Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Clin Exp Immunol ; 214(1): 1-17, 2023 12 11.
Article in English | MEDLINE | ID: mdl-37410892

ABSTRACT

Multiple sclerosis (MS) is characterized by the chronic inflammatory destruction of myelinated axons in the central nervous system. Several ideas have been put forward to clarify the roles of the peripheral immune system and neurodegenerative events in such destruction. Yet, none of the resulting models appears to be consistent with all the experimental evidence. They also do not answer the question of why MS is exclusively seen in humans, how Epstein-Barr virus contributes to its development but does not immediately trigger it, and why optic neuritis is such a frequent early manifestation in MS. Here we describe a scenario for the development of MS that unifies existing experimental evidence as well as answers the above questions. We propose that all manifestations of MS are caused by a series of unfortunate events that usually unfold over a longer period of time after a primary EBV infection and involve periodic weakening of the blood-brain barrier, antibody-mediated CNS disturbances, accumulation of the oligodendrocyte stress protein αB-crystallin and self-sustaining inflammatory damage.


Subject(s)
Epstein-Barr Virus Infections , Multiple Sclerosis , Humans , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human , Central Nervous System , Blood-Brain Barrier/pathology
2.
Front Neurosci ; 14: 574, 2020.
Article in English | MEDLINE | ID: mdl-32595446

ABSTRACT

Progressive neuronal death during tauopathies is associated with aggregation of modified, truncated or mutant forms of tau protein. Such aggregates are neurotoxic, promote spreading of tau aggregation, and trigger release of pro-inflammatory factors by glial cells. Counteracting such pathogenic effects of tau by simultaneously inhibiting protein aggregation as well as pro-inflammatory glial cell responses would be of significant therapeutic interest. Here, we examined the use of the small heat-shock protein HspB5 for this purpose. As a molecular chaperone, HspB5 counteracts aggregation of a wide range of abnormal proteins. As a TLR2 agonist, it selectively activates protective responses by CD14-expressing myeloid cells including microglia. We show that intracerebral infusion of HspB5 in transgenic mice with selective neuronal expression of mutant human P301S tau has significant neuroprotective effects in the superficial, frontal cortical layers. Underlying these effects at least in part, HspB5 induces several potent neuroprotective mediators in both astrocytes and microglia including neurotrophic factors and increased potential for removal of glutamate. Together, these findings highlight the potentially broad therapeutic potential of HspB5 in neurodegenerative proteinopathies.

3.
Front Neurol ; 10: 570, 2019.
Article in English | MEDLINE | ID: mdl-31244750

ABSTRACT

Sensorineural hearing loss is the most common long-term deficit after pneumococcal meningitis (PM), occurring in up to 30% of surviving patients. The infection and the following overshooting inflammatory host response damage the vulnerable sensory cells of the inner ear, resulting in loss of hair cells and spiral ganglion neurons, ultimately leading to elevated hearing thresholds. Here, we tested the oto-protective properties of the small heat shock protein alpha B-crystallin (HspB5) with previously reported anti-inflammatory, anti-apoptotic and neuroprotective functions, in an experimental model of PM-induced hearing loss. We analyzed the effect of local and systemic delivery of HspB5 in an infant rat model of PM, as well as ex vivo, using whole mount cultures. Cytokine secretion profile, hearing thresholds and inner ear damage were assessed at predefined stages of the disease up to 1 month after infection. PM was accompanied by elevated pro-inflammatory cytokine concentrations in the cerebrospinal fluid (CSF), leukocyte and neutrophil infiltration in the perilymphatic spaces of the cochlea with neutrophils extracellular trap formation during the acute phase of the disease. Elevated hearing thresholds were measured after recovery from meningitis. Intracisternal but not intraperitoneal administration of HspB5 significantly reduced the levels of TNF-α, IL-6 IFN-γ and IL-10 in the acute phase of the disease. This resulted in a greater outer hair cell survival, as well as improved hearing thresholds at later stages. These results suggest that high local concentrations of HspB5 are needed to prevent inner ear damage in acute PM. HspB5 represents a promising therapeutic option to improve the auditory outcome and counteract hearing loss after PM.

4.
J Immunol ; 202(8): 2421-2430, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30804043

ABSTRACT

TLR-induced signaling potently activates cells of the innate immune system and is subject to regulation at different levels. Inflammatory conditions are associated with increased levels of extracellular adenosine, which can modulate TLR-induced production of cytokines through adenosine receptor-mediated signaling. There are four adenosine receptor subtypes that induce different signaling cascades. In this study, we demonstrate a pivotal contribution of adenosine A3 receptor (A3R)-mediated signaling to the TLR4-induced expression of IL-12 in different types of human myeloid APC. In dendritic cells, IL-12 and CCL2 responses as evoked by TLR2, 3, 4, 5, and 8, as well as IL-12 responses evoked by whole pathogens, were all reduced when A3R-mediated signaling was blocked. As a result, concomitant production of IFN-γ and IL-17 by T cells was significantly inhibited. We further show that selective inhibition of A3R-mediated signaling reduced TLR-induced phosphorylation of the transcription factor STAT1 at tyrosine 701. Next-generation sequencing revealed that A3R-mediated signaling controls the expression of metallothioneins, known inhibitors of STAT1 phosphorylation. Together our results reveal a novel regulatory layer of innate immune responses, with a central role for metallothioneins and autocrine/paracrine signaling via A3Rs.


Subject(s)
Antigen-Presenting Cells/immunology , Chemokine CCL2/immunology , Interleukin-12/immunology , Myeloid Cells/immunology , Receptor, Adenosine A3/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Antigen-Presenting Cells/cytology , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Myeloid Cells/cytology , THP-1 Cells
5.
Glia ; 65(3): 460-473, 2017 03.
Article in English | MEDLINE | ID: mdl-28063173

ABSTRACT

The glial stress protein alpha B-crystallin (HSPB5) is an endogenous agonist for Toll-like receptor 2 in CD14+ cells. Following systemic administration, HSPB5 acts as a potent inhibitor of neuroinflammation in animal models and reduces lesion development in multiple sclerosis patients. Here, we show that systemically administered HSPB5 rapidly crosses the blood-brain barrier, implicating microglia as additional targets for HSPB5 along with peripheral monocytes and macrophages. To compare key players in the HSPB5-induced protective response of human macrophages and microglia, we applied weighted gene co-expression network analysis on transcript expression data obtained 1 and 4 h after activation. This approach identified networks of genes that are co-expressed in all datasets, thus reducing the complexity of the nonsynchronous waves of transcripts that appear after activation by HSPB5. In both cell types, HSPB5 activates a network of highly connected genes that appear to be functionally equivalent and consistent with the therapeutic effects of HSPB5 in vivo, since both networks include factors that suppress apoptosis, the production of proinflammatory factors, and the development of adaptive immunity. Yet, hub genes at the core of the network in either cell type were strikingly different. They prominently feature the well-known tolerance-promoting programmed-death ligand 1 as a key player in the macrophage response to HSPB5, and the immune-regulatory enzyme cyclooxygenase-2 (COX-2) in that of microglia. This latter finding indicates that despite its reputation as a potential target for nonsteroidal anti-inflammatory drugs, microglial COX-2 plays a central role in the therapeutic effects of HSPB5 during neuroinflammation. GLIA 2017;65:460-473.


Subject(s)
Cyclooxygenase 2/metabolism , Macrophages/drug effects , Macrophages/metabolism , Microglia/drug effects , Microglia/metabolism , alpha-Crystallin B Chain/pharmacology , Animals , Brain/cytology , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Parenchymal Tissue/cytology , Parenchymal Tissue/drug effects , RNA, Messenger/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism , alpha-Crystallin B Chain/metabolism
6.
CNS Neurol Disord Drug Targets ; 16(3): 244-256, 2017.
Article in English | MEDLINE | ID: mdl-27804858

ABSTRACT

Heat shock proteins (HSPs) are families of molecular chaperones that play important homeostatic functions in the central nervous system (CNS) by preventing protein misfolding, promoting degradation of improperly folded proteins, and protecting against apoptosis and inflammatory damage especially during hyperthermia, hypoxia, or oxidative stress. Under stress conditions, HSPs are upregulated to protect cells from damage that accumulates during ageing as well as pathological conditions. An important, yet frequently overlooked function of some HSPs is their ability to function as extracellular messengers (also termed chaperokines) that modulate immune responses within the CNS. Given the strong association between protein aggregation, innate immune cell activation and neurodegeneration, the expression and roles of HSPs in the CNS is attracting attention in many neurodegenerative disorders including inflammatory diseases such as multiple sclerosis, protein folding diseases such as Alzheimer's disease and amyotrophic lateral sclerosis, and genetic white matter diseases. This is especially so since several studies show that HSPs act therapeutically by modulating innate immune activation and may thus serve as neuroprotective agents. Here we review the evidence linking HSPs with neurodegenerative disorders in humans and the experimental animal models of these disorders. We discuss the mechanisms by which HSPs protect cells, and how the knowledge of their endogenous functions can be exploited to treat disorders of the CNS.


Subject(s)
Central Nervous System/metabolism , Heat-Shock Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Animals , Humans
7.
Immunology ; 149(2): 146-56, 2016 10.
Article in English | MEDLINE | ID: mdl-27388634

ABSTRACT

Current therapies for multiple sclerosis (MS) reduce the frequency of relapses by modulating adaptive immune responses but fail to limit the irreversible neurodegeneration driving progressive disability. Experimental autoimmune encephalomyelitis (EAE) in Biozzi ABH mice recapitulates clinical features of MS including relapsing-remitting episodes and secondary-progressive disability. To address the contribution of recurrent inflammatory events and ageing as factors that amplify progressive neurological disease, we examined EAE in 8- to 12-week-old and 12-month-old ABH mice. Compared with the relapsing-remitting (RREAE) and secondary progressive (SPEAE) EAE observed in young mice, old mice developed progressive disease from onset (PEAE) associated with pronounced axonal damage and increased numbers of CD3(+) T cells and microglia/macrophages, but not B cells. Whereas the clinical neurological features of PEAE and SPEAE were comparable, the pathology was distinct. SPEAE was associated with significantly reduced perivascular infiltrates and T-cell numbers in the central nervous system (CNS) compared with PEAE and the acute phase of RREAE. In contrast to perivascular infiltrates that declined during progression from RREAE into SPEAE, the numbers of microglia clusters remained constant. Similar to what is observed during MS, the microglia clusters emerging during EAE were associated with axonal damage and oligodendrocytes expressing heat-shock protein B5, but not lymphocytes. Taken together, our data reveal that the course of EAE is dependent on the age of the mice. Younger mice show a relapsing-remitting phase followed by progressive disease, whereas old mice immediately show progression. This indicates that recurrent episodes of inflammation in the CNS, as well as age, contribute to progressive neurological disease.


Subject(s)
Aging/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Neurogenic Inflammation/immunology , Oligodendroglia/immunology , T-Lymphocytes/immunology , alpha-Crystallin B Chain/metabolism , Animals , Apoptosis , Cells, Cultured , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred Strains , Oxidative Stress , Up-Regulation , alpha-Crystallin B Chain/genetics
8.
Brain Pathol ; 26(2): 248-57, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26780491

ABSTRACT

Neurological dysfunction and motor neuron degeneration in amyotrophic lateral sclerosis (ALS) is strongly associated with neuroinflammation reflected by activated microglia and astrocytes in the CNS. In ALS endogenous triggers in the CNS such as aggregated protein and misfolded proteins activate a pathogenic response by innate immune cells. However, there is also strong evidence for a neuroprotective immune response in ALS. Emerging evidence also reveals changes in the peripheral adaptive immune responses as well as alterations in the blood brain barrier that may aid traffic of lymphocytes and antibodies into the CNS. Understanding the triggers of neuroinflammation is key to controlling neuronal loss. Here, we review the current knowledge regarding the roles of non-neuronal cells as well as the innate and adaptive immune responses in ALS. Existing ALS animal models, in particular genetic rodent models, are very useful to study the underlying pathogenic mechanisms of motor neuron degeneration. We also discuss the approaches used to target the pathogenic immune responses and boost the neuroprotective immune pathways as novel immunotherapies for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Blood-Brain Barrier/metabolism , Immune System/physiopathology , Lymphocytes/physiology , Neuroglia/physiology , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Blood-Brain Barrier/drug effects , Humans , Immune System/drug effects , Lymphocytes/drug effects , Neuroglia/drug effects
9.
Eur J Immunol ; 46(3): 701-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26576501

ABSTRACT

Pentraxin-3 (PTX3), an acute-phase protein released during inflammation, aids phagocytic clearance of pathogens and apoptotic cells, and plays diverse immunoregulatory roles in tissue injury. In neuroinflammatory diseases, like MS, resident microglia could become activated by endogenous agonists for Toll like receptors (TLRs). Previously we showed a strong TLR2-mediated induction of PTX3 in cultured human microglia and macrophages by HspB5, which accumulates in glia during MS. Given the anti-inflammatory effects of HspB5, we examined the contribution of PTX3 to these effects in MS and its animal model EAE. Our data indicate that TLR engagement effectively induces PTX3 expression in human microglia, and that such expression is readily detectable in MS lesions. Enhanced PTX3 expression is prominently expressed in microglia in preactive MS lesions, and in microglia/macrophages engaged in myelin phagocytosis in actively demyelinating lesions. Yet, we did not detect PTX3 in cerebrospinal fluid of MS patients. PTX3 expression is also elevated in spinal cords during chronic relapsing EAE in Biozzi ABH mice, but the EAE severity and time course in PTX3-deficient mice did not differ from WT mice. Moreover, systemic PTX3 administration did not alter the disease onset or severity. Our findings reveal local functions of PTX3 during neuroinflammation in facilitating myelin phagocytosis, but do not point to a role for PTX3 in controlling the development of autoimmune neuroinflammation.


Subject(s)
Brain/immunology , C-Reactive Protein/administration & dosage , C-Reactive Protein/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Serum Amyloid P-Component/administration & dosage , Serum Amyloid P-Component/genetics , Spine/immunology , Animals , Brain/pathology , C-Reactive Protein/cerebrospinal fluid , C-Reactive Protein/immunology , Disease Models, Animal , Humans , Inflammation/immunology , Macrophages/immunology , Mice , Mice, Biozzi , Microglia/immunology , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Phagocytosis , Serum Amyloid P-Component/cerebrospinal fluid , Serum Amyloid P-Component/immunology , Spine/pathology , Toll-Like Receptors/immunology , Up-Regulation
10.
Acta Neuropathol Commun ; 3: 87, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26694816

ABSTRACT

INTRODUCTION: The important protective role of small heat-shock proteins (HSPs) in regulating cellular survival and migration, counteracting protein aggregation, preventing apoptosis, and regulating inflammation in the central nervous system is now well-recognized. Yet, their role in the neuroinflammatory disorder multiple sclerosis (MS) is largely undocumented. With the exception of alpha B-crystallin (HSPB5), little is known about the roles of small HSPs in disease. RESULTS: Here, we examined the expression of four small HSPs during lesion development in MS, focussing on their cellular distribution, and regional differences between white matter (WM) and grey matter (GM). It is well known that MS lesions in these areas differ markedly in their pathology, with substantially more intense blood-brain barrier damage, leukocyte infiltration and microglial activation typifying WM but not GM lesions. We analysed transcript levels and protein distribution profiles for HSPB1, HSPB6, HSPB8 and HSPB11 in MS lesions at different stages, comparing them with normal-appearing brain tissue from MS patients and non-neurological controls. During active stages of demyelination in WM, and especially the centre of chronic active MS lesions, we found significantly increased expression of HSPB1, HSPB6 and HSPB8, but not HSPB11. When induced, small HSPs were exclusively found in astrocytes but not in oligodendrocytes, microglia or neurons. Surprisingly, while the numbers of astrocytes displaying high expression of small HSPs were markedly increased in actively demyelinating lesions in WM, no such induction was observed in GM lesions. This difference was particularly obvious in leukocortical lesions covering both WM and GM areas. CONCLUSIONS: Since induction of small HSPs in astrocytes is apparently a secondary response to damage, their differential expression between WM and GM likely reflects differences in mediators that accompany demyelination in either WM or GM during MS. Our findings also suggest that during MS, cortical structures fail to benefit from the protective actions of small HSPs.


Subject(s)
Gene Expression Regulation/physiology , Gray Matter/metabolism , Heat-Shock Proteins, Small/metabolism , Multiple Sclerosis/pathology , White Matter/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Central Nervous System/pathology , Female , Glial Fibrillary Acidic Protein/metabolism , HLA-DR Antigens/metabolism , Heat-Shock Proteins, Small/genetics , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Myelin Proteolipid Protein/metabolism , RNA, Messenger/metabolism , Statistics, Nonparametric
11.
PLoS One ; 10(11): e0143366, 2015.
Article in English | MEDLINE | ID: mdl-26599332

ABSTRACT

UNLABELLED: As a molecular chaperone and activator of Toll-like receptor 2-mediated protective responses by microglia and macrophages, the small heat shock protein alpha B-crystallin (HspB5) exerts therapeutic effects in different animal models for neuroinflammation, including the model for multiple sclerosis (MS). Yet, HspB5 can also stimulate human antigen-specific memory T cells to release IFN-γ, a cytokine with well-documented detrimental effects during MS. In this study, we explored in a Phase IIa randomized clinical trial the therapeutic application of HspB5 in relapsing-remitting MS (RR-MS), using intravenous doses sufficient to support its protective effects, but too low to trigger pathogenic memory T-cell responses. These sub-immunogenic doses were selected based on in vitro analysis of the dose-response profile of human T cells and macrophages to HspB5, and on the immunological effects of HspB5 in healthy humans as established in a preparatory Phase I study. In a 48-week randomized, placebo-controlled, double-blind Phase IIa trial, three bimonthly intravenous injections of 7.5, 12.5 or 17.5 mg HspB5 were found to be safe and well tolerated in RR-MS patients. While predefined clinical endpoints did not differ significantly between the relatively small groups of MS patients treated with either HspB5 or placebo, repeated administration especially of the lower doses of HspB5 led to a progressive decline in MS lesion activity as monitored by magnetic resonance imaging (MRI), which was not seen in the placebo group. Exploratory linear regression analysis revealed this decline to be significant in the combined group receiving either of the two lower doses, and to result in a 76% reduction in both number and total volumes of active MRI lesions at 9 months into the study. These data provide the first indication for clinical benefit resulting from intervention in RR-MS with HspB5. TRIAL REGISTRATION: ClinicalTrials.gov Phase I: NCT02442557; Phase IIa: NCT02442570.


Subject(s)
Multiple Sclerosis/drug therapy , alpha-Crystallin B Chain/administration & dosage , alpha-Crystallin B Chain/therapeutic use , Adult , Double-Blind Method , Female , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Treatment Outcome
12.
Acta Neuropathol ; 128(2): 215-29, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997049

ABSTRACT

Activated microglia and macrophages play a key role in driving demyelination during multiple sclerosis (MS), but the factors responsible for their activation remain poorly understood. Here, we present evidence for a dual-trigger role of IFN-γ and alpha B-crystallin (HSPB5) in this context. In MS-affected brain tissue, accumulation of the molecular chaperone HSPB5 by stressed oligodendrocytes is a frequent event. We have shown before that this triggers a TLR2-mediated protective response in surrounding microglia, the molecular signature of which is widespread in normal-appearing brain tissue during MS. Here, we show that IFN-γ, which can be released by infiltrated T cells, changes the protective response of microglia and macrophages to HSPB5 into a robust pro-inflammatory classical response. Exposure of cultured microglia and macrophages to IFN-γ abrogated subsequent IL-10 induction by HSPB5, and strongly promoted HSPB5-triggered release of TNF-α, IL-6, IL-12, IL-1ß and reactive oxygen and nitrogen species. In addition, high levels of CXCL9, CXCL10, CXL11, several guanylate-binding proteins and the ubiquitin-like protein FAT10 were induced by combined activation with IFN-γ and HSPB5. As immunohistochemical markers for microglia and macrophages exposed to both IFN-γ and HSPB5, these latter factors were found to be selectively expressed in inflammatory infiltrates in areas of demyelination during MS. In contrast, they were absent from activated microglia in normal-appearing brain tissue. Together, our data suggest that inflammatory demyelination during MS is selectively associated with IFN-γ-induced re-programming of an otherwise protective response of microglia and macrophages to the endogenous TLR2 agonist HSPB5.


Subject(s)
Interferon-gamma/metabolism , Macrophages/physiology , Microglia/physiology , Multiple Sclerosis/immunology , alpha-Crystallin B Chain/metabolism , Brain/immunology , Brain/pathology , Cells, Cultured , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Macrophages/pathology , Microglia/pathology , Multiple Sclerosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Ubiquitins/metabolism
13.
Mult Scler ; 20(8): 1030-2, 2014 07.
Article in English | MEDLINE | ID: mdl-24368758
14.
Immunology ; 142(2): 151-66, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24329535

ABSTRACT

Neurodegeneration, the progressive dysfunction and loss of neurons in the central nervous system (CNS), is the major cause of cognitive and motor dysfunction. While neuronal degeneration is well-known in Alzheimer's and Parkinson's diseases, it is also observed in neurotrophic infections, traumatic brain and spinal cord injury, stroke, neoplastic disorders, prion diseases, multiple sclerosis and amyotrophic lateral sclerosis, as well as neuropsychiatric disorders and genetic disorders. A common link between these diseases is chronic activation of innate immune responses including those mediated by microglia, the resident CNS macrophages. Such activation can trigger neurotoxic pathways leading to progressive degeneration. Yet, microglia are also crucial for controlling inflammatory processes, and repair and regeneration. The adaptive immune response is implicated in neurodegenerative diseases contributing to tissue damage, but also plays important roles in resolving inflammation and mediating neuroprotection and repair. The growing awareness that the immune system is inextricably involved in mediating damage as well as regeneration and repair in neurodegenerative disorders, has prompted novel approaches to modulate the immune system, although it remains whether these approaches can be used in humans. Additional factors in humans include ageing and exposure to environmental factors such as systemic infections that provide additional clues that may be human specific and therefore difficult to translate from animal models. Nevertheless, a better understanding of how immune responses are involved in neuronal damage and regeneration, as reviewed here, will be essential to develop effective therapies to improve quality of life, and mitigate the personal, economic and social impact of these diseases.


Subject(s)
Neurodegenerative Diseases/immunology , Humans , Inflammation/immunology , Neurodegenerative Diseases/therapy
15.
Immunology ; 141(3): 302-13, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23981039

ABSTRACT

Communication between the immune system and the central nervous system (CNS) is exemplified by cross-talk between glia and neurons shown to be essential for maintaining homeostasis. While microglia are actively modulated by neurons in the healthy brain, little is known about the cross-talk between oligodendrocytes and microglia. Oligodendrocytes, the myelin-forming cells in the CNS, are essential for the propagation of action potentials along axons, and additionally serve to support neurons by producing neurotrophic factors. In demyelinating diseases such as multiple sclerosis, oligodendrocytes are thought to be the victims. Here, we review evidence that oligodendrocytes also have strong immune functions, express a wide variety of innate immune receptors, and produce and respond to chemokines and cytokines that modulate immune responses in the CNS. We also review evidence that during stress events in the brain, oligodendrocytes can trigger a cascade of protective and regenerative responses, in addition to responses that elicit progressive neurodegeneration. Knowledge of the cross-talk between microglia and oligodendrocytes may continue to uncover novel pathways of immune regulation in the brain that could be further exploited to control neuroinflammation and degeneration.


Subject(s)
Brain/immunology , Cell Communication , Microglia/immunology , Oligodendroglia/immunology , Signal Transduction , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Cytokines/metabolism , Demyelinating Diseases/immunology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Exosomes/metabolism , Heat-Shock Proteins/metabolism , Humans , Microglia/metabolism , Microglia/pathology , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Stress, Physiological
16.
J Neuropathol Exp Neurol ; 72(10): 970-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24042199

ABSTRACT

Microglial nodules are frequently observed in the normal-appearing white matter of multiple sclerosis (MS) patients. Previously, we have shown that these clusters, which we call "preactive MS lesions," are closely associated with stressed oligodendrocytes and myelin sheaths that contain markedly elevated levels of the small stress protein alpha-B-crystallin (HspB5). Here, we show that microglia in these lesions express the recently identified receptors for HspB5, that is, CD14, Toll-like receptor family 1 and 2 (TLR1 and TLR2), and several molecular markers of the microglial response to HspB5. These markers were identified by genome-wide transcript profiling of 12 primary human microglial cultures at 2 time points after exposure to HspB5. These data indicate that HspB5 activates production by microglia of an array of chemokines, immune-regulatory mediators, and a striking number of antiviral genes that are generally inducible by type I interferons. Together, our data suggest that preactive MS lesions are at least in part driven by HspB5 derived from stressed oligodendrocytes and may reflect a local attempt to restore tissue homeostasis.


Subject(s)
Brain/drug effects , Microglia/drug effects , Multiple Sclerosis/metabolism , Nerve Fibers, Myelinated/drug effects , alpha-Crystallin B Chain/pharmacology , Aged , Aged, 80 and over , Axons/drug effects , Axons/metabolism , Axons/pathology , Brain/metabolism , Brain/pathology , Female , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Male , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , alpha-Crystallin B Chain/metabolism
17.
Biomaterials ; 34(3): 831-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23117214

ABSTRACT

As an extracellular protein, the small heat-shock protein alpha B-crystallin (HSPB5) has anti-inflammatory effects in several mouse models of inflammation. Here, we show that these effects are associated with the ability of HSPB5 to activate an immune-regulatory response in macrophages via endosomal/phagosomal CD14 and Toll-like receptors 1 and 2. Humans, however, possess natural antibodies against HSPB5 that block receptor binding. To protect it from these antibodies, we encapsulated HSPB5 in porous PLGA microparticles. We document here size, morphology, protein loading and release characteristics of such microparticles. Apart from effectively protecting HSPB5 from neutralization, PLGA microparticles also strongly promoted macrophage targeting of HSPB via phagocytosis. As a result, HSPB5 in porous PLGA microparticles was more than 100-fold more effective in activating macrophages than free soluble protein. Yet, the immune-regulatory nature of the macrophage response, as documented here by microarray transcript profiling, remained the same. In mice developing cigarette smoke-induced COPD, HSPB5-loaded PLGA microparticles were selectively taken up by alveolar macrophages upon intratracheal administration, and significantly suppressed lung infiltration by lymphocytes and neutrophils. In contrast, 30-fold higher doses of free soluble HSPB5 remained ineffective. Our data indicate that porous HSPB5-PLGA microparticles hold considerable promise as an anti-inflammatory biomaterial for humans.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Lung/drug effects , Macrophages/drug effects , Pneumonia/complications , Pneumonia/drug therapy , Pulmonary Disease, Chronic Obstructive/complications , alpha-Crystallin B Chain/administration & dosage , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/therapeutic use , Cell Line , Drug Carriers/chemistry , Heat-Shock Proteins, Small/administration & dosage , Heat-Shock Proteins, Small/immunology , Heat-Shock Proteins, Small/therapeutic use , Humans , Lactic Acid/chemistry , Lipopolysaccharide Receptors/immunology , Lung/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Pneumonia/immunology , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Toll-Like Receptor 1/immunology , Toll-Like Receptor 2/immunology , alpha-Crystallin B Chain/immunology , alpha-Crystallin B Chain/therapeutic use
18.
CNS Neurol Disord Drug Targets ; 11(5): 556-69, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22583440

ABSTRACT

Both immune-mediated and neurodegenerative processes play a role in the pathogenesis of multiple sclerosis (MS). There is still considerable debate, however, on how to link these two seemingly unrelated elements in disease. It has also remained unclear how the immune system can be involved without harboring any obvious myelin-directed abnormality in MS patients. Here, we propose that the unique properties of a small heat shock protein, HSPB5, can help reconcile the role of the immune system with the neurodegenerative element in MS, and explain the absence of any peripheral immune abnormality in patients. By being selectively induced as a protective stress protein in oligodendrocytes, and subsequently triggering activation of nearby microglia, HSPB5 accumulation translates neurodegenerative signals into a local innate immune response. The immune-regulatory profile of HSPB5-activated microglia, as well as animal model data, indicate that the HSPB5-induced innate response is neuroprotective. However, the presence of pro-inflammatory HSPB5-reactive memory T cells in the human immune repertoire, a unique feature among mammals, can subvert this response. Recruited by the innate response, such T cells respond to the accumulation of HSPB5 by an adaptive immune response, dominated by IFN-γ production, that ultimately overwhelms the originally protective microglial response, and culminates in tissue damage. Thus, HSPB5 accumulation caused by neurodegeneration can provoke a destructive local adaptive response of an otherwise normal immune system. This scenario is fully consistent with known causative factors and the pathology of MS, and with the effects of various therapies. It also helps explain why MS develops only in humans.


Subject(s)
Autoimmunity , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Nerve Degeneration/physiopathology , Adaptive Immunity , Animals , Autoantigens/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Progression , Humans , Immune Tolerance , Immunity, Innate , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Nerve Tissue Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , alpha-Crystallin B Chain/metabolism
19.
Int J Biochem Cell Biol ; 44(10): 1670-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22233974

ABSTRACT

There is now compelling evidence that members of the family of small heat shock proteins (HSP) can be secreted by a variety of different types of cells. Secretion of small HSP may at times represent altruistic delivery of supporting and stabilizing factors from one cell to another. A probably more general effect of extracellular small HSP, however, is exerted by their ability to activate macrophages and macrophage-like cells. When doing so, small HSP induce an immune-regulatory state of activation, stimulating macrophages to suppress inflammation. For this reason, small HSP deserve consideration as broadly applicable therapeutic agents for inflammatory disorders. In one particular case, however, adaptive immune responses to the small HSP itself may subvert the protective quality of the innate immune response it triggers. This situation only applies to alpha B-crystallin, and is unique for humans as well. In this special case, local concentrations of alpha B-crystallin determine the balance between protective innate responses and destructive adaptive responses, the latter of which are held responsible for the development of multiple sclerosis lesions. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.


Subject(s)
Heat-Shock Proteins, Small/physiology , Immunity, Innate , Inflammation Mediators/physiology , Multiple Sclerosis/metabolism , Adaptive Immunity , Animals , Exosomes/metabolism , Heat-Shock Proteins, Small/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/physiology , Multiple Sclerosis/immunology
20.
J Neurosci Res ; 90(2): 388-98, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21971760

ABSTRACT

Toll-like receptors (TLRs) play a key role in controlling innate immune responses to a wide variety of pathogen-associated molecules as well as endogenous signals. In addition, TLR expression within nonimmune cells has been recognized as as modulator of cell behavior. In this study we have addressed the question of whether functional TLRs are expressed on oligodendrocytes, the myelinating cells of the central nervous system. Primary cultures of rat oligodendrocytes at different maturation stages were found to express TLR2 and, to lesser extent, TLR3. Immunocytochemical analysis revealed that both TLRs were localized at the cell body and primary processes and were excluded from myelin-like membranes. Interestingly, innate immune receptor ligands were able to modulate oligodendrocyte survival, differentiation, and myelin-like membrane formation, indicating that TLRs on oligodendrocytes are functional. In highly purified oligodendrocytes cultures, the TLR2 agonist zymosan promoted survival, differentiation, and myelin-like membrane formation, whereas poly-I:C, a TLR3 ligand, was a potent inducer of apoptosis. Together, these data indicate that, in addition to other neural cell types, also oligodendrocytes express functional TLRs, which play a role in regulating various aspects of oligodendrocyte behavior.


Subject(s)
Cell Differentiation/physiology , Myelin Sheath/physiology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/physiology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/physiology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Rats , Rats, Wistar , Zymosan/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...