Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 572(7769): 335-340, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31316208

RESUMEN

Lamin A/C (LMNA) is one of the most frequently mutated genes associated with dilated cardiomyopathy (DCM). DCM related to mutations in LMNA is a common inherited cardiomyopathy that is associated with systolic dysfunction and cardiac arrhythmias. Here we modelled the LMNA-related DCM in vitro using patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Electrophysiological studies showed that the mutant iPSC-CMs displayed aberrant calcium homeostasis that led to arrhythmias at the single-cell level. Mechanistically, we show that the platelet-derived growth factor (PDGF) signalling pathway is activated in mutant iPSC-CMs compared to isogenic control iPSC-CMs. Conversely, pharmacological and molecular inhibition of the PDGF signalling pathway ameliorated the arrhythmic phenotypes of mutant iPSC-CMs in vitro. Taken together, our findings suggest that the activation of the PDGF pathway contributes to the pathogenesis of LMNA-related DCM and point to PDGF receptor-ß (PDGFRB) as a potential therapeutic target.


Asunto(s)
Cardiomiopatía Dilatada/genética , Lamina Tipo A/genética , Mutación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Calcio/metabolismo , Células Cultivadas , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Haploinsuficiencia/genética , Homeostasis , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/patología , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Degradación de ARNm Mediada por Codón sin Sentido , ARN Mensajero/análisis , ARN Mensajero/genética , Análisis de la Célula Individual
2.
J Allergy Clin Immunol ; 149(3): 923-933.e6, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34902435

RESUMEN

BACKGROUND: Treatments for coronavirus disease 2019, which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are urgently needed but remain limited. SARS-CoV-2 infects cells through interactions of its spike (S) protein with angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) on host cells. Multiple cells and organs are targeted, particularly airway epithelial cells. OM-85, a standardized lysate of human airway bacteria with strong immunomodulating properties and an impeccable safety profile, is widely used to prevent recurrent respiratory infections. We found that airway OM-85 administration inhibits Ace2 and Tmprss2 transcription in the mouse lung, suggesting that OM-85 might hinder SARS-CoV-2/host cell interactions. OBJECTIVES: We sought to investigate whether and how OM-85 treatment protects nonhuman primate and human epithelial cells against SARS-CoV-2. METHODS: ACE2 and TMPRSS2 mRNA and protein expression, cell binding of SARS-CoV-2 S1 protein, cell entry of SARS-CoV-2 S protein-pseudotyped lentiviral particles, and SARS-CoV-2 cell infection were measured in kidney, lung, and intestinal epithelial cell lines, primary human bronchial epithelial cells, and ACE2-transfected HEK293T cells treated with OM-85 in vitro. RESULTS: OM-85 significantly downregulated ACE2 and TMPRSS2 transcription and surface ACE2 protein expression in epithelial cell lines and primary bronchial epithelial cells. OM-85 also strongly inhibited SARS-CoV-2 S1 protein binding to, SARS-CoV-2 S protein-pseudotyped lentivirus entry into, and SARS-CoV-2 infection of epithelial cells. These effects of OM-85 appeared to depend on SARS-CoV-2 receptor downregulation. CONCLUSIONS: OM-85 inhibits SARS-CoV-2 epithelial cell infection in vitro by downregulating SARS-CoV-2 receptor expression. Further studies are warranted to assess whether OM-85 may prevent and/or reduce the severity of coronavirus disease 2019.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , COVID-19/prevención & control , Extractos Celulares/administración & dosificación , Receptores Virales/antagonistas & inhibidores , Receptores Virales/inmunología , SARS-CoV-2/inmunología , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Animales , COVID-19/inmunología , COVID-19/virología , Células CACO-2 , Extractos Celulares/inmunología , Células Cultivadas , Chlorocebus aethiops , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Células HEK293 , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/inmunología , Humanos , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Serina Endopeptidasas/efectos de los fármacos , Serina Endopeptidasas/genética , Serina Endopeptidasas/inmunología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/inmunología , Células Vero
3.
J Mol Cell Cardiol ; 149: 115-122, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33010256

RESUMEN

The ability to reprogram human somatic cells into human induced pluripotent stem cells (hiPSCs) has enabled researchers to generate cell types in vitro that have the potential to faithfully recapitulate patient-specific disease processes and phenotypes. hiPSC-derived cardiomyocytes (hiPSC-CMs) offer the promise of in vitro patient- and disease-specific models for drug testing and the discovery of novel therapeutic approaches for treating cardiovascular diseases. While methods to differentiate hiPSCs into cardiomyocytes have been demonstrated, the heterogeneity and immaturity of these differentiated populations have restricted their potential in reproducing human disease and the associated target cell phenotypes. These barriers may be overcome through comprehensive single-cell characterization to dissect the rich heterogeneity of hiPSC-CMs and to study the source of varying cell fates. In this study, we optimized and validated a new Single-Cell Western method to assess protein expression in hiPSC-CMs. To better understand distinct subpopulations generated from cardiomyocyte differentiations and to track populations at single-cell resolution over time, we measured and quantified the expression of cardiomyocyte subtype-specific proteins (MLC2V and MLC2A) using Single-Cell Westerns. By understanding their heterogeneity through single-cell protein expression and quantification, we may improve upon current cardiomyocyte differentiation protocols, generate hiPSC-CMs that are more representative of in vivo derived cardiomyocytes for disease modeling, and utilize hiPSC-CMs for regenerative medicine purposes. Single-Cell Westerns provide a robust platform for protein expression analysis at single-cell resolution.


Asunto(s)
Western Blotting , Proteínas en la Dieta/metabolismo , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Análisis de la Célula Individual , Calibración , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo
4.
Nat Methods ; 11(8): 855-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24930130

RESUMEN

Existing methods for human induced pluripotent stem cell (hiPSC) cardiac differentiation are efficient but require complex, undefined medium constituents that hinder further elucidation of the molecular mechanisms of cardiomyogenesis. Using hiPSCs derived under chemically defined conditions on synthetic matrices, we systematically developed an optimized cardiac differentiation strategy, using a chemically defined medium consisting of just three components: the basal medium RPMI 1640, L-ascorbic acid 2-phosphate and rice-derived recombinant human albumin. Along with small molecule-based induction of differentiation, this protocol produced contractile sheets of up to 95% TNNT2(+) cardiomyocytes at a yield of up to 100 cardiomyocytes for every input pluripotent cell and was effective in 11 hiPSC lines tested. This chemically defined platform for cardiac specification of hiPSCs will allow the elucidation of cardiomyocyte macromolecular and metabolic requirements and will provide a minimal system for the study of maturation and subtype specification.


Asunto(s)
Miocitos Cardíacos/citología , Diferenciación Celular , Medios de Cultivo , Humanos , Células Madre Pluripotentes Inducidas/citología
5.
Circ Res ; 115(6): 556-66, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25015077

RESUMEN

RATIONALE: Viral myocarditis is a life-threatening illness that may lead to heart failure or cardiac arrhythmias. A major causative agent for viral myocarditis is the B3 strain of coxsackievirus, a positive-sense RNA enterovirus. However, human cardiac tissues are difficult to procure in sufficient enough quantities for studying the mechanisms of cardiac-specific viral infection. OBJECTIVE: This study examined whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) could be used to model the pathogenic processes of coxsackievirus-induced viral myocarditis and to screen antiviral therapeutics for efficacy. METHODS AND RESULTS: hiPSC-CMs were infected with a luciferase-expressing coxsackievirus B3 strain (CVB3-Luc). Brightfield microscopy, immunofluorescence, and calcium imaging were used to characterize virally infected hiPSC-CMs for alterations in cellular morphology and calcium handling. Viral proliferation in hiPSC-CMs was quantified using bioluminescence imaging. Antiviral compounds including interferonß1, ribavirin, pyrrolidine dithiocarbamate, and fluoxetine were tested for their capacity to abrogate CVB3-Luc proliferation in hiPSC-CMs in vitro. The ability of these compounds to reduce CVB3-Luc proliferation in hiPSC-CMs was consistent with reported drug effects in previous studies. Mechanistic analyses via gene expression profiling of hiPSC-CMs infected with CVB3-Luc revealed an activation of viral RNA and protein clearance pathways after interferonß1 treatment. CONCLUSIONS: This study demonstrates that hiPSC-CMs express the coxsackievirus and adenovirus receptor, are susceptible to coxsackievirus infection, and can be used to predict antiviral drug efficacy. Our results suggest that the hiPSC-CM/CVB3-Luc assay is a sensitive platform that can screen novel antiviral therapeutics for their effectiveness in a high-throughput fashion.


Asunto(s)
Antivirales/uso terapéutico , Enterovirus Humano B/aislamiento & purificación , Infecciones por Enterovirus/tratamiento farmacológico , Modelos Cardiovasculares , Miocarditis/tratamiento farmacológico , Miocitos Cardíacos/patología , Células Madre Pluripotentes/patología , Antivirales/farmacología , Calcio/metabolismo , Proliferación Celular , Células Cultivadas , Evaluación Preclínica de Medicamentos , Infecciones por Enterovirus/metabolismo , Humanos , Técnicas In Vitro , Miocarditis/metabolismo , Miocarditis/virología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/virología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/virología , ARN Viral/metabolismo , Resultado del Tratamiento
6.
Eur Heart J ; 36(13): 806-16, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25368203

RESUMEN

AIMS: High-fat diet-induced obesity (DIO) is a major contributor to type II diabetes and micro- and macro-vascular complications leading to peripheral vascular disease (PVD). Metabolic abnormalities of induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) from obese individuals could potentially limit their therapeutic efficacy for PVD. The aim of this study was to compare the function of iPSC-ECs from normal and DIO mice using comprehensive in vitro and in vivo assays. METHODS AND RESULTS: Six-week-old C57Bl/6 mice were fed with a normal or high-fat diet. At 24 weeks, iPSCs were generated from tail tip fibroblasts and differentiated into iPSC-ECs using a directed monolayer approach. In vitro functional analysis revealed that iPSC-ECs from DIO mice had significantly decreased capacity to form capillary-like networks, diminished migration, and lower proliferation. Microarray and ELISA confirmed elevated apoptotic, inflammatory, and oxidative stress pathways in DIO iPSC-ECs. Following hindlimb ischaemia, mice receiving intramuscular injections of DIO iPSC-ECs had significantly decreased reperfusion compared with mice injected with control healthy iPSC-ECs. Hindlimb sections revealed increased muscle atrophy and presence of inflammatory cells in mice receiving DIO iPSC-ECs. When pravastatin was co-administered to mice receiving DIO iPSC-ECs, a significant increase in reperfusion was observed; however, this beneficial effect was blunted by co-administration of the nitric oxide synthase inhibitor, N(ω)-nitro-l-arginine methyl ester. CONCLUSION: This is the first study to provide evidence that iPSC-ECs from DIO mice exhibit signs of endothelial dysfunction and have suboptimal efficacy following transplantation in a hindlimb ischaemia model. These findings may have important implications for future treatment of PVD using iPSC-ECs in the obese population.


Asunto(s)
Células Endoteliales/fisiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Células Madre Pluripotentes Inducidas/fisiología , Óxido Nítrico/fisiología , Obesidad/fisiopatología , Pravastatina/farmacología , Análisis de Varianza , Animales , Apoptosis/fisiología , Diferenciación Celular , Dieta Alta en Grasa , Inhibidores Enzimáticos/farmacología , Fibroblastos/fisiología , Miembro Posterior/irrigación sanguínea , Inyecciones Intramusculares , Isquemia/fisiopatología , Isquemia/prevención & control , Ratones Endogámicos C57BL , Músculo Esquelético , Enfermedades Musculares/prevención & control , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Distribución Aleatoria , Daño por Reperfusión/fisiopatología , Transducción de Señal
7.
Circ Res ; 112(12): 1613-23, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23743227

RESUMEN

High throughput sequencing technologies have become essential in studies on genomics, epigenomics, and transcriptomics. Although sequencing information has traditionally been elucidated using a low throughput technique called Sanger sequencing, high throughput sequencing technologies are capable of sequencing multiple DNA molecules in parallel, enabling hundreds of millions of DNA molecules to be sequenced at a time. This advantage allows high throughput sequencing to be used to create large data sets, generating more comprehensive insights into the cellular genomic and transcriptomic signatures of various diseases and developmental stages. Within high throughput sequencing technologies, whole exome sequencing can be used to identify novel variants and other mutations that may underlie many genetic cardiac disorders, whereas RNA sequencing can be used to analyze how the transcriptome changes. Chromatin immunoprecipitation sequencing and methylation sequencing can be used to identify epigenetic changes, whereas ribosome sequencing can be used to determine which mRNA transcripts are actively being translated. In this review, we will outline the differences in various sequencing modalities and examine the main sequencing platforms on the market in terms of their relative read depths, speeds, and costs. Finally, we will discuss the development of future sequencing platforms and how these new technologies may improve on current sequencing platforms. Ultimately, these sequencing technologies will be instrumental in further delineating how the cardiovascular system develops and how perturbations in DNA and RNA can lead to cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares/genética , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Inmunoprecipitación de Cromatina , Metilación de ADN , Difusión de Innovaciones , Epigénesis Genética , Perfilación de la Expresión Génica/tendencias , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento/tendencias , Humanos , Fenotipo , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
8.
Physiology (Bethesda) ; 28(3): 190-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23636264

RESUMEN

In the present review, we provide an overview of connexin expression during skin development and remodeling in wound healing, and reflect on how loss- or gain-of-function connexin mutations may change cellular phenotypes and lead to diseases of the skin. We also consider the therapeutic value of targeting connexins in wound healing.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Enfermedades de la Piel/metabolismo , Piel/metabolismo , Cicatrización de Heridas , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Conexinas/genética , Uniones Comunicantes/patología , Predisposición Genética a la Enfermedad , Humanos , Mutación , Fenotipo , Transducción de Señal , Piel/crecimiento & desarrollo , Piel/lesiones , Piel/patología , Enfermedades de la Piel/genética , Enfermedades de la Piel/patología
9.
J Biol Chem ; 287(34): 29184-93, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22753409

RESUMEN

Pannexin 1 (Panx1) is a channel-forming glycoprotein expressed in different cell types of mammalian skin. We examined the role of Panx1 in melanoma tumorigenesis and metastasis since qPCR and Western blots revealed that mouse melanocytes exhibited low levels of Panx1 while increased Panx1 expression was correlated with tumor cell aggressiveness in the isogenic melanoma cell lines (B16-F0, -F10, and -BL6). Panx1 shRNA knockdown (Panx1-KD) generated stable BL6 cell lines, with reduced dye uptake, that showed a marked increase in melanocyte-like cell characteristics including higher melanin production, decreased cell migration and enhanced formation of cellular projections. Western blotting and proteomic analyses using 2D-gel/mass spectroscopy identified vimentin and ß-catenin as two of the markers of malignant melanoma that were down-regulated in Panx1-KD cells. Xenograft Panx1-KD cells grown within the chorioallantoic membrane of avian embryos developed tumors that were significantly smaller than controls. Mouse-Alu qPCR of the excised avian embryonic organs revealed that tumor metastasis to the liver was significantly reduced upon Panx1 knockdown. These data suggest that while Panx1 is present in skin melanocytes it is up-regulated during melanoma tumor progression, and tumorigenesis can be inhibited by the knockdown of Panx1 raising the possibility that Panx1 may be a viable target for the treatment of melanoma.


Asunto(s)
Conexinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Melaninas/metabolismo , Melanocitos/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Conexinas/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Melaninas/genética , Melanocitos/patología , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Ratones , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Regulación hacia Arriba/genética , Vimentina/genética , Vimentina/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
10.
Physiol Rep ; 11(20): e15838, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37849042

RESUMEN

Cardiac ischemic reperfusion injury (IRI) is paradoxically instigated by reestablishing blood-flow to ischemic myocardium typically from a myocardial infarction (MI). Although revascularization following MI remains the standard of care, effective strategies remain limited to prevent or attenuate IRI. We hypothesized that epicardial placement of human placental amnion/chorion (HPAC) grafts will protect against IRI. Using a clinically relevant model of IRI, swine were subjected to 45 min percutaneous ischemia followed with (MI + HPAC, n = 3) or without (MI only, n = 3) HPAC. Cardiac function was assessed by echocardiography, and regional punch biopsies were collected 14 days post-operatively. A deep phenotyping approach was implemented by using histological interrogation and incorporating global proteomics and transcriptomics in nonischemic, ischemic, and border zone biopsies. Our results established HPAC limited the extent of cardiac injury by 50% (11.0 ± 2.0% vs. 22.0 ± 3.0%, p = 0.039) and preserved ejection fraction in HPAC-treated swine (46.8 ± 2.7% vs. 35.8 ± 4.5%, p = 0.014). We present comprehensive transcriptome and proteome profiles of infarct (IZ), border (BZ), and remote (RZ) zone punch biopsies from swine myocardium during the proliferative cardiac repair phase 14 days post-MI. Both HPAC-treated and untreated tissues showed regional dynamic responses, whereas only HPAC-treated IZ revealed active immune and extracellular matrix remodeling. Decreased endoplasmic reticulum (ER)-dependent protein secretion and increased antiapoptotic and anti-inflammatory responses were measured in HPAC-treated biopsies. We provide quantitative evidence HPAC reduced cardiac injury from MI in a preclinical swine model, establishing a potential new therapeutic strategy for IRI. Minimizing the impact of MI remains a central clinical challenge. We present a new strategy to attenuate post-MI cardiac injury using HPAC in a swine model of IRI. Placement of HPAC membrane on the heart following MI minimizes ischemic damage, preserves cardiac function, and promotes anti-inflammatory signaling pathways.


Asunto(s)
Lesiones Cardíacas , Infarto del Miocardio , Embarazo , Porcinos , Humanos , Femenino , Animales , Placenta/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Lesiones Cardíacas/tratamiento farmacológico , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Antiinflamatorios/uso terapéutico , Modelos Animales de Enfermedad
11.
Nat Cardiovasc Res ; 2(1): 76-95, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36950336

RESUMEN

Cardiac cell surface proteins are drug targets and useful biomarkers for discriminating among cellular phenotypes and disease states. Here we developed an analytical platform, CellSurfer, that enables quantitative cell surface proteome (surfaceome) profiling of cells present in limited quantities, and we apply it to isolated primary human heart cells. We report experimental evidence of surface localization and extracellular domains for 1,144 N-glycoproteins, including cell-type-restricted and region-restricted glycoproteins. We identified a surface protein specific for healthy cardiomyocytes, LSMEM2, and validated an anti-LSMEM2 monoclonal antibody for flow cytometry and imaging. Surfaceome comparisons among pluripotent stem cell derivatives and their primary counterparts highlighted important differences with direct implications for drug screening and disease modeling. Finally, 20% of cell surface proteins, including LSMEM2, were differentially abundant between failing and non-failing cardiomyocytes. These results represent a rich resource to advance development of cell type and organ-specific targets for drug delivery, disease modeling, immunophenotyping and in vivo imaging.

12.
Exp Dermatol ; 21(8): 612-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22775996

RESUMEN

Transient knock-down of the gap junction protein Cx43 by antisense and siRNA, or gap junction block with mimetic peptides, have been shown to enhance epidermal wound healing. However, patients with oculodentodigital dysplasia (ODDD) express mutant Cx43 that leads to a chronic reduction in gap junctional intercellular communication. To determine whether mutant Cx43 in keratinocytes would impact upon the wound healing process, we localized Cx43 in human and mouse skin tissue expressing mutant Cx43 and assessed the ability of primary keratinocytes derived from a mouse model of ODDD to proliferate, migrate and differentiate. In the epidermis from an ODDD patient and in the epidermis of mice expressing the G60S mutant or in keratinocytes obtained from mutant mice, Cx43 was frequently found within intracellular compartments and rarely localized to punctate sites of cell-cell apposition. Primary keratinocytes derived from G60S mutant mice proliferated faster but migrated similarly to keratinocytes derived from wild-type control mice. Keratinocytes derived from mutant mice expressed abundant Cx43 and higher levels of involucrin and loricrin under low calcium conditions. However, after calcium-induced differentiation, similar levels of Cx43, involucrin and loricrin were observed. Thus, we conclude that during wound healing, mutant Cx43 may enhance keratinocyte proliferation and promote early differentiation of keratinocytes.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Conexina 43/genética , Conexina 43/metabolismo , Hipoplasia del Esmalte Dental/genética , Asimetría Facial/genética , Queratinocitos/metabolismo , Queratinocitos/patología , Microftalmía/genética , Mutación/genética , Sindactilia/genética , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Biopsia , Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Hipoplasia del Esmalte Dental/metabolismo , Hipoplasia del Esmalte Dental/patología , Modelos Animales de Enfermedad , Cara/anomalías , Cara/patología , Asimetría Facial/metabolismo , Asimetría Facial/patología , Humanos , Uniones Intercelulares/metabolismo , Uniones Intercelulares/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Microftalmía/metabolismo , Microftalmía/patología , Precursores de Proteínas/metabolismo , Piel/patología , Sindactilia/metabolismo , Sindactilia/patología , Cicatrización de Heridas/fisiología
13.
Tissue Eng Part B Rev ; 28(4): 891-911, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34476988

RESUMEN

Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. The cardiomyocytes produced have become commonplace as cardiac physiology research models. Accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. Our review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. In this study, our review will examine important considerations in undertaking a cardiovascular tissue engineering project and will present, interpret, and summarize some of the recent advancements in this field. Throughout, we review different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs. Impact statement With advancements in cardiac differentiation protocols, the production of human induced pluripotent stem cell derived cardiomyocytes is becoming cost effective and routine in the laboratory setting. Monolayer based culture methods are rapidly being replaced by three-dimensional (3D) tissue engineered constructs, which are more representative of the heart geometry. In the review presented, we delve into important concepts and tissue engineering principles that should be considered when generating 3D cardiac constructs, interpreting data acquired from, and embarking on a 3D cardiac tissue-based research project.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Evaluación Preclínica de Medicamentos , Humanos , Miocitos Cardíacos , Ingeniería de Tejidos/métodos
14.
Stem Cell Res ; 65: 102944, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36257093

RESUMEN

Pontocerebellar Hypoplasia 1B (PCH1B) is a severe autosomal recessive neurological disorder that is associated with mutations in the exosome complex component RRP40 (EXOSC3) gene. We generated and characterized an iPSC line from an individual with PCH1B that harbors a recessive homozygous c.395 A > C mutation in EXOSC3 and a family matched control from the probands unaffected mother. Each iPSC line presents with normal morphology and karyotype and express high levels of pluripotent markers. UAZTi009-A and UAZTi011-A are capable of directed differentiation and can be used as a vital experimental tool to study the development of PCH1B.


Asunto(s)
Complejo Multienzimático de Ribonucleasas del Exosoma , Proteínas de Unión al ARN , Humanos , Mutación/genética , Células Madre Pluripotentes Inducidas , Línea Celular
15.
Cells ; 11(21)2022 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-36359740

RESUMEN

(1) Background: Cushing's disease (CD) is a serious endocrine disorder caused by an adrenocorticotropic hormone (ACTH)-secreting pituitary neuroendocrine tumor (PitNET) that stimulates the adrenal glands to overproduce cortisol. Chronic exposure to excess cortisol has detrimental effects on health, including increased stroke rates, diabetes, obesity, cognitive impairment, anxiety, depression, and death. The first-line treatment for CD is pituitary surgery. Current surgical remission rates reported in only 56% of patients depending on several criteria. The lack of specificity, poor tolerability, and low efficacy of the subsequent second-line medical therapies make CD a medical therapeutic challenge. One major limitation that hinders the development of specific medical therapies is the lack of relevant human model systems that recapitulate the cellular composition of PitNET microenvironment. (2) Methods: human pituitary tumor tissue was harvested during transsphenoidal surgery from CD patients to generate organoids (hPITOs). (3) Results: hPITOs generated from corticotroph, lactotroph, gonadotroph, and somatotroph tumors exhibited morphological diversity among the organoid lines between individual patients and amongst subtypes. The similarity in cell lineages between the organoid line and the patient's tumor was validated by comparing the neuropathology report to the expression pattern of PitNET specific markers, using spectral flow cytometry and exome sequencing. A high-throughput drug screen demonstrated patient-specific drug responses of hPITOs amongst each tumor subtype. Generation of induced pluripotent stem cells (iPSCs) from a CD patient carrying germline mutation CDH23 exhibited dysregulated cell lineage commitment. (4) Conclusions: The human pituitary neuroendocrine tumor organoids represent a novel approach in how we model complex pathologies in CD patients, which will enable effective personalized medicine for these patients.


Asunto(s)
Tumores Neuroendocrinos , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT) , Neoplasias Hipofisarias , Humanos , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/tratamiento farmacológico , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/cirugía , Organoides , Tumores Neuroendocrinos/tratamiento farmacológico , Hidrocortisona , Microambiente Tumoral
16.
Eur J Hum Genet ; 30(4): 450-457, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35082396

RESUMEN

Dilated cardiomyopathy (DCM) is characterized by cardiac enlargement and impaired ventricular contractility leading to heart failure. A single report identified variants in leiomodin-2 (LMOD2) as a cause of neonatally-lethal DCM. Here, we describe two siblings with DCM who died shortly after birth due to heart failure. Exome sequencing identified a homozygous LMOD2 variant in both siblings, (GRCh38)chr7:g.123656237G > A; NM_207163.2:c.273 + 1G > A, ablating the donor 5' splice-site of intron-1. Pre-mRNA splicing studies and western blot analysis on cDNA derived from proband cardiac tissue, MyoD-transduced proband skin fibroblasts and HEK293 cells transfected with LMOD2 gene constructs established variant-associated absence of canonically spliced LMOD2 mRNA and full-length LMOD2 protein. Immunostaining of proband heart tissue unveiled abnormally short actin-thin filaments. Our data are consistent with LMOD2 c.273 + 1G > A abolishing/reducing LMOD2 transcript expression by: (1) variant-associated perturbation in initiation of transcription due to ablation of the intron-1 donor; and/or (2) degradation of aberrant LMOD2 transcripts (resulting from use of alternative transcription start-sites or cryptic splice-sites) by nonsense-mediated decay. LMOD2 expression is critical for life and the absence of LMOD2 is associated with thin filament shortening and severe cardiac contractile dysfunction. This study describes the first splice-site variant in LMOD2 and confirms the role of LMOD2 variants in DCM.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Cardiomiopatía Dilatada/genética , Células HEK293 , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Homocigoto , Humanos , Recién Nacido
17.
Hum Mutat ; 32(4): 456-66, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21305658

RESUMEN

Oculodentodigital dysplasia (ODDD) is primarily an autosomal dominant human disease caused by any one of over 60 mutations in the GJA1 gene encoding the gap junction protein Cx43. In the present study, wound healing was investigated in a G60S ODDD mutant mouse model and by using dermal fibroblasts isolated from two ODDD patients harboring the p.D3N and p.V216L mutants along with dermal fibroblasts isolated from their respective unaffected relatives. Punch biopsies revealed a delay in wound closure in the G60S mutant mice in comparison to wild-type littermates, and this delay appeared to be due to defects in the dermal fibroblasts. Although both the p.D3N and p.V216L mutants reduced gap junctional intercellular communication in human dermal fibroblasts, immunolocalization studies revealed that Cx43 gap junctions were prevalent at the cell surface of p.D3N expressing fibroblasts but greatly reduced in p.V216L expressing fibroblasts. Mutant expressing fibroblasts were further found to have reduced proliferation and migration capabilities. Finally, in response to TGFß1, mutant expressing fibroblasts expressed significantly less alpha smooth muscle actin suggesting they were inefficient in their ability to differentiate into myofibroblasts. Collectively, our results suggest that ODDD patients may have subclinical defects in wound healing due to impaired function of dermal fibroblasts.


Asunto(s)
Anomalías Múltiples/genética , Fibroblastos/patología , Cicatrización de Heridas/genética , Anomalías Múltiples/metabolismo , Animales , Conexina 43/genética , Conexina 43/metabolismo , Dermis/citología , Fibroblastos/metabolismo , Uniones Comunicantes , Humanos , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/metabolismo , Masculino , Ratones , Anomalías Dentarias/genética , Anomalías Dentarias/metabolismo
18.
J Biol Chem ; 285(12): 9147-60, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20086016

RESUMEN

Pannexins (Panx) are a class of integral membrane proteins that have been proposed to exhibit characteristics similar to those of connexin family members. In this study, we utilized Cx43-positive BICR-M1R(k) cells to stably express Panx1, Panx3, or Panx1-green fluorescent protein (GFP) to assess their trafficking, cell surface dynamics, and interplay with the cytoskeletal network. Expression of a Sar1 dominant negative mutant revealed that endoplasmic reticulum to Golgi transport of Panx1 and Panx3 was mediated via COPII-dependent vesicles. Distinct from Cx43-GFP, fluorescence recovery after photobleaching studies revealed that both Panx1-GFP and Panx3-GFP remained highly mobile at the cell surface. Unlike Cx43, Panx1-GFP exhibited no detectable interrelationship with microtubules. Conversely, cytochalasin B-induced disruption of microfilaments caused a severe loss of cell surface Panx1-GFP, a reduction in the recoverable fraction of Panx1-GFP that remained at the cell surface, and a decrease in Panx1-GFP vesicular transport. Furthermore, co-immunoprecipitation and co-sedimentation assays revealed actin as a novel binding partner of Panx1. Collectively, we conclude that although Panx1 and Panx3 share a common endoplasmic reticulum to Golgi secretory pathway to Cx43, their ultimate cell surface residency appears to be independent of cell contacts and the need for intact microtubules. Importantly, Panx1 has an interaction with actin microfilaments that regulates its cell surface localization and mobility.


Asunto(s)
Membrana Celular/metabolismo , Conexinas/biosíntesis , Citoesqueleto/metabolismo , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/biosíntesis , Citoesqueleto de Actina , Animales , Línea Celular , Línea Celular Tumoral , Conexina 43/metabolismo , Aparato de Golgi/metabolismo , Humanos , Ratones , Ratas
19.
Biochem J ; 429(3): 473-83, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20515445

RESUMEN

Although there are currently 62 mutants of Cx43 (connexin43) that can cause ODDD (oculodentodigital dysplasia), only two mutants have also been reported to cause palmar plantar hyperkeratosis. To determine how mutants of Cx43 can lead to this skin disease, REKs (rat epidermal keratinocytes) were engineered to express an ODDD-associated Cx43 mutant always linked to skin disease (fs260), an ODDD-linked Cx43 mutant which has been reported to sometimes cause skin disease (fs230), Cx43 mutants which cause ODDD only (G21R, G138R), a mouse Cx43 mutant linked to ODDD (G60S), a non-disease-linked truncated Cx43 mutant that is trapped in the endoplasmic reticulum (Delta244*) or full-length Cx43. When grown in organotypic cultures, of all the mutants investigated, only the fs260-expressing REKs consistently developed a thinner stratum corneum and expressed lower levels of Cx43, Cx26 and loricrin in comparison with REKs overexpressing wild-type Cx43. REKs expressing the fs260 mutant also developed a larger organotypic vital layer after acetone-induced injury and exhibited characteristics of parakeratosis. Collectively, our results suggest that the increased skin disease burden exhibited in ODDD patients harbouring the fs260 mutant is probably due to multiple additive effects cause by the mutant during epidermal differentiation.


Asunto(s)
Diferenciación Celular/genética , Conexina 43/fisiología , Queratinocitos/citología , Enfermedades de la Piel/genética , Animales , Secuencia de Bases , Células Cultivadas , Conexina 26 , Conexina 43/genética , Conexinas/metabolismo , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Proteínas de la Membrana/metabolismo , Mutagénesis Sitio-Dirigida , Fosforilación , Ratas , Enfermedades de la Piel/patología
20.
Cardiovasc Res ; 117(1): 188-200, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31995179

RESUMEN

AIMS: Heart failure is a major complication in cancer treatment due to the cardiotoxic effects of anticancer drugs, especially from the anthracyclines such as doxorubicin (DXR). DXR enhances oxidative stress and stimulates matrix metalloproteinase-2 (MMP-2) in cardiomyocytes. We investigated whether MMP inhibitors protect against DXR cardiotoxicity given the role of MMP-2 in proteolyzing sarcomeric proteins in the heart and remodelling the extracellular matrix. METHODS AND RESULTS: Eight-week-old male C57BL/6J mice were treated with DXR weekly with or without MMP inhibitors doxycycline or ONO-4817 by daily oral gavage for 4 weeks. Echocardiography was used to determine cardiac function and left ventricular remodelling before and after treatment. MMP inhibitors ameliorated DXR-induced systolic and diastolic dysfunction by reducing the loss in left ventricular ejection fraction, fractional shortening, and E'/A'. MMP inhibitors attenuated adverse left ventricular remodelling, reduced cardiomyocyte dropout, and prevented myocardial fibrosis. DXR increased myocardial MMP-2 activity in part also by upregulating N-terminal truncated MMP-2. Immunogold transmission electron microscopy showed that DXR elevated MMP-2 levels within the sarcomere and mitochondria which were associated with myofilament lysis, mitochondrial degeneration, and T-tubule distention. DXR-induced myofilament lysis was associated with increased titin proteolysis in the heart which was prevented by ONO-4817. DXR also increased the level and activity of MMP-2 in human embryonic stem cell-derived cardiomyocytes, which was reduced by ONO-4817. CONCLUSIONS: MMP-2 activation is an early event in DXR cardiotoxicity and contributes to myofilament lysis by proteolyzing cardiac titin. Two orally available MMP inhibitors ameliorated DXR cardiotoxicity by attenuating intracellular and extracellular matrix remodelling, suggesting their use may be a potential prophylactic strategy to prevent heart injury during chemotherapy.


Asunto(s)
Doxiciclina/farmacología , Matriz Extracelular/efectos de los fármacos , Cardiopatías/prevención & control , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Miocitos Cardíacos/efectos de los fármacos , Éteres Fenílicos/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiotoxicidad , Línea Celular , Modelos Animales de Enfermedad , Doxorrubicina , Matriz Extracelular/enzimología , Matriz Extracelular/patología , Fibrosis , Cardiopatías/inducido químicamente , Cardiopatías/enzimología , Cardiopatías/fisiopatología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/enzimología , Humanos , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/enzimología , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/ultraestructura , Proteínas Quinasas/metabolismo , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA