Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 115(14): E3173-E3181, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29559533

RESUMEN

Wnts and R-spondins (RSPOs) support intestinal homeostasis by regulating crypt cell proliferation and differentiation. Ex vivo, Wnts secreted by Paneth cells in organoids can regulate the proliferation and differentiation of Lgr5-expressing intestinal stem cells. However, in vivo, Paneth cell and indeed all epithelial Wnt production is completely dispensable, and the cellular source of Wnts and RSPOs that maintain the intestinal stem-cell niche is not known. Here we investigated both the source and the functional role of stromal Wnts and RSPO3 in regulation of intestinal homeostasis. RSPO3 is highly expressed in pericryptal myofibroblasts in the lamina propria and is several orders of magnitude more potent than RSPO1 in stimulating both Wnt/ß-catenin signaling and organoid growth. Stromal Rspo3 ablation ex vivo resulted in markedly decreased organoid growth that was rescued by exogenous RSPO3 protein. Pdgf receptor alpha (PdgfRα) is known to be expressed in pericryptal myofibroblasts. We therefore evaluated if PdgfRα identified the key stromal niche cells. In vivo, Porcn excision in PdgfRα+ cells blocked intestinal crypt formation, demonstrating that Wnt production in the stroma is both necessary and sufficient to support the intestinal stem-cell niche. Mice with Rspo3 excision in the PdgfRα+ cells had decreased intestinal crypt Wnt/ß-catenin signaling and Paneth cell differentiation and were hypersensitive when stressed with dextran sodium sulfate. The data support a model of the intestinal stem-cell niche regulated by both Wnts and RSPO3 supplied predominantly by stromal pericryptal myofibroblasts marked by PdgfRα.


Asunto(s)
Células Epiteliales/citología , Intestinos/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Nicho de Células Madre/fisiología , Células Madre/citología , Células del Estroma/citología , Trombospondinas/metabolismo , Proteína Wnt1/metabolismo , Aciltransferasas/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Células Epiteliales/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides/citología , Organoides/metabolismo , Células Madre/metabolismo , Células del Estroma/metabolismo , Trombospondinas/genética , Proteína Wnt1/genética
2.
Development ; 141(11): 2206-15, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24821987

RESUMEN

Wnt/ß-catenin signaling supports intestinal homeostasis by regulating proliferation in the crypt. Multiple Wnts are expressed in Paneth cells as well as other intestinal epithelial and stromal cells. Ex vivo, Wnts secreted by Paneth cells can support intestinal stem cells when Wnt signaling is enhanced with supplemental R-Spondin 1 (RSPO1). However, in vivo, the source of Wnts in the stem cell niche is less clear. Genetic ablation of Porcn, an endoplasmic reticulum resident O-acyltransferase that is essential for the secretion and activity of all vertebrate Wnts, confirmed the role of intestinal epithelial Wnts in ex vivo culture. Unexpectedly, mice lacking epithelial Wnt activity (Porcn(Del)/Villin-Cre mice) had normal intestinal proliferation and differentiation, as well as successful regeneration after radiation injury, indicating that epithelial Wnts are dispensable for these processes. Consistent with a key role for stroma in the crypt niche, intestinal stromal cells endogenously expressing Wnts and Rspo3 support the growth of Porcn(Del) organoids ex vivo without RSPO1 supplementation. Conversely, increasing pharmacologic PORCN inhibition, affecting both stroma and epithelium, reduced Lgr5 intestinal stem cells, inhibited recovery from radiation injury, and at the highest dose fully blocked intestinal proliferation. We conclude that epithelial Wnts are dispensable and that stromal production of Wnts can fully support normal murine intestinal homeostasis.


Asunto(s)
Epitelio/metabolismo , Proteínas de la Membrana/genética , Nicho de Células Madre , Células del Estroma/citología , Proteínas Wnt/metabolismo , Aciltransferasas , Animales , Apoptosis , Proliferación Celular , Retículo Endoplásmico/metabolismo , Células Epiteliales/citología , Fibroblastos/metabolismo , Eliminación de Gen , Células HEK293 , Homeostasis , Humanos , Intestinos/citología , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Células de Paneth/citología , Transducción de Señal , Células Madre/citología , Trombospondinas/metabolismo
3.
Kidney Int ; 89(5): 1062-1074, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27083283

RESUMEN

Activated Wnt signaling is critical in the pathogenesis of renal fibrosis, a final common pathway for most forms of chronic kidney disease. Therapeutic intervention by inhibition of individual Wnts or downstream Wnt/ß-catenin signaling has been proposed, but these approaches do not interrupt the functions of all Wnts nor block non-canonical Wnt signaling pathways. Alternatively, an orally bioavailable small molecule, Wnt-C59, blocks the catalytic activity of the Wnt-acyl transferase porcupine, and thereby prevents secretion of all Wnt isoforms. We found that inhibiting porcupine dramatically attenuates kidney fibrosis in the murine unilateral ureteral obstruction model. Wnt-C59 treatment similarly blunts collagen mRNA expression in the obstructed kidney. Consistent with its actions to broadly arrest Wnt signaling, porcupine inhibition reduces expression of Wnt target genes and bolsters nuclear exclusion of ß-catenin in the kidney following ureteral obstruction. Importantly, prevention of Wnt secretion by Wnt-C59 blunts expression of inflammatory cytokines in the obstructed kidney that otherwise provoke a positive feedback loop of Wnt expression in collagen-producing fibroblasts and epithelial cells. Thus, therapeutic targeting of porcupine abrogates kidney fibrosis not only by overcoming the redundancy of individual Wnt isoforms but also by preventing upstream cytokine-induced Wnt generation. These findings reveal a novel therapeutic maneuver to protect the kidney from fibrosis by interrupting a pathogenic crosstalk loop between locally generated inflammatory cytokines and the Wnt/ß-catenin signaling pathway.


Asunto(s)
Bencenoacetamidas/farmacología , Inhibidores Enzimáticos/farmacología , Enfermedades Renales/prevención & control , Riñón/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Piridinas/farmacología , Obstrucción Ureteral/tratamiento farmacológico , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Acilación , Aciltransferasas , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/metabolismo , beta Catenina/metabolismo
4.
PLoS Pathog ; 10(1): e1003887, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24465207

RESUMEN

To be able to colonize its host, invading Salmonella enterica serovar Typhimurium must disrupt and severely affect host-microbiome homeostasis. Here we report that S. Typhimurium induces acute infectious colitis by inhibiting peroxisome proliferator-activated receptor gamma (PPARγ) expression in intestinal epithelial cells. Interestingly, this PPARγ down-regulation by S. Typhimurium is independent of TLR-4 signaling but triggers a marked elevation of host innate immune response genes, including that encoding the antimicrobial peptide lipocalin-2 (Lcn2). Accumulation of Lcn2 stabilizes the metalloproteinase MMP-9 via extracellular binding, which further aggravates the colitis. Remarkably, when exposed to S. Typhimurium, Lcn2-null mice exhibited a drastic reduction of the colitis and remained protected even at later stages of infection. Our data suggest a mechanism in which S. Typhimurium hijacks the control of host immune response genes such as those encoding PPARγ and Lcn2 to acquire residence in a host, which by evolution has established a symbiotic relation with its microbiome community to prevent pathogen invasion.


Asunto(s)
Proteínas de Fase Aguda/inmunología , Colitis/inmunología , Evasión Inmune , Lipocalinas/inmunología , Proteínas Oncogénicas/inmunología , PPAR gamma/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Enfermedad Aguda , Proteínas de Fase Aguda/genética , Animales , Línea Celular , Colitis/genética , Colitis/microbiología , Colitis/patología , Humanos , Lipocalina 2 , Lipocalinas/genética , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Noqueados , Proteínas Oncogénicas/genética , PPAR gamma/genética , Infecciones por Salmonella/genética , Infecciones por Salmonella/patología , Salmonella typhimurium/patogenicidad , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
5.
Nanomedicine ; 11(7): 1643-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26115640

RESUMEN

In this study, we report that surface functional groups of single walled carbon nanotubes (f-SWCNT) are critical for mediating survival and ex vivo expansion of hematopoietic stem and progenitor cells (HSPC) in human umbilical cord blood (UCB). In comparison to amide (-O-NH2) and polyethylene-glycol (-PEG) functionalized SWCNT, carboxylic acid (-COOH) variants gave optimal viability support which correlated with maximal reduction of lethal mitochondrial superoxides in HSPC. Cytokine array illustrated that f-SWCNT-COOH maintained higher proportion of HSPC associated cytokines and minimal level of differentiation promoting factors. Transplantation of f-SWCNT-COOH expanded grafts in sub-lethally irradiated immunodeficient mice resulted in higher engraftment of HSPC in bone marrow compared to control when they were co-transplanted with non-expanded cells from the same UCB. Expanded grafts mediated higher survival rate of mice compared to non-expanded grafts due to lower graft-versus-host-disease which is likely a consequence of proportion of immune cells in the grafts. FROM THE CLINICAL EDITOR: Umbilical cord blood (UCB) is a potential source of hematopoietic stem and progenitor (HSPC) cells. One major hurdle for its clinical use is the insufficient yield of cell number. The authors in this study elegantly demonstrated the importance of various functional groups on single-walled carbon nanotubes (f-SWCNT) in enhancing ex vivo expansion of HSPC in UCB. The findings may pave a way for having UCB as a source for HSPC for clinical use in the future.


Asunto(s)
Enfermedad Injerto contra Huésped/terapia , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Nanotubos de Carbono/química , Animales , Diferenciación Celular/efectos de los fármacos , Citocinas/metabolismo , Sangre Fetal/efectos de los fármacos , Enfermedad Injerto contra Huésped/patología , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanotubos de Carbono/efectos adversos , Superóxidos/metabolismo , Trasplante Heterólogo
6.
Blood ; 117(15): 3974-82, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21325603

RESUMEN

Catalytic domain variants of activated factor VII (FVIIa) with enhanced hemostatic properties are highly attractive for the treatment of bleeding disorders via gene-based therapy. To explore this in a hemophilic mouse model, we characterized 2 variants of murine activated FVII (mFVIIa-VEAY and mFVIIa-DVQ) with modified catalytic domains, based on recombinant human FVIIa (rhFVIIa) variants. Using purified recombinant proteins, we showed that murine FVIIa (mFVIIa) and variants had comparable binding to human and murine tissue factor (TF) and exhibited similar extrinsic coagulant activity. In vitro in the absence of TF, the variants showed a 6- to 17-fold enhanced proteolytic and coagulant activity relative to mFVIIa, but increased inactivation by antithrombin. Gene delivery of mFVIIa-VEAY resulted in long-term, effective hemostasis at 5-fold lower expression levels relative to mFVIIa in hemophilia A mice or in hemophilia B mice with inhibitors to factor IX. However, expression of mFVIIa-VEAY at 14-fold higher than therapeutic levels resulted in a progressive mortality to 70% within 6 weeks after gene delivery. These results are the first demonstration of the hemostatic efficacy of continuous expression, in the presence or absence of inhibitors, of a high-activity gene-based FVIIa variant in an animal model of hemophilia.


Asunto(s)
Dominio Catalítico/genética , Factor VIIa/genética , Terapia Genética/métodos , Hemofilia A/terapia , Hemostasis/fisiología , Animales , Línea Celular , Dependovirus/genética , Modelos Animales de Enfermedad , Factor VIIa/química , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/mortalidad , Hemofilia A/sangre , Hemofilia A/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Plásmidos/genética , Estructura Terciaria de Proteína
7.
Nanomedicine ; 9(8): 1304-16, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23732300

RESUMEN

In this study, carboxylic acid functionalized single walled carbon nanotubes (f-SWCNT-COOH) was shown to support the viability and ex vivo expansion of freeze-thawed, non-enriched hematopoietic stem and progenitor cells (HSPC) in human umbilical cord blood-mononucleated cells (UCB-MNC). Our in vitro experiments showed that f-SWCNT-COOH increased the viability of the CD45(+) cells even without cytokine stimulation. It also reduced mitochondrial superoxides and caspase activity in CD45(+) cells. f-SWCNT-COOH drastically reduced the proportions of CD45(-) cells in the non-enriched UCB-MNC. Phenotypic expression analysis and functional colony forming units (CFU) showed significant ex vivo expansion of HSPC, particularly of CD45(+)CD34(+)CD38(-) population and granulocyte-macrophage (GM) colonies, in f-SWCNT-COOH augmented cultures supplemented with basal cytokines. In vivo data suggested that f-SWCNT-COOH expanded UCB-MNC could repopulate immunodeficient mice models with minimal acute or sub-acute symptoms of graft-versus-host disease (GVHD) and f-SWCNT-COOH dependent toxicity. FROM THE CLINICAL EDITOR: In this paper a novel method is presented by using single wall functionalized carbon nanotubes to enhance viability and ex vivo expansion of freeze-thawed, non-enriched hematopoietic stem and progenitor cells in human umbilical cord blood -mononucleated cells. Detailed data is presented about enhanced viability, including improved repopulation of immunodeficient mice models with minimal acute or sub-acute symptoms of graft-versus-host disease.


Asunto(s)
Sangre Fetal/citología , Sangre Fetal/trasplante , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Nanotubos de Carbono/química , ADP-Ribosil Ciclasa 1/análisis , Animales , Antígenos CD34/análisis , Ácidos Carboxílicos/química , Técnicas de Cultivo de Célula/métodos , Supervivencia Celular , Congelación , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Antígenos Comunes de Leucocito/análisis , Ratones , Ratones SCID
8.
PLoS Genet ; 6(3): e1000878, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20300656

RESUMEN

Coordinated cell migration during development is crucial for morphogenesis and largely relies on cells of the neural crest lineage that migrate over long distances to give rise to organs and tissues throughout the body. Recent studies of protein arginylation implicated this poorly understood posttranslational modification in the functioning of actin cytoskeleton and in cell migration in culture. Knockout of arginyltransferase (Ate1) in mice leads to embryonic lethality and severe heart defects that are reminiscent of cell migration-dependent phenotypes seen in other mouse models. To test the hypothesis that arginylation regulates cell migration during morphogenesis, we produced Wnt1-Cre Ate1 conditional knockout mice (Wnt1-Ate1), with Ate1 deletion in the neural crest cells driven by Wnt1 promoter. Wnt1-Ate1 mice die at birth and in the first 2-3 weeks after birth with severe breathing problems and with growth and behavioral retardation. Wnt1-Ate1 pups have prominent defects, including short palate and altered opening to the nasopharynx, and cranial defects that likely contribute to the abnormal breathing and early death. Analysis of neural crest cell movement patterns in situ and cell motility in culture shows an overall delay in the migration of Ate1 knockout cells that is likely regulated by intracellular mechanisms rather than extracellular signaling events. Taken together, our data suggest that arginylation plays a general role in the migration of the neural crest cells in development by regulating the molecular machinery that underlies cell migration through tissues and organs during morphogenesis.


Asunto(s)
Arginina/metabolismo , Movimiento Celular , Crecimiento y Desarrollo , Cresta Neural/patología , Aminoaciltransferasas/metabolismo , Animales , Animales Recién Nacidos , Huesos/anomalías , Huesos/enzimología , Huesos/patología , Adhesión Celular , Células Cultivadas , Técnicas de Cocultivo , Anomalías Craneofaciales/enzimología , Anomalías Craneofaciales/patología , Técnicas de Inactivación de Genes , Mesodermo/enzimología , Mesodermo/patología , Ratones , Ratones Noqueados , Modelos Biológicos , Cresta Neural/crecimiento & desarrollo , Hueso Paladar/anomalías , Hueso Paladar/enzimología , Hueso Paladar/patología , Análisis de Supervivencia , Proteína Wnt1/metabolismo
9.
Elife ; 122023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37267120

RESUMEN

The reasons for poor healing of pressure injuries are poorly understood. Vascular ulcers are worsened by extracellular release of hemoglobin, so we examined the impact of myoglobin (Mb) iron in murine muscle pressure injuries (mPI). Tests used Mb-knockout or treatment with deferoxamine iron chelator (DFO). Unlike acute injuries from cardiotoxin, mPI regenerated poorly with a lack of viable immune cells, persistence of dead tissue (necro-slough), and abnormal deposition of iron. However, Mb-knockout or DFO-treated mPI displayed a reversal of the pathology: decreased tissue death, decreased iron deposition, decrease in markers of oxidative damage, and higher numbers of intact immune cells. Subsequently, DFO treatment improved myofiber regeneration and morphology. We conclude that myoglobin iron contributes to tissue death in mPI. Remarkably, a large fraction of muscle death in untreated mPI occurred later than, and was preventable by, DFO treatment, even though treatment started 12 hr after pressure was removed. This demonstrates an opportunity for post-pressure prevention to salvage tissue viability.


Asunto(s)
Hierro , Úlcera por Presión , Ratones , Humanos , Animales , Deferoxamina/farmacología , Mioglobina , Necrosis , Músculos
10.
J Mol Cell Cardiol ; 53(3): 333-41, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22626847

RESUMEN

Protein arginylation mediated by arginyltransferase (ATE1) is essential for heart formation during embryogenesis, however its cell-autonomous role in cardiomyocytes and the differentiated heart muscle has never been investigated. To address this question, we generated cardiac muscle-specific Ate1 knockout mice, in which Ate1 deletion was driven by α-myosin heavy chain promoter (αMHC-Ate1 mouse). These mice were initially viable, but developed severe cardiac contractility defects, dilated cardiomyopathy, and thrombosis over time, resulting in high rates of lethality after 6months of age. These symptoms were accompanied by severe ultrastructural defects in cardiac myofibrils, seen in the newborns and far preceding the onset of cardiomyopathy, suggesting that these defects were primary and likely underlay the development of the future heart defects. Several major sarcomeric proteins were arginylated in vivo. Moreover, Ate1 deletion in the hearts resulted in a significant reduction of active and passive myofibril forces, suggesting that arginylation is critical for both myofibril structural integrity and contractility. Thus, arginylation is essential for maintaining the heart function by regulation of the major myofibril proteins and myofibril forces, and its absence in the heart muscle leads to progressive heart failure through cardiomyocyte-specific defects.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Corazón/fisiología , Miofibrillas/metabolismo , Aminoaciltransferasas/genética , Aminoaciltransferasas/metabolismo , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/prevención & control , Genes Letales , Ratones , Ratones Noqueados , Contracción Miocárdica/genética , Miocardio/metabolismo , Miocardio/ultraestructura , Miofibrillas/fisiología , Sarcómeros/metabolismo
11.
Biol Blood Marrow Transplant ; 18(5): 674-82, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22240732

RESUMEN

Ex vivo expansion of cord blood (CB) hematopoietic stem cells and cotransplantation of 2 CB units (CBUs) could enhance the applicability of CB transplantation in adult patients. We report an immunodeficient mouse model for cotransplantation of ex vivo expanded and unexpanded human CB, showing enhanced CB engraftment and provide proof of concept for this transplantation strategy as a means of overcoming the limiting cell numbers in each CBU. CBUs were expanded in serum-free medium supplemented with stem cell factor, Flt-3 ligand, thrombopoietin, and insulin growth factor binding protein-2 together with mesenchymal stromal cell coculture. Unexpanded and expanded CB cells were cotransplanted by tail vein injection into 45 sublethally irradiated nonobese diabetic SCID-IL2γ(-/-) (NSG) mice. Submandibular bleeding was performed monthly, and mice were sacrificed 4 months after transplantation to analyze for human hematopoietic engraftment. Expansion of non-CD34(+) selected CB cells yielded 40-fold expansion of CD34(+) cells and 3.1-fold expansion of hematopoietic stem cells based on limiting dilution analysis of NSG engraftment. Mice receiving expanded grafts exhibited 4.30% human cell repopulation, compared with 0.92% in mice receiving only unexpanded grafts at equivalent starting cell doses, even though the unexpanded graft predominated in long-term hematopoiesis (P = .07). Ex vivo expanded grafts with lower initiating cell doses also showed equivalent engraftment to unexpanded grafts with higher cell dose (8.0% versus 7.9%; P = .93). In conclusion, ex vivo expansion resulted in enhanced CB engraftment despite eventual rejection by the unexpanded CBU.


Asunto(s)
Sangre Fetal/trasplante , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Animales , Antígenos CD34/biosíntesis , Antígenos CD34/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Sangre Fetal/citología , Sangre Fetal/inmunología , Supervivencia de Injerto/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Inyecciones Intravenosas , Proteínas de la Membrana/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones SCID , Factor de Células Madre/farmacología , Trombopoyetina/farmacología , Trasplante Heterólogo , Irradiación Corporal Total
12.
Blood ; 115(3): 510-8, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965654

RESUMEN

Proper thymocyte development is required to establish T-cell central tolerance and to generate naive T cells, both of which are essential for T-cell homeostasis and a functional immune system. Here we demonstrate that the loss of transcription factor Foxp1 results in the abnormal development of T cells. Instead of generating naive T cells, Foxp1-deficient single-positive thymocytes acquire an activated phenotype prematurely in the thymus and lead to the generation of peripheral CD4(+) T and CD8(+) T cells that exhibit an activated phenotype and increased apoptosis and readily produce cytokines upon T-cell receptor engagement. These results identify Foxp1 as an essential transcriptional regulator for thymocyte development and the generation of quiescent naive T cells.


Asunto(s)
Diferenciación Celular/genética , Factores de Transcripción Forkhead/fisiología , Proteínas Represoras/fisiología , Linfocitos T/fisiología , Timo/fisiología , Animales , Apoptosis/genética , Apoptosis/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Proteínas Represoras/genética , Linfocitos T/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología
13.
Mol Ther ; 19(9): 1727-36, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21587210

RESUMEN

Vaccines that aim to expand tumor-specific CD8(+) T cells have yielded disappointing results in cancer patients although they showed efficacy in transplantable tumor mouse models. Using a system that more faithfully mimics a progressing cancer and its immunoinhibitory microenvironment, we here show that in transgenic mice, which gradually develop adenocarcinomas due to expression of HPV-16 E7 within their thyroid, a highly immunogenic vaccine expressing E7 only induces low E7-specific CD8(+) T-cell responses, which fail to affect the size of the tumors. In contrast, the same type of vaccine expressing E7 fused to herpes simplex virus (HSV)-1 glycoprotein D (gD), an antagonist of the coinhibitory B- and T-lymphocyte attenuator (BTLA)/CD160-herpes virus entry mediator (HVEM) pathways, stimulates potent E7-specific CD8(+) T-cell responses, which can be augmented by repeated vaccination, resulting in initial regression of even large tumor masses in all mice with sustained regression in more than half of them. These results indicate that active immunization concomitantly with blockade of the immunoinhibitory HVEM-BTLA/CD160 pathways through HSV-1 gD may result in sustained tumor regression.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia Activa/métodos , Neoplasias/terapia , Proteínas E7 de Papillomavirus/inmunología , Animales , Animales Modificados Genéticamente , Biomarcadores , Linfocitos T CD8-positivos/metabolismo , Regulación de la Expresión Génica , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/inmunología , Neoplasias/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Glándula Tiroides/inmunología , Vacunación/métodos
14.
J Clin Invest ; 118(5): 1825-34, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18398505

RESUMEN

Intravenous infusion of recombinant human activated Factor VII (FVIIa) has been used for over a decade in the successful management of bleeding episodes in patients with inhibitory antibodies to Factor VIII or Factor IX. Previously, we showed that expression of murine FVIIa (mFVIIa) from an adeno-associated viral (AAV) vector corrected abnormal hemostatic parameters in hemophilia B mice. To pursue this as a therapeutic approach, we sought to define safe and effective levels of FVIIa for continuous expression. In mice transgenic for mFVIIa or injected with AAV-mFVIIa, we analyzed survival, expression levels, in vitro and in vivo coagulation tests, and histopathology for up to 16 months after birth/mFVIIa expression. We found that continuous expression of mFVIIa at levels at or below 1.5 microg/ml was safe, effective, and compatible with a normal lifespan. However, expression levels of 2 microg/ml or higher were associated with thrombosis and early mortality, with pathologic findings in the heart and lungs that were rescued in a low-factor X (low-FX) mouse background, suggesting a FX-mediated effect. The findings from these mouse models of continuous FVIIa expression have implications for the development of a safe gene transfer approach for hemophilia and are consistent with the possibility of thromboembolic risk of continuously elevated FVIIa levels.


Asunto(s)
Factor VIIa , Hemofilia B , Hemostáticos , Animales , Pruebas de Coagulación Sanguínea , Factor VIIa/genética , Factor VIIa/metabolismo , Factor VIIa/uso terapéutico , Factor VIIa/toxicidad , Hemofilia B/tratamiento farmacológico , Hemofilia B/mortalidad , Hemostasis/fisiología , Hemostáticos/metabolismo , Hemostáticos/uso terapéutico , Hemostáticos/toxicidad , Humanos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microcirculación/fisiología , Miocardio/metabolismo , Miocardio/patología , Tasa de Supervivencia , Resultado del Tratamiento
15.
J Allergy Clin Immunol ; 124(5): 1088-98, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19895996

RESUMEN

BACKGROUND: Lymphocyte cytosolic protein 2, also known as Src homology 2 domain-containing leukocyte phosphoprotein of 76 kilodaltons (SLP-76), is an essential adaptor molecule in myeloid cells, where it regulates FcepsilonRI-induced mast cell (MC) and FcgammaR- and integrin-induced neutrophil (polymorphonuclear leukocyte [PMN]) functions. SLP-76 contains 3 N-terminal tyrosines at residues 112, 128, and 145 that together are critical for its function. OBJECTIVE: We sought to explore the relative importance of tyrosines 112, 128, and 145 of SLP-76 during MC and PMN activation. METHODS: We examined in vitro MC and PMN functions using cells isolated from knock-in mice harboring phenylalanine substitution mutations at tyrosines 112 and 128 (Y112/128F) or 145 (Y145F). We also examined the effects of these mutations on in vivo MC and PMN activation using models of anaphylaxis, dermal inflammation, and serum-induced arthritis. RESULTS: Mutations at Y112/Y128 and Y145 both interfered with SLP-76 activity; however, Y145F had a greater effect than Y112/128F on most in vitro FcR-induced functions. In vitro functional defects were recapitulated in vivo, where mice expressing Y145F exhibited greater attenuation of MC-dependent passive systemic anaphylaxis and PMN-mediated inflammatory responses. Notably, the Y145F mutation completely protected mice against development of joint-specific inflammation in the MC and PMN-dependent K/B x N model of arthritis. CONCLUSION: Our data indicate that Y145 is the most critical tyrosine supporting SLP-76 function in myeloid cells. Future efforts to dissect how Y145 mediates SLP-76-dependent signaling in MCs and PMNs will increase our understanding of these lineages and provide insights into the treatment of allergy and inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Anafilaxia/genética , Artritis/genética , Dermatitis/inmunología , Fosfoproteínas/genética , Anafilaxia/inmunología , Animales , Artritis/inmunología , Artritis/patología , Dermatitis/genética , Integrinas/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Ratones , Ratones Mutantes , Mutación/genética , Mutación/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de IgE/inmunología , Transducción de Señal/inmunología , Tirosina/genética
16.
J Virol ; 82(17): 8431-41, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18562543

RESUMEN

Herpes simplex virus type 1 (HSV-1) produces oral lesions, encephalitis, keratitis, and severe infections in the immunocompromised host. HSV-1 is almost as common as HSV-2 in causing first episodes of genital herpes, a disease that is associated with an increased risk of human immunodeficiency virus acquisition and transmission. No approved vaccines are currently available to protect against HSV-1 or HSV-2 infection. We developed a novel HSV vaccine strategy that uses a replication-competent strain of HSV-1, NS-gEnull, which has a defect in anterograde and retrograde directional spread and cell-to-cell spread. Following scratch inoculation on the mouse flank, NS-gEnull replicated at the site of inoculation without causing disease. Importantly, the vaccine strain was not isolated from dorsal root ganglia (DRG). We used the flank model to challenge vaccinated mice and demonstrated that NS-gEnull was highly protective against wild-type HSV-1. The challenge virus replicated to low titers at the site of inoculation; therefore, the vaccine strain did not provide sterilizing immunity. Nevertheless, challenge by HSV-1 or HSV-2 resulted in less-severe disease at the inoculation site, and vaccinated mice were totally protected against zosteriform disease and death. After HSV-1 challenge, latent virus was recovered by DRG explant cocultures from <10% of vaccinated mice compared with 100% of mock-vaccinated mice. The vaccine provided protection against disease and death after intravaginal challenge and markedly lowered the titers of the challenge virus in the vagina. Therefore, the HSV-1 gEnull strain is an excellent candidate for further vaccine development.


Asunto(s)
Virus Defectuosos/inmunología , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Replicación Viral/fisiología , Adolescente , Animales , Niño , Femenino , Vacunas contra el Virus del Herpes Simple/efectos adversos , Herpesvirus Humano 1/aislamiento & purificación , Humanos , Inmunización Secundaria/métodos , Ratones , Ratones Endogámicos BALB C , Vacunas Atenuadas/inmunología
17.
J Ultrasound Med ; 28(6): 795-800, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19470820

RESUMEN

OBJECTIVE: This study was designed to determine whether the echogenicity of neoplastic tissues changed as a result of low-intensity insonation and whether such alterations were related to an anti-vascular effect. METHODS: In 21 mice, implanted melanomas were insonated at either 1, 2, or 3 MHz using low-intensity ultrasound (spatial-average temporal-average intensity, 2.1 W/cm(2)). B-mode (mean gray scale) and contrast-enhanced power Doppler (percentage area of flow) measurements were made on each tumor before and after therapy. RESULTS: There was an increase in the echogenicity of the tumors with the increase in the frequency of the therapy beam and an accompanying decrease in tumor vascularity. CONCLUSIONS: Although the mechanisms responsible for the echogenicity change are not fully understood, it appears that an increase in the tumor mean gray scale was, at least in part, related to tissue inhomogeneities formed after disruption of the tumor neovasculature.


Asunto(s)
Melanoma Experimental/diagnóstico por imagen , Terapia por Ultrasonido/métodos , Animales , Modelos Animales de Enfermedad , Femenino , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C3H , Trasplante de Neoplasias , Proyectos Piloto , Distribución Aleatoria , Programas Informáticos , Ultrasonografía Doppler en Color
18.
Cell Rep ; 26(12): 3231-3245.e9, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30893597

RESUMEN

Regeneration of injured human heart muscle is limited and an unmet clinical need. There are no methods for the reproducible generation of clinical-quality stem cell-derived cardiovascular progenitors (CVPs). We identified laminin-221 (LN-221) as the most likely expressed cardiac laminin. We produced it as human recombinant protein and showed that LN-221 promotes differentiation of pluripotent human embryonic stem cells (hESCs) toward cardiomyocyte lineage and downregulates pluripotency and teratoma-associated genes. We developed a chemically defined, xeno-free laminin-based differentiation protocol to generate CVPs. We show high reproducibility of the differentiation protocol using time-course bulk RNA sequencing developed from different hESC lines. Single-cell RNA sequencing of CVPs derived from hESC lines supported reproducibility and identified three main progenitor subpopulations. These CVPs were transplanted into myocardial infarction mice, where heart function was measured by echocardiogram and human heart muscle bundle formation was identified histologically. This method may provide clinical-quality cells for use in regenerative cardiology.


Asunto(s)
Laminina/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/patología , Miocitos Cardíacos/patología , Células Madre Pluripotentes/patología , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre
19.
Acad Radiol ; 15(9): 1133-41, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18692754

RESUMEN

RATIONALE AND OBJECTIVES: The goal was to determine whether the tumor vascular disrupting actions of low-intensity ultrasound were frequency dependent. MATERIALS AND METHODS: The effect of the frequency (1 MHz at 2.2 W/cm2 or 3 MHz at 2.4 W/cm2) of low-intensity ultrasound as a neovascular disrupting modality was investigated in 15 murine melanomas (K1735(22)) insonated for 3 minutes after the intravenous injection of a microbubble contrast agent (Definity). In contrast-enhanced power Doppler observations of each tumor (before and after treatment), measurements were made of the size of the area of the tumor that was perfused with blood containing the ultrasound contrast agent (percentage area of flow [PAF]), and the volume of contrast agent flowing through the unit volume of the tumor (color-weighted fractional area [CWFA]). During insonation of the tumor, the temperature was measured with a fine wire thermocouple in an additional eight mice. RESULTS: The antivascular action of low-intensity ultrasound was significantly enhanced (PAF by 64%; CWFA by 106%) when the tumor was treated with 3-MHz ultrasound rather than 1 MHz (analysis of variance: PAF, P=.02; CWFA, P=.04). The average rate of tumor temperature increase was 2.6+/-1.3 degrees C/min for 1 MHz and 5.0+/-1.7 degrees C/min for 3 MHz; these increases were significantly different (P=.04). CONCLUSIONS: Insonation of the tumor at a higher frequency amplified the heating of the neoplasm and led to greater disruption of the tumor vasculature; 3-MHz ultrasound was more efficacious than 1 MHz for antivascular cancer therapy.


Asunto(s)
Melanoma Experimental/terapia , Neovascularización Patológica/terapia , Terapia por Ultrasonido/métodos , Animales , Temperatura Corporal , Medios de Contraste , Femenino , Fluorocarburos , Ratones , Ratones Endogámicos C3H
20.
Dev Cell ; 46(6): 681-695.e5, 2018 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-30146480

RESUMEN

The gut absorbs dietary nutrients and provides a barrier to xenobiotics and microbiome metabolites. To cope with toxin exposures, the intestinal epithelium is one of the most rapidly proliferating tissues in the body. The stem cell niche supplies essential signaling factors including Wnt proteins secreted by subepithelial myofibroblasts. Unexpectedly, therapeutically effective doses of orally administered PORCN inhibitors that block all Wnt secretion do not affect intestinal homeostasis. We find that intestinal myofibroblasts are intrinsically resistant to multiple xenobiotics, including PORCN inhibitors and the anthracycline antibiotic doxorubicin. These myofibroblasts have high expression of a subset of drug transporters; knockout of Mrp1/Abcc1 enhances drug sensitivity. Tamoxifen administration to Rosa26CreERT2;mT/mG mice visually highlights the drug-resistant intestinal stromal compartment and identifies small populations of drug-resistant cells in lung, kidney, and pancreatic islets. Xenobiotic resistance of the Wnt-producing myofibroblasts can protect the intestinal stem cell niche in the face of an unpredictable environment.


Asunto(s)
Aciltransferasas/fisiología , Proliferación Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Mucosa Intestinal/efectos de los fármacos , Proteínas de la Membrana/fisiología , Miofibroblastos/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Aciltransferasas/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Hormonales/farmacología , Broncodilatadores/farmacología , Células Cultivadas , Doxorrubicina/farmacología , Femenino , Homeostasis , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Miofibroblastos/citología , Miofibroblastos/metabolismo , Propionatos/farmacología , Quinolinas/farmacología , Transducción de Señal , Tamoxifeno/farmacología , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA