Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...