Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35216100

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the number one cause of chronic liver disease worldwide, with 25% of these patients developing nonalcoholic steatohepatitis (NASH). NASH significantly increases the risk of cirrhosis and decompensated liver failure. Past studies in rodent models have shown that glycine-N-methyltransferase (GNMT) knockout results in rapid steatosis, fibrosis, and hepatocellular carcinoma progression. However, the attenuation of GNMT in subjects with NASH and the molecular basis for its impact on the disease process is still unclear. To address this knowledge gap, we show the reduction of GNMT protein levels in the liver of NASH subjects compared to healthy controls. To gain insight into the impact of decreased GNMT in the disease process, we performed global label-free proteome studies on the livers from a murine modified amylin diet-based model of NASH. Histological and molecular characterization of the animal model demonstrate a high resemblance to human disease. We found that a reduction of GNMT leads to a significant increase in S-adenosylmethionine (AdoMet), an essential metabolite for transmethylation reactions and a substrate for polyamine synthesis. Further targeted proteomic and metabolomic studies demonstrated a decrease in GNMT transmethylation, increased flux through the polyamine pathway, and increased oxidative stress production contributing to NASH pathogenesis.


Asunto(s)
Cirrosis Hepática/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo/fisiología , Poliaminas/metabolismo , S-Adenosilmetionina/metabolismo , Adulto , Animales , Carcinoma Hepatocelular/metabolismo , Modelos Animales de Enfermedad , Femenino , Glicina N-Metiltransferasa/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Oxidación-Reducción , Proteómica/métodos
2.
Artículo en Inglés | MEDLINE | ID: mdl-39420421

RESUMEN

OBJECTIVE: Caloric restriction (CR) is known to enhance insulin sensitivity and reduce the risk of metabolic disorders; however, its molecular mechanisms are not fully understood. This study aims to elucidate specific proteins and pathways responsible for these benefits. METHODS: We examined adipose tissue from participants in the Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy Phase 2 (CALERIE 2) study, comparing proteomic profiles from individuals after 12 and 24 months of CR with baseline and an ad libitum group. Biochemical and cell-specific physiological approaches complemented these analyses. RESULTS: Our data revealed that CR upregulates prostacyclin synthase (PTGIS) in adipose tissue, an enzyme crucial for producing prostacyclin (PGI2). PGI2 improves the ability of insulin to stimulate the tether-containing UBX domain for GLUT4 (TUG) cleavage pathway, which is essential for glucose uptake regulation. Additionally, iloprost, a PGI2 analog, was shown to increase insulin receptor density on cell membranes, increasing glucose uptake in human adipocytes. CR also reduces carbonylation of GLUT4, a modification that is detrimental to GLUT4 function. CONCLUSIONS: CR enhances insulin sensitivity by promoting PTGIS expression and stimulating the TUG cleavage pathway, leading to increased GLUT4 translocation to the cell surface and decreased GLUT4 carbonylation. These findings shed light on the complex molecular mechanisms through which CR favorably impacts insulin sensitivity and metabolic health.

3.
Nat Struct Mol Biol ; 31(2): 311-322, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38177675

RESUMEN

Targeted protein degradation (TPD) by PROTAC (proteolysis-targeting chimera) and molecular glue small molecules is an emerging therapeutic strategy. To expand the roster of E3 ligases that can be utilized for TPD, we describe the discovery and biochemical characterization of small-molecule ligands targeting the E3 ligase KLHDC2. Furthermore, we functionalize these KLHDC2-targeting ligands into KLHDC2-based BET-family and AR PROTAC degraders and demonstrate KLHDC2-dependent target-protein degradation. Additionally, we offer insight into the assembly of the KLHDC2 E3 ligase complex. Using biochemical binding studies, X-ray crystallography and cryo-EM, we show that the KLHDC2 E3 ligase assembles into a dynamic tetramer held together via its own C terminus, and that this assembly can be modulated by substrate and ligand engagement.


Asunto(s)
Ubiquitina-Proteína Ligasas , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ligandos
4.
Clin Cancer Res ; 30(16): 3549-3563, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38819400

RESUMEN

PURPOSE: Estrogen receptor (ER) alpha signaling is a known driver of ER-positive (ER+)/human epidermal growth factor receptor 2 negative (HER2-) breast cancer. Combining endocrine therapy (ET) such as fulvestrant with CDK4/6, mTOR, or PI3K inhibitors has become a central strategy in the treatment of ER+ advanced breast cancer. However, suboptimal ER inhibition and resistance resulting from the ESR1 mutation dictates that new therapies are needed. EXPERIMENTAL DESIGN: A medicinal chemistry campaign identified vepdegestrant (ARV-471), a selective, orally bioavailable, and potent small molecule PROteolysis-TArgeting Chimera (PROTAC) degrader of ER. We used biochemical and intracellular target engagement assays to demonstrate the mechanism of action of vepdegestrant, and ESR1 wild-type (WT) and mutant ER+ preclinical breast cancer models to demonstrate ER degradation-mediated tumor growth inhibition (TGI). RESULTS: Vepdegestrant induced ≥90% degradation of wild-type and mutant ER, inhibited ER-dependent breast cancer cell line proliferation in vitro, and achieved substantial TGI (87%-123%) in MCF7 orthotopic xenograft models, better than those of the ET agent fulvestrant (31%-80% TGI). In the hormone independent (HI) mutant ER Y537S patient-derived xenograft (PDX) breast cancer model ST941/HI, vepdegestrant achieved tumor regression and was similarly efficacious in the ST941/HI/PBR palbociclib-resistant model (102% TGI). Vepdegestrant-induced robust tumor regressions in combination with each of the CDK4/6 inhibitors palbociclib, abemaciclib, and ribociclib; the mTOR inhibitor everolimus; and the PI3K inhibitors alpelisib and inavolisib. CONCLUSIONS: Vepdegestrant achieved greater ER degradation in vivo compared with fulvestrant, which correlated with improved TGI, suggesting vepdegestrant could be a more effective backbone ET for patients with ER+/HER2- breast cancer.


Asunto(s)
Neoplasias de la Mama , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Transducción de Señal , Serina-Treonina Quinasas TOR , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Femenino , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Ratones , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/antagonistas & inhibidores , Piperazinas/farmacología , Piperazinas/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/administración & dosificación , Receptores de Estrógenos/metabolismo , Piridinas/administración & dosificación , Piridinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proliferación Celular/efectos de los fármacos
5.
J Med Chem ; 67(2): 1262-1313, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38180485

RESUMEN

The identification of VHL-binding proteolysis targeting chimeras (PROTACs) that potently degrade the BRM protein (also known as SMARCA2) in SW1573 cell-based experiments is described. These molecules exhibit between 10- and 100-fold degradation selectivity for BRM over the closely related paralog protein BRG1 (SMARCA4). They also selectively impair the proliferation of the H1944 "BRG1-mutant" NSCLC cell line, which lacks functional BRG1 protein and is thus highly dependent on BRM for growth, relative to the wild-type Calu6 line. In vivo experiments performed with a subset of compounds identified PROTACs that potently and selectively degraded BRM in the Calu6 and/or the HCC2302 BRG1 mutant NSCLC xenograft models and also afforded antitumor efficacy in the latter system. Subsequent PK/PD analysis established a need to achieve strong BRM degradation (>95%) in order to trigger meaningful antitumor activity in vivo. Intratumor quantitation of mRNA associated with two genes whose transcription was controlled by BRM (PLAU and KRT80) also supported this conclusion.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Quimera Dirigida a la Proteólisis , Xenoinjertos , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Neoplasias Pulmonares/genética , Factores de Transcripción/genética , ADN Helicasas/genética , Proteínas Nucleares/genética
6.
Sci Transl Med ; 15(715): eade2966, 2023 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-37756380

RESUMEN

Hepatic fibrosis is the primary determinant of mortality in patients with metabolic dysfunction-associated steatohepatitis (MASH). Transforming growth factor-ß (TGFß), a master profibrogenic cytokine, is a promising therapeutic target that has not yet been translated into an effective therapy in part because of liabilities associated with systemic TGFß antagonism. We have identified that soluble folate receptor γ (FOLR3), which is expressed in humans but not in rodents, is a secreted protein that is elevated in the livers of patients with MASH but not in those with metabolic dysfunction-associated steatotic liver disease, those with type II diabetes, or healthy individuals. Global proteomics showed that FOLR3 was the most highly significant MASH-specific protein and was positively correlated with increasing fibrosis stage, consistent with stimulation of activated hepatic stellate cells (HSCs), which are the key fibrogenic cells in the liver. Exposure of HSCs to exogenous FOLR3 led to elevated extracellular matrix (ECM) protein production, an effect synergistically potentiated by TGFß1. We found that FOLR3 interacts with the serine protease HTRA1, a known regulator of TGFBR, and activates TGFß signaling. Administration of human FOLR3 to mice induced severe bridging fibrosis and an ECM pattern resembling human MASH. Our study thus uncovers a role of FOLR3 in enhancing fibrosis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hígado Graso , Humanos , Animales , Ratones , Factor de Crecimiento Transformador beta , Células Estrelladas Hepáticas , Ácido Fólico
7.
Nat Commun ; 13(1): 6814, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36357397

RESUMEN

The mammalian SWItch/Sucrose Non-Fermentable (SWI/SNF) helicase SMARCA4 is frequently mutated in cancer and inactivation results in a cellular dependence on its paralog, SMARCA2, thus making SMARCA2 an attractive synthetic lethal target. However, published data indicates that achieving a high degree of selective SMARCA2 inhibition is likely essential to afford an acceptable therapeutic index, and realizing this objective is challenging due to the homology with the SMARCA4 paralog. Herein we report the discovery of a potent and selective SMARCA2 proteolysis-targeting chimera molecule (PROTAC), A947. Selective SMARCA2 degradation is achieved in the absence of selective SMARCA2/4 PROTAC binding and translates to potent in vitro growth inhibition and in vivo efficacy in SMARCA4 mutant models, compared to wild type models. Global ubiquitin mapping and proteome profiling reveal no unexpected off-target degradation related to A947 treatment. Our study thus highlights the ability to transform a non-selective SMARCA2/4-binding ligand into a selective and efficacious in vivo SMARCA2-targeting PROTAC, and thereby provides a potential new therapeutic opportunity for patients whose tumors contain SMARCA4 mutations.


Asunto(s)
Neoplasias , Animales , Humanos , Proteolisis , Neoplasias/genética , Mutación , Mamíferos , Factores de Transcripción/genética , ADN Helicasas/genética , Proteínas Nucleares/genética
8.
Neuropsychologia ; 98: 68-84, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27720949

RESUMEN

Understanding the neural processes that underlie learning to read can provide a scientific foundation for literacy education but studying these processes in real-world contexts remains challenging. We present behavioural data from adult participants learning to read artificial words and name artificial objects over two days. Learning profiles and generalisation confirmed that componential learning of visual-verbal associations distinguishes reading from object naming. Functional MRI data collected on the second day allowed us to identify the neural systems that support componential reading as distinct from systems supporting holistic visual-verbal associations in object naming. Results showed increased activation in posterior ventral occipitotemporal (vOT), parietal, and frontal cortices when reading an artificial orthography compared to naming artificial objects, and the reverse profile in anterior vOT regions. However, activation differences between trained and untrained words were absent, suggesting a lack of cortical representations for whole words. Despite this, hippocampal responses provided some evidence for overnight consolidation of both words and objects learned on day 1. The comparison between neural activity for artificial words and objects showed extensive overlap with systems differentially engaged for real object naming and English word/pseudoword reading in the same participants. These findings therefore provide evidence that artificial learning paradigms offer an alternative method for studying the neural systems supporting language and literacy. Implications for literacy acquisition are discussed.


Asunto(s)
Asociación , Mapeo Encefálico , Encéfalo/fisiología , Recuerdo Mental/fisiología , Lectura , Aprendizaje Verbal/fisiología , Estimulación Acústica , Adolescente , Adulto , Encéfalo/diagnóstico por imagen , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Nombres , Oxígeno/sangre , Reconocimiento Visual de Modelos , Fonética , Estimulación Luminosa , Tiempo de Reacción , Vocabulario , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA