Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 144(2): 259-281, 2022 08.
Article in English | MEDLINE | ID: mdl-35666306

ABSTRACT

Oncostatin M (OSM) is an IL-6 family member which exerts neuroprotective and remyelination-promoting effects after damage to the central nervous system (CNS). However, the role of OSM in neuro-inflammation is poorly understood. Here, we investigated OSM's role in pathological events important for the neuro-inflammatory disorder multiple sclerosis (MS). We show that OSM receptor (OSMRß) expression is increased on circulating lymphocytes of MS patients, indicating their elevated responsiveness to OSM signalling. In addition, OSM production by activated myeloid cells and astrocytes is increased in MS brain lesions. In experimental autoimmune encephalomyelitis (EAE), a preclinical model of MS, OSMRß-deficient mice exhibit milder clinical symptoms, accompanied by diminished T helper 17 (Th17) cell infiltration into the CNS and reduced BBB leakage. In vitro, OSM reduces BBB integrity by downregulating the junctional molecules claudin-5 and VE-cadherin, while promoting secretion of the Th17-attracting chemokine CCL20 by inflamed BBB-endothelial cells and reactive astrocytes. Using flow cytometric fluorescence resonance energy transfer (FRET) quantification, we found that OSM-induced endothelial CCL20 promotes activation of lymphocyte function-associated antigen 1 (LFA-1) on Th17 cells. Moreover, CCL20 enhances Th17 cell adhesion to OSM-treated inflamed endothelial cells, which is at least in part ICAM-1 mediated. Together, these data identify an OSM-CCL20 axis, in which OSM contributes significantly to BBB impairment during neuro-inflammation by inducing permeability while recruiting Th17 cells via enhanced endothelial CCL20 secretion and integrin activation. Therefore, care should be taken when considering OSM as a therapeutic agent for treatment of neuro-inflammatory diseases such as MS.


Subject(s)
Blood-Brain Barrier , Encephalomyelitis, Autoimmune, Experimental , Oncostatin M , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oncostatin M/metabolism , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/biosynthesis , Oncostatin M Receptor beta Subunit/genetics , Th17 Cells/metabolism , Th17 Cells/pathology
2.
Proc Natl Acad Sci U S A ; 117(9): 5028-5038, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071226

ABSTRACT

The brain's endogenous capacity to restore damaged myelin deteriorates during the course of demyelinating disorders. Currently, no treatment options are available to establish remyelination. Chronic demyelination leads to damaged axons and irreversible destruction of the central nervous system (CNS). We identified two promising therapeutic candidates which enhance remyelination: oncostatin M (OSM), a member of the interleukin-6 family, and downstream mediator tissue inhibitor of metalloproteinases-1 (TIMP-1). While remyelination was completely abrogated in OSMRß knockout (KO) mice, OSM overexpression in the chronically demyelinated CNS established remyelination. Astrocytic TIMP-1 was demonstrated to play a pivotal role in OSM-mediated remyelination. Astrocyte-derived TIMP-1 drove differentiation of oligodendrocyte precursor cells into mature oligodendrocytes in vitro. In vivo, TIMP-1 deficiency completely abolished spontaneous remyelination, phenocopying OSMRß KO mice. Finally, TIMP-1 was expressed by human astrocytes in demyelinated multiple sclerosis lesions, confirming the human value of our findings. Taken together, OSM and its downstream mediator TIMP-1 have the therapeutic potential to boost remyelination in demyelinating disorders.


Subject(s)
Astrocytes/metabolism , Oncostatin M/metabolism , Remyelination/physiology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Astrocytes/pathology , Axons , Central Nervous System/metabolism , Central Nervous System/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Interleukin-6/metabolism , Mice , Mice, Knockout , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath , Oligodendrocyte Precursor Cells , Tissue Inhibitor of Metalloproteinase-1/genetics
3.
Ann N Y Acad Sci ; 1351: 52-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26100315

ABSTRACT

Multiple sclerosis (MS) is a chronic disabling autoimmune disease of the central nervous system. The interleukin (IL)-6 cytokine family plays a crucial role in regulating the immune response in MS. All members of the IL-6 family share the common signal-transducing receptor protein, glycoprotein 130. Although the intracellular signaling of these cytokines seems to be largely overlapping, they have diverse and contrasting effects on the immune response. This review focuses on the effects of the family members IL-6, leukemia inhibitory factor, oncostatin M, and IL-11 on immune cell subsets and how these effects relate to the pathogenesis of MS. Finally, we propose possible avenues to modulate these family members for future MS therapy.


Subject(s)
Central Nervous System/immunology , Immunomodulation/immunology , Interleukin-6/immunology , Multiple Sclerosis/immunology , Signal Transduction/immunology , Glycoproteins/metabolism , Humans , Interleukin-11/immunology , Leukemia Inhibitory Factor/metabolism , Oncostatin M/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology
4.
Glia ; 63(10): 1729-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25921393

ABSTRACT

Multiple sclerosis (MS) is a chronic disabling disease of the central nervous system (CNS), in which destruction of myelin sheaths leads to disturbed axonal conduction. Available MS therapies modulate the immune response, but are unable to prevent neurological decline. Therefore, great efforts are made to develop therapies that limit demyelination and axonal degeneration. Oncostatin M (OSM), a member of the interleukin (IL)-6 cytokine family, is produced in demyelinating lesions of MS patients and stimulates neuronal survival. In this study, we reveal that the OSM receptor (OSMR) was robustly upregulated on microglia/macrophages and astrocytes in the cuprizone-induced demyelination model. While OSMR deficiency led to aggravated demyelination, CNS-targeted OSM treatment largely prevented demyelination. OSM treatment increased IL-4 expression and induced polarization of myeloid cells towards an anti-inflammatory M2 phenotype in vivo. This study reveals a previously uncharacterized and protective role for OSM during demyelination, and indicates that OSM is a promising therapeutic candidate to limit CNS damage in demyelinating diseases including MS.


Subject(s)
Demyelinating Diseases/pathology , Demyelinating Diseases/prevention & control , Microglia/metabolism , Oncostatin M/pharmacology , Up-Regulation/physiology , Animals , Calcium-Binding Proteins/metabolism , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Chelating Agents/toxicity , Cuprizone/toxicity , Cytokines/genetics , Cytokines/metabolism , Demyelinating Diseases/chemically induced , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Growth Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/drug effects , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism , Phenotype , Time Factors , Transduction, Genetic , Up-Regulation/drug effects , Up-Regulation/genetics
5.
Brain Behav Immun ; 45: 180-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25514345

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS), for which current treatments are unable to prevent disease progression. Based on its neuroprotective and neuroregenerating properties, leukemia inhibitory factor (LIF), a member of the interleukin-6 (IL-6) cytokine family, is proposed as a novel candidate for MS therapy. However, its effect on the autoimmune response remains unclear. In this study, we determined how LIF modulates T cell responses that play a crucial role in the pathogenesis of MS. We demonstrate that expression of the LIF receptor was strongly increased on immune cells of MS patients. LIF treatment potently boosted the number of regulatory T cells (Tregs) in CD4(+) T cells isolated from healthy controls and MS patients with low serum levels of IL-6. Moreover, IL-6 signaling was reduced in the donors that responded to LIF treatment in vitro. Our data together with previous findings revealing that IL-6 inhibits Treg development, suggest an opposing function of LIF and IL-6. In a preclinical animal model of MS we shifted the LIF/IL-6 balance in favor of LIF by CNS-targeted overexpression. This increased the number of Tregs in the CNS during active autoimmune responses and reduced disease symptoms. In conclusion, our data show that LIF downregulates the autoimmune response by enhancing Treg numbers, providing further impetus for the use of LIF as a novel treatment for MS and other autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-6/immunology , Leukemia Inhibitory Factor Receptor alpha Subunit/immunology , Leukemia Inhibitory Factor/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Female , Humans , In Vitro Techniques , Interleukin-6/metabolism , Leukemia Inhibitory Factor/pharmacology , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Male , Mice , Middle Aged , T-Lymphocytes, Regulatory/drug effects
6.
Mol Neurobiol ; 50(3): 1142-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24996996

ABSTRACT

The family of interleukin (IL)-6 like cytokines plays an important role in the neuroinflammatory response to injury by regulating both neural as well as immune responses. Here, we show that expression of the IL-6 family member oncostatin M (OSM) and its receptor is upregulated after spinal cord injury (SCI). To reveal the relevance of increased OSM signaling in the pathophysiology of SCI, OSM was applied locally after spinal cord hemisection in mice. OSM treatment significantly improved locomotor recovery after mild and severe SCI. Improved recovery in OSM-treated mice was associated with a reduced lesion size. OSM significantly diminished astrogliosis and immune cell infiltration. Thus, OSM limits secondary damage after CNS trauma. In vitro viability assays demonstrated that OSM protects primary neurons in culture from cell death, suggesting that the underlying mechanism involves direct neuroprotective effects of OSM. Furthermore, OSM dose-dependently promoted neurite outgrowth in cultured neurons, indicating that the cytokine plays an additional role in CNS repair. Indeed, our in vivo experiments demonstrate that OSM treatment increases plasticity of serotonergic fibers after SCI. Together, our data show that OSM is produced at the lesion site, where it protects the CNS from further damage and promotes recovery.


Subject(s)
Neurites/drug effects , Neuroprotective Agents/pharmacology , Oncostatin M/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Dose-Response Relationship, Drug , Mice , Neurites/physiology , Neuroprotective Agents/therapeutic use , Oncostatin M/metabolism , Oncostatin M/therapeutic use , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Up-Regulation
7.
Mediators Inflamm ; 2013: 685317, 2013.
Article in English | MEDLINE | ID: mdl-23818742

ABSTRACT

Demyelination is one of the pathological hallmarks of multiple sclerosis (MS). To date, no therapy is available which directly potentiates endogenous remyelination. Interleukin-11 (IL-11), a member of the gp130 family of cytokines, is upregulated in MS lesions. Systemic IL-11 treatment was shown to ameliorate clinical symptoms in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. IL-11 modulates immune cells and protects oligodendrocytes in vitro. In this study, the cuprizone-induced demyelination mouse model was used to elucidate effects of IL-11 on de- and remyelination, independent of the immune response. Prophylactic-lentiviral- (LV-) mediated overexpression of IL-11 in mouse brain significantly limited acute demyelination, which was accompanied with the preservation of CC1(+) mature oligodendrocytes (OLs) and a decrease in microglial activation (Mac-2(+)). We further demonstrated that IL-11 directly reduces myelin phagocytosis in vitro. When IL-11 expressing LV was therapeutically applied in animals with extensive demyelination, a significant enhancement of remyelination was observed as demonstrated by Luxol Fast Blue staining and electron microscopy imaging. Our results indicate that IL-11 promotes maturation of NG2(+) OPCs into myelinating CC1(+) OLs and may thus explain the enhanced remyelination. Overall, we demonstrate that IL-11 is of therapeutic interest for MS and other demyelinating diseases by limiting demyelination and promoting remyelination.


Subject(s)
Central Nervous System/metabolism , Demyelinating Diseases/metabolism , Interleukin-11/metabolism , Animals , Cell Proliferation/drug effects , Central Nervous System/ultrastructure , Cuprizone/pharmacology , Demyelinating Diseases/drug therapy , Humans , Immunohistochemistry , Interleukin-11/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Microglia/ultrastructure , Microscopy, Electron, Transmission , Myelin Sheath/metabolism
8.
Toxicology ; 305: 49-64, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23357402

ABSTRACT

A generally accepted and validated laboratory model for smoking-associated pulmonary tumorigenesis would be useful for both basic and applied research applications, such as the development of early diagnostic endpoints or the evaluation of modified risk tobacco products, respectively. The A/J mouse is susceptible for developing both spontaneous and induced lung adenomas and adenocarcinomas, and increased lung tumor multiplicities were also observed in previous cigarette smoke inhalation studies. The present study was designed to collect data useful towards the validation of an 18-month mainstream smoke (MS) inhalation model. Male and female A/J mice were exposed whole-body at three MS concentration levels for 6h/day, and the results were compared to a previous study in the same laboratory and with a similar design. A linear MS concentration-dependent increase in lung tumorigenesis was observed with similar slopes for both sexes and both studies and a maximal 5-fold increase in multiplicity beyond sham control. The minimal detectable difference in lung tumor multiplicity for the current study was 37%. In the larynx, papillomas were detectable in all MS-exposed groups in a non-concentration dependent manner. No other extra-pulmonary MS-dependent neoplastic lesions were found. Gene expression signatures of lung tumor tissues allowed a clear differentiation of sham- and high dose MS-exposed mice. In combination with data from previous smoke inhalation studies with A/J mice, the current data suggest that this model for MS inhalation-induced pulmonary tumorigenesis is reliable and relevant, two crucial requirements towards validation of such a model.


Subject(s)
Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Smoking/adverse effects , Smoking/pathology , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endpoint Determination , Female , Gene Expression Regulation, Neoplastic/genetics , Inhalation Exposure , Male , Mice , Mice, Inbred A , Neoplasms/chemically induced , Neoplasms/pathology , Organ Size , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Reproducibility of Results , Respiratory System/pathology , Sex Characteristics , Tissue Fixation
9.
Toxicol Sci ; 131(2): 596-611, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23104432

ABSTRACT

Cigarette smoking is the leading cause of lung cancer and chronic obstructive pulmonary disease, yet there is little mechanistic information available in the literature. To improve this, laboratory models for cigarette mainstream smoke (MS) inhalation-induced chronic disease development are needed. The current study investigated the effects of exposing male A/J mice to MS (6h/day, 5 days/week at 150 and 300 mg total particulate matter per cubic meter) for 2.5, 5, 10, and 18 months in selected combinations with postinhalation periods of 0, 4, 8, and 13 months. Histopathological examination of step-serial sections of the lungs revealed nodular hyperplasia of the alveolar epithelium and bronchioloalveolar adenoma and adenocarcinoma. At 18 months, lung tumors were found to be enhanced concentration dependently (up to threefold beyond sham exposure), irrespective of whether MS inhalation had been performed for the complete study duration or was interrupted after 5 or 10 months and followed by postinhalation periods. Morphometric analysis revealed an increase in the extent of emphysematous changes after 5 months of MS inhalation, which did not significantly change over the following 13 months of study duration, irrespective of whether MS exposure was continued or not. These changes were found to be accompanied by a complex pattern of transient and sustained pulmonary inflammatory changes that may contribute to the observed pathogeneses. Data from this study suggest that the A/J mouse model holds considerable promise as a relevant model for investigating smoking-related emphysema and adenocarcinoma development.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Emphysema/chemically induced , Lung/drug effects , Nicotiana , Pneumonia/chemically induced , Smoke , Animals , Inhalation Exposure , Male , Mice
10.
Toxicology ; 275(1-3): 10-20, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20594951

ABSTRACT

Knowledge on mechanisms of smoking-induced tumorigenesis and on active smoke constituents may improve the development and evaluation of chemopreventive and therapeutic interventions, early diagnostic markers, and new and potentially reduced-risk tobacco products. A suitable laboratory animal disease model of mainstream cigarette smoke inhalation is needed for this purpose. In order to develop such a model, A/J and Swiss SWR/J mouse strains, with a genetic susceptibility to developing lung adenocarcinoma, were whole-body exposed to diluted cigarette mainstream smoke at 0, 120, and 240 mg total particulate matter per m(3) for 6h per day, 5 days per week. Mainstream smoke is the smoke actively inhaled by the smoker. For etiological reasons, parallel exposures to whole smoke fractions (enriched for particulate or gas/vapor phase) were performed at the higher concentration level. After 5 months of smoke inhalation and an additional 4-month post-inhalation period, both mouse strains responded similarly: no increase in lung tumor multiplicity was seen at the end of the inhalation period; however, there was a concentration-dependent tumorigenic response at the end of the post-inhalation period (up to 2-fold beyond control) in mice exposed to the whole smoke or the particulate phase. Tumors were characterized mainly as pulmonary adenomas. At the end of the inhalation period, epithelial hyperplasia, atrophy, and metaplasia were found in the nasal passages and larynx, and cellular and molecular markers of inflammation were found in the bronchoalveolar lavage fluid. These inflammatory effects were mostly resolved by the end of the post-inhalation period. In summary, these mouse strains responded to mainstream smoke inhalation with enhanced pulmonary adenoma formation. The major tumorigenic potency resided in the particulate phase, which is contrary to the findings published for environmental tobacco smoke surrogate inhalation in these mouse models.


Subject(s)
Gases/adverse effects , Inhalation Exposure/adverse effects , Lung Neoplasms/pathology , Particulate Matter/adverse effects , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects , Animals , Gases/chemistry , Lung Neoplasms/etiology , Male , Mice , Particulate Matter/chemistry , Volatilization
SELECTION OF CITATIONS
SEARCH DETAIL
...