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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(10)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38791146

RESUMEN

Crohn's disease (CD) is a subtype of inflammatory bowel disease (IBD) characterized by transmural disease. The concept of transmural healing (TH) has been proposed as an indicator of deep clinical remission of CD and as a predictor of favorable treatment endpoints. Understanding the pathophysiology involved in transmural disease is critical to achieving these endpoints. However, most studies have focused on the intestinal mucosa, overlooking the contribution of the intestinal wall in Crohn's disease. Multi-omics approaches have provided new avenues for exploring the pathogenesis of Crohn's disease and identifying potential biomarkers. We aimed to use transcriptomic and proteomic technologies to compare immune and mesenchymal cell profiles and pathways in the mucosal and submucosa/wall compartments to better understand chronic refractory disease elements to achieve transmural healing. The results revealed similarities and differences in gene and protein expression profiles, metabolic mechanisms, and immune and non-immune pathways between these two compartments. Additionally, the identification of protein isoforms highlights the complex molecular mechanisms underlying this disease, such as decreased RTN4 isoforms (RTN4B2 and RTN4C) in the submucosa/wall, which may be related to the dysregulation of enteric neural processes. These findings have the potential to inform the development of novel therapeutic strategies to achieve TH.


Asunto(s)
Colon , Enfermedad de Crohn , Mucosa Intestinal , Proteómica , Humanos , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Enfermedad de Crohn/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Proteómica/métodos , Colon/metabolismo , Colon/patología , Transcriptoma , Masculino , Femenino , Adulto , Perfilación de la Expresión Génica , Biomarcadores , Persona de Mediana Edad , Multiómica
2.
Development ; 142(2): 258-67, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25564622

RESUMEN

Basal cells are multipotent airway progenitors that generate distinct epithelial cell phenotypes crucial for homeostasis and repair of the conducting airways. Little is known about how these progenitor cells expand and transition to differentiation to form the pseudostratified airway epithelium in the developing and adult lung. Here, we show by genetic and pharmacological approaches that endogenous activation of Notch3 signaling selectively controls the pool of undifferentiated progenitors of upper airways available for differentiation. This mechanism depends on the availability of Jag1 and Jag2, and is key to generating a population of parabasal cells that later activates Notch1 and Notch2 for secretory-multiciliated cell fate selection. Disruption of this mechanism resulted in aberrant expansion of basal cells and altered pseudostratification. Analysis of human lungs showing similar abnormalities and decreased NOTCH3 expression in subjects with chronic obstructive pulmonary disease suggests an involvement of NOTCH3-dependent events in the pathogenesis of this condition.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/fisiología , Células Epiteliales/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón/embriología , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Mucosa Respiratoria/embriología , Transducción de Señal/fisiología , Animales , Western Blotting , Técnicas de Cultivo de Célula , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Hibridación in Situ , Proteína Jagged-1 , Ratones , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Notch3 , Mucosa Respiratoria/citología , Proteínas Serrate-Jagged , Especificidad de la Especie
3.
FASEB J ; 28(2): 897-907, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24221086

RESUMEN

Children who are exposed to environmental respiratory insults often develop asthma that persists into adulthood. In this study, we used a neonatal mouse model of ovalbumin (OVA)-induced allergic airway inflammation to understand the long-term effects of early childhood insults on airway structure and function. We showed that OVA sensitization and challenge in early life led to a 2-fold increase in airway smooth muscle (ASM) innervation (P<0.05) and persistent airway hyperreactivity (AHR). In contrast, OVA exposure in adult life elicited short-term AHR without affecting innervation levels. We found that postnatal ASM innervation required neurotrophin (NT)-4 signaling through the TrkB receptor and that early-life OVA exposure significantly elevated NT4 levels and TrkB signaling by 5- and 2-fold, respectively, to increase innervation. Notably, blockade of NT4/TrkB signaling in OVA-exposed pups prevented both acute and persistent AHR without affecting baseline airway function or inflammation. Furthermore, biophysical assays using lung slices and isolated cells demonstrated that NT4 was necessary for hyperreactivity of ASM induced by early-life OVA exposure. Together, our findings show that the NT4/TrkB-dependent increase in innervation plays a critical role in the alteration of the ASM phenotype during postnatal growth, thereby linking early-life allergen exposure to persistent airway dysfunction.


Asunto(s)
Músculo Liso/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Receptor trkB/metabolismo , Alérgenos/inmunología , Animales , Asma , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Ratones , Microscopía Confocal , Músculo Liso/efectos de los fármacos , Factores de Crecimiento Nervioso/genética , Ovalbúmina/inmunología , Receptor trkB/genética
4.
Am J Respir Cell Mol Biol ; 50(5): 876-81, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24313705

RESUMEN

An emerging tool in airway biology is the precision-cut lung slice (PCLS). Adoption of the PCLS as a model for assessing airway reactivity has been hampered by the limited time window within which tissues remain viable. Here we demonstrate that the PCLS can be frozen, stored long-term, and then thawed for later experimental use. Compared with the never-frozen murine PCLS, the frozen-thawed PCLS shows metabolic activity that is decreased to an extent comparable to that observed in other cryopreserved tissues but shows no differences in cell viability or in airway caliber responses to the contractile agonist methacholine or the relaxing agonist chloroquine. These results indicate that freezing and long-term storage is a feasible solution to the problem of limited viability of the PCLS in culture.


Asunto(s)
Pulmón/fisiología , Contracción Muscular/fisiología , Animales , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Criopreservación/métodos , Congelación , Ratones , Ratones Endogámicos C57BL
5.
Am J Pathol ; 182(6): 2391-406, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583651

RESUMEN

Pulmonary arterial hypertension (PAH) is a chronic and progressive disease characterized by pulmonary vasculopathy with elevation of pulmonary artery pressure, often culminating in right ventricular failure. GATA-6, a member of the GATA family of zinc-finger transcription factors, is highly expressed in quiescent vasculature and is frequently lost during vascular injury. We hypothesized that endothelial GATA-6 may play a critical role in the molecular mechanisms underlying endothelial cell (EC) dysfunction in PAH. Here we report that GATA-6 is markedly reduced in pulmonary ECs lining both occluded and nonoccluded vessels in patients with idiopathic and systemic sclerosis-associated PAH. GATA-6 transcripts are also rapidly decreased in rodent PAH models. Endothelial GATA-6 is a direct transcriptional regulator of genes controlling vascular tone [endothelin-1, endothelin-1 receptor type A, and endothelial nitric oxide synthase (eNOS)], pro-inflammatory genes, CX3CL1 (fractalkine), 5-lipoxygenease-activating protein, and markers of vascular remodeling, including PAI-1 and RhoB. Mice with the genetic deletion of GATA-6 in ECs (Gata6-KO) spontaneously develop elevated pulmonary artery pressure and increased vessel muscularization, and these features are further exacerbated in response to hypoxia. Furthermore, innate immune cells including macrophages (CD11b(+)/F4/80(+)), granulocytes (Ly6G(+)/CD45(+)), and dendritic cells (CD11b(+)/CD11c(+)) are significantly increased in normoxic Gata6-KO mice. Together, our findings suggest a critical role of endothelial GATA-6 deficiency in development and disease progression in PAH.


Asunto(s)
Endotelio Vascular/metabolismo , Factor de Transcripción GATA6/deficiencia , Hipertensión Pulmonar/metabolismo , Adaptación Fisiológica/fisiología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Estudios de Casos y Controles , Enfermedad Crónica , Progresión de la Enfermedad , Regulación hacia Abajo/fisiología , Células Endoteliales/fisiología , Hipertensión Pulmonar Primaria Familiar , Factor de Transcripción GATA6/metabolismo , Factor de Transcripción GATA6/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Hipertensión Pulmonar/etiología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Hipoxia/complicaciones , Pulmón/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Neumonía/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Esclerodermia Sistémica/complicaciones
6.
J Neurosci ; 31(43): 15407-15, 2011 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22031887

RESUMEN

Dysfunctional neural control of airway smooth muscle (ASM) is involved in inflammatory diseases, such as asthma. However, neurogenesis in the lung is poorly understood. This study uses mouse models to investigate developmental mechanisms of ASM innervation, a process that is highly coordinated with ASM formation during lung branching morphogenesis. We show that brain-derived neurotrophic factor (BDNF) is an essential ASM-derived signal for innervation. Although BDNF mRNA expression is temporally dissociated with ASM formation and innervation, BDNF protein is coordinately produced through post-transcriptional suppression by miR-206. Using a combination of chemical and genetic approaches to modulate sonic hedgehog (Shh) signaling, a pathway essential for lung branching and ASM formation, we show that Shh signaling blocks miR-206 expression, which in turn increases BDNF protein expression. Together, our work uncovers a functional cascade that involves Shh, miR-206 and BDNF to coordinate ASM formation and innervation.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Hedgehog/metabolismo , MicroARNs/metabolismo , Músculo Liso/fisiología , Sistema Respiratorio/anatomía & histología , Sistema Respiratorio/inervación , Factores de Edad , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Hedgehog/genética , Ratones , Ratones Transgénicos , MicroARNs/genética , Músculo Liso/inervación , ARN Mensajero/metabolismo , Sistema Respiratorio/embriología , Transducción de Señal/genética , Tubulina (Proteína)/metabolismo
7.
J Biol Chem ; 286(25): 22678-87, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21536678

RESUMEN

Notch3 signaling is fundamental for arterial specification of systemic vascular smooth muscle cells (VSMCs). However, the developmental role and signaling properties of the Notch3 receptor in the mouse pulmonary artery remain unknown. Here, we demonstrate that Notch3 is expressed selectively in pulmonary artery VSMCs, is activated from late fetal to early postnatal life, and is required to maintain the morphological characteristics and smooth muscle gene expression profile of the pulmonary artery after birth. Using a conditional knock-out mouse model, we show that Notch3 receptor activation in VSMCs is Jagged1-dependent. In vitro VSMC lentivirus-mediated Jagged1 knockdown, confocal localization analysis, and co-culture experiments revealed that Notch3 activation is cell-autonomous and occurs through the physical engagement of Notch3 and VSMC-derived Jagged1 in the interior of the same cell. Although the current models of mammalian Notch signaling involve a two-cell system composed of a signal-receiving cell that expresses a Notch receptor on its surface and a neighboring signal-sending cell that provides membrane-bound activating ligand, our data suggest that pulmonary artery VSMC Notch3 activation is cell-autonomous. This unique mechanism of Notch activation may play an important role in the maturation of the pulmonary artery during the transition to air breathing.


Asunto(s)
Pulmón/irrigación sanguínea , Pulmón/crecimiento & desarrollo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Biomarcadores/metabolismo , Proteínas de Unión al Calcio/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Espacio Intracelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/metabolismo , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Especificidad de Órganos , Embarazo , Transporte de Proteínas , Ratas , Receptor Notch3 , Proteínas Serrate-Jagged , Factores de Tiempo
8.
Proc Natl Acad Sci U S A ; 106(26): 10734-9, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19528638

RESUMEN

The ability to induce durable transplantation tolerance predictably and consistently in the clinic is a highly desired but elusive goal. Progress is hampered by lack of appropriate experimental models in which to study resistance to transplantation tolerance. Here, we demonstrate that T helper 1-associated T box 21 transcription factor (Tbet) KO recipients exhibit allograft tolerance resistance specifically mediated by IL-17-producing CD8 T (T17) cells. Neutralization of IL-17 facilitates long-term cardiac allograft survival with combined T cell co-stimulation (CD28-CD80/86 and CD154-CD40) blockade in Tbet KO recipients. We have used this T17-biased Tbet KO model of allograft tolerance resistance to study the impact of targeting a T cell-co-stimulatory pathway, and demonstrate that targeting T cell Ig and mucin domain-1 (Tim-1) with anti-Tim-1 overcomes this resistance by specifically inhibiting the pathogenic IL-17-producing CD8 T17 cells. These data indicate that in the absence of Th1 immunity, CD8 T17 alloreactivity constitutes a barrier to transplantation tolerance. Targeting TIM-1 provides an approach to overcome resistance to tolerance in clinical transplantation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocitos T CD8-positivos/metabolismo , Interleucina-17/inmunología , Proteínas de la Membrana/inmunología , Tolerancia al Trasplante/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Técnica del Anticuerpo Fluorescente , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Trasplante de Corazón/métodos , Receptor Celular 1 del Virus de la Hepatitis A , Interleucina-17/metabolismo , Estimación de Kaplan-Meier , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Trasplante Homólogo
9.
Mucosal Immunol ; 15(6): 1338-1349, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36372810

RESUMEN

Inflammatory bowel disease (IBD) is characterized by a dysregulated intestinal epithelial barrier leading to breach of barrier immunity. Here we identified similar protein expression changes between IBD and Citrobacter rodentium-infected FVB mice with respect to dysregulation of solute transporters as well as components critical for intestinal barrier integrity. We attribute the disease associated changes in the model to the emergence of undifferentiated intermediate intestinal epithelial cells. Prophylactic treatment with IL-22.Fc in C. rodentium-infected FVB mice reduced disease severity and rescued the mice from lethality. Multi-omics and solute analyses revealed that IL-22.Fc treatment prevented disease-associated changes including disruption of the solute transporter machinery and restored proper physiological functions of the intestine, respectively. Taken together, we established the disease relevance of the C. rodentium-induced colitis model to IBD, demonstrated the protective role of IL-22 in amelioration of epithelial dysfunction and elucidated the molecular mechanisms with IL-22's effect on intestinal epithelial cells.


Asunto(s)
Colitis , Infecciones por Enterobacteriaceae , Enfermedades Inflamatorias del Intestino , Interleucinas , Animales , Ratones , Citrobacter rodentium/fisiología , Colitis/tratamiento farmacológico , Colitis/microbiología , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/metabolismo , Intestinos , Ratones Endogámicos C57BL , Interleucinas/farmacología , Interleucina-22
10.
J Exp Med ; 217(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32106283

RESUMEN

T cells are classically recognized as distinct subsets that express αß or γδ TCRs. We identify a novel population of T cells that coexpress αß and γδ TCRs in mice and humans. These hybrid αß-γδ T cells arose in the murine fetal thymus by day 16 of ontogeny, underwent αß TCR-mediated positive selection into CD4+ or CD8+ thymocytes, and constituted up to 10% of TCRδ+ cells in lymphoid organs. They expressed high levels of IL-1R1 and IL-23R and secreted IFN-γ, IL-17, and GM-CSF in response to canonically restricted peptide antigens or stimulation with IL-1ß and IL-23. Hybrid αß-γδ T cells were transcriptomically distinct from conventional γδ T cells and displayed a hyperinflammatory phenotype enriched for chemokine receptors and homing molecules that facilitate migration to sites of inflammation. These proinflammatory T cells promoted bacterial clearance after infection with Staphylococcus aureus and, by licensing encephalitogenic Th17 cells, played a key role in the development of autoimmune disease in the central nervous system.


Asunto(s)
Inflamación/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología , Animales , Biomarcadores/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Humanos , Inflamación/patología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Fenotipo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/fisiología , Transcripción Genética , Transcriptoma/genética
11.
Crit Rev Immunol ; 28(4): 301-24, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19166382

RESUMEN

Type 1 diabetes (T1D) is caused by the autoimmune-mediated destruction of insulin-producing beta cells in the pancreas. T1D affects as many as 3 million patients in the United States alone, with 15,000 new cases developing every year (Juvenile Diabetes Research Foundation), and presently there is no cure for T1D. In recent years, there has been a great deal of interest in developing gene therapy approaches to treat T1D. Gene therapy approaches tend to fall into three broad categoriesthose aimed at preventing or curing autoimmunity, those aimed at restoring insulin production through islet transplant or genetically engineered insulin production, and approaches that aim to prevent the morbidity and mortality associated with this complex disease. We review these studies here.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Terapia Genética , Células Presentadoras de Antígenos/fisiología , Autoantígenos/inmunología , Autoinmunidad , Citocinas/genética , Complicaciones de la Diabetes/terapia , Diabetes Mellitus Tipo 1/inmunología , Genes MHC Clase II , Humanos , Tolerancia Inmunológica , Insulina/biosíntesis , Linfocitos T Reguladores/fisiología , Cicatrización de Heridas
12.
Inflamm Bowel Dis ; 25(12): 1906-1918, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31173627

RESUMEN

BACKGROUND: Crohn's disease (CD) and ulcerative colitis (UC) are intestinal chronic inflammatory conditions characterized by altered epithelial barrier function and tissue damage. Despite significant efforts to understanding the biological mechanisms responsible for gut inflammation, the pathophysiology of CD and UC remains poorly understood. METHODS: To help elucidate the potential mechanisms responsible for gut inflammation in CD and UC, transcriptomic and proteomic profiling of human colon biopsy specimens was performed. Dysregulated genes and proteins in disease tissues compared with normal tissues were characterized from the expression profiles and further subjected to pathway analysis to identify altered biological processes and signaling pathways. RESULTS: Sample analysis showed 4250 genes with matched protein expression and a wide range of correlation of RNA-protein abundance across samples. Pathway analysis of dysregulated genes and proteins in CD and UC showed alterations in immune and inflammatory responses, complement cascade, and the suppression of metabolic processes and PPAR signaling. In CD, increased T-helper cell differentiation and elevated toll-like receptor and JAK/STAT signaling were observed. Interestingly, increased MAPK signaling was only observed in UC. Weighted gene co-expression network analysis suggested a possible role of epigenetic regulation in UC. Of note, a large discrepancy between regulation of RNA and protein levels in inflamed colon samples was detected for previously identified biomarkers including MMP14 and LAMP1. CONCLUSIONS: With the analysis of dysregulated genes and pathways, the present study unravels key mechanisms contributing to CD and UC pathogenesis and emphasizes that integrative analysis of multi-omics data sets can provide more insight into understanding complex disease mechanisms.


Asunto(s)
Colon/patología , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/metabolismo , Proteoma , Transcriptoma , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Biopsia , Epigénesis Genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN/análisis , Transducción de Señal , Adulto Joven
13.
PLoS One ; 12(8): e0182841, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28792532

RESUMEN

Mutations in the Interleukin (IL)-23/IL-23 receptor loci are associated with increased inflammatory bowel disease (IBD) susceptibility, and IL-23 neutralization has shown efficacy in early clinical trials. To better understand how an excess of IL-23 affects the gastrointestinal tract, we investigated chronic systemic IL-23 exposure in healthy wildtype mice. As expected, IL-23 exposure resulted in early activation of intestinal type 3 innate lymphoid cells (ILC3), followed by infiltration of activated RORγt+ T helper cells. Surprisingly, however, sustained IL-23 stimulus also dramatically reduced classical ILC3 populations within the proximal small intestine, and a phenotypically distinct T-bet expressing ILC3 population emerged. TNFα neutralization, a widely used IBD therapy, reduced several aspects of the IL-23 driven ILC3 response, suggesting a synergy between IL-23 and TNFα in ILC3 activation. In vitro studies supported these findings, revealing previously unappreciated effects of IL-23 and TNFα within the intestine.


Asunto(s)
Interleucina-23/metabolismo , Intestino Delgado/inmunología , Linfocitos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Interleucina-23/administración & dosificación , Intestino Delgado/patología , Linfocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores CCR6/metabolismo , Receptores de Interleucina-7/metabolismo , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
14.
PLoS One ; 8(9): e74469, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040256

RESUMEN

Phenotypes of lung smooth muscle cells in health and disease are poorly characterized. This is due, in part, to a lack of methodologies that allow for the independent and direct isolation of bronchial smooth muscle cells (BSMCs) and vascular smooth muscle cells (VSMCs) from the lung. In this paper, we describe the development of a bi-fluorescent mouse that permits purification of these two cell populations by cell sorting. By subjecting this mouse to an acute allergen based-model of airway inflammation that exhibits many features of asthma, we utilized this tool to characterize the phenotype of so-called asthmatic BSMCs. First, we examined the biophysical properties of single BSMCs from allergen sensitized mice and found increases in basal tone and cell size that were sustained ex vivo. We then generated for the first time, a comprehensive characterization of the global gene expression changes in BSMCs isolated from the bi-fluorescent mice with allergic airway inflammation. Using statistical methods and pathway analysis, we identified a number of differentially expressed mRNAs in BSMCs from allergen sensitized mice that code for key candidate proteins underlying changes in matrix formation, contractility, and immune responses. Ultimately, this tool will provide direction and guidance for the logical development of new markers and approaches for studying human lung smooth muscle.


Asunto(s)
Asma/genética , Bronquios/metabolismo , Hiperreactividad Bronquial/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fenotipo , Proteoma/inmunología , Alérgenos/inmunología , Animales , Asma/inmunología , Asma/patología , Bronquios/inmunología , Bronquios/patología , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Tamaño de la Célula , Modelos Animales de Enfermedad , Fluorescencia , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunización , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Transgénicos , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Ovalbúmina/inmunología , Proteoma/genética , ARN Mensajero/genética , ARN Mensajero/inmunología , Análisis de la Célula Individual
15.
Stem Cells Dev ; 19(3): 341-50, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19788396

RESUMEN

Although the bone marrow (BM) microenvironment is the main inducer niche of early B lymphopoiesis during the adult life, other extramedullar microenvironments, such as the liver, may also have potential for supporting B-cell development. Previously, we reported that murine liver sinusoidal endothelial cells (LSECs) support in vitro and in vivo hematopoietic stem cell (HSC) proliferation and myeloid differentiation. In the present study, we investigated the capacity of LSEC to promote B lymphopoiesis from BM progenitor lineage-negative (Lin(-)) cells. Murine BM Lin(-) cells were co-cultured with LSEC, in the absence of exogenous cytokines. B cells were characterized by flow cytometry and cytokine expression by RT-PCR. We show that BM Lin(-) cells differentiated to early B-lymphoid progenitors (B220(+)) and subsequently to mature (CD19(+)) B cells. Functional studies showed the presence of a high number of non-adherent cells (NACs), collected from lipopolysaccharide (LPS)-treated Lin(-)/LSEC co-cultures, expressing IgM on their surface (sIgM). Colony formation from NAC was observed in the presence of IL-7 (CFU-IL-7). LSEC constitutively express IL-7, Flt-3L, and SCF at the mRNA level, and VCAM-1 on their surface, which may explain the capacity of these cells to promote B lymphopoiesis. These data demonstrate that LSEC promote all stages of B lymphopoiesis. To our knowledge, this is the first report that LSEC constitute an in vitro microenvironment for B lymphopoiesis. Further studies will establish whether LSEC can serve in vivo as a B-lymphopoietic niche under physiological or pathological condition, or when HSC are mobilized.


Asunto(s)
Linfocitos B/citología , Células de la Médula Ósea/citología , Diferenciación Celular , Hígado/citología , Animales , Antígenos CD19/análisis , Linfocitos B/metabolismo , Células de la Médula Ósea/metabolismo , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Endotelio/citología , Endotelio/metabolismo , Femenino , Citometría de Flujo , Expresión Génica , Interleucina-7/genética , Antígenos Comunes de Leucocito/análisis , Hígado/metabolismo , Linfopoyesis , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Molécula 1 de Adhesión Celular Vascular/genética
16.
Endothelium ; 15(4): 165-73, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18663620

RESUMEN

UNLABELLED: Liver sinusoidal endothelial cells (LSECs) constitute an attractive target for gene therapy of several liver and systemic diseases. However, there are few reports showing an efficient plasmid-based or viral methodology to deliver recombinant genes into these cells. In the present study, the authors evaluated in vitro gene transfer efficiency of standard plasmid-based techniques (i.e., electroporation, lipofection, and calcium phosphate) and lentiviral-mediated gene transduction into primary murine LSECs, using reporter genes. The results show that electroporation is the most effective in vitro plasmid-gene transfer method to deliver GFP into LSECs (31%), as compared with lipofection and calcium phosphate transfection (6% and 4%, respectively). However, lentiviral transduction resulted in higher, efficient, and stable gene transfer (70%) as compared with plasmid-based techniques. CONCLUSIONS: The highly efficient gene expression obtained by lentiviral transduction and electroporation shows that these methodologies are highly reliable systems for gene transfer into LSECs.


Asunto(s)
Células Endoteliales/fisiología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Lentivirus/genética , Hígado/fisiología , Plásmidos , Animales , Anticuerpos Monoclonales/genética , Clonación Molecular , Electroporación/métodos , Genes Reporteros , Molécula 1 de Adhesión Intercelular/genética , Liposomas , Ratones , Ratas , Transfección/métodos , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/genética
17.
J Exp Med ; 205(13): 3133-44, 2008 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-19047438

RESUMEN

T-bet plays a crucial role in Th1 development. We investigated the role of T-bet in the development of allograft rejection in an established MHC class II-mismatched (bm12 into B6) model of chronic allograft vasculopathy (CAV). Intriguingly, and in contrast to IFN-gamma(-/-) mice that are protected from CAV, T-bet(-/-) recipients develop markedly accelerated allograft rejection accompanied by early severe vascular inflammation and vasculopathy, and infiltration by predominantly IL-17-producing CD4 T cells. Concurrently, T-bet(-/-) mice exhibit a T helper type 1 (Th1)-deficient environment characterized by profound IFN-gamma deficiency, a Th2 switch characterized by increased production of interleukin (IL) 4, IL-5, IL-10, and IL-13 cytokines, as well as increased production of the proinflammatory cytokines IL-6, IL-12p40, and IL-17. Neutralization of IL-17 inhibits accelerated allograft rejection and vasculopathy in T-bet(-/-) mice. Interestingly, CD4 but not CD8 T cell deficiency in T-bet(-/-) mice affords dramatic protection from vasculopathy and facilitates long-term graft acceptance. This is the first study establishing that in the absence of Th1-mediated alloimmune responses, CD4 Th17 cells mediate an aggressive proinflammatory response culminating in severe accelerated allograft rejection and vasculopathy. These results have important implications for the development of novel therapies to target this intractable problem in clinical solid organ transplantation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Enfermedades Vasculares/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Citocinas/inmunología , Genes MHC Clase II , Memoria Inmunológica/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Ratones , Ratones Noqueados , Fenotipo , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/inmunología , Proteínas de Dominio T Box/genética , Trasplante Homólogo/inmunología
18.
J Immunol ; 179(10): 6762-9, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17982066

RESUMEN

We have previously shown that the development of type 1 diabetes (T1D) can be prevented in nonobese diabetic (NOD) mice by reconstitution with autologous hemopoietic stem cells retrovirally transduced with viruses encoding MHC class II I-A beta-chain molecules associated with protection from the disease. In this study we examined whether a blockade of the programmed death-1 (PD-1)-programmed death ligand-1 (PD-L1) pathway, a major pathway known to control diabetes occurrence, could precipitate T1D in young NOD mice following reconstitution with autologous bone marrow retrovirally transduced with viruses encoding protective MHC class II I-A beta-chain molecules. In addition, we examined whether the expression of protective MHC class II alleles in hemopoietic cells could be used to prevent the recurrence of diabetes in mice with pre-existing disease following islet transplantation. Protection from the occurrence of T1D diabetes in young NOD mice by the expression of protective MHC class II I-A beta-chain molecules in bone marrow-derived hemopoietic cells was resistant to induction by PD-1-PD-L1 blockade. Moreover, reconstitution of NOD mice with pre-existing T1D autologous hemopoietic stem cells transduced with viruses encoding protective MHC class II I-A beta-chains allowed for the successful transplantation of syngeneic islets, resulting in the long-term reversal of T1D. Reversal of diabetes was resistant to induction by PD-1-PDL-1 blockade and depletion of CD25(+) T cells. These data suggest that expression of protective MHC class II alleles in bone marrow-derived cells establishes robust self-tolerance to islet autoantigens and is sufficient to prevent the recurrence of autoimmune diabetes following islet transplantation.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Terapia Genética , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/inmunología , Tolerancia al Trasplante , Alelos , Animales , Antígenos de Diferenciación/inmunología , Antígeno B7-1/inmunología , Antígeno B7-H1 , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/genética , Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Péptidos/inmunología , Receptor de Muerte Celular Programada 1 , Retroviridae , Linfocitos T/inmunología , Transducción Genética , Inmunología del Trasplante , Tolerancia al Trasplante/genética , Trasplante Autólogo , Trasplante Isogénico
19.
J Immunol ; 176(6): 3410-6, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16517709

RESUMEN

Induction of molecular chimerism following reconstitution of mice with autologous bone marrow cells expressing a retrovirally encoded allogeneic MHC class I Ag results in donor-specific tolerance. To investigate the mechanism by which CD4 T cells that recognize allogeneic MHC class I through the indirect pathway of Ag presentation are rendered tolerant in molecular chimeras, transgenic mice expressing a TCR on CD4 T cells specific for peptides derived from K(b) were used. CD4 T cells expressing the transgenic TCR were detected in mice reconstituted with bone marrow cells transduced with retroviruses carrying the gene encoding H-2K(b), albeit detection was at lower levels than in mice receiving mock-transduced bone marrow. Despite the presence of CD4 T cells expressing an alloreactive TCR, mice receiving H-2K(b)-transduced bone marrow permanently accepted K(b) disparate skin grafts. CD4+CD25+ T cells from mice reconstituted with H-2K(b)-transduced bone marrow prevented rejection of K(b) disparate skin grafts when adoptively transferred into immunodeficient mice along with effector T cells, suggesting that induction of molecular chimerism leads to the generation of donor specific regulatory T cells, which may be involved in preventing alloreactive CD4 T cell responses that lead to rejection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimera/inmunología , Tolerancia Inmunológica/inmunología , Isoantígenos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Presentación de Antígeno/inmunología , Médula Ósea/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Trasplante de Células , Células Cultivadas , Femenino , Eliminación de Gen , Ratones , Ratones Transgénicos , Modelos Inmunológicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Tasa de Supervivencia , Timo/metabolismo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA