Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cureus ; 15(9): e45327, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37849592

RESUMEN

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are rare malignancies that arise from secretory endocrine cells of the gastroenteropancreatic system. Clinical outcomes have improved for patients with GEP-NETs due to the development and recent FDA approval of 177Lutetium DOTATATE. However, the response of brain metastases from GEP-NETs from 177Lutetium DOTATATE is unreported. We present the case of an 81-year-old man with low-grade small bowel GEP-NET with liver and brain metastases treated with a total of six cycles of 177Lutetium DOTATATE. With over three years of follow-up from his initial treatment, his brain metastases have had complete or partial responses, with no need for brain radiotherapy or radiosurgery.

2.
Immunol Cell Biol ; 101(1): 36-48, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36214093

RESUMEN

Type 1 diabetes (T1D) is caused by aberrant activation of autoreactive T cells specific for the islet beta cells. How islet-specific T cells evade tolerance to become effector T cells is unknown, but it is believed that an altered gut microbiota plays a role. Possible mechanisms include bystander activation of autoreactive T cells in the gut or "molecular mimicry" from cross-reactivity between gut microbiota-derived peptides and islet-derived epitopes. To investigate these mechanisms, we use two islet-specific CD8+ T cell clones and the non-obese diabetic mouse model of type 1 diabetes. Both insulin-specific G9C8 cells and IGRP-specific 8.3 cells underwent early activation and proliferation in the pancreatic draining lymph nodes but not in the Peyer's patches or mesenteric lymph nodes. Mutation of the endogenous epitope for G9C8 cells abolished their CD69 upregulation and proliferation, ruling out G9C8 cell activation by a gut microbiota derived peptide and molecular mimicry. However, previously activated islet-specific effector memory cells but not naïve cells migrated into the Peyer's patches where they increased their cytotoxic function. Oral delivery of butyrate, a microbiota derived anti-inflammatory metabolite, reduced IGRP-specific cytotoxic function. Thus, while initial activation of islet-specific CD8+ T cells occurred in the pancreatic lymph nodes, activated cells trafficked through the gut lymphoid tissues where they gained additional effector function via non-specific bystander activation influenced by the gut microbiota.


Asunto(s)
Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Ratones , Animales , Linfocitos T CD8-positivos , Diabetes Mellitus Tipo 1/genética , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Péptidos/metabolismo , Ganglios Linfáticos , Epítopos/metabolismo
3.
Semin Liver Dis ; 42(3): 233-249, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36001995

RESUMEN

Nonalcoholic liver disease is a component of metabolic syndrome associated with obesity, insulin resistance, and hyperlipidemia. Excessive alcohol consumption may accelerate the progression of steatosis, steatohepatitis, and fibrosis. While simple steatosis is considered a benign condition, nonalcoholic steatohepatitis with inflammation and fibrosis may progress to cirrhosis, liver failure, and hepatocellular cancer. Studies in rodent experimental models and primary cell cultures have demonstrated several common cellular and molecular mechanisms in the pathogenesis and regression of liver fibrosis. Chronic injury and death of hepatocytes cause the recruitment of myeloid cells, secretion of inflammatory and fibrogenic cytokines, and activation of myofibroblasts, resulting in liver fibrosis. In this review, we discuss the role of metabolically injured hepatocytes in the pathogenesis of nonalcoholic steatohepatitis and alcohol-associated liver disease. Specifically, the role of chemokine production and de novo lipogenesis in the development of steatotic hepatocytes and the pathways of steatosis regulation are discussed.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Cirrosis Hepática/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/complicaciones
4.
Exp Hematol ; 109: 1-10.e1, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35288231

RESUMEN

Acute myeloid leukemia (AML) is an aggressive blood malignancy characterized by the accumulation of immature blood cells that can severely impede the normal functions of the hematopoietic system. AML still has a poor 5-year survival rate of around 30%, and efforts to develop novel targeted therapies have been met with challenges. Allogeneic hematopoietic stem cell transplantation represents a potentially curative treatment for many AML patients. Donor immune cells, namely, T cells and NK cells, can help eliminate residual leukemia cells through the beneficial graft-versus-leukemia (GVL) effect. Nevertheless, malignant cells can still escape allogeneic immune surveillance and lead to disease relapse. Recent studies have provided insights into AML-specific immune evasion mechanisms, many of which are driven by epigenetic changes. This article describes epigenetic regulators as promising therapeutic targets for designing posttransplant maintenance therapies. Therefore, this review aims to summarize AML immune evasion mechanisms with a focus on the allogeneic immune environment. We discuss the roles of epigenetic regulators in driving immune escape and propose targeted strategies for preventing leukemia relapse. We then discuss the diverse immunomodulatory effects of epigenetic inhibitors and their potential to enhance the GVL effect. The current landscape of maintenance therapy trials with epigenetic inhibitors and their clinical prospects is also assessed.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Epigénesis Genética , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Recurrencia , Trasplante Homólogo
5.
Bioorg Med Chem Lett ; 30(13): 127197, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32331932

RESUMEN

A novel series of ethyl ketone based HDACs 1, 2, and 3 selective inhibitors have been identified with good enzymatic and cellular activity and high selectivity over HDACs 6 and 8. These inhibitors contain a spirobicyclic group in the amide region. Compound 13 stands out as a lead due to its good potency, high selectivity, and reasonable rat and dog PK. Compounds 33 and 34 show good potency and rat PK profiles as well.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Cetonas/farmacología , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Animales , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/farmacocinética , Línea Celular Tumoral , Perros , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacocinética , Humanos , Cetonas/síntesis química , Cetonas/farmacocinética , Pruebas de Sensibilidad Microbiana , Ratas , Compuestos de Espiro/síntesis química , Compuestos de Espiro/farmacocinética , Compuestos de Espiro/farmacología
6.
Am J Respir Crit Care Med ; 201(11): 1358-1371, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32105156

RESUMEN

Rationale: Respiratory syncytial virus (RSV) bronchiolitis causes significant infant mortality. Bronchiolitis is characterized by airway epithelial cell (AEC) death; however, the mode of death remains unknown.Objectives: To determine whether necroptosis contributes to RSV bronchiolitis pathogenesis via HMGB1 (high mobility group box 1) release.Methods: Nasopharyngeal samples were collected from children presenting to the hospital with acute respiratory infection. Primary human AECs and neonatal mice were inoculated with RSV and murine Pneumovirus, respectively. Necroptosis was determined via viability assays and immunohistochemistry for RIPK1 (receptor-interacting protein kinase-1), MLKL (mixed lineage kinase domain-like pseudokinase) protein, and caspase-3. Necroptosis was blocked using pharmacological inhibitors and RIPK1 kinase-dead knockin mice.Measurements and Main Results: HMGB1 levels were elevated in nasopharyngeal samples of children with acute RSV infection. RSV-induced epithelial cell death was associated with increased phosphorylated RIPK1 and phosphorylated MLKL but not active caspase-3 expression. Inhibition of RIPK1 or MLKL attenuated RSV-induced HMGB1 translocation and release, and lowered viral load. MLKL inhibition increased active caspase-3 expression in a caspase-8/9-dependent manner. In susceptible mice, Pneumovirus infection upregulated RIPK1 and MLKL expression in the airway epithelium at 8 to 10 days after infection, coinciding with AEC sloughing, HMGB1 release, and neutrophilic inflammation. Genetic or pharmacological inhibition of RIPK1 or MLKL attenuated these pathologies, lowered viral load, and prevented type 2 inflammation and airway remodeling. Necroptosis inhibition in early life ameliorated asthma progression induced by viral or allergen challenge in later life.Conclusions: Pneumovirus infection induces AEC necroptosis. Inhibition of necroptosis may be a viable strategy to limit the severity of viral bronchiolitis and break its nexus with asthma.


Asunto(s)
Bronquiolitis/virología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteína HMGB1/metabolismo , Necroptosis , Mucosa Respiratoria/citología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Animales , Preescolar , Humanos , Lactante , Ratones , Estudios Prospectivos
7.
Sci Transl Med ; 10(440)2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29743346

RESUMEN

Prostaglandin D2 (PGD2) signals through PGD2 receptor 2 (DP2, also known as CRTH2) on type 2 effector cells to promote asthma pathogenesis; however, little is known about its role during respiratory syncytial virus (RSV) bronchiolitis, a major risk factor for asthma development. We show that RSV infection up-regulated hematopoietic prostaglandin D synthase expression and increased PGD2 release by cultured human primary airway epithelial cells (AECs). Moreover, PGD2 production was elevated in nasopharyngeal samples from young infants hospitalized with RSV bronchiolitis compared to healthy controls. In a neonatal mouse model of severe viral bronchiolitis, DP2 antagonism decreased viral load, immunopathology, and morbidity and ablated the predisposition for subsequent asthma onset in later life. This protective response was abolished upon dual DP1/DP2 antagonism and replicated with a specific DP1 agonist. Rather than mediating an effect via type 2 inflammation, the beneficial effects of DP2 blockade or DP1 agonism were associated with increased interferon-λ (IFN-λ) [interleukin-28A/B (IL-28A/B)] expression and were lost upon IL-28A neutralization. In RSV-infected AEC cultures, DP1 activation up-regulated IFN-λ production, which, in turn, increased IFN-stimulated gene expression, accelerating viral clearance. Our findings suggest that DP2 antagonists or DP1 agonists may be useful antivirals for the treatment of viral bronchiolitis and possibly as primary preventatives for asthma.


Asunto(s)
Bronquiolitis Viral/metabolismo , Bronquiolitis Viral/patología , Interferón gamma/biosíntesis , Prostaglandina D2/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Alérgenos , Animales , Animales Recién Nacidos , Antivirales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Humanos , Inmunidad , Lactante , Inflamación/patología , Inflamación/virología , Oxidorreductasas Intramoleculares/metabolismo , Pulmón/patología , Pulmón/virología , Ratones Endogámicos BALB C , Virus de la Neumonía Murina , Receptores Inmunológicos/antagonistas & inhibidores , Receptores de Prostaglandina/antagonistas & inhibidores , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Regulación hacia Arriba
8.
J Allergy Clin Immunol ; 141(5): 1607-1619.e9, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28947081

RESUMEN

BACKGROUND: Rhinovirus infection triggers acute asthma exacerbations. IL-33 is an instructive cytokine of type 2 inflammation whose expression is associated with viral load during experimental rhinovirus infection of asthmatic patients. OBJECTIVE: We sought to determine whether anti-IL-33 therapy is effective during disease progression, established disease, or viral exacerbation using a preclinical model of chronic asthma and in vitro human primary airway epithelial cells (AECs). METHODS: Mice were exposed to pneumonia virus of mice and cockroach extract in early and later life and then challenged with rhinovirus to model disease onset, progression, and chronicity. Interventions included anti-IL-33 or dexamethasone at various stages of disease. AECs were obtained from asthmatic patients and healthy subjects and treated with anti-IL-33 after rhinovirus infection. RESULTS: Anti-IL-33 decreased type 2 inflammation in all phases of disease; however, the ability to prevent airway smooth muscle growth was lost after the progression phase. After the chronic phase, IL-33 levels were persistently high, and rhinovirus challenge exacerbated the type 2 inflammatory response. Treatment with anti-IL-33 or dexamethasone diminished exacerbation severity, and anti-IL-33, but not dexamethasone, promoted antiviral interferon expression and decreased viral load. Rhinovirus replication was higher and IFN-λ levels were lower in AECs from asthmatic patients compared with those from healthy subjects. Anti-IL-33 decreased rhinovirus replication and increased IFN-λ levels at the gene and protein levels. CONCLUSION: Anti-IL-33 or dexamethasone suppressed the magnitude of type 2 inflammation during a rhinovirus-induced acute exacerbation; however, only anti-IL-33 boosted antiviral immunity and decreased viral replication. The latter phenotype was replicated in rhinovirus-infected human AECs, suggesting that anti-IL-33 therapy has the additional benefit of enhancing host defense.


Asunto(s)
Antivirales/farmacología , Asma/tratamiento farmacológico , Asma/inmunología , Inflamación/inmunología , Interleucina-33/inmunología , Virus de la Neumonía Murina/efectos de los fármacos , Virus de la Neumonía Murina/inmunología , Animales , Antivirales/inmunología , Asma/virología , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/virología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Inflamación/tratamiento farmacológico , Inflamación/virología , Ratones , Ratones Endogámicos BALB C , Infecciones por Pneumovirus/tratamiento farmacológico , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/virología , Carga Viral/efectos de los fármacos , Carga Viral/inmunología
9.
Sci Rep ; 7(1): 2353, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28539639

RESUMEN

Respiratory syncytial virus (RSV)-bronchiolitis is a major cause of infant morbidity and mortality and a risk factor for subsequent asthma. We showed previously that toll-like receptor (TLR)7 in plasmacytoid dendritic cells (pDCs) is critical for protection against bronchiolitis and asthma in mice infected with pneumonia virus of mice (PVM), the mouse homolog of RSV. This lack of redundancy was unexpected as interferon-ß promotor stimulator-1 (IPS-1) signalling, downstream of RIG-I-like receptor (RLR) and not TLR7 activation, contributes to host defence in hRSV-inoculated adult mice. To further clarify the role of IPS-1 signalling, we inoculated IPS-1-/- and WT mice with PVM in early-life, and again in later-life, to model the association between bronchiolitis and asthma. IPS-1 deficiency predisposed to severe PVM bronchiolitis, characterised by neutrophilic inflammation and necroptotic airway epithelial cell death, high mobility group box 1 (HMGB1) and IL-33 release, and downstream type-2 inflammation. Secondary infection induced an eosinophilic asthma-like pathophysiology in IPS-1-/- but not WT mice. Mechanistically, we identified that IPS-1 is necessary for pDC recruitment, IFN-α production and viral control. Our findings suggest that TLR7 and RLR signalling work collaboratively to optimally control the host response to pneumovirus infection thereby protecting against viral bronchiolitis and subsequent asthma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Asma/metabolismo , Bronquiolitis/metabolismo , Virus de la Neumonía Murina/fisiología , Infecciones por Pneumovirus/virología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Asma/genética , Bronquiolitis/genética , Proteína 58 DEAD Box/metabolismo , Células Dendríticas/metabolismo , Interacciones Huésped-Patógeno , Interferón-alfa/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Pneumovirus/genética , Infecciones por Pneumovirus/metabolismo , Transducción de Señal , Receptor Toll-Like 7/metabolismo
10.
J Allergy Clin Immunol ; 138(5): 1326-1337, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27236500

RESUMEN

BACKGROUND: Frequent viral lower respiratory infections in early life are an independent risk factor for asthma onset. This risk and the development of persistent asthma are significantly greater in children who later become sensitized. OBJECTIVE: We sought to elucidate the pathogenic processes that underlie the synergistic interplay between allergen exposures and viral infections. METHODS: Mice were inoculated with a murine-specific Pneumovirus species (pneumonia virus of mice [PVM]) and exposed to low-dose cockroach extract (CRE) in early and later life, and airway inflammation, remodeling, and hyperreactivity assessed. Mice were treated with anti-IL-33 or apyrase to neutralize or block IL-33 release. RESULTS: PVM infection or CRE exposure alone did not induce disease, whereas PVM/CRE coexposure acted synergistically to induce the hallmark features of asthma. CRE exposure during viral infection in early life induced a biphasic IL-33 response and impaired IFN-α and IFN-λ production, which in turn increased epithelial viral burden, airway smooth muscle growth, and type 2 inflammation. These features were ameliorated when CRE-induced IL-33 release was blocked or neutralized, whereas substitution of CRE with exogenous IL-33 recapitulated the phenotype observed in PVM/CRE-coexposed mice. Mechanistically, IL-33 downregulated viperin and interferon regulatory factor 7 gene expression and rapidly degraded IL-1 receptor-associated kinase 1 expression in plasmacytoid dendritic cells both in vivo and in vitro, leading to Toll-like receptor 7 hyporesponsiveness and impaired IFN-α production. CONCLUSION: We identified a hitherto unrecognized function of IL-33 as a potent suppressor of innate antiviral immunity and demonstrate that IL-33 contributes significantly to the synergistic interplay between respiratory virus and allergen exposures in the onset and progression of asthma.


Asunto(s)
Alérgenos/inmunología , Asma/inmunología , Cucarachas , Citocinas/inmunología , Proteínas de Insectos/inmunología , Virus de la Neumonía Murina , Infecciones por Pneumovirus/inmunología , Contaminantes Atmosféricos/inmunología , Animales , Asma/virología , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/virología , Células Dendríticas/inmunología , Pulmón/virología , Ratones Endogámicos BALB C , Infecciones por Pneumovirus/virología , Carga Viral
11.
J Allergy Clin Immunol ; 136(4): 1065-73, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25930193

RESUMEN

BACKGROUND: A variant in the IL-6 receptor (IL-6R) gene increases asthma risk and is predicted to decrease IL-6 classic signaling and increase IL-6 trans-signaling. This suggests that inhibition of IL-6 trans-signaling, but not classic signaling, might suppress allergic airway inflammation. OBJECTIVES: We sought to determine whether IL-6 signaling contributes to (1) acute experimental asthma induced by clinically relevant allergens and (2) variation in asthma clinical phenotypes in asthmatic patients. METHODS: Mice were sensitized to house dust mite (HDM) or cockroach at day 0, treated with IL-6R inhibitors at day 13, and challenged with the same allergen at days 14 to 17. End points were measured 3 hours after the final challenge. IL-6 and soluble IL-6 receptor (sIL-6R) expression in induced sputum of asthmatic patients was correlated with asthma clinical phenotypes. RESULTS: Both HDM and cockroach induced a type 2/type 17 cytokine profile and mixed granulocytic inflammation in the airways. Both allergens increased IL-6 expression in the airways, but only cockroach induced sIL-6R expression. Therefore HDM challenge promoted IL-6 classic signaling but not trans-signaling; in this model treatment with anti-IL-6R did not suppress airway inflammation. In contrast, cockroach-induced inflammation involved activation of IL-6 trans-signaling and production of IL-17A by γδ T cells. Anti-IL-6R, selective blockade of sIL-6R, or γδ T-cell deficiency significantly attenuated cockroach-induced inflammation. Asthmatic patients with high airway IL-6 and sIL-6R levels were enriched for the neutrophilic and mixed granulocytic subtypes. CONCLUSION: Experimental asthma associated with both high IL-6 and high sIL-6R levels in the airways is attenuated by treatment with IL-6R inhibitors.


Asunto(s)
Asma/inmunología , Interleucina-6/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Interleucina-6/inmunología , Transducción de Señal/inmunología , Células Th17/inmunología , Células Th2/inmunología , Alérgenos/inmunología , Alérgenos/toxicidad , Animales , Asma/inducido químicamente , Asma/patología , Cucarachas/inmunología , Ratones , Pyroglyphidae/inmunología , Transducción de Señal/efectos de los fármacos , Células Th17/patología , Células Th2/patología
12.
Am J Respir Cell Mol Biol ; 53(5): 615-24, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25789608

RESUMEN

Allergic asthma is underpinned by T helper 2 (Th2) inflammation. Redundancy in Th2 cytokine function and production by innate and adaptive immune cells suggests that strategies aimed at immunomodulation may prove more beneficial. Hence, we sought to determine whether administration of mesenchymal stromal cells (MSCs) to house dust mite (HDM) (Dermatophagoides pteronyssinus)-sensitized mice would suppress the development of Th2 inflammation and airway hyperresponsiveness (AHR) after HDM challenge. We report that the intravenous administration of allogeneic donor MSCs 1 hour before allergen challenge significantly attenuated the features of allergic asthma, including tissue eosinophilia, Th2 cytokine (IL-5 and IL-13) levels in bronchoalveolar lavage fluid, and AHR. The number of infiltrating type 2 innate lymphoid cells was not affected by MSC transfer, suggesting that MSCs may modulate the adaptive arm of Th2 immunity. The effect of MSC administration was long lasting; all features of allergic airway disease were significantly suppressed in response to a second round of HDM challenge 4 weeks after MSC administration. Further, we observed that MSCs decreased the release of epithelial cell-derived alarmins IL-1α and high mobility group box-1 in an IL-1 receptor antagonist-dependent manner. This significantly decreased the expression of the pro-Th2 cytokine IL-25 and reduced the number of activated and antigen-acquiring CD11c(+)CD11b(+) dendritic cells in the lung and mediastinal lymph nodes. Our findings suggest that MSC administration can ameliorate allergic airway inflammation by blunting the amplification of epithelial-derived inflammatory cytokines induced by HDM exposure and may offer long-term protection against Th2-mediated allergic airway inflammation and AHR.


Asunto(s)
Alérgenos/farmacología , Dermatophagoides pteronyssinus/inmunología , Eosinofilia/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Hipersensibilidad Respiratoria/terapia , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Eosinofilia/etiología , Eosinofilia/genética , Eosinofilia/inmunología , Femenino , Expresión Génica , Inmunomodulación/efectos de los fármacos , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Interleucinas/genética , Interleucinas/inmunología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/patología , Trasplante Homólogo
13.
J Immunol ; 194(9): 4567-76, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810394

RESUMEN

Inducible BALT (iBALT) can amplify pulmonary or systemic inflammatory responses to the benefit or detriment of the host. We took advantage of the age-dependent formation of iBALT to interrogate the underlying mechanisms that give rise to this ectopic, tertiary lymphoid organ. In this study, we show that the reduced propensity for weanling as compared with neonatal mice to form iBALT in response to acute LPS exposure is associated with greater regulatory T cell expansion in the mediastinal lymph nodes. Ab- or transgene-mediated depletion of regulatory T cells in weanling mice upregulated the expression of IL-17A and CXCL9 in the lungs, induced a tissue neutrophilia, and increased the frequency of iBALT to that observed in neonatal mice. Remarkably, neutrophil depletion in neonatal mice decreased the expression of the B cell active cytokines, a proliferation-inducing ligand and IL-21, and attenuated LPS-induced iBALT formation. Taken together, our data implicate a role for neutrophils in lymphoid neogenesis. Neutrophilic inflammation is a common feature of many autoimmune diseases in which iBALT are present and pathogenic, and hence the targeting of neutrophils or their byproducts may serve to ameliorate detrimental lymphoid neogenesis in a variety of disease contexts.


Asunto(s)
Inflamación/inmunología , Tejido Linfoide/inmunología , Neutrófilos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Animales Recién Nacidos , Microambiente Celular/inmunología , Citocinas/biosíntesis , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Depleción Linfocítica , Tejido Linfoide/metabolismo , Masculino , Ratones , Neutrófilos/metabolismo , Linfocitos T Reguladores/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
14.
Mol Cancer Ther ; 11(4): 909-20, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22389471

RESUMEN

Recent results from clinical trials with the BRAF inhibitors GSK2118436 (dabrafenib) and PLX4032 (vemurafenib) have shown encouraging response rates; however, the duration of response has been limited. To identify determinants of acquired resistance to GSK2118436 and strategies to overcome the resistance, we isolated GSK2118436 drug-resistant clones from the A375 BRAF(V600E) and the YUSIT1 BRAF(V600K) melanoma cell lines. These clones also showed reduced sensitivity to the allosteric mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitor GSK1120212 (trametinib). Genetic characterization of these clones identified an in-frame deletion in MEK1 (MEK1(K59del)) or NRAS mutation (NRAS(Q61K) and/or NRAS(A146T)) with and without MEK1(P387S) in the BRAF(V600E) background and NRAS(Q61K) in the BRAF(V600K) background. Stable knockdown of NRAS with short hairpin RNA partially restored GSK2118436 sensitivity in mutant NRAS clones, whereas expression of NRAS(Q61K) or NRAS(A146T) in the A375 parental cells decreased sensitivity to GSK2118436. Similarly, expression of MEK1(K59del), but not MEK1(P387S), decreased sensitivity of A375 cells to GSK2118436. The combination of GSK2118436 and GSK1120212 effectively inhibited cell growth, decreased ERK phosphorylation, decreased cyclin D1 protein, and increased p27(kip1) protein in the resistant clones. Moreover, the combination of GSK2118436 or GSK1120212 with the phosphoinositide 3-kinase/mTOR inhibitor GSK2126458 enhanced cell growth inhibition and decreased S6 ribosomal protein phosphorylation in these clones. Our results show that NRAS and/or MEK mutations contribute to BRAF inhibitor resistance in vitro, and the combination of GSK2118436 and GSK1120212 overcomes this resistance. In addition, these resistant clones respond to the combination of GSK2126458 with GSK2118436 or GSK1120212. Clinical trials are ongoing or planned to test these combinations.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Apoptosis/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Imidazoles/administración & dosificación , Imidazoles/farmacología , Indoles/administración & dosificación , Indoles/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Melanoma/enzimología , Oximas/administración & dosificación , Oximas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Vemurafenib
15.
Diabetes ; 56(7): 1751-60, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17456854

RESUMEN

The objective of this study was to further establish and confirm the relationship of adipose mitochondrial biogenesis in diabetes/obesity and the effects of rosiglitazone (RSG), a peroxisome proliferator-activated receptor (PPAR) gamma agonist, by systematically analyzing mitochondrial gene expression and function in two mouse models of obesity and type 2 diabetes. Using microarray technology, adipose mitochondrial gene transcription was studied in db/db, high-fat diet-fed C57BL/6 (HFD) and respective control mice with or without RSG treatment. The findings were extended using mitochondrial staining, DNA quantification, and measurements of citrate synthase activity. In db/db and HFD mice, gene transcripts associated with mitochondrial ATP production, energy uncoupling, mitochondrial ribosomal proteins, outer and inner membrane translocases, and mitochondrial heat-shock proteins were decreased in abundance, compared with db/+ and standard-fat diet-fed control mice, respectively. RSG dose-dependently increased these transcripts in both db/db and HFD mice and induced transcription of mitochondrial structural proteins and cellular antioxidant enzymes responsible for removal of reactive oxygen species generated by increased mitochondrial activity. Transcription factors, including PPAR coactivator (PGC)-1beta, PGC-1alpha, estrogen-related receptor alpha, and PPARalpha, were suppressed in both models and induced by RSG. The effects of RSG on adipose mitochondrial genes were confirmed by quantitative RT-PCR and further supported by mitochondrial staining, mitochondrial DNA quantification, and citrate synthase activity. Adipose mitochondrial biogenesis was overwhelmingly suppressed in both mouse models of diabetes/obesity and globally induced by RSG. These findings suggest an important role of adipose mitochondria in diabetes/obesity and the potential for new treatment approaches targeting adipose mitochondria.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Hipoglucemiantes/farmacología , Mitocondrias/efectos de los fármacos , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , Tejido Adiposo/metabolismo , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Grasas de la Dieta , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Obesidad/tratamiento farmacológico , Rosiglitazona , Transcripción Genética
16.
Exp Eye Res ; 77(1): 77-84, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12823990

RESUMEN

The objective of this study was to determine the cellular localization of P2Y(2) receptor gene expression in rabbit and primate ocular tissues using the technique of non-isotopic in situ hybridization. Fresh frozen whole eye from a New Zealand White rabbit and whole eye and eyelid from a rhesus macaque were cut into 5 microm thick sections and mounted onto glass slides. In situ hybridization was performed on ocular cryosections using digoxigenin-labeled P2Y(2) receptor riboprobes. Alkaline phosphatase-conjugated anti-digoxigenin antibody was used to localize riboprobe hybridization, which was subsequently visualized by staining with a precipitating alkaline phosphatase substrate. Cytoplasmic staining indicative of antisense riboprobe hybridization to P2Y(2) receptor mRNA was observed in the palpebral and bulbar conjunctival epithelium, including goblet cells, the corneal epithelium, and in meibomian gland sebaceous and ductal cells. Staining was also observed in both layers of the ciliary body epithelium, subcapsular epithelium of the lens, and corneal endothelium. In the posterior eye, staining was observed in various layers of the retina, including ganglion cell, inner nuclear, inner segment and retinal pigment epithelium layers, in the optic nerve head, and in a variety of structures within the choroid. No specific staining of sense riboprobe was seen on any of the ocular structures. These results demonstrate that the P2Y(2) receptor gene is expressed in a variety of ocular cells types and suggest that P2Y(2) receptors are associated with diverse physiological functions throughout the eye.


Asunto(s)
Células Epiteliales/química , Ojo/metabolismo , ARN Mensajero/análisis , Receptores Purinérgicos P2/genética , Animales , Cuerpo Ciliar/química , Conjuntiva/química , Córnea/química , Endotelio Corneal/química , Epitelio Corneal/química , Expresión Génica , Células Caliciformes/química , Hibridación in Situ/métodos , Cristalino/química , Macaca mulatta , Glándulas Tarsales/química , Conejos , Receptores Purinérgicos P2Y2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA