Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 291(25): 13028-39, 2016 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-27129247

RESUMO

Developmental signals in metazoans play critical roles in inducing cell differentiation from multipotent progenitors. The existing paradigm posits that the signals operate directly through their downstream transcription factors to activate expression of cell type-specific genes, which are the hallmark of cell identity. We have investigated the mechanism through which Wnt signaling induces osteoblast differentiation in an osteoblast-adipocyte bipotent progenitor cell line. Unexpectedly, Wnt3a acutely suppresses the expression of a large number of genes while inducing osteoblast differentiation. The suppressed genes include Pparg and Cebpa, which encode adipocyte-specifying transcription factors and suppression of which is sufficient to induce osteoblast differentiation. The large scale gene suppression induced by Wnt3a corresponds to a global decrease in histone acetylation, an epigenetic modification that is associated with gene activation. Mechanistically, Wnt3a does not alter histone acetyltransferase or deacetylase activities but, rather, decreases the level of acetyl-CoA in the nucleus. The Wnt-induced decrease in histone acetylation is independent of ß-catenin signaling but, rather, correlates with suppression of glucose metabolism in the tricarboxylic acid cycle. Functionally, preventing histone deacetylation by increasing nucleocytoplasmic acetyl-CoA levels impairs Wnt3a-induced osteoblast differentiation. Thus, Wnt signaling induces osteoblast differentiation in part through histone deacetylation and epigenetic suppression of an alternative cell fate.


Assuntos
Acetilcoenzima A/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Osteoblastos/fisiologia , Via de Sinalização Wnt , Proteína Wnt3A/fisiologia , Acetilação , Animais , Linhagem Celular , Ácido Cítrico/metabolismo , Ciclo do Ácido Cítrico , Expressão Gênica , Inativação Gênica , Glucose/metabolismo , Histonas/metabolismo , Camundongos , Processamento de Proteína Pós-Traducional
2.
PLoS Genet ; 10(1): e1004145, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24497849

RESUMO

WNT signaling has been implicated in both embryonic and postnatal bone formation. However, the pertinent WNT ligands and their downstream signaling mechanisms are not well understood. To investigate the osteogenic capacity of WNT7B and WNT5A, both normally expressed in the developing bone, we engineered mouse strains to express either protein in a Cre-dependent manner. Targeted induction of WNT7B, but not WNT5A, in the osteoblast lineage dramatically enhanced bone mass due to increased osteoblast number and activity; this phenotype began in the late-stage embryo and intensified postnatally. Similarly, postnatal induction of WNT7B in Runx2-lineage cells greatly stimulated bone formation. WNT7B activated mTORC1 through PI3K-AKT signaling. Genetic disruption of mTORC1 signaling by deleting Raptor in the osteoblast lineage alleviated the WNT7B-induced high-bone-mass phenotype. Thus, WNT7B promotes bone formation in part through mTORC1 activation.


Assuntos
Complexos Multiproteicos/genética , Osteogênese/genética , Proteínas Proto-Oncogênicas/genética , Serina-Treonina Quinases TOR/genética , Proteínas Wnt/genética , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/biossíntese , Osteoblastos/citologia , Proteínas Proto-Oncogênicas/biossíntese , Serina-Treonina Quinases TOR/biossíntese , Proteínas Wnt/biossíntese , Via de Sinalização Wnt , Proteína Wnt-5a
3.
Curr Osteoporos Rep ; 12(4): 433-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25200872

RESUMO

Osteoblasts, the chief bone-making cells in the body, are a focus of osteoporosis research. Although teriparatide, a synthetic fragment of the human parathyroid hormone (PTH), has been an effective bone anabolic drug, there remains a clinical need for additional therapeutics that safely stimulates osteoblast number and function. Work in the past several decades has provided unprecedented clarity about the roles of growth factors and transcription factors in regulating osteoblast differentiation and activity, but whether these factors may regulate cellular metabolism to influence cell fate and function has been largely unexplored. The past few years have witnessed a resurgence of interest in the cellular metabolism of osteoblasts, with the hope that elucidation of their metabolic profile may open new avenues for developing bone anabolic agents. Here we review the current understanding about glucose metabolism in osteoblasts.


Assuntos
Glucose/metabolismo , Osteoblastos/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos , Modelos Animais , Osteoblastos/citologia , Osteogênese/fisiologia , Hormônio Paratireóideo/fisiologia
4.
Br J Pharmacol ; 181(13): 2033-2052, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38486310

RESUMO

BACKGROUND AND PURPOSE: RO7502175 is an afucosylated antibody designed to eliminate C-C motif chemokine receptor 8 (CCR8)+ Treg cells in the tumour microenvironment through enhanced antibody-dependent cellular cytotoxicity (ADCC). EXPERIMENTAL APPROACH: We report findings from preclinical studies characterizing pharmacology, pharmacokinetics (PK)/pharmacodynamics (PD) and safety profile of RO7502175 and discuss the translational PK/PD approach used to inform first-in-human (FiH) dosing strategy and clinical development in solid tumour indications. KEY RESULTS: RO7502175 demonstrated selective ADCC against human CCR8+ Treg cells from dissociated tumours in vitro. In cynomolgus monkeys, RO7502175 exhibited a biphasic concentration-time profile consistent with immunoglobulin G1 (IgG1) antibodies, reduced CCR8+ Treg cells in the blood, induced minimal and transient cytokine secretion, and was well tolerated with a no-observed-adverse-effect level (NOAEL) of 100 mg·kg-1. Moreover, RO7502175 caused minimal cytokine release from peripheral blood mononuclear cells (PBMCs) in vitro. A quantitative model was developed to capture surrogate anti-murine CCR8 antibody PK/PD and tumour dynamics in mice and RO7502175 PK/PD in cynomolgus monkeys. Subsequently, the model was used to project RO7502175 human PK and receptor occupancy (RO) in patients. Because traditional approaches resulted in a low FiH dose for this molecule, even with its superior preclinical safety profile, an integrated approach based on the totality of preclinical data and modelling insights was used for starting dose selection. CONCLUSION AND IMPLICATIONS: This work demonstrates a translational research strategy for collecting and utilizing relevant nonclinical data, developing a mechanistic PK/PD model and using a comprehensive approach to inform clinical study design for RO7502175.


Assuntos
Macaca fascicularis , Receptores CCR8 , Linfócitos T Reguladores , Animais , Humanos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Receptores CCR8/antagonistas & inibidores , Receptores CCR8/imunologia , Camundongos , Feminino , Masculino , Pesquisa Translacional Biomédica , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Relação Dose-Resposta a Droga , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos
5.
Gastroenterology ; 139(6): 2038-49, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20816838

RESUMO

BACKGROUND & AIMS: The transition of gastric epithelial mucous neck cells (NCs) to digestive enzyme-secreting zymogenic cells (ZCs) involves an increase in rough endoplasmic reticulum (ER) and formation of many large secretory vesicles. The transcription factor MIST1 is required for granulogenesis of ZCs. The transcription factor XBP1 binds the Mist1 promoter and induces its expression in vitro and expands the ER in other cell types. We investigated whether XBP1 activates Mist1 to regulate ZC differentiation. METHODS: Xbp1 was inducibly deleted in mice using a tamoxifen/Cre-loxP system; effects on ZC size and structure (ER and granule formation) and gastric differentiation were studied and quantified for up to 13 months after deletion using morphologic, immunofluorescence, quantitative reverse-transcriptase polymerase chain reaction, and immunoblot analyses. Interactions between XBP1 and the Mist1 promoter were studied by chromatin immunoprecipitation from mouse stomach and in XBP1-transfected gastric cell lines. RESULTS: Tamoxifen-induced deletion of Xbp1 (Xbp1Δ) did not affect survival of ZCs but prevented formation of their structure. Xbp1Δ ZCs shrank 4-fold, compared with those of wild-type mice, with granulogenesis and cell shape abnormalities and disrupted rough ER. XBP1 was required and sufficient for transcriptional activation of MIST1. ZCs that developed in the absence of XBP1 induced ZC markers (intrinsic factor, pepsinogen C) but showed abnormal retention of progenitor NC markers. CONCLUSIONS: XBP1 controls the transcriptional regulation of ZC structural development; it expands the lamellar rough ER and induces MIST1 expression to regulate formation of large granules. XBP1 is also required for loss of mucous NC markers as ZCs form.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Celulas Principais Gástricas/citologia , Celulas Principais Gástricas/fisiologia , Proteínas de Ligação a DNA/genética , Retículo Endoplasmático Rugoso/fisiologia , Fatores de Transcrição/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Celulas Principais Gástricas/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Integrases/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Regiões Promotoras Genéticas/fisiologia , Fatores de Transcrição de Fator Regulador X , Vesículas Secretórias/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a X-Box
6.
Sci Immunol ; 5(45)2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32220977

RESUMO

During cytotoxic T cell activation, lymphocyte function-associated antigen-1 (LFA-1) engages its ligands on antigen-presenting cells (APCs) or target cells to enhance T cell priming or lytic activity. Inhibiting LFA-1 dampens T cell-dependent symptoms in inflammation, autoimmune diseases, and graft-versus-host disease. However, the therapeutic potential of augmenting LFA-1 function is less explored. Here, we show that genetic deletion or inhibition of mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) enhances LFA-1 activation on CD8 T cells and improves their adherence to APCs or LFA-1 ligand. In addition, loss of Map4k4 increases CD8 T cell priming, which culminates in enhanced antigen-dependent activation, proliferation, cytokine production, and cytotoxic activity, resulting in impaired tumor growth and improved response to viral infection. LFA-1 inhibition reverses these phenotypes. The ERM (ezrin, radixin, and moesin) proteins reportedly regulate T cell-APC conjugation, but the molecular regulator and effector of ERM proteins in T cells have not been defined. In this study, we demonstrate that the ERM proteins serve as mediators between MAP4K4 and LFA-1. Last, systematic analyses of many organs revealed that inducible whole-body deletion of Map4k4 in adult animals is tolerated under homeostatic conditions. Our results uncover MAP4K4 as a potential target to augment antitumor and antiviral immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Neoplasias/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Vírus/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Modelos Animais de Doenças , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética
7.
Sci Signal ; 9(409): ra2, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26732762

RESUMO

Autophagy is a catabolic cellular mechanism that degrades dysfunctional proteins and organelles. Atherosclerotic plaque formation is enhanced in mice with macrophages deficient for the critical autophagy protein ATG5. We showed that exposure of macrophages to lipids that promote atherosclerosis increased the abundance of the autophagy chaperone p62 and that p62 colocalized with polyubiquitinated proteins in cytoplasmic inclusions, which are characterized by insoluble protein aggregates. ATG5-null macrophages developed further p62 accumulation at the sites of large cytoplasmic ubiquitin-positive inclusion bodies. Aortas from atherosclerotic mice and plaques from human endarterectomy samples showed increased abundance of p62 and polyubiquitinated proteins that colocalized with plaque macrophages, suggesting that p62-enriched protein aggregates were characteristic of atherosclerosis. The formation of the cytoplasmic inclusions depended on p62 because lipid-loaded p62-null macrophages accumulated polyubiquitinated proteins in a diffuse cytoplasmic pattern. Lipid-loaded p62-null macrophages also exhibited increased secretion of interleukin-1ß (IL-1ß) and had an increased tendency to undergo apoptosis, which depended on the p62 ubiquitin-binding domain and at least partly involved p62-mediated clearance of NLRP3 inflammasomes. Consistent with our in vitro observations, p62-deficient mice formed greater numbers of more complex atherosclerotic plaques, and p62 deficiency further increased atherosclerotic plaque burden in mice with a macrophage-specific ablation of ATG5. Together, these data suggested that sequestration of cytotoxic ubiquitinated proteins by p62 protects against atherogenesis, a condition in which the clearance of protein aggregates is disrupted.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aterosclerose/metabolismo , Proteínas de Choque Térmico/metabolismo , Corpos de Inclusão/metabolismo , Macrófagos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apoptose/genética , Aterosclerose/genética , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Citoplasma/genética , Citoplasma/metabolismo , Proteínas de Choque Térmico/genética , Humanos , Immunoblotting , Inflamassomos/genética , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR , Poliubiquitina , Interferência de RNA , Proteína Sequestossoma-1 , Proteínas Ubiquitinadas/metabolismo
8.
J Bone Miner Res ; 30(11): 1959-68, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25990470

RESUMO

Teriparatide, a recombinant peptide corresponding to amino acids 1-34 of human parathyroid hormone (PTH), has been an effective bone anabolic drug for over a decade. However, the mechanism whereby PTH stimulates bone formation remains incompletely understood. Here we report that in cultures of osteoblast-lineage cells, PTH stimulates glucose consumption and lactate production in the presence of oxygen, a hallmark of aerobic glycolysis, also known as Warburg effect. Experiments with radioactively labeled glucose demonstrate that PTH suppresses glucose entry into the tricarboxylic acid cycle (TCA cycle). Mechanistically, the increase in aerobic glycolysis is secondary to insulin-like growth factor (Igf) signaling induced by PTH, whereas the metabolic effect of Igf is dependent on activation of mammalian target of rapamycin complex 2 (mTORC2). Importantly, pharmacological perturbation of glycolysis suppresses the bone anabolic effect of intermittent PTH in the mouse. Thus, stimulation of aerobic glycolysis via Igf signaling contributes to bone anabolism in response to PTH.


Assuntos
Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Glicólise/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/metabolismo , Hormônio Paratireóideo/farmacologia , Transdução de Sinais/efeitos dos fármacos , Aerobiose/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Osso e Ossos/diagnóstico por imagem , Dióxido de Carbono/metabolismo , Isótopos de Carbono , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , AMP Cíclico/metabolismo , Glucose/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Tíbia/diagnóstico por imagem , Tíbia/efeitos dos fármacos , Microtomografia por Raio-X
9.
J Clin Invest ; 125(2): 551-62, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25562323

RESUMO

WNT signaling stimulates bone formation by increasing both the number of osteoblasts and their protein-synthesis activity. It is not clear how WNT augments the capacity of osteoblast progenitors to meet the increased energetic and synthetic needs associated with mature osteoblasts. Here, in cultured osteoblast progenitors, we determined that WNT stimulates glutamine catabolism through the tricarboxylic acid (TCA) cycle and consequently lowers intracellular glutamine levels. The WNT-induced reduction of glutamine concentration triggered a general control nonderepressible 2-mediated (GCN2-mediated) integrated stress response (ISR) that stimulated expression of genes responsible for amino acid supply, transfer RNA (tRNA) aminoacylation, and protein folding. WNT-induced glutamine catabolism and ISR were ß-catenin independent, but required mammalian target of rapamycin complex 1 (mTORC1) activation. In a hyperactive WNT signaling mouse model of human osteosclerosis, inhibition of glutamine catabolism or Gcn2 deletion suppressed excessive bone formation. Together, our data indicate that glutamine is both an energy source and a protein-translation rheostat that is responsive to WNT and suggest that manipulation of the glutamine/GCN2 signaling axis may provide a valuable approach for normalizing deranged protein anabolism associated with human diseases.


Assuntos
Ciclo do Ácido Cítrico , Glutamina/metabolismo , Osteoblastos/metabolismo , Osteogênese , Osteosclerose/metabolismo , Via de Sinalização Wnt , Animais , Linhagem Celular , Deleção de Genes , Glutamina/genética , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Osteoblastos/patologia , Osteosclerose/genética , Osteosclerose/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Aminoacil-RNA de Transferência/genética , Aminoacil-RNA de Transferência/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
10.
Cell Metab ; 17(5): 745-55, 2013 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-23623748

RESUMO

WNT signaling controls many biological processes including cell differentiation in metazoans. However, how WNT reprograms cell identity is not well understood. We have investigated the potential role of cellular metabolism in WNT-induced osteoblast differentiation. WNT3A induces aerobic glycolysis known as Warburg effect by increasing the level of key glycolytic enzymes. The metabolic regulation requires LRP5 but not ß-catenin and is mediated by mTORC2-AKT signaling downstream of RAC1. Suppressing WNT3A-induced metabolic enzymes impairs osteoblast differentiation in vitro. Deletion of Lrp5 in the mouse, which decreases postnatal bone mass, reduces mTORC2 activity and glycolytic enzymes in bone cells and lowers serum lactate levels. Conversely, mice expressing a mutant Lrp5 that causes high bone mass exhibit increased glycolysis in bone. Thus, WNT-LRP5 signaling promotes bone formation in part through direct reprogramming of glucose metabolism. Moreover, regulation of cellular metabolism may represent a general mechanism contributing to the wide-ranging functions of WNT proteins.


Assuntos
Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Complexos Multiproteicos/metabolismo , Osteoblastos/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Proteína Wnt3A/metabolismo , Células 3T3-L1 , Animais , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Glucose/metabolismo , Glicólise/fisiologia , Células HEK293 , Humanos , Ácido Láctico/sangue , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA