Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Obes (Lond) ; 48(4): 557-566, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38148333

RESUMEN

BACKGROUND: In obesity, adipose tissue undergoes a remodeling process characterized by increased adipocyte size (hypertrophia) and number (hyperplasia). The ability to tip the balance toward the hyperplastic growth, with recruitment of new fat cells through adipogenesis, seems to be critical for a healthy adipose tissue expansion, as opposed to a hypertrophic growth that is accompanied by the development of inflammation and metabolic dysfunction. However, the molecular mechanisms underlying the fine-tuned regulation of adipose tissue expansion are far from being understood. METHODS: We analyzed by mass spectrometry-based proteomics visceral white adipose tissue (vWAT) samples collected from C57BL6 mice fed with a HFD for 8 weeks. A subset of these mice, called low inflammation (Low-INFL), showed reduced adipose tissue inflammation, as opposed to those developing the expected inflammatory response (Hi-INFL). We identified the discriminants between Low-INFL and Hi-INFL vWAT samples and explored their function in Adipose-Derived human Mesenchymal Stem Cells (AD-hMSCs) differentiated to adipocytes. RESULTS: vWAT proteomics allowed us to quantify 6051 proteins. Among the candidates that most differentiate Low-INFL from Hi-INFL vWAT, we found proteins involved in adipocyte function, including adiponectin and hormone sensitive lipase, suggesting that adipocyte differentiation is enhanced in Low-INFL, as compared to Hi-INFL. The chromatin modifier SET and MYND Domain Containing 3 (SMYD3), whose function in adipose tissue was so far unknown, was another top-scored hit. SMYD3 expression was significantly higher in Low-INFL vWAT, as confirmed by western blot analysis. Using AD-hMSCs in culture, we found that SMYD3 mRNA and protein levels decrease rapidly during the adipocyte differentiation. Moreover, SMYD3 knock-down before adipocyte differentiation resulted in reduced H3K4me3 and decreased cell proliferation, thus limiting the number of cells available for adipogenesis. CONCLUSIONS: Our study describes an important role of SMYD3 as a newly discovered regulator of adipocyte precursor proliferation during the early steps of adipogenesis.


Asunto(s)
Adipocitos , Adipogénesis , Animales , Humanos , Ratones , Adipocitos/metabolismo , Adipogénesis/fisiología , Tejido Adiposo Blanco/metabolismo , Diferenciación Celular/genética , Proliferación Celular , N-Metiltransferasa de Histona-Lisina/metabolismo , Hipertrofia/metabolismo , Inflamación/metabolismo , Ratones Endogámicos C57BL , Obesidad
2.
Proteomics ; 23(3-4): e2200078, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36576318

RESUMEN

Abusive head trauma (AHT) is a leading cause of mortality and morbidity in infants. While the reported incidence is close to 40 cases per 100'000 births/year, misdiagnoses are commonly observed in cases with atypical, subacute, or chronic presentation. Currently, standard clinical evaluation of inflicted intracranial hemorrhagic injury (ICH) in infants urgently requires a screening test able to identify infants who need additional investigations. Blood biomarkers characteristic of AHT may assist in detecting these infants, improving prognosis through early medical care. To date, the application of innovative omics technologies in retrospective studies of AHT in infants is rare, due also to the blood serum and cerebrospinal fluid of AHT cases being scarce and not systematically accessible. Here, we explored the circulating blood proteomes of infants with severe AHT and their atraumatic controls. We discovered 165 circulating serum proteins that display differential changes in AHT cases compared with atraumatic controls. The peripheral blood proteomes of pediatric AHT commonly reflect: (i) potentially secreted proteome from injured brain, and (ii) proteome dysregulated in the system's circulation by successive biological events following acute ICH. This study opens up a novel opportunity for research efforts in clinical screening of AHT cases.


Asunto(s)
Maltrato a los Niños , Traumatismos Craneocerebrales , Humanos , Lactante , Niño , Proteoma , Estudios Retrospectivos , Maltrato a los Niños/diagnóstico , Traumatismos Craneocerebrales/diagnóstico , Traumatismos Craneocerebrales/epidemiología , Hemorragias Intracraneales/diagnóstico
3.
Chem Res Toxicol ; 35(5): 807-816, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35442019

RESUMEN

Cobalt, chromium, and nickel are used in orthopedic prostheses. They can be released, accumulate in many organs, and be toxic. The aim of this study is to evaluate the cytotoxicity of these metals on human hepatocytes and to improve our knowledge of their cellular toxicity mechanisms by metabolomic analysis. HepaRG cells were incubated for 48 h with increasing concentrations of metals to determine their IC50. Then, a nontargeted metabolomic study using liquid chromatography-high-resolution mass spectrometry (LC-HRMS) was done at IC50 and at a lower concentration (100 nM), near to those found in the blood and liver of patients with prostheses. IC50 were defined at 940, 2, and 1380 µM for Co, Cr, and Ni, respectively. In vitro, Cr appears to be much more toxic than Co and Ni. Metabolomic analysis revealed the disruption of metabolic pathways from the low concentration of 100 nM, in particular tryptophan metabolism and lipid metabolism illustrated by an increase in phenylacetylglycine, a marker of phospholipidosis, for all three metals. They also appear to be responsible for oxidative stress. Dysregulation of these pathways impacts hepatocyte metabolism and may result in hepatotoxicity. Further investigations on accessible biological matrices should be conducted to correlate our in vitro results with the clinical data of prostheses-bearing patients.


Asunto(s)
Cromo , Cobalto , Cromo/química , Cromo/toxicidad , Cobalto/toxicidad , Hepatocitos/química , Humanos , Metales , Níquel/toxicidad
4.
Cell Mol Life Sci ; 78(1): 227-247, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32157317

RESUMEN

Chronic inflammation that affects primarily metabolic organs, such as white adipose tissue (WAT), is considered as a major cause of human obesity-associated co-morbidities. However, the molecular mechanisms initiating this inflammation in WAT are poorly understood. By combining transcriptomics, ChIP-seq and modeling approaches, we studied the global early and late responses to a high-fat diet (HFD) in visceral (vWAT) and subcutaneous (scWAT) AT, the first being more prone to obesity-induced inflammation. HFD rapidly triggers proliferation of adipocyte precursors within vWAT. However, concomitant antiadipogenic signals limit vWAT hyperplastic expansion by interfering with the differentiation of proliferating adipocyte precursors. Conversely, in scWAT, residing beige adipocytes lose their oxidizing properties and allow storage of excessive fatty acids. This phase is followed by tissue hyperplastic growth and increased angiogenic signals, which further enable scWAT expansion without generating inflammation. Our data indicate that scWAT and vWAT differential ability to modulate adipocyte number and differentiation in response to obesogenic stimuli has a crucial impact on the different susceptibility to obesity-related inflammation of these adipose tissue depots.


Asunto(s)
Adipogénesis , Tejido Adiposo Blanco/metabolismo , Diferenciación Celular , Inflamación/patología , Obesidad/patología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/patología , Animales , Dieta Alta en Grasa , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Inflamación/etiología , Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Transducción de Señal/genética , Células Madre/citología , Células Madre/metabolismo , Grasa Subcutánea/citología , Grasa Subcutánea/metabolismo , Grasa Subcutánea/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Wnt/metabolismo
5.
BMC Public Health ; 22(1): 2446, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36577956

RESUMEN

BACKGROUND: According to the World Health Organization, road traffic injuries lead to 1.3 million deaths each year and represent the leading cause of death for young adults under 30 years old. The use of psychoactive substances, including alcohol, drugs and pharmaceuticals, is a well-known risk factor for road traffic injuries. Our study aims to assess the prevalence of substances consumed by drivers in western Switzerland. Such studies are pivotal to improving prevention and developing public awareness campaigns. METHODS: To assess the prevalence of psychoactive substances among drivers, roadside controls were performed in collaboration with local police, using their classical sampling procedures to detect drivers under the influence of drugs or alcohol over two time periods (P1: 2006-2008, P2: 2017-2020). When impaired driving was not suspected by the police, minimally invasive sampling strategies (i.e., oral fluids during P1 and dried blood spots during P2) were performed on volunteer drivers after a road safety survey. A posteriori analyses and statistical interpretation were then performed. RESULTS: Among the 1605 drivers included in the study, 1048 volunteers provided an oral fluid sample, while 299 provided a dried blood spot sample. The percentage of drivers testing positive for at least one substance that can impact driving abilities was stable over time, with a rate of 10.5% positivity measured over both periods. Considering the different categories of substances, a slight variation was observed between both periods, with 7.6 and 6.3% of pharmaceuticals and 3.6 and 4.9% of illicit drugs for P1 and P2, respectively. Regarding the consumption of illicit drugs, the highest percentage of positivity was measured in biological fluids of drivers under the age of 35, during nights and week-ends, periods which are considered particularly prone to fatal accidents for this age group. Disturbingly, the road safety survey highlighted that drivers' perception of the risk of getting positively controlled while driving after drug consumption is low (3.3 on a 1-to-10 scale, N = 299). CONCLUSION: The number of positive cases measured in voluntary drivers who passed the preliminary police check demonstrates the importance of systematic biofluid sampling strategies regarding driving under the influence of psychoactive substances. Although the number of fatal road accidents globally has decreased over time, the results of this study reveal the need for both better prevention and deterrent processes that could potentially reduce the risk of fatal road accidents associated with drug consumption.


Asunto(s)
Conducción de Automóvil , Drogas Ilícitas , Trastornos Relacionados con Sustancias , Adulto Joven , Humanos , Lactante , Adulto , Trastornos Relacionados con Sustancias/epidemiología , Prevalencia , Suiza/epidemiología , Detección de Abuso de Sustancias , Etanol , Accidentes de Tránsito
6.
Int J Mol Sci ; 22(18)2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34576136

RESUMEN

Men with nonalcoholic fatty liver disease (NAFLD) are more exposed to nonalcoholic steatohepatitis (NASH) and liver fibrosis than women. However, the underlying molecular mechanisms of NALFD sex dimorphism are unclear. We combined gene expression, histological and lipidomic analyses to systematically compare male and female liver steatosis. We characterized hepatosteatosis in three independent mouse models of NAFLD, ob/ob and lipodystrophic fat-specific (PpargFΔ/Δ) and whole-body PPARγ-null (PpargΔ/Δ) mice. We identified a clear sex dimorphism occurring only in PpargΔ/Δ mice, with females showing macro- and microvesicular hepatosteatosis throughout their entire life, while males had fewer lipid droplets starting from 20 weeks. This sex dimorphism in hepatosteatosis was lost in gonadectomized PpargΔ/Δ mice. Lipidomics revealed hepatic accumulation of short and highly saturated TGs in females, while TGs were enriched in long and unsaturated hydrocarbon chains in males. Strikingly, sex-biased genes were particularly perturbed in both sexes, affecting lipid metabolism, drug metabolism, inflammatory and cellular stress response pathways. Most importantly, we found that the expression of key sex-biased genes was severely affected in all the NAFLD models we tested. Thus, hepatosteatosis strongly affects hepatic sex-biased gene expression. With NAFLD increasing in prevalence, this emphasizes the urgent need to specifically address the consequences of this deregulation in humans.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/patología , PPAR gamma/deficiencia , Caracteres Sexuales , Animales , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Femenino , Regulación de la Expresión Génica , Hormonas Esteroides Gonadales/metabolismo , Inflamación/patología , Gotas Lipídicas/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , PPAR gamma/metabolismo , Fenotipo , Transducción de Señal , Triglicéridos/metabolismo
7.
PLoS Biol ; 15(4): e2001069, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28414715

RESUMEN

Many organisms exhibit temporal rhythms in gene expression that propel diurnal cycles in physiology. In the liver of mammals, these rhythms are controlled by transcription-translation feedback loops of the core circadian clock and by feeding-fasting cycles. To better understand the regulatory interplay between the circadian clock and feeding rhythms, we mapped DNase I hypersensitive sites (DHSs) in the mouse liver during a diurnal cycle. The intensity of DNase I cleavages cycled at a substantial fraction of all DHSs, suggesting that DHSs harbor regulatory elements that control rhythmic transcription. Using chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq), we found that hypersensitivity cycled in phase with RNA polymerase II (Pol II) loading and H3K27ac histone marks. We then combined the DHSs with temporal Pol II profiles in wild-type (WT) and Bmal1-/- livers to computationally identify transcription factors through which the core clock and feeding-fasting cycles control diurnal rhythms in transcription. While a similar number of mRNAs accumulated rhythmically in Bmal1-/- compared to WT livers, the amplitudes in Bmal1-/- were generally lower. The residual rhythms in Bmal1-/- reflected transcriptional regulators mediating feeding-fasting responses as well as responses to rhythmic systemic signals. Finally, the analysis of DNase I cuts at nucleotide resolution showed dynamically changing footprints consistent with dynamic binding of CLOCK:BMAL1 complexes. Structural modeling suggested that these footprints are driven by a transient heterotetramer binding configuration at peak activity. Together, our temporal DNase I mappings allowed us to decipher the global regulation of diurnal transcription rhythms in the mouse liver.


Asunto(s)
Ritmo Circadiano/genética , Regulación de la Expresión Génica , Hígado/fisiología , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Inmunoprecipitación de Cromatina , Relojes Circadianos/genética , Desoxirribonucleasa I/genética , Desoxirribonucleasa I/metabolismo , Ayuno , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Factores de Transcripción/genética , Transcripción Genética
8.
Am J Physiol Renal Physiol ; 314(6): F1154-F1165, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29488390

RESUMEN

Different complex mechanisms control the morphology of podocyte foot processes and their interactions with the underlying basement membrane. Injuries to this system often cause glomerular dysfunction and albuminuria. The present study aimed at identifying early markers of glomerular damage in diabetic nephropathy. For this purpose, we performed a microarray analysis on kidneys of 3-wk-old peroxisome proliferator-activated receptor-γ (PPARγ)-null and AZIP/F1 mice, which are two models of diabetic nephropathy due to lipodystrophy. This was followed by functional annotation of the enriched clusters of genes. One of the significant changes in the early stages of glomerular damage was the increase of hemicentin 1 (HMCN1). Its expression and distribution were then studied by real-time PCR and immunofluorescence in various models of glomerular damage and on podocyte cell cultures. HMCN1 progressively increased in the glomeruli of diabetic mice, according to disease severity, as well as in puromycin aminonucleoside (PA)-treated rats. Studies on murine and human podocytes showed an increased HMCN1 deposition upon different pathological stimuli, such as hyperglycemia, transforming growth factor-ß (TGF-ß), and PA. In vitro silencing studies showed that HMCN1 mediated the rearrangements of podocyte cytoskeleton induced by TGF-ß. Finally, we demonstrated an increased expression of HMCN1 in the kidneys of patients with proteinuric nephropathies. In summary, our studies identified HMCN1 as a new molecule involved in the dynamic changes of podocyte foot processes. Its increased expression associated with podocyte dysfunction points to HMCN1 as a possible marker for the early glomerular damage occurring in different proteinuric nephropathies.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Nefropatías Diabéticas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Inmunoglobulinas/metabolismo , Nefrosis/metabolismo , Podocitos/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Femenino , Glucosa/farmacología , Humanos , Inmunoglobulinas/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrosis/genética , Nefrosis/patología , PPAR gamma/genética , PPAR gamma/metabolismo , Podocitos/efectos de los fármacos , Podocitos/patología , Proteinuria/genética , Proteinuria/metabolismo , Proteinuria/patología , Ratas Sprague-Dawley , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba
9.
Int J Mol Sci ; 19(7)2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-30011852

RESUMEN

Skeletal muscle is a regenerative tissue which can repair damaged myofibers through the activation of tissue-resident muscle stem cells (MuSCs). Many muscle diseases with impaired regeneration cause excessive adipose tissue accumulation in muscle, alter the myogenic fate of MuSCs, and deregulate the cross-talk between MuSCs and fibro/adipogenic progenitors (FAPs), a bi-potent cell population which supports myogenesis and controls intra-muscular fibrosis and adipocyte formation. In order to better characterize the interaction between adipogenesis and myogenesis, we studied muscle regeneration and MuSC function in whole body Pparg null mice generated by epiblast-specific Cre/lox deletion (PpargΔ/Δ). We demonstrate that deletion of PPARγ completely abolishes ectopic muscle adipogenesis during regeneration and impairs MuSC expansion and myogenesis after injury. Ex vivo assays revealed that perturbed myogenesis in PpargΔ/Δ mice does not primarily result from intrinsic defects of MuSCs or from perturbed myogenic support from FAPs. The immune transition from a pro- to anti-inflammatory MuSC niche during regeneration is perturbed in PpargΔ/Δ mice and suggests that PPARγ signaling in macrophages can interact with ectopic adipogenesis and influence muscle regeneration. Altogether, our study demonstrates that a PPARγ-dependent adipogenic response regulates muscle fat infiltration during regeneration and that PPARγ is required for MuSC function and efficient muscle repair.


Asunto(s)
Adipogénesis/genética , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , PPAR gamma/genética , Regeneración/genética , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Mioblastos/citología , Mioblastos/metabolismo , PPAR gamma/metabolismo
10.
PLoS Genet ; 10(3): e1004155, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24603613

RESUMEN

In mammals, the circadian clock allows them to anticipate and adapt physiology around the 24 hours. Conversely, metabolism and food consumption regulate the internal clock, pointing the existence of an intricate relationship between nutrient state and circadian homeostasis that is far from being understood. The Sterol Regulatory Element Binding Protein 1 (SREBP1) is a key regulator of lipid homeostasis. Hepatic SREBP1 function is influenced by the nutrient-response cycle, but also by the circadian machinery. To systematically understand how the interplay of circadian clock and nutrient-driven rhythm regulates SREBP1 activity, we evaluated the genome-wide binding of SREBP1 to its targets throughout the day in C57BL/6 mice. The recruitment of SREBP1 to the DNA showed a highly circadian behaviour, with a maximum during the fed status. However, the temporal expression of SREBP1 targets was not always synchronized with its binding pattern. In particular, different expression phases were observed for SREBP1 target genes depending on their function, suggesting the involvement of other transcription factors in their regulation. Binding sites for Hepatocyte Nuclear Factor 4 (HNF4) were specifically enriched in the close proximity of SREBP1 peaks of genes, whose expression was shifted by about 8 hours with respect to SREBP1 binding. Thus, the cross-talk between hepatic HNF4 and SREBP1 may underlie the expression timing of this subgroup of SREBP1 targets. Interestingly, the proper temporal expression profile of these genes was dramatically changed in Bmal1-/- mice upon time-restricted feeding, for which a rhythmic, but slightly delayed, binding of SREBP1 was maintained. Collectively, our results show that besides the nutrient-driven regulation of SREBP1 nuclear translocation, a second layer of modulation of SREBP1 transcriptional activity, strongly dependent from the circadian clock, exists. This system allows us to fine tune the expression timing of SREBP1 target genes, thus helping to temporally separate the different physiological processes in which these genes are involved.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/genética , Metabolismo de los Lípidos/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Animales , Sitios de Unión , Proteínas CLOCK/genética , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Regulación de la Expresión Génica , Genoma , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Homeostasis , Ratones , Unión Proteica
11.
J Biol Chem ; 289(10): 6908-6920, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24451380

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors regulating lipid and glucose metabolism. Ongoing drug discovery programs aim to develop dual PPARα/γ agonists devoid of the side effects of the marketed antidiabetic agents thiazolidinediones and the dual agonists glitazars. Recently, we described a new dual PPARα/γ ligand, LT175, with a partial agonist profile against PPARγ and interacting with a newly identified region of the PPARγ-ligand binding domain (1). Here we show that LT175 differentially activated PPARγ target genes involved in fatty acid esterification and storage in 3T3-L1-derived adipocytes. This resulted in a less severe lipid accumulation compared with that triggered by rosiglitazone, suggesting that LT175 may have a lower adipogenic activity. Consistent with this hypothesis, in vivo administration of LT175 to mice fed a high-fat diet decreased body weight, adipocyte size, and white adipose tissue mass, as assessed by magnetic resonance imaging. Furthermore, LT175 significantly reduced plasma glucose, insulin, non-esterified fatty acids, triglycerides, and cholesterol and increased circulating adiponectin and fibroblast growth factor 21 levels. Oral glucose and insulin tolerance tests showed that the compound improves glucose homeostasis and insulin sensitivity. Moreover, we demonstrate that the peculiar interaction of LT175 with PPARγ affected the recruitment of the coregulators cyclic-AMP response element-binding protein-binding protein and nuclear corepressor 1 (NCoR1), fundamentals for the PPARγ-mediated adipogenic program. In conclusion, our results describe a new PPAR ligand, modulating lipid and glucose metabolism with reduced adipogenic activity, that may be used as a model for a series of novel molecules with an improved pharmacological profile for the treatment of dyslipidemia and type 2 diabetes.


Asunto(s)
Adipogénesis/efectos de los fármacos , Compuestos de Bifenilo/administración & dosificación , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Insulina/farmacología , PPAR alfa/agonistas , PPAR gamma/agonistas , Fenilpropionatos/administración & dosificación , Células 3T3-L1 , Animales , Compuestos de Bifenilo/metabolismo , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dislipidemias/tratamiento farmacológico , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/metabolismo , Insulina/sangre , Ligandos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Co-Represor 1 de Receptor Nuclear/metabolismo , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Fenilpropionatos/metabolismo
12.
Genome Res ; 22(4): 666-80, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22287103

RESUMEN

The genomic loci occupied by RNA polymerase (RNAP) III have been characterized in human culture cells by genome-wide chromatin immunoprecipitations, followed by deep sequencing (ChIP-seq). These studies have shown that only ∼40% of the annotated 622 human tRNA genes and pseudogenes are occupied by RNAP-III, and that these genes are often in open chromatin regions rich in active RNAP-II transcription units. We have used ChIP-seq to characterize RNAP-III-occupied loci in a differentiated tissue, the mouse liver. Our studies define the mouse liver RNAP-III-occupied loci including a conserved mammalian interspersed repeat (MIR) as a potential regulator of an RNAP-III subunit-encoding gene. They reveal that synteny relationships can be established between a number of human and mouse RNAP-III genes, and that the expression levels of these genes are significantly linked. They establish that variations within the A and B promoter boxes, as well as the strength of the terminator sequence, can strongly affect RNAP-III occupancy of tRNA genes. They reveal correlations with various genomic features that explain the observed variation of 81% of tRNA scores. In mouse liver, loci represented in the NCBI37/mm9 genome assembly that are clearly occupied by RNAP-III comprise 50 Rn5s (5S RNA) genes, 14 known non-tRNA RNAP-III genes, nine Rn4.5s (4.5S RNA) genes, and 29 SINEs. Moreover, out of the 433 annotated tRNA genes, half are occupied by RNAP-III. Transfer RNA gene expression levels reflect both an underlying genomic organization conserved in dividing human culture cells and resting mouse liver cells, and the particular promoter and terminator strengths of individual genes.


Asunto(s)
Perfilación de la Expresión Génica , Genómica/métodos , Hígado/metabolismo , ARN Polimerasa III/genética , Animales , Inmunoprecipitación de Cromatina/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Polimerasa III/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Análisis de Secuencia de ADN/métodos
13.
PLoS Biol ; 10(11): e1001442, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209382

RESUMEN

Interactions of cell-autonomous circadian oscillators with diurnal cycles govern the temporal compartmentalization of cell physiology in mammals. To understand the transcriptional and epigenetic basis of diurnal rhythms in mouse liver genome-wide, we generated temporal DNA occupancy profiles by RNA polymerase II (Pol II) as well as profiles of the histone modifications H3K4me3 and H3K36me3. We used these data to quantify the relationships of phases and amplitudes between different marks. We found that rhythmic Pol II recruitment at promoters rather than rhythmic transition from paused to productive elongation underlies diurnal gene transcription, a conclusion further supported by modeling. Moreover, Pol II occupancy preceded mRNA accumulation by 3 hours, consistent with mRNA half-lives. Both methylation marks showed that the epigenetic landscape is highly dynamic and globally remodeled during the 24-hour cycle. While promoters of transcribed genes had tri-methylated H3K4 even at their trough activity times, tri-methylation levels reached their peak, on average, 1 hour after Pol II. Meanwhile, rhythms in tri-methylation of H3K36 lagged transcription by 3 hours. Finally, modeling profiles of Pol II occupancy and mRNA accumulation identified three classes of genes: one showing rhythmicity both in transcriptional and mRNA accumulation, a second class with rhythmic transcription but flat mRNA levels, and a third with constant transcription but rhythmic mRNAs. The latter class emphasizes widespread temporally gated posttranscriptional regulation in the mouse liver.


Asunto(s)
Ritmo Circadiano , Epigénesis Genética , ARN Polimerasa II/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética , Animales , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Metilación de ADN , Semivida , Histonas/genética , Histonas/metabolismo , Cinética , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Sitio de Iniciación de la Transcripción , Transcriptoma
14.
Subcell Biochem ; 70: 75-102, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24962882

RESUMEN

Retinoid X Receptors (RXR) were initially identified as nuclear receptors binding with stereo-selectivity the vitamin A derivative 9-cis retinoic acid, although the relevance of this molecule as endogenous activator of RXRs is still elusive. Importantly, within the nuclear receptor superfamily, RXRs occupy a peculiar place, as they are obligatory partners for a number of other nuclear receptors, thus integrating the corresponding signaling pathways. In this chapter, we describe the structural features allowing RXR to form homo- and heterodimers, and the functional consequences of this unique ability. Furthermore, we discuss the importance of studying RXR activity at a genome-wide level in order to comprehensively address the biological implications of their action that is fundamental to understand to what extent RXRs could be exploited as new therapeutic targets.


Asunto(s)
Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide/metabolismo , Tretinoina/metabolismo , Alitretinoína , Animales , Regulación de la Expresión Génica , Humanos , Ligandos , Ratones , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Receptores de Ácido Retinoico/genética , Elementos de Respuesta , Receptores X Retinoide/genética , Transducción de Señal , Tretinoina/química
15.
Amino Acids ; 44(3): 1001-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23224824

RESUMEN

To investigate the influence of diet on serum protein pattern, mice were fed for 8 weeks either control chow or a high-fat diet (containing 21 % w/w milk fat and 0.2 % w/w cholesterol); sera were collected and analyzed by 2-DE. The main positive acute-phase reactant proteins, haptoglobin and hemopexin, were significantly up-regulated in animals receiving the high-fat diet. Data on all other proteins also pointed to an inflammatory condition in these animals. The largest change in concentration was observed for carboxylesterase N, a circulating enzyme seldom connected with lipid metabolism in earlier reports. These observations agree with the notion of a link between diet-induced hyperlipidemia and the inflammatory component of its cardiovascular sequels in humans, but the effects in the experimental animals are massive and obviously affect most of the major serum proteins.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Electroforesis en Gel Bidimensional , Humanos , Inflamación/etiología , Inflamación/genética , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteoma/química , Proteoma/genética , Proteoma/metabolismo , Suero/química , Suero/metabolismo
16.
Mol Membr Biol ; 29(7): 257-66, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23095054

RESUMEN

A number of recent studies revealed that epigenetic modifications play a central role in the regulation of lipid and of other metabolic pathways such as cholesterol homeostasis, bile acid synthesis, glucose and energy metabolism. Epigenetics refers to aspects of genome functions regulated in a DNA sequence-independent fashion. Chromatin structure is controlled by epigenetic mechanisms through DNA methylation and histone modifications. The main modifications are histone acetylation and deacetylation on specific lysine residues operated by two different classes of enzymes: Histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. The interaction between these enzymes and histones can activate or repress gene transcription: Histone acetylation opens and activates chromatin, while deacetylation of histones and DNA methylation compact chromatin making it transcriptionally silent. The new evidences on the importance of HDACs in the regulation of lipid and other metabolic pathways will open new perspectives in the comprehension of the pathophysiology of metabolic disorders.


Asunto(s)
Cromatina/metabolismo , Epigénesis Genética/fisiología , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Metabolismo de los Lípidos/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Acetilación , Animales , Cromatina/genética , Metilación de ADN/fisiología , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/genética , Histonas/genética , Humanos
17.
Toxicology ; 500: 153672, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37956786

RESUMEN

Human lifetime exposure to arsenic through drinking water, food supply or industrial pollution leads to its accumulation in many organs such as liver, kidneys, lungs or pancreas but also adipose tissue. Recently, population-based studies revealed the association between arsenic exposure and the development of metabolic diseases such as obesity and type 2 diabetes. To shed light on the molecular bases of such association, we determined the concentration that inhibited 17% of cell viability and investigated the effects of arsenic acute exposure on adipose-derived human mesenchymal stem cells differentiated in vitro into mature adipocytes and treated with sodium arsenite (NaAsO2, 10 nM to 10 µM). Untargeted metabolomics and gene expression analyses revealed a strong dose-dependent inhibition of lipogenesis and lipolysis induction, reducing the cellular ability to store lipids. These dysregulations were emphasized by the inhibition of the cellular response to insulin, as shown by the perturbation of several genes and metabolites involved in the mentioned biological pathways. Our study highlighted the activation of an adaptive oxidative stress response with the strong induction of metallothioneins and increased glutathione levels in response to arsenic accumulation that could exacerbate the decreased insulin sensitivity of the adipocytes. Arsenic exposure strongly affected the expression of arsenic transporters, responsible for arsenic influx and efflux, and induced a pro-inflammatory state in adipocytes by enhancing the expression of the inflammatory interleukin 6 (IL6). Collectively, our data showed that an acute exposure to low levels of arsenic concentrations alters key adipocyte functions, highlighting its contribution to the development of insulin resistance and the pathogenesis of metabolic disorders.


Asunto(s)
Arsénico , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Humanos , Arsénico/metabolismo , Tejido Adiposo/metabolismo , Adipocitos , Insulina/metabolismo , Metaboloma
18.
Toxicology ; 470: 153153, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35301059

RESUMEN

Obesity is considered as a major public health concern with strong economic and social burdens. Exposure to pollutants such as heavy metals can contribute to the development of obesity and its associated metabolic disorders, including type 2 diabetes and cardiovascular diseases. Adipose tissue is an endocrine and paracrine organ that plays a key role in the development of these diseases and is one of the main target of heavy metal accumulation. In this study, we determined by inductively coupled plasma mass spectrometry cadmium concentrations in human subcutaneous and visceral adipose tissues, ranging between 2.5 nM and 2.5 µM. We found a positive correlation between cadmium levels and age, sex and smoking status and a negative correlation between cadmium and body mass index. Based on cadmium adipose tissue concentrations found in humans, we investigated the effects of cadmium exposure, at concentrations between 1 nM and 10 µM, on adipose-derived human mesenchymal stem cells differentiated into mature adipocytes in vitro. Transcriptomic analysis highlighted that such exposure altered the expression of genes involved in trace element homeostasis and heavy metal detoxification, such as Solute Carrier Family transporters and metallothioneins. This effect correlated with zinc level alteration in cells and cellular media. Interestingly, dysregulation of zinc homeostasis and transporters has been particularly associated with the development of obesity and type 2 diabetes. Moreover, we found that cadmium exposure induces the pro-inflammatory state of the adipocytes by enhancing the expression of genes such as IL-6, IL-1B and CCL2, cytokines also induced in obesity. Finally, cadmium modulates various adipocyte functions such as the insulin response signaling pathway and lipid homeostasis. Collectively, our data identified some of the cellular mechanisms by which cadmium alters adipocyte functions, thus highlighting new facets of its potential contribution to the progression of metabolic disorders.


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedades Metabólicas , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Cadmio/toxicidad , Diabetes Mellitus Tipo 2/genética , Humanos , Insulina/metabolismo , Obesidad/inducido químicamente , Obesidad/genética , Transcriptoma , Zinc/metabolismo
19.
Eur J Med Chem ; 235: 114240, 2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35325635

RESUMEN

The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors involved in the regulation of the metabolic homeostasis and therefore represent valuable therapeutic targets for the treatment of metabolic diseases. The development of more balanced drugs interacting with PPARs, devoid of the side-effects showed by the currently marketed PPARγ full agonists, is considered the major challenge for the pharmaceutical companies. Here we present a chemoinformatics search approach for new ligands that let us identify a novel PPAR pan-agonist with a very attractive activity profile being able to reduce lipid accumulation and improve insulin sensitivity. This compound represents, therefore, the potential lead of a new class of drugs for treatment of dyslipidemic type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Quimioinformática , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Ligandos , Lípidos , PPAR gamma/agonistas , Receptores Activados del Proliferador del Peroxisoma/metabolismo
20.
Subcell Biochem ; 51: 109-35, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20213542

RESUMEN

Cholesterol and other cholesterol related metabolites, oxysterols, and bile acids, establish specific interactions with enzymes and other proteins involved in cholesterol and bile acid homeostasis, triggering a variety of biological responses. The substrate-enzyme binding represents the best-characterized type of complementary interaction between proteins and small molecules. Key enzymes in the pathway that converts cholesterol to bile acids belong to the cytochrome P450 superfamily. In contrast to the majority of P450 enzymes, those acting on cholesterol and related metabolites exhibit higher stringency with respect to substrate molecules. This stringency, coupled with the specificity of the reactions, dictates the chemical features of intermediate metabolites (oxysterols) and end products (bile acids). Both oxysterols and bile acids have emerged in recent years as new signalling molecules due to their ability to interact and activate nuclear receptors, and consequently to regulate the transcription of genes involved in cholesterol and bile acid homeostasis and metabolism, but also in glucose and fatty acid metabolism. Interestingly, other proteins function as bile acid or sterol receptors. New findings indicate that bile acids also interact with a membrane G protein-coupled receptor, triggering a signalling cascade that ultimately promote energy expenditure. On the other end, cholesterol and side chain oxysterols establish specific interactions with different proteins residing in the endoplasmic reticulum that result in controlled protein degradation and/or trafficking to the Golgi and the nucleus. These regulatory pathways converge and contribute to adapt cholesterol uptake and synthesis to the cellular needs.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Colesterol/biosíntesis , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Retículo Endoplásmico/metabolismo , Retroalimentación Fisiológica , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Receptores X del Hígado , Receptores Nucleares Huérfanos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Esteroles/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA