Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(3): 176, 2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35247097

RESUMEN

The brain-expressed ubiquilins (UBQLNs) 1, 2 and 4 are a family of ubiquitin adaptor proteins that participate broadly in protein quality control (PQC) pathways, including the ubiquitin proteasome system (UPS). One family member, UBQLN2, has been implicated in numerous neurodegenerative diseases including ALS/FTD. UBQLN2 typically resides in the cytoplasm but in disease can translocate to the nucleus, as in Huntington's disease where it promotes the clearance of mutant Huntingtin. How UBQLN2 translocates to the nucleus and clears aberrant nuclear proteins, however, is not well understood. In a mass spectrometry screen to discover UBQLN2 interactors, we identified a family of small (13 kDa), highly homologous uncharacterized proteins, RTL8, and confirmed the interaction between UBQLN2 and RTL8 both in vitro using recombinant proteins and in vivo using mouse brain tissue. Under endogenous and overexpressed conditions, RTL8 localizes to nucleoli. When co-expressed with UBQLN2, RTL8 promotes nuclear translocation of UBQLN2. RTL8 also facilitates UBQLN2's nuclear translocation during heat shock. UBQLN2 and RTL8 colocalize within ubiquitin-enriched subnuclear structures containing PQC components. The robust effect of RTL8 on the nuclear translocation and subnuclear localization of UBQLN2 does not extend to the other brain-expressed ubiquilins, UBQLN1 and UBQLN4. Moreover, compared to UBQLN1 and UBQLN4, UBQLN2 preferentially stabilizes RTL8 levels in human cell lines and in mouse brain, supporting functional heterogeneity among UBQLNs. As a novel UBQLN2 interactor that recruits UBQLN2 to specific nuclear compartments, RTL8 may regulate UBQLN2 function in nuclear protein quality control.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Nucléolo Celular/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia , Temperatura , Ubiquitina/metabolismo
2.
Sci Rep ; 11(1): 13037, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158522

RESUMEN

Exacerbations of muco-obstructive airway diseases such as COPD and asthma are associated with epithelial changes termed mucous metaplasia (MM). Many molecular pathways triggering MM have been identified; however, the factors that regulate resolution are less well understood. We hypothesized that the autophagy pathway is required for resolution of MM by eliminating excess non-secreted intracellular mucin granules. We found increased intracellular levels of mucins Muc5ac and Muc5b in mice deficient in autophagy regulatory protein, Atg16L1, and that this difference was not due to defects in the known baseline or stimulated mucin secretion pathways. Instead, we found that, in mucous secretory cells, Lc3/Lamp1 vesicles colocalized with mucin granules particularly adjacent to the nucleus, suggesting that some granules were being eliminated in the autophagy pathway rather than secreted. Using a mouse model of MM resolution, we found increased lysosomal proteolytic activity that peaked in the days after mucin production began to decline. In purified lysosomal fractions, Atg16L1-deficient mice had reduced proteolytic degradation of Lc3 and Sqstm1 and persistent accumulation of mucin granules associated with impaired resolution of mucous metaplasia. In normal and COPD derived human airway epithelial cells (AECs), activation of autophagy by mTOR inhibition led to a reduction of intracellular mucin granules in AECs. Our findings indicate that during peak and resolution phases of MM, autophagy activity rather than secretion is required for elimination of some remaining mucin granules. Manipulation of autophagy activation offers a therapeutic target to speed resolution of MM in airway disease exacerbations.


Asunto(s)
Autofagia , Pulmón/metabolismo , Pulmón/patología , Mucina 5AC/metabolismo , Mucina 5B/metabolismo , Moco/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Femenino , Humanos , Inflamación/patología , Interleucina-33/metabolismo , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Metaplasia , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
3.
Sci Immunol ; 6(60)2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34145066

RESUMEN

The nutrient-sensing mammalian target of rapamycin (mTOR) is integral to cell fate decisions after T cell activation. Sustained mTORC1 activity favors the generation of terminally differentiated effector T cells instead of follicular helper and memory T cells. This is particularly pertinent for T cell responses of older adults who have sustained mTORC1 activation despite dysfunctional lysosomes. Here, we show that lysosome-deficient T cells rely on late endosomes rather than lysosomes as an mTORC1 activation platform, where mTORC1 is activated by sensing cytosolic amino acids. T cells from older adults have an increased expression of the plasma membrane leucine transporter SLC7A5 to provide a cytosolic amino acid source. Hence, SLC7A5 and VPS39 deficiency (a member of the HOPS complex promoting early to late endosome conversion) substantially reduced mTORC1 activities in T cells from older but not young individuals. Late endosomal mTORC1 is independent of the negative-feedback loop involving mTORC1-induced inactivation of the transcription factor TFEB that controls expression of lysosomal genes. The resulting sustained mTORC1 activation impaired lysosome function and prevented lysosomal degradation of PD-1 in CD4+ T cells from older adults, thereby inhibiting their proliferative responses. VPS39 silencing of human T cells improved their expansion to pertussis and to SARS-CoV-2 peptides in vitro. Furthermore, adoptive transfer of CD4+ Vps39-deficient LCMV-specific SMARTA cells improved germinal center responses, CD8+ memory T cell generation, and recall responses to infection. Thus, curtailing late endosomal mTORC1 activity is a promising strategy to enhance T cell immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Endosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , SARS-CoV-2/metabolismo , Transducción de Señal/genética , Traslado Adoptivo/métodos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , COVID-19/virología , Células Cultivadas , Femenino , Proteína Forkhead Box O1/deficiencia , Proteína Forkhead Box O1/genética , Voluntarios Sanos , Humanos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/inmunología , Transfección , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética , Adulto Joven
4.
Elife ; 102021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34085925

RESUMEN

Defective autophagy is strongly associated with chronic inflammation. Loss-of-function of the core autophagy gene Atg16l1 increases risk for Crohn's disease in part by enhancing innate immunity through myeloid cells such as macrophages. However, autophagy is also recognized as a mechanism for clearance of certain intracellular pathogens. These divergent observations prompted a re-evaluation of ATG16L1 in innate antimicrobial immunity. In this study, we found that loss of Atg16l1 in myeloid cells enhanced the killing of virulent Shigella flexneri (S.flexneri), a clinically relevant enteric bacterium that resides within the cytosol by escaping from membrane-bound compartments. Quantitative multiplexed proteomics of murine bone marrow-derived macrophages revealed that ATG16L1 deficiency significantly upregulated proteins involved in the glutathione-mediated antioxidant response to compensate for elevated oxidative stress, which simultaneously promoted S.flexneri killing. Consistent with this, myeloid-specific deletion of Atg16l1 in mice accelerated bacterial clearance in vitro and in vivo. Pharmacological induction of oxidative stress through suppression of cysteine import enhanced microbial clearance by macrophages. Conversely, antioxidant treatment of macrophages permitted S.flexneri proliferation. These findings demonstrate that control of oxidative stress by ATG16L1 and autophagy regulates antimicrobial immunity against intracellular pathogens.


Asunto(s)
Proteínas Relacionadas con la Autofagia/deficiencia , Autofagia , Disentería Bacilar/microbiología , Inmunidad Innata , Macrófagos/microbiología , Estrés Oxidativo , Proteoma , Proteómica , Shigella flexneri/patogenicidad , Animales , Proteínas Relacionadas con la Autofagia/genética , Células Cultivadas , Modelos Animales de Enfermedad , Disentería Bacilar/inmunología , Disentería Bacilar/metabolismo , Interacciones Huésped-Patógeno , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Viabilidad Microbiana , Shigella flexneri/inmunología , Shigella flexneri/metabolismo , Virulencia
5.
Nat Commun ; 12(1): 2431, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893273

RESUMEN

Insulin resistance and lower muscle quality (strength divided by mass) are hallmarks of type 2 diabetes (T2D). Here, we explore whether alterations in muscle stem cells (myoblasts) from individuals with T2D contribute to these phenotypes. We identify VPS39 as an important regulator of myoblast differentiation and muscle glucose uptake, and VPS39 is downregulated in myoblasts and myotubes from individuals with T2D. We discover a pathway connecting VPS39-deficiency in human myoblasts to impaired autophagy, abnormal epigenetic reprogramming, dysregulation of myogenic regulators, and perturbed differentiation. VPS39 knockdown in human myoblasts has profound effects on autophagic flux, insulin signaling, epigenetic enzymes, DNA methylation and expression of myogenic regulators, and gene sets related to the cell cycle, muscle structure and apoptosis. These data mimic what is observed in myoblasts from individuals with T2D. Furthermore, the muscle of Vps39+/- mice display reduced glucose uptake and altered expression of genes regulating autophagy, epigenetic programming, and myogenesis. Overall, VPS39-deficiency contributes to impaired muscle differentiation and reduced glucose uptake. VPS39 thereby offers a therapeutic target for T2D.


Asunto(s)
Proteínas Relacionadas con la Autofagia/genética , Autofagia/genética , Diferenciación Celular/genética , Diabetes Mellitus Tipo 2/genética , Epigenómica/métodos , Mioblastos/metabolismo , Células Madre/metabolismo , Proteínas de Transporte Vesicular/genética , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Epigénesis Genética/genética , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Desarrollo de Músculos/genética , Proteínas de Transporte Vesicular/deficiencia
6.
J Mol Biol ; 433(5): 166809, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33484718

RESUMEN

Macroautophagy is a bulk degradation mechanism in eukaryotic cells. Efficiency of an essential step of this process in yeast, Atg8 lipidation, relies on the presence of Atg16, a subunit of the Atg12-Atg5-Atg16 complex acting as the E3-like enzyme in the ubiquitination-like reaction. A current view on the functional structure of Atg16 in the yeast S. cerevisiae comes from the two crystal structures that reveal the Atg5-interacting α-helix linked via a flexible linker to another α-helix of Atg16, which then assembles into a homodimer. This view does not explain the results of previous in vitro studies revealing Atg16-dependent deformations of membranes and liposome-binding of the Atg12-Atg5 conjugate upon addition of Atg16. Here we show that Atg16 acts as both a homodimerizing and peripheral membrane-binding polypeptide. These two characteristics are imposed by the two distinct regions that are disordered in the nascent protein. Atg16 binds to membranes in vivo via the amphipathic α-helix (amino acid residues 113-131) that has a coiled-coil-like propensity and a strong hydrophobic face for insertion into the membrane. The other protein region (residues 64-99) possesses a coiled-coil propensity, but not amphipathicity, and is dispensable for membrane anchoring of Atg16. This region acts as a Leu-zipper essential for formation of the Atg16 homodimer. Mutagenic disruption in either of these two distinct domains renders Atg16 proteins that, in contrast to wild type, completely fail to rescue the autophagy-defective phenotype of atg16Δ cells. Together, the results of this study yield a model for the molecular mechanism of Atg16 function in macroautophagy.


Asunto(s)
Proteína 12 Relacionada con la Autofagia/química , Proteína 5 Relacionada con la Autofagia/química , Familia de las Proteínas 8 Relacionadas con la Autofagia/química , Proteínas Relacionadas con la Autofagia/química , Membrana Celular/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Autofagia/genética , Proteína 12 Relacionada con la Autofagia/genética , Proteína 12 Relacionada con la Autofagia/metabolismo , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Familia de las Proteínas 8 Relacionadas con la Autofagia/genética , Familia de las Proteínas 8 Relacionadas con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Sitios de Unión , Membrana Celular/metabolismo , Regulación Fúngica de la Expresión Génica , Interacciones Hidrofóbicas e Hidrofílicas , Liposomas/química , Liposomas/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
7.
J Biol Chem ; 296: 100153, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33277362

RESUMEN

Familial neurodegenerative diseases commonly involve mutations that result in either aberrant proteins or dysfunctional components of the proteolytic machinery that act on aberrant proteins. UBQLN2 is a ubiquitin receptor of the UBL/UBA family that binds the proteasome through its ubiquitin-like domain and is thought to deliver ubiquitinated proteins to proteasomes for degradation. UBQLN2 mutations result in familial amyotrophic lateral sclerosis (ALS)/frontotemporal dementia in humans through an unknown mechanism. Quantitative multiplexed proteomics was used to provide for the first time an unbiased and global analysis of the role of Ubqln2 in controlling the composition of the proteome. We studied several murine models of Ubqln2-linked ALS and also generated Ubqln2 null mutant mice. We identified impacts of Ubqln2 on diverse physiological pathways, most notably serotonergic signaling. Interestingly, we observed an upregulation of proteasome subunits, suggesting a compensatory response to diminished proteasome output. Among the specific proteins whose abundance is linked to UBQLN2 function, the strongest hits were the ubiquitin ligase TRIM32 and two retroelement-derived proteins, PEG10 and CXX1B. Cycloheximide chase studies using induced human neurons and HEK293 cells suggested that PEG10 and TRIM32 are direct clients. Although UBQLN2 directs the degradation of multiple proteins via the proteasome, it surprisingly conferred strong protection from degradation on the Gag-like protein CXX1B, which is expressed from the same family of retroelement genes as PEG10. In summary, this study charts the proteomic landscape of ALS-related Ubqln2 mutants and identifies candidate client proteins that are altered in vivo in disease models and whose degradation is promoted by UBQLN2.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Esclerosis Amiotrófica Lateral/genética , Proteínas Relacionadas con la Autofagia/genética , Demencia Frontotemporal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica/métodos , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/metabolismo , Línea Celular , Cicloheximida/farmacología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Serotonina/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
8.
Nature ; 585(7824): 251-255, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32848248

RESUMEN

Mutation of C9orf72 is the most prevalent defect associated with amyotrophic lateral sclerosis and frontotemporal degeneration1. Together with hexanucleotide-repeat expansion2,3, haploinsufficiency of C9orf72 contributes to neuronal dysfunction4-6. Here we determine the structure of the C9orf72-SMCR8-WDR41 complex by cryo-electron microscopy. C9orf72 and SMCR8 both contain longin and DENN (differentially expressed in normal and neoplastic cells) domains7, and WDR41 is a ß-propeller protein that binds to SMCR8 such that the whole structure resembles an eye slip hook. Contacts between WDR41 and the DENN domain of SMCR8 drive the lysosomal localization of the complex in conditions of amino acid starvation. The structure suggested that C9orf72-SMCR8 is a GTPase-activating protein (GAP), and we found that C9orf72-SMCR8-WDR41 acts as a GAP for the ARF family of small GTPases. These data shed light on the function of C9orf72 in normal physiology, and in amyotrophic lateral sclerosis and frontotemporal degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas Relacionadas con la Autofagia/química , Proteína C9orf72/química , Proteína C9orf72/genética , Proteínas Portadoras/química , Microscopía por Crioelectrón , Demencia Frontotemporal/genética , Haploinsuficiencia , Complejos Multiproteicos/química , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/ultraestructura , Proteína C9orf72/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/ultraestructura , Demencia Frontotemporal/metabolismo , Humanos , Lisosomas/metabolismo , Modelos Moleculares , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Dominios Proteicos
9.
Prog Mol Biol Transl Sci ; 172: 293-323, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32620246

RESUMEN

Autophagy is a highly conserved lysosomal degradation pathway responsible for rapid elimination of unwanted cytoplasmic materials in response to stressful conditions. This cytoprotective function is essential for maintenance of cellular homeostasis and is mediated by conserved autophagy-related genes (ATG) and autophagic receptors. Impairment of autophagy frequently results in a wide variety of human pathologies. Recent studies have revealed direct links between diverse diseases and genetic defects of core autophagy genes, autophagy-associated genes, and genes encoding autophagic receptors. Here we provide a general description of autophagy-related genes and their mutations or polymorphisms that play a causative role in specific human disorders or may be risk factors for them.


Asunto(s)
Proteínas Relacionadas con la Autofagia/genética , Autofagia/genética , Enfermedades Genéticas Congénitas/patología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Autofagia/fisiología , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/fisiología , Enfermedades Genéticas Congénitas/genética , Homeostasis , Humanos , Inflamación/genética , Inflamación/patología , Ratones , Ratones Noqueados , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Polimorfismo Genético , Estrés Fisiológico/fisiología
10.
Prog Mol Biol Transl Sci ; 172: 87-106, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32620252

RESUMEN

Autophagy is a cellular housekeeping and quality control mechanism that is essential for homeostasis and survival. By virtue of this role, any perturbations to the flow of this process in cardiac or vascular cells can elicit harmful effects on the cardiovascular system, and subsequently affect whole organismal health. In this chapter, we summarize the preclinical evidence supporting the role of autophagy in sustaining cardiovascular health during homeostasis and disease. Furthermore, we discuss how autophagy activation by dietary, genetic and pharmaceutical interventions can be exploited to counteract common cardiovascular disorders, including atherosclerosis, coronary artery disease, diabetic cardiomyopathy, arrhythmia, chemotherapy-induced cardiotoxicity and heart failure.


Asunto(s)
Autofagia , Enfermedades Cardiovasculares/patología , Envejecimiento/fisiología , Animales , Autofagosomas/fisiología , Autofagia/efectos de los fármacos , Autofagia/genética , Autofagia/fisiología , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/fisiología , Restricción Calórica , Enfermedades Cardiovasculares/terapia , Fenómenos Fisiológicos Cardiovasculares , Desarrollo Embrionario/fisiología , Endotelio Vascular/fisiopatología , Homeostasis , Humanos , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo , Sirolimus/farmacología , Sirolimus/uso terapéutico , Espermidina/farmacología , Espermidina/uso terapéutico , Trehalosa/farmacología , Trehalosa/uso terapéutico , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología
11.
Biomed Pharmacother ; 129: 110347, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32535386

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD), which often accompanied by metabolic syndrome, such as obesity, diabetes and dyslipidemia, has become a global health problem. Our previous results show that HCV core protein binding protein 6 (HCBP6) could maintain the triglyceride homeostasis in liver cells. However, the role of HCBP6 in NAFLD and its associated metabolic disorders remains incompletely understood. METHODS: Hepatic HCBP6 expression was determined by qRT-PCR, Western blot and immunohistochemistry analysis. HCBP6 knockout (HCBP6-KO) mice were constructed and fed a high-fat diet (HFD) to induce NAFLD. The effects of HCBP6 on glucose and lipid metabolism were measured by HE staining, qRT-PCR, Western blot and GTT. Wild-type and HCBP6-KO mice kept on a HFD were treated with ginsenosides Rh2, and HE staining and GTT were used to study the function of Rh2 in metabolism disorders. RESULTS: HCBP6 is reduced in HFD-fed mice. HCBP6 deficiency increased the body weight, aggravated fatty liver and deteriorated lipid homeostasis as well as glucose homeostasis in HFD-induced mouse model of NAFLD. Moreover, HCBP6-KO mice failed to maintain body temperature upon cold challenge. Mechanistically, HCBP6 could regulate lipolysis and fatty acid oxidation via activation of AMKP in vivo. In addition, HCBP6 expression was upregulated by ginsenosides Rh2. Accordingly, ginsenosides Rh2 administrations improved HFD-induced fatty liver and glucose tolerance. CONCLUSIONS: These findings indicated that HCBP6 is essential in maintaining lipid and glucose homeostasis and body temperature. HCBP6 augmented by ginsenosides Rh2 may be a promising therapeutic strategy for the treatment of metabolic disorders in NAFLD mice.


Asunto(s)
Proteínas Relacionadas con la Autofagia/deficiencia , Glucemia/metabolismo , Ácidos Grasos/metabolismo , Lipólisis , Hígado/metabolismo , Síndrome Metabólico/metabolismo , Proteínas Mitocondriales/deficiencia , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/genética , Glucemia/efectos de los fármacos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ginsenósidos/farmacología , Células Hep G2 , Humanos , Insulina/sangre , Interleucina-6/metabolismo , Lipólisis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Oxidación-Reducción , Factor de Necrosis Tumoral alfa/metabolismo
12.
Blood ; 135(26): 2388-2401, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32232483

RESUMEN

A goal in precision medicine is to use patient-derived material to predict disease course and intervention outcomes. Here, we use mechanistic observations in a preclinical animal model to design an ex vivo platform that recreates genetic susceptibility to T-cell-mediated damage. Intestinal graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. We found that intestinal GVHD in mice deficient in Atg16L1, an autophagy gene that is polymorphic in humans, is reversed by inhibiting necroptosis. We further show that cocultured allogeneic T cells kill Atg16L1-mutant intestinal organoids from mice, which was associated with an aberrant epithelial interferon signature. Using this information, we demonstrate that pharmacologically inhibiting necroptosis or interferon signaling protects human organoids derived from individuals harboring a common ATG16L1 variant from allogeneic T-cell attack. Our study provides a roadmap for applying findings in animal models to individualized therapy that targets affected tissues.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Enfermedades Intestinales/prevención & control , Organoides , Linfocitos T/inmunología , Acrilamidas/farmacología , Animales , Autofagia , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Trasplante de Médula Ósea/efectos adversos , Técnicas de Cocultivo , Colon/anomalías , Femenino , Predisposición Genética a la Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Imidazoles/farmacología , Indoles/farmacología , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Intestinales/inmunología , Enfermedades Intestinales/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Necroptosis/efectos de los fármacos , Nitrilos , Células de Paneth/patología , Medicina de Precisión , Pirazoles/farmacología , Pirimidinas , Quimera por Radiación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Sulfonamidas/farmacología , Linfocitos T/trasplante
13.
Nat Microbiol ; 5(2): 272-281, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31959973

RESUMEN

Innate and adaptive immune responses that prime myeloid cells, such as macrophages, protect against pathogens1,2. However, if left uncontrolled, these responses may lead to detrimental inflammation3. Macrophages, particularly those resident in tissues, must therefore remain quiescent between infections despite chronic stimulation by commensal microorganisms. The genes required for quiescence of tissue-resident macrophages are not well understood. Autophagy, an evolutionarily conserved cellular process by which cytoplasmic contents are targeted for lysosomal digestion, has homeostatic functions including maintenance of protein and organelle integrity and regulation of metabolism4. Recent research has shown that degradative autophagy, as well as various combinations of autophagy genes, regulate immunity and inflammation5-12. Here, we delineate a function of the autophagy proteins Beclin 1 and FIP200-but not of other essential autophagy components ATG5, ATG16L1 or ATG7-in mediating quiescence of tissue-resident macrophages by limiting the effects of systemic interferon-γ. The perturbation of quiescence in mice that lack Beclin 1 or FIP200 in myeloid cells results in spontaneous immune activation and resistance to Listeria monocytogenes infection. While antibiotic-treated wild-type mice display diminished macrophage responses to inflammatory stimuli, this is not observed in mice that lack Beclin 1 in myeloid cells, establishing the dominance of this gene over effects of the bacterial microbiota. Thus, select autophagy genes, but not all genes essential for degradative autophagy, have a key function in maintaining immune quiescence of tissue-resident macrophages, resulting in genetically programmed susceptibility to bacterial infection.


Asunto(s)
Autofagia/genética , Listeria monocytogenes/patogenicidad , Macrófagos Peritoneales/inmunología , Animales , Autofagia/inmunología , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/inmunología , Beclina-1/deficiencia , Beclina-1/genética , Beclina-1/inmunología , Proliferación Celular , Susceptibilidad a Enfermedades/inmunología , Femenino , Predisposición Genética a la Enfermedad , Interferón gamma/inmunología , Listeria monocytogenes/inmunología , Listeriosis/etiología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/microbiología , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Nat Cell Biol ; 22(2): 187-199, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932738

RESUMEN

Traditionally viewed as an autodigestive pathway, autophagy also facilitates cellular secretion; however, the mechanisms underlying these processes remain unclear. Here, we demonstrate that components of the autophagy machinery specify secretion within extracellular vesicles (EVs). Using a proximity-dependent biotinylation proteomics strategy, we identify 200 putative targets of LC3-dependent secretion. This secretome consists of a highly interconnected network enriched in RNA-binding proteins (RBPs) and EV cargoes. Proteomic and RNA profiling of EVs identifies diverse RBPs and small non-coding RNAs requiring the LC3-conjugation machinery for packaging and secretion. Focusing on two RBPs, heterogeneous nuclear ribonucleoprotein K (HNRNPK) and scaffold-attachment factor B (SAFB), we demonstrate that these proteins interact with LC3 and are secreted within EVs enriched with lipidated LC3. Furthermore, their secretion requires the LC3-conjugation machinery, neutral sphingomyelinase 2 (nSMase2) and LC3-dependent recruitment of factor associated with nSMase2 activity (FAN). Hence, the LC3-conjugation pathway controls EV cargo loading and secretion.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/genética , Vesículas Extracelulares/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Unión al ARN/genética , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Autofagosomas/química , Proteína 7 Relacionada con la Autofagia/deficiencia , Proteína 7 Relacionada con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Transporte Biológico , Biotinilación , Vesículas Extracelulares/química , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/química , Lisosomas/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteómica/métodos , Células RAW 264.7 , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Proteínas de Unión al ARN/clasificación , Proteínas de Unión al ARN/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo
15.
PLoS Biol ; 17(10): e3000508, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31593566

RESUMEN

CDGSH iron-sulfur domain-containing protein 2 (Cisd2) is pivotal to mitochondrial integrity and intracellular Ca2+ homeostasis. In the heart of Cisd2 knockout mice, Cisd2 deficiency causes intercalated disc defects and leads to degeneration of the mitochondria and sarcomeres, thereby impairing its electromechanical functioning. Furthermore, Cisd2 deficiency disrupts Ca2+ homeostasis via dysregulation of sarco/endoplasmic reticulum Ca2+-ATPase (Serca2a) activity, resulting in an increased level of basal cytosolic Ca2+ and mitochondrial Ca2+ overload in cardiomyocytes. Most strikingly, in Cisd2 transgenic mice, a persistently high level of Cisd2 is sufficient to delay cardiac aging and attenuate age-related structural defects and functional decline. In addition, it results in a younger cardiac transcriptome pattern during old age. Our findings indicate that Cisd2 plays an essential role in cardiac aging and in the heart's electromechanical functioning. They highlight Cisd2 as a novel drug target when developing therapies to delay cardiac aging and ameliorate age-related cardiac dysfunction.


Asunto(s)
Envejecimiento Prematuro/genética , Envejecimiento/fisiología , Bloqueo Atrioventricular/genética , Proteínas Relacionadas con la Autofagia/genética , Corazón/fisiopatología , Proteínas del Tejido Nervioso/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/fisiopatología , Animales , Bloqueo Atrioventricular/diagnóstico por imagen , Bloqueo Atrioventricular/metabolismo , Bloqueo Atrioventricular/fisiopatología , Proteínas Relacionadas con la Autofagia/deficiencia , Calcio/metabolismo , Electrocardiografía , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Corazón/fisiología , Homeostasis/fisiología , Masculino , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Proteínas del Tejido Nervioso/deficiencia , Sarcómeros/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transcriptoma
16.
J Biol Chem ; 294(44): 16172-16185, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31515271

RESUMEN

Connections between deficient autophagy and insulin resistance have emerged, however, the mechanism through which reduced autophagy impairs insulin-signaling remains unknown. We examined mouse embryonic fibroblasts lacking Atg16l1 (ATG16L1 KO mouse embryonic fibroblasts (MEFs)), an essential autophagy gene, and observed deficient insulin and insulin-like growth factor-1 signaling. ATG16L1 KO MEFs displayed reduced protein content of insulin receptor substrate-1 (IRS1), pivotal to insulin signaling, whereas IRS1myc overexpression recovered downstream insulin signaling. Endogenous IRS1 protein content and insulin signaling were restored in ATG16L1 KO mouse embryonic fibroblasts (MEF) upon proteasome inhibition. Through proximity-dependent biotin identification (BioID) and co-immunoprecipitation, we found that Kelch-like proteins KLHL9 and KLHL13, which together form an E3 ubiquitin (Ub) ligase complex with cullin 3 (CUL3), are novel IRS1 interactors. Expression of Klhl9 and Klhl13 was elevated in ATG16L1 KO MEFs and siRNA-mediated knockdown of Klhl9, Klhl13, or Cul3 recovered IRS1 expression. Moreover, Klhl13 and Cul3 knockdown increased insulin signaling. Notably, adipose tissue of high-fat fed mice displayed lower Atg16l1 mRNA expression and IRS1 protein content, and adipose tissue KLHL13 and CUL3 expression positively correlated to body mass index in humans. We propose that ATG16L1 deficiency evokes insulin resistance through induction of Klhl9 and Klhl13, which, in complex with Cul3, promote proteasomal IRS1 degradation.


Asunto(s)
Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Animales , Autofagia/fisiología , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas Cullin/metabolismo , Fibroblastos/metabolismo , Genes Reguladores , Células HEK293 , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas/metabolismo , Transducción de Señal , Complejos de Ubiquitina-Proteína Ligasa/metabolismo
17.
Cell ; 178(3): 552-566.e20, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31327526

RESUMEN

Antibacterial autophagy (xenophagy) is an important host defense, but how it is initiated is unclear. Here, we performed a bacterial transposon screen and identified a T3SS effector SopF that potently blocked Salmonella autophagy. SopF was a general xenophagy inhibitor without affecting canonical autophagy. S. Typhimurium ΔsopF resembled S. flexneri ΔvirAΔicsB with the majority of intracellular bacteria targeted by autophagy, permitting a CRISPR screen that identified host V-ATPase as an essential factor. Upon bacteria-caused vacuolar damage, the V-ATPase recruited ATG16L1 onto bacteria-containing vacuole, which was blocked by SopF. Mammalian ATG16L1 bears a WD40 domain required for interacting with the V-ATPase. Inhibiting autophagy by SopF promoted S. Typhimurium proliferation in vivo. SopF targeted Gln124 of ATP6V0C in the V-ATPase for ADP-ribosylation. Mutation of Gln124 also blocked xenophagy, but not canonical autophagy. Thus, the discovery of SopF reveals the V-ATPase-ATG16L1 axis that critically mediates autophagic recognition of intracellular pathogen.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas Bacterianas/genética , Macroautofagia , Salmonella/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Factores de Virulencia/genética , ADP-Ribosilación , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Proteínas Bacterianas/metabolismo , Sistemas CRISPR-Cas/genética , Edición Génica , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Salmonella/patogenicidad , Sistemas de Secreción Tipo III/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Factores de Virulencia/metabolismo
18.
Cell ; 178(3): 536-551.e14, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31257024

RESUMEN

The expression of some proteins in the autophagy pathway declines with age, which may impact neurodegeneration in diseases, including Alzheimer's Disease. We have identified a novel non-canonical function of several autophagy proteins in the conjugation of LC3 to Rab5+, clathrin+ endosomes containing ß-amyloid in a process of LC3-associated endocytosis (LANDO). We found that LANDO in microglia is a critical regulator of immune-mediated aggregate removal and microglial activation in a murine model of AD. Mice lacking LANDO but not canonical autophagy in the myeloid compartment or specifically in microglia have a robust increase in pro-inflammatory cytokine production in the hippocampus and increased levels of neurotoxic ß-amyloid. This inflammation and ß-amyloid deposition were associated with reactive microgliosis and tau hyperphosphorylation. LANDO-deficient AD mice displayed accelerated neurodegeneration, impaired neuronal signaling, and memory deficits. Our data support a protective role for LANDO in microglia in neurodegenerative pathologies resulting from ß-amyloid deposition.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Endocitosis , Proteínas Asociadas a Microtúbulos/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Proteína 5 Relacionada con la Autofagia/deficiencia , Proteína 5 Relacionada con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Antígenos CD36/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Microglía/citología , Microglía/metabolismo , Células RAW 264.7 , Receptores Inmunológicos/metabolismo , Receptor Toll-Like 4/metabolismo
19.
Gene Expr ; 19(3): 229-234, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-30717822

RESUMEN

Studies using genetic mouse models that have defective autophagy have led to the conclusion that macroautophagy/autophagy serves as a tumor suppressor. One of such models is the liver-specific Atg5 or Atg7 knockout mice, and these knockout mice develop spontaneous liver tumors. It has been generally agreed that p62-mediated Nrf2 activation plays a critical role in promoting autophagy deficiency-induced liver injury and liver tumorigenesis. The mechanisms of how persistent Nrf2 activation induces liver injury and tumorigenesis are incompletely known. We discuss the recent progress on the new roles of HMGB1 and Yap in regulating liver injury and tumorigenesis in mice with liver-specific autophagy deficiency.


Asunto(s)
Proteínas Relacionadas con la Autofagia/genética , Carcinogénesis/genética , Neoplasias Hepáticas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Neoplasias Hepáticas/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Señalizadoras YAP
20.
Cardiovasc Res ; 115(8): 1320-1331, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30428088

RESUMEN

AIMS: WD40 repeat and FYVE domain containing 3 (WDFY3) is an adaptor protein involved in selective degradation of protein aggregates by autophagy. Recent studies have revealed that Wdfy3 is critical in the regulation of brain development and osteoclastogenesis in vivo. However, the function of Wdfy3 in cardiac development remains completely unknown. In this study, we explore the role of Wdfy3 in cardiac morphogenesis using Wdfy3-deficient mice. METHODS AND RESULTS: Wdfy3 was expressed in the developing heart in mice and peaked at embryonic day 12.5 (E12.5). Loss of Wdfy3 in mice led to embryonic and neonatal lethality. Wdfy3-deficient mice displayed various congenital heart defects including membranous ventricular septal defect (VSD), aortic overriding (AO), double outlet right ventricle (DORV), thinning of ventricular wall, ventricular dilation, and disorganized ventricular trabeculation at E14.5. Cell proliferation was reduced in the hearts from Wdfy3-deficient mice at E12.5 and E14.5, which was associated with enhanced p21 expression. Cardiomyocyte differentiation was diminished as demonstrated by reduced Myh6 and MLC2v in Wdfy3-deficient mice at E14.5. In addition, Nkx2-5 and Mef2c, two cardiac transcription factors regulating cardiomyocyte differentiation, were decreased in Wdfy3-deficient mice at E14.5. Apoptotic cell death remained unaltered. These data suggest that reduced cell proliferation and cardiomyocyte differentiation contribute to cardiac defects in Wdfy3-deficient mice. Mechanistically, loss of Wdfy3 led to a reduction in protein levels of Notch 1 intracellular domain and its downstream targets Hes1 and Hey1, which was accompanied with enhanced full-length Notch1 protein levels. In vitro luciferase assay showed that Wdfy3 deficiency induced activity of p21 promoter, while diminished activity of Hes1 promoter through modulation of Notch1 signalling. Moreover, Wdfy3 was co-localized with Notch1 in primary embryonic cardiomyocytes. Endogenous Wdfy3 physically interacted with full-length Notch1 in the developing heart. These results suggest that Notch1 signalling is perturbed in the hearts from Wdfy3-deficient mice. No alteration of autophagy was detected in the hearts from Wdfy3-deficient mice. CONCLUSION: Taken together, our data suggest that Wdfy3 plays an essential role in cardiac development, which may be mediated by modulation of Notch1 signalling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Cardiopatías Congénitas/metabolismo , Corazón/embriología , Miocitos Cardíacos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Corazón/fisiopatología , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Miocitos Cardíacos/patología , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transducción de Señal , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA