Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 3.373
Filtrar
Más filtros

Publication year range
1.
Cell ; 178(1): 176-189.e15, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31155231

RESUMEN

RLR-mediated type I IFN production plays a pivotal role in elevating host immunity for viral clearance and cancer immune surveillance. Here, we report that glycolysis, which is inactivated during RLR activation, serves as a barrier to impede type I IFN production upon RLR activation. RLR-triggered MAVS-RIG-I recognition hijacks hexokinase binding to MAVS, leading to the impairment of hexokinase mitochondria localization and activation. Lactate serves as a key metabolite responsible for glycolysis-mediated RLR signaling inhibition by directly binding to MAVS transmembrane (TM) domain and preventing MAVS aggregation. Notably, lactate restoration reverses increased IFN production caused by lactate deficiency. Using pharmacological and genetic approaches, we show that lactate reduction by lactate dehydrogenase A (LDHA) inactivation heightens type I IFN production to protect mice from viral infection. Our study establishes a critical role of glycolysis-derived lactate in limiting RLR signaling and identifies MAVS as a direct sensor of lactate, which functions to connect energy metabolism and innate immunity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína 58 DEAD Box/antagonistas & inhibidores , Proteína 58 DEAD Box/metabolismo , Ácido Láctico/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Animales , Femenino , Glucólisis , Células HEK293 , Humanos , Interferón beta/metabolismo , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células RAW 264.7 , Receptores Inmunológicos , Transducción de Señal/efectos de los fármacos , Transfección
2.
Nat Immunol ; 19(1): 29-40, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29242539

RESUMEN

Although deletion of certain autophagy-related genes has been associated with defects in hematopoiesis, it remains unclear whether hyperactivated mitophagy affects the maintenance and differentiation of hematopoietic stem cells (HSCs) and committed progenitor cells. Here we report that targeted deletion of the gene encoding the AAA+-ATPase Atad3a hyperactivated mitophagy in mouse hematopoietic cells. Affected mice showed reduced survival, severely decreased bone-marrow cellularity, erythroid anemia and B cell lymphopenia. Those phenotypes were associated with skewed differentiation of stem and progenitor cells and an enlarged HSC pool. Mechanistically, Atad3a interacted with the mitochondrial channel components Tom40 and Tim23 and served as a bridging factor to facilitate appropriate transportation and processing of the mitophagy protein Pink1. Loss of Atad3a caused accumulation of Pink1 and activated mitophagy. Notably, deletion of Pink1 in Atad3a-deficient mice significantly 'rescued' the mitophagy defect, which resulted in restoration of the progenitor and HSC pools. Our data indicate that Atad3a suppresses Pink1-dependent mitophagy and thereby serves a key role in hematopoietic homeostasis.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Proteínas Mitocondriales/metabolismo , Mitofagia , Proteínas Quinasas/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , Animales , Apoptosis/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/genética , Proteínas Quinasas/genética
3.
Mol Cell ; 81(18): 3803-3819.e7, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34547240

RESUMEN

Mitochondrial dynamics regulated by mitochondrial fusion and fission maintain mitochondrial functions, whose alterations underline various human diseases. Here, we show that inositol is a critical metabolite directly restricting AMPK-dependent mitochondrial fission independently of its classical mode as a precursor for phosphoinositide generation. Inositol decline by IMPA1/2 deficiency elicits AMPK activation and mitochondrial fission without affecting ATP level, whereas inositol accumulation prevents AMPK-dependent mitochondrial fission. Metabolic stress or mitochondrial damage causes inositol decline in cells and mice to elicit AMPK-dependent mitochondrial fission. Inositol directly binds to AMPKγ and competes with AMP for AMPKγ binding, leading to restriction of AMPK activation and mitochondrial fission. Our study suggests that the AMP/inositol ratio is a critical determinant for AMPK activation and establishes a model in which AMPK activation requires inositol decline to release AMPKγ for AMP binding. Hence, AMPK is an inositol sensor, whose inactivation by inositol serves as a mechanism to restrict mitochondrial fission.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Inositol/metabolismo , Dinámicas Mitocondriales/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Línea Celular , Humanos , Inositol/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Células PC-3 , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Estrés Fisiológico/fisiología
4.
Cell ; 154(3): 556-68, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911321

RESUMEN

Skp2 E3 ligase is overexpressed in numerous human cancers and plays a critical role in cell-cycle progression, senescence, metabolism, cancer progression, and metastasis. In the present study, we identified a specific Skp2 inhibitor using high-throughput in silico screening of large and diverse chemical libraries. This Skp2 inhibitor selectively suppresses Skp2 E3 ligase activity, but not activity of other SCF complexes. It also phenocopies the effects observed upon genetic Skp2 deficiency, such as suppressing survival and Akt-mediated glycolysis and triggering p53-independent cellular senescence. Strikingly, we discovered a critical function of Skp2 in positively regulating cancer stem cell populations and self-renewal ability through genetic and pharmacological approaches. Notably, Skp2 inhibitor exhibits potent antitumor activities in multiple animal models and cooperates with chemotherapeutic agents to reduce cancer cell survival. Our study thus provides pharmacological evidence that Skp2 is a promising target for restricting cancer stem cell and cancer progression.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Neoplasias/enzimología , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Animales , Antineoplásicos/química , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Genes p53 , Glucólisis/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Proteínas Quinasas Asociadas a Fase-S/química , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Trasplante Heterólogo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
5.
Nature ; 602(7898): 632-638, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35140404

RESUMEN

Animals must set behavioural priority in a context-dependent manner and switch from one behaviour to another at the appropriate moment1-3. Here we probe the molecular and neuronal mechanisms that orchestrate the transition from feeding to courtship in Drosophila melanogaster. We find that feeding is prioritized over courtship in starved males, and the consumption of protein-rich food rapidly reverses this order within a few minutes. At the molecular level, a gut-derived, nutrient-specific neuropeptide hormone-Diuretic hormone 31 (Dh31)-propels a switch from feeding to courtship. We further address the underlying kinetics with calcium imaging experiments. Amino acids from food acutely activate Dh31+ enteroendocrine cells in the gut, increasing Dh31 levels in the circulation. In addition, three-photon functional imaging of intact flies shows that optogenetic stimulation of Dh31+ enteroendocrine cells rapidly excites a subset of brain neurons that express Dh31 receptor (Dh31R). Gut-derived Dh31 excites the brain neurons through the circulatory system within a few minutes, in line with the speed of the feeding-courtship behavioural switch. At the circuit level, there are two distinct populations of Dh31R+ neurons in the brain, with one population inhibiting feeding through allatostatin-C and the other promoting courtship through corazonin. Together, our findings illustrate a mechanism by which the consumption of protein-rich food triggers the release of a gut hormone, which in turn prioritizes courtship over feeding through two parallel pathways.


Asunto(s)
Proteínas de Drosophila , Hormonas de Insectos , Animales , Cortejo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Hormonas de Insectos/metabolismo , Masculino , Nutrientes , Conducta Sexual Animal/fisiología
6.
Nature ; 604(7907): 771-778, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35418677

RESUMEN

Adhesion G protein-coupled receptors (aGPCRs) constitute an evolutionarily ancient family of receptors that often undergo autoproteolysis to produce α and ß subunits1-3. A tethered agonism mediated by the 'Stachel sequence' of the ß subunit has been proposed to have central roles in aGPCR activation4-6. Here we present three cryo-electron microscopy structures of aGPCRs coupled to the Gs heterotrimer. Two of these aGPCRs are activated by tethered Stachel sequences-the ADGRG2-ß-Gs complex and the ADGRG4-ß-Gs complex (in which ß indicates the ß subunit of the aGPCR)-and the other is the full-length ADGRG2 in complex with the exogenous ADGRG2 Stachel-sequence-derived peptide agonist IP15 (ADGRG2(FL)-IP15-Gs). The Stachel sequences of both ADGRG2-ß and ADGRG4-ß assume a U shape and insert deeply into the seven-transmembrane bundles. Constituting the FXφφφXφ motif (in which φ represents a hydrophobic residue), five residues of ADGRG2-ß or ADGRG4-ß extend like fingers to mediate binding to the seven-transmembrane domain and activation of the receptor. The structure of the ADGRG2(FL)-IP15-Gs complex reveals the structural basis for the improved binding affinity of IP15 compared with VPM-p15 and indicates that rational design of peptidic agonists could be achieved by exploiting aGPCR-ß structures. By converting the 'finger residues' to acidic residues, we develop a method to generate peptidic antagonists towards several aGPCRs. Collectively, our study provides structural and biochemical insights into the tethered activation mechanism of aGPCRs.


Asunto(s)
Péptidos , Receptores Acoplados a Proteínas G , Microscopía por Crioelectrón , Humanos , Péptidos/metabolismo , Dominios Proteicos , Receptores Acoplados a Proteínas G/metabolismo
7.
Mol Cell ; 80(1): 43-58.e7, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32937100

RESUMEN

Immune cell function depends on specific metabolic programs dictated by mitochondria, including nutrient oxidation, macromolecule synthesis, and post-translational modifications. Mitochondrial adaptations have been linked to acute and chronic inflammation, but the metabolic cues and precise mechanisms remain unclear. Here we reveal that histone deacetylase 3 (HDAC3) is essential for shaping mitochondrial adaptations for IL-1ß production in macrophages through non-histone deacetylation. In vivo, HDAC3 promoted lipopolysaccharide-induced acute inflammation and high-fat diet-induced chronic inflammation by enhancing NLRP3-dependent caspase-1 activation. HDAC3 configured the lipid profile in stimulated macrophages and restricted fatty acid oxidation (FAO) supported by exogenous fatty acids for mitochondria to acquire their adaptations and depolarization. Rather than affecting nuclear gene expression, HDAC3 translocated to mitochondria to deacetylate and inactivate an FAO enzyme, mitochondrial trifunctional enzyme subunit α. HDAC3 may serve as a controlling node that balances between acquiring mitochondrial adaptations and sustaining their fitness for IL-1ß-dependent inflammation.


Asunto(s)
Ácidos Grasos/metabolismo , Histona Desacetilasas/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Mitocondrias/metabolismo , Adulto , Animales , Caspasa 1/metabolismo , Femenino , Humanos , Inflamación/patología , Metabolismo de los Lípidos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Mitocondrias/ultraestructura , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Células Mieloides/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , Adulto Joven
8.
Mol Cell ; 77(2): 213-227.e5, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31735641

RESUMEN

Macrophages form a major cell population in the tumor microenvironment. They can be activated and polarized into tumor-associated macrophages (TAM) by the tumor-derived soluble molecules to promote tumor progression and metastasis. Here, we used comparative metabolomics coupled with biochemical and animal studies to show that cancer cells release succinate into their microenvironment and activate succinate receptor (SUCNR1) signaling to polarize macrophages into TAM. Furthermore, the results from in vitro and in vivo studies revealed that succinate promotes not only cancer cell migration and invasion but also cancer metastasis. These effects are mediated by SUCNR1-triggered PI3K-hypoxia-inducible factor 1α (HIF-1α) axis. Compared with healthy subjects and tumor-free lung tissues, serum succinate levels and lung cancer SUCNR1 expression were elevated in lung cancer patients, suggesting an important clinical relevance. Collectively, our findings indicate that the secreted tumor-derived succinate belongs to a novel class of cancer progression factors, controlling TAM polarization and promoting tumorigenic signaling.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Macrófagos/metabolismo , Metástasis de la Neoplasia/patología , Receptores Acoplados a Proteínas G/metabolismo , Ácido Succínico/metabolismo , Células A549 , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Células HT29 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células MCF-7 , Macrófagos/patología , Ratones Endogámicos C57BL , Células PC-3 , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
9.
Mol Cell ; 80(2): 263-278.e7, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33022274

RESUMEN

Cancer metastasis accounts for the major cause of cancer-related deaths. How disseminated cancer cells cope with hostile microenvironments in secondary site for full-blown metastasis is largely unknown. Here, we show that AMPK (AMP-activated protein kinase), activated in mouse metastasis models, drives pyruvate dehydrogenase complex (PDHc) activation to maintain TCA cycle (tricarboxylic acid cycle) and promotes cancer metastasis by adapting cancer cells to metabolic and oxidative stresses. This AMPK-PDHc axis is activated in advanced breast cancer and predicts poor metastasis-free survival. Mechanistically, AMPK localizes in the mitochondrial matrix and phosphorylates the catalytic alpha subunit of PDHc (PDHA) on two residues S295 and S314, which activates the enzymatic activity of PDHc and alleviates an inhibitory phosphorylation by PDHKs, respectively. Importantly, these phosphorylation events mediate PDHc function in cancer metastasis. Our study reveals that AMPK-mediated PDHA phosphorylation drives PDHc activation and TCA cycle to empower cancer cells adaptation to metastatic microenvironments for metastasis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Ciclo del Ácido Cítrico , Complejo Piruvato Deshidrogenasa/metabolismo , Animales , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular , Activación Enzimática , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Fosforilación , Fosfoserina/metabolismo , Transducción de Señal , Estrés Fisiológico , Análisis de Supervivencia
10.
Cell ; 149(5): 1098-111, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22632973

RESUMEN

Akt kinase plays a central role in cell growth, metabolism, and tumorigenesis. The TRAF6 E3 ligase orchestrates IGF-1-mediated Akt ubiquitination and activation. Here, we show that Akt ubiquitination is also induced by activation of ErbB receptors; unexpectedly, and in contrast to IGF-1 induced activation, the Skp2 SCF complex, not TRAF6, is a critical E3 ligase for ErbB-receptor-mediated Akt ubiquitination and membrane recruitment in response to EGF. Skp2 deficiency impairs Akt activation, Glut1 expression, glucose uptake and glycolysis, and breast cancer progression in various tumor models. Moreover, Skp2 overexpression correlates with Akt activation and breast cancer metastasis and serves as a marker for poor prognosis in Her2-positive patients. Finally, Skp2 silencing sensitizes Her2-overexpressing tumors to Herceptin treatment. Our study suggests that distinct E3 ligases are utilized by diverse growth factors for Akt activation and that targeting glycolysis sensitizes Her2-positive tumors to Herceptin treatment.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Transformación Celular Neoplásica , Proteínas F-Box/metabolismo , Glucólisis , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Receptor ErbB-2/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Trastuzumab , Ubiquitinación
11.
Mol Cell ; 74(5): 996-1009.e7, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-30975460

RESUMEN

Nucleotide-binding site leucine-rich repeat (NLR) receptors perceive pathogen effectors and trigger plant immunity. However, the mechanisms underlying NLR-triggered defense responses remain obscure. The recently discovered Pigm locus in rice encodes a cluster of NLRs, including PigmR, which confers broad-spectrum resistance to blast fungus. Here, we identify PIBP1 (PigmR-INTERACTING and BLAST RESISTANCE PROTEIN 1), an RRM (RNA-recognition motif) protein that specifically interacts with PigmR and other similar NLRs to trigger blast resistance. PigmR-promoted nuclear accumulation of PIBP1 ensures full blast resistance. We find that PIBP1 and a homolog, Os06 g02240, bind DNA and function as unconventional transcription factors at the promoters of the defense genes OsWAK14 and OsPAL1, activating their expression. Knockout of PIBP1 and Os06 g02240 greatly attenuated blast resistance. Collectively, our study discovers previously unappreciated RRM transcription factors that directly interact with NLRs to activate plant defense, establishing a direct link between transcriptional activation of immune responses with NLR-mediated pathogen perception.


Asunto(s)
Resistencia a la Enfermedad/genética , Proteínas NLR/genética , Oryza/genética , Enfermedades de las Plantas/genética , Proteínas de Plantas/genética , Factores de Transcripción/genética , Sitios de Unión , Hongos/patogenicidad , Regulación de la Expresión Génica de las Plantas , Oryza/microbiología , Enfermedades de las Plantas/microbiología , Inmunidad de la Planta/genética , Regiones Promotoras Genéticas , Unión Proteica/genética , Transducción de Señal/genética
12.
PLoS Biol ; 21(3): e3002033, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36928253

RESUMEN

Aging is a systemic process, which is a risk factor for impaired physiological functions, and finally death. The molecular mechanisms driving aging process and the associated cognitive decline are not fully understood. The hypothalamus acts as the arbiter that orchestrates systemic aging through neuroinflammatory signaling. Our recent findings revealed that Menin plays important roles in neuroinflammation and brain development. Here, we found that the hypothalamic Menin signaling diminished in aged mice, which correlates with systemic aging and cognitive deficits. Restoring Menin expression in ventromedial nucleus of hypothalamus (VMH) of aged mice extended lifespan, improved learning and memory, and ameliorated aging biomarkers, while inhibiting Menin in VMH of middle-aged mice induced premature aging and accelerated cognitive decline. We further found that Menin epigenetically regulates neuroinflammatory and metabolic pathways, including D-serine metabolism. Aging-associated Menin reduction led to impaired D-serine release by VMH-hippocampus neural circuit, while D-serine supplement rescued cognitive decline in aged mice. Collectively, VMH Menin serves as a key regulator of systemic aging and aging-related cognitive decline.


Asunto(s)
Envejecimiento , Disfunción Cognitiva , Hipotálamo , Animales , Ratones , Envejecimiento/genética , Envejecimiento/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Hipotálamo/metabolismo , Serina/metabolismo , Factores de Transcripción/metabolismo
13.
Circ Res ; 132(2): 187-204, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36583388

RESUMEN

BACKGROUND: NOTCH1 pathogenic variants are implicated in multiple types of congenital heart defects including hypoplastic left heart syndrome, where the left ventricle is underdeveloped. It is unknown how NOTCH1 regulates human cardiac cell lineage determination and cardiomyocyte proliferation. In addition, mechanisms by which NOTCH1 pathogenic variants lead to ventricular hypoplasia in hypoplastic left heart syndrome remain elusive. METHODS: CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 genome editing was utilized to delete NOTCH1 in human induced pluripotent stem cells. Cardiac differentiation was carried out by sequential modulation of WNT signaling, and NOTCH1 knockout and wild-type differentiating cells were collected at day 0, 2, 5, 10, 14, and 30 for single-cell RNA-seq. RESULTS: Human NOTCH1 knockout induced pluripotent stem cells are able to generate functional cardiomyocytes and endothelial cells, suggesting that NOTCH1 is not required for mesoderm differentiation and cardiovascular development in vitro. However, disruption of NOTCH1 blocks human ventricular-like cardiomyocyte differentiation but promotes atrial-like cardiomyocyte generation through shortening the action potential duration. NOTCH1 deficiency leads to defective proliferation of early human cardiomyocytes, and transcriptomic analysis indicates that pathways involved in cell cycle progression and mitosis are downregulated in NOTCH1 knockout cardiomyocytes. Single-cell transcriptomic analysis reveals abnormal cell lineage determination of cardiac mesoderm, which is manifested by the biased differentiation toward epicardial and second heart field progenitors at the expense of first heart field progenitors in NOTCH1 knockout cell populations. CONCLUSIONS: NOTCH1 is essential for human ventricular-like cardiomyocyte differentiation and proliferation through balancing cell fate determination of cardiac mesoderm and modulating cell cycle progression. Because first heart field progenitors primarily contribute to the left ventricle, we speculate that pathogenic NOTCH1 variants lead to biased differentiation of first heart field progenitors, blocked ventricular-like cardiomyocyte differentiation, and defective cardiomyocyte proliferation, which collaboratively contribute to left ventricular hypoplasia in hypoplastic left heart syndrome.


Asunto(s)
Síndrome del Corazón Izquierdo Hipoplásico , Células Madre Pluripotentes Inducidas , Humanos , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Diferenciación Celular/fisiología , Miocitos Cardíacos/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo
14.
PLoS Genet ; 18(6): e1010236, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35737725

RESUMEN

Congenital heart disease (CHD) is a common group of birth defects with a strong genetic contribution to their etiology, but historically the diagnostic yield from exome studies of isolated CHD has been low. Pleiotropy, variable expressivity, and the difficulty of accurately phenotyping newborns contribute to this problem. We hypothesized that performing exome sequencing on selected individuals in families with multiple members affected by left-sided CHD, then filtering variants by population frequency, in silico predictive algorithms, and phenotypic annotations from publicly available databases would increase this yield and generate a list of candidate disease-causing variants that would show a high validation rate. In eight of the nineteen families in our study (42%), we established a well-known gene/phenotype link for a candidate variant or performed confirmation of a candidate variant's effect on protein function, including variants in genes not previously described or firmly established as disease genes in the body of CHD literature: BMP10, CASZ1, ROCK1 and SMYD1. Two plausible variants in different genes were found to segregate in the same family in two instances suggesting oligogenic inheritance. These results highlight the need for functional validation and demonstrate that in the era of next-generation sequencing, multiplex families with isolated CHD can still bring high yield to the discovery of novel disease genes.


Asunto(s)
Exoma , Cardiopatías Congénitas , Proteínas Morfogenéticas Óseas/genética , Proteínas de Unión al ADN/genética , Exoma/genética , Frecuencia de los Genes , Estudios de Asociación Genética , Cardiopatías Congénitas/genética , Humanos , Recién Nacido , Linaje , Factores de Transcripción/genética , Secuenciación del Exoma , Quinasas Asociadas a rho/genética
15.
Proc Natl Acad Sci U S A ; 119(15): e2117004119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35394864

RESUMEN

GPR126 is a member of the adhesion G protein-coupled receptors (aGPCRs) that is essential for the normal development of diverse tissues, and its mutations are implicated in various pathological processes. Here, through screening 34 steroid hormones and their derivatives for cAMP production, we found that progesterone (P4) and 17-hydroxyprogesterone (17OHP) could specifically activate GPR126 and trigger its downstream Gi signaling by binding to the ligand pocket in the seven-transmembrane domain of the C-terminal fragment of GPR126. A detailed mutagenesis screening according to a computational simulated structure model indicated that K1001ECL2 and F1012ECL2 are key residues that specifically recognize 17OHP but not progesterone. Finally, functional analysis revealed that progesterone-triggered GPR126 activation promoted cell growth in vitro and tumorigenesis in vivo, which involved Gi-SRC pathways in a triple-negative breast cancer model. Collectively, our work identified a membrane receptor for progesterone/17OHP and delineated the mechanisms by which GPR126 participated in potential tumor progression in triple-negative breast cancer, which will enrich our understanding of the functions and working mechanisms of both the aGPCR member GPR126 and the steroid hormone progesterone.


Asunto(s)
Progesterona , Receptores Acoplados a Proteínas G , Receptores de Progesterona , Neoplasias de la Mama Triple Negativas , 17-alfa-Hidroxiprogesterona/metabolismo , Línea Celular Tumoral , Humanos , Progesterona/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
16.
Pharmacol Rev ; 74(3): 462-505, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35710133

RESUMEN

The concept of local formation of angiotensin II in the kidney has changed over the last 10-15 years. Local synthesis of angiotensinogen in the proximal tubule has been proposed, combined with prorenin synthesis in the collecting duct. Binding of prorenin via the so-called (pro)renin receptor has been introduced, as well as megalin-mediated uptake of filtered plasma-derived renin-angiotensin system (RAS) components. Moreover, angiotensin metabolites other than angiotensin II [notably angiotensin-(1-7)] exist, and angiotensins exert their effects via three different receptors, of which angiotensin II type 2 and Mas receptors are considered renoprotective, possibly in a sex-specific manner, whereas angiotensin II type 1 (AT1) receptors are believed to be deleterious. Additionally, internalized angiotensin II may stimulate intracellular receptors. Angiotensin-converting enzyme 2 (ACE2) not only generates angiotensin-(1-7) but also acts as coronavirus receptor. Multiple, if not all, cardiovascular diseases involve the kidney RAS, with renal AT1 receptors often being claimed to exert a crucial role. Urinary RAS component levels, depending on filtration, reabsorption, and local release, are believed to reflect renal RAS activity. Finally, both existing drugs (RAS inhibitors, cyclooxygenase inhibitors) and novel drugs (angiotensin receptor/neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors, soluble ACE2) affect renal angiotensin formation, thereby displaying cardiovascular efficacy. Particular in the case of the latter three, an important question is to what degree they induce renoprotection (e.g., in a renal RAS-dependent manner). This review provides a unifying view, explaining not only how kidney angiotensin formation occurs and how it is affected by drugs but also why drugs are renoprotective when altering the renal RAS. SIGNIFICANCE STATEMENT: Angiotensin formation in the kidney is widely accepted but little understood, and multiple, often contrasting concepts have been put forward over the last two decades. This paper offers a unifying view, simultaneously explaining how existing and novel drugs exert renoprotection by interfering with kidney angiotensin formation.


Asunto(s)
Angiotensinógeno , Enfermedades Cardiovasculares , Femenino , Humanos , Masculino , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Angiotensinógeno/metabolismo , Enfermedades Cardiovasculares/metabolismo , Sistemas de Liberación de Medicamentos , Riñón/irrigación sanguínea , Riñón/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina , Inhibidores del Cotransportador de Sodio-Glucosa 2/metabolismo
17.
J Neurosci ; 43(46): 7831-7841, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37714708

RESUMEN

Languages come in different forms but have shared meanings to convey. Some meanings are expressed by sentence structure and morphologic inflections rather than content words, such as indicating time frame using tense. This fMRI study investigates whether there is cross-language common representation of grammatical meanings that can be identified from neural signatures in the bilingual human brain. Based on the representations in intersentence neural similarity space, identifying grammatical construction of a sentence in one language by models trained on the other language resulted in reliable accuracy. By contrast, cross-language identification of grammatical construction by spatially matched activation patterns was only marginally accurate. Brain locations representing grammatical meaning in the two languages were interleaved in common regions bilaterally. The locations of voxels representing grammatical features in the second language were more varied across individuals than voxels representing the first language. These findings suggest grammatical meaning is represented by language-specific activation patterns, which is different from lexical semantics. Commonality of grammatical meaning is neurally reflected only in the interstimulus similarity space.SIGNIFICANCE STATEMENT Whether human brain encodes sentence-level meanings beyond content words in different languages similarly has been a long-standing question. We characterize the neural representations of similar grammatical meanings in different languages. Using complementary analytic approaches on fMRI data, we show that the same grammatical meaning is neurally represented as the common pattern of neural distances between sentences. The results suggest the possibility of identifying specific grammatical meaning expressed by different morphologic and syntactic implementations of different languages. The neural realization of grammatical meanings is constrained by the specific language being used, but the relationships between the neural representations of sentences are preserved across languages. These findings have some theoretical implications on a distinction between grammar and lexical meanings.


Asunto(s)
Lenguaje , Semántica , Humanos , Lingüística , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Mapeo Encefálico
18.
Am J Physiol Cell Physiol ; 326(2): C331-C347, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38047307

RESUMEN

Diabetic cardiomyopathy (dCM) is a major complication of diabetes; however, specific treatments for dCM are currently lacking. RTA 408, a semisynthetic triterpenoid, has shown therapeutic potential against various diseases by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. We established in vitro and in vivo models using high glucose toxicity and db/db mice, respectively, to simulate dCM. Our results demonstrated that RTA 408 activated Nrf2 and alleviated various dCM-related cardiac dysfunctions, both in vivo and in vitro. Additionally, it was found that silencing the Nrf2 gene eliminated the cardioprotective effect of RTA 408. RTA 408 ameliorated oxidative stress in dCM mice and high glucose-exposed H9C2 cells by activating Nrf2, inhibiting mitochondrial fission, exerting anti-inflammatory effects through the Nrf2/NF-κB axis, and ultimately suppressing apoptosis, thereby providing cardiac protection against dCM. These findings provide valuable insights for potential dCM treatments.NEW & NOTEWORTHY We demonstrated first that the nuclear factor erythroid 2-related factor 2 (Nrf2) activator RTA 408 has a protective effect against diabetic cardiomyopathy. We found that RTA 408 could stimulate the nuclear entry of Nrf2 protein, regulate the mitochondrial fission-fusion balance, and redistribute p65, which significantly alleviated the oxidative stress level in cardiomyocytes, thereby reducing apoptosis and inflammation, and protecting the systolic and diastolic functions of the heart.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Triterpenos , Ratones , Animales , FN-kappa B/genética , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Dinámicas Mitocondriales , Estrés Oxidativo , Inflamación/metabolismo , Triterpenos/metabolismo , Triterpenos/farmacología , Triterpenos/uso terapéutico , Miocitos Cardíacos/metabolismo , Glucosa/metabolismo , Diabetes Mellitus/metabolismo
19.
J Biol Chem ; 299(4): 104570, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36870679

RESUMEN

Liver kinase B1 (LKB1) is a serine-threonine kinase that participates in multiple cellular and biological processes, including energy metabolism, cell polarity, cell proliferation, cell migration, and many others. LKB1 is initially identified as a germline-mutated causative gene in Peutz-Jeghers syndrome and is commonly regarded as a tumor suppressor due to frequent inactivation in a variety of cancers. LKB1 directly binds and activates its downstream kinases including the AMP-activated protein kinase (AMPK) and AMPK-related kinases by phosphorylation, which has been intensively investigated for the past decades. An increasing number of studies have uncovered the posttranslational modifications (PTMs) of LKB1 and consequent changes in its localization, activity, and interaction with substrates. The alteration in LKB1 function as a consequence of genetic mutations and aberrant upstream signaling regulation leads to tumor development and progression. Here, we review current knowledge about the mechanism of LKB1 in cancer and the contributions of PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, prenylation, and others, to the regulation of LKB1 function, offering new insights into the therapeutic strategies in cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Neoplasias , Procesamiento Proteico-Postraduccional , Humanos , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Hígado/metabolismo , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias/enzimología
20.
J Biol Chem ; 299(6): 104814, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37178919

RESUMEN

Epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma (LUAD) patients often respond to EGFR tyrosine kinase inhibitors (TKIs) initially but eventually develop resistance to TKIs. The switch of EGFR downstream signaling from TKI-sensitive to TKI-insensitive is a critical mechanism-driving resistance to TKIs. Identification of potential therapies to target EGFR effectively is a potential strategy to treat TKI-resistant LUADs. In this study, we developed a small molecule diarylheptanoid 35d, a curcumin derivative, that effectively suppressed EGFR protein expression, killed multiple TKI-resistant LUAD cells in vitro, and suppressed tumor growth of EGFR-mutant LUAD xenografts with variant TKI-resistant mechanisms including EGFR C797S mutations in vivo. Mechanically, 35d triggers heat shock protein 70-mediated lysosomal pathway through transcriptional activation of several components in the pathway, such as HSPA1B, to induce EGFR protein degradation. Interestingly, higher HSPA1B expression in LUAD tumors associated with longer survival of EGFR-mutant, TKI-treated patients, suggesting the role of HSPA1B on retarding TKI resistance and providing a rationale for combining 35d with EGFR TKIs. Our data showed that combination of 35d significantly inhibits tumor reprogression on osimertinib and prolongs mice survival. Overall, our results suggest 35d as a promising lead compound to suppress EGFR expression and provide important insights into the development of combination therapies for TKI-resistant LUADs, which could have translational potential for the treatment of this deadly disease.


Asunto(s)
Adenocarcinoma del Pulmón , Diarilheptanoides , Resistencia a Antineoplásicos , Neoplasias Pulmonares , Animales , Humanos , Ratones , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Línea Celular Tumoral , Diarilheptanoides/farmacología , Receptores ErbB/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Lisosomas/metabolismo , Mutación , Inhibidores de Proteínas Quinasas/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda