Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 144(8): 2361-2374, 2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-34145876

RESUMEN

Autoantibodies are a hallmark of numerous neurological disorders, including multiple sclerosis, autoimmune encephalitides and neuromyelitis optica. Whilst well understood in peripheral myeloid cells, the pathophysiological significance of autoantibody-induced Fc receptor signalling in microglia remains unknown, in part due to the lack of a robust in vivo model. Moreover, the application of therapeutic antibodies for neurodegenerative disease also highlights the importance of understanding Fc receptor signalling in microglia. Here, we describe a novel in vivo experimental paradigm that allows for selective engagement of Fc receptors within the CNS by peripherally injecting anti-myelin oligodendrocyte glycoprotein (MOG) monoclonal antibodies into normal wild-type mice. MOG antigen-bound immunoglobulins were detected throughout the CNS and triggered a rapid and tightly regulated proliferative response in both brain and spinal cord microglia. This microglial response was abrogated when anti-MOG antibodies were deprived of Fc receptor effector function or injected into Fcγ receptor knockout mice and was associated with the downregulation of Fc receptors in microglia, but not peripheral myeloid cells, establishing that this response was dependent on central Fc receptor engagement. Downstream of the Fc receptors, BTK was a required signalling node for this response, as microglia proliferation was amplified in BtkE41K knock-in mice expressing a constitutively active form of the enzyme and blunted in mice treated with a CNS-penetrant small molecule inhibitor of BTK. Finally, this response was associated with transient and stringently regulated changes in gene expression predominantly related to cellular proliferation, which markedly differed from transcriptional programs typically associated with Fc receptor engagement in peripheral myeloid cells. Together, these results establish a physiologically-meaningful functional response to Fc receptor and BTK signalling in microglia, while providing a novel in vivo tool to further dissect the roles of microglia-specific Fc receptor and BTK-driven responses to both pathogenic and therapeutic antibodies in CNS homeostasis and disease.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Autoanticuerpos/inmunología , Encéfalo/patología , Microglía/patología , Glicoproteína Mielina-Oligodendrócito/inmunología , Receptores Fc/metabolismo , Médula Espinal/patología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Proliferación Celular/fisiología , Ratones , Microglía/inmunología , Microglía/metabolismo , Médula Espinal/inmunología , Médula Espinal/metabolismo
2.
Immunity ; 34(2): 163-74, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21349429

RESUMEN

V(D)J gene segment recombination is linked to the cell cycle by the periodic phosphorylation and destruction of the RAG-2 protein at the G1-to-S cell cycle transition. To examine the function of this coupling, we constructed mice in which the phosphorylation site at threonine 490 of RAG-2 was mutated to alanine. The RAG-2(T490A) mutation uncoupled DNA cleavage from cell cycle and promoted aberrant recombination. Similar aberrant recombination products were observed in mice deficient in the Skp2 ubiquitin ligase subunit, which is required for periodic destruction of RAG-2. On a p53-deficient background, the RAG-2(T490A) mutation induced lymphoid malignancies characterized by clonal chromosomal translocations involving antigen receptor genes. Taken together, these observations provide a direct link between the periodic destruction of RAG-2 and lymphoid tumorigenesis. We infer that cell cycle control of the V(D)J recombinase limits the potential genomic damage that could otherwise result from RAG-mediated DNA cleavage.


Asunto(s)
Ciclo Celular , Proteínas de Unión al ADN/fisiología , Reordenamiento Génico , Inestabilidad Genómica , Linfoma no Hodgkin/genética , Receptores de Antígenos de Linfocitos T/genética , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Técnicas de Sustitución del Gen , Genes p53 , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Quinasas Asociadas a Fase-S/genética , Organismos Libres de Patógenos Específicos , Linfocitos T/metabolismo , Translocación Genética
3.
J Immunol ; 197(10): 3806-3819, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27815440

RESUMEN

Despite being one of the most common rheumatologic diseases, there is still no disease-modifying drug for primary Sjögren's syndrome (pSS). Advancing our knowledge of the target tissue has been limited by the low dimensionality of histology techniques and the small size of human salivary gland biopsies. In this study, we took advantage of a molecularly validated mouse model of pSS to characterize tissue-infiltrating CD4+ T cells and their regulation by the lymphotoxin/LIGHT signaling axis. Novel cell subsets were identified by combining highly dimensional flow and mass cytometry with transcriptomic analyses. Pharmacologic modulation of the LTßR signaling pathway was achieved by treating mice with LTßR-Ig, a therapeutic intervention currently being tested in pSS patients (Baminercept trial NCT01552681). Using these approaches, we identified two novel CD4+ T cell subsets characterized by high levels of PD1: Prdm1+ effector regulatory T cells expressing immunoregulatory factors, such as Il10, Areg, Fgl2, and Itgb8, and Il21+ effector conventional T cells expressing a pathogenic transcriptional signature. Mirroring these observations in mice, large numbers of CD4+PD1+ T cells were detected in salivary glands from Sjögren's patients but not in normal salivary glands or kidney biopsies from lupus nephritis patients. Unexpectedly, LTßR-Ig selectively halted the recruitment of PD1- naive, but not PD1+, effector T cells to the target tissue, leaving the cells with pathogenic potential unaffected. Altogether, this study revealed new cellular players in pSS pathogenesis, their transcriptional signatures, and differential dependency on the lymphotoxin/LIGHT signaling axis that help to interpret the negative results of the Baminercept trial and will guide future therapeutic interventions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-alfa/metabolismo , Glándulas Salivales/inmunología , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/fisiopatología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Anfirregulina/genética , Animales , Biopsia , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Interleucina-10/genética , Interleucinas/genética , Riñón/patología , Nefritis Lúpica/inmunología , Linfotoxina-alfa/genética , Ratones , Glándulas Salivales/patología , Transducción de Señal , Síndrome de Sjögren/terapia , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores
4.
J Am Soc Nephrol ; 28(6): 1741-1752, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28062569

RESUMEN

Ischemia-reperfusion injury (IRI) is a leading cause of AKI. This common clinical complication lacks effective therapies and can lead to the development of CKD. The αvß5 integrin may have an important role in acute injury, including septic shock and acute lung injury. To examine its function in AKI, we utilized a specific function-blocking antibody to inhibit αvß5 in a rat model of renal IRI. Pretreatment with this anti-αvß5 antibody significantly reduced serum creatinine levels, diminished renal damage detected by histopathologic evaluation, and decreased levels of injury biomarkers. Notably, therapeutic treatment with the αvß5 antibody 8 hours after IRI also provided protection from injury. Global gene expression profiling of post-ischemic kidneys showed that αvß5 inhibition affected established injury markers and induced pathway alterations previously shown to be protective. Intravital imaging of post-ischemic kidneys revealed reduced vascular leak with αvß5 antibody treatment. Immunostaining for αvß5 in the kidney detected evident expression in perivascular cells, with negligible expression in the endothelium. Studies in a three-dimensional microfluidics system identified a pericyte-dependent role for αvß5 in modulating vascular leak. Additional studies showed αvß5 functions in the adhesion and migration of kidney pericytes in vitro Initial studies monitoring renal blood flow after IRI did not find significant effects with αvß5 inhibition; however, future studies should explore the contribution of vasomotor effects. These studies identify a role for αvß5 in modulating injury-induced renal vascular leak, possibly through effects on pericyte adhesion and migration, and reveal αvß5 inhibition as a promising therapeutic strategy for AKI.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Riñón/irrigación sanguínea , Receptores de Vitronectina/antagonistas & inhibidores , Daño por Reperfusión/prevención & control , Animales , Masculino , Ratas , Ratas Sprague-Dawley
5.
J Allergy Clin Immunol ; 137(6): 1809-1821.e12, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27045581

RESUMEN

BACKGROUND: Mass cytometry has recently emerged as a promising tool for clinical research. However, few studies have demonstrated its benefit for patient stratification and biomarker identification. Primary Sjögren's syndrome (pSS) is a prototype of chronic autoimmune disease, the pathogenesis of which remains unclear and for which treatment does not exist. OBJECTIVE: This observational case-control study was designed to discover new cellular biomarkers and therapeutic targets in patients with pSS. METHODS: Forty-nine patients with pSS and 45 control subjects were enrolled for clinical evaluation and mass cytometry quantification of 34 protein markers in blood. For a third of these subjects, matched labial salivary gland biopsy specimens were also analyzed by mass cytometry and immunohistochemistry. RESULTS: In salivary gland biopsy specimens from patients with pSS, we identified a high number of activated CD8(+) T cells, terminally differentiated plasma cells, and activated epithelial cells, pointing to new pathogenic mechanisms for future clinical intervention. In blood, we identified a 6-cell disease signature defined by decreased numbers of CD4 and memory B lymphocytes, decreased plasmacytoid dendritic cell numbers, and increased representation of activated CD4 and CD8 T cells and plasmablasts. These blood cellular components correlated with clinical parameters and, when taken together, clustered patients into subsets with distinct disease activity and glandular inflammation. CONCLUSION: This first application of mass cytometry to a well-stratified clinical cohort and small biopsy tissues establishes the benefits of such an approach for the discovery of new biomarkers and therapeutic targets. Similar high-dimensional immunophenotyping strategies could be implemented in longitudinal and interventional clinical settings in this and other disease areas.


Asunto(s)
Biomarcadores , Inmunofenotipificación , Glándulas Salivales/metabolismo , Glándulas Salivales/patología , Síndrome de Sjögren/diagnóstico , Adulto , Biopsia , Linfocitos T CD8-positivos , Análisis por Conglomerados , Biología Computacional/métodos , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación/métodos , Masculino , Persona de Mediana Edad , Células Plasmáticas , Reproducibilidad de los Resultados , Glándulas Salivales Menores/metabolismo , Glándulas Salivales Menores/patología
6.
Clin Immunol ; 169: 69-79, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27352977

RESUMEN

B-cell activating factor (BAFF) levels are increased in rheumatoid arthritis, lupus and primary Sjögren's syndrome (pSS). However, BAFF contribution to pathogenesis is not completely understood. In pSS, immune infiltration of the salivary and lacrimal glands leads to xerostomia and xerophtalmia. Glandular B cell hyperactivation, differentiation into germinal center (GC)-like structures and plasma cell accumulation are histopathological hallmarks that were attributed to increased BAFF. Here, we experimentally tested this hypothesis by overexpressing BAFF in a mouse model of pSS. BAFF overexpression enhanced lymphocytic infiltration and MHCII expression on B cells. Increased BAFF also induced B cell differentiation into GC B cells within the autoimmune target tissue. However, even in these conditions, GC B cells only accounted for <1% of glandular B cells, demonstrating that BAFF is not efficiently promoting ectopic GC formation in pSS and warranting further investigation of therapeutics targeting both BAFF and the related TNF-family member APRIL.


Asunto(s)
Factor Activador de Células B/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Síndrome de Sjögren/inmunología , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Linfocitos B/metabolismo , Linfocitos B/patología , Diferenciación Celular/genética , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica/métodos , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inmunohistoquímica , Aparato Lagrimal/inmunología , Aparato Lagrimal/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Análisis de Secuencia por Matrices de Oligonucleótidos , Síndrome de Sjögren/genética , Síndrome de Sjögren/metabolismo , Xeroftalmia/genética , Xeroftalmia/inmunología , Xeroftalmia/metabolismo , Xerostomía/genética , Xerostomía/inmunología , Xerostomía/metabolismo
7.
Lupus Sci Med ; 8(1)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33658303

RESUMEN

OBJECTIVE: Punch biopsy, a standard diagnostic procedure for patients with cutaneous lupus erythematosus (CLE) carries an infection risk, is invasive, uncomfortable and potentially scarring, and impedes patient recruitment in clinical trials. Non-invasive tape sampling is an alternative that could enable serial evaluation of specific lesions. This cross-sectional pilot research study evaluated the use of a non-invasive adhesive tape device to collect messenger RNA (mRNA) from the skin surface of participants with CLE and healthy volunteers (HVs) and investigated its feasibility to detect biologically meaningful differences between samples collected from participants with CLE and samples from HVs. METHODS: Affected and unaffected skin tape samples and simultaneous punch biopsies were collected from 10 participants with CLE. Unaffected skin tape and punch biopsies were collected from 10 HVs. Paired samples were tested using quantitative PCR for a candidate immune gene panel and semi-quantitative immunohistochemistry for hallmark CLE proteins. RESULTS: mRNA collected using the tape device was of sufficient quality for amplification of 94 candidate immune genes. Among these, we found an interferon (IFN)-dominant gene cluster that differentiated CLE-affected from HV (23-fold change; p<0.001) and CLE-unaffected skin (sevenfold change; p=0.002), respectively. We found a CLE-associated gene cluster that differentiated CLE-affected from HV (fourfold change; p=0.005) and CLE-unaffected skin (fourfold change; p=0.012), respectively. Spearman's correlation between per cent area myxovirus 1 protein immunoreactivity and IFN-dominant mRNA gene cluster expression was highly significant (dermis, rho=0.86, p<0.001). In total, skin tape-derived RNA expression comprising both IFN-dominant and CLE-associated gene clusters correlated with per cent area immunoreactivity of some hallmark CLE-associated proteins in punch biopsies from the same lesions. CONCLUSIONS: A non-invasive tape RNA collection technique is a potential tool for repeated skin biomarker measures throughout a clinical trial.


Asunto(s)
Lupus Eritematoso Cutáneo , Estudios Transversales , Femenino , Voluntarios Sanos , Humanos , Masculino , ARN , Piel
8.
Sci Rep ; 11(1): 11154, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34045583

RESUMEN

Sjögren's syndrome (SS) is a complex autoimmune disease associated with lymphocytic infiltration and secretory dysfunction of salivary and lacrimal glands. Although the etiology of SS remains unclear, evidence suggests that epithelial damage of the glands elicits immune and fibrotic responses in SS. To define molecular changes underlying epithelial tissue damage in SS, we laser capture microdissected (LCM) labial salivary gland epithelia from 8 SS and 8 non-SS controls for analysis by RNA sequencing (RNAseq). Computational interrogation of gene expression signatures revealed that, in addition to a division of SS and non-SS samples, there was a potential intermediate state overlapping clustering of SS and non-SS samples. Differential expression analysis uncovered signaling events likely associated with distinct SS pathogenesis. Notable signals included the enrichment of IFN-γ and JAK/STAT-regulated genes, and the induction of genes encoding secreted factors, such as LTF, BMP3, and MMP7, implicated in immune responses, matrix remodeling and tissue destruction. Identification of gene expression signatures of salivary epithelia associated with mixed clinical and histopathological characteristics suggests that SS pathology may be defined by distinct molecular subtypes. We conclude that gene expression changes arising in the damaged salivary epithelia may offer novel insights into the signals contributing to SS development and progression.


Asunto(s)
Regulación de la Expresión Génica , Expresión Génica , Glándulas Salivales/metabolismo , Síndrome de Sjögren/genética , Adulto , Anciano , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Glándulas Salivales/patología , Transducción de Señal/fisiología , Síndrome de Sjögren/metabolismo , Síndrome de Sjögren/patología
9.
PLoS One ; 14(8): e0220125, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31408457

RESUMEN

BIN1 is the most important risk locus for Late Onset Alzheimer's Disease (LOAD), after ApoE. BIN1 AD-associated SNPs correlate with Tau deposition as well as with brain atrophy. Furthermore, the level of neuronal-specific BIN1 isoform 1 protein is decreased in sporadic AD cases in parallel with neuronal loss, despite an overall increase in BIN1 total mRNA. To address the relationship between reduction of BIN1 and neuronal cell loss in the context of Tau pathology, we knocked-down endogenous murine Bin1 via stereotaxic injection of AAV-Bin1 shRNA in the hippocampus of mice expressing Tau P301S (PS19). We observed a statistically significant reduction in the number of neurons in the hippocampus of mice injected with AAV-Bin1 shRNA in comparison with mice injected with AAV control. To investigate whether neuronal loss is due to deletion of Bin1 selectively in neurons in presence Tau P301S, we bred Bin1flox/flox with Thy1-Cre and subsequently with PS19 mice. Mice lacking neuronal Bin1 and expressing Tau P301S showed increased mortality, without increased neuropathology, when compared to neuronal Bin1 and Tau P301S-expressing mice. The loss of Bin1 isoform 1 resulted in reduced excitability in primary neurons in vitro, reduced neuronal c-fos expression as well as in altered microglia transcriptome in vivo. Taken together, our data suggest that the contribution of genetic variation in BIN1 locus to AD risk could result from a cell-autonomous reduction of neuronal excitability due to Bin1 decrease, exacerbated by the presence of aggregated Tau, coupled with a non-cell autonomous microglia activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Enfermedad de Alzheimer/patología , Biomarcadores/metabolismo , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Proteínas del Tejido Nervioso/fisiología , Neuronas/patología , Proteínas Supresoras de Tumor/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Conducta Animal , Encéfalo/metabolismo , Femenino , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Ratas , Proteínas tau/metabolismo
10.
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
11.
J Clin Invest ; 129(3): 1359-1371, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30645203

RESUMEN

BACKGROUND: Plasmacytoid DCs (pDC) produce large amounts of type I IFN (IFN-I), cytokines convincingly linked to systemic lupus erythematosus (SLE) pathogenesis. BIIB059 is a humanized mAb that binds blood DC antigen 2 (BDCA2), a pDC-specific receptor that inhibits the production of IFN-I and other inflammatory mediators when ligated. A first-in-human study was conducted to assess safety, tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) effects of single BIIB059 doses in healthy volunteers (HV) and patients with SLE with active cutaneous disease as well as proof of biological activity and preliminary clinical response in the SLE cohort. METHODS: A randomized, double-blind, placebo-controlled clinical trial was conducted in HV (n = 54) and patients with SLE (n = 12). All subjects were monitored for adverse events. Serum BIIB059 concentrations, BDCA2 levels on pDCs, and IFN-responsive biomarkers in whole blood and skin biopsies were measured. Skin disease activity was determined using the Cutaneous Lupus Erythematosus Disease Area and Severity Index Activity (CLASI-A). RESULTS: Single doses of BIIB059 were associated with favorable safety and PK profiles. BIIB059 administration led to BDCA2 internalization on pDCs, which correlated with circulating BIIB059 levels. BIIB059 administration in patients with SLE decreased expression of IFN response genes in blood, normalized MxA expression, reduced immune infiltrates in skin lesions, and decreased CLASI-A score. CONCLUSIONS: Single doses of BIIB059 were associated with favorable safety and PK/PD profiles and robust target engagement and biological activity, supporting further development of BIIB059 in SLE. The data suggest that targeting pDCs may be beneficial for patients with SLE, especially those with cutaneous manifestations. TRIAL REGISTRATION: ClinicalTrials.gov NCT02106897. FUNDING: Biogen Inc.


Asunto(s)
Anticuerpos Monoclonales , Lectinas Tipo C/antagonistas & inhibidores , Lupus Eritematoso Sistémico/tratamiento farmacológico , Glicoproteínas de Membrana/antagonistas & inhibidores , Receptores Inmunológicos/antagonistas & inhibidores , Enfermedades de la Piel/tratamiento farmacológico , Piel/inmunología , Adolescente , Adulto , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Células Dendríticas/inmunología , Células Dendríticas/patología , Método Doble Ciego , Femenino , Humanos , Lectinas Tipo C/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Masculino , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Receptores Inmunológicos/inmunología , Piel/patología , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/patología
12.
Sci Rep ; 9(1): 9477, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31263146

RESUMEN

Despite Bridging INtegrator 1 (BIN1) being the second most statistically-significant locus associated to Late Onset Alzheimer's Disease, its role in disease pathogenesis remains to be clarified. As reports suggest a link between BIN1, Tau and extracellular vesicles, we investigated whether BIN1 could affect Tau spreading via exosomes secretion. We observed that BIN1-associated Tau-containing extracellular vesicles purified from cerebrospinal fluid of AD-affected individuals are seeding-competent. We showed that BIN1 over-expression promotes the release of Tau via extracellular vesicles in vitro as well as exacerbation of Tau pathology in vivo in PS19 mice. Genetic deletion of Bin1 from microglia resulted in reduction of Tau secretion via extracellular vesicles in vitro, and in decrease of Tau spreading in vivo in male, but not female, mice, in the context of PS19 background. Interestingly, ablation of Bin1 in microglia of male mice resulted in significant reduction in the expression of heat-shock proteins, previously implicated in Tau proteostasis. These observations suggest that BIN1 could contribute to the progression of AD-related Tau pathology by altering Tau clearance and promoting release of Tau-enriched extracellular vesicles by microglia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Enfermedad de Alzheimer/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Proteínas tau/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Vesículas Extracelulares/genética , Vesículas Extracelulares/patología , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Proteostasis , Caracteres Sexuales , Proteínas Supresoras de Tumor/genética , Proteínas tau/genética
13.
Elife ; 62017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28492365

RESUMEN

Salivary glands, such as submandibular glands (SMGs), are composed of branched epithelial ductal networks that terminate in acini that together produce, transport and secrete saliva. Here, we show that the transcriptional regulator Yap, a key effector of the Hippo pathway, is required for the proper patterning and morphogenesis of SMG epithelium. Epithelial deletion of Yap in developing SMGs results in the loss of ductal structures, arising from reduced expression of the EGF family member Epiregulin, which we show is required for the expansion of Krt5/Krt14-positive ductal progenitors. We further show that epithelial deletion of the Lats1 and Lats2 genes, which encode kinases that restrict nuclear Yap localization, results in morphogenesis defects accompanied by an expansion of Krt5/Krt14-positive cells. Collectively, our data indicate that Yap-induced Epiregulin signaling promotes the identity of SMG ductal progenitors and that removal of nuclear Yap by Lats1/2-mediated signaling is critical for proper ductal maturation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Epirregulina/metabolismo , Epitelio/embriología , Morfogénesis , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Glándula Submandibular/embriología , Proteínas Supresoras de Tumor/metabolismo , Animales , Tipificación del Cuerpo , Proteínas de Ciclo Celular , Eliminación de Gen , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Células Madre/fisiología , Proteínas Supresoras de Tumor/genética , Proteínas Señalizadoras YAP
14.
PLoS One ; 11(9): e0162758, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27618690

RESUMEN

Systemic sclerosis (SSc) is a chronic autoimmune disorder that can result in extensive tissue damage in the skin and, in advanced cases, internal organs. Vasculopathy, aberrant immune activation, and tissue fibrosis are three hallmarks of the disease that have been identified, with vasculopathy and aberrant immunity being amongst the earliest events. However, a mechanistic link between these processes has not been established. Here, we have identified a novel role of platelet derived growth factor-BB (PDGF-BB)/PDGFRß activation in combination with dermal injury induced by bleomycin as a driver of early, aberrant expression of interferon stimulatory genes (ISGs) and inflammatory monocyte infiltration. Activation of PDGFRß in combination with bleomycin-induced dermal injury resulted in increased dermal thickness, vascular density, monocyte/macrophage infiltration, and exacerbation of tissue injury. Many of these features were dependent on IFNAR-signaling, and an increase in the number of interferon-beta (IFN-ß) producing monocytes cells was found in the skin lesions. Taken together, these results identify a novel link between PDGFRß activation, and Type I IFN-driven vascular maintenance and monocyte/macrophage cell recruitment, and provide a potential explanation linking key features of SSc that were previously thought to be unrelated.


Asunto(s)
Vasos Sanguíneos/patología , Modelos Animales de Enfermedad , Interferón Tipo I/fisiología , Monocitos/patología , Proteínas Proto-Oncogénicas c-sis/fisiología , Esclerodermia Sistémica/patología , Enfermedades de la Piel/patología , Animales , Becaplermina , Bleomicina/farmacología , Fibrosis , Ratones , Ratones Endogámicos C57BL , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
15.
PLoS One ; 11(10): e0164423, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27760209

RESUMEN

Mouse models lupus nephritis (LN) have provided important insights into disease pathogenesis, although none have been able to recapitulate all features of the human disease. Using comprehensive longitudinal analyses, we characterized a novel accelerated mouse model of lupus using pristane treatment in SNF1 (SWR X NZB F1) lupus prone mice (pristane-SNF1 mice). Pristane treatment in SNF1 mice accelerated the onset and progression of proteinuria, autoantibody production, immune complex deposition and development of renal lesions. At week 14, the pristane-SNF1 model recapitulated kidney disease parameters and molecular signatures seen in spontaneous disease in 36 week-old SNF1 mice and in a traditional IFNα-accelerated NZB X NZW F1 (BWF1) model. Blood transcriptome analysis revealed interferon, plasma cell, neutrophil, T-cell and protein synthesis signatures in the pristane-SNF1 model, all known to be present in the human disease. The pristane-SNF1 model appears to be particularly useful for preclinical research, robustly exhibiting many characteristics reminiscent of human disease. These include i) a stronger upregulation of the cytosolic nucleic acid sensing pathway, which is thought to be key component of the pathogenesis of the human disease, and ii) more prominent kidney interstitial inflammation and fibrosis, which have been both associated with poor prognosis in human LN. To our knowledge, this is the only accelerated model of LN that exhibits a robust tubulointerstitial inflammatory and fibrosis response. Taken together our data show that the pristane-SNF1 model is a novel accelerated model of LN with key features similar to human disease.


Asunto(s)
Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Nefritis Lúpica/patología , Terpenos/farmacología , Animales , Autoanticuerpos/biosíntesis , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Fibrosis , Glomerulonefritis/inducido químicamente , Glomerulonefritis/complicaciones , Humanos , Hipergammaglobulinemia/inducido químicamente , Hipergammaglobulinemia/complicaciones , Inflamación/inducido químicamente , Inflamación/complicaciones , Nefritis Lúpica/complicaciones , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
16.
J Wildl Dis ; 41(3): 654-8, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16244081

RESUMEN

A stranded 5-month-old female Pacific harbor seal (Phoca vitulina richardsi) was presented displaying tachypnea and diminished lung sounds. No neurological abnormalities were noted. The animal was treated for verminous pneumonia, but died 2 wk later. Gross necropsy examination revealed a severe obstructive verminous pneumonia associated with large numbers of Otostrongylus circumlitus. In addition, the majority of the right cerebral hemisphere was absent, with hypoplasia of the left cerebellar hemisphere, absence of the right pyramid, and malformation of the right occipital bone. Histopathologic findings included multifocal thrombosis and inflammation of pulmonary arteries, verminous pneumonia, and mild vacuolation of the subependymal white matter in the third ventricle representing swelling of myelin sheaths and edema. This is the first report of a hemicerebral anomaly in a marine mammal.


Asunto(s)
Encéfalo/anomalías , Phoca/anomalías , Animales , Encéfalo/patología , Resultado Fatal , Femenino , Hidrocefalia/etiología , Metastrongyloidea/aislamiento & purificación , Infecciones por Strongylida/diagnóstico , Infecciones por Strongylida/veterinaria
17.
Dis Aquat Organ ; 56(2): 95-104, 2003 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-14598985

RESUMEN

The results of an investigation of a new coral disease affecting Indo-Pacific reefs are presented. Porites ulcerative white spot disease (PUWS) is characterized by discrete, bleached, round foci, 3 to 5 mm in diameter, that may either regress or progress to full tissue-thickness ulcerations that coalesce, occasionally resulting in colony mortality. Monitoring of 25 diseased and 5 healthy reference colonies for 17 mo revealed that advanced stages of the disease were characterized by lesion coalescence, partial colony death (i.e. portions of the colony still alive; n = 17) and total colony death (n = 2). Field transmission experiments revealed that 95% of healthy colonies developed lesions within 5 wk after continual exposure to diseased branches, while 60% of the reference colonies remained healthy. The host range of PUWS includes branching and massive Porites spp., and prevalence per species was positively correlated with species density. On 10 reefs surveyed in the Central Philippines, 22 +/- 7% (mean +/- SE) of poritid colonies were infected, and the disease was present on 80% of the surveyed reefs. Poritids are dominant Indo-Pacific reef builders; a disease targeting this genus could cause major shifts in community structure over time. This report contributes to the limited knowledge of PUWS impacts in this region.


Asunto(s)
Antozoos/microbiología , Animales , Geografía , Océanos y Mares , Filipinas , Estaciones del Año
18.
Mol Cancer Res ; 11(10): 1223-34, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23858098

RESUMEN

UNLABELLED: The DNA damage response (DDR) factors ataxia telangiectasia mutated (ATM) and p53 binding protein 1 (53BP1) function as tumor suppressors in humans and mice, but the significance of their mutual interaction to the suppression of oncogenic translocations in vivo has not been investigated. To address this question, the phenotypes of compound mutant mice lacking 53BP1 and ATM (Trp53bp1(-/-)/Atm(-/-)), relative to single mutants, were examined. These analyses revealed that loss of 53BP1 markedly decreased the latency of T-lineage lymphomas driven by RAG-dependent oncogenic translocations in Atm(-/-) mice (average survival, 14 and 23 weeks for Trp53bp1(-/-)/Atm(-/-) and Atm(-/-) mice, respectively). Mechanistically, 53BP1 deficiency aggravated the deleterious effect of ATM deficiency on nonhomologous end-joining (NHEJ)-mediated double-strand break repair. Analysis of V(D)J recombinase-mediated coding joints and signal joints in Trp53bp1(-/-)/Atm(-/-) primary thymocytes is, however, consistent with canonical NHEJ-mediated repair. Together, these findings indicate that the greater NHEJ defect in the double mutant mice resulted from decreased efficiency of rejoining rather than switching to an alternative NHEJ-mediated repair mechanism. Complementary analyses of irradiated primary cells indicated that defects in cell-cycle checkpoints subsequently function to amplify the NHEJ defect, resulting in more frequent chromosomal breaks and translocations in double mutant cells throughout the cell cycle. Finally, it was determined that 53BP1 is dispensable for the formation of RAG-mediated hybrid joints in Atm(-/-) thymocytes but is required to suppress large deletions in a subset of hybrid joints. IMPLICATIONS: The current study uncovers novel ATM-independent functions for 53BP1 in the suppression of oncogenic translocations and in radioprotection.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Daño del ADN , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/fisiología , Rayos gamma , Estrés Fisiológico , Animales , Proteínas de la Ataxia Telangiectasia Mutada/deficiencia , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Células Cultivadas , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN por Unión de Extremidades/efectos de la radiación , Humanos , Linfoma/genética , Linfoma/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Estrés Fisiológico/genética , Timocitos/metabolismo , Timocitos/efectos de la radiación , Translocación Genética , Proteína 1 de Unión al Supresor Tumoral P53
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA