Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Int J Mol Sci ; 22(13)2021 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-34199142

RESUMEN

Metabolism is the central engine of living organisms as it provides energy and building blocks for many essential components of each cell, which are required for specific functions in different tissues. Mitochondria are the main site for energy production in living organisms and they also provide intermediate metabolites required for the synthesis of other biologically relevant molecules. Such cellular processes are finely tuned at different levels, including allosteric regulation, posttranslational modifications, and transcription of genes encoding key proteins in metabolic pathways. Peroxisome proliferator activated receptor γ coactivator 1 (PGC1) proteins are transcriptional coactivators involved in the regulation of many cellular processes, mostly ascribable to metabolic pathways. Here, we will discuss some aspects of the cellular processes regulated by PGC1s, bringing up some examples of their role in mitochondrial and cellular metabolism, and how metabolic regulation in mitochondria by members of the PGC1 family affects the immune system. We will analyze how PGC1 proteins are regulated at the transcriptional and posttranslational level and will also examine other regulators of mitochondrial metabolism and the related cellular functions, considering approaches to identify novel mitochondrial regulators and their role in physiology and disease. Finally, we will analyze possible therapeutical perspectives currently under assessment that are applicable to different disease states.


Asunto(s)
Metabolismo Energético , Mitocondrias/genética , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Animales , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunomodulación , Redes y Vías Metabólicas , Especificidad de Órganos , Termogénesis
2.
Int J Mol Sci ; 22(17)2021 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-34502211

RESUMEN

Obesity is a condition characterized by uncontrolled expansion of adipose tissue mass resulting in pathological weight gain. Histone deacetylases (HDACs) have emerged as crucial players in epigenetic regulation of adipocyte metabolism. Previously, we demonstrated that selective inhibition of class I HDACs improves white adipocyte functionality and promotes the browning phenotype of murine mesenchymal stem cells (MSCs) C3H/10T1/2 differentiated to adipocytes. These effects were also observed in db/db and diet induced obesity mouse models and in mice with adipose-selective inactivation of HDAC3, a member of class I HDACs. The molecular basis of class I HDACs action in adipose tissue is not deeply characterized and it is not known whether the effects of their inhibition are exerted on adipocyte precursors or mature adipocytes. Therefore, the aim of the present work was to explore the molecular mechanism of class I HDAC action in adipocytes by evaluating the effects of HDAC3-specific silencing at different stages of differentiation. HDAC3 was silenced in C3H/10T1/2 MSCs at different stages of differentiation to adipocytes. shRNA targeting HDAC3 was used to generate the knock-down model. Proper HDAC3 silencing was assessed by measuring both mRNA and protein levels of mouse HDAC3 via qPCR and western blot, respectively. Mitochondrial DNA content and gene expression were quantified via qPCR. HDAC3 silencing at the beginning of differentiation enhanced adipocyte functionality by amplifying the expression of genes regulating differentiation, oxidative metabolism, browning and mitochondrial activity, starting from 72 h after induction of differentiation and silencing. Insulin signaling was enhanced as demonstrated by increased AKT phosphorylation following HDAC3 silencing. Mitochondrial content/density did not change, while the increased expression of the transcriptional co-activator Ppargc1b suggests the observed phenotype was related to enhanced mitochondrial activity, which was confirmed by increased maximal respiration and proton leak linked to reduced coupling efficiency. Moreover, the expression of pro-inflammatory markers increased with HDAC3 early silencing. To the contrary, no differences in terms of gene expression were found when HDAC3 silencing occurred in terminally differentiated adipocyte. Our data demonstrated that early epigenetic events mediated by class I HDAC inhibition/silencing are crucial to commit adipocyte precursors towards the above-mentioned metabolic phenotype. Moreover, our data suggest that these effects are exerted on adipocyte precursors.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Diferenciación Celular , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Mitocondrias/metabolismo , Fenotipo , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Histona Desacetilasas/genética , Ratones , Ratones Endogámicos C3H
3.
EMBO Rep ; 19(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29507079

RESUMEN

Mitochondria are the energy-generating hubs of the cell. In spite of considerable advances, our understanding of the factors that regulate the molecular circuits that govern mitochondrial function remains incomplete. Using a genome-wide functional screen, we identify the poorly characterized protein Zinc finger CCCH-type containing 10 (Zc3h10) as regulator of mitochondrial physiology. We show that Zc3h10 is upregulated during physiological mitochondriogenesis as it occurs during the differentiation of myoblasts into myotubes. Zc3h10 overexpression boosts mitochondrial function and promotes myoblast differentiation, while the depletion of Zc3h10 results in impaired myoblast differentiation, mitochondrial dysfunction, reduced expression of electron transport chain (ETC) subunits, and blunted TCA cycle flux. Notably, we have identified a loss-of-function mutation of Zc3h10 in humans (Tyr105 to Cys105) that is associated with increased body mass index, fat mass, fasting glucose, and triglycerides. Isolated peripheral blood mononuclear cells from individuals homozygotic for Cys105 display reduced oxygen consumption rate, diminished expression of some ETC subunits, and decreased levels of some TCA cycle metabolites, which all together derive in mitochondrial dysfunction. Taken together, our study identifies Zc3h10 as a novel mitochondrial regulator.


Asunto(s)
Proteínas Portadoras/metabolismo , Mitocondrias/metabolismo , Anciano , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Línea Celular , Ciclo del Ácido Cítrico , Biología Computacional/métodos , Metabolismo Energético , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Masculino , Ratones , Mitocondrias/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Mutación , Mioblastos/citología , Mioblastos/metabolismo , Proteoma , Proteómica/métodos
4.
Int J Mol Sci ; 22(1)2020 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-33375647

RESUMEN

Metabolic disorders such as obesity and type 2 diabetes (T2D) are considered the major risk factors for the development of cardiovascular diseases (CVD). Although the pathological mechanisms underlying the mutual development of obesity and T2D are difficult to define, a better understanding of the molecular aspects is of utmost importance to identify novel therapeutic targets. Recently, a class of non-coding RNAs, called microRNAs (miRNAs), are emerging as key modulators of metabolic abnormalities. There is increasing evidence supporting the role of intra- and extracellular miRNAs as determinants of the crosstalk between adipose tissues, liver, skeletal muscle and other organs, triggering the paracrine communication among different tissues. miRNAs may be considered as risk factors for CVD due to their correlation with cardiovascular events, and in particular, may be related to the most prominent risk factors. In this review, we describe the associations observed between miRNAs expression levels and the most common cardiovascular risk factors. Furthermore, we sought to depict the molecular aspect of the interplay between obesity and diabetes, investigating the role of microRNAs in the interorgan crosstalk. Finally, we discussed the fascinating hypothesis of the loss of protective factors, such as antioxidant defense systems regulated by such miRNAs.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , MicroARNs/genética , Obesidad/etiología , Interferencia de ARN , Adipogénesis/genética , Tejido Adiposo/metabolismo , Animales , Antioxidantes/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Perfilación de la Expresión Génica , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Obesidad/metabolismo
5.
Biochim Biophys Acta ; 1851(1): 51-60, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25150974

RESUMEN

Lipids in the nervous system accomplish a great number of key functions, from synaptogenesis to impulse conduction, and more. Most of the lipids of the nervous system are localized in myelin sheaths. It has long been known that myelin structure and brain homeostasis rely on specific lipid-protein interactions and on specific cell-to-cell signaling. In more recent years, the growing advances in large-scale technologies and genetically modified animal models have provided valuable insights into the role of lipids in the nervous system. Key findings recently emerged in these areas are here summarized. In addition, we briefly discuss how this new knowledge can open novel approaches for the treatment of diseases associated with alteration of lipid metabolism/homeostasis in the nervous system. This article is part of a Special Issue entitled Linking transcription to physiology in lipidomics.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Sistema Nervioso/metabolismo , Sistema Nervioso/fisiopatología , Animales , Humanos
6.
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
7.
IUBMB Life ; 66(2): 89-99, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24488813

RESUMEN

After the completion of the human genome sequence and that from many other organisms, last decade has witnessed a spectacular gain of knowledge on gene functions. These studies provided new insights on the roles of genes in physiology and disease. Nonetheless, the availability of genetically modified models and of "omics" technologies such as next generation sequencing unveiled clear evidences on epigenetic regulation of many cellular functions. At this regard, sirtuins, belonging to class III histone deacetylase family, have emerged as regulators of metabolism as well as other cellular processes and seem ideally suited as targets of future therapeutical interventions. This review deals on general aspects of the biology of sirtuins and focuses on their relevance in lipid metabolism in different tissues, pointing to their exploitation as potential pharmacological targets of compounds that could be used as new therapeutic alternatives in several disorders ranging from type 2 diabetes and obesity to age-related cardiovascular and neurodegenerative diseases.


Asunto(s)
Histona Desacetilasas/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Sirtuinas/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Metabolismo Energético , Histona Desacetilasas/química , Histona Desacetilasas/clasificación , Humanos , Hígado/patología , Terapia Molecular Dirigida , Obesidad/metabolismo , Obesidad/patología , Conformación Proteica , Sirtuinas/química , Sirtuinas/clasificación
8.
Curr Genomics ; 15(6): 436-56, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25646072

RESUMEN

Energy metabolism and mitochondrial function hold a core position in cellular homeostasis. Oxidative metabolism is regulated at multiple levels, ranging from gene transcription to allosteric modulation. To accomplish the fine tuning of these multiple regulatory circuits, the nuclear and mitochondrial compartments are tightly and reciprocally controlled. The fact that nuclear encoded factors, PPARγ coactivator 1α and mitochondrial transcription factor A, play pivotal roles in the regulation of oxidative metabolism and mitochondrial biogenesis is paradigmatic of this crosstalk. Here we provide an updated survey of the genetic and epigenetic mechanisms involved in the control of energy metabolism and mitochondrial function. Chromatin dynamics highly depends on post-translational modifications occurring at specific amino acids in histone proteins and other factors associated to nuclear DNA. In addition to the well characterized enzymes responsible for histone methylation/demethylation and acetylation/deacetylation, other factors have gone on the "metabolic stage". This is the case of the new class of α-ketoglutarate-regulated demethylases (Jumonji C domain containing demethylases) and of the NAD+-dependent deacetylases, also known as sirtuins. Moreover, unexpected features of the machineries involved in mitochondrial DNA (mtDNA) replication and transcription, mitochondrial RNA processing and maturation have recently emerged. Mutations or defects of any component of these machineries profoundly affect mitochondrial activity and oxidative metabolism. Finally, recent evidences support the importance of mtDNA packaging in replication and transcription. These observations, along with the discovery that non-classical CpG islands present in mtDNA undergo methylation, indicate that epigenetics also plays a role in the regulation of the mitochondrial genome function.

9.
Mol Metab ; 79: 101862, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38141849

RESUMEN

BACKGROUND AND OBJECTIVES: Since white adipose tissue (WAT) lacks parasympathetic cholinergic innervation, the source of the acetylcholine (ACh) acting on white adipocyte cholinergic receptors is unknown. This study was designed to identify ACh-producing cells in mouse and human visceral WAT and to determine whether a non-neuronal cholinergic system becomes activated in obese inflamed WAT. METHODS: Mouse epididymal WAT (eWAT) and human omental fat were studied in normal and obese subjects. The expression of the key molecules involved in cholinergic signaling was evaluated by qRT-PCR and western blotting whereas their tissue distribution and cellular localization were investigated by immunohistochemistry, confocal microscopy and in situ hybridization. ACh levels were measured by liquid chromatography/tandem mass spectrometry. The cellular effects of ACh were assessed in cultured human multipotent adipose-derived stem cell (hMADS) adipocytes. RESULTS: In mouse eWAT, diet-induced obesity modulated the expression of key cholinergic molecular components and, especially, raised the expression of choline acetyltransferase (ChAT), the ACh-synthesizing enzyme, which was chiefly detected in interstitial macrophages, in macrophages forming crown-like structures (CLSs), and in multinucleated giant cells (MGCs). The stromal vascular fraction of obese mouse eWAT contained significantly higher ACh and choline levels than that of control mice. ChAT was undetectable in omental fat from healthy subjects, whereas it was expressed in a number of interstitial macrophages, CLSs, and MGCs from some obese individuals. In hMADS adipocytes stressed with tumor necrosis factor α, ACh, alone or combined with rivastigmine, significantly blunted monocyte chemoattractant protein 1 and interleukin 6 expression, it partially but significantly, restored adiponectin and GLUT4 expression, and promoted glucose uptake. CONCLUSIONS: In mouse and human visceral WAT, obesity induces activation of a macrophage-dependent non-neuronal cholinergic system that is capable of exerting anti-inflammatory and insulin-sensitizing effects on white adipocytes.


Asunto(s)
Tejido Adiposo Blanco , Sistema Colinérgico no Neuronal , Humanos , Ratones , Animales , Ratones Obesos , Tejido Adiposo Blanco/metabolismo , Obesidad/metabolismo , Colinérgicos/metabolismo
10.
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
11.
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
12.
J Lipid Res ; 53(2): 300-10, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22158827

RESUMEN

Diabetic peripheral neuropathy (DPN) is characterized by myelin abnormalities; however, the molecular mechanisms underlying such deficits remain obscure. To uncover the effects of diabetes on myelin alterations, we have analyzed myelin composition. In a streptozotocin-treated rat model of diabetic neuropathy, analysis of sciatic nerve myelin lipids revealed that diabetes alters myelin's phospholipid, FA, and cholesterol content in a pattern that can modify membrane fluidity. Reduced expression of relevant genes in the FA biosynthetic pathway and decreased levels of the transcriptionally active form of the lipogenic factor sterol-regulatory element binding factor-1c (SREBF-1c) were found in diabetic sciatic nerve. Expression of myelin's major protein, myelin protein zero (P0), was also suppressed by diabetes. In addition, we confirmed that diabetes induces sciatic nerve myelin abnormalities, primarily infoldings that have previously been associated with altered membrane fluidity. In a diabetic setting, synthetic activator of the nuclear receptor liver X receptor (LXR) increased SREBF-1c function and restored myelin lipid species and P0 expression levels to normal. These LXR-modulated improvements were associated with restored myelin structure in sciatic nerve and enhanced performance in functional tests such as thermal nociceptive threshold and nerve conduction velocity. These findings demonstrate an important role for the LXR-SREBF-1c axis in protection from diabetes-induced myelin abnormalities.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Vaina de Mielina/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Animales , Colesterol/metabolismo , Diabetes Mellitus Experimental/genética , Regulación de la Expresión Génica , Lípidos/química , Receptores X del Hígado , Masculino , Proteína P0 de la Mielina/metabolismo , Vaina de Mielina/química , Fosfolípidos/metabolismo , Proteínas Quinasas/genética , Ratas , Ratas Sprague-Dawley , Nervio Ciático/patología , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Estreptozocina
14.
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
15.
Sci Rep ; 11(1): 13310, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172806

RESUMEN

The use of cell-free scaffolds for the regeneration of clinically relevant volumes of soft tissue has been challenged, particularly in the case of synthetic biomaterials, by the difficulty of reconciling the manufacturing and biological performance requirements. Here, we investigated in vivo the importance of biomechanical and biochemical cues for conditioning the 3D regenerative microenvironment towards soft tissue formation. In particular, we evaluated the adipogenesis changes related to 3D mechanical properties by creating a gradient of 3D microenvironments with different stiffnesses using 3D Poly(Urethane-Ester-ether) PUEt scaffolds. Our results showed a significant increase in adipose tissue proportions while decreasing the stiffness of the 3D mechanical microenvironment. This mechanical conditioning effect was also compared with biochemical manipulation by loading extracellular matrices (ECMs) with a PPAR-γ activating molecule. Notably, results showed mechanical and biochemical conditioning equivalency in promoting adipose tissue formation in the conditions tested, suggesting that adequate mechanical signaling could be sufficient to boost adipogenesis by influencing tissue remodeling. Overall, this work could open a new avenue in the design of synthetic 3D scaffolds for microenvironment conditioning towards the regeneration of large volumes of soft and adipose tissue, with practical and direct implications in reconstructive and cosmetic surgery.


Asunto(s)
Microambiente Celular/fisiología , Regeneración/fisiología , Células 3T3-L1 , Adipogénesis/fisiología , Tejido Adiposo/fisiología , Animales , Línea Celular , Matriz Extracelular/fisiología , Ratones , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Cicatrización de Heridas/fisiología
16.
J Cell Biol ; 220(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33566069

RESUMEN

The commitment of mesenchymal stem cells to preadipocytes is stimulated by hormonal induction. Preadipocytes induced to differentiate repress protein synthesis, remodel their cytoskeleton, and increase mitochondrial function to support anabolic pathways. These changes enable differentiation into mature adipocytes. Our understanding of the factors that coordinately regulate the early events of adipocyte differentiation remains incomplete. Here, by using multipronged approaches, we have identified zinc finger CCCH-type containing 10 (Zc3h10) as a critical regulator of the early stages of adipogenesis. Zc3h10 depletion in preadipocytes resulted in increased protein translation and impaired filamentous (F)-actin remodeling, with the latter detrimental effect leading to mitochondrial and metabolic dysfunction. These defects negatively affected differentiation to mature adipocytes. In contrast, Zc3h10 overexpression yielded mature adipocytes with remarkably increased lipid droplet size. Overall, our study establishes Zc3h10 as a fundamental proadipogenic transcription factor that represses protein synthesis and promotes F-actin/mitochondria dynamics to ensure proper energy metabolism and favor lipid accumulation.


Asunto(s)
Actinas/metabolismo , Adipogénesis , Mitocondrias/metabolismo , Biosíntesis de Proteínas , Proteínas de Unión al ARN/metabolismo , Citoesqueleto de Actina/metabolismo , Adipocitos/metabolismo , Adipogénesis/genética , Tejido Adiposo Blanco/metabolismo , Animales , Línea Celular , Ciclo del Ácido Cítrico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Metabolismo Energético/genética , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Masculino , Metaboloma , Ratones Endogámicos C57BL , Dinámicas Mitocondriales , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Transcriptoma/genética , Proteínas de Unión al GTP rho/metabolismo
17.
Cells ; 9(11)2020 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-33233365

RESUMEN

Mitochondria represent the energy hub of cells and their function is under the constant influence of their tethering with other subcellular organelles. Mitochondria interact with the endoplasmic reticulum, lysosomes, cytoskeleton, peroxisomes, and nucleus in several ways, ranging from signal transduction, vesicle transport, and membrane contact sites, to regulate energy metabolism, biosynthetic processes, apoptosis, and cell turnover. Tumorigenesis is often associated with mitochondrial dysfunction, which could likely be the result of an altered interaction with different cell organelles or structures. The purpose of the present review is to provide an updated overview of the links between inter-organellar communications and interactions and metabolism in cancer cells, with a focus on mitochondria. The very recent publication of several reviews on these aspects testifies the great interest in the area. Here, we aim at (1) summarizing recent evidence supporting that the metabolic rewiring and adaptation observed in tumors deeply affect organelle dynamics and cellular functions and vice versa; (2) discussing insights on the underlying mechanisms, when available; and (3) critically presenting the gaps in the field that need to be filled, for a comprehensive understanding of tumor cells' biology. Chemo-resistance and druggable vulnerabilities of cancer cells related to the aspects mentioned above is also outlined.


Asunto(s)
Mitocondrias/metabolismo , Neoplasias/metabolismo , Orgánulos/metabolismo , Apoptosis , Carcinogénesis , Supervivencia Celular , Humanos
18.
Artículo en Inglés | MEDLINE | ID: mdl-31904421

RESUMEN

Obesity is characterized by uncontrolled expansion of adipose tissue mass, resulting in adipocyte hypertrophy (increased adipocyte size) and hyperplasia (increased number of adipocytes). The number of adipose cells is directly related to adipocyte differentiation process from stromal vascular cells to mature adipocytes. It is known that epigenetic factors influence adipose differentiation program. However, how specific epigenome modifiers affect white adipocyte differentiation and metabolic phenotype is still matter of research. Here, we provide evidence that class I histone deacetylases (HDACs) are involved both in the differentiation of adipocytes and in determining the metabolic features of these cells. We demonstrate that inhibition of class I HDACs from the very first stage of differentiation amplifies the differentiation process and imprints cells toward a highly oxidative phenotype. These effects are related to the capacity of the inhibitor to modulate H3K27 acetylation on enhancer regions regulating Pparg and Ucp1 genes. These epigenomic modifications result in improved white adipocyte functionality and metabolism and induce browning. Collectively, our results show that modulation of class I HDAC activity regulates the metabolic phenotype of white adipocytes via epigenetic imprinting on a key histone mark.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Animales , Línea Celular , Histona Desacetilasas/genética , Humanos , Ratones , Obesidad/tratamiento farmacológico , Obesidad/genética , Obesidad/metabolismo , Estrés Oxidativo/efectos de los fármacos
19.
Animals (Basel) ; 10(8)2020 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32731342

RESUMEN

The aim of this study was to evaluate the effects of mannan oligosaccharides (MOS) on gut health and performance in post-weaning piglets. In total, 40 piglets were divided into two experimental groups and fed a basal diet with (TRT) or without (CON) 0.2% mannan oligosaccharides for 35 days. Growth performance was determined weekly and faecal microbial composition on days 0, 14 and 35. On day 36, histometrical evaluations were performed on duodenal, jejunal, ileal, and colon samples. mRNA gene expression of inflammation-related genes was evaluated in samples of ileal Peyer's patches (IPP). MOS administration improved feed efficiency in the last two weeks of the trial (p < 0.05), and a decreased clostridia content was found in faeces at day 14 (p = 0.05). TRT piglets showed increased duodenal villi height (p < 0.05), and reduced mRNA levels of Tumour Necrosis Factor α (p < 0.05) and Toll-Like Receptor 4 (p < 0.01) in IPP. Our results suggest beneficial effects of MOS supplementation on gut morphology and the expression of inflammation-related genes in post-weaning piglets, accompanied by increased feed efficiency.

20.
FEBS Lett ; 593(22): 3173-3189, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31432511

RESUMEN

The metabolic transition from anaerobic glycolysis and fatty acid ß-oxidation to glycolysis coupled to oxidative phosphorylation is a key process for the transition of quiescent neural stem cells to proliferative neural progenitor cells. However, a full characterization of the metabolic shift and the involvement of mitochondria during the last step of neurogenesis, from neuroblasts to neuron maturation, is still elusive. Here, we describe a model of neuroblasts, Neuro2a cells, with impaired differentiation capacity due to mitochondrial dysfunction. Using a detailed biochemical characterization consisting of steady-state metabolomics and metabolic flux analysis, we find increased fatty acid ß-oxidation as a peculiar feature of neuroblasts with altered mitochondria. The consequent metabolic switch favors neuroblast proliferation at the expense of neuron maturation.


Asunto(s)
Ácidos Grasos/metabolismo , Mitocondrias/metabolismo , Células-Madre Neurales/citología , Línea Celular , Proliferación Celular , Metabolismo Energético , Humanos , Metabolómica , Modelos Biológicos , Células-Madre Neurales/metabolismo , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA