Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Immunol ; 14: 1130735, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033925

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental , Microglía , Humanos , Ratones , Animales , Microglía/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Diabetes Mellitus Experimental/patología , Inflamación/metabolismo , Retina/patología , Proteínas Portadoras/metabolismo , Fibrinógeno/metabolismo
2.
Methods Mol Biol ; 1936: 23-36, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30820891

RESUMEN

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.


Asunto(s)
Encéfalo/citología , Remielinización , Análisis de Secuencia de ARN/métodos , Animales , Separación Celular , Sistema Nervioso Central/química , Citometría de Flujo , Regulación de la Expresión Génica , Macrófagos/química , Macrófagos/citología , Ratones , Microglía/química , Microglía/citología
3.
PLoS One ; 10(7): e0133392, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26197437

RESUMEN

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.


Asunto(s)
Astrocitos/citología , Barrera Hematoencefálica/metabolismo , Células Endoteliales/citología , Lisofosfolípidos/metabolismo , Esclerosis Múltiple/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Adulto , Astrocitos/metabolismo , Movimiento Celular , Supervivencia Celular , Citocinas/metabolismo , Clorhidrato de Fingolimod/química , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Voluntarios Sanos , Humanos , Inflamación , Leucocitos/citología , Microcirculación , Persona de Mediana Edad , Transducción de Señal , Esfingolípidos/química , Esfingosina/metabolismo , Estrés Mecánico , Adulto Joven
4.
J Exp Med ; 212(3): 287-95, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25732305

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/patología , Macrófagos/metabolismo , Macrófagos/patología , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Factores de Edad , Anciano , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Antígenos Comunes de Leucocito/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Ratones Transgénicos , Receptores Inmunológicos/genética , Regulación hacia Arriba , Proteínas tau/metabolismo
5.
J Exp Med ; 211(8): 1533-49, 2014 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-25002752

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/patología , Inflamación/patología , Microglía/patología , Monocitos/patología , Animales , Receptor 1 de Quimiocinas CX3C , Forma de la Célula , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Homeostasis/genética , Humanos , Inflamación/genética , Cinética , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Microglía/ultraestructura , Monocitos/ultraestructura , Nódulos de Ranvier/patología , Receptores CCR2/metabolismo , Receptores de Quimiocina/metabolismo , Transducción de Señal/genética , Factores de Tiempo
6.
Neurocrit Care ; 21(2): 285-93, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24710655

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Líquido Cefalorraquídeo/metabolismo , Hierro/metabolismo , Hemorragia Subaracnoidea/metabolismo , Vasoespasmo Intracraneal/metabolismo , Anciano , Isquemia Encefálica/etiología , Ceruloplasmina/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Hemorragia Subaracnoidea/complicaciones , Vasoespasmo Intracraneal/etiología
7.
Genesis ; 51(8): 587-95, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23650205

RESUMEN

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.


Asunto(s)
Eliminación de Gen , Neutrófilos/metabolismo , Receptores de Interleucina-8B/genética , Animales , Movimiento Celular/genética , Ratones , Ratones Noqueados , Neutrófilos/fisiología , Receptores de Interleucina-8B/metabolismo
8.
Sci Transl Med ; 4(119): 119ra14, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22301555

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica/inmunología , Quimiocina CXCL12/metabolismo , Células Endoteliales/inmunología , Mediadores de Inflamación/metabolismo , Inflamación/inmunología , Linfocitos/inmunología , Monocitos/inmunología , Transducción de Señal , Migración Transendotelial y Transepitelial , Adulto , Anticuerpos Neutralizantes/farmacología , Antígenos CD19/metabolismo , Antígenos Transformadores de Poliomavirus/genética , Linfocitos B/inmunología , Barrera Hematoencefálica/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Humanos , Receptores de Lipopolisacáridos/metabolismo , Linfocitos/efectos de los fármacos , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Perfusión , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Flujo Sanguíneo Regional , Transducción de Señal/efectos de los fármacos , Virus 40 de los Simios/genética , Virus 40 de los Simios/inmunología , Estrés Mecánico , Factores de Tiempo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Transfección , Adulto Joven
9.
J Neuroimmunol ; 130(1-2): 211-23, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12225904

RESUMEN

Many autoimmune diseases preferentially affect women; however, the underlying mechanisms for the sex differences are poorly understood. We examined sex-dependent differences in the immunologic response to myelin proteins in 22 multiple sclerosis (MS) patients and 22 healthy controls. Using ELISA spot assay (ELISPOT) methodology, interferon (IFN) gamma and IL-5 secretions were examined at the single cell level in response to overlapping proteolipid protein (PLP) peptides. As previously reported, we observed an overall disease effect in the IFNgamma response, such that MS patients were significantly higher than controls. With respect to PLP-induced IFNgamma secretion, both MS and control females responded higher than their corresponding males. Female MS patients demonstrated the highest responses compared to MS males or healthy controls of either sex. Although MS females had high IFNgamma responses to PLP, they had no IL-5 responses at all, suggesting strong Th1 skewing. In contrast, MS males had more IL-5 than control males, who lacked IL-5 responses. These IL-5 responses suggested that disease and gender are not independent, but rather interact to influence the cytokine response to myelin. The data suggest a gender bias towards Th1 responses in MS, which may contribute to the female predominance in this disease.


Asunto(s)
Sistema Nervioso Central/inmunología , Citocinas/inmunología , Esclerosis Múltiple/inmunología , Proteínas de la Mielina/inmunología , Caracteres Sexuales , Linfocitos T/inmunología , Adulto , Células Cultivadas , Sistema Nervioso Central/fisiopatología , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-5/inmunología , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/genética , Esclerosis Múltiple/fisiopatología , Proteína Proteolipídica de la Mielina/inmunología , Péptidos/inmunología , Células TH1/citología , Células TH1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA