Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
EMBO J ; 39(9): e102808, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32154941

ABSTRACT

Defects in transcriptional regulators of pancreatic exocrine differentiation have been implicated in pancreatic tumorigenesis, but the molecular mechanisms are poorly understood. The locus encoding the transcription factor HNF1A harbors susceptibility variants for pancreatic ductal adenocarcinoma (PDAC), while KDM6A, encoding Lysine-specific demethylase 6A, carries somatic mutations in PDAC. Here, we show that pancreas-specific Hnf1a null mutant transcriptomes phenocopy those of Kdm6a mutations, and both defects synergize with KrasG12D to cause PDAC with sarcomatoid features. We combine genetic, epigenomic, and biochemical studies to show that HNF1A recruits KDM6A to genomic binding sites in pancreatic acinar cells. This remodels the acinar enhancer landscape, activates differentiated acinar cell programs, and indirectly suppresses oncogenic and epithelial-mesenchymal transition genes. We also identify a subset of non-classical PDAC samples that exhibit the HNF1A/KDM6A-deficient molecular phenotype. These findings provide direct genetic evidence that HNF1A deficiency promotes PDAC. They also connect the tumor-suppressive role of KDM6A deficiency with a cell-specific molecular mechanism that underlies PDAC subtype definition.


Subject(s)
Acinar Cells/metabolism , Carcinoma, Pancreatic Ductal/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Histone Demethylases/genetics , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Hepatocyte Nuclear Factor 1-alpha/metabolism , Histone Demethylases/metabolism , Humans , Mice , Mutation , Organ Specificity , Pancreas/metabolism , Pancreatic Neoplasms/metabolism
2.
PLoS Biol ; 16(7): e2004455, 2018 07.
Article in English | MEDLINE | ID: mdl-29979672

ABSTRACT

Adipose tissue has emerged as an important regulator of whole-body metabolism, and its capacity to dissipate energy in the form of heat has acquired a special relevance in recent years as potential treatment for obesity. In this context, the p38MAPK pathway has arisen as a key player in the thermogenic program because it is required for the activation of brown adipose tissue (BAT) thermogenesis and participates also in the transformation of white adipose tissue (WAT) into BAT-like depot called beige/brite tissue. Here, using mice that are deficient in p38α specifically in adipose tissue (p38αFab-KO), we unexpectedly found that lack of p38α protected against high-fat diet (HFD)-induced obesity. We also showed that p38αFab-KO mice presented higher energy expenditure due to increased BAT thermogenesis. Mechanistically, we found that lack of p38α resulted in the activation of the related protein kinase family member p38δ. Our results showed that p38δ is activated in BAT by cold exposure, and lack of this kinase specifically in adipose tissue (p38δ Fab-KO) resulted in overweight together with reduced energy expenditure and lower body and skin surface temperature in the BAT region. These observations indicate that p38α probably blocks BAT thermogenesis through p38δ inhibition. Consistent with the results obtained in animals, p38α was reduced in visceral and subcutaneous adipose tissue of subjects with obesity and was inversely correlated with body mass index (BMI). Altogether, we have elucidated a mechanism implicated in physiological BAT activation that has potential clinical implications for the treatment of obesity and related diseases such as diabetes.


Subject(s)
Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/physiology , Mitogen-Activated Protein Kinase 13/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Thermogenesis , Adipocytes, Brown/enzymology , Adult , Animals , Body Mass Index , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/prevention & control , Diet , Energy Metabolism , Enzyme Activation , Humans , MAP Kinase Signaling System , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 13/metabolism , Models, Biological , Obesity/enzymology , Obesity/prevention & control , Uncoupling Protein 1/metabolism
3.
EMBO J ; 35(5): 536-52, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26843485

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a major health problem and the main cause of liver disease in Western countries. Although NAFLD is strongly associated with obesity and insulin resistance, its pathogenesis remains poorly understood. The disease begins with an excessive accumulation of triglycerides in the liver, which stimulates an inflammatory response. Alternative p38 mitogen-activated kinases (p38γ and p38δ) have been shown to contribute to inflammation in different diseases. Here we demonstrate that p38δ is elevated in livers of obese patients with NAFLD and that mice lacking p38γ/δ in myeloid cells are resistant to diet-induced fatty liver, hepatic triglyceride accumulation and glucose intolerance. This protective effect is due to defective migration of p38γ/δ-deficient neutrophils to the damaged liver. We further show that neutrophil infiltration in wild-type mice contributes to steatosis development by means of inflammation and liver metabolic changes. Therefore, p38γ and p38δ in myeloid cells provide a potential target for NAFLD therapy.


Subject(s)
Liver/metabolism , Mitogen-Activated Protein Kinase 12/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Neutrophil Infiltration , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Adult , Aged , Animals , Female , Glucose Intolerance , Humans , Male , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/immunology , Mitogen-Activated Protein Kinase 13/genetics , Mitogen-Activated Protein Kinase 13/immunology , Non-alcoholic Fatty Liver Disease/immunology , Obesity/immunology , RNA, Messenger/metabolism , Triglycerides/metabolism
4.
Alcohol Clin Exp Res ; 39(10): 1939-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26365275

ABSTRACT

BACKGROUND: Alcohol binge drinking is one of the most common patterns of excessive alcohol use and recent data would suggest that histone deacetylases (HDACs) gene expression profiling could be useful as a biomarker for psychiatric disorders. METHODS: This study aimed to characterize the gene expression patterns of Hdac 1-11 in samples of rat peripheral blood, liver, heart, prefrontal cortex, and amygdala following repeated binge alcohol consumption and to determine the parallelism of Hdac gene expression between rats and humans in peripheral blood. To accomplish this goal, we examined Hdac gene expression following 1, 4, or 8 alcohol binges (3 g/kg, orally) in the rat, in patients who were admitted to the hospital emergency department for acute alcohol intoxication, and in rats trained in daily operant alcohol self-administration. RESULTS: We primarily found that acute alcohol binging reduced gene expression (Hdac1-10) in the peripheral blood of alcohol-naïve rats and that this effect was attenuated following repeated alcohol binges. There was also a reduction of Hdac gene expression in the liver (Hdac2,4,5), whereas there was increased expression in the heart (Hdac1,7,8) and amygdala (Hdac1,2,5). Additionally, increased blood alcohol concentrations were measured in rat blood at 1 to 4 hours following repeated alcohol binging, and the only group that developed hepatic steotosis (fatty liver) were those animals exposed to 8 alcohol binge events. Finally, both binge consumption of alcohol in humans and daily operant alcohol self-administration in rats increased Hdac gene expression in peripheral blood. CONCLUSIONS: Our results suggest that increases in HDAC gene expression within the peripheral blood are associated with chronic alcohol consumption, whereas HDAC gene expression is reduced following initial exposure to alcohol.


Subject(s)
Binge Drinking/genetics , Ethanol/administration & dosage , Ethanol/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylases/genetics , Alcoholic Intoxication/blood , Alcoholic Intoxication/enzymology , Alcoholic Intoxication/genetics , Amygdala/drug effects , Amygdala/metabolism , Animals , Binge Drinking/blood , Binge Drinking/enzymology , Ethanol/blood , Fatty Liver/chemically induced , Female , Histone Deacetylases/blood , Humans , Liver/drug effects , Liver/metabolism , Male , Myocardium/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Self Administration , Young Adult
5.
Dev Cell ; 57(16): 1922-1936.e9, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35998583

ABSTRACT

Sequence variants in cis-acting enhancers are important for polygenic disease, but their role in Mendelian disease is poorly understood. Redundancy between enhancers that regulate the same gene is thought to mitigate the pathogenic impact of enhancer mutations. Recent findings, however, have shown that loss-of-function mutations in a single enhancer near PTF1A cause pancreas agenesis and neonatal diabetes. Using mouse and human genetic models, we show that this enhancer activates an entire PTF1A enhancer cluster in early pancreatic multipotent progenitors. This leading role, therefore, precludes functional redundancy. We further demonstrate that transient expression of PTF1A in multipotent progenitors sets in motion an epigenetic cascade that is required for duct and endocrine differentiation. These findings shed insights into the genome regulatory mechanisms that drive pancreas differentiation. Furthermore, they reveal an enhancer that acts as a regulatory master key and is thus vulnerable to pathogenic loss-of-function mutations.


Subject(s)
Diabetes Mellitus , Transcription Factors , Animals , Cell Differentiation/genetics , Diabetes Mellitus/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Humans , Infant, Newborn , Mice , Mutation/genetics , Pancreas/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
6.
J Gastrointest Surg ; 26(2): 286-297, 2022 02.
Article in English | MEDLINE | ID: mdl-34882294

ABSTRACT

BACKGROUND: The peroxisome proliferator-activated receptor (PPAR)-γ plays a key role in adipose tissue differentiation and fat metabolism. However, it is unclear which factors may regulate its expression and whether obese patients have changes in adipose tissue expression of PPAR-γor potential regulators such as miR-27. Thus, our aims were to analyze PPAR-γ and miR-27 expression in adipose tissue of obese patients, and to correlate their levels with clinical variables. SUBJECTS AND METHODS: We included 43 morbidly obese subjects who underwent sleeve gastrectomy (31 of them completed 1-year follow-up) and 19 non-obese subjects. mRNA expression of PPAR-γ1 and PPAR-γ2, miR-27a, and miR-27b was measured by qPCR in visceral and subcutaneous adipose tissue. Clinical variables and serum adipokine and hormone levels were correlated with PPAR-γ and miR-27 expression. In addition, a systematic review of the literature regarding PPAR-γ expression in adipose tissue of obese patients was performed. RESULTS: We found no differences in the expression of PPAR-γ and miR-27 in adipose tissue of obese patients vs. controls. The literature review revealed discrepant results regarding PPAR-γ expression in adipose tissue of obese patients. Of note, we described a significant negative correlation between pre-operative PPAR-γ1 expression in adipose tissue of obese patients and post-operative weight loss, potentially linked with insulin resistance markers. CONCLUSION: PPAR-γ1 expression in adipose tissue is associated with weight loss after sleeve gastrectomy and may be used as a biomarker for response to surgery.


Subject(s)
Adipose Tissue , Obesity, Morbid , Peroxisome Proliferator-Activated Receptors , Weight Loss , Adipose Tissue/metabolism , Gastrectomy , Gene Expression , Humans , MicroRNAs , Obesity, Morbid/genetics , Obesity, Morbid/surgery , PPAR gamma , Peroxisome Proliferator-Activated Receptors/metabolism
7.
Am J Respir Cell Mol Biol ; 45(6): 1133-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21642586

ABSTRACT

In addition to immune cells, airway epithelial cells can contribute to and shape the immune response in the lung by secreting specific cytokines. IL-6 is a key factor in determining the effector fate of CD4(+) T cells. Here we show that under basal conditions, the IL-6 gene is already highly expressed in lung epithelial cells, but not in immune cells resident in the lung. However, upon exposure of the lungs to fungal allergens, the direct contact of ß-glucans present in the fungus cell wall with lung epithelial cells is sufficient to trigger the rapid synthesis and secretion of IL-6 protein. This posttranscriptional regulation of IL-6 in response to fungal extracts is mediated by the p38 mitogen-activated protein kinase pathway. The inhalation of ß-glucans with a nonallergenic antigen is sufficient to provide an adjuvant effect that leads to mucous hyperplasia in the airways. Thus, ß-glucans may constitute a common determinant of the fungal and plant-derived allergens responsible for some of the pathological features in allergic asthma.


Subject(s)
Allergens/immunology , Aspergillus fumigatus/immunology , Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Gene Expression Regulation/immunology , Interleukin-6/immunology , Respiratory Mucosa/immunology , beta-Glucans/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Allergens/chemistry , Allergens/pharmacology , Animals , Aspergillus fumigatus/chemistry , Asthma/metabolism , Asthma/pathology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Interleukin-6/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , beta-Glucans/chemistry , beta-Glucans/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Front Cell Dev Biol ; 9: 708844, 2021.
Article in English | MEDLINE | ID: mdl-35111744

ABSTRACT

IL6 is an essential cytokine in metabolism regulation and for intercommunication among different organs and tissues. IL6 produced by different tissues has different functions and therefore it is very important to understand the mechanism of its expression in adipose tissue. In this work we demonstrated that IL6 expression in mouse preadipocytes, like in human, is partially dependent on Wnt5a and JNK. Using mouse preadipocytes lacking each one of the p38 SAPK family members, we have shown that IL6 expression is also p38γ and p38δ dependent. In fact, the lack of some of these two kinases increases IL6 expression without altering that of Wnt5a. Moreover, we show that the absence of p38δ promotes greater ERK1/2 phosphorylation in a MEK1/2 independent manner, and that this increased ERK1/2 phosphorylation state is contributing to the higher IL6 expression in p38δ-/- preadipocytes. These results suggest a new crosstalk between two MAPK signaling pathway, p38δ and ERK1/2, where p38δ modulates the phosphorylation state of ERK1/2.

9.
F1000Res ; 9: 1336, 2020.
Article in English | MEDLINE | ID: mdl-34745570

ABSTRACT

The COVID-19 pandemic has posed and is continuously posing enormous societal and health challenges worldwide. The research community has mobilized to develop novel projects to find a cure or a vaccine, as well as to contribute to mass testing, which has been a critical measure to contain the infection in several countries. Through this article, we share our experiences and learnings as a group of volunteers at the Centre for Genomic Regulation (CRG) in Barcelona, Spain. As members of the ORFEU project, an initiative by the Government of Catalonia to achieve mass testing of people at risk and contain the epidemic in Spain, we share our motivations, challenges and the key lessons learnt, which we feel will help better prepare the global society to address similar situations in the future.


Subject(s)
COVID-19 , COVID-19 Testing , Genomics , Humans , Pandemics , SARS-CoV-2 , Volunteers
10.
Nat Commun ; 8(1): 856, 2017 10 11.
Article in English | MEDLINE | ID: mdl-29021624

ABSTRACT

Increasing the thermogenic capacity of adipose tissue to enhance organismal energy expenditure is considered a promising therapeutic strategy to combat obesity. Here, we report that expression of the p38 MAPK activator MKK6 is elevated in white adipose tissue of obese individuals. Using knockout animals and shRNA, we show that Mkk6 deletion increases energy expenditure and thermogenic capacity of white adipose tissue, protecting mice against diet-induced obesity and the development of diabetes. Deletion of Mkk6 increases T3-stimulated UCP1 expression in adipocytes, thereby increasing their thermogenic capacity. Mechanistically, we demonstrate that, in white adipose tissue, p38 is activated by an alternative pathway involving AMPK, TAK, and TAB. Our results identify MKK6 in adipocytes as a potential therapeutic target to reduce obesity.Brown and beige adipose tissues dissipate heat via uncoupling protein 1 (UCP1). Here the authors show that the stress activated kinase MKK6 acts as a repressor of UCP1 expression, suggesting that its inhibition promotes adipose tissue browning and increases organismal energy expenditure.


Subject(s)
Adipose Tissue, White/enzymology , MAP Kinase Kinase 6/metabolism , Obesity/enzymology , Uncoupling Protein 1/metabolism , Adipocytes, White/metabolism , Adult , Aged , Animals , Case-Control Studies , Diabetes Mellitus/etiology , Diet, High-Fat , Energy Metabolism , Female , Humans , MAP Kinase Signaling System , Male , Metabolic Syndrome/etiology , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/etiology , Triiodothyronine/physiology , Ventromedial Hypothalamic Nucleus/metabolism
11.
Mol Cell Biol ; 33(11): 2302-14, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23530063

ABSTRACT

Mitochondria are the main engine that generates ATP through oxidative phosphorylation within the respiratory chain. Mitochondrial respiration is regulated according to the metabolic needs of cells and can be modulated in response to metabolic changes. Little is known about the mechanisms that regulate this process. Here, we identify MCJ/DnaJC15 as a distinct cochaperone that localizes at the mitochondrial inner membrane, where it interacts preferentially with complex I of the electron transfer chain. We show that MCJ impairs the formation of supercomplexes and functions as a negative regulator of the respiratory chain. The loss of MCJ leads to increased complex I activity, mitochondrial membrane potential, and ATP production. Although MCJ is dispensable for mitochondrial function under normal physiological conditions, MCJ deficiency affects the pathophysiology resulting from metabolic alterations. Thus, enhanced mitochondrial respiration in the absence of MCJ prevents the pathological accumulation of lipids in the liver in response to both fasting and a high-cholesterol diet. Impaired expression or loss of MCJ expression may therefore result in a "rapid" metabolism that mitigates the consequences of metabolic disorders.


Subject(s)
Electron Transport Complex I/metabolism , HSP40 Heat-Shock Proteins/genetics , Lipid Metabolism/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Respiration/genetics , Cholesterol/adverse effects , Diet , Electron Transport Complex I/genetics , Fatty Liver/genetics , Female , Gene Expression Regulation , Humans , Intracellular Membranes/metabolism , Male , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rotenone/pharmacology
12.
Fortaleza; s.n; 2016. 250 p. map, ilus, tab.
Thesis in Portuguese | LILACS | ID: biblio-971963

ABSTRACT

O desenvolvimento como forma de intervenção do capital constitui uma ideologia que se difunde nos marcos do colonialismo, ao longo dos tempos históricos. Assim, marca a relação Norte-Sul, redefinindo-se, hoje, na cooperação Sul-Sul, a constituir-se um dos eixos da nova ordem económica e política. Este trabalho vincula-se, fundamentalmente a uma discussão crítica da cooperação Sul-Sul. Neste sentido, considera, como um marco, na configuração geopolítica contemporânea, os BRICS, na condição de bloco constituído pelo Brasil, Rússia, Índia, China e África do Sul, efetivando novas formas de imperialismo que se constituem em nome da cooperação. Neste contexto de relações assimétricas Sul-Sul, esta dissertação problematiza a chamada cooperação dos países do Sul global, tendo como foco específico as relações Brasil-Moçambique, a partir da atuação da empresa multinacional Vale S.A que opera no território moçambicano, na extração de carvão mineral...


The development as a form of capital intervention is an ideology that is spreading in colonial landmarks over the historical times. This marks the North-South relationship, resetting itself today in South-South cooperation, to constitute one of the pillarsof the new economic and political order. This work is linked to fundamentally a critical discussion of South-South cooperation. Therefore considers, as a milestone in contemporary geopolitical configuration, the BRICS, in block condition comprising Brazil, Russia, India, China and South Africa, making effective new forms of imperialism which are in the name of the cooperation. In this context of asymmetric South-South, this dissertation problematizes the so-called cooperation of the global South countries,with the specific focus of the Brazil-Mozambique relations, from the activities of multinational Vale SA operating in Mozambique in coal mining mineral...


Subject(s)
Humans , Economic Development , Natural Resources Exploitation , Health , Environment , Work
SELECTION OF CITATIONS
SEARCH DETAIL