201
|
Moehle MS, West AB. M1 and M2 immune activation in Parkinson's Disease: Foe and ally? Neuroscience 2014; 302:59-73. [PMID: 25463515 DOI: 10.1016/j.neuroscience.2014.11.018] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/03/2014] [Accepted: 11/06/2014] [Indexed: 12/20/2022]
Abstract
Parkinson's Disease (PD) is a chronic and progressive neurodegenerative disorder of unknown etiology. Autopsy findings, genetics, retrospective studies, and molecular imaging all suggest a role for inflammation in the neurodegenerative process. However, relatively little is understood about the causes and implications of neuroinflammation in PD. Understanding how inflammation arises in PD, in particular the activation state of cells of the innate immune system, may provide an exciting opportunity for novel neuroprotective therapeutics. We analyze the evidence of immune system involvement in PD susceptibility, specifically in the context of M1 and M2 activation states. Tracking and modulating these activation states may provide new insights into both PD etiology and therapeutic strategies.
Collapse
Affiliation(s)
- M S Moehle
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States.
| | - A B West
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
202
|
Nakagawa Y, Chiba K. Role of microglial m1/m2 polarization in relapse and remission of psychiatric disorders and diseases. Pharmaceuticals (Basel) 2014; 7:1028-48. [PMID: 25429645 PMCID: PMC4276905 DOI: 10.3390/ph7121028] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/27/2022] Open
Abstract
Psychiatric disorders such as schizophrenia and major depressive disorder were thought to be caused by neurotransmitter abnormalities. Patients with these disorders often experience relapse and remission; however the underlying molecular mechanisms of relapse and remission still remain unclear. Recent advanced immunological analyses have revealed that M1/M2 polarization of macrophages plays an important role in controlling the balance between promotion and suppression in inflammation. Microglial cells share certain characteristics with macrophages and contribute to immune-surveillance in the central nervous system (CNS). In this review, we summarize immunoregulatory functions of microglia and discuss a possible role of microglial M1/M2 polarization in relapse and remission of psychiatric disorders and diseases. M1 polarized microglia can produce pro-inflammatory cytokines, reactive oxygen species, and nitric oxide, suggesting that these molecules contribute to dysfunction of neural network in the CNS. Alternatively, M2 polarized microglia express cytokines and receptors that are implicated in inhibiting inflammation and restoring homeostasis. Based on these aspects, we propose a possibility that M1 and M2 microglia are related to relapse and remission, respectively in psychiatric disorders and diseases. Consequently, a target molecule skewing M2 polarization of microglia may provide beneficial therapies for these disorders and diseases in the CNS.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Research Strategy and Planning Department, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| | - Kenji Chiba
- Advanced Medical Research Laboratories, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| |
Collapse
|
203
|
Zhang XM, Lund H, Mia S, Parsa R, Harris RA. Adoptive transfer of cytokine-induced immunomodulatory adult microglia attenuates experimental autoimmune encephalomyelitis in DBA/1 mice. Glia 2014; 62:804-17. [PMID: 24677019 PMCID: PMC4237117 DOI: 10.1002/glia.22643] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 01/20/2014] [Indexed: 12/19/2022]
Abstract
Microglia are resident antigen-presenting cells in the central nervous system (CNS) that either suppress or promote disease depending on their activation phenotype and the microenvironment. Multiple sclerosis (MS) is a chronic inflammatory disease causing demyelination and nerve loss in the CNS, and experimental autoimmune encephalomyelitis (EAE) is an animal model of MS that is widely used to investigate pathogenic mechanisms and therapeutic effects. We isolated and cultured microglia from adult mouse brains and exposed them to specific combinations of stimulatory molecules and cytokines, the combination of IL-4, IL-10, and TGF-β yielding the optimal regime for induction of an immunosuppressive phenotype (M2). M2 microglia were characterized by decreased expression or production of CD86, PD-L1, nitric oxide, and IL-6, increased expression of PD-L2, and having a potent capacity to retain their phenotype on secondary proinflammatory stimulation. M2 microglia induced regulatory T cells, suppressed T-cell proliferation, and downmodulated M1-associated receptor expression in M1 macrophages. Myelin oligodendrocyte glycoprotein (MOG)-induced EAE was induced in DBA/1 mice and at different time points (0, 5, 12, or 15 days postimmunization) 3 × 105 M2 microglia were transferred intranasally. A single transfer of M2 microglia attenuated the severity of established EAE, which was particularly obvious when the cells were injected at 15 days postimmunization. M2 microglia-treated mice had reduced inflammatory responses and less demyelination in the CNS. Our findings demonstrate that adult M2 microglia therapy represents a novel intervention that alleviated established EAE and that this therapeutic principle may have relevance for treatment of MS patients.
Collapse
|
204
|
Hu X, Leak RK, Shi Y, Suenaga J, Gao Y, Zheng P, Chen J. Microglial and macrophage polarization—new prospects for brain repair. Nat Rev Neurol 2014; 11:56-64. [PMID: 25385337 DOI: 10.1038/nrneurol.2014.207] [Citation(s) in RCA: 1018] [Impact Index Per Article: 92.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The traditional view of the adult brain as a static organ has changed in the past three decades, with the emergence of evidence that it remains plastic and has some regenerative capacity after injury. In the injured brain, microglia and macrophages clear cellular debris and orchestrate neuronal restorative processes. However, activation of these cells can also hinder CNS repair and expand tissue damage. Polarization of macrophage populations toward different phenotypes at different stages of injury might account for this dual role. This Perspectives article highlights the specific roles of polarized microglial and macrophage populations in CNS repair after acute injury, and argues that therapeutic approaches targeting cerebral inflammation should shift from broad suppression of microglia and macrophages towards subtle adjustment of the balance between their phenotypes. Breakthroughs in the identification of regulatory molecules that control these phenotypic shifts could ultimately accelerate research towards curing brain disorders.
Collapse
Affiliation(s)
- Xiaoming Hu
- Centre of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Rehana K Leak
- Centre of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Yejie Shi
- Centre of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Jun Suenaga
- Centre of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, 220 Handan Road, Fudan University, Shanghai 200032, China
| | - Ping Zheng
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, 220 Handan Road, Fudan University, Shanghai 200032, China
| | - Jun Chen
- Centre of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
205
|
Liu CY, Guo SD, Yu JZ, Li YH, Zhang H, Feng L, Chai Z, Yuan HJ, Yang WF, Feng QJ, Xiao BG, Ma CG. Fasudil mediates cell therapy of EAE by immunomodulating encephalomyelitic T cells and macrophages. Eur J Immunol 2014; 45:142-52. [PMID: 25287052 DOI: 10.1002/eji.201344429] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 08/28/2014] [Accepted: 09/29/2014] [Indexed: 01/05/2023]
Abstract
Although Fasudil has shown therapeutic potential in EAE mice, the mechanism of action are still not fully understood. Here, we examined the immunomodulatory effect of Fasudil on encephalitogenic mononuclear cells (MNCs), and tested the therapeutic potential of Fasudil-treated MNCs in active EAE. Fasudil inhibited expression of CCL20 on T cells and migration of T cells, decreased CD4(+) IFN-γ(+) and CD4(+) IL-17(+) T cells, but increased CD4(+) IL-10(+) and CD4(+) TGF-β(+) T cells. Fasudil reduced expression of CD16/32 and IL-12, while elevating expression of CD206, CD23, and IL-10. Fasudil also decreased levels of iNOS/NO, enhanced levels of Arg-1, and inhibited the TLR-4/NF-κB signaling and TNF-α, shifting M1 macrophage to M2 phenotype. These modulatory effects of Fasudil on T cells and macrophages were not altered by adding autoantigen MOG35-55 to the culture, i.e., autoantigen-independent. Further, we observed that, in vitro, Fasudil inhibited the capacity of encephalitogenic MNCs to adoptively transfer EAE and reduced TLR-4/p-NF-κB/p65 and inflammatory cytokines in spinal cords. Importantly, Fasudil-treated encephalitogenic MNCs exhibited therapeutic potential when injected into actively induced EAE mice. Together, our results not only provide evidence that Fasudil mediates the polarization of macrophages and the regulation of T cells, but also reveal a novel strategy for cell therapy in MS.
Collapse
Affiliation(s)
- Chun-Yun Liu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Barbour M, Allan D, Xu H, Pei C, Chen M, Niedbala W, Fukada SY, Besnard AG, Alves-Filho JC, Tong X, Forrester JV, Liew FY, Jiang HR. IL-33 attenuates the development of experimental autoimmune uveitis. Eur J Immunol 2014; 44:3320-9. [PMID: 25116404 PMCID: PMC4449115 DOI: 10.1002/eji.201444671] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/25/2014] [Accepted: 08/08/2014] [Indexed: 12/22/2022]
Abstract
Interleukin-33 (IL-33) is associated with several important immune-mediated disorders. However, its role in uveitis, an important eye inflammatory disease, is unknown. Here, we investigated the function of IL-33 in the development of experimental autoimmune uveitis (EAU). IL-33 and IL-33 receptor (ST2) were expressed in murine retinal pigment epithelial (RPE) cells in culture, and IL-33 increased the expression of Il33 and Mcp1 mRNA in RPE cells. In situ, IL-33 was highly expressed in the inner nuclear cells of the retina of naïve mice, and its expression was elevated in EAU mice. ST2-deficient mice developed exacerbated EAU compared with WT mice, and administration of IL-33 to WT mice significantly reduced EAU severity. The attenuated EAU in IL-33-treated mice was accompanied by decreased frequency of IFN-γ+ and IL-17(+) CD4+ T cells and reduced IFN-γ and IL-17 production but with increased frequency of IL-5(+) and IL-4(+) CD4 T cells and IL-5 production in the draining lymph node and spleen. Macrophages from the IL-33-treated mice show a significantly higher polarization toward an alternatively activated macrophage phenotype. Our results therefore demonstrate that the endogenous IL-33/ST2 pathway plays an important role in EAU, and suggest that IL-33 represents a potential option for treatment of uveitis.
Collapse
Affiliation(s)
- Mark Barbour
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of StrathclydeGlasgow, UK
| | - Debbie Allan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of StrathclydeGlasgow, UK
| | - Heping Xu
- Centre for Experimental Medicine, Queen's University BelfastBelfast, UK
| | - Cheng Pei
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of StrathclydeGlasgow, UK
| | - Mei Chen
- Centre for Experimental Medicine, Queen's University BelfastBelfast, UK
| | - Wanda Niedbala
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgow, UK
| | - Sandra Y Fukada
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgow, UK
| | - Anne-Galle Besnard
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgow, UK
| | - Jose C Alves-Filho
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgow, UK
| | - Xiaoguang Tong
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of StrathclydeGlasgow, UK
| | - John V Forrester
- Institute of Medical Science, University of AberdeenUK
- Centre for Ophthalmology and Visual Science, The University of Western AustraliaAustralia
- Centre for Experimental Immunology, Lions Eye InstituteNedlands, Australia
| | - Foo Yew Liew
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgow, UK
- CEGMR, King Abdulaziz UniversityJeddah, Saudi Arabia
- Institute of Biology and Medical Sciences, Soochow UniversitySuzhou, China
| | - Hui-Rong Jiang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of StrathclydeGlasgow, UK
| |
Collapse
|
207
|
Fasudil regulates T cell responses through polarization of BV-2 cells in mice experimental autoimmune encephalomyelitis. Acta Pharmacol Sin 2014; 35:1428-38. [PMID: 25263338 DOI: 10.1038/aps.2014.68] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/22/2014] [Indexed: 01/22/2023] Open
Abstract
AIM Fasudil, a selective Rho kinase (ROCK) inhibitor, has been shown to alleviate the severity of experimental autoimmune encephalomyelitis (EAE) via attenuating demyelination and neuroinflammation. The aim of this study was to investigate the effects of fasudil on interactions between macrophages/microglia and T cells in a mice EAE model. METHODS Mouse BV-2 microglia were treated with IFN-γ and fasudil. Cell viability was detected with MTT assay. BV-2 microglia polarization was analyzed using flow cytometry. Cytokines and other proteins were detected with ELISA and Western blotting, respectively. Mice were immunized with MOG35-55 to induce EAE, and then treated with fasudil (40 mg/kg, ip) every other day from d 3 to d 27 pi. Encephalomyelitic T cells were prepared from the spleen of mice immunized with MOG35-55 on d 9 pi. RESULTS Treatment of mouse BV-2 microglia with fasudil (15 μg/mL) induced significant phenotype polarization and functional plasticity, shifting M1 to M2 polarization. When co-cultured with the encephalomyelitic T cells, fasudil-treated BV-2 microglia significantly inhibited the proliferation of antigen-reactive T cells, and down-regulated IL-17-expressing CD4(+) T cells and IL-17 production. Furthermore, fasudil-treated BV-2 microglia significantly up-regulated CD4(+)CD25(high) and CD4(+)IL-10(+) regulatory T cells (Tregs) and IL-10 production, suggesting that the encephalomyelitic T cells had converted to Tregs. In EAE mice, fasudil administration significantly decreased both CD11b(+)iNOS(+) and CD11b(+)TNF-α(+) M1 microglia, and increased CD11b(+)IL-10(+) M2 microglia. CONCLUSION Fasudil polarizes BV-2 microglia into M2 cells, which convert the encephalomyelitic T cells into Tregs in the mice EAE model.
Collapse
|
208
|
Efficacy of the selective progesterone receptor agonist Nestorone for chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 276:89-97. [DOI: 10.1016/j.jneuroim.2014.08.619] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 11/17/2022]
|
209
|
Okuneva O, Körber I, Li Z, Tian L, Joensuu T, Kopra O, Lehesjoki AE. Abnormal microglial activation in the Cstb(-/-) mouse, a model for progressive myoclonus epilepsy, EPM1. Glia 2014; 63:400-11. [PMID: 25327891 DOI: 10.1002/glia.22760] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/26/2014] [Indexed: 12/12/2022]
Abstract
Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal-recessively inherited neurodegenerative disorder characterized by severely incapacitating myoclonus, seizures, and ataxia, and caused by loss-of-function mutations in the cystatin B gene (CSTB). A central neuropathological finding in the Cstb(-/-) mouse, an animal model for EPM1, is early microglial activation, which precedes astroglial activation, neuronal loss, and onset of myoclonus, thus implying a critical role for microglia in EPM1 pathogenesis. Here, we characterized phenotypic and functional properties of microglia from Cstb(-/-) mice utilizing brain tissue, microglia directly isolated from the brain, and primary microglial cultures. Our results show significantly higher Cstb mRNA expression in microglia than in neurons and astrocytes. In Cstb(-/-) mouse brain, expression of the inflammatory marker p-p38 MAPK and the proportion of both pro-inflammatory M1 and anti-inflammatory M2 microglia is higher than in control mice. Moreover, M1/M2 polarization of microglia in presymptomatic Cstb(-/-) mice is, compared to control mice, skewed towards M2 type at postnatal day 14 (P14), but towards M1 type at P30, a time point associated with onset of myoclonus. At this age, the high expression of both pro-inflammatory inducible nitric oxide synthase (iNOS) and anti-inflammatory arginase 1 (ARG1) in Cstb(-/-) mouse cortex is accompanied by the presence of peripheral immune cells. Consistently, activated Cstb(-/-) microglia show elevated chemokine release and chemotaxis. However, their MHCII surface expression is suppressed. Taken together, our results link CSTB deficiency to neuroinflammation with early activation and dysfunction of microglia and will open new avenues for therapeutic interventions for EPM1.
Collapse
Affiliation(s)
- Olesya Okuneva
- Folkhälsan Institute of Genetics, Haartmaninkatu 8, 00014, Helsinki, Finland; Haartman Institute, Department of Medical Genetics and Research Program's Unit, Molecular Neurology, University of Helsinki, Haartmaninkatu 8, 00014, Helsinki, Finland; Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014, Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
210
|
The contribution of immune and glial cell types in experimental autoimmune encephalomyelitis and multiple sclerosis. Mult Scler Int 2014; 2014:285245. [PMID: 25374694 PMCID: PMC4211315 DOI: 10.1155/2014/285245] [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: 08/07/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system characterised by widespread areas of focal demyelination. Its aetiology and pathogenesis remain unclear despite substantial insights gained through studies of animal models, most notably experimental autoimmune encephalomyelitis (EAE). MS is widely believed to be immune-mediated and pathologically attributable to myelin-specific autoreactive CD4+ T cells. In recent years, MS research has expanded beyond its focus on CD4+ T cells to recognise the contributions of multiple immune and glial cell types to the development, progression, and amelioration of the disease. This review summarises evidence of T and B lymphocyte, natural killer cell, macrophage/microglial, astrocytic, and oligodendroglial involvement in both EAE and MS and the intercommunication and influence of each cell subset in the inflammatory process. Despite important advances in the understanding of the involvement of these cell types in MS, many questions still remain regarding the various subsets within each cell population and their exact contribution to different stages of the disease.
Collapse
|
211
|
Donders R, Vanheusden M, Bogie JFJ, Ravanidis S, Thewissen K, Stinissen P, Gyselaers W, Hendriks JJA, Hellings N. Human Wharton's Jelly-Derived Stem Cells Display Immunomodulatory Properties and Transiently Improve Rat Experimental Autoimmune Encephalomyelitis. Cell Transplant 2014; 24:2077-98. [PMID: 25310756 DOI: 10.3727/096368914x685104] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Umbilical cord matrix or Wharton's jelly-derived stromal cells (WJ-MSCs) are an easily accessible source of mesenchymal-like stem cells. Recent studies describe a hypoimmunogenic phenotype, multipotent differentiation potential, and trophic support function for WJ-MSCs, with variable clinical benefit in degenerative disease models such as stroke, myocardial infarction, and Parkinson's disease. It remains unclear whether WJ-MSCs have therapeutic value for multiple sclerosis (MS), where autoimmune-mediated demyelination and neurodegeneration need to be halted. In this study, we investigated whether WJ-MSCs possess the required properties to effectively and durably reverse these pathological hallmarks and whether they survive in an inflammatory environment after transplantation. WJ-MSCs displayed a lowly immunogenic phenotype and showed intrinsic expression of neurotrophic factors and a variety of anti-inflammatory molecules. Furthermore, they dose-dependently suppressed proliferation of activated T cells using contact-dependent and paracrine mechanisms. Indoleamine 2,3-dioxygenase 1 was identified as one of the main effector molecules responsible for the observed T-cell suppression. The immune-modulatory phenotype of WJ-MSCs was further enhanced after proinflammatory cytokine treatment in vitro (licensing). In addition to their effect on adaptive immunity, WJ-MSCs interfered with dendritic cell differentiation and maturation, thus directly affecting antigen presentation and therefore T-cell priming. Systemically infused WJ-MSCs potently but transiently ameliorated experimental autoimmune encephalomyelitis (EAE), an animal model for MS, when injected at onset or during chronic disease. This protective effect was paralleled with a reduction in autoantigen-induced T-cell proliferation, confirming their immunomodulatory activity in vivo. Surprisingly, in vitro licensed WJ-MSCs did not ameliorate EAE, indicative of a fast rejection as a result of enhanced immunogenicity. Collectively, we show that WJ-MSCs have trophic support properties and effectively modulate immune cell functioning both in vitro and in the EAE model, suggesting WJ-MSC may hold promise for MS therapy. Future research is needed to optimize survival of stem cells and enhance clinical durability.
Collapse
Affiliation(s)
- Raf Donders
- Hasselt University, Biomedical Research Institute/Transnational University Limburg, School of Life Sciences, Diepenbeek, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Kim J, Yang M, Son Y, Jang H, Kim D, Kim JC, Kim SH, Kang MJ, Im HI, Shin T, Moon C. Glial activation with concurrent up-regulation of inflammatory mediators in trimethyltin-induced neurotoxicity in mice. Acta Histochem 2014; 116:1490-500. [PMID: 25265880 DOI: 10.1016/j.acthis.2014.09.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Trimethyltin (TMT), a potent neurotoxic chemical, causes dysfunction and neuroinflammation in the brain, particularly in the hippocampus. The present study assessed TMT-induced glial cell activation and inflammatory cytokine alterations in the mouse hippocampus, BV-2 microglia, and primary cultured astrocytes. In the mouse hippocampus, TMT treatment significantly increased the expression of glial cell markers, including the microglial marker ionized calcium-binding adapter molecule 1 and the astroglial marker glial fibrillary acidic protein. The expression of M1 and M2 microglial markers (inducible nitric oxide synthase [iNOS] and CD206, respectively) and pro-inflammatory cytokines (interleukin [IL]-1β, IL-6 and tumor necrosis factor [TNF]-α) were significantly increased in the mouse hippocampus following TMT treatment. In BV-2 microglia, iNOS, IL-1β, TNF-α, and IL-6 expression increased significantly, whereas arginase-1 and CD206 expression decreased significantly after TMT treatment in a time- and concentration-dependent manner. In primary cultured astrocytes, iNOS, arginase-1, IL-1β, TNF-α, and IL-6 expression increased significantly, whereas IL-10 expression decreased significantly after TMT treatment in a time- and concentration-dependent manner. These results indicate that significant up-regulation of pro-inflammatory signals in TMT-induced neurotoxicity may be associated with pathological processing of TMT-induced neurodegeneration.
Collapse
|
213
|
Gao Z, Nissen JC, Ji K, Tsirka SE. The experimental autoimmune encephalomyelitis disease course is modulated by nicotine and other cigarette smoke components. PLoS One 2014; 9:e107979. [PMID: 25250777 PMCID: PMC4176721 DOI: 10.1371/journal.pone.0107979] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/16/2014] [Indexed: 12/12/2022] Open
Abstract
Epidemiological studies have reported that cigarette smoking increases the risk of developing multiple sclerosis (MS) and accelerates its progression. However, the molecular mechanisms underlying these effects remain unsettled. We have investigated here the effects of the nicotine and the non-nicotine components in cigarette smoke on MS using the experimental autoimmune encephalomyelitis (EAE) model, and have explored their underlying mechanism of action. Our results show that nicotine ameliorates the severity of EAE, as shown by reduced demyelination, increased body weight, and attenuated microglial activation. Nicotine administration after the development of EAE symptoms prevented further disease exacerbation, suggesting that it might be useful as an EAE/MS therapeutic. In contrast, the remaining components of cigarette smoke, delivered as cigarette smoke condensate (CSC), accelerated and increased adverse clinical symptoms during the early stages of EAE, and we identify a particular cigarette smoke compound, acrolein, as one of the potential mediators. We also show that the mechanisms underlying the opposing effects of nicotine and CSC on EAE are likely due to distinct effects on microglial viability, activation, and function.
Collapse
Affiliation(s)
- Zhen Gao
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- Program in Neuroscience, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Jillian C. Nissen
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Kyungmin Ji
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Stella E. Tsirka
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- Program in Neuroscience, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
214
|
Lewis ND, Hill JD, Juchem KW, Stefanopoulos DE, Modis LK. RNA sequencing of microglia and monocyte-derived macrophages from mice with experimental autoimmune encephalomyelitis illustrates a changing phenotype with disease course. J Neuroimmunol 2014; 277:26-38. [PMID: 25270668 DOI: 10.1016/j.jneuroim.2014.09.014] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/07/2014] [Accepted: 09/15/2014] [Indexed: 12/11/2022]
Abstract
The role of microglia and monocyte-derived macrophages in experimental autoimmune encephalomyelitis pathogenesis has been controversial. To gain insight into their respective roles, we developed a method for differentiating between microglia and monocyte-derived macrophages in the CNS by flow cytometry utilizing anti-CD44 antibodies. We used this system to monitor changes in cell number, activation status, and gene expression by RNA sequencing over the course of disease. This in vivo characterization and RNA-Seq dataset improves our understanding of macrophage biology in the brain under inflammatory conditions and may lead to strategies to identify therapies for neuroinflammatory diseases.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Base Sequence/genetics
- Base Sequence/physiology
- Cell Proliferation
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Flow Cytometry
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred C57BL
- Microglia/metabolism
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Peptide Fragments/toxicity
- Signal Transduction/immunology
- Time Factors
Collapse
Affiliation(s)
- Nuruddeen D Lewis
- Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877-0368, USA
| | - Jonathan D Hill
- Department of Research Networking, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, 06877-0368, USA
| | - Kathryn W Juchem
- Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877-0368, USA
| | - Dimitria E Stefanopoulos
- Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877-0368, USA
| | - Louise K Modis
- Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877-0368, USA.
| |
Collapse
|
215
|
Obesity in autoimmune diseases: Not a passive bystander. Autoimmun Rev 2014; 13:981-1000. [DOI: 10.1016/j.autrev.2014.07.001] [Citation(s) in RCA: 440] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/20/2014] [Indexed: 02/06/2023]
|
216
|
Dungan LS, McGuinness NC, Boon L, Lynch MA, Mills KHG. Innate IFN-γ promotes development of experimental autoimmune encephalomyelitis: A role for NK cells and M1 macrophages. Eur J Immunol 2014; 44:2903-17. [DOI: 10.1002/eji.201444612] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/09/2014] [Accepted: 07/18/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Lara S. Dungan
- Immune Regulation Research Group; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin Ireland
| | - Niamh C. McGuinness
- Immune Regulation Research Group; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin Ireland
- Trinity College Institute of Neuroscience; Trinity College Dublin; Dublin Ireland
| | - Louis Boon
- Bioceros Holding BV; Utrecht The Netherlands
| | - Marina A. Lynch
- Trinity College Institute of Neuroscience; Trinity College Dublin; Dublin Ireland
| | - Kingston H. G. Mills
- Immune Regulation Research Group; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin Ireland
| |
Collapse
|
217
|
Sexton M, Cudaback E, Abdullah RA, Finnell J, Mischley LK, Rozga M, Lichtman AH, Stella N. Cannabis use by individuals with multiple sclerosis: effects on specific immune parameters. Inflammopharmacology 2014; 22:295-303. [PMID: 25135301 DOI: 10.1007/s10787-014-0214-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/04/2014] [Indexed: 11/30/2022]
Abstract
Cannabinoids affect immune responses in ways that may be beneficial for autoimmune diseases. We sought to determine whether chronic Cannabis use differentially modulates a select number of immune parameters in healthy controls and individuals with multiple sclerosis (MS cases). Subjects were enrolled and consented to a single blood draw, matched for age and BMI. We measured monocyte migration isolated from each subject, as well as plasma levels of endocannabinoids and cytokines. Cases met definition of MS by international diagnostic criteria. Monocyte cell migration measured in control subjects and individuals with MS was similarly inhibited by a set ratio of phytocannabinoids. The plasma levels of CCL2 and IL17 were reduced in non-naïve cannabis users irrespective of the cohorts. We detected a significant increase in the endocannabinoid arachidonoylethanolamine (AEA) in serum from individuals with MS compared to control subjects, and no significant difference in levels of other endocannabinoids and signaling lipids irrespective of Cannabis use. Chronic Cannabis use may affect the immune response to similar extent in individuals with MS and control subjects through the ability of phytocannabinoids to reduce both monocyte migration and cytokine levels in serum. From a panel of signaling lipids, only the levels of AEA are increased in individuals with MS, irrespective of Cannabis use or not. Our results suggest that both MS cases and controls respond similarly to chronic Cannabis use with respect to the immune parameters measured in this study.
Collapse
Affiliation(s)
- Michelle Sexton
- Center for the Study of Cannabis and Social Policy, Seattle, WA, 98028, USA,
| | | | | | | | | | | | | | | |
Collapse
|
218
|
Brunn A, Mihelcic M, Carstov M, Hummel L, Geier F, Schmidt A, Saupe L, Utermöhlen O, Deckert M. IL-10, IL-4, and STAT6 promote an M2 milieu required for termination of P0(106-125)-induced murine experimental autoimmune neuritis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2627-40. [PMID: 25108223 DOI: 10.1016/j.ajpath.2014.06.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/06/2014] [Accepted: 06/12/2014] [Indexed: 01/24/2023]
Abstract
The role of the type 2 helper T cell (Th2)-polarizing cytokines IL-4 and IL-10 has not yet been studied in P0106-125-induced murine experimental autoimmune neuritis (EAN). We, therefore, addressed the functional relevance of these cytokines and signaling via the IL-4-associated transcription factor STAT6. The clinical course of P0106-125-induced EAN in mice deficient for IL-10(0/0), IL-4(0/0), or STAT6(0/0) was significantly aggravated compared with that of wild-type control mice. In addition, treatment of P0106-125-immunized C57BL/6 mice at the onset of clinical symptoms with a monoclonal IL-10 neutralizing antibody aggravated symptoms and prolonged disease to a similar degree as in IL-10(0/0) mice. This exacerbated course was attributed to a more prominent Th1 immune response associated with a persistent M1 milieu in the sciatic nerve and in the regional and systemic lymphatic system. These data suggest a Th2-polarized milieu being required to prevent axonal damage of the sciatic nerve and to terminate the P0106-125-specific immune response in EAN. Beyond the already known role of macrophages as pathogenic effector cells in EAN, these data suggest that M2-differentiated macrophages do not damage and may even protect neural tissues in EAN. Thus, these data highlight the pathogenetic relevance of the macrophage polarization status in EAN. Therapeutic modulation of immune responses from an M1 toward an M2 milieu may be a promising novel strategy in peripheral nervous system neuritis.
Collapse
Affiliation(s)
- Anna Brunn
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany.
| | - Mirna Mihelcic
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| | - Mariana Carstov
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| | - Lea Hummel
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| | - Frank Geier
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| | - Annika Schmidt
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| | - Lisa Saupe
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| | - Olaf Utermöhlen
- Institute for Medical Microbiology, Immunology, and Hygiene, Medical Center, Cologne, Germany; Center for Molecular Medicine Colgne, University of Cologne, Cologne, Germany
| | - Martina Deckert
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
219
|
Strachan-Whaley M, Rivest S, Yong VW. Interactions Between Microglia and T Cells in Multiple Sclerosis Pathobiology. J Interferon Cytokine Res 2014; 34:615-22. [DOI: 10.1089/jir.2014.0019] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Megan Strachan-Whaley
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Serge Rivest
- Laboratory of Molecular Endocrinology and Department of Anatomy and Physiology, Laval University, Quebec, Canada
| | - V. Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| |
Collapse
|
220
|
Di Lorenzo D, Biasiotto G, Zanella I. Source of iron overload in multiple sclerosis. Cell Mol Life Sci 2014; 71:3187-9. [PMID: 24830704 PMCID: PMC11113671 DOI: 10.1007/s00018-014-1641-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/29/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Diego Di Lorenzo
- Biotechnology/3rd Laboratory, Department of Diagnostics, Civic Hospital of Brescia, 25123 Brescia, Italy
| | - Giorgio Biasiotto
- Biotechnology/3rd Laboratory, Department of Diagnostics, Civic Hospital of Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Biotechnology/3rd Laboratory, Department of Diagnostics, Civic Hospital of Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
221
|
Miron VE. Dissecting the damaging versus regenerative roles of CNS macrophages: implications for the use of immunomodulatory therapeutics. Regen Med 2014; 8:673-6. [PMID: 24147520 DOI: 10.2217/rme.13.73] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Veronique E Miron
- MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
222
|
New aspects of the pathogenesis of canine distemper leukoencephalitis. Viruses 2014; 6:2571-601. [PMID: 24992230 PMCID: PMC4113784 DOI: 10.3390/v6072571] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/11/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022] Open
Abstract
Canine distemper virus (CDV) is a member of the genus morbillivirus, which is known to cause a variety of disorders in dogs including demyelinating leukoencephalitis (CDV-DL). In recent years, substantial progress in understanding the pathogenetic mechanisms of CDV-DL has been made. In vivo and in vitro investigations provided new insights into its pathogenesis with special emphasis on axon-myelin-glia interaction, potential endogenous mechanisms of regeneration, and astroglial plasticity. CDV-DL is characterized by lesions with a variable degree of demyelination and mononuclear inflammation accompanied by a dysregulated orchestration of cytokines as well as matrix metalloproteinases and their inhibitors. Despite decades of research, several new aspects of the neuropathogenesis of CDV-DL have been described only recently. Early axonal damage seems to represent an initial and progressive lesion in CDV-DL, which interestingly precedes demyelination. Axonopathy may, thus, function as a potential trigger for subsequent disturbed axon-myelin-glia interactions. In particular, the detection of early axonal damage suggests that demyelination is at least in part a secondary event in CDV-DL, thus challenging the dogma of CDV as a purely primary demyelinating disease. Another unexpected finding refers to the appearance of p75 neurotrophin (NTR)-positive bipolar cells during CDV-DL. As p75NTR is a prototype marker for immature Schwann cells, this finding suggests that Schwann cell remyelination might represent a so far underestimated endogenous mechanism of regeneration, though this hypothesis still remains to be proven. Although it is well known that astrocytes represent the major target of CDV infection in CDV-DL, the detection of infected vimentin-positive astrocytes in chronic lesions indicates a crucial role of this cell population in nervous distemper. While glial fibrillary acidic protein represents the characteristic intermediate filament of mature astrocytes, expression of vimentin is generally restricted to immature or reactive astrocytes. Thus, vimentin-positive astrocytes might constitute an important cell population for CDV persistence and spread, as well as lesion progression. In vitro models, such as dissociated glial cell cultures, as well as organotypic brain slice cultures have contributed to a better insight into mechanisms of infection and certain morphological and molecular aspects of CDV-DL. Summarized, recent in vivo and in vitro studies revealed remarkable new aspects of nervous distemper. These new perceptions substantially improved our understanding of the pathogenesis of CDV-DL and might represent new starting points to develop novel treatment strategies.
Collapse
|
223
|
Sawyer AJ, Tian W, Saucier-Sawyer JK, Rizk PJ, Saltzman WM, Bellamkonda RV, Kyriakides TR. The effect of inflammatory cell-derived MCP-1 loss on neuronal survival during chronic neuroinflammation. Biomaterials 2014; 35:6698-706. [PMID: 24881026 DOI: 10.1016/j.biomaterials.2014.05.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 05/01/2014] [Indexed: 01/22/2023]
Abstract
Intracranial implants elicit neurodegeneration via the foreign body response (FBR) that includes BBB leakage, macrophage/microglia accumulation, and reactive astrogliosis, in addition to neuronal degradation that limit their useful lifespan. Previously, monocyte chemoattractant protein 1 (MCP-1, also CCL2), which plays an important role in monocyte recruitment and propagation of inflammation, was shown to be critical for various aspects of the FBR in a tissue-specific manner. However, participation of MCP-1 in the brain FBR has not been evaluated. Here we examined the FBR to intracortical silicon implants in MCP-1 KO mice at 1, 2, and 8 weeks after implantation. MCP-1 KO mice had a diminished FBR compared to WT mice, characterized by reductions in BBB leakage, macrophage/microglia accumulation, and astrogliosis, and an increased neuronal density. Moreover, pharmacological inhibition of MCP-1 in implant-bearing WT mice maintained the increased neuronal density. To elucidate the relative contribution of microglia and macrophages, bone marrow chimeras were generated between MCP-1 KO and WT mice. Increased neuronal density was observed only in MCP-1 knockout mice transplanted with MCP-1 knockout marrow, which indicates that resident cells in the brain are major contributors. We hypothesized that these improvements are the result of a phenotypic switch of the macrophages/microglia polarization state, which we confirmed using PCR for common activation markers. Our observations suggest that MCP-1 influences neuronal loss, which is integral to the progression of neurological disorders like Alzheimer's and Parkinson disease, via BBB leakage and macrophage polarization.
Collapse
Affiliation(s)
- Andrew J Sawyer
- Department of Pathology, Yale School of Medicine, 310 Cedar Street LH 108, New Haven, CT 06520-8023, USA
| | - Weiming Tian
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, PR China
| | | | - Paul J Rizk
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Themis R Kyriakides
- Department of Pathology, Yale School of Medicine, 310 Cedar Street LH 108, New Haven, CT 06520-8023, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
224
|
Onore CE, Schwartzer JJ, Careaga M, Berman RF, Ashwood P. Maternal immune activation leads to activated inflammatory macrophages in offspring. Brain Behav Immun 2014; 38:220-6. [PMID: 24566386 PMCID: PMC4321784 DOI: 10.1016/j.bbi.2014.02.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/24/2014] [Accepted: 02/10/2014] [Indexed: 02/07/2023] Open
Abstract
Several epidemiological studies have shown an association between infection or inflammation during pregnancy and increased risk of autism in the child. In addition, animal models have illustrated that maternal inflammation during gestation can cause autism-relevant behaviors in the offspring; so called maternal immune activation (MIA) models. More recently, permanent changes in T cell cytokine responses were reported in children with autism and in offspring of MIA mice; however, the cytokine responses of other immune cell populations have not been thoroughly investigated in these MIA models. Similar to changes in T cell function, we hypothesized that following MIA, offspring will have long-term changes in macrophage function. To test this theory, we utilized the poly (I:C) MIA mouse model in C57BL/6J mice and examined macrophage cytokine production in adult offspring. Pregnant dams were given either a single injection of 20mg/kg polyinosinic-polycytidylic acid, poly (I:C), or saline delivered intraperitoneally on gestational day 12.5. When offspring of poly (I:C) treated dams reached 10weeks of age, femurs were collected and bone marrow-derived macrophages were generated. Cytokine production was measured in bone marrow-derived macrophages incubated for 24h in either growth media alone, LPS, IL-4/LPS, or IFN-γ/LPS. Following stimulation with LPS alone, or the combination of IFN-γ/LPS, macrophages from offspring of poly (I:C) treated dams produced higher levels of IL-12(p40) (p<0.04) suggesting an increased M1 polarization. In addition, even without the presence of a polarizing cytokine or LPS stimulus, macrophages from offspring of poly (I:C) treated dams exhibited a higher production of CCL3 (p=0.05). Moreover, CCL3 levels were further increased when stimulated with LPS, or polarized with either IL-4/LPS or IFN-γ/LPS (p<0.05) suggesting a general increase in production of this chemokine. Collectively, these data suggest that MIA can produce lasting changes in macrophage function that are sustained into adulthood.
Collapse
Affiliation(s)
- Charity E Onore
- Department of Medical Microbiology and Immunology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States; The M.I.N.D. Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Jared J Schwartzer
- The M.I.N.D. Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States; Department of Psychiatry and Behavioral Sciences, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States; Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, United States
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States; The M.I.N.D. Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Robert F Berman
- The M.I.N.D. Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States; Department of Psychiatry and Behavioral Sciences, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States; Department of Neurological Surgery, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States; The M.I.N.D. Institute, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States.
| |
Collapse
|
225
|
Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat Rev Neurosci 2014; 15:300-12. [PMID: 24713688 DOI: 10.1038/nrn3722] [Citation(s) in RCA: 970] [Impact Index Per Article: 88.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mononuclear phagocytic cells in the CNS used to be defined according to their anatomical location and surface marker expression. Recently, this concept has been challenged by the results of developmental and gene expression profiling studies that have used novel molecular biological tools to unravel the origin of microglia and to define their role as specialized tissue macrophages with long lifespans. Here, we describe how these results have redefined microglia and helped us to understand how different myeloid cell populations operate in the CNS based on their cell-specific gene expression signatures, distinct ontogeny and differential functions. Moreover, we describe the vulnerability of microglia to dysfunction and propose that myelomonocytic cells might be used in the treatment of neurological and psychiatric disorders that are characterized by primary or secondary 'microgliopathy'.
Collapse
|
226
|
Macrophages: a double-edged sword in experimental autoimmune encephalomyelitis. Immunol Lett 2014; 160:17-22. [PMID: 24698730 DOI: 10.1016/j.imlet.2014.03.006] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/28/2014] [Accepted: 03/17/2014] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is a debilitating neurological disorder of the central nervous system (CNS), characterized by activation and infiltration of leukocytes and dendritic cells into the CNS. In the initial phase of MS and its animal model, experimental autoimmune encephalomyelitis (EAE), peripheral macrophages infiltrate into the CNS, where, together with residential microglia, they participate in the induction and development of disease. During the early phase, microglia/macrophages are immediately activated to become classically activated macrophages (M1 cells), release pro-inflammatory cytokines and damage CNS tissue. During the later phase, microglia/macrophages in the inflamed CNS are less activated, present as alternatively activated macrophage phenotype (M2 cells), releasing anti-inflammatory cytokines, accompanied by inflammation resolution and tissue repair. The balance between activation and polarization of M1 cells and M2 cells in the CNS is important for disease progression. Pro-inflammatory IFN-γ and IL-12 drive M1 cell polarization, while IL-4 and IL-13 drive M2 cell polarization. Given that polarized macrophages are reversible in a well-defined cytokine environment, macrophage phenotypes in the CNS can be modulated by molecular intervention. This review summarizes the detrimental and beneficial roles of microglia and macrophages in the CNS, with an emphasis on the role of M2 cells in EAE and MS patients.
Collapse
|
227
|
Miron VE, Franklin RJM. Macrophages and CNS remyelination. J Neurochem 2014; 130:165-71. [PMID: 24601941 DOI: 10.1111/jnc.12705] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 02/24/2014] [Indexed: 12/14/2022]
Abstract
Microglia are the resident macrophages of the central nervous system that survey the microenvironment for signals of injury or infection. The response to such signals induces an inflammatory response involving macrophages derived from both resident microglia and recruited circulating monocytes. Although implicated as contributors to autoimmune-mediated injury, microglia/ macrophages have recently been shown to be critical for the important central nervous system regenerative process of remyelination. This functional dichotomy may reflect their ability to be polarized along a continuum of activation states including the well-characterized cytotoxic M1 and regenerative M2 phenotypes. Here, we review the roles of microglia, monocytes and the macrophages which they give rise to in creating lesion environments favourable to remyelination, highlighting the specific roles of M1 and M2 phenotypes and how the pro-regenerative role of the innate immune system is altered by ageing. Here, we review the roles of microglia, monocytes and the macrophages, which they give rise to in creating lesion environments favourable to remyelination, highlighting the specific roles of activation phenotypes and how the pro-regenerative role of the innate immune system is altered by ageing.
Collapse
Affiliation(s)
- Veronique E Miron
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
228
|
Jensen SK, Yong VW. Microglial modulation as a mechanism behind the promotion of central nervous system well-being by physical exercise. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/cen3.12093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Samuel K. Jensen
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences; University of Calgary; Calgary AB Canada
| | - V. Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences; University of Calgary; Calgary AB Canada
| |
Collapse
|
229
|
Hedström AK, Lima Bomfim I, Barcellos L, Gianfrancesco M, Schaefer C, Kockum I, Olsson T, Alfredsson L. Interaction between adolescent obesity and HLA risk genes in the etiology of multiple sclerosis. Neurology 2014; 82:865-72. [PMID: 24500647 PMCID: PMC3959752 DOI: 10.1212/wnl.0000000000000203] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 11/01/2013] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE We investigated potential interactions between human leukocyte antigen (HLA) genotype and body mass index (BMI) status in relation to the risk of developing multiple sclerosis (MS). METHODS We used 2 case-control studies, one with incident cases (1,510 cases, 2,017 controls) and one with prevalent cases (937 cases, 609 controls). Subjects with different genotypes and BMI were compared with regard to incidence of MS by calculating odds ratios (ORs) with 95% confidence intervals (CIs) employing logistic regression. Potential interactions between genotypes and BMI were evaluated by calculating the attributable proportion due to interaction. RESULTS In both cohorts, a significant interaction was observed between HLA-DRB1*15 and obesity, regardless of HLA-A*02 status. Similarly, there was a significant interaction between absence of A*02 and obesity, regardless of DRB1*15 status. In the incident cohort, obese subjects with the most susceptible genotype (carriage of DRB1*15 and absence of A*02) had an OR of 16.2 (95% CI 7.5-35.2) compared to nonobese subjects without the genetic risk factors. The corresponding OR in the prevalent study was 13.8 (95% CI 4.1-46.8). CONCLUSIONS We observed striking interactions between BMI status and HLA genotype with regard to MS risk. Hypothetically, a low-grade inflammatory response inherent to obesity synergizes with the adaptive, HLA molecule-restricted arm of the immune system, causing MS. Prevention of adolescent obesity may thus lower the risk of developing MS, predominantly among people with a genetic susceptibility to the disease.
Collapse
Affiliation(s)
- Anna Karin Hedström
- From the Institute of Environmental Medicine (A.K.H.), Neuroimmunology Unit, Department of Clinical Neuroscience (I.L.B., I.K.), and the Institute of Environmental Medicine (L.A.), Karolinska Institutet, Stockholm, Sweden; Genetic Epidemiology and Genomics Lab (L.B., M.G.), Division of Epidemiology, School of Public Health, University of California, Berkeley; Kaiser Permanente Division of Research (C.S.), Oakland, CA; Center for Molecular Medicine (I.K.) and Neuroimmunology Unit, Department of Clinical Neuroscience and Center for Molecular Medicine (T.O.), Karolinska Institutet at Karolinska University Hospital, Solna; and Centre for Occupational and Environmental Medicine (L.A.), Stockholm County Council, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Iannitti T, Kerr BJ, Taylor BK. Mechanisms and pharmacology of neuropathic pain in multiple sclerosis. Curr Top Behav Neurosci 2014; 20:75-97. [PMID: 24590824 PMCID: PMC4464806 DOI: 10.1007/7854_2014_288] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The neuropathic pain of multiple sclerosis is quite prevalent and severely impacts quality of life. A few randomized, placebo-controlled, blinded clinical trials suggest that cannabis- and anticonvulsant-based treatments provide partial pain relief, but at the expense of adverse events. An even smaller, but emerging, number of translational studies are using rodent models of experimental autoimmune encephalomyelitis (EAE), which exhibit pain-like behaviors resembling those of Multiple sclerosis (MS) patients. These studies not only support the possible effectiveness of anticonvulsants, but also compel further clinical trials with serotonin-norepinephrine reuptake inhibitors, the immunosuppressant drug rapamycin, or drugs which interfere with glutamatergic neurotransmission. Future behavioral studies in EAE models are essential toward a new pharmacotherapy of multiple sclerosis pain.
Collapse
Affiliation(s)
- T Iannitti
- Department of Physiology, School of Medicine, University of Kentucky Medical Center, Lexington, KY, 40536-0298, USA
| | | | | |
Collapse
|
231
|
Biber K, Owens T, Boddeke E. What is microglia neurotoxicity (Not)? Glia 2014; 62:841-54. [PMID: 24590682 DOI: 10.1002/glia.22654] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/10/2014] [Accepted: 02/14/2014] [Indexed: 01/19/2023]
Abstract
Microglia most likely appeared early in evolution as they are not only present in vertebrates, but are also found in nervous systems of various nonvertebrate organisms. Mammalian microglia are derived from a specific embryonic, self-renewable myeloid cell population that is throughout lifetime not replaced by peripheral myeloid cells. These phylogenic and ontogenic features suggest that microglia serve vital functions. Yet, microglia often are described as neurotoxic cells, that actively kill (healthy) neurons. Since it is from an evolutionary point of view difficult to understand why an important and vulnerable organ like the brain should host numerous potential killers, we here review the concept of microglia neurotoxicity. On one hand it is discussed that most of our understanding about how microglia kill neurons is based on in vitro experiments or correlative staining studies that suffer from the difficulty to discriminate microglia and peripheral myeloid cells in the diseased brain. On the other hand it is described that a more functional approach by mutating, inactivating or deleting microglia is seldom associated with a beneficial outcome in an acute injury situation, suggesting that microglia are normally important protective elements in the brain. This might change in chronic disease or the aged brain, where; however, it remains to be established whether microglia simply lose their protective capacities or whether microglia become truly neurotoxic cells.
Collapse
Affiliation(s)
- Knut Biber
- Department of Psychiatry and Psychotherapy, University Hospital Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department of Neuroscience, University Medical Center Groningen, Ant. Deusinglaan 1, 9713, AV Groningen, The Netherlands
| | | | | |
Collapse
|
232
|
Hamilton AM, Mallett C, Foster PJ. High-resolution MRI and nanoparticles: the future of brain imaging. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT: Cellular MRI uses superparamagnetic iron oxide nanoparticles to label cells (in vitro or in vivo) for detection in magnetic resonance images. The infiltration of inflammatory macrophages can be visualized in brain diseases, such as multiple sclerosis, stroke and Alzheimer‘s disease, and correlates with disease severity and responses to treatments. Mesenchymal stromal cells, neural stem cells and immune cells used as cell therapies in CNS diseases can be tracked in vivo over time to determine their migration and dispersion. Tracking labeled cancer cells provides information about metastasis and proliferative status in preclinical tumor models. Ongoing technical improvements come from the development of new particles, the use of fluorine-based contrast agents and the refinement of high-field MRI for cell tracking.
Collapse
Affiliation(s)
- Amanda M Hamilton
- Imaging Research Laboratories, Robarts Research Institute, London, ON, N6A 5K8, Canada
| | - Christiane Mallett
- Imaging Research Laboratories, Robarts Research Institute, London, ON, N6A 5K8, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Western University, London, ON, Canada
| |
Collapse
|
233
|
Vogel DYS, Heijnen PDAM, Breur M, de Vries HE, Tool ATJ, Amor S, Dijkstra CD. Macrophages migrate in an activation-dependent manner to chemokines involved in neuroinflammation. J Neuroinflammation 2014; 11:23. [PMID: 24485070 PMCID: PMC3937114 DOI: 10.1186/1742-2094-11-23] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/23/2014] [Indexed: 11/10/2022] Open
Abstract
Background In neuroinflammatory diseases, macrophages can play a dual role in the process of tissue damage, depending on their activation status (M1 / M2). M1 macrophages are considered to exert damaging effects to neurons, whereas M2 macrophages are reported to aid regeneration and repair of neurons. Their migration within the central nervous system may be of critical importance in the final outcome of neurodegeneration in neuroinflammatory diseases e.g. multiple sclerosis (MS). To provide insight into this process, we examined the migratory capacity of human monocyte-derived M1 and M2 polarised macrophages towards chemoattractants, relevant for neuroinflammatory diseases like MS. Methods Primary cultures of human monocyte-derived macrophages were exposed to interferon gamma and lipopolysaccharide (LPS) to evoke proinflammatory (M1) activation or IL-4 to evoke anti-inflammatory (M2) activation. In a TAXIScan assay, migration of M0, M1 and M2 towards chemoattractants was measured and quantified. Furthermore the adhesion capacity and the expression levels of integrins as well as chemokine receptors of M0, M1 and M2 were assessed. Alterations in cell morphology were analysed using fluorescent labelling of the cytoskeleton. Results Significant differences were observed between M1 and M2 macrophages in the migration towards chemoattractants. We show that M2 macrophages migrated over longer distances towards CCL2, CCL5, CXCL10, CXCL12 and C1q compared to non-activated (M0) and M1 macrophages. No differences were observed in the adhesion of M0, M1 and M2 macrophages to multiple matrix components, nor in the expression of integrins and chemokine receptors. Significant changes were observed in the cytoskeleton organization upon stimulation with CCL2, M0, M1 and M2 macrophages adopt a spherical morphology and the cytoskeleton is rapidly rearranged. M0 and M2 macrophages are able to form filopodia, whereas M1 macrophages only adapt a spherical morphology. Conclusions Together our results indicate that the alternative activation status of macrophages promotes their migratory properties to chemoattractants relevant for neuroinflammatory diseases like MS. Conversely, classically activated, proinflammatory macrophages have reduced migratory properties. Based on our results, we postulate that the activation status of the macrophage influences the capacity of the macrophages to rearrange their cytoskeleton. This is the first step in understanding how modulation of macrophage activation affects macrophage migration in neuroinflammatory diseases like MS.
Collapse
Affiliation(s)
- Daphne Y S Vogel
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam VU University Medical Centre, MF J283, P,O, Box 7057, 1007, MB, Amsterdam, Netherlands.
| | | | | | | | | | | | | |
Collapse
|
234
|
Dagley LF, Croft NP, Isserlin R, Olsen JB, Fong V, Emili A, Purcell AW. Discovery of novel disease-specific and membrane-associated candidate markers in a mouse model of multiple sclerosis. Mol Cell Proteomics 2013; 13:679-700. [PMID: 24361864 DOI: 10.1074/mcp.m113.033340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Multiple sclerosis is a chronic demyelinating disorder characterized by the infiltration of auto-reactive immune cells from the periphery into the central nervous system resulting in axonal injury and neuronal cell death. Experimental autoimmune encephalomyelitis represents the best characterized animal model as common clinical, histological, and immunological features are recapitulated. A label-free mass spectrometric proteomics approach was used to detect differences in protein abundance within specific fractions of disease-affected tissues including the soluble lysate derived from the spinal cord and membrane protein-enriched peripheral blood mononuclear cells. Tissues were harvested from actively induced experimental autoimmune encephalomyelitis mice and sham-induced ("vehicle" control) counterparts at the disease peak followed by subsequent analysis by nanoflow liquid chromatography tandem mass spectrometry. Relative protein quantitation was performed using both intensity- and fragmentation-based approaches. After statistical evaluation of the data, over 500 and 250 differentially abundant proteins were identified in the spinal cord and peripheral blood mononuclear cell data sets, respectively. More than half of these observations have not previously been linked to the disease. The biological significance of all candidate disease markers has been elucidated through rigorous literature searches, pathway analysis, and validation studies. Results from comprehensive targeted mass spectrometry analyses have confirmed the differential abundance of ∼ 200 candidate markers (≥ twofold dysregulated expression) at a 70% success rate. This study is, to our knowledge, the first to examine the cell-surface proteome of peripheral blood mononuclear cells in experimental autoimmune encephalomyelitis. These data provide a unique mechanistic insight into the dynamics of peripheral immune cell infiltration into CNS-privileged sites at a molecular level and has identified several candidate markers, which represent promising targets for future multiple sclerosis therapies. The mass spectrometry proteomics data associated with this manuscript have been deposited to the ProteomeXchange Consortium with the data set identifier PXD000255.
Collapse
Affiliation(s)
- Laura F Dagley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | | | | | | | | | | |
Collapse
|
235
|
Côté CH, Bouchard P, van Rooijen N, Marsolais D, Duchesne E. Monocyte depletion increases local proliferation of macrophage subsets after skeletal muscle injury. BMC Musculoskelet Disord 2013; 14:359. [PMID: 24354415 PMCID: PMC3878260 DOI: 10.1186/1471-2474-14-359] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 12/08/2013] [Indexed: 11/10/2022] Open
Abstract
Background Sequential accumulation of M1 and M2 macrophages is critical for skeletal muscle recovery after an acute injury. While M1 accumulation is believed to rely on monocyte infiltration, the mechanisms of M2 accumulation remain controversial, but could involve an infiltrating precursor. Yet, strong depletion of monocytes only partially impairs skeletal muscle healing, supporting the existence of alternative mechanisms to palliate the loss of infiltrating macrophage progenitors. The aims of this study are thus to investigate if proliferation occurs in macrophage subsets within injured skeletal muscles; and to determine if monocyte depletion leads to increased proliferation of macrophages after injury. Methods Injury was induced by bupivacaine injection in the tibialis anterior muscle of rats. Blood monocytes were depleted by daily intravenous injections of liposome-encapsulated clodronate, starting 24 h prior to injury. In separate experiments, irradiation of hind limb was also performed to prevent resident cell proliferation. Upon euthanasia, blood and muscles were collected for flow cytometric analyses of macrophage/monocyte subsets. Results Clodronate induced a 80%-90% depletion of monocyte but only led to 57% and 41% decrease of M1 and M2 macrophage accumulation, respectively, 2 d following injury. Conversely, the number of M1 macrophages in monocyte-depleted rats was 2.4-fold higher than in non-depleted rats 4 d after injury. This was associated with a 16-fold increase in the number of proliferative M1 macrophages, which was reduced by 46% in irradiated animals. Proliferation of M2 macrophages was increased tenfold by clodronate treatment 4 d post injury. The accumulation of M2 macrophages was partially impaired by irradiation, regardless of monocyte depletion. Conclusions M1 and M2 subsets proliferate after skeletal muscle injury and their proliferation is enhanced under condition of monocyte depletion. Our study supports the conclusion that both infiltrating and resident precursors could contribute to M1 or M2 macrophage accumulation in muscle injury.
Collapse
Affiliation(s)
| | | | | | | | - Elise Duchesne
- Centre de Recherche du CHUL (CHUQ), 2705 Boulevard Laurier, RC-9800 Québec, Québec, Canada.
| |
Collapse
|
236
|
Abstract
Traumatic injury or disease of the spinal cord and brain elicits multiple cellular and biochemical reactions that together cause or are associated with neuropathology. Specifically, injury or disease elicits acute infiltration and activation of immune cells, death of neurons and glia, mitochondrial dysfunction, and the secretion of substrates that inhibit axon regeneration. In some diseases, inflammation is chronic or non-resolving. Ligands that target PPARs (peroxisome proliferator-activated receptors), a group of ligand-activated transcription factors, are promising therapeutics for neurologic disease and CNS injury because their activation affects many, if not all, of these interrelated pathologic mechanisms. PPAR activation can simultaneously weaken or reprogram the immune response, stimulate metabolic and mitochondrial function, promote axon growth and induce progenitor cells to differentiate into myelinating oligodendrocytes. PPAR activation has beneficial effects in many pre-clinical models of neurodegenerative diseases and CNS injury; however, the mechanisms through which PPARs exert these effects have yet to be fully elucidated. In this review we discuss current literature supporting the role of PPAR activation as a therapeutic target for treating traumatic injury and degenerative diseases of the CNS.
Collapse
|
237
|
Liu Y, Holdbrooks AT, De Sarno P, Rowse AL, Yanagisawa LL, McFarland BC, Harrington LE, Raman C, Sabbaj S, Benveniste EN, Qin H. Therapeutic efficacy of suppressing the Jak/STAT pathway in multiple models of experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 192:59-72. [PMID: 24323580 DOI: 10.4049/jimmunol.1301513] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Pathogenic Th cells and myeloid cells are involved in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The JAK/STAT pathway is used by numerous cytokines for signaling and is critical for development, regulation, and termination of immune responses. Dysregulation of the JAK/STAT pathway has pathological implications in autoimmune and neuroinflammatory diseases. Many of the cytokines involved in MS/EAE, including IL-6, IL-12, IL-23, IFN-γ, and GM-CSF, use the JAK/STAT pathway to induce biological responses. Thus, targeting JAKs has implications for treating autoimmune inflammation of the brain. We have used AZD1480, a JAK1/2 inhibitor, to investigate the therapeutic potential of inhibiting the JAK/STAT pathway in models of EAE. AZD1480 treatment inhibits disease severity in myelin oligodendrocyte glycoprotein-induced classical and atypical EAE models by preventing entry of immune cells into the brain, suppressing differentiation of Th1 and Th17 cells, deactivating myeloid cells, inhibiting STAT activation in the brain, and reducing expression of proinflammatory cytokines and chemokines. Treatment of SJL/J mice with AZD1480 delays disease onset of PLP-induced relapsing-remitting disease, reduces relapses and diminishes clinical severity. AZD1480 treatment was also effective in reducing ongoing paralysis induced by adoptive transfer of either pathogenic Th1 or Th17 cells. In vivo AZD1480 treatment impairs both the priming and expansion of T cells and attenuates Ag presentation functions of myeloid cells. Inhibition of the JAK/STAT pathway has clinical efficacy in multiple preclinical models of MS, suggesting the feasibility of the JAK/STAT pathway as a target for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Keratan sulfate expression in microglia is diminished in the spinal cord in experimental autoimmune neuritis. Cell Death Dis 2013; 4:e946. [PMID: 24309933 PMCID: PMC3877550 DOI: 10.1038/cddis.2013.479] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 10/29/2013] [Accepted: 11/05/2013] [Indexed: 01/07/2023]
Abstract
Experimental autoimmune neuritis (EAN) is an animal model of Guillain–Barré syndrome, an inflammatory demyelination disease of the peripheral nervous system. Although this disease has been extensively studied on peripheral nerves, the pathology of the central nervous system has not been fully understood. Previous studies demonstrate that expression of keratan sulfate (KS), the sugar chain of proteoglycan, is associated with activated microglia/macrophages accumulated after neuronal injuries. Unexpectedly, we found here that KS is rather diminished in rat EAN. KS was restrictively expressed in microglia in the spinal cord of normal rats. KS was positive in 50% microglia in the ventral horn and 20% in the dorsal horn. In EAN, microglia increased in number and expressed the activation marker CD68, but KS expression was abolished. Concomitantly, pro-inflammatory cytokines, i.e., interferon (IFN)-γ, interleukin (IL)-1β, and tumor necrosis factor (TNF)-α, were increased in the spinal cord of EAN rats, whereas anti-inflammatory cytokines, such as IL-4 and IL-10, were decreased. In addition, silencing of KSGal6ST attenuated KS expression on the primary cultured microglia and upregulated expression of some activation markers (TNF-α, IL-1β, and iNOS) under the stimulation with lipopolysaccharide and IFN-γ. This study demonstrates for the first time a close association of EAN and disappearance of KS on microglia. KS expression could be a useful marker to evaluate the status of polyneuropathy.
Collapse
|
239
|
Wang G, Zhang J, Hu X, Zhang L, Mao L, Jiang X, Liou AKF, Leak RK, Gao Y, Chen J. Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J Cereb Blood Flow Metab 2013; 33:1864-74. [PMID: 23942366 PMCID: PMC3851898 DOI: 10.1038/jcbfm.2013.146] [Citation(s) in RCA: 361] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 01/05/2023]
Abstract
Mononuclear phagocytes are a population of multi-phenotypic cells and have dual roles in brain destruction/reconstruction. The phenotype-specific roles of microglia/macrophages in traumatic brain injury (TBI) are, however, poorly characterized. In the present study, TBI was induced in mice by a controlled cortical impact (CCI) and animals were killed at 1 to 14 days post injury. Real-time polymerase chain reaction (RT-PCR) and immunofluorescence staining for M1 and M2 markers were performed to characterize phenotypic changes of microglia/macrophages in both gray and white matter. We found that the number of M1-like phagocytes increased in cortex, striatum and corpus callosum (CC) during the first week and remained elevated until at least 14 days after TBI. In contrast, M2-like microglia/macrophages peaked at 5 days, but decreased rapidly thereafter. Notably, the severity of white matter injury (WMI), manifested by immunohistochemical staining for neurofilament SMI-32, was strongly correlated with the number of M1-like phagocytes. In vitro experiments using a conditioned medium transfer system confirmed that M1 microglia-conditioned media exacerbated oxygen glucose deprivation-induced oligodendrocyte death. Our results indicate that microglia/macrophages respond dynamically to TBI, experiencing a transient M2 phenotype followed by a shift to the M1 phenotype. The M1 phenotypic shift may propel WMI progression and represents a rational target for TBI treatment.
Collapse
Affiliation(s)
- Guohua Wang
- 1] State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China [2] Department of Neurology, Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA [3] Department of Neuropharmacology, Institute of Nautical Medicine, Nantong University, Nantong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
von Leden RE, Cooney SJ, Ferrara TM, Zhao Y, Dalgard CL, Anders JJ, Byrnes KR. 808 nm wavelength light induces a dose-dependent alteration in microglial polarization and resultant microglial induced neurite growth. Lasers Surg Med 2013; 45:253-63. [PMID: 23619903 DOI: 10.1002/lsm.22133] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2013] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND OBJECTIVE Despite the success of using photobiomodulation (PBM), also known as low level light therapy, in promoting recovery after central nervous system (CNS) injury, the effect of PBM on microglia, the primary mediators of immune and inflammatory response in the CNS, remains unclear. Microglia exhibit a spectrum of responses to injury, with partial or full polarization into pro- and anti-inflammatory phenotypes. Pro-inflammatory (M1 or classically activated) microglia contribute to chronic inflammation and neuronal toxicity, while anti-inflammatory (M2 or alternatively activated) microglia play a role in wound healing and tissue repair; microglia can fall anywhere along this spectrum in response to stimulation. MATERIALS AND METHODS The effect of PBM on microglial polarization therefore was investigated using colorimetric assays, immunocytochemistry, proteomic profiling and RT-PCR in vitro after exposure of primary microglia or BV2 microglial cell line to PBM of differing energy densities (0.2, 4, 10, and 30 J/cm(2) , 808 nm wavelength, 50 mW output power). RESULTS PBM has a dose-dependent effect on the spectrum of microglial M1 and M2 polarization. Specifically, PBM with energy densities between 4 and 30 J/cm(2) induced expression of M1 markers in microglia. Markers of the M2 phenotype, including CD206 and TIMP1, were observed at lower energy densities of 0.2-10 J/cm(2) . In addition, co-culture of PBM or control-treated microglia with primary neuronal cultures demonstrated a dose-dependent effect of PBM on microglial-induced neuronal growth and neurite extension. CONCLUSION These data suggest that the Arndt-Schulz law as applied to PBM for a specific bioassay does not hold true in cells with a spectrum of responses, and that PBM can alter microglial phenotype across this spectrum in a dose-dependent manner. These data are therefore of important relevance to not only therapies in the CNS but also to understanding of PBM effects and mechanisms.
Collapse
Affiliation(s)
- Ramona E von Leden
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Jones Bridge Road, Bethesda, Maryland 20814, USA
| | | | | | | | | | | | | |
Collapse
|
241
|
Microglia: an active player in the regulation of synaptic activity. Neural Plast 2013; 2013:627325. [PMID: 24303218 PMCID: PMC3835777 DOI: 10.1155/2013/627325] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 09/05/2013] [Accepted: 09/19/2013] [Indexed: 12/18/2022] Open
Abstract
Synaptic plasticity is critical for elaboration and adaptation in the developing and developed brain. It is well established that astrocytes play an important role in the maintenance of what has been dubbed “the tripartite synapse”. Increasing evidence shows that a fourth cell type, microglia, is critical to this maintenance as well. Microglia are the resident macrophages of the central nervous system (CNS). Because of their well-characterized inflammatory functions, research has primarily focused on their innate immune properties. The role of microglia in the maintenance of synapses in development and in homeostasis is not as well defined. A number of significant findings have shed light on the critical role of microglia at the synapse. It is becoming increasingly clear that microglia play a seminal role in proper synaptic development and elimination.
Collapse
|
242
|
Carvalho SCD, Apolinário LM, Matheus SMM, Santo Neto H, Marques MJ. EPA protects against muscle damage in the mdx mouse model of Duchenne muscular dystrophy by promoting a shift from the M1 to M2 macrophage phenotype. J Neuroimmunol 2013; 264:41-7. [PMID: 24090650 DOI: 10.1016/j.jneuroim.2013.09.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/05/2013] [Accepted: 09/10/2013] [Indexed: 12/20/2022]
Abstract
In dystrophic mdx mice and in Duchenne muscular dystrophy, inflammation contributes to myonecrosis. Previously, we demonstrated that eicosapentaenoic acid (EPA) decreased inflammation and necrosis in dystrophic muscle. In the present study, we examined the effects of EPA and the corticoid deflazacort (DFZ) as modulators of M1 (iNOS-expressing cells) and M2 (CD206-expressing cells) macrophages. Mdx mice (14 days old) received EPA or DFZ for 16 days. The diaphragm, biceps brachii and quadriceps muscles were studied. Immunofluorescence, immunoblotting and ELISA assays showed that EPA increased interleucin-10, reduced interferon-γ and was more effective than DFZ in promoting a shift from M1 to M2.
Collapse
Affiliation(s)
- Samara Camaçari de Carvalho
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | | | | | | | | |
Collapse
|
243
|
Onore CE, Careaga M, Babineau BA, Schwartzer JJ, Berman RF, Ashwood P. Inflammatory macrophage phenotype in BTBR T+tf/J mice. Front Neurosci 2013; 7:158. [PMID: 24062633 PMCID: PMC3774991 DOI: 10.3389/fnins.2013.00158] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/16/2013] [Indexed: 12/05/2022] Open
Abstract
Although autism is a behaviorally defined disorder, many studies report an association with increased pro-inflammatory cytokine production. Recent characterization of the BTBR T+tf/J (BTBR) inbred mouse strain has revealed several behavioral characteristics including social deficits, repetitive behavior, and atypical vocalizations which may be relevant to autism. We therefore hypothesized that, asocial BTBR mice, which exhibit autism-like behaviors, may have an inflammatory immune profile similar to that observed in children with autism. The objectives of this study were to characterize the myeloid immune profile of BTBR mice and to explore their associations with autism-relevant behaviors. C57BL/6J (C57) mice and BTBR mice were tested for social interest and repetitive self-grooming behavior. Cytokine production was measured in bone-marrow derived macrophages incubated for 24 h in either growth media alone, LPS, IL-4/LPS, or IFNγ/LPS to ascertain any M1/M2 skewing. After LPS stimulation, BTBR macrophages produced higher levels of IL-6, MCP-1, and MIP-1α and lower IL-10 (p < 0.01) than C57 mice, suggesting an exaggerated inflammatory profile. After exposure to IL-4/LPS BTBR macrophages produced less IL-10 (p < 0.01) than C57 macrophages and more IL-12p40 (p < 0.01) suggesting poor M2 polarization. Levels of IL-12(p70) (p < 0.05) were higher in BTBR macrophages after IFNγ/LPS stimulation, suggesting enhanced M1 polarization. We further observed a positive correlation between grooming frequency, and production of IL-12(p40), IL-12p70, IL-6, and TNFα (p < 0.05) after treatment with IFNγ/LPS across both strains. Collectively, these data suggest that the asocial BTBR mouse strain exhibits a more inflammatory, or M1, macrophage profile in comparison to the social C57 strain. We have further demonstrated a relationship between this relative increase in inflammation and repetitive grooming behavior, which may have relevance to repetitive and stereotyped behavior of autism.
Collapse
Affiliation(s)
- Charity E Onore
- The MIND Institute, University of California Davis, CA, USA ; Department of Medical Microbiology and Immunology, University of California Davis, CA, USA
| | | | | | | | | | | |
Collapse
|
244
|
Kim H, Walczak P, Kerr C, Galpoththawela C, Gilad AA, Muja N, Bulte JWM. Immunomodulation by transplanted human embryonic stem cell-derived oligodendroglial progenitors in experimental autoimmune encephalomyelitis. Stem Cells 2013; 30:2820-9. [PMID: 22949039 DOI: 10.1002/stem.1218] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 08/09/2012] [Indexed: 12/18/2022]
Abstract
Transplantation of embryonic stem cells and their neural derivatives can lead to amelioration of the disease symptoms of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). Oligodendroglial progenitors (OPs), derived from human embryonic stem cells (hESC, HES-1), were labeled with superparamagnetic iron oxide and transduced with luciferase. At 7 days following induction of EAE in C57/BL6 mice, 1 × 10(6) cells were transplanted in the ventricles of C57/BL6 mice and noninvasively monitored by magnetic resonance and bioluminescence imaging. Cells were found to remain within the cerebroventricular system and did not survive for more than 10 days. However, EAE mice that received hESC-OPs showed a significant improvement in neurological disability scores (0.9 ± 0.2; n = 12) compared to that of control animals (3.3 ± 0.4; n = 12) at day 15 post-transplantation. Histopathologically, transplanted hESC-OPs generated TREM2-positive CD45 cells, increased TIMP-1 expression, confined inflammatory cells within the subarachnoid space, and gave rise to higher numbers of Foxp3-positive regulatory T cells in the spinal cord and spleen. Our results suggest that transplantation of hESC-OPs can alter the pathogenesis of EAE through immunomodulation, potentially providing new avenues for stem cell-based treatment of MS.
Collapse
Affiliation(s)
- Heechul Kim
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2195, USA
| | | | | | | | | | | | | |
Collapse
|
245
|
Abstract
Microglia are critical nervous system-specific cells influencing brain development, maintenance of the neural environment, response to injury, and repair. They contribute to neuronal proliferation and differentiation, pruning of dying neurons, synaptic remodeling and clearance of debris and aberrant proteins. Colonization of the brain occurs during gestation with an expansion following birth with localization stimulated by programmed neuronal death, synaptic pruning, and axonal degeneration. Changes in microglia phenotype relate to cellular processes including specific neurotransmitter, pattern recognition, or immune-related receptor activation. Upon activation, microglia cells have the capacity to release a number of substances, e.g., cytokines, chemokines, nitric oxide, and reactive oxygen species, which could be detrimental or beneficial to the surrounding cells. With aging, microglia shift their morphology and may display diminished capacity for normal functions related to migration, clearance, and the ability to shift from a pro-inflammatory to an anti-inflammatory state to regulate injury and repair. This shift in microglia potentially contributes to increased susceptibility and neurodegeneration as a function of age. In the current review, information is provided on the colonization of the brain by microglia, the expression of various pattern recognition receptors to regulate migration and phagocytosis, and the shift in related functions that occur in normal aging.
Collapse
Affiliation(s)
- G Jean Harry
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, MD C1-04, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
246
|
Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, van Wijngaarden P, Wagers AJ, Williams A, Franklin RJM, Ffrench-Constant C. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci 2013; 16:1211-1218. [PMID: 23872599 DOI: 10.1038/nn.3469] [Citation(s) in RCA: 1256] [Impact Index Per Article: 104.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/14/2013] [Indexed: 02/08/2023]
Abstract
The lack of therapies for progressive multiple sclerosis highlights the need to understand the regenerative process of remyelination that can follow CNS demyelination. This involves an innate immune response consisting of microglia and macrophages, which can be polarized to distinct functional phenotypes: pro-inflammatory (M1) and anti-inflammatory or immunoregulatory (M2). We found that a switch from an M1- to an M2-dominant response occurred in microglia and peripherally derived macrophages as remyelination started. Oligodendrocyte differentiation was enhanced in vitro with M2 cell conditioned media and impaired in vivo following intra-lesional M2 cell depletion. M2 cell densities were increased in lesions of aged mice in which remyelination was enhanced by parabiotic coupling to a younger mouse and in multiple sclerosis lesions that normally show remyelination. Blocking M2 cell-derived activin-A inhibited oligodendrocyte differentiation during remyelination in cerebellar slice cultures. Thus, our results indicate that M2 cell polarization is essential for efficient remyelination and identify activin-A as a therapeutic target for CNS regeneration.
Collapse
Affiliation(s)
- Veronique E Miron
- MRC Centre for Regenerative Medicine/MS Society Centre for Translational Research, University of Edinburgh, Edinburgh, UK
| | - Amanda Boyd
- MRC Centre for Regenerative Medicine/MS Society Centre for Translational Research, University of Edinburgh, Edinburgh, UK
| | - Jing-Wei Zhao
- Wellcome Trust and MRC Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Tracy J Yuen
- MRC Centre for Regenerative Medicine/MS Society Centre for Translational Research, University of Edinburgh, Edinburgh, UK.,Wellcome Trust and MRC Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Julia M Ruckh
- Wellcome Trust and MRC Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jennifer L Shadrach
- Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute & Joslin Diabetes Center, Cambridge, USA
| | - Peter van Wijngaarden
- Wellcome Trust and MRC Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Amy J Wagers
- Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute & Joslin Diabetes Center, Cambridge, USA
| | - Anna Williams
- MRC Centre for Regenerative Medicine/MS Society Centre for Translational Research, University of Edinburgh, Edinburgh, UK
| | - Robin J M Franklin
- Wellcome Trust and MRC Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Charles Ffrench-Constant
- MRC Centre for Regenerative Medicine/MS Society Centre for Translational Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
247
|
Podojil JR, Miller SD. Targeting the B7 family of co-stimulatory molecules: successes and challenges. BioDrugs 2013; 27:1-13. [PMID: 23329394 DOI: 10.1007/s40259-012-0001-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As more patient data is cross-referenced with animal models of disease, the primary focus on T(h)1 autoreactive effector cell function in autoimmune diseases, such as rheumatoid arthritis and multiple sclerosis, has shifted towards the role of T(h)17 autoreactive effector cells and the ability of regulatory T cells (T(reg)) to modulate the pro-inflammatory autoimmune response. Therefore, the currently favored hypothesis is that a delicate balance between T(h)1/17 effector cells and T(reg) cell function is critical in the regulation of inflammatory autoimmune disease. An intensive area of research with regard to the T(h)1/17:T(reg) cell balance is the utilization of blockade and/or ligation of various co-stimulatory or co-inhibitory molecules, respectively, during ongoing disease to skew the immune response toward a more tolerogenic/regulatory state. Currently, FDA-approved therapies for multiple sclerosis patients are all aimed at the suppression of immune cell function. The other favored method of treatment is a modulation or deletion of autoreactive immune cells via short-term blockade of activating co-stimulatory receptors via treatment with fusion proteins such as CTLA4-Ig and CTLA4-FasL. Based on the initial success of CTLA4-Ig, there are additional fusion proteins that are currently under development. Examples of the more recently identified B7/CD28 family members are PD-L1, PD-L2, inducible co-stimulatory molecule-ligand (ICOS-L), B7-H3, and B7-H4, all of which may emerge as potential fusion protein therapeutics, each with unique, yet often overlapping functions. The expression of both stimulatory and inhibitory B7 molecules seems to play an essential role in modulating immune cell function through a variety of mechanisms, which is supported by findings that suggest each B7 molecule has developed its own indispensable niche in the immune system. As more data are generated, the diagnostic and therapeutic potential of the above B7 family-member-derived fusion proteins becomes ever more apparent. Besides defining the biology of these B7/CD28 family members in vivo, additional difficulty in the development of these therapies lies in maintaining the normal immune functions of recognition and reaction to non-self-antigens following viral or bacterial infection in the patient. Further complicating the clinical translation of these therapies, the mechanism of action identified for a particular reagent may depend upon the method of immune-cell activation and the subset of immune cells targeted in the study.
Collapse
Affiliation(s)
- Joseph R Podojil
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Tarry 6-718, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | | |
Collapse
|
248
|
Al Faraj A, Luciani N, Kolosnjaj-Tabi J, Mattar E, Clement O, Wilhelm C, Gazeau F. Real-time high-resolution magnetic resonance tracking of macrophage subpopulations in a murine inflammation model: a pilot study with a commercially available cryogenic probe. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:193-203. [PMID: 23281292 DOI: 10.1002/cmmi.1516] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 09/12/2012] [Accepted: 10/10/2012] [Indexed: 12/14/2022]
Abstract
Macrophages present different polarization states exhibiting distinct functions in response to environmental stimuli. However, the dynamic of their migration to sites of inflammation is not fully elucidated. Here we propose a real-time in vivo cell tracking approach, using high-resolution (HR)-MRI obtained with a commercially available cryogenic probe (Cryoprobe™), to monitor trafficking of differently polarized macrophages after systemic injection into mice. Murine bone marrow-derived mononuclear cells were differentiated ex vivo into nonpolarized M0, pro-inflammatory M1 and immunomodulator M2 macrophage subsets and labeled with citrate-coated anionic iron oxide nanoparticles (AMNP). These cells were subsequently intravenously injected to mice bearing calf muscle inflammation. Whole body migration dynamics of macrophage subsets was monitored by MRI at 4.7 T with a volume transmission/reception radiofrequency coil and macrophage infiltration to the inflamed paw was monitored with the cryogenic probe, allowing 3D spatial resolution of 50 µm with a scan time of only 10 min. Capture of AMNP was rapid and efficient regardless of macrophage polarization, with the highest uptake in M2 macrophages. Flow cytometry confirmed that macrophages preserved their polarization hallmarks after labeling. Migration kinetics of labeled cells differed from that of free AMNP. A preferential homing of M2-polarized macrophages to inflammation sites was observed. Our in vivo HR-MRI protocol highlights the extent of macrophage infiltration to the inflammation site. Coupled to whole body imaging, HR-MRI provides quantitative information on the time course of migration of ex vivo-polarized intravenously injected macrophages.
Collapse
Affiliation(s)
- Achraf Al Faraj
- Laboratoire Matière et Systèmes Complexes MSC, CNRS UMR7057, Université Paris Diderot, Paris, France
| | | | | | | | | | | | | |
Collapse
|
249
|
In vitro and in vivo models of cerebral ischemia show discrepancy in therapeutic effects of M2 macrophages. PLoS One 2013; 8:e67063. [PMID: 23825621 PMCID: PMC3692438 DOI: 10.1371/journal.pone.0067063] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/13/2013] [Indexed: 01/16/2023] Open
Abstract
THE INFLAMMATORY RESPONSE FOLLOWING ISCHEMIC STROKE IS DOMINATED BY INNATE IMMUNE CELLS: resident microglia and blood-derived macrophages. The ambivalent role of these cells in stroke outcome might be explained in part by the acquisition of distinct functional phenotypes: classically (M1) and alternatively activated (M2) macrophages. To shed light on the crosstalk between hypoxic neurons and macrophages, an in vitro model was set up in which bone marrow-derived macrophages were co-cultured with hippocampal slices subjected to oxygen and glucose deprivation. The results showed that macrophages provided potent protection against neuron cell loss through a paracrine mechanism, and that they expressed M2-type alternative polarization. These findings raised the possibility of using bone marrow-derived M2 macrophages in cellular therapy for stroke. Therefore, 2 million M2 macrophages (or vehicle) were intravenously administered during the subacute stage of ischemia (D4) in a model of transient middle cerebral artery occlusion. Functional neuroscores and magnetic resonance imaging endpoints (infarct volumes, blood-brain barrier integrity, phagocytic activity assessed by iron oxide uptake) were longitudinally monitored for 2 weeks. This cell-based treatment did not significantly improve any outcome measure compared with vehicle, suggesting that this strategy is not relevant to stroke therapy.
Collapse
|
250
|
The benefits and detriments of macrophages/microglia in models of multiple sclerosis. Clin Dev Immunol 2013; 2013:948976. [PMID: 23840244 PMCID: PMC3694375 DOI: 10.1155/2013/948976] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022]
Abstract
The central nervous system (CNS) is immune privileged with access to leukocytes being limited. In several neurological diseases, however, infiltration of immune cells from the periphery into the CNS is largely observed and accounts for the increased representation of macrophages within the CNS. In addition to extensive leukocyte infiltration, the activation of microglia is frequently observed. The functions of activated macrophages/microglia within the CNS are complex. In three animal models of multiple sclerosis (MS), namely, experimental autoimmune encephalomyelitis (EAE) and cuprizone- and lysolecithin-induced demyelination, there have been many reported detrimental roles associated with the involvement of macrophages and microglia. Such detriments include toxicity to neurons and oligodendrocyte precursor cells, release of proteases, release of inflammatory cytokines and free radicals, and recruitment and reactivation of T lymphocytes in the CNS. Many studies, however, have also reported beneficial roles of macrophages/microglia, including axon regenerative roles, assistance in promoting remyelination, clearance of inhibitory myelin debris, and the release of neurotrophic factors. This review will discuss the evidence supporting the detrimental and beneficial aspects of macrophages/microglia in models of MS, provide a discussion of the mechanisms underlying the dichotomous roles, and describe a few therapies in clinical use in MS that impinge on the activity of macrophages/microglia.
Collapse
|