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.
bioRxiv ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38464291

RESUMEN

Lung cancer, the leading cause of cancer mortality, exhibits diverse histological subtypes and genetic complexities. Numerous preclinical mouse models have been developed to study lung cancer, but data from these models are disparate, siloed, and difficult to compare in a centralized fashion. Here we established the Lung Cancer Mouse Model Database (LCMMDB), an extensive repository of 1,354 samples from 77 transcriptomic datasets covering 974 samples from genetically engineered mouse models (GEMMs), 368 samples from carcinogen-induced models, and 12 samples from a spontaneous model. Meticulous curation and collaboration with data depositors have produced a robust and comprehensive database, enhancing the fidelity of the genetic landscape it depicts. The LCMMDB aligns 859 tumors from GEMMs with human lung cancer mutations, enabling comparative analysis and revealing a pressing need to broaden the diversity of genetic aberrations modeled in GEMMs. Accompanying this resource, we developed a web application that offers researchers intuitive tools for in-depth gene expression analysis. With standardized reprocessing of gene expression data, the LCMMDB serves as a powerful platform for cross-study comparison and lays the groundwork for future research, aiming to bridge the gap between mouse models and human lung cancer for improved translational relevance.

2.
Sci Adv ; 10(1): eadj4686, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38170783

RESUMEN

Type 1 diabetes mellitus (T1DM) is characterized by insulin deficiency leading to hyperglycemia and several metabolic defects. Insulin therapy remains the cornerstone of T1DM management, yet it increases the risk of life-threatening hypoglycemia and the development of major comorbidities. Here, we report an insulin signaling-independent pathway able to improve glycemic control in T1DM rodents. Co-treatment with recombinant S100 calcium-binding protein A9 (S100A9) enabled increased adherence to glycemic targets with half as much insulin and without causing hypoglycemia. Mechanistically, we demonstrate that the hyperglycemia-suppressing action of S100A9 is due to a Toll-like receptor 4-dependent increase in glucose uptake in specific skeletal muscles (i.e., soleus and diaphragm). In addition, we found that T1DM mice have abnormal systemic inflammation, which is resolved by S100A9 therapy alone (or in combination with low insulin), hence uncovering a potent anti-inflammatory action of S100A9 in T1DM. In summary, our findings reveal the S100A9-TLR4 skeletal muscle axis as a promising therapeutic target for improving T1DM treatment.


Asunto(s)
Diabetes Mellitus Tipo 1 , Hiperglucemia , Hipoglucemia , Animales , Ratones , Insulina/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Hipoglucemia/complicaciones , Hipoglucemia/tratamiento farmacológico , Hiperglucemia/tratamiento farmacológico , Calgranulina B
3.
Nat Commun ; 13(1): 4107, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35840613

RESUMEN

Unrestrained ketogenesis leads to life-threatening ketoacidosis whose incidence is high in patients with diabetes. While insulin therapy reduces ketogenesis this approach is sub-optimal. Here, we report an insulin-independent pathway able to normalize diabetic ketogenesis. By generating insulin deficient male mice lacking or re-expressing Toll-Like Receptor 4 (TLR4) only in liver or hepatocytes, we demonstrate that hepatic TLR4 in non-parenchymal cells mediates the ketogenesis-suppressing action of S100A9. Mechanistically, S100A9 acts extracellularly to activate the mechanistic target of rapamycin complex 1 (mTORC1) in a TLR4-dependent manner. Accordingly, hepatic-restricted but not hepatocyte-restricted loss of Tuberous Sclerosis Complex 1 (TSC1, an mTORC1 inhibitor) corrects insulin-deficiency-induced hyperketonemia. Therapeutically, recombinant S100A9 administration restrains ketogenesis and improves hyperglycemia without causing hypoglycemia in diabetic mice. Also, circulating S100A9 in patients with ketoacidosis is only marginally increased hence unveiling a window of opportunity to pharmacologically augment S100A9 for preventing unrestrained ketogenesis. In summary, our findings reveal the hepatic S100A9-TLR4-mTORC1 axis in non-parenchymal cells as a promising therapeutic target for restraining diabetic ketogenesis.


Asunto(s)
Diabetes Mellitus Experimental , Cetosis , Animales , Calgranulina B/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Insulina/metabolismo , Cuerpos Cetónicos/metabolismo , Hígado/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
4.
Cell Rep ; 30(11): 3851-3863.e6, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32187554

RESUMEN

Cancer therapy is limited, in part, by lack of specificity. Thus, identifying molecules that are selectively expressed by, and relevant for, cancer cells is of paramount medical importance. Here, we show that peptidyl-prolyl-cis-trans-isomerase (PPIase) FK506-binding protein 10 (FKBP10)-positive cells are present in cancer lesions but absent in the healthy parenchyma of human lung. FKBP10 expression negatively correlates with survival of lung cancer patients, and its downregulation causes a dramatic diminution of lung tumor burden in mice. Mechanistically, our results from gain- and loss-of-function assays show that FKBP10 boosts cancer growth and stemness via its PPIase activity. Also, FKBP10 interacts with ribosomes, and its downregulation leads to reduction of translation elongation at the beginning of open reading frames (ORFs), particularly upon insertion of proline residues. Thus, our data unveil FKBP10 as a cancer-selective molecule with a key role in translational reprogramming, stem-like traits, and growth of lung cancer.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Biosíntesis de Proteínas , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Isomerasa de Peptidilprolil/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ribosomas/metabolismo
5.
Nat Commun ; 10(1): 3545, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31391467

RESUMEN

Tens of millions suffer from insulin deficiency (ID); a defect leading to severe metabolic imbalance and death. The only means for management of ID is insulin therapy; yet, this approach is sub-optimal and causes life-threatening hypoglycemia. Hence, ID represents a great medical and societal challenge. Here we report that S100A9, also known as Calgranulin B or Myeloid-Related Protein 14 (MRP14), is a leptin-induced circulating cue exerting beneficial anti-diabetic action. In murine models of ID, enhanced expression of S100A9 alone (i.e. without administered insulin and/or leptin) slightly improves hyperglycemia, and normalizes key metabolic defects (e.g. hyperketonemia, hypertriglyceridemia, and increased hepatic fatty acid oxidation; FAO), and extends lifespan by at least a factor of two. Mechanistically, we report that Toll-Like Receptor 4 (TLR4) is required, at least in part, for the metabolic-improving and pro-survival effects of S100A9. Thus, our data identify the S100A9/TLR4 axis as a putative target for ID care.


Asunto(s)
Calgranulina B/metabolismo , Diabetes Mellitus Experimental/metabolismo , Hiperglucemia/metabolismo , Longevidad/fisiología , Receptor Toll-Like 4/metabolismo , Animales , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/etiología , Toxina Diftérica/toxicidad , Ácidos Grasos/metabolismo , Humanos , Hiperglucemia/sangre , Hiperglucemia/etiología , Insulina/deficiencia , Leptina/administración & dosificación , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Oxidación-Reducción , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estreptozocina/toxicidad , Receptor Toll-Like 4/genética
6.
Cell Rep ; 27(8): 2385-2398.e3, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31116983

RESUMEN

Loss of synchrony between geophysical time and insulin action predisposes to metabolic diseases. Yet the brain and peripheral pathways linking proper insulin effect to diurnal changes in light-dark and feeding-fasting inputs are poorly understood. Here, we show that the insulin sensitivity of several metabolically relevant tissues fluctuates during the 24 h period. For example, in mice, the insulin sensitivity of skeletal muscle, liver, and adipose tissue is lowest during the light period. Mechanistically, by performing loss- and gain-of-light-action and food-restriction experiments, we demonstrate that SIRT1 in steroidogenic factor 1 (SF1) neurons of the ventromedial hypothalamic nucleus (VMH) convey photic inputs to entrain the biochemical and metabolic action of insulin in skeletal muscle. These findings uncover a critical light-SF1-neuron-skeletal-muscle axis that acts to finely tune diurnal changes in insulin sensitivity and reveal a light regulatory mechanism of skeletal muscle function.


Asunto(s)
Insulina/metabolismo , Músculo Esquelético/metabolismo , Fototerapia/métodos , Núcleo Hipotalámico Ventromedial/fisiopatología , Animales , Ritmo Circadiano , Humanos , Ratones
7.
Nat Commun ; 8(1): 1820, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29180649

RESUMEN

Obesity-induced inflammation engenders insulin resistance and type 2 diabetes mellitus (T2DM) but the inflammatory effectors linking obesity to insulin resistance are incompletely understood. Here, we show that hepatic expression of Protein Tyrosine Phosphatase Receptor Gamma (PTPR-γ) is stimulated by inflammation in obese/T2DM mice and positively correlates with indices of inflammation and insulin resistance in humans. NF-κB binds to the promoter of Ptprg and is required for inflammation-induced PTPR-γ expression. PTPR-γ loss-of-function lowers glycemia and insulinemia by enhancing insulin-stimulated suppression of endogenous glucose production. These phenotypes are rescued by re-expression of Ptprg only in liver of mice lacking Ptprg globally. Hepatic PTPR-γ overexpression that mimics levels found in obesity is sufficient to cause severe hepatic and systemic insulin resistance. We propose hepatic PTPR-γ as a link between obesity-induced inflammation and insulin resistance and as potential target for treatment of T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Obesidad/metabolismo , Proteínas Tirosina Fosfatasas Similares a Receptores/metabolismo , Adulto , Anciano , Animales , Glucemia , Línea Celular , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Células Hep G2 , Humanos , Inflamación/metabolismo , Insulina/sangre , Interleucina-6/metabolismo , Metabolismo de los Lípidos , Lipopolisacáridos/efectos adversos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Persona de Mediana Edad , Modelos Animales , FN-kappa B/metabolismo , Obesidad/sangre , Obesidad/complicaciones , Proteínas Tirosina Fosfatasas/metabolismo , ARN Mensajero/biosíntesis , Proteínas Tirosina Fosfatasas Similares a Receptores/genética , Sirtuina 1/metabolismo
8.
Cell Rep ; 18(8): 1858-1868, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228253

RESUMEN

Cancer stem cells (CSCs) have high tumorigenic capacity. Here, we show that stem-like traits of specific human cancer cells are reduced by overexpression of the histone deacetylase sirtuin 6 (SIRT6). SIRT6-sensitive cancer cells bear mutations that activate phosphatidylinositol-3-kinase (PI3K) signaling, and overexpression of SIRT6 reduces growth, progression, and grade of breast cancer in a mouse model with PI3K activation. Tumor metabolomic and transcriptomic analyses reveal that SIRT6 overexpression dampens PI3K signaling and stem-like characteristics and causes metabolic rearrangements in this cancer model. Ablation of a PI3K activating mutation in otherwise isogenic cancer cells is sufficient to convert SIRT6-sensitive into SIRT6-insensitive cells. SIRT6 overexpression suppresses PI3K signaling at the transcriptional level and antagonizes tumor sphere formation independent of its histone deacetylase activity. Our data identify SIRT6 as a putative molecular target that hinders stemness of tumors with PI3K activation.


Asunto(s)
Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Sirtuinas/metabolismo , Acetilación , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación/fisiología , Transducción de Señal/fisiología , Transcripción Genética/fisiología
9.
Cell Metab ; 23(3): 467-78, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26959185

RESUMEN

Organismal homeostasis relies on coherent interactions among tissues, specifically between brain-driven functions and peripheral metabolic organs. Hypothalamic circuits compute metabolic information to optimize energetic resources, but the role of the circadian clock in these pathways remains unclear. We have generated mice with targeted ablation of the core-clock gene Bmal1 within Sf1-neurons of the ventromedial hypothalamus (VMH). While this mutation does not affect the central clock in the suprachiasmatic nucleus (SCN), the VMH clock controls cyclic thermogenesis in brown adipose tissue (BAT), a tissue that governs energy balance by dissipating chemical energy as heat. VMH-driven control is exerted through increased adrenergic signaling within the sympathetic nervous system, without affecting the BAT's endogenous clock. Moreover, we show that the VMH circadian clock computes light and feeding inputs to modulate basal energy expenditure. Thus, we reveal a previously unsuspected circuit where an SCN-independent, hypothalamic circadian clock controls BAT function, energy expenditure, and thermogenesis.


Asunto(s)
Metabolismo Energético , Núcleo Supraquiasmático/fisiología , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Animales , Relojes Circadianos , Ritmo Circadiano , Homeostasis , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Proteínas Circadianas Period/fisiología , Sistema Nervioso Simpático/fisiología , Termogénesis
10.
Mol Metab ; 4(11): 846-56, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26629408

RESUMEN

OBJECTIVE: Available treatment for obesity and type 2 diabetes mellitus (T2DM) is suboptimal. Thus, identifying novel molecular target(s) exerting protective effects against these metabolic imbalances is of enormous medical significance. Sirt6 loss- and gain-of-function studies have generated confounding data regarding the role of this sirtuin on energy and glucose homeostasis, leaving unclear whether activation or inhibition of SIRT6 may be beneficial for the treatment of obesity and/or T2DM. METHODS: To address these issues, we developed and studied a novel mouse model designed to produce eutopic and physiological overexpression of SIRT6 (Sirt6BAC mice). These mutants and their controls underwent several metabolic analyses. These include whole-blood reverse phase high-performance liquid chromatography assay, glucose and pyruvate tolerance tests, hyperinsulinemic-euglycemic clamp assays, and assessment of basal and insulin-induced level of phosphorylated AKT (p-AKT)/AKT in gastrocnemius muscle. RESULTS: Sirt6BAC mice physiologically overexpress functionally competent SIRT6 protein. While Sirt6BAC mice have normal body weight and adiposity, they are protected from developing high-caloric-diet (HCD)-induced hyperglycemia and glucose intolerance. Also, Sirt6BAC mice display increased circulating level of the polyamine spermidine. The ability of insulin to suppress endogenous glucose production was significantly enhanced in Sirt6BAC mice compared to wild-type controls. Insulin-stimulated glucose uptake was increased in Sirt6BAC mice in both gastrocnemius and soleus muscle, but not in brain, interscapular brown adipose, or epididymal adipose tissue. Insulin-induced p-AKT/AKT ratio was increased in gastrocnemius muscle of Sirt6BAC mice compared to wild-type controls. CONCLUSIONS: Our data indicate that moderate, physiological overexpression of SIRT6 enhances insulin sensitivity in skeletal muscle and liver, engendering protective actions against diet-induced T2DM. Hence, the present study provides support for the anti-T2DM effect of SIRT6 and suggests SIRT6 as a putative molecular target for anti-T2DM treatment.

11.
Endocrinology ; 156(6): 2174-84, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25763637

RESUMEN

Circadian rhythms govern homeostasis and organism physiology. Nutritional cues act as time givers, contributing to the synchronization between central and peripheral clocks. Neuronal food-synchronized clocks are thought to reside in hypothalamic nuclei such as the ventromedial hypothalamus (VMH) and the dorsomedial hypothalamus or extrahypothalamic brain areas such as nucleus accumbens. Interestingly, the metabolic sensor of nicotinamide adenine dinucleotide-dependent deacetylase sirtuin-1 (SIRT1) is highly expressed in the VMH and was shown to contribute to both control of energy balance and clock function. We used mice with targeted ablation of Sirt1 in the steroidogenic factor 1 neurons of the VMH to gain insight on the role played by this deacetylase in the modulation of the central clock by nutritional inputs. By studying circadian behavior and circadian gene expression, we reveal that SIRT1 operates as a metabolic sensor connecting food intake to circadian behavior. Indeed, under food restriction and absence of light, SIRT1 in the VMH contributes to activity behavior and circadian gene expression in the suprachiasmatic nucleus. Thus, under specific physiological conditions, SIRT1 contributes to the modulation of the circadian clock by nutrients.


Asunto(s)
Relojes Circadianos/fisiología , Proteínas de Unión al ADN/metabolismo , Neuronas/metabolismo , Sirtuina 1/metabolismo , Factores de Transcripción/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Encéfalo/metabolismo , Relojes Circadianos/genética , Proteínas de Unión al ADN/genética , Masculino , Ratones , Factores de Empalme de ARN , Sirtuina 1/genética , Factores de Transcripción/genética
12.
Cell Metab ; 21(1): 117-25, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25533479

RESUMEN

Dietary effects on tumor biology can be exploited to unravel cancer vulnerabilities. Here, we present surprising evidence for anti-proliferative action of high-calorie-diet (HCD) feeding on KRAS-driven lung tumors. Tumors of mice that commenced HCD feeding before tumor onset displayed defective unfolded protein response (UPR) and unresolved endoplasmic reticulum (ER) stress. Unresolved ER stress and reduced proliferation are reversed by chemical chaperone treatment. Whole-genome transcriptional analyses revealed FKBP10 as one of the most downregulated chaperones in tumors of the HCD-pre-tumor-onset group. FKBP10 downregulation dampens tumor growth in vitro and in vivo. Providing translational value to these results, we report that FKBP10 is expressed in human KRAS-positive and -negative lung cancers, but not in healthy parenchyma. Collectively, our data shed light on an unexpected anti-tumor action of HCD imposed before tumor onset and identify FKBP10 as a putative therapeutic target to selectively hinder lung cancer.


Asunto(s)
Dieta , Neoplasias Pulmonares/patología , Fenilbutiratos/toxicidad , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Regulación hacia Abajo , Doxiciclina/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones SCID , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Trasplante Heterólogo , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteínas ras/genética
13.
Cell Metab ; 18(3): 431-44, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-24011077

RESUMEN

The dogma that life without insulin is incompatible has recently been challenged by results showing the viability of insulin-deficient rodents undergoing leptin monotherapy. Yet, the mechanisms underlying these actions of leptin are unknown. Here, the metabolic outcomes of intracerebroventricular (i.c.v.) administration of leptin in mice devoid of insulin and lacking or re-expressing leptin receptors (LEPRs) only in selected neuronal groups were assessed. Our results demonstrate that concomitant re-expression of LEPRs only in hypothalamic γ-aminobutyric acid (GABA) and pro-opiomelanocortin (POMC) neurons is sufficient to fully mediate the lifesaving and antidiabetic actions of leptin in insulin deficiency. Our analyses indicate that enhanced glucose uptake by brown adipose tissue and soleus muscle, as well as improved hepatic metabolism, underlies these effects of leptin. Collectively, our data elucidate a hypothalamic-dependent pathway enabling life without insulin and hence pave the way for developing better treatments for diseases of insulin deficiency.


Asunto(s)
Hipotálamo/efectos de los fármacos , Insulina/metabolismo , Leptina/farmacología , Neuronas/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Glucosa/análisis , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/mortalidad , Hipotálamo/metabolismo , Estimación de Kaplan-Meier , Leptina/uso terapéutico , Hígado/metabolismo , Ratones , Músculo Esquelético/metabolismo , Neuronas/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
14.
Cancer Discov ; 3(4): 444-57, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23358651

RESUMEN

UNLABELLED: Non-small cell lung cancer (NSCLC) often expresses mutant KRAS together with tumor-associated mutations of the CDKN2A locus, which are associated with aggressive, therapy-resistant tumors. Here, we unravel specific requirements for the maintenance of NSCLC that carries this genotype. We establish that the extracellular signal-regulated kinase (ERK)/RHOA/focal adhesion kinase (FAK) network is deregulated in high-grade lung tumors. Suppression of RHOA or FAK induces cell death selectively in mutant KRAS;INK4A/ARF-deficient lung cancer cells. Furthermore, pharmacologic inhibition of FAK caused tumor regression specifically in the high-grade lung cancer that developed in mutant Kras;Cdkn2a-null mice. These findings provide a rationale for the rapid implementation of genotype-specific targeted therapies using FAK inhibitors in patients with cancer. SIGNIFICANCE: Targeted therapies are effective for only a small fraction of patients with cancer. We report that FAK inhibitors exert potent antitumor effects in NSCLCs that express mutant KRAS in association with INK4A/ARF deficiency. These results reveal a novel genotype-specific vulnerability of cancer cells that can be exploited for therapeutic purposes.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Genes ras/genética , Proteína de Unión al GTP rhoA/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Vaccines (Basel) ; 1(4): 463-80, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-26344342

RESUMEN

In veterinary medicine, there have been different experiences with the plasmid DNA vaccination. In this area and with the hypothesis to demonstrate the effectiveness of different plasmids encoding porcine respiratory and reproductive syndrome (PRRS), five DNA vaccines against PRRS were evaluated for their innocuity and efficacy in pigs. Eighteen animals were divided into five groups which were injected with five (A, B, C, D, E) different DNA vaccines. Albeit, none of the proposed vaccines were able to protect the animals against PRRS virus. Only vaccines A and B were able to reduce the clinical signs of the infection. ELISA IgM were detected 30 days after the first vaccination in the pigs injected by Vaccine A or B. ELISA IgG were detected 90 days after the first vaccination in the pigs injected by Vaccine B or C. Neutralizing antibody were detected Post Challenge Days 61 (PCD) in all groups. In the pigs inoculated with Vaccine C, IFN-g were detected 90 days after first vaccination, and after challenge exposure they increased. In the other groups, the IFN-g were detected after challenge infection. Pigs injected with each of the vaccines A, B, C, D and E showed a significantly higher level of CD4(-)CD8⁺ lymphocytes (p < 0.001) after infection in comparison with their controls.

16.
Cell Metab ; 15(5): 675-90, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22560220

RESUMEN

Resveratrol induces mitochondrial biogenesis and protects against metabolic decline, but whether SIRT1 mediates these benefits is the subject of debate. To circumvent the developmental defects of germline SIRT1 knockouts, we have developed an inducible system that permits whole-body deletion of SIRT1 in adult mice. Mice treated with a moderate dose of resveratrol showed increased mitochondrial biogenesis and function, AMPK activation, and increased NAD(+) levels in skeletal muscle, whereas SIRT1 knockouts displayed none of these benefits. A mouse overexpressing SIRT1 mimicked these effects. A high dose of resveratrol activated AMPK in a SIRT1-independent manner, demonstrating that resveratrol dosage is a critical factor. Importantly, at both doses of resveratrol no improvements in mitochondrial function were observed in animals lacking SIRT1. Together these data indicate that SIRT1 plays an essential role in the ability of moderate doses of resveratrol to stimulate AMPK and improve mitochondrial function both in vitro and in vivo.


Asunto(s)
Mitocondrias/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Proteínas Quinasas/metabolismo , Sirtuina 1/metabolismo , Estilbenos/farmacología , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Células Cultivadas , Activación Enzimática , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/enzimología , Mitocondrias/genética , Músculo Esquelético/enzimología , NAD/metabolismo , Proteínas Quinasas/genética , Resveratrol , Transducción de Señal/efectos de los fármacos , Sirtuina 1/genética
17.
Cell Metab ; 14(3): 301-12, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21907137

RESUMEN

Chronic feeding on high-calorie diets causes obesity and type 2 diabetes mellitus (T2DM), illnesses that affect hundreds of millions. Thus, understanding the pathways protecting against diet-induced metabolic imbalance is of paramount medical importance. Here, we show that mice lacking SIRT1 in steroidogenic factor 1 (SF1) neurons are hypersensitive to dietary obesity owing to maladaptive energy expenditure. Also, mutant mice have increased susceptibility to developing dietary T2DM due to insulin resistance in skeletal muscle. Mechanistically, these aberrations arise, in part, from impaired metabolic actions of the neuropeptide orexin-A and the hormone leptin. Conversely, mice overexpressing SIRT1 in SF1 neurons are more resistant to diet-induced obesity and insulin resistance due to increased energy expenditure and enhanced skeletal muscle insulin sensitivity. Our results unveil important protective roles of SIRT1 in SF1 neurons against dietary metabolic imbalance.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/farmacología , Neuronas/metabolismo , Neuropéptidos/farmacología , Obesidad/metabolismo , Sirtuina 1/deficiencia , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/metabolismo , Grasas de la Dieta/farmacología , Metabolismo Energético , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Inmunohistoquímica , Insulina/metabolismo , Insulina/farmacología , Resistencia a la Insulina , Leptina/farmacología , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Obesidad/complicaciones , Obesidad/patología , Orexinas , Técnicas de Placa-Clamp , Sirtuina 1/genética , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo
18.
Aging (Albany NY) ; 3(3): 325-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21464518

RESUMEN

In virtually all organisms, life expectancy is profoundly affected by caloric intake. For example, dietary restriction (DR; a feeding regimen of fewer calories compared to the ad libitum level without causing malnutrition) has been shown to lengthen, whereas hypercaloric (HC) diet feeding to shorten, lifespan. Recent findings in invertebrates indicate that specialized groups of cells (e.g.: metabolic-sensing neurons) detect changes in caloric intake and convey energy-status-variation signals to other cells in the body to regulate lifespan. In mammals, whether metabolic-sensing neurons govern aging in a cell-non-autonomous fashion is unknown. Yet, this is a captivating and testable hypothesis.


Asunto(s)
Envejecimiento/fisiología , Hipotálamo/fisiología , Sirtuina 1/fisiología , Tejido Adiposo Pardo/fisiología , Animales , Restricción Calórica , Humanos , Longevidad/fisiología , Modelos Biológicos , Neuronas/fisiología
19.
PLoS One ; 5(11): e14131, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21152434

RESUMEN

BACKGROUND: Mounting clinical and experimental evidence suggests that the shift of carcinomas towards a mesenchymal phenotype is a common paradigm for both resistance to therapy and tumor recurrence. However, the mesenchymalization of carcinomas has not yet entered clinical practice as a crucial diagnostic paradigm. METHODOLOGY/PRINCIPAL FINDINGS: By integrating in silico and in vitro studies with our epithelial and mesenchymal tumor models, we compare herein crucial molecular pathways of previously described carcinoma-derived mesenchymal tumor cells (A17) with that of both carcinomas and other mesenchymal phenotypes, such as mesenchymal stem cells (MSCs), breast stroma, and various types of sarcomas. We identified three mesenchymal/stromal-signatures which A17 cells shares with MSCs and breast stroma. By using a recently developed computational approach with publicly available microarray data, we show that these signatures: 1) significantly relates to basal-like breast cancer subtypes; 2) significantly relates to bone metastasis; 3) are up-regulated after hormonal treatment; 4) predict resistance to neoadjuvant therapies. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that mesenchymalization is an intrinsic property of the most aggressive tumors and it relates to therapy resistance as well as bone metastasis.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias de la Mama/genética , Perfilación de la Expresión Génica , Mesodermo/metabolismo , Células del Estroma/metabolismo , Animales , Western Blotting , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Análisis por Conglomerados , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Resistencia a Antineoplásicos/genética , Células Epiteliales/metabolismo , Femenino , Humanos , Células Madre Mesenquimatosas/metabolismo , Mesodermo/patología , Ratones , Terapia Neoadyuvante/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células del Estroma/patología
20.
Proc Natl Acad Sci U S A ; 107(40): 17391-6, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20855609

RESUMEN

Leptin monotherapy reverses the deadly consequences and improves several of the metabolic imbalances caused by insulin-deficient type 1 diabetes (T1D) in rodents. However, the mechanism(s) underlying these effects is totally unknown. Here, we report that intracerebroventricular (icv) infusion of leptin reverses lethality and greatly improves hyperglycemia, hyperglucagonemia, hyperketonemia, and polyuria caused by insulin deficiency in mice. Notably, icv leptin administration leads to increased body weight while suppressing food intake, thus correcting the catabolic consequences of T1D. Also, icv leptin delivery improves expression of the metabolically relevant hypothalamic neuropeptides proopiomelanocortin, neuropeptide Y, and agouti-related peptide in T1D mice. Furthermore, this treatment normalizes phosphoenolpyruvate carboxykinase 1 contents without affecting glycogen levels in the liver. Pancreatic ß-cell regeneration does not underlie these beneficial effects of leptin, because circulating insulin levels were undetectable at basal levels and following a glucose overload. Also, pancreatic preproinsulin mRNA was completely absent in these icv leptin-treated T1D mice. Furthermore, the antidiabetic effects of icv leptin administration rapidly vanished (i.e., within 48 h) after leptin treatment was interrupted. Collectively, these results unveil a key role for the brain in mediating the antidiabetic actions of leptin in the context of T1D.


Asunto(s)
Sistema Nervioso Central/fisiología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Leptina/uso terapéutico , Animales , Modelos Animales de Enfermedad , Humanos , Inyecciones Intraventriculares , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Leptina/farmacología , Masculino , Ratones , Músculo Esquelético/metabolismo , Páncreas/metabolismo , Placebos , Precursores de Proteínas/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA