Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Am J Pathol ; 188(9): 2097-2108, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29935165

RESUMEN

Sepsis remains a major public health concern, characterized by marked immune dysfunction. Innate lymphoid cells develop from a common lymphoid precursor but have a role in orchestrating inflammation during innate response to infection. Here, we investigate the pathologic contribution of the group 2 innate lymphoid cells (ILC2s) in a murine model of acute septic shock (cecal ligation and puncture). Flow cytometric data revealed that ILC2s increase in number and percentage in the small intestine and in the peritoneal cells and inversely decline in the liver at 24 hours after septic insult. Sepsis also resulted in changes in ILC2 effector cytokine (IL-13) and activating cytokine (IL-33) in the plasma of mice and human patients in septic shock. Of interest, the sepsis-induced changes in cytokines were abrogated in mice deficient in functionally invariant natural killer T cells. Mice deficient in IL-13-producing cells, including ILC2s, had a survival advantage after sepsis along with decreased morphologic evidence of tissue injury and reduced IL-10 levels in the peritoneal fluid. Administration of a suppressor of tumorigenicity 2 (IL-33R) receptor-blocking antibody led to a transient survival advantage. Taken together, these findings suggest that ILC2s may play an unappreciated role in mediating the inflammatory response in both mice and humans; further, modulating ILC2 response in vivo may allow development of immunomodulatory strategies directed against sepsis.


Asunto(s)
Modelos Animales de Enfermedad , Inmunidad Innata/inmunología , Inflamación/inmunología , Hígado/inmunología , Linfocitos/inmunología , Sepsis/complicaciones , Animales , Estudios de Casos y Controles , Células Cultivadas , Citocinas/metabolismo , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Interleucina-33/inmunología , Masculino , Ratones , Células T Asesinas Naturales/inmunología , Sepsis/microbiología
2.
Proc Natl Acad Sci U S A ; 112(5): E440-9, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605905

RESUMEN

With the wide availability of massively parallel sequencing technologies, genetic mapping has become the rate limiting step in mammalian forward genetics. Here we introduce a method for real-time identification of N-ethyl-N-nitrosourea-induced mutations that cause phenotypes in mice. All mutations are identified by whole exome G1 progenitor sequencing and their zygosity is established in G2/G3 mice before phenotypic assessment. Quantitative and qualitative traits, including lethal effects, in single or multiple combined pedigrees are then analyzed with Linkage Analyzer, a software program that detects significant linkage between individual mutations and aberrant phenotypic scores and presents processed data as Manhattan plots. As multiple alleles of genes are acquired through mutagenesis, pooled "superpedigrees" are created to analyze the effects. Our method is distinguished from conventional forward genetic methods because it permits (1) unbiased declaration of mappable phenotypes, including those that are incompletely penetrant (2), automated identification of causative mutations concurrent with phenotypic screening, without the need to outcross mutant mice to another strain and backcross them, and (3) exclusion of genes not involved in phenotypes of interest. We validated our approach and Linkage Analyzer for the identification of 47 mutations in 45 previously known genes causative for adaptive immune phenotypes; our analysis also implicated 474 genes not previously associated with immune function. The method described here permits forward genetic analysis in mice, limited only by the rates of mutant production and screening.


Asunto(s)
Mutación Puntual , Alelos , Animales , Femenino , Genes Letales , Ligamiento Genético , Masculino , Ratones , Linaje , Fenotipo , Sitios de Carácter Cuantitativo
3.
Am J Physiol Gastrointest Liver Physiol ; 311(2): G237-45, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27288425

RESUMEN

Recent studies suggest that coinhibitory receptors appear to be important in contributing sepsis-induced immunosuppression. Our laboratory reported that mice deficient in programmed cell death receptor (PD)-1 have increased bacterial clearance and improved survival in experimental sepsis induced by cecal ligation and puncture (CLP). In response to infection, the liver clears the blood of bacteria and produces cytokines. Kupffer cells, the resident macrophages in the liver, are strategically situated to perform the above functions. However, it is not known if PD-1 expression on Kupffer cells is altered by septic stimuli, let alone if PD-1 ligation contributes to the altered microbial handling seen. Here we report that PD-1 is significantly upregulated on Kupffer cells during sepsis. PD-1-deficient septic mouse Kupffer cells displayed markedly enhanced phagocytosis and restoration of the expression of major histocompatibility complex II and CD86, but reduced CD80 expression compared with septic wild-type (WT) mouse Kupffer cells. In response to ex vivo LPS stimulation, the cytokine productive capacity of Kupffer cells derived from PD-1-/- CLP mice exhibited a marked, albeit partial, restoration of the release of IL-6, IL-12, IL-1ß, monocyte chemoattractant protein-1, and IL-10 compared with septic WT mouse Kupffer cells. In addition, PD-1 gene deficiency decreased LPS-induced apoptosis of septic Kupffer cells, as indicated by decreased levels of cleaved caspase-3 and reduced terminal deoxynucleotidyl transferase dUTP nick end-labeling-positive cells. Exploring the signal pathways involved, we found that, after ex vivo LPS stimulation, septic PD-1-/- mouse Kupffer cells exhibited an increased Akt phosphorylation and a reduced p38 phosphorylation compared with septic WT mouse Kupffer cells. Together, these results indicate that PD-1 appears to play an important role in regulating the development of Kupffer cell dysfunction seen in sepsis.


Asunto(s)
Coinfección/metabolismo , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Sepsis/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Células Cultivadas , Coinfección/genética , Coinfección/microbiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Macrófagos del Hígado/microbiología , Hígado/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Fenotipo , Fosforilación , Receptor de Muerte Celular Programada 1/deficiencia , Receptor de Muerte Celular Programada 1/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sepsis/genética , Sepsis/microbiología , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Am J Pathol ; 182(3): 742-54, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23306157

RESUMEN

Endothelial cell (EC) dysfunction is a key feature of multiple organ injury, the primary cause of fatality seen in critically ill patients. Although the development of EC dysfunction in the heart and lung is well studied in sepsis, it remains unclear in the liver. Herein, we report that liver sinusoidal ECs (LSECs; defined as CD146(+)CD45(-)) exhibit increased intercellular adhesion molecule-1 (CD54) and Fas in response to sepsis induced by cecal ligation and puncture (CLP). By using magnetically enriched LSEC (CD146(+)) populations, we show evidence of marked apoptosis, with a twofold decline in viable LSECs in CLP animals compared with sham controls. These changes and increased serum alanine aminotransferase levels were all mitigated in septic Fas(-/-) and Fas ligand(-/-) animals. Although we previously reported increased numbers of Fas ligand expressing CD8(+) T lymphocytes in the septic liver, CD8(+) T-cell deficiency did not reverse the onset of LSEC apoptosis/damage. However, Kupffer cell depletion with clodronate liposomes resulted in greater apoptosis and Fas expression after CLP and a decrease in glycoprotein 130 expression on LSECs, suggesting that STAT3 activation may protect these cells from injury. Our results document a critical role for death receptor-mediated LSEC injury and show the first evidence that Kupffer cells are essential to the viability of LSECs, which appears to be mediated through glycoprotein 130 expression in sepsis.


Asunto(s)
Apoptosis , Receptor gp130 de Citocinas/metabolismo , Regulación hacia Abajo , Células Endoteliales/patología , Macrófagos del Hígado/patología , Sepsis/patología , Receptor fas/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Ciego/patología , Separación Celular , Citoprotección , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Proteína Ligando Fas/deficiencia , Proteína Ligando Fas/metabolismo , Humanos , Macrófagos del Hígado/metabolismo , Ligadura , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Fenotipo , Punciones , Reproducibilidad de los Resultados , Sepsis/inmunología
5.
J Mol Biol ; 360(4): 760-73, 2006 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16793062

RESUMEN

Depletion of the transcriptional co-activator LEDGF/p75 by RNA interference alters the genome-wide pattern of HIV-1 integration, reducing integration into active genes, reducing integration into LEDGF/p75-regulated genes, and increasing integration into G+C-rich sequences. LEDGF/p75 is also able to act as a molecular tether linking HIV-1 integrase protein to chromatin, a phenomenon likely to underlie the integration site distribution effects. The LEDGF/p75 integrase-binding domain has been established but the domain or domains responsible for the chromatin-binding component of tethering are unknown. Here, we identify and characterize these domains. Complementary methods were used to assess condensed and uncondensed chromatin, and to determine the stringency of chromatin binding. Immuno-localization analyses revealed that an N-terminal PWWP domain and its beta-barrel substructure are needed for binding to metaphase chromatin. However, the PWWP domain is insufficient to transfer metaphase chromatin binding to green fluorescent protein, which requires addition of a downstream charged region (CR1). Biochemical analysis showed that full-length LEDGF/p75 resists Triton X-100 extraction from chromatin. To transfer Triton-resistant chromatin binding to green fluorescent protein, PWWP-CR1 is necessary but not sufficient. Further inclusion of a tandem pair of AT-hooks in combination with at least one of two identified downstream charged regions (CR2 or CR3) is needed. Deletion of just the PWWP or the AT-hook domain from full-length LEDGF/p75 reduced Triton-resistant chromatin binding, while deletion of both elements abolished it, underscoring their dominant and cooperative role. The results establish a molecular mechanism for LEDGF/p75-mediated tethering of HIV-1 integrase to chromatin.


Asunto(s)
Cromatina/metabolismo , Integrasa de VIH/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Aminoácidos/metabolismo , Células Cultivadas , Cromosomas Humanos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Señales de Localización Nuclear/genética , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Eliminación de Secuencia/genética
6.
Trends Mol Med ; 20(4): 224-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24485901

RESUMEN

Sepsis is the leading cause of death among critically ill patients in intensive care units, and treatment options are limited. Therapies developed against the proinflammatory stage have failed clinically; therefore, new approaches that target the host immune response in sepsis are necessary. Increasing evidence suggests that a major pathophysiological event in sepsis is immune suppression, often resulting in secondary fungal, bacterial, or viral infections. Recent studies from animal sepsis models and patient samples suggest that cytokines such as interleukin-7 (IL-7), IL-15, granulocyte macrophage colony-stimulating factor (GM-CSF), as well as co-inhibitory molecule blockade, such as anti-programmed cell death receptor-1 (anti-PD-1) and anti-B and T lymphocyte attenuator (anti-BTLA), may have utility in alleviating the clinical morbidity associated with sustained sepsis. This review discusses some of these novel immunomodulatory agents and evaluates their potential use as therapeutics.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Animales , Citocinas/inmunología , Citocinas/uso terapéutico , Humanos , Factores Inmunológicos/inmunología , Inmunomodulación/efectos de los fármacos
7.
J Leukoc Biol ; 94(5): 963-70, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23766529

RESUMEN

PD-1 and PD-L1 have been reported to provide peripheral tolerance by inhibiting TCR-mediated activation. We have reported that PD-L1-/- animals are protected from sepsis-induced mortality and immune suppression. Whereas studies indicate that LSECs normally express PD-L1, which is also thought to maintain local immune liver tolerance by ligating the receptor PD-1 on T lymphocytes, the role of PD-L1 in the septic liver remains unknown. Thus, we hypothesized initially that PD-L1 expression on LSECs protects them from sepsis-induced injury. We noted that the increased vascular permeability and pSTAT3 protein expression in whole liver from septic animals were attenuated in the absence of PD-L1. Isolated LSECs taken from septic animals, which exhibited increased cell death, declining cell numbers, reduced cellular proliferation, and VEGFR2 expression (an angiogenesis marker), also showed improved cell numbers, proliferation, and percent VEGFR2(+) levels in the absence of PD-L1. We also observed that sepsis induced an increase of liver F4/80(+)PD-1(+)-expressing KCs and increased PD-L1 expression on LSECs. Interestingly, PD-L1 expression levels on LSECs decreased when PD-1(+)-expressing KCs were depleted with clodronate liposomes. Contrary to our original hypothesis, we document here that increased interactions between PD-1(+) KCs and PD-L1(+) LSECs appear to lead to the decline of normal endothelial function-essential to sustain vascular integrity and prevent ALF. Importantly, we uncover an underappreciated pathological aspect of PD-1:PD-L1 ligation during inflammation that is independent of its normal, immune-suppressive activity.


Asunto(s)
Antígeno B7-H1/fisiología , Células Endoteliales/patología , Macrófagos del Hígado/fisiología , Hígado/patología , Sepsis/patología , Animales , Apoptosis , Permeabilidad Capilar , Proliferación Celular , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/metabolismo , Receptor fas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA