of Th cells dedicated to production of other pro-inflammatory cytokines such as GM-CSF have led to these advances. Here, we review recent progress in this area and focus specifically upon evidence for chemokine receptors that drive recruitment of these various pro-inflammatory Th cell subsets to sites of autoimmune inflammation in the CNS. We discuss expression of specific chemokine receptors by subsets of pro-inflammatory Th cells and highlight which receptors may be tractable targets of therapeutic interventions to limit pathogenic Th cell recruitment in autoimmunity.
Collapse
Affiliation(s)
- Aaron H S Heng
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Caleb W Han
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Caitlin Abbott
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Shaun R McColl
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Iain Comerford
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
8
|
Manouchehri N, Salinas VH, Rabi Yeganeh N, Pitt D, Hussain RZ, Stuve O. Efficacy of Disease Modifying Therapies in Progressive MS and How Immune Senescence May Explain Their Failure. Front Neurol 2022; 13:854390. [PMID: 35432156 PMCID: PMC9009145 DOI: 10.3389/fneur.2022.854390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
The advent of disease modifying therapies (DMT) in the past two decades has been the cornerstone of successful clinical management of multiple sclerosis (MS). Despite the great strides made in reducing the relapse frequency and occurrence of new signal changes on neuroimaging in patients with relapsing remitting MS (RRMS) by approved DMT, it has been challenging to demonstrate their effectiveness in non-active secondary progressive MS (SPMS) and primary progressive MS (PPMS) disease phenotypes. The dichotomy of DMT effectiveness between RRMS and progressive MS informs on distinct pathogeneses of the different MS phenotypes. Conversely, factors that render patients with progressive MS resistant to therapy are not understood. Thus far, age has emerged as the main correlate of the transition from RRMS to SPMS. Whether it is aging and age-related factors or the underlying immune senescence that qualitatively alter immune responses as the disease transitions to SPMS, that diminish DMT effectiveness, or both, is currently not known. Here, we will discuss the role of immune senescence on different arms of the immune system, and how it may explain relative DMT resistance.
Collapse
Affiliation(s)
- Navid Manouchehri
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Victor H. Salinas
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Negar Rabi Yeganeh
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - David Pitt
- Department of Neurology, Yale University, New Haven, CT, United States
| | - Rehana Z. Hussain
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Medical Service Dallas, Veterans Affairs Medical Center, Dallas, TX, United States
- *Correspondence: Olaf Stuve
| |
Collapse
|
9
|
Kunkl M, Amormino C, Tedeschi V, Fiorillo MT, Tuosto L. Astrocytes and Inflammatory T Helper Cells: A Dangerous Liaison in Multiple Sclerosis. Front Immunol 2022; 13:824411. [PMID: 35211120 PMCID: PMC8860818 DOI: 10.3389/fimmu.2022.824411] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/13/2022] [Indexed: 11/15/2022] Open
Abstract
Multiple Sclerosis (MS) is a neurodegenerative autoimmune disorder of the central nervous system (CNS) characterized by the recruitment of self-reactive T lymphocytes, mainly inflammatory T helper (Th) cell subsets. Once recruited within the CNS, inflammatory Th cells produce several inflammatory cytokines and chemokines that activate resident glial cells, thus contributing to the breakdown of blood-brain barrier (BBB), demyelination and axonal loss. Astrocytes are recognized as key players of MS immunopathology, which respond to Th cell-defining cytokines by acquiring a reactive phenotype that amplify neuroinflammation into the CNS and contribute to MS progression. In this review, we summarize current knowledge of the astrocytic changes and behaviour in both MS and experimental autoimmune encephalomyelitis (EAE), and the contribution of pathogenic Th1, Th17 and Th1-like Th17 cell subsets, and CD8+ T cells to the morphological and functional modifications occurring in astrocytes and their pathological outcomes.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Valentina Tedeschi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
10
|
Astrocytes in Multiple Sclerosis-Essential Constituents with Diverse Multifaceted Functions. Int J Mol Sci 2021; 22:ijms22115904. [PMID: 34072790 PMCID: PMC8198285 DOI: 10.3390/ijms22115904] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 11/19/2022] Open
Abstract
In multiple sclerosis (MS), astrocytes respond to the inflammatory stimulation with an early robust process of morphological, transcriptional, biochemical, and functional remodeling. Recent studies utilizing novel technologies in samples from MS patients, and in an animal model of MS, experimental autoimmune encephalomyelitis (EAE), exposed the detrimental and the beneficial, in part contradictory, functions of this heterogeneous cell population. In this review, we summarize the various roles of astrocytes in recruiting immune cells to lesion sites, engendering the inflammatory loop, and inflicting tissue damage. The roles of astrocytes in suppressing excessive inflammation and promoting neuroprotection and repair processes is also discussed. The pivotal roles played by astrocytes make them an attractive therapeutic target. Improved understanding of astrocyte function and diversity, and the mechanisms by which they are regulated may lead to the development of novel approaches to selectively block astrocytic detrimental responses and/or enhance their protective properties.
Collapse
|
11
|
Heo YJ, Choi SE, Lee N, Jeon JY, Han SJ, Kim DJ, Kang Y, Lee KW, Kim HJ. CCL20 induced by visfatin in macrophages via the NF-κB and MKK3/6-p38 signaling pathways contributes to hepatic stellate cell activation. Mol Biol Rep 2020; 47:4285-4293. [PMID: 32418112 PMCID: PMC7295719 DOI: 10.1007/s11033-020-05510-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/08/2020] [Indexed: 12/16/2022]
Abstract
Chemokines interact with hepatic resident cells during inflammation and fibrosis. CC chemokine ligand (CCL) 20 has been reported to be important in inflammation and fibrosis in the liver. We hypothesized that visfatin, an adipocytokine, could play a role in hepatic fibrosis via CCL20. We investigated the effect of visfatin on CCL20 in THP-1 human promonocytic cells and examined the molecular mechanisms involved. Following treatment of THP-1 cells with visfatin, CCL20 expression and secretion were assessed. We assessed the intracellular signaling molecules IKK/NF-κB, JAK2/STAT3, MAPKs, and MKK3/6 by western blotting. We treated THP-1 cells with visfatin and signaling inhibitors, and examined CCL20 mRNA and protein levels. To investigate the effect of visfatin-induced CCL20 expression in hepatic stellate cells (HSCs), LX-2 cells were co-cultured with the culture supernatant of THP-1 cells with or without anti-CCL20 neutralizing antibodies, and fibrosis markers were examined by RT-PCR and immunoblotting. In THP-1 cells, visfatin increased the CCL20 mRNA and protein levels. visfatin increased the activities of the NF-κB, p38, and MLK3/6 signaling pathways but not those of the JAK2/STAT3 and ERK pathways. Visfatin treatment together with an NF-κB, p38, or MLK3 inhibitor reduced the mRNA and protein levels of CCL20. The visfatin-induced CCL20 increased the expression of fibrosis markers and CCR6 in HSCs. Following neutralization of CCL20, the levels of fibrosis markers and CCR6 were decreased. Visfatin increases the expression of CCL20 via the NF-κB and MKK3/6-p38 signaling pathways in macrophages, and visfatin-induced CCL20 expression promotes the fibrosis markers in HSCs.
Collapse
Affiliation(s)
- Yu Jung Heo
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Sung-E Choi
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Nami Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Ja Young Jeon
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Kwan Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea.
| |
Collapse
|
12
|
Goshi N, Morgan RK, Lein PJ, Seker E. A primary neural cell culture model to study neuron, astrocyte, and microglia interactions in neuroinflammation. J Neuroinflammation 2020; 17:155. [PMID: 32393376 PMCID: PMC7216677 DOI: 10.1186/s12974-020-01819-z] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background Interactions between neurons, astrocytes, and microglia critically influence neuroinflammatory responses to insult in the central nervous system. In vitro astrocyte and microglia cultures are powerful tools to study specific molecular pathways involved in neuroinflammation; however, in order to better understand the influence of cellular crosstalk on neuroinflammation, new multicellular culture models are required. Methods Primary cortical cells taken from neonatal rats were cultured in a serum-free “tri-culture” medium formulated to support neurons, astrocytes, and microglia, or a “co-culture” medium formulated to support only neurons and astrocytes. Caspase 3/7 activity and morphological changes were used to quantify the response of the two culture types to different neuroinflammatory stimuli mimicking sterile bacterial infection (lipopolysaccharide (LPS) exposure), mechanical injury (scratch), and seizure activity (glutamate-induced excitotoxicity). The secreted cytokine profile of control and LPS-exposed co- and tri-cultures were also compared. Results The tri-culture maintained a physiologically relevant representation of neurons, astrocytes, and microglia for 14 days in vitro, while the co-cultures maintained a similar population of neurons and astrocytes, but lacked microglia. The continuous presence of microglia did not negatively impact the overall health of the neurons in the tri-culture, which showed reduced caspase 3/7 activity and similar neurite outgrowth as the co-cultures, along with an increase in the microglia-secreted neurotrophic factor IGF-1 and a significantly reduced concentration of CX3CL1 in the conditioned media. LPS-exposed tri-cultures showed significant astrocyte hypertrophy, increase in caspase 3/7 activity, and the secretion of a number of pro-inflammatory cytokines (e.g., TNF, IL-1α, IL-1β, and IL-6), none of which were observed in LPS-exposed co-cultures. Following mechanical trauma, the tri-culture showed increased caspase 3/7 activity, as compared to the co-culture, along with increased astrocyte migration towards the source of injury. Finally, the microglia in the tri-culture played a significant neuroprotective role during glutamate-induced excitotoxicity, with significantly reduced neuron loss and astrocyte hypertrophy in the tri-culture. Conclusions The tri-culture consisting of neurons, astrocytes, and microglia more faithfully mimics in vivo neuroinflammatory responses than standard mono- and co-cultures. This tri-culture can be a useful tool to study neuroinflammation in vitro with improved accuracy in predicting in vivo neuroinflammatory phenomena.
Collapse
Affiliation(s)
- Noah Goshi
- Department of Biomedical Engineering, University of California - Davis, Davis, CA, 95616, USA
| | - Rhianna K Morgan
- Department of Molecular Biosciences, University of California - Davis, Davis, CA, 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California - Davis, Davis, CA, 95616, USA
| | - Erkin Seker
- Department of Electrical and Computer Engineering, University of California - Davis, 3177 Kemper Hall, Davis, CA, 95616, USA.
| |
Collapse
|
13
|
Watson AES, Goodkey K, Footz T, Voronova A. Regulation of CNS precursor function by neuronal chemokines. Neurosci Lett 2020; 715:134533. [DOI: 10.1016/j.neulet.2019.134533] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
|
14
|
Raphael I, Gomez-Rivera F, Raphael RA, Robinson RR, Nalawade S, Forsthuber TG. TNFR2 limits proinflammatory astrocyte functions during EAE induced by pathogenic DR2b-restricted T cells. JCI Insight 2019; 4:132527. [PMID: 31852844 DOI: 10.1172/jci.insight.132527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/13/2019] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune neuroinflammatory disease where the underlying mechanisms driving disease progression have remained unresolved. HLA-DR2b (DRB1*15:01) is the most common genetic risk factor for MS. Additionally, TNF and its receptors TNFR1 and TNFR2 play key roles in MS and its preclinical animal model, experimental autoimmune encephalomyelitis (EAE). TNFR2 is believed to ameliorate CNS pathology by promoting remyelination and Treg function. Here, we show that transgenic mice expressing the human MHC class II (MHC-II) allele HLA-DR2b and lacking mouse MHC-II and TNFR2 molecules, herein called DR2bΔR2, developed progressive EAE, while disease was not progressive in DR2b littermates. Mechanistically, expression of the HLA-DR2b favored Th17 cell development, whereas T cell-independent TNFR2 expression was critical for restraining of an astrogliosis-induced proinflammatory milieu and Th17 cell responses, while promoting remyelination. Our data suggest the TNFR2 signaling pathway as a potentially novel mechanism for curtailing astrogliosis and promoting remyelination, thus providing new insights into mechanisms limiting progressive MS.
Collapse
Affiliation(s)
- Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, UPMC Children's Hospital, Pittsburgh, Pennsylvania, USA.,Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Francisco Gomez-Rivera
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rebecca A Raphael
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Saisha Nalawade
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
15
|
Gharagozloo M, Mahmoud S, Simard C, Yamamoto K, Bobbala D, Ilangumaran S, Smith MD, Lamontagne A, Jarjoura S, Denault JB, Blais V, Gendron L, Vilariño-Güell C, Sadovnick AD, Ting JP, Calabresi PA, Amrani A, Gris D. NLRX1 inhibits the early stages of CNS inflammation and prevents the onset of spontaneous autoimmunity. PLoS Biol 2019; 17:e3000451. [PMID: 31525189 PMCID: PMC6762215 DOI: 10.1371/journal.pbio.3000451] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 09/26/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022] Open
Abstract
Nucleotide-binding, leucine-rich repeat containing X1 (NLRX1) is a mitochondria-located innate immune sensor that inhibits major pro-inflammatory pathways such as type I interferon and nuclear factor-κB signaling. We generated a novel, spontaneous, and rapidly progressing mouse model of multiple sclerosis (MS) by crossing myelin-specific T-cell receptor (TCR) transgenic mice with Nlrx1−/− mice. About half of the resulting progeny developed spontaneous experimental autoimmune encephalomyelitis (spEAE), which was associated with severe demyelination and inflammation in the central nervous system (CNS). Using lymphocyte-deficient mice and a series of adoptive transfer experiments, we demonstrate that genetic susceptibility to EAE lies within the innate immune compartment. We show that NLRX1 inhibits the subclinical stages of microglial activation and prevents the generation of neurotoxic astrocytes that induce neuronal and oligodendrocyte death in vitro. Moreover, we discovered several mutations within NLRX1 that run in MS-affected families. In summary, our findings highlight the importance of NLRX1 in controlling the early stages of CNS inflammation and preventing the onset of spontaneous autoimmunity. NLRX1 is a guardian protein that inhibits the inflammatory response of glial cells within the central nervous system and prevents the onset of a spontaneous multiple sclerosis–like disease in mice. This study uses a novel mouse model to provide mechanistic insights into the neurodegenerative origin of multiple sclerosis.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shaimaa Mahmoud
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Camille Simard
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Kenzo Yamamoto
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Diwakar Bobbala
- Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Subburaj Ilangumaran
- Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Albert Lamontagne
- Department of Neurology, Faculty of Medicine, MS Clinic, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Samir Jarjoura
- Department of Neurology, Faculty of Medicine, MS Clinic, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Bernard Denault
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Véronique Blais
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Louis Gendron
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - A. Dessa Sadovnick
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Jenny P. Ting
- Department of Microbiology and Immunology, Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Abdelaziz Amrani
- Department of Pediatrics, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Denis Gris
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- * E-mail:
| |
Collapse
|
16
|
Kimura A, Takemura M, Yamamoto Y, Hayashi Y, Saito K, Shimohata T. Cytokines and biological markers in autoimmune GFAP astrocytopathy: The potential role for pathogenesis and therapeutic implications. J Neuroimmunol 2019; 334:576999. [DOI: 10.1016/j.jneuroim.2019.576999] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/07/2019] [Accepted: 06/20/2019] [Indexed: 10/26/2022]
|
17
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
18
|
Cabrera‐Pastor A, Llansola M, Montoliu C, Malaguarnera M, Balzano T, Taoro‐Gonzalez L, García‐García R, Mangas‐Losada A, Izquierdo‐Altarejos P, Arenas YM, Leone P, Felipo V. Peripheral inflammation induces neuroinflammation that alters neurotransmission and cognitive and motor function in hepatic encephalopathy: Underlying mechanisms and therapeutic implications. Acta Physiol (Oxf) 2019; 226:e13270. [PMID: 30830722 DOI: 10.1111/apha.13270] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022]
Abstract
Several million patients with liver cirrhosis suffer minimal hepatic encephalopathy (MHE), with mild cognitive and coordination impairments that reduce their quality of life and life span. Hyperammonaemia and peripheral inflammation act synergistically to induce these neurological alterations. We propose that MHE appearance is because of the changes in peripheral immune system, which are transmitted to brain, leading to neuroinflammation that alters neurotransmission leading to cognitive and motor alterations. We summarize studies showing that MHE in cirrhotic patients is associated with alterations in the immune system and that patients died with HE show neuroinflammation in cerebellum, with microglial and astrocytic activation and Purkinje cell loss. We also summarize studies in animal models of MHE on the role of peripheral inflammation in neuroinflammation induction, how neuroinflammation alters neurotransmission and how this leads to cognitive and motor alterations. These studies identify therapeutic targets and treatments that improve cognitive and motor function. Rats with MHE show neuroinflammation in hippocampus and altered NMDA and AMPA receptor membrane expression, which impairs spatial learning and memory. Neuroinflammation in cerebellum is associated with altered GABA transporters and extracellular GABA, which impair motor coordination and learning in a Y maze. These alterations are reversed by treatments that reduce peripheral inflammation (anti-TNFα, ibuprofen), neuroinflammation (sulphoraphane, p38 inhibitors), GABAergic tone (bicuculline, pregnenolone sulphate) or increase extracellular cGMP (sildenafil or cGMP). The mechanisms identified would also occur in other chronic diseases associated with inflammation, aging and some mental and neurodegenerative diseases. Treatments that improve MHE may also be beneficial to treat these pathologies.
Collapse
Affiliation(s)
- Andrea Cabrera‐Pastor
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
- Fundacion Investigacion Hospital Clinico Valencia, INCLIVA Valencia Spain
| | - Marta Llansola
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| | - Carmina Montoliu
- Fundacion Investigacion Hospital Clinico Valencia, INCLIVA Valencia Spain
| | - Michele Malaguarnera
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| | - Tiziano Balzano
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| | - Lucas Taoro‐Gonzalez
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| | - Raquel García‐García
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| | - Alba Mangas‐Losada
- Fundacion Investigacion Hospital Clinico Valencia, INCLIVA Valencia Spain
| | | | - Yaiza M. Arenas
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| | - Paola Leone
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| | - Vicente Felipo
- Laboratory of Neurobiology Centro de Investigación Principe Felipe Valencia Spain
| |
Collapse
|
19
|
The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol 2019; 137:757-783. [PMID: 30847559 DOI: 10.1007/s00401-019-01980-7] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation is the coordinated response of the central nervous system (CNS) to threats to its integrity posed by a variety of conditions, including autoimmunity, pathogens and trauma. Activated astrocytes, in concert with other cellular elements of the CNS and immune system, are important players in the modulation of the neuroinflammatory response. During neurological disease, they produce and respond to cellular signals that often lead to dichotomous processes, which can promote further damage or contribute to repair. This occurs also in multiple sclerosis (MS), where astrocytes are now recognized as key components of its immunopathology. Evidence supporting this role has emerged not only from studies in MS patients, but also from animal models, among which the experimental autoimmune encephalomyelitis (EAE) model has proved especially instrumental. Based on this premise, the purpose of the present review is to summarize the current knowledge of astrocyte behavior in MS and EAE. Following a brief description of the pathological characteristics of the two diseases and the main functional roles of astrocytes in CNS physiology, we will delve into the specific responses of this cell population, analyzing MS and EAE in parallel. We will define the temporal and anatomical profile of astroglial activation, then focus on key processes they participate in. These include: (1) production and response to soluble mediators (e.g., cytokines and chemokines), (2) regulation of oxidative stress, and (3) maintenance of BBB integrity and function. Finally, we will review the state of the art on the available methods to measure astroglial activation in vivo in MS patients, and how this could be exploited to optimize diagnosis, prognosis and treatment decisions. Ultimately, we believe that integrating the knowledge obtained from studies in MS and EAE may help not only better understand the pathophysiology of MS, but also uncover new signals to be targeted for therapeutic intervention.
Collapse
|
20
|
De Luca C, Colangelo AM, Alberghina L, Papa M. Neuro-Immune Hemostasis: Homeostasis and Diseases in the Central Nervous System. Front Cell Neurosci 2018; 12:459. [PMID: 30534057 PMCID: PMC6275309 DOI: 10.3389/fncel.2018.00459] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/12/2018] [Indexed: 01/08/2023] Open
Abstract
Coagulation and the immune system interact in several physiological and pathological conditions, including tissue repair, host defense, and homeostatic maintenance. This network plays a key role in diseases of the central nervous system (CNS) by involving several cells (CNS resident cells, platelets, endothelium, and leukocytes) and molecular pathways (protease activity, complement factors, platelet granule content). Endothelial damage prompts platelet activation and the coagulation cascade as the first physiological step to support the rescue of damaged tissues, a flawed rescuing system ultimately producing neuroinflammation. Leukocytes, platelets, and endothelial cells are sensitive to the damage and indeed can release or respond to chemokines and cytokines (platelet factor 4, CXCL4, TNF, interleukins), and growth factors (including platelet-derived growth factor, vascular endothelial growth factor, and brain-derived neurotrophic factor) with platelet activation, change in capillary permeability, migration or differentiation of leukocytes. Thrombin, plasmin, activated complement factors and matrix metalloproteinase-1 (MMP-1), furthermore, activate intracellular transduction through complement or protease-activated receptors. Impairment of the neuro-immune hemostasis network induces acute or chronic CNS pathologies related to the neurovascular unit, either directly or by the systemic activation of its main steps. Neurons, glial cells (astrocytes and microglia) and the extracellular matrix play a crucial function in a “tetrapartite” synaptic model. Taking into account the neurovascular unit, in this review we thoroughly analyzed the influence of neuro-immune hemostasis on these five elements acting as a functional unit (“pentapartite” synapse) in the adaptive and maladaptive plasticity and discuss the relevance of these events in inflammatory, cerebrovascular, Alzheimer, neoplastic and psychiatric diseases. Finally, based on the solid reviewed data, we hypothesize a model of neuro-immune hemostatic network based on protein–protein interactions. In addition, we propose that, to better understand and favor the maintenance of adaptive plasticity, it would be useful to construct predictive molecular models, able to enlighten the regulating logic of the complex molecular network, which belongs to different cellular domains. A modeling approach would help to define how nodes of the network interact with basic cellular functions, such as mitochondrial metabolism, autophagy or apoptosis. It is expected that dynamic systems biology models might help to elucidate the fine structure of molecular events generated by blood coagulation and neuro-immune responses in several CNS diseases, thereby opening the way to more effective treatments.
Collapse
Affiliation(s)
- Ciro De Luca
- Laboratory of Morphology of Neuronal Network, Department of Public Medicine, University of Campania-Luigi Vanvitelli, Naples, Italy
| | - Anna Maria Colangelo
- Laboratory of Neuroscience "R. Levi-Montalcini", Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Milan, Italy
| | - Lilia Alberghina
- Laboratory of Neuroscience "R. Levi-Montalcini", Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Milan, Italy
| | - Michele Papa
- Laboratory of Morphology of Neuronal Network, Department of Public Medicine, University of Campania-Luigi Vanvitelli, Naples, Italy.,SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
21
|
Jafarzadeh A, Nemati M. Therapeutic potentials of ginger for treatment of Multiple sclerosis: A review with emphasis on its immunomodulatory, anti-inflammatory and anti-oxidative properties. J Neuroimmunol 2018; 324:54-75. [PMID: 30243185 DOI: 10.1016/j.jneuroim.2018.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/24/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is characterized by chronic inflammatory response-induced demyelination of the neurons and degeneration of the axons within the central nervous system (CNS). A complex network of immunopathological-, inflammatory- and oxidative parameters involve in the development and advancement of MS. The anti-inflammatory, immunomodulatory and anti-oxidative characteristics of the ginger and several of its components have been indicated in some of experimental and clinical investigations. The possible therapeutic potentials of ginger and its ingredients in the treatment of MS may exert mainly through the regulation of the Th1-, Th2-, Th9-, Th17-, Th22- and Treg cell-related immune responses, down-regulation of the B cell-related immune responses, modulation of the macrophages-related responses, modulation of the production of pro- and anti-inflammatory cytokines, down-regulation of the arachidonic acid-derived mediators, interfering with the toll like receptor-related signaling pathways, suppression of the inflammasomes, down-regulation of the oxidative stress, reduction of the adhesion molecules expression, and down-regulation of the expression of the chemokines and chemokine receptors. This review aimed to provide a comprehensive knowledge regarding the immunomodulatory-, anti-inflammatory and anti-oxidative properties of ginger and its components, and highlight novel insights into the possible therapeutic potentials of this plant for treatment of MS. The review encourages more investigations to consider the therapeutic potentials of ginger and its effective components for managing of MS.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Maryam Nemati
- Department of Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
22
|
El Sharkawi FZ, Ali SA, Hegazy MI, Atya HB. The combined effect of IL-17F and CCL20 gene polymorphism in susceptibility to multiple sclerosis in Egypt. Gene 2018; 685:164-169. [PMID: 30399422 DOI: 10.1016/j.gene.2018.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/01/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Levels of CCL20 and its CCR6 receptor are elevated in many autoimmune diseases which help in the recruitment of T helper (Th17) cells to site of inflammation. OBJECTIVES Determine the value of single nucleotide polymorphism of CCL20 (rs6749704) and IL-17F (rs763780) genes and their concomitant effect on the serum CCL20 level and susceptibility to MS in Egyptian patients. SUBJECTS AND METHODS Blood samples were collected from 83 patients and 95 healthy subjects. Serum levels of CCL20 were measured by ELISA. The DNA was analyzed for rs6749704 and rs763780 using Genotyping Taqman assay. RESULTS The mean serum levels of CCL20 in the MS group were significantly higher than healthy group (P < 0.001). Frequencies of CT genotype of rs6749704 in CCL20 gene and C allele in MS patients were significantly higher compared to controls. Also significant increase of rs763780 in IL-17F gene was detected in MS patients. Concomitant polymorphism in both genes in MS patients showed an increase risk to MS rather than individual locus. CONCLUSION CCL20 may play an important role in the pathogenesis of MS. Both allelic variation of (rs6749704) within CCL20 gene and (rs763780) within IL-17F gene can be considered risk factor for development of MS in Egyptian patients.
Collapse
Affiliation(s)
| | - Sahar A Ali
- Biochemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| | - Mohamed I Hegazy
- Neurology Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Hanaa B Atya
- Biochemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| |
Collapse
|
23
|
Park JH, Noh Y, Kim SS, Ahn JH, Ohk TG, Cho JH, Lee TK, Kim H, Song M, Lee JC, Won MH, Lee CH. Time-Course Changes and New Expressions of MIP-3α and Its Receptor, CCR6, in the Gerbil Hippocampal CA1 Area Following Transient Global Cerebral Ischemia. Neurochem Res 2018; 43:2102-2110. [PMID: 30203401 DOI: 10.1007/s11064-018-2632-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/13/2018] [Accepted: 09/06/2018] [Indexed: 12/25/2022]
Abstract
Macrophage inflammatory protein-3α (MIP-3α) and its sole receptor, CCR6, play pivotal roles in neuroinflammatory processes induced by brain ischemic insults. In this study, we investigated transient ischemia-induced changes in MIP-3α and CCR6 protein expressions in the hippocampal CA1 area following 5 min of transient global cerebral ischemia (tgCI) in gerbils. Both MIP-3α and CCR6 immunoreactivities were very strongly expressed in pyramidal neurons of the CA1 area from 6 h to 1 day after tgCI and were hardly shown 4 days after tgCI. In addition, strong MIP-3α immunoreactivity was newly expressed in astrocytes 6 h after tgCI. These results indicate that tgCI causes apparent changes in MIP-3α and CCR6 expressions in pyramidal neurons and astrocytes in the hippocampal CA1 area and suggest that tgCI-induced changes in MIP-3α and CCR6 expressions might be closely associated with neuroinflammatory processes in brain ischemic regions.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Republic of Korea
| | - YooHun Noh
- Famenity Company, Gwacheon, Geyonggi, 13837, Republic of Korea
| | - Sung-Su Kim
- Famenity Company, Gwacheon, Geyonggi, 13837, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea.
| |
Collapse
|
24
|
Wang K, Wang H, Lou W, Ma L, Li Y, Zhang N, Wang C, Li F, Awais M, Cao S, She R, Fu ZF, Cui M. IP-10 Promotes Blood-Brain Barrier Damage by Inducing Tumor Necrosis Factor Alpha Production in Japanese Encephalitis. Front Immunol 2018; 9:1148. [PMID: 29910805 PMCID: PMC5992377 DOI: 10.3389/fimmu.2018.01148] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 05/07/2018] [Indexed: 01/21/2023] Open
Abstract
Japanese encephalitis is a neuropathological disorder caused by Japanese encephalitis virus (JEV), which is characterized by severe pathological neuroinflammation and damage to the blood-brain barrier (BBB). Inflammatory cytokines/chemokines can regulate the expression of tight junction (TJ) proteins and are believed to be a leading cause of BBB disruption, but the specific mechanisms remain unclear. IP-10 is the most abundant chemokine produced in the early stage of JEV infection, but its role in BBB disruption is unknown. The administration of IP-10-neutralizing antibody ameliorated the decrease in TJ proteins and restored BBB integrity in JEV-infected mice. In vitro study showed IP-10 and JEV treatment did not directly alter the permeability of the monolayers of endothelial cells. However, IP-10 treatment promoted tumor necrosis factor alpha (TNF-α) production and IP-10-neutralizing antibody significantly reduced the production of TNF-α. Thus, TNF-α could be a downstream cytokine of IP-10, which decreased TJ proteins and damaged BBB integrity. Further study indicated that JEV infection can stimulate upregulation of the IP-10 receptor CXCR3 on astrocytes, resulting in TNF-α production through the JNK-c-Jun signaling pathway. Consequently, TNF-α affected the expression and cellular distribution of TJs in brain microvascular endothelial cells and led to BBB damage during JEV infection. Regarding regulation of the BBB, the IP-10/TNF-α cytokine axis could be considered a potential target for the development of novel therapeutics in BBB-related neurological diseases.
Collapse
Affiliation(s)
- Ke Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Haili Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Wenjuan Lou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Longhuan Ma
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yunchuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Nan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Chong Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Fang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Muhammad Awais
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China.,College of Veterinary and Animal Sciences Jhang, Jhang, Pakistan
| | - Shengbo Cao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Ruiping She
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhen F Fu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China.,Departments of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Min Cui
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| |
Collapse
|
25
|
Jin P, Shin SH, Chun YS, Shin HW, Shin YJ, Lee Y, Kim D, Nam DH, Park JW. Astrocyte-derived CCL20 reinforces HIF-1-mediated hypoxic responses in glioblastoma by stimulating the CCR6-NF-κB signaling pathway. Oncogene 2018. [PMID: 29535421 DOI: 10.1038/s41388-018-0182-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
During tumor development, stromal cells are co-opted to the tumor milieu and provide favorable conditions for the tumor. Hypoxia stimulates cancer cells to acquire a more malignant phenotype via activation of hypoxia-inducible factor 1 (HIF-1). Given that cancer cells and astrocytes in glioblastomas coexist in a hypoxic microenvironment, we examined whether astrocytes affect the adaptation of glioblastoma cells to hypoxia. Immunoblotting, reporter assays, quantitative RT-PCR, and chromatin immunoprecipitation were performed to evaluate HIF-1 signaling in glioblastoma cells. Astrocyte-derived chemokine C-C motif ligand 20 (CCL20) was identified using cytokine arrays, and its role in glioblastoma development was evaluated in orthotopic xenografts. Astrocytes augmented HIF-1α expression in glioblastoma cells under hypoxia. The expression of HIF-1 downstream genes, cancer colony formation, and Matrigel invasion of glioblastoma cells were stimulated by conditioned medium from astrocytes pre-exposed to hypoxia. CCL20 was secreted in a hypoxia-dependent manner from astrocytes and busted the hypoxic induction of HIF-1α in glioblastoma cells. Mechanistically, the CCL20/CCR6 signaling pathway upregulates HIF-1α by stimulating nuclear factor kappa B-driven transactivation of the HIF1A gene. Compared with the control tumors, CCR6-deficient glioblastoma xenografts grew more slowly, with poor vascularization, and expressed lower levels of HIF-1α and its downstream proteins. Furthermore, CCR6 expression was correlated with HIF-1α expression in GEO and TCGA datasets from human glioblastoma tissues. These results suggest that glioblastoma cells adapt well to hypoxic stress by virtue of CCL20 derived from neighboring astrocytes.
Collapse
Affiliation(s)
- Peng Jin
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Hyun Shin
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yang-Sook Chun
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Woo Shin
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Jae Shin
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeri Lee
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Donggeon Kim
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Do-Hyun Nam
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea. .,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea. .,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
26
|
Jafarzadeh A, Azizi SV, Arabi Z, Ahangar-Parvin R, Mohammadi-Kordkhayli M, Larussa T, Khatami F, Nemati M. Vitamin D down-regulates the expression of some Th17 cell-related cytokines, key inflammatory chemokines, and chemokine receptors in experimental autoimmune encephalomyelitis. Nutr Neurosci 2018; 22:725-737. [DOI: 10.1080/1028415x.2018.1436237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Abdollah Jafarzadeh
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Sayyed Vahab Azizi
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Arabi
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Rayhaneh Ahangar-Parvin
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Tiziana Larussa
- Department of Health Science, University of Catanzaro ‘Magna Graecia’, Catanzaro, Italy
| | - Fariba Khatami
- Department of Pathology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Maryam Nemati
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
- Department of Laboratory Sciences, Para-Medicine School, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
27
|
Li R, Sun X, Shu Y, Wang Y, Xiao L, Wang Z, Hu X, Kermode AG, Qiu W. Serum CCL20 and its association with SIRT1 activity in multiple sclerosis patients. J Neuroimmunol 2017; 313:56-60. [PMID: 29153609 DOI: 10.1016/j.jneuroim.2017.10.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 01/08/2023]
Abstract
CCL20 is a potentially important component in the pathogenesis of multiple sclerosis (MS). SIRT1 exhibits a negative regulatory effect on a variety of inflammatory cytokines and can relieve experimental autoimmune encephalomyelitis. The association between the level of CCL20 and SIRT1 activity in MS patients has not been investigated. In the present study, blood samples were collected from 38 RRMS patients and 40 healthy controls. The serum CCL20 levels were measured by ELISA. SIRT1 activity was evaluated by fluorometric assay. We revealed elevated serum CCL20 concentrations in MS, and discovered an inverse correlation between CCL20 and SIRT1 activity in MS patients.
Collapse
Affiliation(s)
- Rui Li
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xiaobo Sun
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yaqing Shu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yuge Wang
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Li Xiao
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhanhang Wang
- Department of Neurology, Guangdong 999 Brain Hospital, China
| | - Xueqiang Hu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Allan G Kermode
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China; Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Department of Neurology, Sir Charles Gairdner Hospital, Queen Elizabeth II Medical Centre, Perth, Australia; Institute of Immunology and Infectious Diseases, Murdoch University, Perth, Australia
| | - Wei Qiu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
28
|
Natural and induced immunization against CCL20 ameliorate experimental autoimmune encephalitis and may confer protection against multiple sclerosis. Clin Immunol 2017; 183:316-324. [DOI: 10.1016/j.clim.2017.09.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 09/11/2017] [Accepted: 09/17/2017] [Indexed: 12/24/2022]
|
29
|
Nacka-Aleksić M, Stojić-Vukanić Z, Pilipović I, Vujnović I, Bufan B, Dimitrijević M, Leposavić G. Strain specificities in cellular and molecular immunopathogenic mechanisms underlying development of experimental autoimmune encephalomyelitis in aged rats. Mech Ageing Dev 2017; 164:146-163. [DOI: 10.1016/j.mad.2017.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/21/2017] [Accepted: 03/03/2017] [Indexed: 11/29/2022]
|
30
|
Stojić-Vukanić Z, Pilipović I, Vujnović I, Nacka-Aleksić M, Petrović R, Arsenović-Ranin N, Dimitrijević M, Leposavić G. GM-CSF-Producing Th Cells in Rats Sensitive and Resistant to Experimental Autoimmune Encephalomyelitis. PLoS One 2016; 11:e0166498. [PMID: 27832210 PMCID: PMC5104330 DOI: 10.1371/journal.pone.0166498] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 10/28/2016] [Indexed: 12/13/2022] Open
Abstract
Given that granulocyte macrophage colony-stimulating factor (GM-CSF) is identified as the key factor to endow auto-reactive Th cells with the potential to induce neuroinflammation in experimental autoimmune encephalomyelitis (EAE) models, the frequency and phenotype of GM-CSF-producing (GM-CSF+) Th cells in draining lymph nodes (dLNs) and spinal cord (SC) of Albino Oxford (AO) and Dark Agouti (DA) rats immunized for EAE were examined. The generation of neuroantigen-specific GM-CSF+ Th lymphocytes was impaired in dLNs of AO rats (relatively resistant to EAE induction) compared with their DA counterparts (susceptible to EAE) reflecting impaired CD4+ lymphocyte proliferation and less supportive of GM-CSF+ Th cell differentiation dLN cytokine microenvironment. Immunophenotyping of GM-CSF+ Th cells showed their phenotypic heterogeneity in both strains and revealed lower frequency of IL-17+IFN-γ+, IL-17+IFN-γ-, and IL-17-IFN-γ+ cells accompanied by higher frequency of IL-17-IFN-γ- cells among them in AO than in DA rats. Compared with DA, in AO rats was also found (i) slightly lower surface density of CCR2 (drives accumulation of highly pathogenic GM-CSF+IFN-γ+ Th17 cells in SC) on GM-CSF+IFN-γ+ Th17 lymphocytes from dLNs, and (ii) diminished CCL2 mRNA expression in SC tissue, suggesting their impaired migration into the SC. Moreover, dLN and SC cytokine environments in AO rats were shown to be less supportive of GM-CSF+IFN-γ+ Th17 cell differentiation (judging by lower expression of mRNAs for IL-1β, IL-6 and IL-23/p19). In accordance with the (i) lower frequency of GM-CSF+ Th cells in dLNs and SC of AO rats and their lower GM-CSF production, and (ii) impaired CCL2 expression in the SC tissue, the proportion of proinflammatory monocytes among peripheral blood cells and their progeny (CD45hi cells) among the SC CD11b+ cells were reduced in AO compared with DA rats. Collectively, the results indicate that the strain specificities in efficacy of several mechanisms controlling (auto)reactive CD4+ lymphocyte expansion/differentiation into the cells with pathogenic phenotype and migration of the latter to the SC contribute to AO rat resistance to EAE.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Center “Branislav Janković”, Institute of Virology, Vaccines and Sera “Torlak”, Belgrade, Serbia
| | - Ivana Vujnović
- Immunology Research Center “Branislav Janković”, Institute of Virology, Vaccines and Sera “Torlak”, Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Department of Physiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Raisa Petrović
- Immunology Research Center “Branislav Janković”, Institute of Virology, Vaccines and Sera “Torlak”, Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Gordana Leposavić
- Immunology Research Center “Branislav Janković”, Institute of Virology, Vaccines and Sera “Torlak”, Belgrade, Serbia
- Department of Physiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
31
|
Guerrini MM, Okamoto K, Komatsu N, Sawa S, Danks L, Penninger JM, Nakashima T, Takayanagi H. Inhibition of the TNF Family Cytokine RANKL Prevents Autoimmune Inflammation in the Central Nervous System. Immunity 2015; 43:1174-85. [PMID: 26680207 DOI: 10.1016/j.immuni.2015.10.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 08/25/2015] [Accepted: 10/26/2015] [Indexed: 12/11/2022]
Abstract
The central nervous system (CNS) is an immunologically privileged site protected from uncontrolled access of T cells by the blood-brain barrier (BBB), which is breached upon autoimmune inflammation. Here we have shown that receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL) on T cells regulates C-C type chemokine ligand 20 (CCL20) production by astrocytes and T cell localization in the CNS. Importantly, mice specifically lacking RANKL in T cells were resistant to experimental autoimmune encephalomyelitis (EAE) due to altered T cell trafficking. Pharmacological inhibition of RANKL prevented the development of EAE without affecting the peripheral immune response, indicating that RANKL is a potential therapeutic target for treating autoimmune diseases in the CNS.
Collapse
Affiliation(s)
- Matteo M Guerrini
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan; Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO) Program, Takayanagi Osteonetwork Project, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kazuo Okamoto
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan; Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO) Program, Takayanagi Osteonetwork Project, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shinichiro Sawa
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan; Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO) Program, Takayanagi Osteonetwork Project, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Lynett Danks
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan; Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO) Program, Takayanagi Osteonetwork Project, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Josef M Penninger
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030, Vienna, Austria
| | - Tomoki Nakashima
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan; Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO) Program, Takayanagi Osteonetwork Project, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
32
|
Jha MK, Lee WH, Suk K. Functional polarization of neuroglia: Implications in neuroinflammation and neurological disorders. Biochem Pharmacol 2015; 103:1-16. [PMID: 26556658 DOI: 10.1016/j.bcp.2015.11.003] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
Abstract
Recent neuroscience research has established the adult brain as a dynamic organ having a unique ability to undergo changes with time. Neuroglia, especially microglia and astrocytes, provide dynamicity to the brain. Activation of these glial cells is a major component of the neuroinflammatory responses underlying brain injury and neurodegeneration. Glial cells execute functional reaction programs in response to diverse microenvironmental signals manifested by neuropathological conditions. Activated microglia exist along a continuum of two functional states of polarization namely M1-type (classical/proinflammatory activation) and M2-type (alternative/anti-inflammatory activation) as in macrophages. The balance between classically and alternatively activated microglial phenotypes influences disease progression in the CNS. The classically activated state of microglia drives the neuroinflammatory response and mediates the detrimental effects on neurons, whereas in their alternative activation state, which is apparently a beneficial activation state, the microglia play a crucial role in tissue maintenance and repair. Likewise, in response to immune or inflammatory microenvironments astrocytes also adopt neurotoxic or neuroprotective phenotypes. Reactive astrocytes exhibit two distinctive functional phenotypes defined by pro- or anti-inflammatory gene expression profile. In this review, we have thoroughly covered recent advances in the understanding of the functional polarization of brain and peripheral glia and its implications in neuroinflammation and neurological disorders. The identifiable phenotypes adopted by neuroglia in response to specific insult or injury can be exploited as promising diagnostic markers of neuroinflammatory diseases. Furthermore, harnessing the beneficial effects of the polarized glia could undoubtedly pave the way for the formulation of novel glia-based therapeutic strategies for diverse neurological disorders.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
33
|
Borgmann K, Ghorpade A. HIV-1, methamphetamine and astrocytes at neuroinflammatory Crossroads. Front Microbiol 2015; 6:1143. [PMID: 26579077 PMCID: PMC4621459 DOI: 10.3389/fmicb.2015.01143] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022] Open
Abstract
As a popular psychostimulant, methamphetamine (METH) use leads to long-lasting, strong euphoric effects. While METH abuse is common in the general population, between 10 and 15% of human immunodeficiency virus-1 (HIV-1) patients report having abused METH. METH exacerbates the severity and onset of HIV-1-associated neurocognitive disorders (HAND) through direct and indirect mechanisms. Repetitive METH use impedes adherence to antiretroviral drug regimens, increasing the likelihood of HIV-1 disease progression toward AIDS. METH exposure also directly affects both innate and adaptive immunity, altering lymphocyte numbers and activity, cytokine signaling, phagocytic function and infiltration through the blood brain barrier. Further, METH triggers the dopamine reward pathway and leads to impaired neuronal activity and direct toxicity. Concurrently, METH and HIV-1 alter the neuroimmune balance and induce neuroinflammation, which modulates a wide range of brain functions including neuronal signaling and activity, glial activation, viral infection, oxidative stress, and excitotoxicity. Pathologically, reactive gliosis is a hallmark of both HIV-1- and METH-associated neuroinflammation. Significant commonality exists in the neurotoxic mechanisms for both METH and HAND; however, the pathways dysregulated in astroglia during METH exposure are less clear. Thus, this review highlights alterations in astrocyte intracellular signaling pathways, gene expression and function during METH and HIV-1 comorbidity, with special emphasis on HAND-associated neuroinflammation. Importantly, this review carefully evaluates interventions targeting astrocytes in HAND and METH as potential novel therapeutic approaches. This comprehensive overview indicates, without a doubt, that during HIV-1 infection and METH abuse, a complex dialog between all neural cells is orchestrated through astrocyte regulated neuroinflammation.
Collapse
Affiliation(s)
- Kathleen Borgmann
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| |
Collapse
|
34
|
Abstract
Multiple sclerosis is a neurologic disease caused by immune cell infiltration into the central nervous system, resulting in gray and white matter inflammation, progressive demyelination, and neuronal loss. Astrocytes, the most abundant cell population in the central nervous system (CNS), have been considered inert scaffold or housekeeping cells for many years. However, recently, it has become clear that this cell population actively modulates the immune response in the CNS at multiple levels. While being exposed to a plethora of cytokines during ongoing autoimmune inflammation, astrocytes modulate local CNS inflammation by secreting cytokines and chemokines, among other factors. This review article gives an overview of the most recent understanding about cytokine networks operational in astrocytes during autoimmune neuroinflammation and highlights potential targets for immunomodulatory therapies for multiple sclerosis.
Collapse
Affiliation(s)
- Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Ave. Louis Pasteur, HIM 714, Boston, MA, 02115, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Ave. Louis Pasteur, HIM 714, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Marsigliante S, Vetrugno C, Muscella A. Paracrine CCL20 loop induces epithelial-mesenchymal transition in breast epithelial cells. Mol Carcinog 2015; 55:1175-86. [DOI: 10.1002/mc.22360] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 05/26/2015] [Accepted: 06/15/2015] [Indexed: 12/30/2022]
Affiliation(s)
- S. Marsigliante
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.); Laboratorio di Fisiologia Cellulare; Università del Salento; Via Provinciale per Monteroni; Lecce Italy
| | - C. Vetrugno
- Unità di Neuropatologia; Istituto di Neurologia sperimentale e Divisione di Neuroscienze; Istituto Scientifico IRCCS San Raffaele; Milano Italy
| | - A. Muscella
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.); Laboratorio di Patologia Molecolare; Università del Salento; Via Provinciale per Monteroni; Lecce Italy
| |
Collapse
|
36
|
Zhao L, Xia J, Wang X, Xu F. Transcriptional regulation of CCL20 expression. Microbes Infect 2014; 16:864-70. [DOI: 10.1016/j.micinf.2014.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/03/2014] [Accepted: 08/07/2014] [Indexed: 12/19/2022]
|
37
|
Mony JT, Khorooshi R, Owens T. Chemokine receptor expression by inflammatory T cells in EAE. Front Cell Neurosci 2014; 8:187. [PMID: 25071447 PMCID: PMC4081975 DOI: 10.3389/fncel.2014.00187] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/19/2014] [Indexed: 11/13/2022] Open
Abstract
Chemokines direct cellular infiltration to tissues, and their receptors and signaling pathways represent targets for therapy in diseases such as multiple sclerosis (MS). The chemokine CCL20 is expressed in choroid plexus, a site of entry of T cells to the central nervous system (CNS). The CCL20 receptor CCR6 has been reported to be selectively expressed by CD4(+) T cells that produce the cytokine IL-17 (Th17 cells). Th17 cells and interferon-gamma (IFNγ)-producing Th1 cells are implicated in induction of MS and its animal model experimental autoimmune encephalomyelitis (EAE). We have assessed whether CCR6 identifies specific inflammatory T cell subsets in EAE. Our approach was to induce EAE, and then examine chemokine receptor expression by cytokine-producing T cells sorted from CNS at peak disease. About 7% of CNS-infiltrating CD4(+) T cells produced IFNγ in flow cytometric cytokine assays, whereas less than 1% produced IL-17. About 1% of CD4(+) T cells produced both cytokines. CCR6 was expressed by Th1, Th1+17 and by Th17 cells, but not by CD8(+) T cells. CD8(+) T cells expressed CXCR3, which was also expressed by CD4(+) T cells, with no correlation to cytokine profile. Messenger RNA for IFNγ, IL-17A, and the Th1 and Th17-associated transcription factors T-bet and RORγt was detected in both CCR6(+) and CXCR3(+) CD4(+) T cells. IFNγ, but not IL-17A mRNA expression was detected in CD8(+) T cells in CNS. CCR6 and CD4 were co-localized in spinal cord infiltrates by double immunofluorescence. Consistent with flow cytometry data some but not all CD4(+) T cells expressed CCR6 within infiltrates. CD4-negative CCR6(+) cells included macrophage/microglial cells. Thus we have for the first time directly studied CD4(+) and CD8(+) T cells in the CNS of mice with peak EAE, and determined IFNγ and IL17 expression by cells expressing CCR6 and CXCR3. We show that neither CCR6 or CXCR3 align with CD4 T cell subsets, and Th1 or mixed Th1+17 predominate in EAE.
Collapse
Affiliation(s)
- Jyothi Thyagabhavan Mony
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Reza Khorooshi
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Trevor Owens
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| |
Collapse
|
38
|
García-Berrocoso T, Giralt D, Llombart V, Bustamante A, Penalba A, Flores A, Ribó M, Molina CA, Rosell A, Montaner J. Chemokines after human ischemic stroke: From neurovascular unit to blood using protein arrays. TRANSLATIONAL PROTEOMICS 2014. [DOI: 10.1016/j.trprot.2014.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
39
|
Guo MF, Meng J, Li YH, Yu JZ, Liu CY, Feng L, Yang WF, Li JL, Feng QJ, Xiao BG, Ma CG. The inhibition of Rho kinase blocks cell migration and accumulation possibly by challenging inflammatory cytokines and chemokines on astrocytes. J Neurol Sci 2014; 343:69-75. [PMID: 24952673 DOI: 10.1016/j.jns.2014.05.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 04/17/2014] [Accepted: 05/14/2014] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), are autoimmune diseases characterized by the immune-mediated demyelination and neurodegeneration of the CNS. Our previous studies showed that Rho kinase inhibitor Fasudil can delay onset, and ameliorate severity of EAE, accompanied by the improvement in myelination and the inhibition of inflammatory responses in the CNS. In this study, we found that Fasudil inhibited the migration of T cells indirectly by affecting the production of inflammatory factors and the expression of chemokines in astrocytes functions, indicating that Fasudil treatment reduced inflammatory cytokines such as TNF-α and IL-6, reactive oxygen species (NO) and chemokines like MIP-3α (CCL-20), RANTES (CCL5), MIP-1α (CCL-3) and MCP-1 (CCL2) in vitro, and blocked the chemotaxis of reactive mononuclear cells in EAE mice. Further studies found that Fasudil treatment reduced the infiltration and accumulation of pathogenic T cells into the CNS. Astrocytes expressing GFAP and CCL-20 were inhibited in Fasudil-treated EAE compared with control mice. These results demonstrate that Fasudil alleviates the pathogenesis of EAE possibly by blocking astrocyte-derived chemokine-mediated migration of inflammatory macrophages and pathogenic T cells, and might be used to treat MS.
Collapse
Affiliation(s)
- Min-Fang Guo
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Jian Meng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Yan-Hua Li
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Jie-Zhong Yu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Chun-Yun Liu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Ling Feng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Wan-Fang Yang
- Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Jun-Lian Li
- Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Qian-Jin Feng
- Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| | - Cun-Gen Ma
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China; Department of Encephalopathy and National Major Clinical Department of Ministry of Health, Third Hospital, Department of Neurology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China.
| |
Collapse
|
40
|
Li R, Xu W, Chen Y, Qiu W, Shu Y, Wu A, Dai Y, Bao J, Lu Z, Hu X. Raloxifene suppresses experimental autoimmune encephalomyelitis and NF-κB-dependent CCL20 expression in reactive astrocytes. PLoS One 2014; 9:e94320. [PMID: 24722370 PMCID: PMC3983123 DOI: 10.1371/journal.pone.0094320] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/14/2014] [Indexed: 12/19/2022] Open
Abstract
Recent clinical data have led to the consideration of sexual steroids as new potential therapeutic tools for multiple sclerosis. Selective estrogen receptor modulators can exhibit neuroprotective effects like estrogen, with fewer systemic estrogen side effects than estrogen, offering a more promising therapeutic modality for multiple sclerosis. The important role of astrocytes in a proinflammatory effect mediated by CCL20 signaling on inflammatory cells has been documented. Their potential contribution to selective estrogen receptor modulator-mediated protection is still unknown. Using a mouse model of chronic neuroinflammation, we report that raloxifene, a selective estrogen receptor modulator, alleviated experimental autoimmune encephalomyelitis–an animal model of multiple sclerosis–and decreased astrocytic production of CCL20. Enzyme-linked immunosorbent assay, immunohistochemistry imaging and transwell migration assays revealed that reactive astrocytes express CCL20, which promotes Th17 cell migration. In cultured rodent astrocytes, raloxifene inhibited IL-1β-induced CCL20 expression and chemotaxis ability for Th17 migration, whereas the estrogen receptor antagonist ICI 182,780 blocked this effect. Western blotting further indicated that raloxifene suppresses IL-1β-induced NF-κB activation (phosphorylation of p65) and translocation but does not affect phosphorylation of IκB. In conclusion, these data demonstrate that raloxifene provides robust neuroprotection against experimental autoimmune encephalomyelitis, partially via an inhibitory action on CCL20 expression and NF-κB pathways in reactive astrocytes. Our results contribute to a better understanding of the critical roles of raloxifene in treating experimental autoimmune encephalomyelitis and uncover reactive astrocytes as a new target for the inhibitory action of estrogen receptors on chemokine CCL20 expression.
Collapse
MESH Headings
- Animals
- Astrocytes/drug effects
- Astrocytes/pathology
- Cell Movement/drug effects
- Cells, Cultured
- Chemokine CCL20/antagonists & inhibitors
- Chemokine CCL20/genetics
- Chemokine CCL20/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Female
- Fulvestrant
- Gene Expression Regulation
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/genetics
- Multiple Sclerosis/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Raloxifene Hydrochloride/pharmacology
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/genetics
- Receptors, Estrogen/immunology
- Selective Estrogen Receptor Modulators/pharmacology
- Signal Transduction
- Th17 Cells/drug effects
- Th17 Cells/pathology
Collapse
Affiliation(s)
- Rui Li
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wen Xu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Ying Chen
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wei Qiu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yaqing Shu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Aimin Wu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yongqiang Dai
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Jian Bao
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhengqi Lu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xueqiang Hu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- * E-mail:
| |
Collapse
|
41
|
Interactions between neutrophils, Th17 cells, and chemokines during the initiation of experimental model of multiple sclerosis. Mediators Inflamm 2014; 2014:590409. [PMID: 24692851 PMCID: PMC3945772 DOI: 10.1155/2014/590409] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 01/07/2014] [Accepted: 01/12/2014] [Indexed: 11/18/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis (MS) in which activated T cell and neutrophil interactions lead to neuroinflammation. In this study the expression of CCR6, CXCR2, and CXCR6 in Th17 cells and neutrophils migrating to the brain during EAE was measured, alongside an evaluation of the production of IL-17, IL-23, CCL-20, and CXCL16 in the brain. Next, inflammatory cell subpopulations accumulating in the brain after intracerebral injections of IL-17 or CXCL1, as well as during modulation of EAE with anti-IL-23R or anti-CXCR2 antibodies, were analyzed. Th17 cells upregulate CXCR2 during the preclinical phase of EAE and a significant migration of these cells to the brain was observed. Neutrophils upregulated CCR6, CXCR2, and CXCR6 during EAE, accumulating in the brain both prior to and during acute EAE attacks. Production of IL-17, IL-23, CCL20, and CXCL16 in the CNS was increased during both preclinical and acute EAE. Intracerebral delivery of CXCL1 stimulated the early accumulation of neutrophils in normal and preclinical EAE brains but reduced the migration of Th17 cells to the brain during the preclinical stage of EAE. Modulation of EAE by anti-IL-23R antibodies ameliorated EAE by decreasing the intracerebral accumulation of Th17 cells.
Collapse
|
42
|
Glabinski A, Jalosinski M, Ransohoff RM. Chemokines and chemokine receptors in inflammation of the CNS. Expert Rev Clin Immunol 2014; 1:293-301. [DOI: 10.1586/1744666x.1.2.293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
43
|
Higher Circulating Levels of Chemokine CCL20 in Patients with Multiple Sclerosis: Evaluation of the Influences of Chemokine Gene Polymorphism, Gender, Treatment and Disease Pattern. J Mol Neurosci 2014; 53:500-5. [DOI: 10.1007/s12031-013-0214-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 12/15/2013] [Indexed: 01/01/2023]
|
44
|
Enteric neural disruption in necrotizing enterocolitis occurs in association with myenteric glial cell CCL20 expression. J Pediatr Gastroenterol Nutr 2013; 57:788-93. [PMID: 24280992 DOI: 10.1097/mpg.0b013e3182a86fd4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE The aetiology of necrotising enterocolitis (NEC) is unknown, but luminal factors and epithelial leakiness appear critical triggers of an inflammatory cascade. A separate finding has been suggested in mouse models, in which disruption of glial cells in the myenteric plexus induced a severe NEC-like lesion. We have thus looked for evidence of neuroglial abnormality in NEC. METHODS We studied full-thickness resected specimens from 20 preterm infants with acute NEC and from 13 control infants undergoing resection for other indications. Immunohistochemical analysis was performed for immunological (CD3, syndecan-1, human leucocyte antigen-DR), neural (glial fibrillary acidic protein [GFAP], nerve growth factor receptor, neurofilament protein, neuron-specific enolase), and functional markers (Ki67), and for potential inflammatory regulators (interleukin-12, transforming growth factor-β, CCL20, CCR6). RESULTS Expression of the chemokine CCL20 and its receptor CCR6 was significantly upregulated in myenteric plexus in NEC, with CCL20 strongly expressed by glial cells. In 9 of 20 cases with NEC, myenteric plexus architecture and GFAP+ glial cells were normal, with preserved submucosal and mucosal innervation; however, 11 cases showed disrupted myenteric plexus architecture, reduced GFAP expression, and loss of submucosal and mucosal innervation. Persistent abnormalities were identified in the 2 infants who had ongoing inflammation at ileostomy closure. CONCLUSIONS Our findings identified heterogeneity among patients with NEC. Approximately half showed evidence of marked neural abnormality extending from the deeper layers of the intestine, associated with glial activation and myenteric plexus disruption. The factors that may activate enteric glia in this manner, potentially including bacterial products or viruses, remain to be determined.
Collapse
|
45
|
Comerford I, Kara EE, McKenzie DR, McColl SR. Advances in understanding the pathogenesis of autoimmune disorders: focus on chemokines and lymphocyte trafficking. Br J Haematol 2013; 164:329-41. [PMID: 24164387 DOI: 10.1111/bjh.12616] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Lymphocyte trafficking is a key step in the pathogenesis of various autoimmune diseases. Recruitment of autoreactive lymphocytes to inflamed tissues is a defining feature of numerous persistent organ-specific autoimmune conditions and various therapies are now used in several of these diseases which appear to specifically block lymphocyte migration. Thus, better understanding of the molecular events involved in homing of autoreactive pathogenic lymphocytes may present novel opportunities for pharmacological intervention in autoimmune diseases, such as multiple sclerosis, rheumatoid arthritis, type-1 diabetes and psoriasis. This review describes recent progress in understanding lymphocyte trafficking in autoimmunity, focusing on the involvement of the chemokine and chemokine receptor superfamily. Possible strategies to improve therapeutics for autoimmune diseases arising from these studies are discussed.
Collapse
Affiliation(s)
- Iain Comerford
- Chemokine Biology Laboratory, School of Molecular and Biomedical Science, Centre for Molecular Pathology, The University of Adelaide, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
46
|
Sulkowski G, Dąbrowska-Bouta B, Chalimoniuk M, Strużyńska L. Effects of antagonists of glutamate receptors on pro-inflammatory cytokines in the brain cortex of rats subjected to experimental autoimmune encephalomyelitis. J Neuroimmunol 2013; 261:67-76. [PMID: 23746391 DOI: 10.1016/j.jneuroim.2013.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/18/2013] [Accepted: 05/10/2013] [Indexed: 10/26/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Inflammatory cytokines and glutamate neurotoxicity have been proposed as major determinants accompanying the demyelination and axonal degeneration observed during the course of MS. The present study using the animal model of MS known as experimental autoimmune encephalomyelitis (EAE) demonstrates that pharmacological inhibition of ionotropic NMDA glutamate receptors by their antagonists (amantadine and memantine) suppresses neurological symptoms of disease in EAE rats and reduces expression of pro-inflammatory cytokines in the brain. Conversely, antagonists of group I metabotropic glutamate receptors, mGluRs (LY 367385 and MPEP), do not affect the inflammatory process and the neurological condition of EAE rats.
Collapse
Affiliation(s)
- Grzegorz Sulkowski
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawińskiego str., 02-106 Warsaw, Poland.
| | | | | | | |
Collapse
|
47
|
Moriguchi K, Miyamoto K, Tanaka N, Yoshie O, Kusunoki S. The importance of CCR4 and CCR6 in experimental autoimmune encephalomyelitis. J Neuroimmunol 2013; 257:53-8. [PMID: 23477966 DOI: 10.1016/j.jneuroim.2013.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/19/2013] [Accepted: 02/11/2013] [Indexed: 01/12/2023]
Abstract
Chemokine receptors (CCRs) play important roles in the pathogenesis of immune-mediated diseases, as well as in normal immune response. We examined the role of CCR6 and CCR4 in experimental autoimmune encephalomyelitis (EAE) by using CCR6(-/-)CCR4(-/-) double knockout (DKO) and single knockout mice. DKO mice developed less severe EAE and presented repressed recall response in the induction phase, especially in the activity of T helper 17 (Th17) cells. CCR6 expression in central nervous system (CNS)-infiltrated cells was diminished in DKO. Our results suggest that CCR6 and CCR4 were involved in a more rapid progression of EAE and that their regulation might be a therapeutic target of human inflammatory demyelinating diseases.
Collapse
Affiliation(s)
- Kota Moriguchi
- Division of Neurology, Department of Internal Medicine 3, National Defense Medical College, Tokorozawa, Japan
| | | | | | | | | |
Collapse
|
48
|
Sagar D, Foss C, El Baz R, Pomper MG, Khan ZK, Jain P. Mechanisms of dendritic cell trafficking across the blood-brain barrier. J Neuroimmune Pharmacol 2012; 7:74-94. [PMID: 21822588 PMCID: PMC3276728 DOI: 10.1007/s11481-011-9302-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 07/25/2011] [Indexed: 12/14/2022]
Abstract
Although the central nervous system (CNS) is considered to be an immunoprivileged site, it is susceptible to a host of autoimmune as well as neuroinflammatory disorders owing to recruitment of immune cells across the blood-brain barrier into perivascular and parenchymal spaces. Dendritic cells (DCs), which are involved in both primary and secondary immune responses, are the most potent immune cells in terms of antigen uptake and processing as well as presentation to T cells. In light of the emerging importance of DC traficking into the CNS, these cells represent good candidates for targeted immunotherapy against various neuroinflammatory diseases. This review focuses on potential physiological events and receptor interactions between DCs and the microvascular endothelial cells of the brain as they transmigrate into the CNS during degeneration and injury. A clear understanding of the underlying mechanisms involved in DC migration may advance the development of new therapies that manipulate these mechanistic properties via pharmacologic intervention. Furthermore, therapeutic validation should be in concurrence with the molecular imaging techniques that can detect migration of these cells in vivo. Since the use of noninvasive methods to image migration of DCs into CNS has barely been explored, we highlighted potential molecular imaging techniques to achieve this goal. Overall, information provided will bring this important leukocyte population to the forefront as key players in the immune cascade in the light of the emerging contribution of DCs to CNS health and disease.
Collapse
Affiliation(s)
- Divya Sagar
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Catherine Foss
- Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Rasha El Baz
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Martin G. Pomper
- Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Zafar K. Khan
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Pooja Jain
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
- Department of Microbiology & Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, 3805 Old Easton Road, Doylestown, PA 18902, USA
| |
Collapse
|
49
|
Meares GP, Ma X, Qin H, Benveniste EN. Regulation of CCL20 expression in astrocytes by IL-6 and IL-17. Glia 2012; 60:771-81. [PMID: 22319003 DOI: 10.1002/glia.22307] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 01/13/2012] [Accepted: 01/21/2012] [Indexed: 12/22/2022]
Abstract
Astrocytes have an important role in the regulation of inflammation within the central nervous system (CNS). In neuroinflammatory conditions such as multiple sclerosis, numerous cytokines and chemokines are elevated including IL-6, IL-17, and CCL20. IL-17 enhances IL-6 signaling and subsequent IL-6 expression in astrocytes. CCL20 is a CC motif chemokine that functions as a chemoattractant to facilitate the recruitment of CCR6-expressing cells, including Th17 cells. In this study, we examined the role of IL-6 and IL-17 on CCL20 production in primary murine astrocytes. IL-6 in combination with the IL-6 soluble receptor (sIL-6R) stimulated CCL20 expression in part through STAT3 activation, whereas IL-17 alone had no effect. However, the combination of IL-6, sIL-6R, and IL-17 led to a robust increase in CCL20 production. IL-17 increased the activation-associated phosphorylation of NF-κB, and inhibition of the NF-κB pathway significantly inhibited the enhancement of CCL20 expression by IL-17. In addition, chromatin immunoprecipitation revealed that stimulation of primary astrocytes with IL-6 plus the sIL-6R induced STAT3 binding to the CCL20 promoter. Combined stimulation with IL-6, sIL-6R, and IL-17 increased the recruitment of phosphorylated NF-κB to the CCL20 promoter, increased binding of coactivators such as p300 and CBP, and enhanced H3 and H4 histone acetylation, consistent with a transcriptionally active gene. The astrocyte-produced CCL20 increased T cell migration as determined by transwell migration assay. Collectively, these results suggest that astrocytes, in response to IL-6, sIL-6R, and IL-17, may shift chemokine production to that favoring T cell recruitment to the CNS.
Collapse
Affiliation(s)
- Gordon P Meares
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | |
Collapse
|
50
|
Dalgard CL, Cole JT, Kean WS, Lucky JJ, Sukumar G, McMullen DC, Pollard HB, Watson WD. The cytokine temporal profile in rat cortex after controlled cortical impact. Front Mol Neurosci 2012; 5:6. [PMID: 22291617 PMCID: PMC3265961 DOI: 10.3389/fnmol.2012.00006] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 01/12/2012] [Indexed: 12/30/2022] Open
Abstract
Cerebral inflammatory responses may initiate secondary cascades following traumatic brain injury (TBI). Changes in the expression of both cytokines and chemokines may activate, regulate, and recruit innate and adaptive immune cells associated with secondary degeneration, as well as alter a host of other cellular processes. In this study, we quantified the temporal expression of a large set of inflammatory mediators in rat cortical tissue after brain injury. Following a controlled cortical impact (CCI) on young adult male rats, cortical and hippocampal tissue of the injured hemisphere and matching contralateral material was harvested at early (4, 12, and 24 hours) and extended (3 and 7 days) time points post-procedure. Naïve rats that received only anesthesia were used as controls. Processed brain homogenates were assayed for chemokine and cytokine levels utilizing an electrochemiluminescence-based multiplex ELISA platform. The temporal profile of cortical tissue samples revealed a multi-phasic injury response following brain injury. CXCL1, IFN-γ, TNF-α levels significantly peaked at four hours post-injury compared to levels found in naïve or contralateral tissue. CXCL1, IFN-γ, and TNF-α levels were then observed to decrease at least 3-fold by 12 hours post-injury. IL-1β, IL-4, and IL-13 levels were also significantly elevated at four hours post-injury although their expression did not decrease more than 3-fold for up to 24 hours post-injury. Additionally, IL-1β and IL-4 levels displayed a biphasic temporal profile in response to injury, which may suggest their involvement in adaptive immune responses. Interestingly, peak levels of CCL2 and CCL20 were not observed until after four hours post-injury. CCL2 levels in injured cortical tissue were significantly higher than peak levels of any other inflammatory mediator measured, thus suggesting a possible use as a biomarker. Fully elucidating chemokine and cytokine signaling properties after brain injury may provide increased insight into a number of secondary cascade events that are initiated or regulated by inflammatory responses.
Collapse
Affiliation(s)
- Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda MD, USA
| | | | | | | | | | | | | | | |
Collapse
|