Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(1): 113626, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38157298

RESUMEN

Exercise training can stimulate the formation of fatty-acid-oxidizing slow-twitch skeletal muscle fibers, which are inversely correlated with obesity, but the molecular mechanism underlying this transformation requires further elucidation. Here, we report that the downregulation of the mitochondrial disulfide relay carrier CHCHD4 by exercise training decreases the import of TP53-regulated inhibitor of apoptosis 1 (TRIAP1) into mitochondria, which can reduce cardiolipin levels and promote VDAC oligomerization in skeletal muscle. VDAC oligomerization, known to facilitate mtDNA release, can activate cGAS-STING/NFKB innate immune signaling and downregulate MyoD in skeletal muscle, thereby promoting the formation of oxidative slow-twitch fibers. In mice, CHCHD4 haploinsufficiency is sufficient to activate this pathway, leading to increased oxidative muscle fibers and decreased fat accumulation with aging. The identification of a specific mediator regulating muscle fiber transformation provides an opportunity to understand further the molecular underpinnings of complex metabolic conditions such as obesity and could have therapeutic implications.


Asunto(s)
Fibras Musculares Esqueléticas , Músculo Esquelético , Animales , Ratones , Apoptosis , Inmunidad Innata , Músculo Esquelético/metabolismo , Obesidad/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(34): e2302738120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37579159

RESUMEN

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is characterized by various disabling symptoms including exercise intolerance and is diagnosed in the absence of a specific cause, making its clinical management challenging. A better understanding of the molecular mechanism underlying this apparent bioenergetic deficiency state may reveal insights for developing targeted treatment strategies. We report that overexpression of Wiskott-Aldrich Syndrome Protein Family Member 3 (WASF3), here identified in a 38-y-old woman suffering from long-standing fatigue and exercise intolerance, can disrupt mitochondrial respiratory supercomplex formation and is associated with endoplasmic reticulum (ER) stress. Increased expression of WASF3 in transgenic mice markedly decreased their treadmill running capacity with concomitantly impaired respiratory supercomplex assembly and reduced complex IV levels in skeletal muscle mitochondria. WASF3 induction by ER stress using endotoxin, well known to be associated with fatigue in humans, also decreased skeletal muscle complex IV levels in mice, while decreasing WASF3 levels by pharmacologic inhibition of ER stress improved mitochondrial function in the cells of the patient with chronic fatigue. Expanding on our findings, skeletal muscle biopsy samples obtained from a cohort of patients with ME/CFS showed increased WASF3 protein levels and aberrant ER stress activation. In addition to revealing a potential mechanism for the bioenergetic deficiency in ME/CFS, our study may also provide insights into other disorders associated with fatigue such as rheumatic diseases and long COVID.


Asunto(s)
COVID-19 , Síndrome de Fatiga Crónica , Animales , Femenino , Humanos , Ratones , COVID-19/metabolismo , Síndrome de Fatiga Crónica/diagnóstico , Mitocondrias/metabolismo , Síndrome Post Agudo de COVID-19 , Respiración , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Ratones Transgénicos
3.
J Biol Chem ; 299(8): 104946, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37348559

RESUMEN

Dysregulated bile acid (BA)/lipid metabolism and gut bacteria dysbiosis are tightly associated with the development of obesity and non-alcoholic fatty liver disease (NAFLD). The orphan nuclear receptor, Small Heterodimer Partner (SHP/NR0B2), is a key regulator of BA/lipid metabolism, and its gene-regulating function is markedly enhanced by phosphorylation at Thr-58 mediated by a gut hormone, fibroblast growth factor-15/19 (FGF15/19). To investigate the role of this phosphorylation in whole-body energy metabolism, we generated transgenic SHP-T58A knock-in mice. Compared with wild-type (WT) mice, the phosphorylation-defective SHP-T58A mice gained weight more rapidly with decreased energy expenditure and increased lipid/BA levels. This obesity-prone phenotype was associated with the upregulation of lipid/BA synthesis genes and downregulation of lipophagy/ß-oxidation genes. Mechanistically, defective SHP phosphorylation selectively impaired its interaction with LRH-1, resulting in de-repression of SHP/LRH-1 target BA/lipid synthesis genes. Remarkably, BA composition and selective gut bacteria which are known to impact obesity, were also altered in these mice. Upon feeding a high-fat diet, fatty liver developed more severely in SHP-T58A mice compared to WT mice. Treatment with antibiotics substantially improved the fatty liver phenotypes in both groups but had greater effects in the T58A mice so that the difference between the groups was largely eliminated. These results demonstrate that defective phosphorylation at a single nuclear receptor residue can impact whole-body energy metabolism by altering BA/lipid metabolism and gut bacteria, promoting complex metabolic disorders like NAFLD. Since posttranslational modifications generally act in gene- and context-specific manners, the FGF15/19-SHP phosphorylation axis may allow more targeted therapy for NAFLD.


Asunto(s)
Ácidos y Sales Biliares , Microbioma Gastrointestinal , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Ácidos y Sales Biliares/análisis , Ácidos y Sales Biliares/genética , Lípidos/sangre , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/microbiología , Obesidad/microbiología , Fosforilación , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Masculino , Antibacterianos/farmacología
4.
J Biol Chem ; 299(3): 103018, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36796514

RESUMEN

The endosymbiotic theory posits that ancient eukaryotic cells engulfed O2-consuming prokaryotes, which protected them against O2 toxicity. Previous studies have shown that cells lacking cytochrome c oxidase (COX), required for respiration, have increased DNA damage and reduced proliferation, which could be improved by reducing O2 exposure. With recently developed fluorescence lifetime microscopy-based probes demonstrating that the mitochondrion has lower [O2] than the cytosol, we hypothesized that the perinuclear distribution of mitochondria in cells may create a barrier for O2 to access the nuclear core, potentially affecting cellular physiology and maintaining genomic integrity. To test this hypothesis, we utilized myoglobin-mCherry fluorescence lifetime microscopy O2 sensors without subcellular targeting ("cytosol") or with targeting to the mitochondrion or nucleus for measuring their localized O2 homeostasis. Our results showed that, similar to the mitochondria, the nuclear [O2] was reduced by ∼20 to 40% compared with the cytosol under imposed O2 levels of ∼0.5 to 18.6%. Pharmacologically inhibiting respiration increased nuclear O2 levels, and reconstituting O2 consumption by COX reversed this increase. Similarly, genetic disruption of respiration by deleting SCO2, a gene essential for COX assembly, or restoring COX activity in SCO2-/- cells by transducing with SCO2 cDNA replicated these changes in nuclear O2 levels. The results were further supported by the expression of genes known to be affected by cellular O2 availability. Our study reveals the potential for dynamic regulation of nuclear O2 levels by mitochondrial respiratory activity, which in turn could affect oxidative stress and cellular processes such as neurodegeneration and aging.


Asunto(s)
Mitocondrias , Oxígeno , Oxígeno/metabolismo , Mitocondrias/metabolismo , Respiración , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Núcleo Celular/metabolismo , Consumo de Oxígeno , Respiración de la Célula
5.
EMBO J ; 41(17): e109997, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35686465

RESUMEN

Lysosome-mediated macroautophagy, including lipophagy, is activated under nutrient deprivation but is repressed after feeding. We show that, unexpectedly, feeding activates intestinal autophagy/lipophagy in a manner dependent on both the orphan nuclear receptor, small heterodimer partner (SHP/NR0B2), and the gut hormone, fibroblast growth factor-15/19 (FGF15/19). Furthermore, postprandial intestinal triglycerides (TGs) and apolipoprotein-B48 (ApoB48), the TG-rich chylomicron marker, were elevated in SHP-knockout and FGF15-knockout mice. Genomic analyses of the mouse intestine indicated that SHP partners with the key lysosomal activator, transcription factor-EB (TFEB) to upregulate the transcription of autophagy/lipolysis network genes after feeding. FGF19 treatment activated lipophagy, reducing TG and ApoB48 levels in HT29 intestinal cells, which was dependent on TFEB. Mechanistically, feeding-induced FGF15/19 signaling increased the nuclear localization of TFEB and SHP via PKC beta/zeta-mediated phosphorylation, leading to increased transcription of the TFEB/SHP target lipophagy genes, Ulk1 and Atgl. Collectively, these results demonstrate that paradoxically after feeding, FGF15/19-activated SHP and TFEB activate gut lipophagy, limiting postprandial TGs. As excess postprandial lipids cause dyslipidemia and obesity, the FGF15/19-SHP-TFEB axis that reduces intestinal TGs via lipophagic activation provides promising therapeutic targets for obesity-associated metabolic disease.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Ingestión de Alimentos , Factores de Crecimiento de Fibroblastos , Tracto Gastrointestinal , Receptores Citoplasmáticos y Nucleares , Animales , Apolipoproteína B-48/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Tracto Gastrointestinal/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Obesidad/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
6.
Mitochondrial DNA B Resour ; 7(3): 498-500, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35311211

RESUMEN

In this study, we use a specimen from wild-caught individual to determine the complete mitochondrial genome of the Amur soft-shelled turtle (Pelodiscus maackii). The complete mitogenome of P. maackii has 16,258 bp in length and consists of 13 protein-coding genes (PCGs), 22 tRNAs, two rRNAs, and one control region. The arrangement of genes of P. maackii is identical with previously reported mitogenomes in the family Trionychoidea. According to our result, the ML tree for the phylogenetic reconstruction revealed that the individuals used in present study is closely related with the previously reported sequences of P. sinensis (AY962573 and MG431983) in p-distance 0.7% and 2.5%.

7.
Diabetes ; 70(3): 733-744, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33328206

RESUMEN

Aberrantly elevated expression in obesity of microRNAs (miRNAs), including the miRNA miR-802, contributes to obesity-associated metabolic complications, but the mechanisms underlying the elevated expression are unclear. Farnesoid X receptor (FXR), a key regulator of hepatic energy metabolism, has potential for treatment of obesity-related diseases. We examined whether a nuclear receptor cascade involving FXR and FXR-induced small heterodimer partner (SHP) regulates expression of miR-802 to maintain glucose and lipid homeostasis. Hepatic miR-802 levels are increased in FXR-knockout (KO) or SHP-KO mice and are decreased by activation of FXR in a SHP-dependent manner. Mechanistically, transactivation of miR-802 by aromatic hydrocarbon receptor (AHR) is inhibited by SHP. In obese mice, activation of FXR by obeticholic acid treatment reduced miR-802 levels and improved insulin resistance and hepatosteatosis, but these beneficial effects were largely abolished by overexpression of miR-802. In patients with nonalcoholic fatty liver disease (NAFLD) and in obese mice, occupancy of SHP is reduced and that of AHR is modestly increased at the miR-802 promoter, consistent with elevated hepatic miR-802 expression. These results demonstrate that normal inhibition of miR-802 by FXR-SHP is defective in obesity, resulting in increased miR-802 levels, insulin resistance, and fatty liver. This FXR-SHP-miR-802 pathway may present novel targets for treating type 2 diabetes and NAFLD.


Asunto(s)
Hígado Graso/metabolismo , Glucosa/metabolismo , Hígado/metabolismo , MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Animales , Células Cultivadas , Hígado Graso/genética , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/genética , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Activación Transcripcional/genética , Activación Transcripcional/fisiología
8.
Nat Commun ; 11(1): 5969, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33235221

RESUMEN

Hepatic lipogenesis is normally tightly regulated but is aberrantly elevated in obesity. Fibroblast Growth Factor-15/19 (mouse FGF15, human FGF19) are bile acid-induced late fed-state gut hormones that decrease hepatic lipid levels by unclear mechanisms. We show that FGF15/19 and FGF15/19-activated Small Heterodimer Partner (SHP/NR0B2) have a role in transcriptional repression of lipogenesis. Comparative genomic analyses reveal that most of the SHP cistrome, including lipogenic genes repressed by FGF19, have overlapping CpG islands. FGF19 treatment or SHP overexpression in mice inhibits lipogenesis in a DNA methyltransferase-3a (DNMT3A)-dependent manner. FGF19-mediated activation of SHP via phosphorylation recruits DNMT3A to lipogenic genes, leading to epigenetic repression via DNA methylation. In non-alcoholic fatty liver disease (NAFLD) patients and obese mice, occupancy of SHP and DNMT3A and DNA methylation at lipogenic genes are low, with elevated gene expression. In conclusion, FGF15/19 represses hepatic lipogenesis by activating SHP and DNMT3A physiologically, which is likely dysregulated in NAFLD.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Lipogénesis , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Receptores Citoplasmáticos y Nucleares/genética
9.
Nat Commun ; 11(1): 807, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-32042044

RESUMEN

Autophagy is essential for cellular survival and energy homeostasis under nutrient deprivation. Despite the emerging importance of nuclear events in autophagy regulation, epigenetic control of autophagy gene transcription remains unclear. Here, we report fasting-induced Fibroblast Growth Factor-21 (FGF21) signaling activates hepatic autophagy and lipid degradation via Jumonji-D3 (JMJD3/KDM6B) histone demethylase. Upon FGF21 signaling, JMJD3 epigenetically upregulates global autophagy-network genes, including Tfeb, Atg7, Atgl, and Fgf21, through demethylation of histone H3K27-me3, resulting in autophagy-mediated lipid degradation. Mechanistically, phosphorylation of JMJD3 at Thr-1044 by FGF21 signal-activated PKA increases its nuclear localization and interaction with the nuclear receptor PPARα to transcriptionally activate autophagy. Administration of FGF21 in obese mice improves defective autophagy and hepatosteatosis in a JMJD3-dependent manner. Remarkably, in non-alcoholic fatty liver disease patients, hepatic expression of JMJD3, ATG7, LC3, and ULK1 is substantially decreased. These findings demonstrate that FGF21-JMJD3 signaling epigenetically links nutrient deprivation with hepatic autophagy and lipid degradation in mammals.


Asunto(s)
Autofagia/genética , Ayuno/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Hígado/metabolismo , Animales , Autofagia/efectos de los fármacos , Epigénesis Genética , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/deficiencia , Hepatocitos/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Klotho , Lipólisis , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Obesos , PPAR alfa/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal , Regulación hacia Arriba
10.
Mitochondrial DNA B Resour ; 5(3): 2145-2146, 2020 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-33457760

RESUMEN

The complete mitochondrial genome of Muskrat, Ondatra zibethicus (Rodentia: Cricetidae) in Korea was sequenced for the first time using the next-generation sequencing method to understand its evolutionary relationship and to be helpful to establish a management plan. This mitogenome was 16,350 base pairs in length, containing 13 protein-coding genes, 22 transfer RNA genes, two ribosomal RNA genes, and one control region. Its overall A, C, G, and T contents were 32.0%, 26.9%, 12.6%, and 28.5%, respectively. A + T content (63.7%) was higher than G + C content (36.3%). We made the phylogenetic tree of muskrat and other 12 species of order Rodentia distributed in Korea.

11.
Hepatology ; 71(6): 2118-2134, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31549733

RESUMEN

BACKGROUND AND AIMS: Bile acids (BAs) are important regulators of metabolism and energy balance, but excess BAs cause cholestatic liver injury. The histone methyltransferase mixed-lineage leukemia-4 (MLL4) is a transcriptional coactivator of the BA-sensing nuclear receptor farnesoid X receptor (FXR) and epigenetically up-regulates FXR targets important for the regulation of BA levels, small heterodimer partner (SHP), and bile salt export pump (BSEP). MLL4 expression is aberrantly down-regulated and BA homeostasis is disrupted in cholestatic mice, but the underlying mechanisms are unclear. APPROACH AND RESULTS: We examined whether elevated microRNA-210 (miR-210) in cholestatic liver promotes BA-induced pathology by inhibiting MLL4 expression. miR-210 was the most highly elevated miR in hepatic SHP-down-regulated mice with elevated hepatic BA levels. MLL4 was identified as a direct target of miR-210, and overexpression of miR-210 inhibited MLL4 and, subsequently, BSEP and SHP expression, resulting in defective BA metabolism and hepatotoxicity with inflammation. miR-210 levels were elevated in cholestatic mouse models, and in vivo silencing of miR-210 ameliorated BA-induced liver pathology and decreased hydrophobic BA levels in an MLL4-dependent manner. In gene expression studies, SHP inhibited miR-210 expression by repressing a transcriptional activator, Kruppel-like factor-4 (KLF4). In patients with primary biliary cholangitis/cirrhosis (PBC), hepatic levels of miR-210 and KLF4 were highly elevated, whereas nuclear levels of SHP and MLL4 were reduced. CONCLUSIONS: Hepatic miR-210 is physiologically regulated by SHP but elevated in cholestatic mice and patients with PBC, promoting BA-induced liver injury in part by targeting MLL4. The miR-210-MLL4 axis is a potential target for the treatment of BA-associated hepatobiliary disease.


Asunto(s)
Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/metabolismo , Ácidos y Sales Biliares , Colestasis/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Cirrosis Hepática Biliar/metabolismo , MicroARNs/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/metabolismo , Colestasis/complicaciones , Perfilación de la Expresión Génica , Humanos , Factor 4 Similar a Kruppel , Cirrosis Hepática Biliar/etiología , Ratones , Activación Transcripcional
12.
Endocrinology ; 160(7): 1573-1589, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31050726

RESUMEN

The involvement of small heterodimer partner (SHP) in the inhibition of hepatic bile acid synthesis from cholesterol has been established. However, extrahepatic expression of SHP implies that SHP may have regulatory functions other than those in the liver. Here, we find that SHP mRNA expression is high in murine bone marrow cells, suggesting a physiological role within macrophages. Indeed, expression of SHP in macrophages decreases the transcriptional activity and nuclear localization of nuclear factor κB, whereas downregulation of SHP has the opposite effects. Expression of genes associated with macrophage-T cell crosstalk were altered by overexpression or downregulation of SHP. Intriguingly, increasing SHP expression in macrophages resulted in decreased T cell expansion, a hallmark of T cell activation, whereas knockdown of SHP resulted in increased expansion. Analyses of the expanded T cells revealed a dichotomous skewing between effector T cells and regulatory T cells (Tregs), with SHP overexpression reducing Tregs and downregulation of SHP increasing their expansion. The expanded Tregs were confirmed to be suppressive via adoptive transfers. IL-2 and TGF-ß, known inducers of Treg differentiation, were found to be regulated by SHP. Furthermore, SHP occupancy at the promoter region of IL-2 was increased after macrophages were challenged with lipopolysaccharide. Neutralizing antibodies to IL-2 and TGF-ß inhibited the expansion of Tregs mediated by downregulation of SHP. This study demonstrates that expression and activity of SHP within macrophages can alter T cell fate and identifies SHP as a potential therapeutic target for autoimmune diseases or solid cancers.


Asunto(s)
Células de la Médula Ósea/metabolismo , Macrófagos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Linfocitos T/metabolismo , Animales , Línea Celular , Proliferación Celular , Regulación hacia Abajo , Regulación de la Expresión Génica , Ratones , FN-kappa B/metabolismo
13.
J Biol Chem ; 294(22): 8732-8744, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-30996006

RESUMEN

The bile acid (BA) nuclear receptor, farnesoid X receptor (FXR/NR1H4), maintains metabolic homeostasis by transcriptional control of numerous genes, including an intestinal hormone, fibroblast growth factor-19 (FGF19; FGF15 in mice). Besides activation by BAs, the gene-regulatory function of FXR is also modulated by hormone or nutrient signaling-induced post-translational modifications. Recently, phosphorylation at Tyr-67 by the FGF15/19 signaling-activated nonreceptor tyrosine kinase Src was shown to be important for FXR function in BA homeostasis. Here, we examined the role of this FXR phosphorylation in cholesterol regulation. In both hepatic FXR-knockout and FXR-knockdown mice, reconstitution of FXR expression up-regulated cholesterol transport genes for its biliary excretion, including scavenger receptor class B member 1 (Scarb1) and ABC subfamily G member 8 (Abcg5/8), decreased hepatic and plasma cholesterol levels, and increased biliary and fecal cholesterol levels. Of note, these sterol-lowering effects were blunted by substitution of Phe for Tyr-67 in FXR. Moreover, consistent with Src's role in phosphorylating FXR, Src knockdown impaired cholesterol regulation in mice. In hypercholesterolemic apolipoprotein E-deficient mice, expression of FXR, but not Y67F-FXR, ameliorated atherosclerosis, whereas Src down-regulation exacerbated it. Feeding or treatment with an FXR agonist induced Abcg5/8 and Scarb1 expression in WT, but not FGF15-knockout, mice. Furthermore, FGF19 treatment increased occupancy of FXR at Abcg5/8 and Scarb1, expression of these genes, and cholesterol efflux from hepatocytes. These FGF19-mediated effects were blunted by the Y67F-FXR substitution or Src down-regulation or inhibition. We conclude that phosphorylation of hepatic FXR by FGF15/19-induced Src maintains cholesterol homeostasis and protects against atherosclerosis.


Asunto(s)
Colesterol/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hepatocitos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Familia-src Quinasas/metabolismo , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8/genética , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8/metabolismo , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Ácidos y Sales Biliares/metabolismo , Colesterol/sangre , Regulación hacia Abajo , Factores de Crecimiento de Fibroblastos/deficiencia , Factores de Crecimiento de Fibroblastos/genética , Lipoproteínas/genética , Lipoproteínas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo , Transducción de Señal , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
14.
Gastroenterology ; 156(4): 1052-1065, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30521806

RESUMEN

BACKGROUND & AIMS: The nuclear receptor subfamily 0 group B member 2 (NR0B2, also called SHP) is expressed at high levels in the liver and intestine. Postprandial fibroblast growth factor 19 (human FGF19, mouse FGF15) signaling increases the transcriptional activity of SHP. We studied the functions of SHP and FGF19 in the intestines of mice, including their regulation of expression of the cholesterol transporter NPC1L1 )NPC1-like intracellular cholesterol transporter 1) and cholesterol absorption. METHODS: We performed histologic and biochemical analyses of intestinal tissues from C57BL/6 and SHP-knockout mice and performed RNA-sequencing analyses to identify genes regulated by SHP. The effects of fasting and refeeding on intestinal expression of NPC1L1 were examined in C57BL/6, SHP-knockout, and FGF15-knockout mice. Mice were given FGF19 daily for 1 week; fractional cholesterol absorption, cholesterol and bile acid (BA) levels, and composition of BAs were measured. Intestinal organoids were generated from C57BL/6 and SHP-knockout mice, and cholesterol uptake was measured. Luciferase reporter assays were performed with HT29 cells. RESULTS: We found that the genes that regulate lipid and ion transport in intestine, including NPC1L1, were up-regulated and that cholesterol absorption was increased in SHP-knockout mice compared with C57BL/6 mice. Expression of NPC1L1 was reduced in C57BL/6 mice after refeeding after fasting but not in SHP-knockout or FGF15-knockout mice. SHP-knockout mice had altered BA composition compared with C57BL/6 mice. FGF19 injection reduced expression of NPC1L1, decreased cholesterol absorption, and increased levels of hydrophilic BAs, including tauro-α- and -ß-muricholic acids; these changes were not observed in SHP-knockout mice. SREBF2 (sterol regulatory element binding transcription factor 2), which regulates cholesterol, activated transcription of NPC1L1. FGF19 signaling led to phosphorylation of SHP, which inhibited SREBF2 activity. CONCLUSIONS: Postprandial FGF19 and SHP inhibit SREBF2, which leads to repression of intestinal NPC1L1 expression and cholesterol absorption. Strategies to increase FGF19 signaling to activate SHP might be developed for treatment of hypercholesterolemia.


Asunto(s)
Colesterol/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Proteínas de Transporte de Membrana/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Colesterol/análisis , Colesterol/sangre , HDL-Colesterol/análisis , HDL-Colesterol/sangre , LDL-Colesterol/análisis , LDL-Colesterol/sangre , Ingestión de Alimentos , Ayuno , Heces/química , Factores de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/genética , Células HT29 , Humanos , Íleon/metabolismo , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/genética , Yeyuno/metabolismo , Yeyuno/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides/metabolismo , Fosforilación , Periodo Posprandial , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/genética , Regulación hacia Arriba
15.
Nat Commun ; 9(1): 2590, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29968724

RESUMEN

Farnesoid-X-Receptor (FXR) plays a central role in maintaining bile acid (BA) homeostasis by transcriptional control of numerous enterohepatic genes, including intestinal FGF19, a hormone that strongly represses hepatic BA synthesis. How activation of the FGF19 receptor at the membrane is transmitted to the nucleus for transcriptional regulation of BA levels and whether FGF19 signaling posttranslationally modulates FXR function remain largely unknown. Here we show that FXR is phosphorylated at Y67 by non-receptor tyrosine kinase, Src, in response to postprandial FGF19, which is critical for its nuclear localization and transcriptional regulation of BA levels. Liver-specific expression of phospho-defective Y67F-FXR or Src downregulation in mice results in impaired homeostatic responses to acute BA feeding, and exacerbates cholestatic pathologies upon drug-induced hepatobiliary insults. Also, the hepatic FGF19-Src-FXR pathway is defective in primary biliary cirrhosis (PBC) patients. This study identifies Src-mediated FXR phosphorylation as a potential therapeutic target and biomarker for BA-related enterohepatic diseases.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Cirrosis Hepática Biliar/patología , Receptores Citoplasmáticos y Nucleares/metabolismo , Familia-src Quinasas/metabolismo , 1-Naftilisotiocianato/toxicidad , Animales , Biomarcadores/metabolismo , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/genética , Voluntarios Sanos , Hepatocitos , Humanos , Mucosa Intestinal/metabolismo , Isoxazoles/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática Biliar/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fosforilación/fisiología , Periodo Posprandial/fisiología , Cultivo Primario de Células , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/fisiología , Tirosina/metabolismo
16.
J Clin Invest ; 128(7): 3144-3159, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29911994

RESUMEN

Jumonji D3 (JMJD3) histone demethylase epigenetically regulates development and differentiation, immunity, and tumorigenesis by demethylating a gene repression histone mark, H3K27-me3, but a role for JMJD3 in metabolic regulation has not been described. SIRT1 deacetylase maintains energy balance during fasting by directly activating both hepatic gluconeogenic and mitochondrial fatty acid ß-oxidation genes, but the underlying epigenetic and gene-specific mechanisms remain unclear. In this study, JMJD3 was identified unexpectedly as a gene-specific transcriptional partner of SIRT1 and epigenetically activated mitochondrial ß-oxidation, but not gluconeogenic, genes during fasting. Mechanistically, JMJD3, together with SIRT1 and the nuclear receptor PPARα, formed a positive autoregulatory loop upon fasting-activated PKA signaling and epigenetically activated ß-oxidation-promoting genes, including Fgf21, Cpt1a, and Mcad. Liver-specific downregulation of JMJD3 resulted in intrinsic defects in ß-oxidation, which contributed to hepatosteatosis as well as glucose and insulin intolerance. Remarkably, the lipid-lowering effects by JMJD3 or SIRT1 in diet-induced obese mice were mutually interdependent. JMJD3 histone demethylase may serve as an epigenetic drug target for obesity, hepatosteatosis, and type 2 diabetes that allows selective lowering of lipid levels without increasing glucose levels.


Asunto(s)
Ayuno/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Mitocondrias Hepáticas/metabolismo , Animales , Regulación hacia Abajo , Epigénesis Genética , Ácidos Grasos/metabolismo , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Código de Histonas , Humanos , Histona Demetilasas con Dominio de Jumonji/deficiencia , Histona Demetilasas con Dominio de Jumonji/genética , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Obesidad/genética , Obesidad/metabolismo , Oxidación-Reducción , PPAR alfa/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo
17.
Nat Commun ; 9(1): 540, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416063

RESUMEN

Phosphatidylcholines (PC) and S-adenosylmethionine (SAM) are critical determinants of hepatic lipid levels, but how their levels are regulated is unclear. Here, we show that Pemt and Gnmt, key one-carbon cycle genes regulating PC/SAM levels, are downregulated after feeding, leading to decreased PC and increased SAM levels, but these effects are blunted in small heterodimer partner (SHP)-null or FGF15-null mice. Further, aryl hydrocarbon receptor (AhR) is translocated into the nucleus by insulin/PKB signaling in the early fed state and induces Pemt and Gnmt expression. This induction is blocked by FGF15 signaling-activated SHP in the late fed state. Adenoviral-mediated expression of AhR in obese mice increases PC levels and exacerbates steatosis, effects that are blunted by SHP co-expression or Pemt downregulation. PEMT, AHR, and PC levels are elevated in simple steatosis patients, but PC levels are robustly reduced in steatohepatitis-fibrosis patients. This study identifies AhR and SHP as new physiological regulators of PC/SAM levels.


Asunto(s)
Ciclo del Carbono , Fosfatidilcolinas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , S-Adenosilmetionina/metabolismo , Animales , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Glicina N-Metiltransferasa/genética , Glicina N-Metiltransferasa/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Fosfatidiletanolamina N-Metiltransferasa/genética , Fosfatidiletanolamina N-Metiltransferasa/metabolismo , Unión Proteica/efectos de los fármacos , Receptores de Hidrocarburo de Aril/genética , Receptores Citoplasmáticos y Nucleares/genética
18.
EMBO J ; 36(12): 1755-1769, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28446510

RESUMEN

Lysosome-mediated autophagy is essential for cellular survival and homeostasis upon nutrient deprivation, but is repressed after feeding. Despite the emerging importance of transcriptional regulation of autophagy by nutrient-sensing factors, the role for epigenetic control is largely unexplored. Here, we show that Small Heterodimer Partner (SHP) mediates postprandial epigenetic repression of hepatic autophagy by recruiting histone demethylase LSD1 in response to a late fed-state hormone, FGF19 (hFGF19, mFGF15). FGF19 treatment or feeding inhibits macroautophagy, including lipophagy, but these effects are blunted in SHP-null mice or LSD1-depleted mice. In addition, feeding-mediated autophagy inhibition is attenuated in FGF15-null mice. Upon FGF19 treatment or feeding, SHP recruits LSD1 to CREB-bound autophagy genes, including Tfeb, resulting in dissociation of CRTC2, LSD1-mediated demethylation of gene-activation histone marks H3K4-me2/3, and subsequent accumulation of repressive histone modifications. Both FXR and SHP inhibit hepatic autophagy interdependently, but while FXR acts early, SHP acts relatively late after feeding, which effectively sustains postprandial inhibition of autophagy. This study demonstrates that the FGF19-SHP-LSD1 axis maintains homeostasis by suppressing unnecessary autophagic breakdown of cellular components, including lipids, under nutrient-rich postprandial conditions.


Asunto(s)
Autofagia , Represión Epigenética , Factores de Crecimiento de Fibroblastos/metabolismo , Histona Demetilasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Hepatocitos/ultraestructura , Histonas/metabolismo , Hígado/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Proteínas del Tejido Nervioso/deficiencia , Procesamiento Proteico-Postraduccional , Factores de Transcripción/metabolismo
19.
Mol Endocrinol ; 30(1): 92-103, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26505219

RESUMEN

The bile acid (BA)-sensing nuclear receptor, farnesoid X receptor (FXR), regulates postprandial metabolic responses, including inhibition of BA synthesis, by inducing the intestinal hormone, fibroblast growth factor (FGF)15 (FGF19 in human). In this study, we tested a novel hypothesis that FXR not only induces intestinal FGF15 but also primes the liver for effectively responding to the signal by transcriptional induction of the obligate coreceptor for FGF15, ß-Klotho (ßKL). Activation of FXR by a synthetic agonist, GW4064, in mice increased occupancy of FXR and its DNA-binding partner, retinoid X receptor-α, at FGF15-signaling component genes, particularly ßKL, and induced expression of these genes. Interestingly, mRNA levels of Fgfr4, the FGF15 receptor, were not increased by GW4064, but protein levels increased as a result of ßKL-dependent increased protein stability. Both FGF receptor 4 and ßKL protein levels were substantially decreased in FXR-knockout (KO) mice, and FGF19 signaling, monitored by phosphorylated ERK, was blunted in FXR-KO mice, FXR-KO mouse hepatocytes, and FXR-down-regulated human hepatocytes. Overexpression of ßKL in FXR-lacking hepatocytes partially restored FGF19 signaling and inhibition by FGF19 of Cyp7a1, which encodes the rate-limiting BA biosynthetic enzyme. In mice, transient inductions of intestinal Fgf15 and hepatic ßKL were temporally correlated after GW4064 treatment, and pretreatment of hepatocytes with GW4064 before FGF19 treatment enhanced FGF19 signaling, which was abolished by transcriptional inhibition or ßKL down-regulation. This study identifies FXR as a gut-liver metabolic coordinator for FGF15/19 action that orchestrates transient induction of hepatic ßKL and intestinal Fgf15/19 in a temporally correlated manner.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología , Animales , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Intestinos/efectos de los fármacos , Isoxazoles/farmacología , Proteínas Klotho , Hígado/efectos de los fármacos , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/efectos de los fármacos
20.
Genome Biol ; 16: 268, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26634251

RESUMEN

BACKGROUND: Fibroblast growth factor-19 (FGF19) is an intestinal hormone that mediates postprandial metabolic responses in the liver. The unusual orphan nuclear receptor, small heterodimer partner (SHP), acts as a co-repressor for many transcriptional factors and has been implicated in diverse biological pathways including FGF19-mediated repression of bile acid synthesis. To explore global functions of SHP in mediating FGF19 action, we identify genome-wide SHP binding sites in hepatic chromatin in mice treated with vehicle or FGF19 by ChIP-seq analysis. RESULTS: The overall pattern of SHP binding sites between these two groups is similar, but SHP binding is enhanced at the sites by addition of FGF19. SHP binding is detected preferentially in promoter regions that are enriched in motifs for unexpected non-nuclear receptors. We observe global co-localization of SHP sites with published sites for SREBP-2, a master transcriptional activator of cholesterol biosynthesis. FGF19 increases functional interaction between endogenous SHP and SREBP-2 and inhibits SREBP-2 target genes, and these effects were blunted in SHP-knockout mice. Furthermore, FGF19-induced phosphorylation of SHP at Thr-55 is shown to be important for its functional interaction with SREBP-2 and reduction of liver/serum cholesterol levels. CONCLUSION: This study reveals SHP as a global transcriptional partner of SREBP-2 in regulation of sterol biosynthetic gene networks and provides a potential mechanism for cholesterol-lowering action of FGF19.


Asunto(s)
Colesterol/biosíntesis , Factores de Crecimiento de Fibroblastos/genética , Proteínas de Choque Térmico Pequeñas/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Transcripción Genética , Animales , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/metabolismo , Sitios de Unión , Colesterol/genética , Inmunoprecipitación de Cromatina/métodos , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de Choque Térmico Pequeñas/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Unión Proteica , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...