Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Arch Microbiol ; 206(9): 385, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39177836

RESUMEN

Hydrophobins (HFBs) and cerato-platanins (CPs) are surface-active extracellular proteins produced by filamentous fungi. This study identified two HFB genes (pshyd1 and pshyd2) and one CP gene (pscp) in the marine fungus Paradendryphiella salina. The proteins PsCP, PsHYD2, and PsHYD1 had molecular weights of 12.70, 6.62, and 5.98 kDa, respectively, with isoelectric points below 7. PsHYD1 and PsHYD2 showed hydrophobicity (GRAVY score 0.462), while PsCP was hydrophilic (GRAVY score - 0.202). Stability indices indicated in-solution stability. Mass spectrometry identified 2,922 proteins, including CP but not HFB proteins. qPCR revealed differential gene expression influenced by developmental stage and substrate, with pshyd1 consistently expressed. These findings suggest P. salina's adaptation to marine ecosystems with fewer hydrophobin genes than other fungi but capable of producing surface-active proteins from seaweed carbohydrates. These proteins have potential applications in medical biocoatings, food industry foam stabilizers, and environmental bioremediation.


Asunto(s)
Proteínas Fúngicas , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/química , Ascomicetos/genética , Ascomicetos/metabolismo , Ascomicetos/química , Algas Marinas/microbiología , Algas Marinas/química , Organismos Acuáticos/genética , Organismos Acuáticos/metabolismo , Regulación Fúngica de la Expresión Génica , Agua de Mar/microbiología
2.
Diabetes ; 70(1): 204-213, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33033064

RESUMEN

The aim of the current study was to evaluate the effect of sustained physiologic increase of ∼50 mg/dL in plasma glucose concentration on insulin secretion in normal glucose-tolerant (NGT) subjects. Twelve NGT subjects without family history of type 2 diabetes mellitus (T2DM; FH-) and 8 NGT with family history of T2DM (FH+) received an oral glucose tolerance test and two-step hyperglycemic clamp (100 and 300 mg/dL) followed by intravenous arginine bolus before and after 72-h glucose infusion. Fasting plasma glucose increased from 94 ± 2 to 142 ± 4 mg/dL for 72 h. First-phase insulin secretion (0-10 min) increased by 70%, while second-phase insulin secretion during the first (10-80 min) and second (90-160 min) hyperglycemic clamp steps increased by 3.8-fold and 1.9-fold, respectively, following 72 h of physiologic hyperglycemia. Insulin sensitivity during hyperglycemic clamp declined by ∼30% and ∼55% (both P < 0.05), respectively, during the first and second hyperglycemic clamp steps. Insulin secretion/insulin resistance (disposition) index declined by 60% (second clamp step) and by 62% following arginine (both P < 0.005) following 72-h glucose infusion. The effect of 72-h glucose infusion on insulin secretion and insulin sensitivity was similar in subjects with and without FH of T2DM. Following 72 h of physiologic hyperglycemia, metabolic clearance rate of insulin was markedly reduced (P < 0.01). These results demonstrate that sustained physiologic hyperglycemia for 72 h 1) increases absolute insulin secretion but impairs ß-cell function, 2) causes insulin resistance, and 3) reduces metabolic clearance rate of insulin.


Asunto(s)
Glucemia/metabolismo , Hiperglucemia/metabolismo , Resistencia a la Insulina/fisiología , Secreción de Insulina/fisiología , Insulina/metabolismo , Adulto , Femenino , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Voluntarios Sanos , Humanos , Hiperglucemia/sangre , Masculino , Persona de Mediana Edad
3.
Mol Metab ; 45: 101154, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359401

RESUMEN

OBJECTIVE: Insulin resistance and altered hepatic mitochondrial function are central features of type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD), but the etiological role of these processes in disease progression remains unclear. Here we investigated the molecular links between insulin resistance, mitochondrial remodeling, and hepatic lipid accumulation. METHODS: Hepatic insulin sensitivity, endogenous glucose production, and mitochondrial metabolic fluxes were determined in wild-type, obese (ob/ob) and pioglitazone-treatment obese mice using a combination of radiolabeled tracer and stable isotope NMR approaches. Mechanistic studies of pioglitazone action were performed in isolated primary hepatocytes, whilst molecular hepatic lipid species were profiled using shotgun lipidomics. RESULTS: Livers from obese, insulin-resistant mice displayed augmented mitochondrial content and increased tricarboxylic acid cycle (TCA) cycle and pyruvate dehydrogenase (PDH) activities. Insulin sensitization with pioglitazone mitigated pyruvate-driven TCA cycle activity and PDH activation via both allosteric (intracellular pyruvate availability) and covalent (PDK4 and PDP2) mechanisms that were dependent on PPARγ activity in isolated primary hepatocytes. Improved mitochondrial function following pioglitazone treatment was entirely dissociated from changes in hepatic triglycerides, diacylglycerides, or fatty acids. Instead, we highlight a role for the mitochondrial phospholipid cardiolipin, which underwent pathological remodeling in livers from obese mice that was reversed by insulin sensitization. CONCLUSION: Our findings identify targetable mitochondrial features of T2D and NAFLD and highlight the benefit of insulin sensitization in managing the clinical burden of obesity-associated disease.


Asunto(s)
Resistencia a la Insulina/fisiología , Hígado/metabolismo , Mitocondrias Hepáticas/metabolismo , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Glucemia/metabolismo , Cardiolipinas , Ciclo del Ácido Cítrico , Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos/metabolismo , Hepatocitos/metabolismo , Insulina/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Tiazolidinedionas , Triglicéridos/metabolismo
4.
Stem Cells ; 38(4): 542-555, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31828876

RESUMEN

A comprehensive characterization of the molecular processes controlling cell fate decisions is essential to derive stable progenitors and terminally differentiated cells that are functional from human pluripotent stem cells (hPSCs). Here, we report the use of quantitative proteomics to describe early proteome adaptations during hPSC differentiation toward pancreatic progenitors. We report that the use of unbiased quantitative proteomics allows the simultaneous profiling of numerous proteins at multiple time points, and is a valuable tool to guide the discovery of signaling events and molecular signatures underlying cellular differentiation. We also monitored the activity level of pathways whose roles are pivotal in the early pancreas differentiation, including the Hippo signaling pathway. The quantitative proteomics data set provides insights into the dynamics of the global proteome during the transition of hPSCs from a pluripotent state toward pancreatic differentiation.


Asunto(s)
Páncreas/metabolismo , Células Madre Pluripotentes/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Diferenciación Celular , Humanos , Páncreas/citología
5.
Mol Metab ; 18: 153-163, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30316806

RESUMEN

OBJECTIVES: Insulin receptor (IR)-mediated signaling is involved in the regulation of pluripotent stem cells; however, its direct effects on regulating the maintenance of pluripotency and lineage development are not fully understood. The main objective of this study is to understand the role of IR signaling in pluripotency and lineage development. METHODS: To explore the role of IR signaling, we generated IR knock-out (IRKO) mouse induced pluripotent stem cells (miPSCs) from E14.5 mouse embryonic fibroblasts (MEFs) of global IRKO mice using a cocktail of four reprogramming factors: Oct4, Sox2, Klf4, cMyc. We performed pluripotency characterization and directed the differentiation of control and IRKO iPSCs into neural progenitors (ectoderm), adipocyte progenitors (mesoderm), and pancreatic beta-like cells (endoderm). We mechanistically confirmed these findings via phosphoproteomics analyses of control and IRKO iPSCs. RESULTS: Interestingly, expression of pluripotency markers including Klf4, Lin28a, Tbx3, and cMyc were upregulated, while abundance of Oct4 and Nanog were enhanced by 4-fold and 3-fold, respectively, in IRKO iPSCs. Analyses of signaling pathways demonstrated downregulation of phospho-STAT3, p-mTor and p-Erk and an increase in the total mTor and Erk proteins in IRKO iPSCs in the basal unstimulated state. Stimulation with leukemia inhibitory factor (LIF) showed a ∼33% decrease of phospho-ERK in IRKO iPSCs. On the contrary, Erk phosphorylation was increased during in vitro spontaneous differentiation of iPSCs lacking IRs. Lineage-specific directed differentiation of the iPSCs revealed that cells lacking IR showed enhanced expression of neuronal lineage markers (Pax6, Tubb3, Ascl1 and Oligo2) while exhibiting a decrease in adipocyte (Fas, Acc, Pparγ, Fabp4, C/ebpα, and Fsp27) and pancreatic beta cell markers (Ngn3, Isl1, and Sox9). Further molecular characterization by phosphoproteomics confirmed the novel IR-mediated regulation of the global pluripotency network including several key proteins involved in diverse aspects of growth and embryonic development. CONCLUSION: We report, for the first time to our knowledge, the phosphoproteome of insulin, IGF1, and LIF stimulation in mouse iPSCs to reveal the importance of insulin receptor signaling for the maintenance of pluripotency and lineage determination.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal , Adipocitos/citología , Adipocitos/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Factor 4 Similar a Kruppel , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Receptor de Insulina/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Cell Rep ; 15(3): 460-470, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27068459

RESUMEN

A major goal of diabetes research is to develop strategies that replenish pancreatic insulin-producing beta cells. One emerging strategy is to harness pancreatic plasticity-the ability of pancreatic cells to undergo cellular interconversions-a phenomenon implicated in physiological stress and pancreatic injury. Here, we investigate the effects of inflammatory cytokine stress on the differentiation potential of ductal cells in a human cell line, in mouse ductal cells by pancreatic intraductal injection, and during the progression of autoimmune diabetes in the non-obese diabetic (NOD) mouse model. We find that inflammatory cytokine insults stimulate epithelial-to-mesenchymal transition (EMT) as well as the endocrine program in human pancreatic ductal cells via STAT3-dependent NGN3 activation. Furthermore, we show that inflammatory cytokines activate ductal-to-endocrine cell reprogramming in vivo independent of hyperglycemic stress. Together, our findings provide evidence that inflammatory cytokines direct ductal-to-endocrine cell differentiation, with implications for beta cell regeneration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Citocinas/farmacología , Sistema Endocrino/citología , Mediadores de Inflamación/farmacología , Proteínas del Tejido Nervioso/metabolismo , Conductos Pancreáticos/citología , Factor de Transcripción STAT3/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/patología , Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
7.
Stem Cell Reports ; 6(3): 357-67, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26876668

RESUMEN

Patients with an HNF1B(S148L/+) mutation (MODY5) typically exhibit pancreatic hypoplasia. However, the molecular mechanisms are unknown due to inaccessibility of patient material and because mouse models do not fully recapitulate MODY5. Here, we differentiated MODY5 human-induced pluripotent stem cells (hiPSCs) into pancreatic progenitors, and show that the HNF1B(S148L/+) mutation causes a compensatory increase in several pancreatic transcription factors, and surprisingly, a decrease in PAX6 pancreatic gene expression. The lack of suppression of PDX1, PTF1A, GATA4, and GATA6 indicates that MODY5-mediated pancreatic hypoplasia is mechanistically independent. Overexpression studies demonstrate that a compensatory increase in PDX1 gene expression is due to mutant HNF1B(S148L/+) but not wild-type HNF1B or HNF1A. Furthermore, HNF1B does not appear to directly regulate PAX6 gene expression necessary for glucose tolerance. Our results demonstrate compensatory mechanisms in the pancreatic transcription factor network due to mutant HNF1B(S148L/+) protein. Thus, patients typically develop MODY5 but not neonatal diabetes despite exhibiting pancreatic hypoplasia.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Páncreas/patología , Células Cultivadas , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Páncreas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Cell Metab ; 22(2): 239-52, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26244933

RESUMEN

The mechanisms underlying the development of complications in type 1 diabetes (T1D) are poorly understood. Disease modeling of induced pluripotent stem cells (iPSCs) from patients with longstanding T1D (disease duration ≥ 50 years) with severe (Medalist +C) or absent to mild complications (Medalist -C) revealed impaired growth, reprogramming, and differentiation in Medalist +C. Genomics and proteomics analyses suggested differential regulation of DNA damage checkpoint proteins favoring protection from cellular apoptosis in Medalist -C. In silico analyses showed altered expression patterns of DNA damage checkpoint factors among the Medalist groups to be targets of miR200, whose expression was significantly elevated in Medalist +C serum. Notably, neurons differentiated from Medalist +C iPSCs exhibited enhanced susceptibility to genotoxic stress that worsened upon miR200 overexpression. Furthermore, knockdown of miR200 in Medalist +C fibroblasts and iPSCs rescued checkpoint protein expression and reduced DNA damage. We propose miR200-regulated DNA damage checkpoint pathway as a potential therapeutic target for treating complications of diabetes.


Asunto(s)
Puntos de Control del Ciclo Celular , Daño del ADN , Diabetes Mellitus Tipo 1/metabolismo , Regulación de la Expresión Génica , MicroARNs/biosíntesis , Modelos Biológicos , Anciano , Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/patología , Complicaciones de la Diabetes/prevención & control , Diabetes Mellitus Tipo 1/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Masculino , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología
9.
Sci Transl Med ; 7(273): 273ps2, 2015 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25653218

RESUMEN

Controversy has long surrounded research on pancreatic beta cell regeneration. Some groups have used nonphysiological experimental methodologies to build support for the existence of pancreatic progenitor cells within the adult pancreas that constantly replenish the beta cell pool; others argue strongly against this mode of regeneration. Recent research has reinvigorated enthusiasm for the harnessing of pancreatic plasticity for therapeutic application--for example, the transdifferentiation of human pancreatic exocrine cells into insulin-secreting beta-like cells in vitro; the conversion of mouse pancreatic acinar cells to beta-like cells in vivo via cytokine treatment; and the potential redifferentiation of dedifferentiated mouse beta cells in vivo. Here, we highlight key findings in this provocative field and provide a perspective on possible exploitation of human pancreatic plasticity for therapeutic beta cell regeneration.


Asunto(s)
Páncreas/citología , Estrés Fisiológico , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Páncreas/metabolismo , Páncreas Exocrino/citología , Páncreas Exocrino/metabolismo , Regeneración
10.
Stem Cell Reports ; 3(1): 5-14, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-25068117

RESUMEN

There is considerable interest in differentiating human pluripotent stem cells (hPSCs) into definitive endoderm (DE) and pancreatic cells for in vitro disease modeling and cell replacement therapy. Numerous protocols use fetal bovine serum, which contains poorly defined factors to induce DE formation. Here, we compared Wnt and BMP in their ability to cooperate with Activin signaling to promote DE formation in a chemically defined medium. Varying concentrations of WNT3A, glycogen synthase kinase (GSK)-3 inhibitors CHIR99021 and 6-bromoindirubin-3'-oxime (BIO), and BMP4 could independently co-operate with Activin to effectively induce DE formation even in the absence of serum. Overall, CHIR99021 is favored due to its cost effectiveness. Surprisingly, WNT3A was ineffective in suppressing E-CADHERIN/CDH1 and pluripotency factor gene expression unlike GSK-3 inhibitors or BMP4. Our findings indicate that both Wnt and BMP effectively synergize with Activin signaling to generate DE from hPSCs, although WNT3A requires additional factors to suppress the pluripotency program inherent in hPSCs.


Asunto(s)
Activinas/farmacología , Proteínas Morfogenéticas Óseas/metabolismo , Endodermo/citología , Endodermo/metabolismo , Western Blotting , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Línea Celular , Citometría de Flujo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Humanos , Indoles/farmacología , Oximas/farmacología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suero/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
11.
Biofarbo ; 17(1): 47-53, 2009. graf
Artículo en Español | LILACS | ID: lil-544859

RESUMEN

Bertholletia excelsa, es un árbol originario de la selva húmeda tropical de la amazonía sudamericana. Sus frutos conocidos como castaña constituyen uno de los productos forestales no maderables más importantes del norte de Boliva.


Asunto(s)
Bertholletia , Cromatografía de Gases , Ácidos Grasos , Cromatografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA