Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Front Immunol ; 14: 1130735, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37033925

RESUMO

Diabetic retinopathy, a microvascular disease characterized by irreparable vascular damage, neurodegeneration and neuroinflammation, is a leading complication of diabetes mellitus. There is no cure for DR, and medical interventions marginally slow the progression of disease. Microglia-mediated inflammation in the diabetic retina is regulated via CX3CR1-FKN signaling, where FKN serves as a calming signal for microglial activation in several neuroinflammatory models. Polymorphic variants of CX3CR1, hCX3CR1I249/M280 , found in 25% of the human population, result in a receptor with lower binding affinity for FKN. Furthermore, disrupted CX3CR1-FKN signaling in CX3CR1-KO and FKN-KO mice leads to exacerbated microglial activation, robust neuronal cell loss and substantial vascular damage in the diabetic retina. Thus, studies to characterize the effects of hCX3CR1I249/M280 -expression in microglia-mediated inflammation in the diseased retina are relevant to identify mechanisms by which microglia contribute to disease progression. Our results show that hCX3CR1I249/M280 mice are significantly more susceptible to microgliosis and production of Cxcl10 and TNFα under acute inflammatory conditions. Inflammation is exacerbated under diabetic conditions and coincides with robust neuronal loss in comparison to CX3CR1-WT mice. Therefore, to further investigate the role of hCX3CR1I249/M280 -expression in microglial responses, we pharmacologically depleted microglia using PLX-5622, a CSF-1R antagonist. PLX-5622 treatment led to a robust (~70%) reduction in Iba1+ microglia in all non-diabetic and diabetic mice. CSF-1R antagonism in diabetic CX3CR1-WT prevented TUJ1+ axonal loss, angiogenesis and fibrinogen deposition. In contrast, PLX-5622 microglia depletion in CX3CR1-KO and hCX3CR1I249/M280 mice did not alleviate TUJ1+ axonal loss or angiogenesis. Interestingly, PLX-5622 treatment reduced fibrinogen deposition in CX3CR1-KO mice but not in hCX3CR1I249/M280 mice, suggesting that hCX3CR1I249/M280 expressing microglia influences vascular pathology differently compared to CX3CR1-KO microglia. Currently CX3CR1-KO mice are the most commonly used strain to investigate CX3CR1-FKN signaling effects on microglia-mediated inflammation and the results in this study indicate that hCX3CR1I249/M280 receptor variants may serve as a complementary model to study dysregulated CX3CR1-FKN signaling. In summary, the protective effects of microglia depletion is CX3CR1-dependent as microglia depletion in CX3CR1-KO and hCX3CR1I249/M280 mice did not alleviate retinal degeneration nor microglial morphological activation as observed in CX3CR1-WT mice.


Assuntos
Diabetes Mellitus Experimental , Microglia , Humanos , Camundongos , Animais , Microglia/metabolismo , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Diabetes Mellitus Experimental/patologia , Inflamação/metabolismo , Retina/patologia , Proteínas de Transporte/metabolismo , Fibrinogênio/metabolismo
2.
Life Sci Alliance ; 2(6)2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31792059

RESUMO

CX3CR1, one of the highest expressed genes in microglia in mice and humans, is implicated in numerous microglial functions. However, the molecular mechanisms underlying Cx3cr1 signaling are not well understood. Here, we analyzed transcriptomes of Cx3cr1-deficient microglia under varying conditions by RNA-sequencing (RNA-seq). In 2-mo-old mice, Cx3cr1 deletion resulted in the down-regulation of a subset of immune-related genes, without substantial epigenetic changes in markers of active chromatin. Surprisingly, Cx3cr1-deficient microglia from young mice exhibited a transcriptome consistent with that of aged Cx3cr1-sufficient animals, suggesting a premature aging transcriptomic signature. Immunohistochemical analysis of microglia in young and aged mice revealed that loss of Cx3cr1 modulates microglial morphology in a comparable fashion. Our results suggest that CX3CR1 may regulate microglial function in part by modulating the expression levels of a subset of inflammatory genes during chronological aging, making Cx3cr1-deficient mice useful for studying aged microglia.


Assuntos
Senilidade Prematura/genética , Receptor 1 de Quimiocina CX3C/deficiência , Microglia/metabolismo , Animais , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Modelos Animais de Doenças , Feminino , Deleção de Genes , Perfil Genético , Inflamação/genética , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios/metabolismo , Receptores de Quimiocinas/deficiência , Transdução de Sinais , Transcriptoma
3.
Methods Mol Biol ; 1936: 23-36, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30820891

RESUMO

Remyelination is the regenerative process whereby myelin sheaths are restored around axons following nervous system injury, allowing reinstatement of electrical impulse conduction, trophic/metabolic support, and axon health. Failure of remyelination in progressive multiple sclerosis is considered to contribute to axon loss, a correlate of clinical decline. Lack of approved pro-regenerative therapies for MS highlights the need to understand the cellular and molecular mechanisms underpinning successful remyelination. One approach is to conduct nonbiased gene expression analyses of cell types which regulate remyelination, such as microglia and monocyte-derived macrophages. Recent technological advances address the challenges of RNA sequencing of small tissue samples, thus allowing relatively small numbers of cells to be isolated from discrete lesions for analysis. Here, we present methods for FACS-based isolation of cells from focal remyelinating lesions of the adult mouse brain and subsequent RNA extraction for sequencing, using isolation of microglia/macrophages as an example.


Assuntos
Encéfalo/citologia , Remielinização , Análise de Sequência de RNA/métodos , Animais , Separação Celular , Sistema Nervoso Central/química , Citometria de Fluxo , Regulação da Expressão Gênica , Macrófagos/química , Macrófagos/citologia , Camundongos , Microglia/química , Microglia/citologia
4.
Sci Transl Med ; 9(397)2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28679661

RESUMO

Neuromyelitis optica (NMO) is an inflammatory disorder mediated by antibodies to aquaporin-4 (AQP4) with prominent blood-brain barrier (BBB) breakdown in the acute phase of the disease. Anti-AQP4 antibodies are produced mainly in the periphery, yet they target the astrocyte perivascular end feet behind the BBB. We reasoned that an endothelial cell-targeted autoantibody might promote BBB transit of AQP4 antibodies and facilitate NMO attacks. Using monoclonal recombinant antibodies (rAbs) from patients with NMO, we identified two that strongly bound to the brain microvascular endothelial cells (BMECs). Exposure of BMECs to these rAbs resulted in nuclear translocation of nuclear factor κB p65, decreased claudin-5 protein expression, and enhanced transit of macromolecules. Unbiased membrane proteomics identified glucose-regulated protein 78 (GRP78) as the rAb target. Using immobilized GRP78 to deplete GRP78 antibodies from pooled total immunoglobulin G (IgG) of 50 NMO patients (NMO-IgG) reduced the biological effect of NMO-IgG on BMECs. GRP78 was expressed on the surface of murine BMECs in vivo, and repeated administration of a GRP78-specific rAb caused extravasation of serum albumin, IgG, and fibrinogen into mouse brains. Our results identify GRP78 antibodies as a potential component of NMO pathogenesis and GRP78 as a candidate target for promoting central nervous system transit of therapeutic antibodies.


Assuntos
Autoanticorpos/metabolismo , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Proteínas de Choque Térmico/imunologia , Neuromielite Óptica/imunologia , Neuromielite Óptica/patologia , Adulto , Albuminas/metabolismo , Animais , Aquaporina 4/metabolismo , Membrana Celular/metabolismo , Chaperona BiP do Retículo Endoplasmático , Células Endoteliais/patologia , Fibrinogênio/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imunoglobulina G/metabolismo , Camundongos Endogâmicos C57BL , Microvasos/patologia , Neuromielite Óptica/líquido cefalorraquidiano , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia
6.
Neurol Neuroimmunol Neuroinflamm ; 4(1): e311, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28018943

RESUMO

OBJECTIVE: To address the hypothesis that physiologic interactions between astrocytes and endothelial cells (EC) at the blood-brain barrier (BBB) are afflicted by pathogenic inflammatory signaling when astrocytes are exposed to aquaporin-4 (AQP4) antibodies present in the immunoglobulin G (IgG) fraction of serum from patients with neuromyelitis optica (NMO), referred to as NMO-IgG. METHODS: We established static and flow-based in vitro BBB models incorporating co-cultures of conditionally immortalized human brain microvascular endothelial cells and human astrocyte cell lines with or without AQP4 expression. RESULTS: In astrocyte-EC co-cultures, exposure of astrocytes to NMO-IgG decreased barrier function, induced CCL2 and CXCL8 expression by EC, and promoted leukocyte migration under flow, contingent on astrocyte expression of AQP4. NMO-IgG selectively induced interleukin (IL)-6 production by AQP4-positive astrocytes. When EC were exposed to IL-6, we observed decreased barrier function, increased CCL2 and CXCL8 expression, and enhanced leukocyte transmigration under flow. These effects were reversed after application of IL-6 neutralizing antibody. CONCLUSIONS: Our results indicate that NMO-IgG induces IL-6 production by AQP4-positive astrocytes and that IL-6 signaling to EC decreases barrier function, increases chemokine production, and enhances leukocyte transmigration under flow.

7.
Neurol Neuroimmunol Neuroinflamm ; 2(6): e174, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26668819

RESUMO

BACKGROUND: Residual CXCR2 expression on CNS cells in Cxcr2 (+/-) →Cxcr2 (-/-) chimeric animals slowed remyelination after both experimental autoimmune encephalomyelitis and cuprizone-induced demyelination. METHODS: We generated Cxcr2 (fl/-) :PLPCre-ER(T) mice enabling an inducible, conditional deletion of Cxcr2 on oligodendrocyte lineage cells of the CNS. Cxcr2 (fl/-) :PLPCre-ER(T) mice were evaluated in 2 demyelination/remyelination models: cuprizone-feeding and in vitro lysophosphatidylcholine (LPC) treatment of cerebellar slice cultures. RESULTS: Cxcr2 (fl/-) :PLPCre-ER(T)(+) (termed Cxcr2-cKO) mice showed better myelin repair 4 days after LPC-induced demyelination of cerebellar slice cultures. Cxcr2-cKOs also displayed enhanced hippocampal remyelination after a 2-week recovery from 6-week cuprizone feeding. CONCLUSION: Using 2 independent demyelination/remyelination models, our data document enhanced myelin repair in Cxcr2-cKO mice, consistent with the data obtained from radiation chimerism studies of germline CXCR2. Further experiments are appropriate to explore how CXCR2 function in the oligodendrocyte lineage accelerates myelin repair.

8.
PLoS One ; 10(7): e0133392, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26197437

RESUMO

The ability of the Blood Brain Barrier (BBB) to maintain proper barrier functions, keeping an optimal environment for central nervous system (CNS) activity and regulating leukocytes' access, can be affected in CNS diseases. Endothelial cells and astrocytes are the principal BBB cellular constituents and their interaction is essential to maintain its function. Both endothelial cells and astrocytes express the receptors for the bioactive sphingolipid S1P. Fingolimod, an immune modulatory drug whose structure is similar to S1P, has been approved for treatment in multiple sclerosis (MS): fingolimod reduces the rate of MS relapses by preventing leukocyte egress from the lymph nodes. Here, we examined the ability of S1P and fingolimod to act on the BBB, using an in vitro co-culture model that allowed us to investigate the effects of S1P on endothelial cells, astrocytes, and interactions between the two. Acting selectively on endothelial cells, S1P receptor signaling reduced cell death induced by inflammatory cytokines. When acting on astrocytes, fingolimod treatment induced the release of a factor, granulocyte macrophage colony-stimulating factor (GM-CSF) that reduced the effects of cytokines on endothelium. In an in vitro BBB model incorporating shear stress, S1P receptor modulation reduced leukocyte migration across the endothelial barrier, indicating a novel mechanism that might contribute to fingolimod efficacy in MS treatment.


Assuntos
Astrócitos/citologia , Barreira Hematoencefálica/metabolismo , Células Endoteliais/citologia , Lisofosfolipídeos/metabolismo , Esclerose Múltipla/imunologia , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Adulto , Astrócitos/metabolismo , Movimento Celular , Sobrevivência Celular , Citocinas/metabolismo , Cloridrato de Fingolimode/química , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Voluntários Saudáveis , Humanos , Inflamação , Leucócitos/citologia , Microcirculação , Pessoa de Meia-Idade , Transdução de Sinais , Esfingolipídeos/química , Esfingosina/metabolismo , Estresse Mecânico , Adulto Jovem
9.
J Exp Med ; 212(3): 287-95, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25732305

RESUMO

Variants in triggering receptor expressed on myeloid cells 2 (TREM2) confer high risk for Alzheimer's disease (AD) and other neurodegenerative diseases. However, the cell types and mechanisms underlying TREM2's involvement in neurodegeneration remain to be established. Here, we report that TREM2 is up-regulated on myeloid cells surrounding amyloid deposits in AD mouse models and human AD tissue. TREM2 was detected on CD45(hi)Ly6C(+) myeloid cells, but not on P2RY12(+) parenchymal microglia. In AD mice deficient for TREM2, the CD45(hi)Ly6C(+) macrophages are virtually eliminated, resulting in reduced inflammation and ameliorated amyloid and tau pathologies. These data suggest a functionally important role for TREM2(+) macrophages in AD pathogenesis and an unexpected, detrimental role of TREM2 in AD pathology. These findings have direct implications for future development of TREM2-targeted therapeutics.


Assuntos
Doença de Alzheimer/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Fatores Etários , Idoso , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Camundongos Transgênicos , Receptores Imunológicos/genética , Regulação para Cima , Proteínas tau/metabolismo
10.
J Exp Med ; 211(8): 1533-49, 2014 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-25002752

RESUMO

In the human disorder multiple sclerosis (MS) and in the model experimental autoimmune encephalomyelitis (EAE), macrophages predominate in demyelinated areas and their numbers correlate to tissue damage. Macrophages may be derived from infiltrating monocytes or resident microglia, yet are indistinguishable by light microscopy and surface phenotype. It is axiomatic that T cell-mediated macrophage activation is critical for inflammatory demyelination in EAE, yet the precise details by which tissue injury takes place remain poorly understood. In the present study, we addressed the cellular basis of autoimmune demyelination by discriminating microglial versus monocyte origins of effector macrophages. Using serial block-face scanning electron microscopy (SBF-SEM), we show that monocyte-derived macrophages associate with nodes of Ranvier and initiate demyelination, whereas microglia appear to clear debris. Gene expression profiles confirm that monocyte-derived macrophages are highly phagocytic and inflammatory, whereas those arising from microglia demonstrate an unexpected signature of globally suppressed cellular metabolism at disease onset. Distinguishing tissue-resident macrophages from infiltrating monocytes will point toward new strategies to treat disease and promote repair in diverse inflammatory pathologies in varied organs.


Assuntos
Sistema Nervoso Central/patologia , Inflamação/patologia , Microglia/patologia , Monócitos/patologia , Animais , Receptor 1 de Quimiocina CX3C , Forma Celular , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Homeostase/genética , Humanos , Inflamação/genética , Cinética , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/ultraestrutura , Monócitos/ultraestrutura , Nós Neurofibrosos/patologia , Receptores CCR2/metabolismo , Receptores de Quimiocinas/metabolismo , Transdução de Sinais/genética , Fatores de Tempo
11.
J Neurosci Methods ; 232: 165-72, 2014 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-24858797

RESUMO

BACKGROUND: In vitro blood-brain barrier (BBB) models can be useful for understanding leukocyte-endothelial interactions at this unique vascular-tissue interface. Desirable features of such a model include shear stress, non-transformed cells and co-cultures of brain microvascular endothelial cells with astrocytes. Recovery of transmigrated leukocytes for further analysis is also appealing. NEW METHODS: We report an in vitro BBB model for leukocyte transmigration incorporating shear stress with co-culture of conditionally immortalized human endothelial cell line (hBMVEC) and human astrocyte cell line (hAST). Transmigrated leukocytes can be recovered for comparison with input and non-transmigrated cells. RESULT: hBMVEC and hAST exhibited physiological and morphological BBB properties when cocultured back-to-back on membranes. In particular, astrocyte processes protruded through 3 µm membrane pores, terminating in close proximity to the hBMVEC with a morphology reminiscent of end-feet. Co-culture with hAST also decreased the permeability of hBMVEC. In our model, astrocytes promoted transendothelial leukocyte transmigration. COMPARISON WITH EXISTING METHODS: This model offers the opportunity to evaluate whether BBB properties and leukocyte transmigration across cytokine-activated hBMVEC are influenced by human astrocytes. CONCLUSIONS: We present a BBB model for leukocyte transmigration incorporating shear stress with co-culture of hBMVEC and hAST. We demonstrate that hAST promoted leukocyte transmigration and also increased certain barrier functions of hBMVEC. This model provides reproducible assays for leukocyte transmigration with robust results, which will enable further defining the relationships among leukocytes and the cellular elements of the BBB.


Assuntos
Astrócitos/fisiologia , Barreira Hematoencefálica/fisiologia , Células Endoteliais/fisiologia , Estresse Mecânico , Adulto , Transporte Biológico , Barreira Hematoencefálica/ultraestrutura , Células Cultivadas , Claudina-5/metabolismo , Técnicas de Cocultura , Células Endoteliais/ultraestrutura , Feminino , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Microvasos/citologia , Pessoa de Meia-Idade , Modelos Biológicos , Ocludina/metabolismo , Temperatura , Migração Transendotelial e Transepitelial , Adulto Jovem , Proteína da Zônula de Oclusão-1/metabolismo , Fator de von Willebrand/metabolismo
12.
Neurocrit Care ; 21(2): 285-93, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24710655

RESUMO

INTRODUCTION: Iron-mediated oxidative damage has been implicated in the genesis of cerebral vasospasm in animal models of SAH. We sought to explore the relationship between levels of non-protein bound iron in cerebrospinal fluid and the development of brain injury in patients with aneurysmal SAH. METHODS: Patients admitted with aneurysmal subarachnoid hemorrhage to a Neurointensive care unit of an academic, tertiary medical center, with Hunt and Hess grades 2-4 requiring ventriculostomy insertion as part of their clinical management were included in this pilot study. Samples of cerebrospinal fluid (CSF) were obtained on days 1, 3, and 5. A fluorometric assay that relies on an oxidation sensitive probe was used to measure unbound iron, and levels of iron-handling proteins were measured by means of enzyme-linked immunosorbent assays. We prospectively collected and recorded demographic, clinical, and radiological data. RESULTS: A total of 12 patients were included in this analysis. Median Hunt and Hess score on admission was 3.5 (IQR: 1) and median modified Fisher scale score was 4 (IQR: 1). Seven of 12 patients (58 %) developed delayed cerebral ischemia (DCI). Day 5 non-transferrin bound iron (NTBI) (7.88 ± 1 vs. 3.58 ± 0.8, p = 0.02) and mean NTBI (7.39 ± 0.4 vs. 3.34 + 0.4 p = 0.03) were significantly higher in patients who developed DCI. Mean redox-active iron, as well as day 3 levels of redox-active iron correlated with development of angiographic vasospasm in logistic regression analysis (p = 0.02); while mean redox-active iron and lower levels of ceruloplasmin on days 3, 5, and peak concentration were correlated with development of deep cerebral infarcts. CONCLUSIONS: Our preliminary data indicate a causal relationship between unbound iron and brain injury following SAH and suggest a possible protective role for ceruloplasmin in this setting, particularly in the prevention of cerebral ischemia. Further studies are needed to validate these findings and to probe their clinical significance.


Assuntos
Isquemia Encefálica/metabolismo , Líquido Cefalorraquidiano/metabolismo , Ferro/metabolismo , Hemorragia Subaracnóidea/metabolismo , Vasoespasmo Intracraniano/metabolismo , Idoso , Isquemia Encefálica/etiologia , Ceruloplasmina/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/etiologia
13.
Genesis ; 51(8): 587-95, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23650205

RESUMO

Type 2 CXC chemokine receptor CXCR2 plays roles in development, tumorigenesis, and inflammation. CXCR2 also promotes demyelination and decreases remyelination by actions toward hematopoietic cells and nonhematopoietic cells. Germline CXCR2 deficient (Cxcr2(-/-) ) mice reported in 1994 revealed the complexity of CXCR2 function and its differential expression in varied cell-types. Here, we describe Cxcr2(fl/fl) mice for which the targeting construct was generated by recombineering based on homologous recombination in E. coli. Without recombination Cxcr2(fl/fl) mice have CXCR2 expression on neutrophils in peripheral blood, bone marrow and spleen. Cxcr2(fl/fl) mice were crossed to Mx-Cre mice in which Cre recombinase is induced by Type I interferons, elicited by injection with polyinosinic-polycytidylic acid (poly(I:C)). CXCR2-deficient neutrophils were observed in poly(I:C) treated Cxcr2(fl/fl) ::Mx-Cre(+) (Cxcr2-CKO) mice, but not in poly(I:C) treated Cxcr2(f//+) ::Mx-Cre(+) mice. CXCR2 deletion was mainly observed peripherally but not in the CNS. Cxcr2-CKO mice showed impaired neutrophil migration in sterile peritonitis. Cxcr2-CKO mice reported here will provide a genetic reagent to dissect roles of CXCR2 in the neutrophil granulocyte lineage. Furthermore Cxcr2(fl/fl) mice will provide useful genetic models to evaluate CXCR2 function in varied cell populations.


Assuntos
Deleção de Genes , Neutrófilos/metabolismo , Receptores de Interleucina-8B/genética , Animais , Movimento Celular/genética , Camundongos , Camundongos Knockout , Neutrófilos/fisiologia , Receptores de Interleucina-8B/metabolismo
14.
Sci Transl Med ; 4(119): 119ra14, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22301555

RESUMO

The accumulation of inflammatory cells in the brain parenchyma is a critical step in the pathogenesis of neuroinflammatory diseases such as multiple sclerosis (MS). Chemokines and adhesion molecules orchestrate leukocyte transmigration across the blood-brain barrier (BBB), but the dynamics of chemokine receptor expression during leukocyte transmigration are unclear. We describe an in vitro BBB model system using human brain microvascular endothelial cells that incorporates shear forces mimicking blood flow to elucidate how chemokine receptor expression is modulated during leukocyte transmigration. In the presence of the chemokine CXCL12, we examined modulation of its receptor CXCR4 on human T cells, B cells, and monocytes transmigrating across the BBB under flow conditions. CXCL12 stimulated transmigration of CD4(+) and CD8(+) T cells, CD19(+) B cells, and CD14(+) monocytes. Transmigration was blocked by CXCR4-neutralizing antibodies. Unexpectedly, CXCL12 selectively down-regulated CXCR4 on transmigrating monocytes, but not T cells. Monocytes underwent preferential CXCL12-mediated adhesion to the BBB in vitro compared with lymphocytes. These findings provide new insights into leukocyte-endothelial interactions at the BBB under conditions mimicking blood flow and suggest that in vitro BBB models may be useful for identifying chemokine receptors that could be modulated therapeutically to reduce neuroinflammation in diseases such as MS.


Assuntos
Barreira Hematoencefálica/imunologia , Quimiocina CXCL12/metabolismo , Células Endoteliais/imunologia , Mediadores da Inflamação/metabolismo , Inflamação/imunologia , Linfócitos/imunologia , Monócitos/imunologia , Transdução de Sinais , Migração Transendotelial e Transepitelial , Adulto , Anticorpos Neutralizantes/farmacologia , Antígenos CD19/metabolismo , Antígenos Transformantes de Poliomavirus/genética , Linfócitos B/imunologia , Barreira Hematoencefálica/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/efeitos dos fármacos , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Linfócitos/efeitos dos fármacos , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Perfusão , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Fluxo Sanguíneo Regional , Transdução de Sinais/efeitos dos fármacos , Vírus 40 dos Símios/genética , Vírus 40 dos Símios/imunologia , Estresse Mecânico , Fatores de Tempo , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Transfecção , Adulto Jovem
15.
Obstet Gynecol Int ; 2012: 612946, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21941558

RESUMO

The local route of stem cell administration utilized presently in clinical trials for stress incontinence may not take full advantage of the capabilities of these cells. The goal of this study was to evaluate if intravenously injected mesenchymal stem cells (MSCs) home to pelvic organs after simulated childbirth injury in a rat model. Female rats underwent either vaginal distension (VD) or sham VD. All rats received 2 million GFP-labeled MSCs intravenously 1 hour after injury. Four or 10 days later pelvic organs and muscles were imaged for visualization of GFP-positive cells. Significantly more MSCs home to the urethra, vagina, rectum, and levator ani muscle 4 days after VD than after sham VD. MSCs were present 10 days after injection but GFP intensity had decreased. This study provides basic science evidence that intravenous administration of MSCs could provide an effective route for cell-based therapy to facilitate repair after injury and treat stress incontinence.

16.
Clin Transl Sci ; 4(5): 317-22, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22029801

RESUMO

BACKGROUND: Approximately 20% of patients with idiopathic dilated cardiomyopathy (iDCM) have autoantibodies (AAbs) specific to cardiac troponin-I (cTnI). However, there has been no work evaluating active cellular autoimmunity. We aimed to identify a cTnI-stimulated cellular autoimmune response and to correlate our findings with cTnI AAb production. METHODS: Samples were obtained from stable ambulatory iDCM patients and healthy controls. Peripheral blood monocytes were incubated with cTnI, and cellular proliferation was measured using flow cytometry. AAbs against cTnI were detected by enzyme-linked immunosorbent assay. RESULTS: A positive cellular proliferative response to cTnI was identified in 20.5% (9/44) patients with iDCM and 5.7% (2/35) of healthy controls (p < 0.05). Positive cTnI AAbs were identified in 20% (7/35) of healthy controls and 13.6% (6/44) of patients with iDCM (p = NS). The presence of cTnI AAbs did not correlate with a positive cellular proliferative response. However, patients with iDCM who had an AAb response to cTnI were less likely to be taking a statin (p < 0.05). CONCLUSIONS: A cellular autoimmune response to cTnI is demonstrated in a subset of patients with iDCM. However, the presence of a cellular response did not correlate with the presence of AAbs to the same antigen.


Assuntos
Cardiomiopatia Dilatada/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Troponina I/farmacologia , Autoanticorpos/imunologia , Autoimunidade/efeitos dos fármacos , Cardiomiopatia Dilatada/imunologia , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Imunidade Humoral/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade
17.
J Card Fail ; 17(5): 359-65, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21549291

RESUMO

BACKGROUND: Autoimmune mechanisms, particularly through generation of autoantibodies, may contribute to the pathophysiology of idiopathic dilated cardiomyopathy (iDCM). The precise role of cellular autoimmune responses to cardiac-specific antigens has not been well described in humans. The purpose of this study was to characterize the cellular autoimmune response to cardiac troponin I (cTnI), specifically, the release of cytokines by peripheral blood mononuclear cells (PBMCs), in subjects with iDCM and healthy control subjects. METHODS AND RESULTS: We performed enzyme-linked immunospot assays on PBMCs isolated from subjects with iDCM and healthy control subjects to examine the ex vivo interferon-gamma (IFN-γ) and interleukin-10 (IL-10) production in response to cTnI exposure. Thirty-five consecutive subjects with iDCM (mean age 53 ± 11 years, 60% male, left ventricular ejection fraction 23 ± 7%) and 26 control subjects (mean age 46 ± 13 years, 46% male) were prospectively enrolled. IFN-γ production in response to cTnI did not differ between the groups (number of secreting cells 26 ± 49 vs 38 ± 53, respectively; P = .1). In contrast, subjects with iDCM showed significantly higher IL-10 responses to cTnI compared with control subjects (number of secreting cells 386 ± 428 vs 152 ± 162, respectively; P < .05). Among iDCM subjects, heightened IL-10 response to cTnI was associated with reduced systemic inflammation and lower prevalence of advanced diastolic dysfunction compared with those with normal IL-10 response to cTnI. CONCLUSIONS: Our preliminary findings suggest that a heightened cellular autoimmune IL-10 response to cTnI is detectable in a subset of patients with iDCM, which may be associated with reduced systemic levels of high-sensitivity C-reactive protein and lower prevalence of advanced diastolic dysfunction.


Assuntos
Cardiomiopatia Dilatada/imunologia , Interferon gama/fisiologia , Interleucina-10/fisiologia , Leucócitos Mononucleares/imunologia , Troponina I/farmacologia , Adulto , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/prevenção & controle , Estudos de Casos e Controles , Estudos de Coortes , Estudos Transversais , Feminino , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Estudos Prospectivos , Troponina I/fisiologia
18.
Glia ; 59(1): 108-18, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21046557

RESUMO

CXC chemokine receptor CXCR4 is expressed in vitro in both human and rodent adult neural progenitor cells (NPCs). It has been suggested that the CXCL12-CXCR4 axis potentially enhances the proliferation of NPCs. However, whether CXCR4 is expressed in the neural stem cells (NSCs), a subset of self-renewing and multipotent NPCs, and whether CXCR4 signaling is directly required for their proliferation are not clear. In this study, we report that CXCR4 is expressed in a subpopulation of NPCs in the early embryonic ventricular zone. In studies of a CXCR4(eGFP) bacterial artificial chromosomal (BAC) transgenic mouse line, we further isolated NPCs from E12.5 transgenic telencephalon and GFP(+) cells demonstrated self-renewal and multipotency in neurosphere assays in vitro. Consistent with these observations, we enriched GFP(+)/CXCR4(+) cells by fluorescence activated cell sorting (FACS) with either CXCR4 antibody 12G5 or GFP. Furthermore, we observed that CXCL12 alone did not activate the self-renewal of NPCs or increase the proliferation of NPCs that are induced by bFGF/EGF. However, we found that blocking CXCR4 receptor with antagonist AMD3100 impaired the bFGF/EGF-induced expansion of GFP(+) NPCs through modulating their cell cycling. In addition, AMD3100 treatment of pregnant mice reduced the generation of neurospheres from E12.5 embryos. Our data suggest that CXCR4 is a potential cell surface marker for early embryonic NSCs and modulates growth-factor signaling.


Assuntos
Ciclo Celular/fisiologia , Células-Tronco Multipotentes/metabolismo , Células-Tronco Neurais/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Citometria de Fluxo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos
19.
PLoS One ; 5(10): e13693, 2010 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-21060874

RESUMO

BACKGROUND: Monocyte subpopulations distinguished by differential expression of chemokine receptors CCR2 and CX3CR1 are difficult to track in vivo, partly due to lack of CCR2 reagents. METHODOLOGY/PRINCIPAL FINDINGS: We created CCR2-red fluorescent protein (RFP) knock-in mice and crossed them with CX3CR1-GFP mice to investigate monocyte subset trafficking. In mice with experimental autoimmune encephalomyelitis, CCR2 was critical for efficient intrathecal accumulation and localization of Ly6C(hi)/CCR2(hi) monocytes. Surprisingly, neutrophils, not Ly6C(lo) monocytes, largely replaced Ly6C(hi) cells in the central nervous system of these mice. CCR2-RFP expression allowed the first unequivocal distinction between infiltrating monocytes/macrophages from resident microglia. CONCLUSION/SIGNIFICANCE: These results refine the concept of monocyte subsets, provide mechanistic insight about monocyte entry into the central nervous system, and present a novel model for imaging and quantifying inflammatory myeloid populations.


Assuntos
Sistema Nervoso Central/metabolismo , Receptores CCR2/metabolismo , Animais , Sistema Nervoso Central/citologia , Proteínas Luminescentes/genética , Camundongos , Receptores CCR2/genética , Proteína Vermelha Fluorescente
20.
Nat Neurosci ; 13(3): 319-26, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20154684

RESUMO

Multiple sclerosis is an inflammatory demyelinating disorder of the CNS. Recent studies have suggested diverse mechanisms as underlying demyelination, including a subset of lesions induced by an interaction between metabolic insult to oligodendrocytes and inflammatory mediators. For mice of susceptible strains, cuprizone feeding results in oligodendrocyte cell loss and demyelination of the corpus callosum. Remyelination ensues and has been extensively studied. Cuprizone-induced demyelination remains incompletely characterized. We found that mice lacking the type 2 CXC chemokine receptor (CXCR2) were relatively resistant to cuprizone-induced demyelination and that circulating CXCR2-positive neutrophils were important for cuprizone-induced demyelination. Our findings support a two-hit process of cuprizone-induced demyelination, supporting the idea that multiple sclerosis pathogenesis features extensive oligodendrocyte cell loss. These data suggest that cuprizone-induced demyelination is useful for modeling certain aspects of multiple sclerosis pathogenesis.


Assuntos
Cuprizona/toxicidade , Doenças Desmielinizantes/induzido quimicamente , Inibidores da Monoaminoxidase/toxicidade , Bainha de Mielina/efeitos dos fármacos , Neutrófilos/fisiologia , Receptores de Interleucina-8B/metabolismo , Animais , Quimera , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/fisiopatologia , Corpo Caloso/ultraestrutura , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Esclerose Múltipla/fisiopatologia , Proteínas da Mielina/metabolismo , Bainha de Mielina/fisiologia , Bainha de Mielina/ultraestrutura , Neutrófilos/efeitos dos fármacos , Neutrófilos/ultraestrutura , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Oligodendroglia/ultraestrutura , RNA Mensageiro/metabolismo , Receptores de Interleucina-8B/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...