Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Immunol ; 207(2): 493-504, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34215653

RESUMEN

The availability of Ags on the surface of tumor cells is crucial for the efficacy of cancer immunotherapeutic approaches using large molecules, such as T cell bispecific Abs (TCBs). Tumor Ags are processed through intracellular proteasomal protein degradation and are displayed as peptides on MHC class I (MHC I). Ag recognition through TCRs on the surface of CD8+ T cells can elicit a tumor-selective immune response. In this article, we show that proteolysis-targeting chimeras (PROTACs) that target bromo- and extraterminal domain proteins increase the abundance of the corresponding target-derived peptide Ags on MHC I in both liquid and solid tumor-derived human cell lines. This increase depends on the engagement of the E3 ligase to bromo- and extraterminal domain protein. Similarly, targeting of a doxycycline-inducible Wilms tumor 1 (WT1)-FKBP12F36V fusion protein, by a mutant-selective FKBP12F36V degrader, increases the presentation of WT1 Ags in human breast cancer cells. T cell-mediated response directed against cancer cells was tested on treatment with a TCR-like TCB, which was able to bridge human T cells to a WT1 peptide displayed on MHC I. FKBP12F36V degrader treatment increased the expression of early and late activation markers (CD69, CD25) in T cells; the secretion of granzyme ß, IFN-γ, and TNF-α; and cancer cell killing in a tumor-T cell coculture model. This study supports harnessing targeted protein degradation in tumor cells, for modulation of T cell effector function, by investigating for the first time, to our knowledge, the potential of combining a degrader and a TCB in a cancer immunotherapy setting.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Quimera/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/inmunología , Línea Celular Tumoral , Epítopos de Linfocito T/inmunología , Humanos , Proteolisis , Receptores de Antígenos de Linfocitos T/inmunología
2.
Gastroenterology ; 152(6): 1462-1476.e10, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28130067

RESUMEN

BACKGROUND & AIMS: The nuclear receptor subfamily 1 group H member 4 (NR1H4 or farnesoid X receptor [FXR]) regulates bile acid synthesis, transport, and catabolism. FXR also regulates postprandial lipid and glucose metabolism. We performed quantitative proteomic analyses of liver tissues from mice to evaluate these functions and investigate whether FXR regulates amino acid metabolism. METHODS: To study the role of FXR in mouse liver, we used mice with a disruption of Nr1h4 (FXR-knockout mice) and compared them with floxed control mice. Mice were gavaged with the FXR agonist obeticholic acid or vehicle for 11 days. Proteome analyses, as well as targeted metabolomics and chromatin immunoprecipitation, were performed on the livers of these mice. Primary rat hepatocytes were used to validate the role of FXR in amino acid catabolism by gene expression and metabolomics studies. Finally, control mice and mice with liver-specific disruption of Nr1h4 (liver FXR-knockout mice) were re-fed with a high-protein diet after 6 hours fasting and gavaged a 15NH4Cl tracer. Gene expression and the metabolome were studied in the livers and plasma from these mice. RESULTS: In livers of control mice and primary rat hepatocytes, activation of FXR with obeticholic acid increased expression of proteins that regulate amino acid degradation, ureagenesis, and glutamine synthesis. We found FXR to bind to regulatory sites of genes encoding these proteins in control livers. Liver tissues from FXR-knockout mice had reduced expression of urea cycle proteins, and accumulated precursors of ureagenesis, compared with control mice. In liver FXR-knockout mice on a high-protein diet, the plasma concentration of newly formed urea was significantly decreased compared with controls. In addition, liver FXR-knockout mice had reduced hepatic expression of enzymes that regulate ammonium detoxification compared with controls. In contrast, obeticholic acid increased expression of genes encoding enzymes involved in ureagenesis compared with vehicle in C57Bl/6 mice. CONCLUSIONS: In livers of mice, FXR regulates amino acid catabolism and detoxification of ammonium via ureagenesis and glutamine synthesis. Failure of the urea cycle and hyperammonemia are common in patients with acute and chronic liver diseases; compounds that activate FXR might promote ammonium clearance in these patients.


Asunto(s)
Amoníaco/metabolismo , Glutamina/biosíntesis , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Urea/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacología , Proteínas en la Dieta/administración & dosificación , Expresión Génica , Hepatocitos , Hígado/enzimología , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoma , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores
3.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 45-59, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28986309

RESUMEN

The Farnesoid X receptor (FXR) is a nuclear receptor activated by bile acids (BAs). BAs are amphipathic molecules that serve as fat solubilizers in the intestine under postprandial conditions. In the post-absorptive state, BAs bind FXR in the hepatocytes, which in turn provides feedback signals on BA synthesis and transport and regulates lipid, glucose and amino acid metabolism. Therefore, FXR acts as a homeostat of all three classes of nutrients, fats, sugars and proteins. Here we re-analyze the function of FXR in the perspective of nutritional metabolism, and discuss the role of FXR in liver energy homeostasis in postprandial, post-absorptive and fasting/starvation states. FXR, by regulating nutritional metabolism, represses autophagy in conditions of nutrient abundance, and controls the metabolic needs of proliferative cells. In addition, FXR regulates inflammation via direct effects and via its impact on nutrient metabolism. These functions indicate that FXR is an attractive therapeutic target for liver diseases.


Asunto(s)
Metabolismo Energético/genética , Homeostasis/genética , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Ácidos y Sales Biliares/metabolismo , Alimentos , Glucosa/metabolismo , Hepatocitos/metabolismo , Humanos , Mucosa Intestinal/metabolismo
4.
Biochim Biophys Acta ; 1862(2): 166-73, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26554605

RESUMEN

Inflammatory Bowel Disease (IBD) is a multifactorial disorder involving dysregulation of the immune response and bacterial translocation through the intestinal mucosal barrier. Previously, we have shown that activation of the bile acid sensor Farnesoid X Receptor (FXR), which belongs to the family of nuclear receptors, improves experimental intestinal inflammation, decreasing expression of pro-inflammatory cytokines and protecting the intestinal barrier. Here, we aimed to investigate the immunological mechanisms that ameliorate colitis when FXR is activated. We analyzed by FACS immune cell populations in mesenteric lymph nodes (MLN) and in the spleen to understand whether FXR activation alters the systemic immune response. We show that FXR activation by obeticholic acid (OCA) has systemic anti-inflammatory effects that include increased levels of plasma IL-10, inhibition of both DSS-colitis associated decrease in splenic dendritic cells (DCs) and increase in Tregs. Impact of OCA on DC relative abundance was seen in spleen but not MLN, possibly related to the increased FXR expression in splenic DCs compared to MLN DCs. Moreover, FXR activation modulates the chemotactic environment in the colonic site of inflammation, as Madcam1 expression is decreased, while Ccl25 is upregulated. Together, our data suggest that OCA treatment elicits an anti-inflammatory immune status including retention of DCs in the spleen, which is associated with decreased colonic inflammation. Pharmacological FXR activation is therefore an attractive new drug target for treatment of IBD.


Asunto(s)
Colitis/inducido químicamente , Colitis/inmunología , Células Dendríticas/inmunología , Sulfato de Dextran , Receptores Citoplasmáticos y Nucleares/inmunología , Bazo/inmunología , Animales , Quimiotaxis , Colitis/patología , Colon/citología , Colon/inmunología , Colon/patología , Células Dendríticas/patología , Interleucina-10/inmunología , Masculino , Ratones Endogámicos C57BL , Bazo/citología , Bazo/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
5.
Mol Metab ; 30: 221-229, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31767173

RESUMEN

OBJECTIVE: Coupling metabolic and reproductive pathways is essential for the survival of species. However, the functions of steroidogenic enzymes expressed in metabolic tissues are largely unknown. METHODS AND RESULTS: Here, we show that in the liver, the classical steroidogenic enzyme Cyp17a1 forms an essential nexus for glucose and ketone metabolism during feed-fast cycles. Both gain- and loss-of-function approaches are used to show that hepatic Cyp17a1 is induced by fasting, catalyzes the production of at least one hormone-ligand (DHEA) for the nuclear receptor PPARα, and is ultimately required for maintaining euglycemia and ketogenesis during nutrient deprivation. The feedback-loop that terminates Cyp17a1-PPARα activity, and re-establishes anabolic liver metabolism during re-feeding is mapped to postprandial bile acid-signaling, involving the receptors FXR, SHP and LRH-1. CONCLUSIONS: Together, these findings represent a novel paradigm of homeostatic control in which nutritional cues feed-forward on to metabolic pathways by influencing extragonadal steroidogenesis.


Asunto(s)
Hígado/metabolismo , PPAR alfa/metabolismo , Esteroide 17-alfa-Hidroxilasa/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Glucosa/metabolismo , Células HEK293 , Hepatocitos/metabolismo , Homeostasis , Humanos , Cetonas/metabolismo , Lipogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxidación-Reducción , Receptores Citoplasmáticos y Nucleares , Transducción de Señal , Esteroide 17-alfa-Hidroxilasa/fisiología
6.
Pharmacol Ther ; 191: 162-177, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29933033

RESUMEN

Bile acids are amphipathic molecules that were previously known to serve as fat solubilizers in the intestine in postprandial conditions. In the last two decades, bile acids have been recognized as signaling molecules regulating energy metabolism pathways via, amongst others, the farnesoid X receptor (FXR). Upon bile acid activation, FXR controls expression of genes involved in bile acid, lipid, glucose and amino acid metabolism. In addition, FXR activation has been shown to limit the inflammatory response. The central role of FXR in various aspects of metabolism and inflammation makes FXR an attractive drug target for several diseases, such as obesity, metabolic syndrome, non-alcoholic steatohepatitis, cholestasis and chronic inflammatory diseases of the liver and intestine. However, most of the currently available compounds impact on all discovered FXR-mediated functions and may have, on top of beneficial effects, undesired biological actions depending on the disease. Therefore, research efforts are increasingly focused on the development of selective FXR modulators, i.e. selective bile acid receptor modulators (SBARMs), aimed at limiting the potential side-effects of conventional full FXR agonists upon chronic treatment. Here, we review the rationale for the design of SBARMs comprising dissociation between metabolic and inflammatory signaling, gene-selective and tissue-specific targeting. We discuss the potential structural mechanisms underlying the binding properties of dissociating ligands of FXR in light of ongoing efforts on the generation of dissociated ligands for otxher nuclear receptors, as well as their pharmacological and therapeutic potential.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Diseño de Fármacos , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Metabolismo Energético/fisiología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Ligandos , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/patología , Terapia Molecular Dirigida , Receptores Citoplasmáticos y Nucleares/metabolismo
7.
PLoS One ; 12(2): e0171185, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28178326

RESUMEN

Fibroblast growth factor 19 (FGF19) is a gut-derived peptide hormone that is produced following activation of Farnesoid X Receptor (FXR). FGF19 is secreted and signals to the liver, where it contributes to the homeostasis of bile acid (BA), lipid and carbohydrate metabolism. FGF19 is a promising therapeutic target for the metabolic syndrome and cholestatic diseases, but enthusiasm for its use has been tempered by FGF19-mediated induction of proliferation and hepatocellular carcinoma. To inform future rational design of FGF19-variants, we have conducted temporal quantitative proteomic and gene expression analyses to identify FGF19-targets related to metabolism and proliferation. Mice were fasted for 16 hours, and injected with human FGF19 (1 mg/kg body weight) or vehicle. Liver protein extracts (containing "light" lysine) were mixed 1:1 with a spike-in protein extract from 13C6-lysine metabolically labelled mouse liver (containing "heavy" lysine) and analysed by LC-MS/MS. Our analyses provide a resource of FGF19 target proteins in the liver. 189 proteins were upregulated (≥ 1.5 folds) and 73 proteins were downregulated (≤ -1.5 folds) by FGF19. FGF19 treatment decreased the expression of proteins involved in fatty acid (FA) synthesis, i.e., Fabp5, Scd1, and Acsl3 and increased the expression of Acox1, involved in FA oxidation. As expected, FGF19 increased the expression of proteins known to drive proliferation (i.e., Tgfbi, Vcam1, Anxa2 and Hdlbp). Importantly, many of the FGF19 targets (i.e., Pdk4, Apoa4, Fas and Stat3) have a dual function in both metabolism and cell proliferation. Therefore, our findings challenge the development of FGF19-variants that fully uncouple metabolic benefit from mitogenic potential.


Asunto(s)
Metabolismo Energético , Factores de Crecimiento de Fibroblastos/farmacología , Hígado/metabolismo , Proteoma , Proteómica , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Expresión Génica , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones , Proteómica/métodos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes
8.
PLoS One ; 9(8): e104449, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25116592

RESUMEN

BACKGROUND & AIMS: Liver regeneration (LR) is a valuable model for studying mechanisms modulating hepatocyte proliferation. Nuclear receptors (NRs) are key players in the control of cellular functions, being ideal modulators of hepatic proliferation and carcinogenesis. METHODS & RESULTS: We used a previously validated RT-qPCR platform to profile modifications in the expression of all 49 members of the NR superfamily in mouse liver during LR. Twenty-nine NR transcripts were significantly modified in their expression during LR, including fatty acid (peroxisome proliferator-activated receptors, PPARs) and oxysterol (liver X receptors, Lxrs) sensors, circadian masters RevErbα and RevErbß, glucocorticoid receptor (Gr) and constitutive androxane receptor (Car). In order to detect the NRs that better characterize proliferative status vs. proliferating liver, we used the novel Random Forest (RF) analysis to selected a trio of down-regulated NRs (thyroid receptor alpha, Trα; farsenoid X receptor beta, Fxrß; Pparδ) as best discriminators of the proliferating status. To validate our approach, we further studied PPARδ role in modulating hepatic proliferation. We first confirmed the suppression of PPARδ both in LR and human hepatocellular carcinoma at protein level, and then demonstrated that PPARδ agonist GW501516 reduces the proliferative potential of hepatoma cells. CONCLUSIONS: Our data suggest that NR transcriptome is modulated in proliferating liver and is a source of biomarkers and bona fide pharmacological targets for the management of liver disease affecting hepatocyte proliferation.


Asunto(s)
Carcinoma Hepatocelular/genética , Hepatocitos/metabolismo , Neoplasias Hepáticas/genética , Regeneración Hepática/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular , Proliferación Celular , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Receptores Citoplasmáticos y Nucleares/metabolismo , Transcriptoma
9.
Mol Cell Endocrinol ; 368(1-2): 108-19, 2013 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22789748

RESUMEN

A comprehensive understanding of the pathways underlying hepatocyte turnover and liver regeneration is essential for the development of innovative and effective therapies in the management of chronic liver disease, and the prevention of hepatocellular carcinoma (HCC) in cirrhosis. Nuclear receptors (NRs) are master transcriptional regulators of liver development, differentiation and function. NRs have been implicated in the modulation of hepatocyte priming and proliferation in regenerating liver, chronic hepatitis and HCC development. In this review, we focus on NRs and their pathways regulating hepatocyte proliferation and liver regeneration, with a perspective view on NRs as candidate biomarkers and novel pharmacological targets in the management of liver disease and HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Regeneración Hepática , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Ácidos y Sales Biliares/fisiología , Carcinoma Hepatocelular/patología , Proliferación Celular , Regulación de la Expresión Génica , Hepatocitos/fisiología , Humanos , Lípidos/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA