Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 47(6): 1182-1196.e10, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262351

RESUMEN

CD4+ T cells are tightly regulated by microbiota in the intestine, but whether intestinal T cells interface with host-derived metabolites is less clear. Here, we show that CD4+ T effector (Teff) cells upregulated the xenobiotic transporter, Mdr1, in the ileum to maintain homeostasis in the presence of bile acids. Whereas wild-type Teff cells upregulated Mdr1 in the ileum, those lacking Mdr1 displayed mucosal dysfunction and induced Crohn's disease-like ileitis following transfer into Rag1-/- hosts. Mdr1 mitigated oxidative stress and enforced homeostasis in Teff cells exposed to conjugated bile acids (CBAs), a class of liver-derived emulsifying agents that actively circulate through the ileal mucosa. Blocking ileal CBA reabsorption in transferred Rag1-/- mice restored Mdr1-deficient Teff cell homeostasis and attenuated ileitis. Further, a subset of ileal Crohn's disease patients displayed MDR1 loss of function. Together, these results suggest that coordinated interaction between mucosal Teff cells and CBAs in the ileum regulate intestinal immune homeostasis.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/inmunología , Ácidos y Sales Biliares/inmunología , Linfocitos T CD4-Positivos/inmunología , Enfermedad de Crohn/inmunología , Ileítis/inmunología , Mucosa Intestinal/inmunología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/deficiencia , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Acridinas/farmacología , Adulto , Animales , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/farmacología , Transporte Biológico , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/patología , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Homeostasis/inmunología , Humanos , Ileítis/genética , Ileítis/patología , Íleon/inmunología , Íleon/patología , Inmunidad Mucosa , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Estrés Oxidativo , Transducción de Señal , Tetrahidroisoquinolinas/farmacología
3.
EMBO Rep ; 21(10): e49332, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32875703

RESUMEN

Autotaxin (ATX) converts lysophosphatidylcholine and sphingosyl-phosphorylcholine into lysophosphatidic acid and sphingosine 1-phosphate, respectively. Despite the pivotal function of ATX in lipid metabolism, mechanisms by which ATX regulates immune and inflammatory disorders remain elusive. Here, using myeloid cell lineage-restricted Atx knockout mice, we show that Atx deficiency disrupts membrane microdomains and lipid rafts, resulting in the inhibition of Toll-like receptor 4 (TLR4) complex formation and the suppression of adaptor recruitment, thereby inhibiting TLR4-mediated responses in macrophages. Accordingly, TLR4-induced innate immune functions, including phagocytosis and iNOS expression, are attenuated in Atx-deficient macrophages. Consequently, Atx-/- mice exhibit a higher bacterial prevalence in the intestinal mucosa compared to controls. When combined with global Il10-/- mice, which show spontaneous colitis due to the translocation of luminal commensal microbes into the mucosa, myeloid cell lineage-restricted Atx knockout accelerates colitis development compared to control littermates. Collectively, our data reveal that Atx deficiency compromises innate immune responses, thereby promoting microbe-associated gut inflammation.


Asunto(s)
Colitis , Receptor Toll-Like 4 , Animales , Colitis/genética , Inmunidad , Inflamación/genética , Ratones , Ratones Noqueados , Receptor Toll-Like 4/genética
4.
Development ; 145(5)2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29519889

RESUMEN

The advent of human induced pluripotent stem cells (iPSCs) presents unprecedented opportunities to model human diseases. Differentiated cells derived from iPSCs in two-dimensional (2D) monolayers have proven to be a relatively simple tool for exploring disease pathogenesis and underlying mechanisms. In this Spotlight article, we discuss the progress and limitations of the current 2D iPSC disease-modeling platform, as well as recent advancements in the development of human iPSC models that mimic in vivo tissues and organs at the three-dimensional (3D) level. Recent bioengineering approaches have begun to combine different 3D organoid types into a single '4D multi-organ system'. We summarize the advantages of this approach and speculate on the future role of 4D multi-organ systems in human disease modeling.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Enfermedad , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Modelos Teóricos , Organoides/citología , Bioingeniería/instrumentación , Bioingeniería/métodos , Materiales Biomiméticos , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/tendencias , Diferenciación Celular , Matriz Extracelular/fisiología , Humanos , Células Madre Pluripotentes Inducidas/patología , Andamios del Tejido
5.
Circulation ; 139(21): 2451-2465, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30866650

RESUMEN

BACKGROUND: Molecular targeted chemotherapies have been shown to significantly improve the outcomes of patients who have cancer, but they often cause cardiovascular side effects that limit their use and impair patients' quality of life. Cardiac dysfunction induced by these therapies, especially trastuzumab, shows a distinct cardiotoxic clinical phenotype in comparison to the cardiotoxicity induced by conventional chemotherapies. METHODS: We used the human induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) platform to determine the underlying cellular mechanisms in trastuzumab-induced cardiac dysfunction. We assessed the effects of trastuzumab on structural and functional properties in iPSC-CMs from healthy individuals and performed RNA-sequencing to further examine the effect of trastuzumab on iPSC-CMs. We also generated human induced pluripotent stem cells from patients receiving trastuzumab and examined whether patients' phenotype could be recapitulated in vitro by using patient-specific iPSC-CMs. RESULTS: We found that clinically relevant doses of trastuzumab significantly impaired the contractile and calcium-handling properties of iPSC-CMs without inducing cardiomyocyte death or sarcomeric disorganization. RNA-sequencing and subsequent functional analysis revealed mitochondrial dysfunction and altered the cardiac energy metabolism pathway as primary causes of trastuzumab-induced cardiotoxic phenotype. Human iPSC-CMs generated from patients who received trastuzumab and experienced severe cardiac dysfunction were more vulnerable to trastuzumab treatment than iPSC-CMs generated from patients who did not experience cardiac dysfunction following trastuzumab therapy. It is important to note that metabolic modulation with AMP-activated protein kinase activators could avert the adverse effects induced by trastuzumab. CONCLUSIONS: Our results indicate that alterations in cellular metabolic pathways in cardiomyocytes could be a key mechanism underlying the development of cardiac dysfunction following trastuzumab therapy; therefore, targeting the altered metabolism may be a promising therapeutic approach for trastuzumab-induced cardiac dysfunction.


Asunto(s)
Antineoplásicos Inmunológicos/toxicidad , Neoplasias de la Mama/tratamiento farmacológico , Cardiopatías/inducido químicamente , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Trastuzumab/toxicidad , Proteínas Quinasas Activadas por AMP/metabolismo , Señalización del Calcio/efectos de los fármacos , Cardiotoxicidad , Estudios de Casos y Controles , Línea Celular , Metabolismo Energético/efectos de los fármacos , Femenino , Cardiopatías/metabolismo , Cardiopatías/patología , Cardiopatías/fisiopatología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Contracción Miocárdica/efectos de los fármacos , Fenotipo , Factores de Riesgo , Transcriptoma/efectos de los fármacos
6.
Circulation ; 139(6): 799-811, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30586709

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is frequently caused by mutations in myosin-binding protein C3 ( MYBPC3) resulting in a premature termination codon (PTC). The underlying mechanisms of how PTC mutations in MYBPC3 lead to the onset and progression of HCM are poorly understood. This study's aim was to investigate the molecular mechanisms underlying the pathogenesis of HCM associated with MYBPC3 PTC mutations by utilizing human isogenic induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). METHODS: Isogenic iPSC lines were generated from HCM patients harboring MYBPC3 PTC mutations (p.R943x; p.R1073P_Fsx4) using genome editing. Comprehensive phenotypic and transcriptome analyses were performed in the iPSC-CMs. RESULTS: We observed aberrant calcium handling properties with prolonged decay kinetics and elevated diastolic calcium levels in the absence of structural abnormalities or contracile dysfunction in HCM iPSC-CMs as compared to isogenic controls. The mRNA expression levels of MYBPC3 were significantly reduced in mutant iPSC-CMs, but the protein levels were comparable among isogenic iPSC-CMs, suggesting that haploinsufficiency of MYBPC3 does not contribute to the pathogenesis of HCM in vitro. Furthermore, truncated MYBPC3 peptides were not detected. At the molecular level, the nonsense-mediated decay pathway was activated, and a set of genes involved in major cardiac signaling pathways was dysregulated in HCM iPSC-CMs, indicating an HCM gene signature in vitro. Specific inhibition of the nonsense-mediated decay pathway in mutant iPSC-CMs resulted in reversal of the molecular phenotype and normalization of calcium-handling abnormalities. CONCLUSIONS: iPSC-CMs carrying MYBPC3 PTC mutations displayed aberrant calcium signaling and molecular dysregulations in the absence of significant haploinsufficiency of MYBPC3 protein. Here we provided the first evidence of the direct connection between the chronically activated nonsense-mediated decay pathway and HCM disease development.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Proteínas Portadoras/genética , Codón sin Sentido/genética , Mutación/genética , Miocitos Cardíacos/fisiología , Células Madre Pluripotentes/fisiología , ARN Mensajero/genética , Señalización del Calcio , Diferenciación Celular , Línea Celular , Progresión de la Enfermedad , Edición Génica , Perfilación de la Expresión Génica , Haploinsuficiencia , Humanos
7.
J Mol Cell Cardiol ; 133: 174-187, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31220468

RESUMEN

The mammalian heart undergoes complex structural and functional remodeling to compensate for stresses such as pressure overload. While studies suggest that, at best, the adult mammalian heart is capable of very limited regeneration arising from the proliferation of existing cardiomyocytes, how myocardial stress affects endogenous cardiac regeneration or repair is unknown. To define the relationship between left ventricular afterload and cardiac repair, we induced left ventricle pressure overload in adult mice by constriction of the ascending aorta (AAC). One week following AAC, we normalized ventricular afterload in a subset of animals through removal of the aortic constriction (de-AAC). Subsequent monitoring of cardiomyocyte cell cycle activity via thymidine analog labeling revealed that an acute increase in ventricular afterload induced cardiomyocyte proliferation. Intriguingly, a release in ventricular overload (de-AAC) further increases cardiomyocyte proliferation. Following both AAC and de-AAC, thymidine analog-positive cardiomyocytes exhibited characteristics of newly generated cardiomyocytes, including single diploid nuclei and reduced cell size as compared to age-matched, sham-operated adult mouse myocytes. Notably, those smaller cardiomyocytes frequently resided alongside one another, consistent with local stimulation of cellular proliferation. Collectively, our data demonstrate that adult cardiomyocyte proliferation can be locally stimulated by an acute increase or decrease of ventricular pressure, and this mode of stimulation can be harnessed to promote cardiac repair.


Asunto(s)
Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Presión Ventricular , Remodelación Ventricular , Animales , Biomarcadores , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Proliferación Celular , Modelos Animales de Enfermedad , Ecocardiografía , Técnica del Anticuerpo Fluorescente , Expresión Génica , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo
8.
Mol Ther ; 26(7): 1624-1634, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29699941

RESUMEN

Cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Regenerative therapy has been applied to restore lost cardiac muscle and cardiac performance. Induced pluripotent stem cells (iPSCs) can provide an unlimited source of cardiomyocytes and therefore play a key role in cardiac regeneration. Despite initial encouraging results from pre-clinical studies, progress toward clinical applications has been hampered by issues such as tumorigenesis, arrhythmogenesis, immune rejection, scalability, low graft-cell survival, and poor engraftment. Here, we review recent developments in iPSC research on regenerating injured heart tissue, including novel advances in cell therapy and potential strategies to overcome current obstacles in the field.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Humanos , Miocardio/citología , Regeneración/fisiología , Medicina Regenerativa/métodos
9.
BMC Gastroenterol ; 17(1): 63, 2017 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-28494754

RESUMEN

BACKGROUND: Cathelicidin (LL-37) is an antimicrobial peptide known to be associated with various autoimmune diseases. We attempt to determine if cathelicidin can accurately reflect IBD disease activity. We hypothesize that serum cathelicidin correlates with mucosal disease activity, stricture, and clinical prognosis of IBD patients. METHODS: Serum samples were collected from two separate cohorts of patients at the University of California, Los Angeles. Cohort 1 consisted of 50 control, 23 UC, and 28 CD patients. Cohort 2 consisted of 20 control, 57 UC, and 67 CD patients. LL-37 levels were determined by ELISA. Data from both cohorts were combined for calculation of accuracies in indicating mucosal disease activity, relative risks of stricture, and odds ratios of predicting disease development. RESULTS: Serum cathelicidin levels were inversely correlated with Partial Mayo Scores of UC patients and Harvey-Bradshaw Indices of CD patients. Among IBD patients with moderate or severe initial disease activity, the patients with high initial LL-37 levels had significantly better recovery than the patients with low initial LL-37 levels after 6-18 months, suggesting that high LL-37 levels correlate with good prognosis. Co-evaluation of LL-37 and CRP levels was more accurate than CRP alone or LL-37 alone in the correlation with Mayo Endoscopic Score of UC patients. Low LL-37 levels indicated a significantly elevated risk of intestinal stricture in CD patients. CONCLUSION: Co-evaluation of LL-37 and CRP can indicate mucosal disease activity in UC patients. LL-37 can predict future clinical activity in IBD patients and indicate risk of intestinal stricture in CD patients.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/sangre , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/complicaciones , Intestinos/patología , Adulto , Anciano , Proteína C-Reactiva/metabolismo , Colitis Ulcerosa/sangre , Colitis Ulcerosa/complicaciones , Colitis Ulcerosa/patología , Constricción Patológica/etiología , Enfermedad de Crohn/sangre , Enfermedad de Crohn/complicaciones , Enfermedad de Crohn/patología , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Catelicidinas
11.
Gastroenterology ; 149(4): 981-92.e11, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26055138

RESUMEN

BACKGROUND & AIMS: Persistent activation of the inflammatory response contributes to the development of inflammatory bowel diseases, which increase the risk of colorectal cancer. We aimed to identify microRNAs that regulate inflammation during the development of ulcerative colitis (UC) and progression to colitis-associated colon cancer (CAC). METHODS: We performed a quantitative polymerase chain reaction analysis to measure microRNAs in 401 colon specimens from patients with UC, Crohn's disease, irritable bowel syndrome, sporadic colorectal cancer, or CAC, as well as subjects without these disorders (controls); levels were correlated with clinical features and disease activity of patients. Colitis was induced in mice by administration of dextran sodium sulfate (DSS), and carcinogenesis was induced by addition of azoxymethane; some mice also were given an inhibitor of microRNA214 (miR214). RESULTS: A high-throughput functional screen of the human microRNAome found that miR214 regulated the activity of nuclear factor-κB. Higher levels of miR214 were detected in colon tissues from patients with active UC or CAC than from patients with other disorders or controls and correlated with disease progression. Bioinformatic and genome-wide profile analyses showed that miR214 activates an inflammatory response and is amplified through a feedback loop circuit mediated by phosphatase and tensin homolog (PTEN) and PDZ and LIM domain 2 (PDLIM2). Interleukin-6 induced signal transducer and activator of transcription 3 (STAT3)-mediated transcription of miR214. A miR214 chemical inhibitor blocked this circuit and reduced the severity of DSS-induced colitis in mice, as well as the number and size of tumors that formed in mice given azoxymethane and DSS. In fresh colonic biopsy specimens from patients with active UC, the miR214 inhibitor reduced inflammation by increasing levels of PDLIM2 and PTEN. CONCLUSIONS: Interleukin-6 up-regulates STAT3-mediated transcription of miR214 in colon tissues, which reduces levels of PDLIM2 and PTEN, increases phosphorylation of AKT, and activates nuclear factor-κB. The activity of this circuit correlates with disease activity in patients with UC and progression to colorectal cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Colitis Ulcerosa/prevención & control , Colon/metabolismo , Neoplasias del Colon/prevención & control , MicroARNs/metabolismo , Tratamiento con ARN de Interferencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Azoximetano , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Línea Celular , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/genética , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Proteínas con Dominio LIM/metabolismo , Ratones , MicroARNs/genética , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transcripción Genética , Transfección , Células Tumorales Cultivadas
12.
Gut ; 64(7): 1095-104, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25112884

RESUMEN

OBJECTIVE: Neurotensin (NT) mediates colonic inflammation through its receptor neurotensin receptor 1 (NTR1). NT stimulates miR-133α expression in colonic epithelial cells. We investigated the role of miR-133α in NT-associated colonic inflammation in vitro and in vivo. DESIGN: miR-133α and aftiphilin (AFTPH) levels were measured by quantitative PCR. Antisense (as)-miR-133α was administrated intracolonicaly prior to induction of 2, 4, 6-trinitrobenzene sulfonic acid (TNBS)-induced colitis and dextran sodium sulfate (DSS)-induced colitis. The effect of AFTPH was examined by gene silencing in vitro. RESULTS: NT increased miR-133α levels in NCM-460 overexpressing NTR1 (NCM460-NTR1) and HCT-116 cells. NT-induced p38, ERK1/2, c-Jun, and NF-κB activation, as well as IL-6, IL-8 and IL-1ß messenger RNA (mRNA) expression in NCM-460-NTR1 cells were reduced in miR-133α-silenced cells, while overexpression of miR-133α reversed these effects. MiR-133α levels were increased in TNBS (2 day) and DSS (5 day) colitis, while NTR1 deficient DSS-exposed mice had reduced miR-133α levels, compared to wild-type colitic mice. Intracolonic as-miR-133α attenuated several parameters of colitis as well expression of proinflammatory mediators in the colonic mucosa. In silico search coupled with qPCR identified AFTPH as a downstream target of miR-133α, while NT decreased AFTPH expression in NCM-460-NTR1 colonocytes. Gene silencing of AFTPH enhanced NT-induced proinflammatory responses and AFTPH levels were downregulated in experimental colitis. Levels of miR-133α were significantly upregulated, while AFTPH levels were downregulated in colonic biopsies of patients with ulcerative colitis compared to controls. CONCLUSIONS: NT-associated colitis and inflammatory signalling are regulated by miR-133α-AFTPH interactions. Targeting of miR-133α or AFTPH may represent a novel therapeutic approach in inflammatory bowel disease.


Asunto(s)
Colitis/fisiopatología , Colon/fisiología , Células Epiteliales/fisiología , Animales , Colon/citología , Células HCT116 , Humanos , Ratones , Ratones Noqueados , MicroARNs , FN-kappa B/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores de Neurotensina/genética , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
13.
Circ Res ; 112(1): 27-34, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23136123

RESUMEN

RATIONALE: After cardiac injury, cardiac progenitor cells are acutely reduced and are replenished in part by regulated self-renewal and proliferation, which occurs through symmetric and asymmetric cellular division. Understanding the molecular cues controlling progenitor cell self-renewal and lineage commitment is critical for harnessing these cells for therapeutic regeneration. We previously have found that the cell surface ATP-binding cassette G-subfamily transporter 2 (Abcg2) influences the proliferation of cardiac side population (CSP) progenitor cells, but through unclear mechanisms. OBJECTIVE: To determine the role of Abcg2 on cell cycle progression and mode of division in mouse CSP cells. METHODS AND RESULTS: Herein, using CSP cells isolated from wild-type and Abcg2 knockout mice, we found that Abcg2 regulates G1-S cell cycle transition by fluorescence ubiquitination cell cycle indicators, cell cycle-focused gene expression arrays, and confocal live-cell fluorescent microscopy. Moreover, we found that modulation of cell cycle results in transition from symmetric to asymmetric cellular division in CSP cells lacking Abcg2. CONCLUSIONS: Abcg2 modulates CSP cell cycle progression and asymmetric cell division, establishing a mechanistic link between this surface transporter and cardiac progenitor cell function. Greater understanding of progenitor cell biology and, in particular, the regulation of resident progenitor cell homeostasis is vital for guiding the future development of cell-based therapies for cardiac regeneration.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , División Celular Asimétrica , Puntos de Control del Ciclo Celular , Miocardio/metabolismo , Células de Población Lateral/metabolismo , Transducción de Señal , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/deficiencia , Transportadoras de Casetes de Unión a ATP/genética , Animales , División Celular Asimétrica/genética , Puntos de Control del Ciclo Celular/genética , Células Cultivadas , Citometría de Flujo , Puntos de Control de la Fase G1 del Ciclo Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Cinética , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Fluorescente , Microscopía por Video , Miocardio/citología , Interferencia de ARN , Transducción de Señal/genética , Imagen de Lapso de Tiempo , Transfección
14.
Circ Res ; 112(9): 1253-62, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23463815

RESUMEN

RATIONALE: Multiple progenitors derived from the heart and bone marrow (BM) have been used for cardiac repair. Despite this, not much is known about the molecular identity and relationship among these progenitors. To develop a robust stem cell therapy for the heart, it is critical to understand the molecular identity of the multiple cardiogenic progenitor cells. OBJECTIVE: This study is the first report of high-throughput transcriptional profiling of cardiogenic progenitor cells carried out on an identical platform. METHOD AND RESULTS: Microarray-based transcriptional profiling was carried out for 3 cardiac (ckit(+), Sca1(+), and side population) and 2 BM (ckit(+) and mesenchymal stem cell) progenitors, obtained from age- and sex-matched wild-type C57BL/6 mice. Analysis indicated that cardiac-derived ckit(+) population was very distinct from Sca1(+) and side population cells in the downregulation of genes encoding for cell-cell and cell-matrix adhesion proteins, and in the upregulation of developmental genes. Significant enrichment of transcripts involved in DNA replication and repair was observed in BM-derived progenitors. The BM ckit(+) cells seemed to have the least correlation with the other progenitors, with enrichment of immature neutrophil-specific molecules. CONCLUSIONS: Our study indicates that cardiac ckit(+) cells represent the most primitive population in the rodent heart. Primitive cells of cardiac versus BM origin differ significantly with respect to stemness and cardiac lineage-specific genes, and molecules involved in DNA replication and repair. The detailed molecular profile of progenitors reported here will serve as a useful reference to determine the molecular identity of progenitors used in future preclinical and clinical studies.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo , Animales , Antígenos Ly/metabolismo , Biomarcadores/metabolismo , Adhesión Celular/genética , Comunicación Celular/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Reparación del ADN/genética , Replicación del ADN/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Separación Inmunomagnética , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/genética
15.
Curr Opin Gastroenterol ; 29(4): 384-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23666365

RESUMEN

PURPOSE OF REVIEW: Stem cell therapy has emerged as a promising therapeutic strategy for inflammatory bowel diseases (IBDs). Currently, stem cell therapy is not part of the standard of care and is usually only performed as a part of clinical trials. In this review, clinical results, proposed underlying mechanisms, and future research directions will be discussed. RECENT FINDINGS: Administration of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) has been evaluated for IBD treatment over the past years. MSC therapy is being explored as a treatment option for fistulizing Crohn's disease and for luminal Crohn's disease. It is shown to be well tolerated, but results on efficacy are inconsistent. HSC transplantation seems to be very effective, but serious adverse events are common. Therefore, future research should focus on improving efficacy of MSC therapy, and on improvement of safety of HSC therapy. SUMMARY: Both MSC and HSC therapy offer clinical potential, but currently are not routinely used for IBD treatment. MSC therapy seems well tolerated but results on efficacy are conflicting. HSC transplantation is shown to be effective but safety concerns remain. Nonetheless, for severe refractory IBD cases, stem cell therapy could well become the next-generation treatment option.


Asunto(s)
Enfermedades Inflamatorias del Intestino/terapia , Trasplante de Células Madre/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/inmunología , Trasplante de Células Madre Mesenquimatosas/métodos , Trasplante de Células Madre/tendencias
16.
Circ Res ; 109(12): 1363-74, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22034491

RESUMEN

RATIONALE: Recent work in animal models and humans has demonstrated the presence of organ-specific progenitor cells required for the regenerative capacity of the adult heart. In response to tissue injury, progenitor cells differentiate into specialized cells, while their numbers are maintained through mechanisms of self-renewal. The molecular cues that dictate the self-renewal of adult progenitor cells in the heart, however, remain unclear. OBJECTIVE: We investigate the role of canonical Wnt signaling on adult cardiac side population (CSP) cells under physiological and disease conditions. METHODS AND RESULTS: CSP cells isolated from C57BL/6J mice were used to study the effects of canonical Wnt signaling on their proliferative capacity. The proliferative capacity of CSP cells was also tested after injection of recombinant Wnt3a protein (r-Wnt3a) in the left ventricular free wall. Wnt signaling was found to decrease the proliferation of adult CSP cells, both in vitro and in vivo, through suppression of cell cycle progression. Wnt stimulation exerted its antiproliferative effects through a previously unappreciated activation of insulin-like growth factor binding protein 3 (IGFBP3), which requires intact IGF binding site for its action. Moreover, injection of r-Wnt3a after myocardial infarction in mice showed that Wnt signaling limits CSP cell renewal, blocks endogenous cardiac regeneration and impairs cardiac performance, highlighting the importance of progenitor cells in maintaining tissue function after injury. CONCLUSIONS: Our study identifies canonical Wnt signaling and the novel downstream mediator, IGFBP3, as key regulators of adult cardiac progenitor self-renewal in physiological and pathological states.


Asunto(s)
Proliferación Celular , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Miocitos Cardíacos/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Proteínas Wnt/fisiología , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Proliferación Celular/efectos de los fármacos , Femenino , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Homeostasis/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Proteínas Recombinantes/farmacología , Células Madre/citología , Proteína Wnt3A/farmacología
17.
Cell Rep ; 40(9): 111264, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36044843

RESUMEN

As our closest living relatives, non-human primates uniquely enable explorations of human health, disease, development, and evolution. Considerable effort has thus been devoted to generating induced pluripotent stem cells (iPSCs) from multiple non-human primate species. Here, we establish improved culture methods for chimpanzee (Pan troglodytes) and pig-tailed macaque (Macaca nemestrina) iPSCs. Such iPSCs spontaneously differentiate in conventional culture conditions, but can be readily propagated by inhibiting endogenous WNT signaling. As a unique functional test of these iPSCs, we injected them into the pre-implantation embryos of another non-human species, rhesus macaques (Macaca mulatta). Ectopic expression of gene BCL2 enhances the survival and proliferation of chimpanzee and pig-tailed macaque iPSCs within the pre-implantation embryo, although the identity and long-term contribution of the transplanted cells warrants further investigation. In summary, we disclose transcriptomic and proteomic data, cell lines, and cell culture resources that may be broadly enabling for non-human primate iPSCs research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Pan troglodytes , Animales , Macaca mulatta , Macaca nemestrina/genética , Proteómica
18.
Circ Res ; 103(8): 825-35, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18787193

RESUMEN

Recently, the side population (SP) phenotype has been introduced as a reliable marker to identify subpopulations of cells with stem/progenitor cell properties in various tissues. We and others have identified SP cells from postmitotic tissues, including adult myocardium, in which they have been suggested to contribute to cellular regeneration following injury. SP cells are identified and characterized by a unique efflux of Hoechst 33342 dye. Abcg2 belongs to the ATP-binding cassette (ABC) transporter superfamily and constitutes the molecular basis for the dye efflux, hence the SP phenotype, in hematopoietic stem cells. Although Abcg2 is also expressed in cardiac SP (cSP) cells, its role in regulating the SP phenotype and function of cSP cells is unknown. Herein, we demonstrate that regulation of the SP phenotype in cSP cells occurs in a dynamic, age-dependent fashion, with Abcg2 as the molecular determinant of the cSP phenotype in the neonatal heart and another ABC transporter, Mdr1, as the main contributor to the SP phenotype in the adult heart. Using loss- and gain-of-function experiments, we find that Abcg2 tightly regulates cell fate and function. Adult cSP cells isolated from mice with genetic ablation of Abcg2 exhibit blunted proliferation capacity and augmented cell death. Conversely, overexpression of Abcg2 is sufficient to enhance cell proliferation, although with a limitation of cardiomyogenic differentiation. In summary, for the first time, we reveal a functional role for Abcg2 in modulating the proliferation, differentiation, and survival of adult cSP cells that goes beyond its distinct role in Hoechst dye efflux.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Miocardio/metabolismo , Células Madre/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Factores de Edad , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Bencimidazoles/metabolismo , Muerte Celular , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Colorantes Fluorescentes/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocardio/citología , Fenotipo , Transducción Genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
19.
J Am Coll Cardiol ; 72(1): 62-75, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29957233

RESUMEN

BACKGROUND: The long QT syndrome (LQTS) is an arrhythmogenic disorder of QT interval prolongation that predisposes patients to life-threatening ventricular arrhythmias such as Torsades de pointes and sudden cardiac death. Clinical genetic testing has emerged as the standard of care to identify genetic variants in patients suspected of having LQTS. However, these results are often confounded by the discovery of variants of uncertain significance (VUS), for which there is insufficient evidence of pathogenicity. OBJECTIVES: The purpose of this study was to demonstrate that genome editing of patient-specific induced pluripotent stem cells (iPSCs) can be a valuable approach to delineate the pathogenicity of VUS in cardiac channelopathy. METHODS: Peripheral blood mononuclear cells were isolated from a carrier with a novel missense variant (T983I) in the KCNH2 (LQT2) gene and an unrelated healthy control subject. iPSCs were generated using an integration-free Sendai virus and differentiated to iPSC-derived cardiomyocytes (CMs). RESULTS: Whole-cell patch clamp recordings revealed significant prolongation of the action potential duration (APD) and reduced rapidly activating delayed rectifier K+ current (IKr) density in VUS iPSC-CMs compared with healthy control iPSC-CMs. ICA-105574, a potent IKr activator, enhanced IKr magnitude and restored normal action potential duration in VUS iPSC-CMs. Notably, VUS iPSC-CMs exhibited greater propensity to proarrhythmia than healthy control cells in response to high-risk torsadogenic drugs (dofetilide, ibutilide, and azimilide), suggesting a compromised repolarization reserve. Finally, the selective correction of the causal variant in iPSC-CMs using CRISPR/Cas9 gene editing (isogenic control) normalized the aberrant cellular phenotype, whereas the introduction of the homozygous variant in healthy control cells recapitulated hallmark features of the LQTS disorder. CONCLUSIONS: The results suggest that the KCNH2T983I VUS may be classified as potentially pathogenic.


Asunto(s)
Canalopatías/genética , Canal de Potasio ERG1/genética , Edición Génica , Células Madre Pluripotentes Inducidas , Síndrome de QT Prolongado/genética , Adulto , Humanos , Masculino
20.
Circ Res ; 97(11): 1090-2, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16269652

RESUMEN

Tissue-specific progenitor cells contribute to local cellular regeneration and maintain organ function. Recently, we have determined that cardiac side-population (CSP) cells represent a distinct cardiac progenitor cell population, capable of in vitro differentiation into functional cardiomyocytes. The response of endogenous CSP to myocardial injury, however, and the cellular mechanisms that maintain this cardiac progenitor cell pool in vivo remain unknown. In this report we demonstrate that local progenitor cell proliferation maintains CSP under physiologic conditions, with little contribution from extracardiac stem cell sources. Following myocardial infarction in adult mice, however, CSP cells are acutely depleted, both within the infarct and noninfarct areas. CSP pools are subsequently reconstituted to baseline levels within 7 days after myocardial infarction, through both proliferation of resident CSP cells, as well as through homing of bone marrow-derived stem cells (BMC) to specific areas of myocardial injury and immunophenotypic conversion of BMC to adopt a CSP phenotype. We, therefore, conclude that following myocardial injury, cardiac progenitor cell populations are acutely depleted and are reconstituted to normal levels by both self-proliferation and selective homing of BMC. Understanding and enhancing such processes hold enormous potential for therapeutic myocardial regeneration.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Células Madre/citología , Animales , Proliferación Celular , Antígenos Comunes de Leucocito/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/terapia , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Regeneración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA