Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(8): 104999, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37394005

RESUMEN

Hepatocytes on exposure to high levels of lipids reorganize the metabolic program while fighting against the toxicity associated with elevated cellular lipids. The mechanism of this metabolic reorientation and stress management in lipid-challenged hepatocytes has not been well explored. We have noted the lowering of miR-122, a liver-specific miRNA, in the liver of mice fed with either a high-fat diet or a methionine-choline-deficient diet that is associated with increased fat accumulation in mice liver. Interestingly, low miR-122 levels are attributed to the enhanced extracellular export of miRNA processor enzyme Dicer1 from hepatocytes in the presence of high lipids. Export of Dicer1 can also account for the increased cellular levels of pre-miR-122-the substrate of Dicer1. Interestingly, restoration of Dicer1 levels in the mouse liver resulted in a strong inflammatory response and cell death in the presence of high lipids. Increasing death of hepatocytes was found to be caused by increased miR-122 levels in hepatocytes restored for Dicer1. Thus, the Dicer1 export by hepatocytes seems to be a key mechanism to combat lipotoxic stress by shunting out miR-122 from stressed hepatocytes. Finally, as part of this stress management, we determined that the Ago2-interacting pool of Dicer1, responsible for mature microribonucleoprotein formation in mammalian cells, gets depleted. miRNA-binder and exporter protein HuR is found to accelerate Ago2-Dicer1 uncoupling to ensure export of Dicer1 via extracellular vesicles in lipid-loaded hepatocytes.


Asunto(s)
MicroARNs , Animales , Ratones , Muerte Celular , ARN Helicasas DEAD-box/metabolismo , Dieta Alta en Grasa , Hepatocitos/metabolismo , Lípidos , Mamíferos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Ribonucleasa III/genética , Ratones Endogámicos C57BL , Humanos , Masculino , Línea Celular Tumoral
2.
Proc Natl Acad Sci U S A ; 117(12): 6509-6520, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32152128

RESUMEN

Among all of the Super Elongation Complex (SEC) components, ELL1 (also known as ELL) is the only bona fide elongation factor that directly stimulates transcription elongation by RNA polymerase II. However, the mechanism(s) of functional regulation of ELL1 (referred to as ELL hereafter), through its stabilization, is completely unknown. Here, we report a function of human DBC1 in regulating ELL stability involving HDAC3, p300, and Siah1. Mechanistically, we show that p300-mediated site-specific acetylation increases, whereas HDAC3-mediated deacetylation decreases, ELL stability through polyubiquitylation by the E3 ubiquitin ligase Siah1. DBC1 competes with HDAC3 for the same binding sites on ELL and thus increases its acetylation and stability. Knockdown of DBC1 reduces ELL levels and expression of a significant number of genes, including those involved in glucose metabolism. Consistently, Type 2 diabetes patient-derived peripheral blood mononuclear cells show reduced expression of DBC1 and ELL and associated key target genes required for glucose homeostasis. Thus, we describe a pathway of regulating stability and functions of key elongation factor ELL for expression of diverse sets of genes, including ones that are linked to Type 2 diabetes pathogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Elongación Transcripcional/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Acetilación , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sitios de Unión , Línea Celular , Diabetes Mellitus Tipo 2/patología , Proteína p300 Asociada a E1A/genética , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Histona Desacetilasas/genética , Humanos , Leucocitos Mononucleares/metabolismo , Mutación , Unión Proteica , Estabilidad Proteica , Transcripción Genética , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/genética , Ubiquitinación
3.
Int J Mol Sci ; 24(9)2023 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-37175791

RESUMEN

Pancreatic beta cell function is an important component of glucose homeostasis. Here, we investigated the function of PIMT (PRIP-interacting protein with methyl transferase domain), a transcriptional co-activator binding protein, in the pancreatic beta cells. We observed that the protein levels of PIMT, along with key beta cell markers such as PDX1 (pancreatic and duodenal homeobox 1) and MafA (MAF bZIP transcription factor A), were reduced in the beta cells exposed to hyperglycemic and hyperlipidemic conditions. Consistently, PIMT levels were reduced in the pancreatic islets isolated from high fat diet (HFD)-fed mice. The RNA sequencing analysis of PIMT knockdown beta cells identified that the expression of key genes involved in insulin secretory pathway, Ins1 (insulin 1), Ins2 (insulin 2), Kcnj11 (potassium inwardly-rectifying channel, subfamily J, member 11), Kcnn1 (potassium calcium-activated channel subfamily N member 1), Rab3a (member RAS oncogene family), Gnas (GNAS complex locus), Syt13 (synaptotagmin 13), Pax6 (paired box 6), Klf11 (Kruppel-Like Factor 11), and Nr4a1 (nuclear receptor subfamily 4, group A, member 1) was attenuated due to PIMT depletion. PIMT ablation in the pancreatic beta cells and in the rat pancreatic islets led to decreased protein levels of PDX1 and MafA, resulting in the reduction in glucose-stimulated insulin secretion (GSIS). The results from the immunoprecipitation and ChIP experiments revealed the interaction of PIMT with PDX1 and MafA, and its recruitment to the insulin promoter, respectively. Importantly, PIMT ablation in beta cells resulted in the nuclear translocation of insulin. Surprisingly, forced expression of PIMT in beta cells abrogated GSIS, while Ins1 and Ins2 transcript levels were subtly enhanced. On the other hand, the expression of genes, PRIP/Asc2/Ncoa6 (nuclear receptor coactivator 6), Pax6, Kcnj11, Syt13, Stxbp1 (syntaxin binding protein 1), and Snap25 (synaptosome associated protein 25) associated with insulin secretion, was significantly reduced, providing an explanation for the decreased GSIS upon PIMT overexpression. Our findings highlight the importance of PIMT in the regulation of insulin synthesis and secretion in beta cells.


Asunto(s)
Células Secretoras de Insulina , Insulina , Animales , Ratones , Ratas , Genes Homeobox , Glucosa/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Insulina/metabolismo , Insulina Regular Humana , Células Secretoras de Insulina/metabolismo , Potasio/metabolismo , Transactivadores/metabolismo , Histonas
4.
Biochem J ; 478(17): 3297-3317, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34409981

RESUMEN

Alzheimer's Disease (AD) and Type 2 Diabetes (T2D) share a common hallmark of insulin resistance. Reportedly, two non-canonical Receptor Tyrosine Kinases (RTKs), ALK and RYK, both targets of the same micro RNA miR-1271, exhibit significant and consistent functional down-regulation in post-mortem AD and T2D tissues. Incidentally, both have Grb2 as a common downstream adapter and NOX4 as a common ROS producing factor. Here we show that Grb2 and NOX4 play critical roles in reducing the severity of both the diseases. The study demonstrates that the abundance of Grb2 in degenerative conditions, in conjunction with NOX4, reverse cytoskeletal degradation by counterbalancing the network of small GTPases. PAX4, a transcription factor for both Grb2 and NOX4, emerges as the key link between the common pathways of AD and T2D. Down-regulation of both ALK and RYK through miR-1271, elevates the PAX4 level by reducing its suppressor ARX via Wnt/ß-Catenin signaling. For the first time, this study brings together RTKs beyond Insulin Receptor (IR) family, transcription factor PAX4 and both AD and T2D pathologies on a common regulatory platform.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Quinasa de Linfoma Anaplásico/metabolismo , Citoesqueleto/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Regulación hacia Abajo , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Vía de Señalización Wnt/genética , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Quinasa de Linfoma Anaplásico/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas del Citoesqueleto/metabolismo , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Células Hep G2 , Proteínas de Homeodominio/genética , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Factores de Transcripción Paired Box/genética , Proteínas Tirosina Quinasas Receptoras/genética , Transfección
5.
Inflammopharmacology ; 29(5): 1427-1442, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34350509

RESUMEN

BACKGROUND: Imbalance between apoptosis and autophagy in fibroblast-like synoviocytes (FLS) is one of the pathogenic mechanisms responsible for their abnormal proliferation in rheumatoid arthritis (RA). Methotrexate (MTX) demonstrated limited efficacy in amending this imbalance in fluid-derived (fd)-FLS. The active compound of black tea Theaflavin 3,3'-digallate (TF3) may be effective in restoring apoptosis-autophagy imbalance in (fd)-FLS. The combined effect of MTX + TF3 upon the same is yet to be elucidated. OBJECTIVE: To evaluate the effect of MTX + TF3 on fd-FLS to induce apoptosis and inhibit autophagy through Endoplasmic Reticulum (ER) stress-mediated pathways. METHODS: FLS from synovial fluid of 11 RA and 10 osteoarthritis patients were cultured after treatment with MTX/TF3 or a combination of MTX (125 nM) and TF3(10 µM) and the following parameters were evaluated. C-reactive protein, cytokines (TNF-α, IL-6), angiogenic markers were quantified by ELISA. fd-FLS viability was determined by MTT assay and apoptosis by flow cytometry. ER stress markers were estimated by RT-PCR (IRE1A, spliced-XBP-1) and immunoblotting (Grp78, Hsp70, CHOP, HIF-1α). Immunoblot studies were done to evaluate apoptotic (Bcl-2, Bax, Caspases) and autophagic (Beclin1, LC3b, p62) proteins. RESULTS: MTX (IC25) and TF3 (IC50) both in single doses could down-regulate the levels of pro-inflammatory and angiogenic markers. Combinatorial treatment modulated autophagosomal proteins in fd-FLS and induced apoptosis by regulating ER stress response. CONCLUSION: Disruption in homeostasis between apoptosis and autophagy in fd-FLS might be an underlying phenomenon in the progression of pathophysiology in RA. Co-administration of MTX + TF3 successfully restored the homeostasis by inducing apoptosis.


Asunto(s)
Antirreumáticos/farmacología , Artritis Reumatoide/tratamiento farmacológico , Biflavonoides/farmacología , Catequina/análogos & derivados , Metotrexato/farmacología , Adulto , Antirreumáticos/administración & dosificación , Apoptosis/efectos de los fármacos , Artritis Reumatoide/fisiopatología , Autofagia/efectos de los fármacos , Biflavonoides/administración & dosificación , Catequina/administración & dosificación , Catequina/farmacología , Células Cultivadas , Progresión de la Enfermedad , Sinergismo Farmacológico , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Masculino , Metotrexato/administración & dosificación , Persona de Mediana Edad , Osteoartritis/tratamiento farmacológico , Osteoartritis/fisiopatología , Líquido Sinovial/citología , Líquido Sinovial/efectos de los fármacos , Sinoviocitos/citología , Sinoviocitos/efectos de los fármacos
6.
J Mol Cell Cardiol ; 139: 148-163, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31958467

RESUMEN

Chronic pressure overload-induced left ventricular hypertrophy in heart is preceded by a metabolic perturbation that prefers glucose over lipid as substrate for energy requirement. Here, we establish C/EBPß (CCAAT/enhancer-binding protein ß) as an early marker of the metabolic derangement that triggers the imbalance in fatty acid (FA) oxidation and glucose uptake with increased lipid accumulation in cardiomyocytes during pathological hypertrophy, leading to contractile dysfunction and endoplasmic reticulum (ER) stress. This is the first study that shows that myocardium-targeted C/EBPß knockdown prevents the impaired cardiac function during cardiac hypertrophy led by maladaptive metabolic response with persistent hypertrophic stimuli, whereas its targeted overexpression in control increases lipid accumulation significantly compared to control hearts. A new observation from this study was the dual and opposite transcriptional regulation of the alpha and gamma isoforms of Peroxisomal proliferator activated receptors (PPARα and PPARγ) by C/EBPß in hypertrophied cardiomyocytes. Before the functional and structural remodeling sets in the diseased myocardium, C/EBPß aggravates lipid accumulation with the aid of the increased FA uptake involving induced PPARγ expression and decreased fatty acid oxidation (FAO) by suppressing PPARα expression. Glucose uptake into cardiomyocytes was greatly increased by C/EBPß via PPARα suppression. The activation of mammalian target of rapamycin complex-1 (mTORC1) during increased workload in presence of glucose as the only substrate was prevented by C/EBPß knockdown, thereby abating contractile dysfunction in cardiomyocytes. Our study thus suggests that C/EBPß may be considered as a novel cellular marker for deranged metabolic milieu before the heart pathologically remodels itself during hypertrophy.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Corazón/fisiopatología , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Cardiomegalia/genética , Cardiomegalia/patología , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Metabolismo de los Lípidos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Miocardio/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Ratas Wistar , Estrés Fisiológico
7.
Org Biomol Chem ; 18(36): 7151-7164, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32966514

RESUMEN

Convergent strategies for the first total synthesis of biselyngbyolide C and an alternative route for the total synthesis of biselyngbyolide A have been developed. The key strategic feature in this study is Heck macrocyclization. The use of intramolecular Heck coupling for biselyngbyolide B was demonstrated by us earlier; however such a strategy has not been explored further for the other members of this family of natural products, in particular, where sensitive skipped olefins are involved. The other highlights of this synthetic study include iterative Crimmins acetate aldol and Wittig olefination processes, followed by the less explored cobalt-hydride-based reduction of an activated olefin and Shiina esterification. Our synthetic study enabled us to amend the reported NMR data of biselyngbyolides A and C. An evaluation of the anticancer activities of both biselyngbyolides A and C revealed that the apoptosis generated in cancer cells followed an intrinsic pathway.


Asunto(s)
Macrólidos
8.
J Comput Aided Mol Des ; 34(8): 915-927, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32270361

RESUMEN

Insulin aggregation is the leading cause of considerable reduction in the amount of active drug molecules in liquid formulations manufactured for diabetes management. Phenolic compounds, such as phenol and m-cresol, are routinely used to stabilize insulin in a hexameric form during its commercial preparation. However, long term usage of commercial insulin results in various adverse secondary responses, for which toxicity of the phenolic excipients is primarily responsible. In this study we aimed to find out a nontoxic insulin stabilizer. To that end, we have selected resveratrol, a natural polyphenol, as a prospective nontoxic insulin stabilizer because of its structural similarity with commercially used phenolic compounds. Atomic force microscopy visualization of resveratrol-treated human insulin revealed that resveratrol has a unique ability to arrest hINS in a soluble oligomeric form having discrete spherical morphology. Most importantly, resveratrol-treated insulin is nontoxic for HepG2 cells and it effectively maintains low blood glucose in a mouse model. Cryo-electron microscopy revealed 3D morphology of resveratrol-stabilized insulin that strikingly resembles crystal structures of insulin hexamer formulated with m-cresol. Significantly, we found that, in a condition inductive to amyloid fibrillation at physiological pH, resveratrol is capable of stabilizing insulin more efficiently than m-cresol. Thus, this study describes resveratrol as an effective nontoxic natural molecule that can be used for stabilizing insulin in a bioactive oligomeric form during its commercial formulation.


Asunto(s)
Excipientes/química , Insulina/química , Insulina/farmacocinética , Resveratrol/química , Animales , Rastreo Diferencial de Calorimetría , Microscopía por Crioelectrón , Liberación de Fármacos , Estabilidad de Medicamentos , Dispersión Dinámica de Luz , Células Hep G2 , Humanos , Masculino , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Estabilidad Proteica
9.
Biochem Biophys Res Commun ; 512(4): 806-811, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-30926171

RESUMEN

Adipose triglyceride lipase (ATGL) plays a compelling role in hepatic lipid turnover and in the pathophysiology of non-alcoholic fatty liver disease. Hepatic ATGL is post-transcriptionally regulated by E3 ubiquitin ligase constitutive photomorphogenic1 (COP1) through polyubiquitylation and proteasomal degradation. However the physiological cue for COP1-mediated hepatocellular degradation of ATGL remained unknown. Here we checked for the role of pigment epithelium-derived factor (PEDF), a moonlighting hepatokine and the so-called ligand of ATGL for its stability in hepatocytes. We show that PEDF diminishes ATGL protein stability by promoting its proteasomal degradation in COP1-dependent manner. Despite being a secretory glycoprotein, PEDF is also sequestered in the nuclear compartment so as COP1. Interestingly, PEDF enhances nuclear import of predominantly cytosolic ATGL protein for its subsequent proteasomal degradation in the nucleus. PEDF also controls cell autonomous hepatocyte lipid accumulation and mobilization through COP1-ATGL axis, thereby unraveling a novel pathway for hepatic lipid metabolism.


Asunto(s)
Proteínas del Ojo/metabolismo , Lipasa/metabolismo , Hígado/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Núcleo Celular/metabolismo , Proteínas del Ojo/genética , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos , Masculino , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/genética , Serpinas/genética , Triglicéridos/metabolismo , Ubiquitinación
10.
FASEB J ; : fj201800190, 2018 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-29897812

RESUMEN

Hepatocellular death or ballooning distinguishes the transition of simple steatosis to irreversible nonalcoholic steatohepatitis (NASH). However, the molecular mechanism of hepatocellular apoptosis in NASH is largely unclear, and discovery of endogenous mediators that could prevent or inhibit cell death is thereby critical in intercepting NASH progression. Here, we identified pigment epithelium-derived factor (PEDF), a secreted, moonlighting hepatokine as 1 hepatoprotective agent in mice with diet-induced NASH. Hepatic PEDF expression is induced by IL-1ß, which is derived from inflammasome activation in liver-resident Kupffer cells, an effect that is negatively regulated by TNF-α and predominantly secreted by monocyte-derived, recruited, hepatic macrophages. Mechanistically, reciprocal and opposing roles for IL-1ß and TNF-α in PEDF expression are mediated by differential activation of NF-κB. Although augmented TNF-α production leads to temporal reduction of PEDF expression in NASH, PEDF conversely abrogates TNF-α-mediated hepatocyte death by modulating the extrinsic apoptosis pathway. Thus, our study highlights PEDF as a functionally important hepatokine in NASH progression by linking inflammasome activation and hepatocellular death.-Adak, M., Das, D., Niyogi, S., Nagalakshmi, C., Ray, D., Chakrabarti, P. Inflammasome activation in Kupffer cells confers a protective response in nonalcoholic steatohepatitis through pigment epithelium-derived factor expression.

11.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2040-2052, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29555211

RESUMEN

Glutamine, a well-established oncometabolite, anaplerotically fuels mitochondrial energy metabolism and modulates activity of mammalian/mechanistic target of rapamycin complexes (mTOR). Currently, mTOR inhibitors are in clinical use for certain types of cancer but with limited success. Since glutamine is essential for growth of many cancers, we reasoned that glutamine deprivation under conditions of mTOR inhibition should be more detrimental to cancer cell survival. However, our results show that when cells are deprived of glutamine concomitant with mTOR inhibition, hepatocarcinoma cells elicit an adaptive response which aids in their survival due to enhanced autophagic flux. Moreover, inhibition of mTOR promotes Akt ubiquitination and its proteasomal degradation however we show that Akt degradation is abrogated by increased autophagy following glutamine withdrawal. Under conditions of glutamine deficiency and mTOR inhibition, the enhanced stability of Akt protein may provide survival cues to cancer cells. Thus, our data uncovers a novel molecular link between glutamine metabolism, autophagy and stability of Akt with cancer cell survival.


Asunto(s)
Glutamina/deficiencia , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Ratones , Células 3T3 NIH , Neoplasias/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Estabilidad Proteica , Proteolisis/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Ubiquitinación/efectos de los fármacos
12.
Chembiochem ; 2018 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-29897151

RESUMEN

Nonalcoholic fatty liver disease (NAFLD), which is characterized by excess accumulation of triglyceride in hepatocytes, is the major cause of chronic liver disease worldwide and no approved drug is available. The mechanistic target of rapamycin (mTOR) complexes has been implicated in promoting lipogenesis and fat accumulation in the liver, and thus, serve as attractive drug targets. The generation of non- or low cytotoxic mTOR inhibitors is required because existing cytotoxic mTOR inhibitors are not useful for NAFLD therapy. New compounds based on the privileged adenosine triphosphate (ATP) site binder quinoline scaffold conjugated to glucose and galactosamine derivatives, which have significantly low cytotoxicity, but strong mTORC1 inhibitory activity at low micromolar concentrations, have been synthesized. These compounds also effectively inhibit the rate of lipogenesis and lipid accumulation in cultured hepatocytes. This is the first report of glycomimetic-quinoline derivatives that reduce lipid load in hepatocytes.

13.
IUBMB Life ; 70(2): 112-119, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29331088

RESUMEN

Dipeptidyl peptidase 4 (DPP4), also known as CD26 is a type II transmembrane protein that is released from the cell membrane in a nonclassical secretory mechanism. This exopeptidase selectively degrades varieties of substrates including incretin hormones, growth factors, and cytokines. A significant detectable amount of DPP4 activity can be measured in plasma as well as in different tissues such as intestinal epithelium, vascular endothelium, lymphocytes, monocytes, kidney, liver, adipose, lung, thymus, spleen, prostate, etc. Enzymatically active circulatory DPP4 is shed from the plasma membrane via proteolytic cleavage, a process responsible for the enhanced plasma DPP4 levels and activity. Elevated circulatory DPP4 activity as well as levels has been found in wide spectrum of metabolic diseases including diabetes, obesity, cardiovascular diseases, and nonalcoholic fatty liver diseases. Moreover, recent preclinical studies have further expanded the repertoire for the usage of DPP4 inhibitors in the treatment of other metabolic diseases and in their consequent complications. In the present review we highlight the reason behind the elevated circulatory DPP4 levels in metabolic diseases with a focus on the tissue of origin. We also underscore the discrepancy of protein levels with enzyme activity of circulatory DPP4 in metabolic diseases. © 2018 IUBMB Life, 70(2):112-119, 2018.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Enfermedades Metabólicas/metabolismo , Adipocitos/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Dipeptidil Peptidasa 4/sangre , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Linfocitos/metabolismo , Monocitos/metabolismo , Obesidad/metabolismo
14.
Biochem J ; 474(11): 1919-1934, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28432260

RESUMEN

Enhanced migratory potential and invasiveness of cancer cells contribute crucially to cancer progression. These phenotypes are achieved by precise alteration of invasion-associated genes through local epigenetic modifications which are recognized by a class of proteins termed a chromatin reader. ZMYND8 [zinc finger MYND (myeloid, Nervy and DEAF-1)-type containing 8], a key component of the transcription regulatory network, has recently been shown to be a novel reader of H3.1K36Me2/H4K16Ac marks. Through differential gene expression analysis upon silencing this chromatin reader, we identified a subset of genes involved in cell proliferation and invasion/migration regulated by ZMYND8. Detailed analysis uncovered its antiproliferative activity through BrdU incorporation, alteration in the expression of proliferation markers, and cell cycle regulating genes and cell viability assays. In addition, performing wound healing and invasion/migration assays, its anti-invasive nature is evident. Interestingly, epithelial-mesenchymal transition (EMT), a key mechanism of cellular invasion, is regulated by ZMYND8 where we identified its selective enrichment on promoters of CLDN1/CDH1 genes, rich in H3K36Me2/H4K16Ac marks, leading to their up-regulation. Thus, the presence of ZMYND8 could be implicated in maintaining the epithelial phenotype of cells. Furthermore, syngeneic mice, injected with ZMYND8-overexpressed invasive breast cancer cells, showed reduction in tumor volume and weight. In concert with this, we observed a significant down-regulation of ZMYND8 in invasive ductal and lobular breast cancer tissues compared with normal tissue. Taken together, our study elucidates a novel function of ZMYND8 in regulating EMT and invasion of cancer cells, possibly through its chromatin reader function.


Asunto(s)
Neoplasias de la Mama/metabolismo , Cadherinas/agonistas , Claudina-1/agonistas , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Antígenos CD , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/patología , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Claudina-1/genética , Claudina-1/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica/patología , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Interferencia de ARN , Receptores de Cinasa C Activada , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Supresoras de Tumor
15.
FASEB J ; 30(11): 3822-3834, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27485820

RESUMEN

Adipose triglyceride lipase (ATGL) maintains an optimum mitochondrial function putatively by generating cognate ligands for peroxisome proliferator-activated receptor α (PPARα), which, together with PPARγ coactivator-1α (PGC1α), regulate muscle mitochondrial biogenesis. However, the cross-talk between ATGL and PPARα in skeletal muscle mitochondrial metabolism and its implication in chronological aging is poorly understood. The role of ATGL in muscle mitochondrial metabolism was studied by overexpressing and depleting the gene and studying its downstream effect in cultured myotubes and in murine skeletal muscle. We found that PPARα directly induces ATGL expression during myogenesis. Overexpression of ATGL significantly enhanced while depletion of ATGL attenuated mitochondrial oxidative phosphorylation and fatty acid oxidation without alteration in mitochondrial content, and it rendered PPARα and PGC1α redundant in promoting mitochondrial oxidative function. However, ATGL did not alter PPARα-dependent lipid accumulation and insulin sensitivity. In middle-aged rats, ATGL expression was higher and correlated with PPARα expression and sustained fatty acid oxidation in oxidative soleus muscle. Fenofibrate feeding further induced ATGL expression selectively in this muscle compartment. These findings illustrate that PPARα and ATGL constitute a regulatory pathway in skeletal muscle, suggesting their role as a mitochondrial metabolic reserve.-Biswas, D., Ghosh, M., Kumar, S., Chakrabarti, P. PPARα-ATGL pathway improves muscle mitochondrial metabolism: implication in aging.


Asunto(s)
Envejecimiento/fisiología , Lipasa/metabolismo , Metabolismo de los Lípidos/fisiología , Mitocondrias Musculares/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , PPAR alfa/metabolismo , Transducción de Señal , Animales , Fenofibrato/farmacología , Resistencia a la Insulina/fisiología , Mitocondrias Musculares/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Ratas , Factores de Transcripción/metabolismo
16.
J Biol Chem ; 290(28): 17331-8, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25814662

RESUMEN

Early growth response transcription factor Egr1 controls multiple aspects of cell physiology and metabolism. In particular, Egr1 suppresses lipolysis and promotes fat accumulation in adipocytes by inhibiting the expression of adipose triglyceride lipase. According to current dogma, regulation of the Egr1 expression takes place primarily at the level of transcription. Correspondingly, treatment of cultured adipocytes with insulin stimulates expression of Egr1 mRNA and protein. Unexpectedly, the MEK inhibitor PD98059 completely blocks insulin-stimulated increase in the Egr1 mRNA but has only a moderate effect on the Egr1 protein. At the same time, mTORC1 inhibitors rapamycin and PP242 suppress expression of the Egr1 protein and have an opposite effect on the Egr1 mRNA. Mouse embryonic fibroblasts with genetic ablations of TSC2 or 4E-BP1/2 express less Egr1 mRNA but more Egr1 protein than wild type controls. (35)S-labeling has confirmed that translation of the Egr1 mRNA is much more effective in 4E-BP1/2-null cells than in control. A selective agonist of the CB1 receptors, ACEA, up-regulates Egr1 mRNA, but does not activate mTORC1 and does not increase Egr1 protein in adipocytes. These data suggest that although insulin activates both the Erk and the mTORC1 signaling pathways in adipocytes, regulation of the Egr1 expression takes place predominantly via the mTORC1/4E-BP-mediated axis. In confirmation of this model, we show that 4E-BP1/2-null MEFs express less ATGL and accumulate more fat than control cells, while knock down of Egr1 in 4E-BP1/2-null MEFs increases ATGL expression and decreases fat storage.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Lipasa/metabolismo , Metabolismo de los Lípidos , Fosfoproteínas/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/metabolismo , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/antagonistas & inhibidores , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Factores Eucarióticos de Iniciación/antagonistas & inhibidores , Factores Eucarióticos de Iniciación/genética , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Insulina/metabolismo , Lipólisis , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
17.
IEEE J Biomed Health Inform ; 28(7): 4184-4193, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38593020

RESUMEN

Detecting Alzheimer's disease (AD) accurately at an early stage is critical for planning and implementing disease-modifying treatments that can help prevent the progression to severe stages of the disease. In the existing literature, diagnostic test scores and clinical status have been provided for specific time points, and predicting the disease progression poses a significant challenge. However, few studies focus on longitudinal data to build deep-learning models for AD detection. These models are not stable to be relied upon in real medical settings due to a lack of adaptive training and testing. We aim to predict the individual's diagnostic status for the next six years in an adaptive manner where prediction performance improves with the number of patient visits. This study presents a Sequence-Length Adaptive Encoder-Decoder Long Short-Term Memory (SLA-ED LSTM) deep-learning model on longitudinal data obtained from the Alzheimer's Disease Neuroimaging Initiative archive. In the suggested approach, decoder LSTM dynamically adjusts to accommodate variations in training sequence length and inference length rather than being constrained to a fixed length. We evaluated the model performance for various sequence lengths and found that for inference length one, sequence length nine gives the highest average test accuracy and area under the receiver operating characteristic curves of 0.920 and 0.982, respectively. This insight suggests that data from nine visits effectively captures meaningful cognitive status changes and is adequate for accurate model training. We conducted a comparative analysis of the proposed model against state-of-the-art methods, revealing a significant improvement in disease progression prediction over the previous methods.


Asunto(s)
Enfermedad de Alzheimer , Aprendizaje Profundo , Progresión de la Enfermedad , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/diagnóstico , Humanos , Anciano , Masculino , Femenino
18.
BMJ Open Diabetes Res Care ; 12(4)2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39209774

RESUMEN

INTRODUCTION: Adipsin or complement factor D is an adipokine that augments insulin secretion, is altered in various degrees of obesity, and is involved in alternative complement pathway. However, whether adipsin has any independent association with risk factors and biomarkers in patients with type 2 diabetes (T2D) remains elusive. RESEARCH DESIGN AND METHODS: We performed an oral glucose tolerance test on a subset of 43 patients with T2D from the community health cohort to access the role of adipsin in insulin secretion. We further cross-sectionally examined the role of adipsin in plasma, adipose tissue (AT), and secretion in a community cohort of 353 subjects and a hospital cohort of 52 subjects. RESULTS: We found that plasma adipsin has no significant correlation with insulin secretion in people with diabetes. Among the risk factors of T2D, adipsin levels were independently associated only with age, and a positive correlation between plasma adipsin and age among subjects without T2D was lost in patients with T2D. Plasma adipsin levels, AT adipsin expression, and secretion were upregulated both in T2D and aging, with a corresponding drop in Homeostatic Model Assessment for assessing ß-cell function. Adipsin expression was positively associated with other aging biomarkers, such as ß-galactosidase, p21, and p16. These results also corroborated with existing plasma proteomic signatures of aging, including growth, and differentiation factor-15, which strongly correlated with adipsin. CONCLUSIONS: Our results demonstrate an increase in circulating adipsin in T2D and aging, and it scores as a candidate plasma marker for aging specifically in non-T2D population.


Asunto(s)
Tejido Adiposo , Envejecimiento , Biomarcadores , Factor D del Complemento , Diabetes Mellitus Tipo 2 , Humanos , Factor D del Complemento/metabolismo , Factor D del Complemento/análisis , Masculino , Femenino , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/sangre , Persona de Mediana Edad , Biomarcadores/análisis , Biomarcadores/sangre , Envejecimiento/metabolismo , Estudios Transversales , Tejido Adiposo/metabolismo , Anciano , Adulto , Prueba de Tolerancia a la Glucosa , Factores de Riesgo , Insulina/metabolismo , Insulina/sangre
19.
ACS Med Chem Lett ; 15(5): 696-705, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38746877

RESUMEN

A systematic structure-activity relationship study of the potent anticancer marine macrolide biselyngbyolide B has been accomplished. A total of 11 structural variants of the parent natural product, of which 2 are natural analogues, have been studied against a human colorectal carcinoma cell line. The requisite functional units of the parent molecule responsible for the cytotoxic activities have been disclosed. Biselyngbyolide C, one of the natural analogues of biselyngbyolide B, has been studied in depth to explore its molecular mechanism. Interestingly, the in vitro data demonstrated an induction of dynamin-related protein 1-mediated mitochondrial fission and reactive oxygen species production which led to activation of ASK1/P38/JNK-mediated apoptosis in colon cancer cells as an important pathway for biselyngbyolide B-mediated cytotoxicity. Notably, this study revealed that a macrolide participated in mitochondrial fission to promote apoptosis of cancer cells, providing new insight.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA