Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
J Card Fail ; 17(5): 359-65, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21549291

RESUMEN

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.


Asunto(s)
Cardiomiopatía Dilatada/inmunología , Interferón gamma/fisiología , Interleucina-10/fisiología , Leucocitos Mononucleares/inmunología , Troponina I/farmacología , Adulto , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/prevención & control , Estudios de Casos y Controles , Estudios de Cohortes , Estudios Transversales , Femenino , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Troponina I/fisiología
4.
Life Sci Alliance ; 2(6)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31792059

RESUMEN

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.


Asunto(s)
Envejecimiento Prematuro/genética , Receptor 1 de Quimiocinas CX3C/deficiencia , Microglía/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Perfil Genético , Inflamación/genética , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/metabolismo , Receptores de Quimiocina/deficiencia , Transducción de Señal , Transcriptoma
5.
Arch Neurol ; 65(3): 349-57, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18332247

RESUMEN

BACKGROUND: Interferon (IFN) gamma (IFNG) allelic variants are associated with susceptibility to multiple sclerosis (MS) in men but not in women. OBJECTIVES: To conduct a high-density linkage disequilibrium association study of IFNG and the surrounding region for sex-associated MS susceptibility bias and to evaluate whether IFNG allelic variants associated with MS susceptibility are associated with expression. DESIGN: Genotype case-control study, quantitative polymerase chain reaction (qPCR), and enzyme-linked immunosorbent assay expression analyses for IFN gamma. SETTING: Three independently ascertained populations from the Mayo Clinic, Rochester, Minnesota, Queen's University of Belfast, Belfast, Ireland, and University of Leuven, Leuven, Belgium. PATIENTS: For linkage disequilibrium, 861 patients with MS (293 men and 568 women) and 843 controls (340 men and 503 women) derived from the US (population-based) and the Northern Ireland and Belgium (clinic-based) cohorts were studied. For expression analyses, 50 US patients were selected to enrich for homozygotes and to achieve a balance between men and women. INTERVENTIONS: Twenty markers were genotyped over the 120-kilobase region harboring IFNG and the interleukin 26 gene (IL26). MAIN OUTCOME MEASURES: Expression of IFN gamma was evaluated by qPCR and enzyme-linked immunosorbent assay in stimulated peripheral blood mononuclear cells. RESULTS: Multiple markers were associated with MS susceptibility in men but not in women. The sex-specific susceptibility markers, of which rs2069727 was the strongest, were confined to IFNG. Carriers of rs2069727*G had higher expression than noncarriers. The effect of genotype in the qPCR experiments was also evident in men but not in women. CONCLUSIONS: IFNG is associated with sex bias in MS susceptibility and with expression of IFN gamma in MS. These observations add to a growing body of literature that implicates an interaction between sex and IFN gamma expression in a variety of disease states.


Asunto(s)
Expresión Génica/genética , Predisposición Genética a la Enfermedad , Interferón gamma/genética , Esclerosis Múltiple/genética , Polimorfismo de Nucleótido Simple/genética , Caracteres Sexuales , Alelos , Bélgica/epidemiología , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Frecuencia de los Genes , Humanos , Interferón gamma/metabolismo , Irlanda/epidemiología , Desequilibrio de Ligamiento , Masculino , Estados Unidos/epidemiología
6.
J Neuroimmunol ; 193(1-2): 161-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18022700

RESUMEN

Multiple sclerosis affects more women than men. The reasons for this are unknown. Previously, we have shown significant differences in women versus men in inflammatory cytokine responses to the major protein component of myelin, proteolipid protein (PLP), which is thought to be a target in MS patients. Here, using the ELISPOT assay, we examined sex differences in single-cell secretion of Th1 and Th2 cytokines from freshly isolated PBMC between relapsing remitting (RR) MS patients and healthy individuals. Cells were stimulated with MS-associated antigens including proteolipid protein (PLP), myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), and non-disease related antigens. Our data show a sex bias in the cytokine responses to multiple MS-relevant myelin antigens: Women with MS show IFNgamma-skewed responses and men with MS show IL-5-skewed responses. These data extend our previous findings [Pelfrey, C.M., Cotleur, A.C., Lee, J.C., Rudick, R.A. 2002. Sex differences in cytokine responses to myelin peptides in multiple sclerosis. J. Neuroimmunol. 130, 211-223.]: (1) by demonstrating gender skewing in cytokine responses to an expanded myelin antigen repertoire, which includes MBP, MOG and PLP; (2) by showing TNFalpha and IL-10 do not display comparable gender skewing compared to IFNgamma and IL5; (3) by defining the patient population as early, untreated RRMS patients to avoid confounding factors, such as different disease stages/disability and immunomodulatory therapy; and (4) by showing HLA type does not appear to underlie the gender differences. These findings may explain increased susceptibility to MS in women and could contribute to the differences in disease severity between men and women.


Asunto(s)
Citocinas/biosíntesis , Esclerosis Múltiple/inmunología , Proteínas de la Mielina/inmunología , Adulto , Femenino , Antígenos HLA-DR/análisis , Humanos , Interferón gamma/biosíntesis , Interleucina-5/biosíntesis , Masculino , Persona de Mediana Edad , Proteína Básica de Mielina/inmunología , Proteína Proteolipídica de la Mielina/inmunología , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Caracteres Sexuales , Células TH1/inmunología , Células Th2/inmunología
8.
Neurol Neuroimmunol Neuroinflamm ; 4(1): e311, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28018943

RESUMEN

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.

9.
Sci Transl Med ; 9(397)2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28679661

RESUMEN

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.


Asunto(s)
Autoanticuerpos/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Proteínas de Choque Térmico/inmunología , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/patología , Adulto , Albúminas/metabolismo , Animales , Acuaporina 4/metabolismo , Membrana Celular/metabolismo , Chaperón BiP del Retículo Endoplásmico , Células Endoteliales/patología , Fibrinógeno/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Ratones Endogámicos C57BL , Microvasos/patología , Neuromielitis Óptica/líquido cefalorraquídeo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología
10.
J Neuroimmunol ; 175(1-2): 192-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16644023

RESUMEN

Mitoxantrone (MX) has demonstrated efficacy in multiple sclerosis (MS), but its immunologic mechanisms of action are poorly understood. Furthermore, no study has examined the immunological effects of MX in primary progressive MS (PPMS). This study investigated the immunological effects of MX therapy in PPMS patients. Lymphocyte phenotypes and chemokine receptor (CCR) expression were evaluated by flow cytometry on fresh PBMC from 20 PPMS patients enrolled in a placebo (PLC)-controlled trial of MX. Longitudinal data were collected at weeks 0, 12, 24, 36 and short-term data (pre-/post-infusion) were collected at weeks 0 and 36. CXCR3, CCR1, CCR2 and CCR5 on CD4 and CD8 T cells and CD14 monocytes were evaluated. MX therapy induced a significant increase in the proportion of CD8 T cells (CD45RO(-)) over 9 months. Expression of several CCR increased following MX treatment. Two of the eight MX-treated patients demonstrated dramatic upregulation (70-76%) of CCR2 on monocytes. These two patients were the only MX-treated patients to demonstrate active inflammation by magnetic resonance imaging (MRI). PLC patients did not show significant changes in CCR expression. MX therapy was not associated with selective loss of CD4, CD8 or CD14 cells 1 month after treatment or over 9 months. These results suggest that MX therapy leads to a longitudinal increase in CD8 T cells and may increase CCR in patients with inflammation on MRI. Overall, MX did not show extensive immune changes in PPMS, although patients with active disease (gadolinium enhancing lesions) may identify a subset of PPMS subjects who respond immunologically to MX therapy. An improved understanding of MX may help identify markers of disease activity and response to therapy.


Asunto(s)
Mitoxantrona/uso terapéutico , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Esclerosis Múltiple Crónica Progresiva/inmunología , Adulto , Relación CD4-CD8 , Método Doble Ciego , Femenino , Humanos , Antígenos Comunes de Leucocito/biosíntesis , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Receptores CCR2 , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/inmunología
11.
J Neuroimmunol ; 167(1-2): 190-203, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16046001

RESUMEN

Sex hormones play a central role as modulators of immune responses and autoimmune diseases. We hypothesized that suppression of MS disease during pregnancy may be mediated by sex steroid hormones via regulation of costimulatory molecules such as CD40L or CD80/CD86 (B7-1/B7-2). We tested two sex hormones that are implicated in immune suppression during pregnancy: estriol and progesterone. We also examined whether this regulation is gender-specific or disease-related. PBMC from untreated relapsing remitting multiple sclerosis (RR MS) patients and controls were examined for expression of T cell and monocyte costimulatory molecules following mitogen stimulation in the presence or absence of sex hormones. In the absence of hormones, we confirmed that mitogen stimulation induced significantly more CD40L on the surface of CD4(+)T cells in MS patients compared to controls, and we extend these findings by showing there were no gender differences in induction of CD40L. Although supra-physiologic doses of hormones mildly suppressed CD40L expression on activated T cells, in vitro exposure to typical pregnancy-related physiologic doses of estriol or progesterone showed very little or no suppression of CD40L. On monocytes, neither estriol nor progesterone significantly altered the expression of CD80/CD86. These results suggest that physiologic doses of estriol or progesterone cannot alter CD40L on T cells or CD80/CD86 on monocytes sufficiently to explain the improvement observed in MS during pregnancy. Thus, although amelioration of MS and other autoimmune diseases during pregnancy is thought to be due to increased sex hormones, the present results do not support a role for suppression of costimulation via estriol or progesterone.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hormonas Esteroides Gonadales/farmacología , Esclerosis Múltiple/metabolismo , Adulto , Antígenos CD/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología , Factores Sexuales , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
12.
Neurol Neuroimmunol Neuroinflamm ; 2(6): e174, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26668819

RESUMEN

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.

13.
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
14.
J Neuroimmunol ; 141(1-2): 150-4, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12965266

RESUMEN

The expression of CCR5 and CXCR3, two chemokine receptors involved with homing of T cells to inflamed tissue, was examined longitudinally on CD4+ and CD8+ T cells in patients with a first demyelinating event of the central nervous system (CNS) randomized to receive i.m. injections of interferon-beta1a (IFN-beta1a) or placebo. Blood for analysis was collected before and 48 h after injection at baseline and after 3 and 12 months of treatment. The results showed that treatment with IFN-beta1a did not affect numbers of T cells expressing CCR5 and CXCR3 during the first 12 months of treatment, either at the peak of biological response or at the trough of interferon effect, at steady-state.


Asunto(s)
Regulación hacia Abajo , Interferón beta/administración & dosificación , Receptores CCR5/biosíntesis , Receptores de Quimiocina/biosíntesis , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Relación CD4-CD8/estadística & datos numéricos , Regulación hacia Abajo/inmunología , Esquema de Medicación , Estudios de Seguimiento , Humanos , Inyecciones Intramusculares , Interferón beta-1a , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Receptores CCR5/sangre , Receptores CXCR3 , Receptores de Quimiocina/sangre , Factores de Tiempo
15.
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
16.
J Neuroimmunol ; 141(1-2): 132-40, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12965264

RESUMEN

The relationship between autoreactivity to myelin antigens and disease progression in multiple sclerosis (MS) is not fully understood. We addressed this relationship by cross-sectionally comparing an objective measure of MS disability with immune cytokine responses to myelin proteins. The ELISPOT assay was used to determine the ex vivo interferon gamma (IFNgamma) and interleukin-10 (IL-10) production by peripheral blood mononuclear cells (PBMCs) in response to peptides spanning the entire proteolipid protein (PLP) and myelin basic protein (MBP) molecules in 20 patients with relapsing-remitting (RR) MS and 27 age- and sex-matched healthy controls. MS patients showed significantly higher MBP-induced IFNgamma responses and PLP-induced IL-10 responses compared with healthy controls. Using the Multiple Sclerosis Functional Composite (MSFC), a new multifactorial measure of disability, MS patients showed a significant correlation between the IFNgamma response to PLP peptides and MBP peptides, and disability. In contrast, in MS patients, there was no correlation between the MSFC and the response to unrelated control antigens or mitogens. These data show that myelin-specific T lymphocytes secreting the inflammatory cytokine IFNgamma correlate with functional impairment in MS, supporting an antigen-specific link between the immune response to myelin and disability in MS.


Asunto(s)
Evaluación de la Discapacidad , Interferón gamma/biosíntesis , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Esclerosis Múltiple Recurrente-Remitente/fisiopatología , Proteína Básica de Mielina/farmacología , Proteína Proteolipídica de la Mielina/farmacología , Fragmentos de Péptidos/farmacología , Adolescente , Adulto , Células Cultivadas , Enfermedad Crónica , Estudios Transversales , Citocinas/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-10/biosíntesis , Interleucina-10/metabolismo , Activación de Linfocitos/fisiología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Linfocitos T/metabolismo , Regulación hacia Arriba/fisiología
17.
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
18.
PLoS One ; 5(10): e13693, 2010 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21060874

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/metabolismo , Receptores CCR2/metabolismo , Animales , Sistema Nervioso Central/citología , Proteínas Luminiscentes/genética , Ratones , Receptores CCR2/genética , Proteína Fluorescente Roja
19.
Nat Neurosci ; 13(3): 319-26, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20154684

RESUMEN

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.


Asunto(s)
Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Inhibidores de la Monoaminooxidasa/toxicidad , Vaina de Mielina/efectos de los fármacos , Neutrófilos/fisiología , Receptores de Interleucina-8B/metabolismo , Animales , Quimera , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/fisiopatología , Cuerpo Calloso/ultraestructura , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Esclerosis Múltiple/fisiopatología , Proteínas de la Mielina/metabolismo , Vaina de Mielina/fisiología , Vaina de Mielina/ultraestructura , Neutrófilos/efectos de los fármacos , Neutrófilos/ultraestructura , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Oligodendroglía/ultraestructura , ARN Mensajero/metabolismo , Receptores de Interleucina-8B/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA