Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Oncogene ; 42(6): 434-448, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36509998

RESUMEN

Small cell lung cancer (SCLC) remains a lethal disease with a dismal overall survival rate of 6% despite promising responses to upfront combination chemotherapy. The key drivers of such rapid mortality include early metastatic dissemination in the natural course of the disease and the near guaranteed emergence of chemoresistant disease. Here, we found that we could model the regression and relapse seen in clinical SCLC in vitro. We utilized time-course resolved RNA-sequencing to globally profile transcriptome changes as SCLC cells responded to a combination of cisplatin and etoposide-the standard-of-care in SCLC. Comparisons across time points demonstrated a distinct transient transcriptional state resembling embryonic diapause. Differential gene expression analysis revealed that expression of the PEA3 transcription factors ETV4 and ETV5 were transiently upregulated in the surviving fraction of cells which we determined to be necessary for efficient clonogenic expansion following chemotherapy. The FGFR-PEA3 signaling axis guided the identification of a pan-FGFR inhibitor demonstrating in vitro and in vivo efficacy in delaying progression following combination chemotherapy, observed inhibition of phosphorylation of the FGFR adaptor FRS2 and corresponding downstream MAPK and PI3K-Akt signaling pathways. Taken together, these data nominate PEA3 transcription factors as key mediators of relapse progression in SCLC and identify a clinically actionable small molecule candidate for delaying relapse of SCLC.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Fosfatidilinositol 3-Quinasas/genética , Recurrencia Local de Neoplasia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Línea Celular Tumoral
2.
Methods Mol Biol ; 2386: 1-16, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34766261

RESUMEN

Immunohistochemistry (IHC) is a highly sensitive protein detection technique developed using the principle of antigen-antibody binding reaction. With immunohistochemistry, it is possible to visualize the abundance, distribution, and localization of proteins in situ. This chapter discusses the standard protocols involved in IHC detection using chromogenic substrates, including pre-treatment of tissues, types of chromogenic substrates, and troubleshooting at various stages of the protocol.


Asunto(s)
Inmunoquímica , Compuestos Cromogénicos , Inmunohistoquímica
3.
Nat Med ; 27(5): 806-814, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33958799

RESUMEN

Cystic fibrosis (CF) is a lethal autosomal recessive disorder that afflicts more than 70,000 people. People with CF experience multi-organ dysfunction resulting from aberrant electrolyte transport across polarized epithelia due to mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CF-related lung disease is by far the most important determinant of morbidity and mortality. Here we report results from a multi-institute consortium in which single-cell transcriptomics were applied to define disease-related changes by comparing the proximal airway of CF donors (n = 19) undergoing transplantation for end-stage lung disease with that of previously healthy lung donors (n = 19). Disease-dependent differences observed include an overabundance of epithelial cells transitioning to specialized ciliated and secretory cell subsets coupled with an unexpected decrease in cycling basal cells. Our study yields a molecular atlas of the proximal airway epithelium that will provide insights for the development of new targeted therapies for CF airway disease.


Asunto(s)
Fibrosis Quística/genética , Fibrosis Quística/patología , Células Epiteliales/citología , Pulmón/patología , Mucosa Respiratoria/patología , Diferenciación Celular/genética , Cilios/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/biosíntesis , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/patología , Perfilación de la Expresión Génica , Humanos , Análisis de la Célula Individual/métodos , Transcriptoma/genética
4.
Cell Stem Cell ; 27(6): 869-875.e4, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33259798

RESUMEN

Current smoking is associated with increased risk of severe COVID-19, but it is not clear how cigarette smoke (CS) exposure affects SARS-CoV-2 airway cell infection. We directly exposed air-liquid interface (ALI) cultures derived from primary human nonsmoker airway basal stem cells (ABSCs) to short term CS and then infected them with SARS-CoV-2. We found an increase in the number of infected airway cells after CS exposure with a lack of ABSC proliferation. Single-cell profiling of the cultures showed that the normal interferon response was reduced after CS exposure with infection. Treatment of CS-exposed ALI cultures with interferon ß-1 abrogated the viral infection, suggesting one potential mechanism for more severe viral infection. Our data show that acute CS exposure allows for more severe airway epithelial disease from SARS-CoV-2 by reducing the innate immune response and ABSC proliferation and has implications for disease spread and severity in people exposed to CS.


Asunto(s)
COVID-19/fisiopatología , Mucosa Respiratoria/fisiopatología , Fumar/efectos adversos , Células Madre/virología , COVID-19/genética , COVID-19/inmunología , COVID-19/terapia , Células Cultivadas , Regulación hacia Abajo , Humanos , Inmunidad Innata , Interferón beta/uso terapéutico , Gravedad del Paciente , Mucosa Respiratoria/virología
5.
bioRxiv ; 2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32766588

RESUMEN

Most demographic studies are now associating current smoking status with increased risk of severe COVID-19 and mortality from the disease but there remain many questions about how direct cigarette smoke exposure affects SARS-CoV-2 airway cell infection. We directly exposed mucociliary air-liquid interface (ALI) cultures derived from primary human nonsmoker airway basal stem cells (ABSCs) to short term cigarette smoke and infected them with live SARS-CoV-2. We found an increase in the number of infected airway cells after cigarette smoke exposure as well as an increased number of apoptotic cells. Cigarette smoke exposure alone caused airway injury that resulted in an increased number of ABSCs, which proliferate to repair the airway. But we found that acute SARS-CoV-2 infection or the combination of exposure to cigarette smoke and SARS-CoV-2 did not induce ABSC proliferation. We set out to examine the underlying mechanism governing the increased susceptibility of cigarette smoke exposed ALI to SARS-CoV-2 infection. Single cell profiling of the cultures showed that infected airway cells displayed a global reduction in gene expression across all airway cell types. Interestingly, interferon response genes were induced in SARS-CoV-2 infected airway epithelial cells in the ALI cultures but smoking exposure together with SARS-CoV-2 infection reduced the interferon response. Treatment of cigarette smoke-exposed ALI cultures with Interferon ß-1 abrogated the viral infection, suggesting that the lack of interferon response in the cigarette smoke-exposed ALI cultures allows for more severe viral infection and cell death. In summary, our data show that acute smoke exposure allows for more severe proximal airway epithelial disease from SARS-CoV-2 by reducing the mucosal innate immune response and ABSC proliferation and has implications for disease spread and severity in people exposed to cigarette smoke.

6.
Cell Stem Cell ; 27(3): 413-429.e4, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32721381

RESUMEN

Our understanding of dynamic interactions between airway basal stem cells (ABSCs) and their signaling niches in homeostasis, injury, and aging remains elusive. Using transgenic mice and pharmacologic studies, we found that Wnt/ß-catenin within ABSCs was essential for proliferation post-injury in vivo. ABSC-derived Wnt ligand production was dispensable for epithelial proliferation. Instead, the PDGFRα+ lineage in the intercartilaginous zone (ICZ) niche transiently secreted Wnt ligand necessary for ABSC proliferation. Strikingly, ABSC-derived Wnt ligand later drove early progenitor differentiation to ciliated cells. We discovered additional changes in aging, as glandular-like epithelial invaginations (GLEIs) derived from ABSCs emerged exclusively in the ICZ of aged mice and contributed to airway homeostasis and repair. Further, ABSC Wnt ligand secretion was necessary for GLEI formation, and constitutive activation of ß-catenin in young mice induced their formation in vivo. Collectively, these data underscore multiple spatiotemporally dynamic Wnt-secreting niches that regulate functionally distinct phases of airway regeneration and aging.


Asunto(s)
Células Madre , beta Catenina , Envejecimiento , Animales , Diferenciación Celular , Proliferación Celular , Ratones , Ratones Transgénicos , Células Madre/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
7.
Cell Rep ; 30(7): 2055-2064.e5, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32075752

RESUMEN

Mechanisms underpinning airway epithelial homeostatic maintenance and ways to prevent its dysregulation remain elusive. Herein, we identify that ß-catenin phosphorylated at Y489 (p-ß-cateninY489) emerges during human squamous lung cancer progression. This led us to develop a model of airway basal stem cell (ABSC) hyperproliferation by driving Wnt/ß-catenin signaling, resulting in a morphology that resembles premalignant lesions and loss of ciliated cell differentiation. To identify small molecules that could reverse this process, we performed a high-throughput drug screen for inhibitors of Wnt/ß-catenin signaling. Our studies unveil Wnt inhibitor compound 1 (WIC1), which suppresses T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) activity, reduces ABSC proliferation, induces ciliated cell differentiation, and decreases nuclear p-ß-cateninY489. Collectively, our work elucidates a dysregulated Wnt/p-ß-cateninY489 axis in lung premalignancy that can be modeled in vitro and identifies a Wnt/ß-catenin inhibitor that promotes airway homeostasis. WIC1 may therefore serve as a tool compound in regenerative medicine studies with implications for restoring normal airway homeostasis after injury.


Asunto(s)
Pulmón/efectos de los fármacos , Pulmón/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Vía de Señalización Wnt/efectos de los fármacos , Animales , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Bronquios/patología , Diferenciación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Femenino , Ensayos Analíticos de Alto Rendimiento/métodos , Homeostasis/efectos de los fármacos , Humanos , Pulmón/citología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Bibliotecas de Moléculas Pequeñas/farmacología , Células Madre/citología , Células Madre/patología , Transfección , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo
8.
Cell Rep ; 29(11): 3488-3505.e9, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31825831

RESUMEN

Progressive organ fibrosis accounts for one-third of all deaths worldwide, yet preclinical models that mimic the complex, progressive nature of the disease are lacking, and hence, there are no curative therapies. Progressive fibrosis across organs shares common cellular and molecular pathways involving chronic injury, inflammation, and aberrant repair resulting in deposition of extracellular matrix, organ remodeling, and ultimately organ failure. We describe the generation and characterization of an in vitro progressive fibrosis model that uses cell types derived from induced pluripotent stem cells. Our model produces endogenous activated transforming growth factor ß (TGF-ß) and contains activated fibroblastic aggregates that progressively increase in size and stiffness with activation of known fibrotic molecular and cellular changes. We used this model as a phenotypic drug discovery platform for modulators of fibrosis. We validated this platform by identifying a compound that promotes resolution of fibrosis in in vivo and ex vivo models of ocular and lung fibrosis.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Fibrosis Pulmonar/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular , Células Cultivadas , Descubrimiento de Drogas/métodos , Femenino , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta/metabolismo
9.
Stem Cell Res Ther ; 10(1): 52, 2019 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755264

RESUMEN

The original article [1] contains an error in the legend of Fig 5 whereby the descriptions for panels 5d and 5e are incorrect; as such, the corrected legend can be viewed below with its respective figure images.

10.
Stem Cell Res Ther ; 8(1): 217, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28969679

RESUMEN

BACKGROUND: Dysfunction of the retinal pigment epithelium (RPE) is implicated in numerous forms of retinal degeneration. The readily accessible environment of the eye makes it particularly suitable for the transplantation of RPE cells, which can now be derived from autologous induced pluripotent stem cells (iPSCs), to treat retinal degeneration. For RPE transplantation to become feasible in the clinic, patient-specific somatic cells should be reprogrammed to iPSCs without the introduction of reprogramming genes into the genome of the host cell, and then subsequently differentiated into RPE cells that are well characterized for safety and functionality prior to transplantation. METHODS: We have reprogrammed human dermal fibroblasts to iPSCs using nonintegrating RNA, and differentiated the iPSCs toward an RPE fate (iPSC-RPE), under Good Manufacturing Practice (GMP)-compatible conditions. RESULTS: Using highly sensitive assays for cell polarity, structure, organelle trafficking, and function, we found that iPSC-RPE cells in culture exhibited key characteristics of native RPE. Importantly, we demonstrate for the first time with any stem cell-derived RPE cell that live cells are able to support dynamic organelle transport. This highly sensitive test is critical for RPE cells intended for transplantation, since defects in intracellular motility have been shown to promote RPE pathogenesis akin to that found in macular degeneration. To test their capabilities for in-vivo transplantation, we injected the iPSC-RPE cells into the subretinal space of a mouse model of retinal degeneration, and demonstrated that the transplanted cells are capable of rescuing lost RPE function. CONCLUSIONS: This report documents the successful generation, under GMP-compatible conditions, of human iPSC-RPE cells that possess specific characteristics of healthy RPE. The report adds to a growing literature on the utility of human iPSC-RPE cells for cell culture investigations on pathogenicity and for therapeutic transplantation, by corroborating findings of others, and providing important new information on essential RPE cell biological properties.


Asunto(s)
Reprogramación Celular/genética , Virus de la Encefalitis Equina Venezolana/genética , Células Epiteliales/efectos de los fármacos , Fibroblastos/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Degeneración Retiniana/terapia , Animales , Diferenciación Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/metabolismo , Células Epiteliales/citología , Células Epiteliales/fisiología , Células Epiteliales/trasplante , Fibroblastos/citología , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Inyecciones Intraoculares , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Cultivo Primario de Células , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/fisiología , Piel/citología
11.
Cell Stem Cell ; 21(1): 1-3, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28686859

RESUMEN

Lung tissue can robustly regenerate functional alveolar units after injury, but the mechanisms are unknown. Lechner et al. (2017) in this issue of Cell Stem Cell demonstrate that lung regeneration is facilitated by bone-marrow-derived myeloid cells that are recruited to the lung through a CCL2-CCR2 chemokine axis and by IL-13 expressing innate lymphoid cells.


Asunto(s)
Quimiocina CCL2/inmunología , Lesión Pulmonar/inmunología , Pulmón/fisiología , Macrófagos Alveolares/inmunología , Receptores CCR2/inmunología , Regeneración/inmunología , Animales , Humanos , Interleucina-13/inmunología , Lesión Pulmonar/patología , Macrófagos Alveolares/patología
12.
Stem Cells Transl Med ; 6(2): 622-633, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28191779

RESUMEN

Stem cell technologies, especially patient-specific, induced stem cell pluripotency and directed differentiation, hold great promise for changing the landscape of medical therapies. Proper exploitation of these methods may lead to personalized organ transplants, but to regenerate organs, it is necessary to develop methods for assembling differentiated cells into functional, organ-level tissues. The generation of three-dimensional human tissue models also holds potential for medical advances in disease modeling, as full organ functionality may not be necessary to recapitulate disease pathophysiology. This is specifically true of lung diseases where animal models often do not recapitulate human disease. Here, we present a method for the generation of self-assembled human lung tissue and its potential for disease modeling and drug discovery for lung diseases characterized by progressive and irreversible scarring such as idiopathic pulmonary fibrosis (IPF). Tissue formation occurs because of the overlapping processes of cellular adhesion to multiple alveolar sac templates, bioreactor rotation, and cellular contraction. Addition of transforming growth factor-ß1 to single cell-type mesenchymal organoids resulted in morphologic scarring typical of that seen in IPF but not in two-dimensional IPF fibroblast cultures. Furthermore, this lung organoid may be modified to contain multiple lung cell types assembled into the correct anatomical location, thereby allowing cell-cell contact and recapitulating the lung microenvironment. Our bottom-up approach for synthesizing patient-specific lung tissue in a scalable system allows for the development of relevant human lung disease models with the potential for high throughput drug screening to identify targeted therapies. Stem Cells Translational Medicine 2017;6:622-633.


Asunto(s)
Técnicas de Cultivo de Célula , Fibroblastos/patología , Fibrosis Pulmonar Idiopática/patología , Células Madre Pluripotentes Inducidas/patología , Pulmón/patología , Organoides/patología , Ingeniería de Tejidos/métodos , Reactores Biológicos , Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Fibroblastos/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Pulmón/fisiopatología , Organoides/efectos de los fármacos , Fenotipo , Factores de Tiempo , Ingeniería de Tejidos/instrumentación , Factor de Crecimiento Transformador beta1/farmacología
13.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L186-L195, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27941077

RESUMEN

Bronchopulmonary dysplasia (BPD) is a common complication of premature birth. The histopathology of BPD is characterized by an arrest of alveolarization with fibroblast activation. The Wnt/ß-catenin signaling pathway is important in early lung development. When Wnt signaling is active, phosphorylation of ß-catenin by tyrosine kinases at activating sites, specifically at tyrosine 489 (Y489), correlates with nuclear localization of ß-catenin. We examined fetal lung tissue, lung tissue from term newborns, and lung tissue from infants who died with BPD; we found nuclear ß-catenin phosphorylation at Y489 in epithelial and mesenchymal cells in fetal tissue and BPD tissue, but not in the lungs of term infants. Using a 3D human organoid model, we found increased nuclear localization of ß-catenin phosphorylated at Y489 (p-ß-cateninY489) after exposure to alternating hypoxia and hyperoxia compared with organoids cultured in normoxia. Exogenous stimulation of the canonical Wnt pathway in organoids was sufficient to cause nuclear localization of p-ß-cateninY489 in normoxia and mimicked the pattern of α-smooth muscle actin (α-SMA) expression seen with fibroblastic activation from oxidative stress. Treatment of organoids with a tyrosine kinase inhibitor prior to cyclic hypoxia-hyperoxia inhibited nuclear localization of p-ß-cateninY489 and prevented α-SMA expression by fibroblasts. Posttranslational phosphorylation of ß-catenin is a transient feature of normal lung development. Moreover, the persistence of p-ß-cateninY489 is a durable marker of fibroblast activation in BPD and may play an important role in BPD disease pathobiology.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Procesamiento Proteico-Postraduccional , beta Catenina/metabolismo , Actinas/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Dasatinib/farmacología , Fibroblastos/efectos de los fármacos , Humanos , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Hiperoxia/patología , Hipoxia/complicaciones , Hipoxia/metabolismo , Hipoxia/patología , Recién Nacido , Pulmón/efectos de los fármacos , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Pulmón/patología , Organoides/efectos de los fármacos , Organoides/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
15.
Am J Physiol Lung Cell Mol Physiol ; 310(10): L889-98, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26968771

RESUMEN

Bronchopulmonary dysplasia (BPD) is a leading complication of premature birth and occurs primarily in infants delivered during the saccular stage of lung development. Histopathology shows decreased alveolarization and a pattern of fibroblast proliferation and differentiation to the myofibroblast phenotype. Little is known about the molecular pathways and cellular mechanisms that define BPD pathophysiology and progression. We have developed a novel three-dimensional human model of the fibroblast activation associated with BPD, and using this model we have identified the Notch pathway as a key driver of fibroblast activation and proliferation in response to changes in oxygen. Fetal lung fibroblasts were cultured on sodium alginate beads to generate lung organoids. After exposure to alternating hypoxia and hyperoxia, the organoids developed a phenotypic response characterized by increased α-smooth muscle actin (α-SMA) expression and other genes known to be upregulated in BPD and also demonstrated increased expression of downstream effectors of the Notch pathway. Inhibition of Notch with a γ-secretase inhibitor prevented the development of the pattern of cellular proliferation and α-SMA expression in our model. Analysis of human autopsy tissue from the lungs of infants who expired with BPD demonstrated evidence of Notch activation within fibrotic areas of the alveolar septae, suggesting that Notch may be a key driver of BPD pathophysiology.


Asunto(s)
Displasia Broncopulmonar/patología , Transducción de Señal , Alginatos/química , Displasia Broncopulmonar/metabolismo , Técnicas de Cultivo de Célula , Hipoxia de la Célula , Células Cultivadas , Medios de Cultivo/química , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos , Receptores Notch/metabolismo
16.
J Cell Biol ; 211(5): 1057-75, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26644517

RESUMEN

Keratin intermediate filaments (KIFs) protect the epidermis against mechanical force, support strong adhesion, help barrier formation, and regulate growth. The mechanisms by which type I and II keratins contribute to these functions remain incompletely understood. Here, we report that mice lacking all type I or type II keratins display severe barrier defects and fragile skin, leading to perinatal mortality with full penetrance. Comparative proteomics of cornified envelopes (CEs) from prenatal KtyI(-/-) and KtyII(-/-)(K8) mice demonstrates that absence of KIF causes dysregulation of many CE constituents, including downregulation of desmoglein 1. Despite persistence of loricrin expression and upregulation of many Nrf2 targets, including CE components Sprr2d and Sprr2h, extensive barrier defects persist, identifying keratins as essential CE scaffolds. Furthermore, we show that KIFs control mitochondrial lipid composition and activity in a cell-intrinsic manner. Therefore, our study explains the complexity of keratinopathies accompanied by barrier disorders by linking keratin scaffolds to mitochondria, adhesion, and CE formation.


Asunto(s)
Epidermis/metabolismo , Queratinas/metabolismo , Lípidos/química , Mitocondrias/metabolismo , Animales , Adhesión Celular , Membrana Celular/metabolismo , Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Filamentos Intermedios/metabolismo , Queratinocitos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoma/metabolismo , Factores de Transcripción/metabolismo
17.
Mol Biol Cell ; 25(15): 2260-71, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899640

RESUMEN

Molecular chaperones play key roles during growth, development, and stress survival. The ability to induce chaperone expression enables cells to cope with the accumulation of nonnative proteins under stress and complete developmental processes with an increased requirement for chaperone assistance. Here we generate and analyze transgenic mice that lack the cochaperone HSPBP1, a nucleotide-exchange factor of HSP70 proteins and inhibitor of chaperone-assisted protein degradation. Male HSPBP1(-/-) mice are sterile because of impaired meiosis and massive apoptosis of spermatocytes. HSPBP1 deficiency in testes strongly reduces the expression of the inducible, antiapoptotic HSP70 family members HSPA1L and HSPA2, the latter of which is essential for synaptonemal complex disassembly during meiosis. We demonstrate that HSPBP1 affects chaperone expression at a posttranslational level by inhibiting the ubiquitylation and proteasomal degradation of inducible HSP70 proteins. We further provide evidence that the cochaperone BAG2 contributes to HSP70 stabilization in tissues other than testes. Our findings reveal that chaperone expression is determined not only by regulated transcription, but also by controlled degradation, with degradation-inhibiting cochaperones exerting essential prosurvival functions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Espermatogénesis , Ubiquitinación , Animales , Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Humanos , Infertilidad Masculina/genética , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis , Testículo/metabolismo , Testículo/patología
18.
Development ; 139(21): 3973-85, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22932696

RESUMEN

During murine embryogenesis, the Ets factor Erg is highly expressed in endothelial cells of the developing vasculature and in articular chondrocytes of developing bone. We identified seven isoforms for the mouse Erg gene. Four share a common translational start site encoded by exon 3 (Ex3) and are enriched in chondrocytes. The other three have a separate translational start site encoded by Ex4 and are enriched in endothelial cells. Homozygous Erg(ΔEx3/ΔEx3) knockout mice are viable, fertile and do not display any overt phenotype. By contrast, homozygous Erg(ΔEx4/ΔEx4) knockout mice are embryonic lethal, which is associated with a marked reduction in endocardial-mesenchymal transformation (EnMT) during cardiac valve morphogenesis. We show that Erg is required for the maintenance of the core EnMT regulatory factors that include Snail1 and Snail2 by binding to their promoter and intronic regions.


Asunto(s)
Endocardio/metabolismo , Válvulas Cardíacas/embriología , Válvulas Cardíacas/metabolismo , Mesodermo/metabolismo , Proteínas Oncogénicas/metabolismo , Animales , Endocardio/embriología , Genotipo , Mesodermo/embriología , Ratones , Ratones Noqueados , Morfogénesis , Proteínas Oncogénicas/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulador Transcripcional ERG
19.
Am J Pathol ; 178(4): 1578-90, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21435445

RESUMEN

The mammalian placenta represents the interface between maternal and embryonic tissues and provides nutrients and gas exchange during embryo growth. Recently, keratin intermediate filament proteins were found to regulate embryo growth upstream of the mammalian target of rapamycin pathway through glucose transporter relocalization and to contribute to yolk sac vasculogenesis through altered bone morphogenetic protein 4 signaling. Whether keratins have vital functions in extraembryonic tissues is not well understood. Here, we report that keratins are essential for placental function. In the absence of keratins, we find hyperoxia in the decidual tissue directly adjacent to the placenta, because of an increased maternal vasculature. Hyperoxia causes impaired vasculogenesis through defective hypoxia-inducible factor 1α and vascular endothelial growth factor signaling, resulting in invagination defects of fetal blood vessels into the chorion. In turn, the reduced labyrinth, together with impaired gas exchange between maternal and embryonic blood, led to increased hypoxia in keratin-deficient embryos. We provide evidence that keratin-positive trophoblast secretion of prolactin-like protein a (Prlpa) and placental growth factor (PlGF) during decidualization are altered in the absence of keratins, leading to increased infiltration of uterine natural killer cells into placental vicinity and increased vascularization of the maternal decidua. Our findings suggest that keratin mutations might mediate conditions leading to early pregnancy loss due to hyperoxia in the decidua.


Asunto(s)
Decidua/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hiperoxia/metabolismo , Queratinas/metabolismo , Placenta/irrigación sanguínea , Animales , Linaje de la Célula , Corion/metabolismo , Femenino , Hibridación in Situ/métodos , Ratones , Ratones Transgénicos , Microscopía Fluorescente/métodos , Mutación , Factor de Crecimiento Placentario , Embarazo , Proteínas Gestacionales/metabolismo
20.
Vasc Med ; 15(4): 321-31, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20621955

RESUMEN

The differentiation of embryonic stem cells along the endothelial cell lineage requires a tightly coordinated sequence of events that are regulated in both space and time. Although significant gaps remain in this process, major strides have been made over the past 10 years in identifying the growth factors, signal transduction pathways, and transcription factors that function together as critical mediators of this process. Examples of some of the signal transduction pathways include the hedgehog (HH), WNT, BMP, and Notch pathways. A complex interplay between growth factors, and activation of a variety of signal transduction pathways leads to the induction of transcriptional programs that promote the differentiation of embryonic stem cells along the endothelial lineage and ultimately into arterial, venous, and lymphatic endothelial cells. The purpose of this review is to summarize the recent advances in our understanding of the molecular mechanisms underlying endothelial differentiation.


Asunto(s)
Células Madre Embrionarias/citología , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Transducción de Señal/fisiología , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...