Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Trauma Case Rep ; 44: 100800, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36895863

RESUMEN

Traumatic maternal cardiac arrest (MCA) is a challenging scenario for the healthcare team. Expanding the focused assessment with sonography for trauma (FAST) and modifying cardiopulmonary resuscitation (CPR) is necessary. Critical components in the resuscitation of reproductive-age women with traumatic cardiac arrest are highlighted using recommendations from Obstetric Life Support™. A morbidly obese female presented to the Emergency Department (ED) with ongoing CPR and massive hemorrhage from two gunshot wounds to the chest. Ultrasound used during secondary survey, revealed an intrauterine pregnancy, with uterine fundus palpated above the umbilicus. Four minutes after arrival at the ED, the trauma surgeon initiated a resuscitative cesarean delivery (RCD) by transverse abdominal incision. The on-call obstetrician completed the procedure, and the neonate was resuscitated and transferred to the neonatal intensive care unit (NICU). Multiple agents and surgical techniques were required to control ongoing uterine and abdominal wall hemorrhage during intermittent return of spontaneous circulation (ROSC). Despite ongoing CPR and management of the patient's chest, pelvic and abdominal wounds, eventually, there was no return of cardiac activity, no organized cardiac rhythm, no measurable end-tidal carbon dioxide, and no palpable pulse. Further resuscitation and initiation of extracorporeal cardiopulmonary resuscitation (ECPR) were deemed futile by the multidisciplinary team and stopped at the 60-minute mark. Our case summarizes essential techniques addressing MCA recommended in OBLS™ courses. Including 1) expanding the FAST exam to assess for pregnancy status, 2) estimating gestational age by fundal height or point-of-care ultrasound, 3) performing a RCD via midline vertical incision at 4 min if pregnancy is suspected to be ≥20 weeks' gestation (fundal height at or above the umbilicus, femoral length of ≥30 mm or biparietal diameter of ≥45 mm), and 4) execution of ECPR for refractory cardiac arrest.

2.
Cells ; 12(2)2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36672221

RESUMEN

The serine/threonine protein kinase calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) plays critical roles in a range of biological processes. Despite its importance, only a handful of inhibitors of CAMKK2 have been disclosed. Having a selective small molecule tool to interrogate this kinase will help demonstrate that CAMKK2 inhibition can be therapeutically beneficial. Herein, we disclose SGC-CAMKK2-1, a selective chemical probe that targets CAMKK2.

3.
J Biol Methods ; 9(2): e160, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733440

RESUMEN

The regulation of cellular energetics is a complex process that requires the coordinated function of multiple organelles. Historically, studies focused on understanding cellular energy utilization and production have been overwhelmingly concentrated on the mitochondria. While mitochondria account for the majority of intracellular energy production, they alone are incapable of maintaining the variable energetic demands of the cell. The peroxisome has recently emerged as a secondary metabolic organelle that complements and improves mitochondrial performance. Although mitochondria and peroxisomes are structurally distinct organelles, they share key functional similarities that allows for the potential to repurpose readily available tools initially developed for mitochondrial assessment to interrogate peroxisomal metabolic function in a novel manner. To this end, we report here on procedures for the isolation, purification and real-time metabolic assessment of peroxisomal ß-oxidation using the Agilent Seahorse® system. When used together, these protocols provide a straightforward, reproducible and highly quantifiable method for measuring the contributions of peroxisomes to cellular and organismal metabolism.

4.
Mol Metab ; 62: 101513, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35562082

RESUMEN

OBJECTIVE: The liver is the primary internal metabolic organ that coordinates whole body energy homeostasis in response to feeding and fasting. Genetic ablation or pharmacological inhibition of calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) has been shown to significantly improve hepatic health and peripheral insulin sensitivity upon overnutrition with high fat diet. However, the precise molecular underpinnings that explain this metabolic protection have remained largely undefined. METHODS: To characterize the role of CaMKK2 in hepatic metabolism, we developed and challenged liver-specific CaMKK2 knockout (CaMKK2LKO) mice with high fat diet and performed glucose and insulin tolerance tests to evaluate peripheral insulin sensitivity. We used a combination of RNA-Sequencing, glucose and fatty acid istotopic tracer studies, a newly developed Seahorse assay for measuring the oxidative capacity of purified peroxisomes, and a degenerate peptide libarary to identify putative CaMKK2 substrates that mechanistically explain the protective effects of hepatic CaMKK2 ablation. RESULTS: Consistent with previous findings, we show that hepatic CaMKK2 ablation significantly improves indices of peripheral insulin sensitivity. Mechanistically, we found that CaMKK2 phosphorylates and regulates GAPDH to promote glucose metabolism and PEX3 to blunt peroxisomal fatty acid catabolism in the liver. CONCLUSION: CaMKK2 is a central metabolic fuel sensor in the liver that significantly contributes to whole body systems metabolism.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Resistencia a la Insulina , Animales , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Ácidos Grasos , Glucosa/metabolismo , Resistencia a la Insulina/fisiología , Ratones
5.
Circulation ; 145(6): 448-464, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35034472

RESUMEN

BACKGROUND: The nuclear receptor Rev-erbα/ß, a key component of the circadian clock, emerges as a drug target for heart diseases, but the function of cardiac Rev-erb has not been studied in vivo. Circadian disruption is implicated in heart diseases, but it is unknown whether cardiac molecular clock dysfunction is associated with the progression of any naturally occurring human heart diseases. Obesity paradox refers to the seemingly protective role of obesity for heart failure, but the mechanism is unclear. METHODS: We generated mouse lines with cardiac-specific Rev-erbα/ß knockout (KO), characterized cardiac phenotype, conducted multi-omics (RNA-sequencing, chromatin immunoprecipitation sequencing, proteomics, and metabolomics) analyses, and performed dietary and pharmacological rescue experiments to assess the time-of-the-day effects. We compared the temporal pattern of cardiac clock gene expression with the cardiac dilation severity in failing human hearts. RESULTS: KO mice display progressive dilated cardiomyopathy and lethal heart failure. Inducible ablation of Rev-erbα/ß in adult hearts causes similar phenotypes. Impaired fatty acid oxidation in the KO myocardium, in particular, in the light cycle, precedes contractile dysfunctions with a reciprocal overreliance on carbohydrate utilization, in particular, in the dark cycle. Increasing dietary lipid or sugar supply in the dark cycle does not affect cardiac dysfunctions in KO mice. However, obesity coupled with systemic insulin resistance paradoxically ameliorates cardiac dysfunctions in KO mice, associated with rescued expression of lipid oxidation genes only in the light cycle in phase with increased fatty acid availability from adipose lipolysis. Inhibition of glycolysis in the light cycle and lipid oxidation in the dark cycle, but not vice versa, ameliorate cardiac dysfunctions in KO mice. Altered temporal patterns of cardiac Rev-erb gene expression correlate with the cardiac dilation severity in human hearts with dilated cardiomyopathy. CONCLUSIONS: The study delineates temporal coordination between clock-mediated anticipation and nutrient-induced response in myocardial metabolism at multi-omics levels. The obesity paradox is attributable to increased cardiac lipid supply from adipose lipolysis in the fasting cycle due to systemic insulin resistance and adiposity. Cardiac molecular chronotypes may be involved in human dilated cardiomyopathy. Myocardial bioenergetics downstream of Rev-erb may be a chronotherapy target in treating heart failure and dilated cardiomyopathy.


Asunto(s)
Ritmo Circadiano/fisiología , Miocardio/patología , Obesidad/fisiopatología , Animales , Relojes Circadianos , Cardiopatías , Humanos , Ratones , Ratones Noqueados
6.
Sci Rep ; 11(1): 3441, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33564037

RESUMEN

A subset of CD4 + lymphocytes, regulatory T cells (Tregs), are necessary for central tolerance and function as suppressors of autoimmunity against self-antigens. The SRC-3 coactivator is an oncogene in multiple cancers and is capable of potentiating numerous transcription factors in a wide variety of cell types. Src-3 knockout mice display broad lymphoproliferation and hypersensitivity to systemic inflammation. Using publicly available bioinformatics data and directed cellular approaches, we show that SRC-3 also is highly enriched in Tregs in mice and humans. Human Tregs lose phenotypic characteristics when SRC-3 is depleted or pharmacologically inhibited, including failure of induction from resting T cells and loss of the ability to suppress proliferation of stimulated T cells. These data support a model for SRC-3 as a coactivator that actively participates in protection from autoimmunity and may support immune evasion of cancers by contributing to the biology of Tregs.


Asunto(s)
Proliferación Celular , Coactivador 3 de Receptor Nuclear/inmunología , Linfocitos T Reguladores/inmunología , Animales , Humanos , Ratones , Ratones Noqueados , Coactivador 3 de Receptor Nuclear/genética
7.
Nat Commun ; 11(1): 6215, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277471

RESUMEN

A distinct 12-hour clock exists in addition to the 24-hour circadian clock to coordinate metabolic and stress rhythms. Here, we show that liver-specific ablation of X-box binding protein 1 (XBP1) disrupts the hepatic 12-hour clock and promotes spontaneous non-alcoholic fatty liver disease (NAFLD). We show that hepatic XBP1 predominantly regulates the 12-hour rhythmicity of gene transcription in the mouse liver and demonstrate that perturbation of the 12-hour clock, but not the core circadian clock, is associated with the onset and progression of this NAFLD phenotype. Mechanistically, we provide evidence that the spliced form of XBP1 (XBP1s) binds to the hepatic 12-hour cistrome to directly regulate the 12-hour clock, with a periodicity paralleling the harmonic activation of the 12-hour oscillatory transcription of many rate-limiting metabolic genes known to have perturbations in human metabolic disease. Functionally, we show that Xbp1 ablation significantly reduces cellular membrane fluidity and impairs lipid homeostasis via rate-limiting metabolic processes in fatty acid monounsaturated and phospholipid remodeling pathways. These findings reveal that genetic disruption of the hepatic 12-hour clock links to the onset and progression of NAFLD development via transcriptional regulator XBP1, and demonstrate a role for XBP1 and the 12-hour clock in the modulation of phospholipid composition and the maintenance of lipid homeostasis.


Asunto(s)
Relojes Circadianos , Homeostasis , Fluidez de la Membrana , Enfermedad del Hígado Graso no Alcohólico/genética , Fosfolípidos/metabolismo , Proteína 1 de Unión a la X-Box/genética , Animales , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/patología , Proteína 1 de Unión a la X-Box/metabolismo
8.
Proc Natl Acad Sci U S A ; 117(49): 31353-31364, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229578

RESUMEN

Progressive remodeling of the heart, resulting in cardiomyocyte (CM) loss and increased inflammation, fibrosis, and a progressive decrease in cardiac function, are hallmarks of myocardial infarction (MI)-induced heart failure. We show that MCB-613, a potent small molecule stimulator of steroid receptor coactivators (SRCs) attenuates pathological remodeling post-MI. MCB-613 decreases infarct size, apoptosis, hypertrophy, and fibrosis while maintaining significant cardiac function. MCB-613, when given within hours post MI, induces lasting protection from adverse remodeling concomitant with: 1) inhibition of macrophage inflammatory signaling and interleukin 1 (IL-1) signaling, which attenuates the acute inflammatory response, 2) attenuation of fibroblast differentiation, and 3) promotion of Tsc22d3-expressing macrophages-all of which may limit inflammatory damage. SRC stimulation with MCB-613 (and derivatives) is a potential therapeutic approach for inhibiting cardiac dysfunction after MI.


Asunto(s)
Ciclohexanonas/farmacología , Infarto del Miocardio/fisiopatología , Piridinas/farmacología , Receptores de Esteroides/metabolismo , Remodelación Ventricular/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Pruebas de Función Cardíaca , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Células RAW 264.7 , ARN/genética , ARN/metabolismo , Transcripción Genética/efectos de los fármacos
9.
PLoS Biol ; 18(1): e3000580, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31935211

RESUMEN

Our group recently characterized a cell-autonomous mammalian 12-h clock independent from the circadian clock, but its function and mechanism of regulation remain poorly understood. Here, we show that in mouse liver, transcriptional regulation significantly contributes to the establishment of 12-h rhythms of mRNA expression in a manner dependent on Spliced Form of X-box Binding Protein 1 (XBP1s). Mechanistically, the motif stringency of XBP1s promoter binding sites dictates XBP1s's ability to drive 12-h rhythms of nascent mRNA transcription at dawn and dusk, which are enriched for basal transcription regulation, mRNA processing and export, ribosome biogenesis, translation initiation, and protein processing/sorting in the Endoplasmic Reticulum (ER)-Golgi in a temporal order consistent with the progressive molecular processing sequence described by the central dogma information flow (CEDIF). We further identified GA-binding proteins (GABPs) as putative novel transcriptional regulators driving 12-h rhythms of gene expression with more diverse phases. These 12-h rhythms of gene expression are cell autonomous and evolutionarily conserved in marine animals possessing a circatidal clock. Our results demonstrate an evolutionarily conserved, intricate network of transcriptional control of the mammalian 12-h clock that mediates diverse biological pathways. We speculate that the 12-h clock is coopted to accommodate elevated gene expression and processing in mammals at the two rush hours, with the particular genes processed at each rush hour regulated by the circadian and/or tissue-specific pathways.


Asunto(s)
Relojes Biológicos/genética , Regulación de la Expresión Génica , Ritmo Ultradiano/genética , Proteína 1 de Unión a la X-Box/fisiología , Animales , Células Cultivadas , Ritmo Circadiano/genética , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Factores de Tiempo , Transcripción Genética , Proteína 1 de Unión a la X-Box/genética
10.
Nat Commun ; 10(1): 2450, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164648

RESUMEN

Tumor-associated myeloid cells regulate tumor growth and metastasis, and their accumulation is a negative prognostic factor for breast cancer. Here we find calcium/calmodulin-dependent kinase kinase (CaMKK2) to be highly expressed within intratumoral myeloid cells in mouse models of breast cancer, and demonstrate that its inhibition within myeloid cells suppresses tumor growth by increasing intratumoral accumulation of effector CD8+ T cells and immune-stimulatory myeloid subsets. Tumor-associated macrophages (TAMs) isolated from Camkk2-/- mice expressed higher levels of chemokines involved in the recruitment of effector T cells compared to WT. Similarly, in vitro generated Camkk2-/- macrophages recruit more T cells, and have a reduced capability to suppress T cell proliferation, compared to WT. Treatment with CaMKK2 inhibitors blocks tumor growth in a CD8+ T cell-dependent manner, and facilitates a favorable reprogramming of the immune cell microenvironment. These data, credential CaMKK2 as a myeloid-selective checkpoint, the inhibition of which may have utility in the immunotherapy of breast cancer.


Asunto(s)
Neoplasias de la Mama/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/inmunología , Carcinoma/inmunología , Neoplasias Mamarias Animales/inmunología , Células Mieloides/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Linfocitos T CD8-positivos/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Proliferación Celular , Quimiocinas/inmunología , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Escape del Tumor/genética
11.
PLoS One ; 13(9): e0198503, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30231032

RESUMEN

Circadian rhythmicity, the 24-hour cycle responsive to light and dark, is determined by periodic oscillations in gene transcription. This phenomenon has broad ramifications in physiologic function. Recent work has disclosed more cycles in gene transcription, and to the uncovering of these we apply a novel signal processing methodology known as the pencil method and compare it to conventional parametric, nonparametric, and statistical methods. METHODS: In order to assess periodicity of gene expression over time, we analyzed a database derived from livers of mice entrained to a 12-hour light/12-hour dark cycle. We also analyzed artificially generated signals to identify differences between the pencil decomposition and other alternative methods. RESULTS: The pencil decomposition revealed hitherto-unsuspected oscillations in gene transcription with 12-hour periodicity. The pencil method was robust in detecting the 24-hour circadian cycle that was known to exist, as well as confirming the existence of shorter-period oscillations. A key consequence of this approach is that orthogonality of the different oscillatory components can be demonstrated. thus indicating a biological independence of these oscillations, that has been subsequently confirmed empirically by knocking out the gene responsible for the 24-hour clock. CONCLUSION: System identification techniques can be applied to biological systems and can uncover important characteristics that may elude visual inspection of the data. SIGNIFICANCE: The pencil method provides new insights on the essence of gene expression and discloses a wide variety of oscillations in addition to the well-studied circadian pattern. This insight opens the door to the study of novel mechanisms by which oscillatory gene expression signals exert their regulatory effect on cells to influence human diseases.


Asunto(s)
Simulación por Computador , Modelos Genéticos , Periodicidad , Fotoperiodo , Activación Transcripcional , Algoritmos , Animales , Ritmo Circadiano , Ratones , Transcripción Genética
12.
Diabetes ; 67(12): 2541-2553, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30002134

RESUMEN

Chronic inflammation accompanies obesity and limits subcutaneous white adipose tissue (WAT) expandability, accelerating the development of insulin resistance and type 2 diabetes mellitus. MicroRNAs (miRNAs) influence expression of many metabolic genes in fat cells, but physiological roles in WAT remain poorly characterized. Here, we report that expression of the miRNA miR-30a in subcutaneous WAT corresponds with insulin sensitivity in obese mice and humans. To examine the hypothesis that restoration of miR-30a expression in WAT improves insulin sensitivity, we injected adenovirus (Adv) expressing miR-30a into the subcutaneous fat pad of diabetic mice. Exogenous miR-30a expression in the subcutaneous WAT depot of obese mice coupled improved insulin sensitivity and increased energy expenditure with decreased ectopic fat deposition in the liver and reduced WAT inflammation. High-throughput proteomic profiling and RNA-Seq suggested that miR-30a targets the transcription factor STAT1 to limit the actions of the proinflammatory cytokine interferon-γ (IFN-γ) that would otherwise restrict WAT expansion and decrease insulin sensitivity. We further demonstrated that miR-30a opposes the actions of IFN-γ, suggesting an important role for miR-30a in defending adipocytes against proinflammatory cytokines that reduce peripheral insulin sensitivity. Together, our data identify a critical molecular signaling axis, elements of which are involved in uncoupling obesity from metabolic dysfunction.


Asunto(s)
Resistencia a la Insulina/fisiología , Hígado/metabolismo , MicroARNs/metabolismo , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/fisiología , Ratones , MicroARNs/genética , Obesidad/etiología , Obesidad/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
13.
14.
Sci Rep ; 7(1): 11793, 2017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28924233

RESUMEN

Binding of calcium to its intracellular receptor calmodulin (CaM) activates a family of Ca2+/CaM-dependent protein kinases. CaMKK2 (Ca2+/CaM-dependent protein kinase kinase 2) is a central member of this kinase family as it controls the actions of a CaMK cascade involving CaMKI, CaMKIV or AMPK. CaMKK2 controls insulin signaling, metabolic homeostasis, inflammation and cancer cell growth highlighting its potential as a therapeutic target for a variety of diseases. STO-609 is a selective, small molecule inhibitor of CaMKK2. Although STO-609 has been used extensively in vitro and in cells to characterize and define new mechanistic functions of CaMKK2, only a few studies have reported the in vivo use of STO-609. We synthesized functional STO-609 and assessed its pharmacological properties through in vitro (kinase assay), ex vivo (human liver microsomes) and in vivo (mouse) model systems. We describe the metabolic processing of STO-609, its toxicity, pharmacokinetics and bioavailability in a variety of mouse tissues. Utilizing these data, we show STO-609 treatment to inhibit CaMKK2 function confers protection against non-alcoholic fatty liver disease. These data provide a valuable resource by establishing criteria for use of STO-609 to inhibit the in vivo functions of CaMKK2 and demonstrate its utility for treating metabolically-related hepatic disease.


Asunto(s)
Bencimidazoles , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Naftalimidas , Enfermedad del Hígado Graso no Alcohólico , Animales , Bencimidazoles/farmacocinética , Bencimidazoles/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/patología , Naftalimidas/farmacocinética , Naftalimidas/farmacología , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/prevención & control
15.
Cell Metab ; 25(6): 1305-1319.e9, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28591634

RESUMEN

Besides circadian rhythms, oscillations cycling with a 12 hr period exist. However, the prevalence, origin, regulation, and function of mammalian 12 hr rhythms remain elusive. Utilizing an unbiased mathematical approach identifying all superimposed oscillations, we uncovered prevalent 12 hr gene expression and metabolic rhythms in mouse liver, coupled with a physiological 12 hr unfolded protein response oscillation. The mammalian 12 hr rhythm is cell autonomous, driven by a dedicated 12 hr pacemaker distinct from the circadian clock, and can be entrained in vitro by metabolic and ER stress cues. Mechanistically, we identified XBP1s as a transcriptional regulator of the mammalian 12 hr clock. Downregulation of the 12 hr gene expression strongly correlates with human hepatic steatosis and steatohepatitis, implying its importance in maintaining metabolic homeostasis. The mammalian 12 hr rhythm of gene expression also is conserved in nematodes and crustaceans, indicating an ancient origin of the 12 hr clock. Our work sheds new light on how perturbed biological rhythms contribute to human disease.


Asunto(s)
Relojes Circadianos/fisiología , Estrés del Retículo Endoplásmico/fisiología , Regulación de la Expresión Génica/fisiología , Respuesta de Proteína Desplegada/fisiología , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Línea Celular , Humanos , Ratones , Ratones Transgénicos , Proteína 1 de Unión a la X-Box/genética
16.
Nat Rev Endocrinol ; 13(8): 445-457, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28524171

RESUMEN

A growing epidemic of nonalcoholic fatty liver disease (NAFLD) is paralleling the increase in the incidence of obesity and diabetes mellitus in countries that consume a Western diet. As NAFLD can lead to life-threatening conditions such as cirrhosis and hepatocellular carcinoma, an understanding of the factors that trigger its development and pathological progression is needed. Although by definition this disease is not associated with alcohol consumption, exposure to environmental agents that have been linked to other diseases might have a role in the development of NAFLD. Here, we focus on one class of these agents, endocrine-disrupting chemicals (EDCs), and their potential to influence the initiation and progression of a cascade of pathological conditions associated with hepatic steatosis (fatty liver). Experimental studies have revealed several potential mechanisms by which EDC exposure might contribute to disease pathogenesis, including the modulation of nuclear hormone receptor function and the alteration of the epigenome. However, many questions remain to be addressed about the causal link between acute and chronic EDC exposure and the development of NAFLD in humans. Future studies that address these questions hold promise not only for understanding the linkage between EDC exposure and liver disease but also for elucidating the molecular mechanisms that underpin NAFLD, which in turn could facilitate the development of new prevention and treatment opportunities.


Asunto(s)
Disruptores Endocrinos/efectos adversos , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/metabolismo , Obesidad/patología , Factores de Riesgo
17.
PLoS Genet ; 13(3): e1006650, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28273073

RESUMEN

Hepatocellular carcinoma (HCC) is the fifth most common solid tumor in the world and the third leading cause of cancer-associated deaths. A Sleeping Beauty-mediated transposon mutagenesis screen previously identified mutations that cooperate with MYC to accelerate liver tumorigenesis. This revealed a tumor suppressor role for Steroid Receptor Coactivator 2/Nuclear Receptor Coactivator 2 (Src-2/Ncoa2) in liver cancer. In contrast, SRC-2 promotes survival and metastasis in prostate cancer cells, suggesting a tissue-specific and context-dependent role for SRC-2 in tumorigenesis. To determine if genetic loss of SRC-2 is sufficient to accelerate MYC-mediated liver tumorigenesis, we bred Src-2-/- mice with a MYC-induced liver tumor model and observed a significant increase in liver tumor burden. RNA sequencing of liver tumors and in vivo chromatin immunoprecipitation assays revealed a set of direct target genes that are bound by SRC-2 and exhibit downregulated expression in Src-2-/- liver tumors. We demonstrate that activation of SHP (Small Heterodimer Partner), DKK4 (Dickkopf-4), and CADM4 (Cell Adhesion Molecule 4) by SRC-2 suppresses tumorigenesis in vitro and in vivo. These studies suggest that SRC-2 may exhibit oncogenic or tumor suppressor activity depending on the target genes and nuclear receptors that are expressed in distinct tissues and illuminate the mechanisms of tumor suppression by SRC-2 in liver.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Alelos , Animales , Antineoplásicos/química , Carcinogénesis , Carcinoma Hepatocelular/genética , Moléculas de Adhesión Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Inmunoprecipitación de Cromatina , Elementos Transponibles de ADN , Femenino , Eliminación de Gen , Células Hep G2 , Humanos , Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Mutagénesis , Metástasis de la Neoplasia , Trasplante de Neoplasias , Coactivador 2 del Receptor Nuclear/genética , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Análisis de Secuencia de ARN
18.
J Biol Chem ; 291(47): 24747-24755, 2016 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-27758866

RESUMEN

The acquisition of beige adipocyte features by white fat cells corresponds to protection against obesity-induced metabolic diseases in humans and animal models of type 2 diabetes. In adipose tissue, expression of the E2 small ubiquitin-like modifier ligase ubiquitin carrier protein 9 (Ubc9) is positively correlated with markers of insulin resistance and corresponds with impaired browning of human white adipocytes. However, the molecular regulation of Ubc9 expression in adipocytes and other cells remains unclear. In this study, we demonstrate that the mRNA and protein expression of Ubc9 are regulated by the microRNA miRNA-30a (miR-30a) in human subcutaneous adipocytes. Ubc9 and miR-30a exhibit inverse expression in adipose tissue, with miR-30a robustly elevated in brown fat. Depletion of Ubc9 by siRNA or enforced expression of a miR-30a mimic augments mitochondrial volume and respiration in human white adipocytes, reflecting features of brown fat cells. Furthermore, Ubc9 depletion induces a brown fat gene program in human subcutaneous adipocytes. Induction of the beige-selective gene program corresponds to stabilization of the PR domain-containing 16 (PRDM16) protein, an obligate transcriptional regulator of the brown/beige fat metabolic program in white adipocytes that interacts with Ubc9. Taken together, our data demonstrate a previously unappreciated molecular axis that controls browning of human white adipocytes.


Asunto(s)
Adipocitos Blancos/metabolismo , Regulación de la Expresión Génica/fisiología , MicroARNs/biosíntesis , Mitocondrias/metabolismo , Enzimas Ubiquitina-Conjugadoras/biosíntesis , Adipocitos Blancos/citología , Animales , Proteínas de Unión al ADN/metabolismo , Humanos , Masculino , Ratones , Factores de Transcripción/metabolismo
19.
J Biol Rhythms ; 31(5): 443-60, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27432117

RESUMEN

Circadian rhythmicity is a fundamental process that synchronizes behavioral cues with metabolic homeostasis. Disruption of daily cycles due to jet lag or shift work results in severe physiological consequences including advanced aging, metabolic syndrome, and even cancer. Our understanding of the molecular clock, which is regulated by intricate positive feedforward and negative feedback loops, has expanded to include an important metabolic transcriptional coregulator, Steroid Receptor Coactivator-2 (SRC-2), that regulates both the central clock of the suprachiasmatic nucleus (SCN) and peripheral clocks including the liver. We hypothesized that an environmental uncoupling of the light-dark phases, termed chronic circadian disruption (CCD), would lead to pathology similar to the genetic circadian disruption observed with loss of SRC-2 We found that CCD and ablation of SRC-2 in mice led to a common comorbidity of metabolic syndrome also found in humans with circadian disruption, non-alcoholic fatty liver disease (NAFLD). The combination of SRC-2(-/-) and CCD results in a more robust phenotype that correlates with human non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) gene signatures. Either CCD or SRC-2 ablation produces an advanced aging phenotype leading to increased mortality consistent with other circadian mutant mouse models. Collectively, our studies demonstrate that SRC-2 provides an essential link between the behavioral activities influenced by light cues and the metabolic homeostasis maintained by the liver.


Asunto(s)
Envejecimiento , Hígado/patología , Coactivador 2 del Receptor Nuclear/genética , Coactivador 2 del Receptor Nuclear/fisiología , Animales , Carcinoma Hepatocelular/genética , Relojes Circadianos , Ritmo Circadiano/fisiología , Modelos Animales de Enfermedad , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/genética , Ratones , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Coactivador 2 del Receptor Nuclear/deficiencia , Proteínas Circadianas Period/genética , Fotoperiodo , Núcleo Supraquiasmático/fisiología
20.
Trends Endocrinol Metab ; 27(10): 706-718, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27449752

RESUMEN

Calcium (Ca(2+)) is an essential ligand that binds its primary intracellular receptor calmodulin (CaM) to trigger a variety of downstream processes and pathways. Central to the actions of Ca(2+)/CaM is the activation of a highly conserved Ca(2+)/CaM kinase (CaMK) cascade that amplifies Ca(2+) signals through a series of subsequent phosphorylation events. Proper regulation of Ca(2+) flux is necessary for whole-body metabolism and disruption of Ca(2+) homeostasis has been linked to various metabolic diseases. Here we provide a synthesis of recent advances that highlight the roles of the Ca(2+)/CaMK axis in key metabolic tissues. An appreciation of this information is critical to understanding the mechanisms by which Ca(2+)/CaM-dependent signaling contributes to metabolic homeostasis and disease.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Humanos , Fosforilación , Transducción de Señal/genética , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...