Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Endocr Disord ; 21(1): 47, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33711989

RESUMEN

BACKGROUND: NKX6.1 is a transcription factor for insulin, as well as a marker for ß cell maturity. Abnormal NKX6.1 expression in ß cells, such as translocation from the nucleus to cytoplasm or lost expression, has been shown as a marker for ß cell dedifferentiation. METHODS: We obtained pancreatic sections from organ donors and immunofluorescence staining with NKX6.1 and insulin was performed to characterize NKX6.1 expression in subjects with or without type 2 diabetes mellitus (T2DM). RESULTS: Our results showed that cells with insulin expression but no nucleic NKX6.1 expression (NKX6.1Nuc-Ins+), and cells with cytoplasmic NKX6.1 expression but no insulin expression (NKX6.1cytIns-) were significantly increased in T2DM subjects and positively correlated with glycated hemoglobin (HbA1c), indicating the elevated ß cell dedifferentiation with NKX6.1 inactivation in T2DM. To investigate whether ß cell dedifferentiation has initiated in subjects with higher risks for T2DM, we next analyzed the association between ß-cell dedifferentiation level in ND subjects with different ages, body mass index, and HbA1c. The results showed the absolute number and percentage of dedifferentiated ß cells with NKX6.1 inactivation did not significantly change in subjects with advanced aging, obesity, or modest hyperglycemia, indicating that the ß cell dedifferentiation might mainly occur after T2DM was diagnosed. CONCLUSION: Our results suggested that NKX6.1 expression in ß cells was changed in type 2 diabetic subjects, evidenced by significantly increased NKX6.1Nuc-Ins+ and NKX6.1cytIns- cells. This abnormality did not occur more frequently in subjects with a higher risk for T2DM, suggesting that ß cell dedifferentiation might be secondary to the pathological changes in T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/metabolismo , Estado Prediabético/metabolismo , Adulto , Anciano , Autopsia , Estudios de Casos y Controles , Recuento de Células , Diferenciación Celular , Diabetes Mellitus Tipo 2/patología , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Masculino , Persona de Mediana Edad , Estado Prediabético/patología , Factores de Riesgo
2.
J Nat Prod ; 83(9): 2696-2705, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32869646

RESUMEN

Co-cultivation has been used as a promising tool to turn on or up-regulate cryptic biosynthetic pathways for microbial natural product discovery. Recently, a modified culturing strategy similar to co-cultivation was investigated, where heat-killed inducer cultures were supplemented to the culture medium of producer fermentations to induce cryptic pathways. In the present study, the repeatability and effectiveness of both methods in turning on cryptic biosynthetic pathways were unbiasedly assessed using UHPLC-HRESIMS-based metabolomics analysis. Both induction methods had good repeatability, and they resulted in very different induced metabolites from the tested producers. Co-cultivation generated more induced mass features than the heat-killed inducer cultures, while both methods resulted in the induction of mass features not observed using the other induction method. As examples, pathways leading to two new natural products, N-carbamoyl-2-hydroxy-3-methoxybenzamide (1) and carbazoquinocin G (5), were induced and up-regulated through co-culturing a producer Streptomyces sp. RKND-216 with inducers Alteromonas sp. RKMC-009 and M. smegmatis ATCC 120515, respectively.


Asunto(s)
Redes y Vías Metabólicas , Metaboloma , Alteromonas/metabolismo , Antineoplásicos/farmacología , Bacterias/efectos de los fármacos , Productos Biológicos , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Técnicas de Cocultivo , Descubrimiento de Drogas , Calor , Humanos , Pruebas de Sensibilidad Microbiana , Mycobacterium smegmatis/efectos de los fármacos , Reproducibilidad de los Resultados , Espectrometría de Masa por Ionización de Electrospray , Esterilización , Streptomyces/metabolismo
3.
Int J Stem Cells ; 13(2): 268-278, 2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32587133

RESUMEN

BACKGROUND AND OBJECTIVES: Adipose tissue-derived mesenchymal stem cells (ASCs) are recognized as an advantaged source for the prevention and treatment of diverse diseases including type 2 diabetes mellitus (T2DM). However, alterations in characteristics of ASCs from the aforementioned T2DM patients are still obscure, which also hinder the rigorous and systematic illumination of progression and pathogenesis. METHODS AND RESULTS: In this study, we originally isolated peripancreatic adipose tissue-derived mesenchymal stem cells from both human type 2 diabetic and non-diabetic donors (T2DM-ASCs, ND-ASCs) with the parental consent, respectively. We noticed that T2DM-ASCs exhibited indistinguishable immunophenotype, cell vitality, chondrogenic differentiation and stemness as ND-ASCs. Simultaneously, there's merely alterations in migration and immunoregulatory capacities in T2DM-ASCs. However, differing from ND-ASCs, T2DM-ASCs exhibited deficiency in adipogenic and osteogenic differentiation, and in particular, the delayed cell cycle and different cytokine expression spectrum. CONCLUSIONS: The conservative alterations of T2DM-ASCs in multifaceted characteristics indicated the possibility of autologous application of ASCs for cell-based T2DM treatment in the future.

4.
Cell Transplant ; 29: 963689720903691, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32364405

RESUMEN

During the process of human islet isolation a cascade of stressful events are triggered and negatively influence islet yield, viability, and function, including the production of proinflammatory cytokines and activation of apoptosis. Carbon monoxide-releasing molecule 2 (CORM-2) is a donor of carbon monoxide (CO) and can release CO spontaneously. Accumulating studies suggest that CORM-2 exerts cytoprotective and anti-inflammatory properties. However, the effect of CORM-2 on islet isolation is still unclear. In this study, we found that CORM-2 pretreatment significantly decreased the expression of critical inflammatory genes, including tissue factor, intercellular adhesion molecule-1, chemokine (C-C motif) ligand 2, C-X-C motif chemokine 10, Toll-like receptor 4, interleukin-1ß, interleukin-6, and tumor necrosis factor-α (TNF-α). The isolated islets of the CORM-2 pretreatment group showed reduced apoptotic rate, improved viability, and higher glucose-stimulated insulin secretion, and functional gene expression in comparison to control group. Importantly, CORM-2 pretreatment prevented the impairment caused by TNF-α, evidenced by the improved glucose-stimulated index and transplantation outcomes. The present study demonstrated the anti-inflammatory property of CORM-2 during human islet isolation, and we suggest that CORM-2 pretreatment is an appealing treatment to mitigate inflammation-mediated islet dysfunction during isolation and culture ex vivo and to preserve long-term islet survival and function.


Asunto(s)
Inflamación/tratamiento farmacológico , Compuestos Organometálicos/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Citometría de Flujo , Prueba de Tolerancia a la Glucosa , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Trasplante de Islotes Pancreáticos , Masculino , Ratones Endogámicos BALB C , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Pancreas ; 49(5): 692-698, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32433408

RESUMEN

OBJECTIVES: The present study aimed to investigate the dynamic change of α cells and ß cells, and their ratios in prediabetes and type 2 diabetes in the Chinese population. METHODS: Pancreata from 27 nondiabetic (ND), 8 prediabetic (PreD), and 19 type 2 diabetic (T2D) organ donors were subjected to immunofluorescence staining with insulin and glucagon. RESULTS: The ß to α ratio in islets (ß/α) in PreD was significantly higher than that in ND, resulting from an increase of ß cells and a decrease of α cells per islet, but that in T2D was significantly lower than that in ND, resulting from a decrease of ß cells and an increase of α cells per islet. The ß-cell percentage and ß/α ratio positively correlated and α-cell percentage negatively correlated with HbA1c (glycated hemoglobin) in ND and PreD, but these correlations disappeared when T2D subjects were included. CONCLUSIONS: The islet ß to α ratio increased in PreD individuals because of a relative α-cell loss and ß-cell compensation and decreased after T2D onset because of both ß-cell loss and α-cell reexpansion.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Células Secretoras de Glucagón/patología , Células Secretoras de Insulina/patología , Estado Prediabético/patología , Adulto , Pueblo Asiatico , Recuento de Células , China , Diabetes Mellitus Tipo 2/etnología , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Glucagón/metabolismo , Hemoglobina Glucada/metabolismo , Humanos , Insulina/metabolismo , Masculino , Persona de Mediana Edad , Estado Prediabético/etnología , Estado Prediabético/metabolismo
6.
EBioMedicine ; 51: 102615, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31918404

RESUMEN

BACKGROUND: A physiological hallmark of patients with type 2 diabetes mellitus (T2DM) is ß cell dysfunction. Despite adequate treatment, it is an irreversible process that follows disease progression. Therefore, the development of novel therapies that restore ß cell function is of utmost importance. METHODS: This study aims to unveil the mechanistic action of mesenchymal stem cells (MSCs) by investigating its impact on isolated human T2DM islets ex vivo and in vivo. FINDINGS: We propose that MSCs can attenuate ß cell dysfunction by reversing ß cell dedifferentiation in an IL-1Ra-mediated manner. In response to the elevated expression of proinflammatory cytokines in human T2DM islet cells, we observed that MSCs was activated to secret IL-1R antagonist (IL-1Ra) which acted on the inflammed islets and reversed ß cell dedifferentiation, suggesting a crosstalk between MSCs and human T2DM islets. The co-transplantation of MSCs with human T2DM islets in diabetic SCID mice and intravenous infusion of MSCs in db/db mice revealed the reversal of ß cell dedifferentiation and improved glycaemic control in the latter. INTERPRETATION: This evidence highlights the potential of MSCs in future cell-based therapies regarding the amelioration of ß cell dysfunction.


Asunto(s)
Desdiferenciación Celular , Diabetes Mellitus Tipo 2/patología , Células Secretoras de Insulina/patología , Células Madre Mesenquimatosas/metabolismo , Animales , Diabetes Mellitus Tipo 2/terapia , Femenino , Humanos , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1beta/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones SCID , Persona de Mediana Edad , Factor de Necrosis Tumoral alfa/metabolismo
7.
Biochem Biophys Res Commun ; 523(2): 382-388, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31866014

RESUMEN

Hypoxia affects the function of pancreatic ß cells, and the molecular mechanism underlying hypoxia-related ß cell dysfunction in human type 2 diabetes mellitus (T2DM) remains to be elucidated. In this study, by comparing the gene expression profiles of islets from nondiabetic and T2D subjects using gene chip array, we aimed to elucidate that hypoxia signaling pathways are activated in human T2DM islets. CoCl2 treatment, which was employed to mimic hypoxic stimulation in human islets, decreased insulin secretion, insulin content, and the functional gene expression of human islets. In parallel, the expression of mature ß cell-disallowed genes was upregulated by CoCl2, including progenitor cell marker NGN3, ß cell differentiation marker ALDH1A3, and genes that are typically inhibited in mature ß cells, namely, GLUT1 and LDHA, indicating that CoCl2-mimicked hypoxia induced ß cell dedifferentiation of human islets. This finding in human islets was confirmed in mouse ß cell line NIT-1. By using Dimethyloxalylglycine (DMOG) to activate hypoxia-inducible factor-1α (HIF-1α) or siRNAs to knockdown HIF-1α, we found that HIF-1α was a key regulator of hypoxia-induced dedifferentiation of ß cells by upregulating mature ß cell-disallowed genes. Our findings suggested that HIF-1α activation might be an important contributor to ß cell dedifferentiation in human T2DM islets, and HIF-1α-targeted therapies may have the potential to reverse ß cell dedifferentiation of human T2DM islets.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Animales , Desdiferenciación Celular/efectos de los fármacos , Desdiferenciación Celular/genética , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Línea Celular , Cobalto/toxicidad , Diabetes Mellitus Tipo 2/genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Transducción de Señal
8.
Endocr J ; 66(8): 691-699, 2019 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-31105125

RESUMEN

The cyclooxygenase2 (COX-2) enzyme catalyzes the first step of prostanoid biosynthesis, and is known for its crucial role in the pathogenesis of several inflammatory diseases including type 2 diabetes mellitus (T2DM). Although a variety of studies revealed that COX-2 played a role in the IL-1ß induced ß cell dysfunction, the molecular mechanism remains unclear. Here, using a cDNA microarray and in silico analysis, we demonstrated that inflammatory responses were upregulated in human T2DM islets compared with non-diabetic (ND) islets. COX-2 expression was significantly enhanced in human T2DM islets, correlated with the high inflammation level. PGE2, the catalytic product of COX-2, downregulated the functional gene expression of PDX1, NKX6.1, and MAFA and blunted the glucose induced insulin secretion of human islets. Conversely, inhibition of COX-2 activity by a pharmaceutical inhibitor prevented the ß-cell dysfunction induced by IL-1ß. COX-2 inhibitor also abrogated the IL-1ß autostimulation in ß cells, which further resulted in reduced COX-2 expression in ß cells. Together, our results revealed that COX-2/PGE2 signaling was involved in the regulation of IL-1ß autostimulation, thus forming an IL-1ß/COX-2/PGE2 pathway loop, which may result in the high inflammation level in human T2DM islets and the inflammatory impairment of ß cells. Breaking this IL-1ß/COX-2/PGE2 pathway loop provides a potential therapeutic strategy to improve ß cell function in the treatment of T2DM patients.


Asunto(s)
Ciclooxigenasa 2/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Dinoprostona/fisiología , Interleucina-1beta/fisiología , Islotes Pancreáticos/fisiopatología , Adulto , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/patología , Dinoprostona/metabolismo , Retroalimentación Fisiológica/fisiología , Femenino , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Persona de Mediana Edad , Transducción de Señal/fisiología
9.
J Phys Chem B ; 122(49): 11137-11146, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30047727

RESUMEN

PI3Kα is a key lipid kinase in the PI3K/Akt pathway. Its frequent oncogenic mutations make it a primary drug target. Calmodulin (CaM) activates PI3Kα independently of extracellular signals, indicating a significant role in oncogenic PI3Kα activation. Here, we reveal the atomic-scale structures of CaM in complexes with the nSH2 and cSH2 domains of the regulatory p85α subunit of PI3Kα, and illustrate how CaM activates PI3Kα by targeting the "soft 1-5-10" CaM-binding motifs in both nSH2 and cSH2 domains. Experiment observed CaM binding cSH2 first, followed by nSH2 binding hours later. CaM typically prefers binding helical peptides. Here we observe that, unlike in cSH2, the CaM-binding motif in nSH2 populates a mixed ß-sheet/α-helix/random coil structure. The population shift from a ß-sheet toward CaM's favored α-helical conformation explains why the nSH2 domain needs a longer time for CaM binding in the experiments. The "soft" CaM-binding motifs in both nSH2 and cSH2 domains establish strong CaM-PI3Kα interactions, collectively facilitating PI3Kα activation. This work uncovers the structural basis for CaM-driven PI3Kα activation.


Asunto(s)
Secuencias de Aminoácidos , Calmodulina/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Dominios Homologos src , Calmodulina/química , Fosfatidilinositol 3-Quinasa Clase I/química , Humanos , Simulación de Dinámica Molecular , Conformación Proteica
10.
Endocr J ; 65(9): 943-952, 2018 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-29998909

RESUMEN

Type 1 diabetes mellitus (T1DM) is a syndrome of loss of glucose homeostasis caused by the loss of ß cell chronic autoimmunity against islet cells. Islet-specific epitopes coupled antigen presenting cells by Ethylenecarbodiimide (ECDI) is a promising strategy to induce antigen-specific tolerance. However, single epitope induced tolerance is insufficient to prevent the onset of T1DM. The aim of this study is to evaluate the efficacy of whole islet antigens in preventing the onset and progression of T1DM and identify the underlying immune mechanism in NOD mice. In this study, the whole islet antigens, derived from islet lysate isolated from BALB/c mice, were coupled to splenocytes of BALB/c mice by ECDI fixation (SP-Islet lysate), and then intravenously administrated to NOD mice. The results showed that, compared with control group, SP-Islet lysate group significantly decreased T1DM incidence and improved the survival of NOD mice. SP-Islet lysate treated mice had reduced insulitis score and autoantibody levels, and improved glucose tolerance and insulin/glucagon production. Furthermore, the effector memory T cells (TEMs) were downregulated and regulatory T cells (Tregs) were upregulated by the SP-Islet lysate treatment, with reduced populations of Th1&Th17 cells. In conclusion, ECDI-fixed splenocytes carrying whole islet antigens effectively prevented the onset of T1DM in NOD mice, via suppressing the production of autoantibodies and inducing anergy of autoreactive T cells.


Asunto(s)
Autoanticuerpos/metabolismo , Carbodiimidas/química , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/terapia , Etilenos/química , Bazo/citología , Bazo/trasplante , Linfocitos T Reguladores/patología , Animales , Antígenos/metabolismo , Reactivos de Enlaces Cruzados/química , Diabetes Mellitus Experimental/patología , Regulación hacia Abajo/inmunología , Femenino , Tolerancia Inmunológica/fisiología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Endogámicos NOD , Bazo/inmunología , Bazo/metabolismo , Fijación del Tejido
11.
Biochemistry ; 57(12): 1917-1928, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29494137

RESUMEN

Calmodulin (CaM) is a calcium sensor protein that directly interacts with the dual-specificity (lipid and protein) kinase PI3Kα through the SH2 domains of the p85 regulatory subunit. In adenocarcinomas, the CaM interaction removes the autoinhibition of the p110 catalytic subunit of PI3Kα, leading to activation of PI3Kα and promoting cell proliferation, survival, and migration. Here we demonstrate that the cSH2 domain of p85α engages its two CaM-binding motifs in the interaction with the N- and C-lobes of CaM as well as the flexible central linker, and our nuclear magnetic resonance experiments provide structural details. We show that in response to binding CaM, cSH2 exposes its tryptophan residue at the N-terminal region to the solvent. Because of the flexible nature of both CaM and cSH2, multiple binding modes of the interactions are possible. Binding of CaM to the cSH2 domain can help release the inhibition imposed on the p110 subunit, similar to the binding of the phosphorylated motif of RTK, or phosphorylated CaM (pCaM), to the SH2 domains. Amino acid sequence analysis shows that CaM-binding motifs are common in SH2 domains of non-RTKs. We speculate that CaM can also activate these kinases through similar mechanisms.


Asunto(s)
Adenocarcinoma/química , Calmodulina/química , Fosfatidilinositol 3-Quinasa Clase Ia/química , Proteínas de Neoplasias/química , Dominios Homologos src , Adenocarcinoma/metabolismo , Secuencias de Aminoácidos , Calmodulina/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo
12.
Sci Rep ; 7(1): 4351, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659588

RESUMEN

Glucagon like peptide-1 (GLP-1) plays a vital role in glucose homeostasis and sustaining ß-cell function. Currently there are two major methods to enhance endogenous GLP-1 activity; inhibiting dipeptidyl peptidase-4 (DPP4) or activating G protein-coupled receptor 119 (GPR119). Here we describe and validate a novel dual-target compound, HBK001, which can both inhibit DPP4 and activate GPR119 ex and in vivo. We show that HBK001 can promote glucose-stimulated insulin secretion in mouse and human primary islets. A single administration of HBK001 in ICR mice can increase plasma incretins levels much more efficiently than linagliptin, a classic DPP4 inhibitor. Long-term treatment of HBK001 in KKAy mice can ameliorate hyperglycemia as well as improve glucose tolerance, while linagliptin fails to achieve such glucose-lowing effects despite inhibiting 95% of serum DPP4 activity. Moreover, HBK001 can increase first-phase insulin secretion in KKAy mice, suggesting a direct effect on islet ß-cells via GPR119 activation. Furthermore, HBK001 can improve islet morphology, increase ß-cell proliferation and up-regulate genes involved in improved ß-cell function. Thus, we have identified, designed and synthesized a novel dual-target compound, HBK001, which represents a promising therapeutic candidate for type 2 diabetes, especially for patients who are insensitive to current DPP4 inhibitors.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Glucemia , AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/química , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Insulina/biosíntesis , Ratones , Transducción de Señal
13.
Trends Cancer ; 3(3): 214-224, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28462395

RESUMEN

Calmodulin (CaM) uniquely promotes signaling of oncogenic K-Ras; but not N-Ras or H-Ras. How CaM interacts with K-Ras and how this stimulates cell proliferation are among the most challenging questions in KRAS-driven cancers. Earlier data pointed to formation of a ternary complex consisting of K-Ras, PI3Kα and CaM. Recent data point to phosphorylated CaM binding to the SH2 domains of the p85 subunit of PI3Kα and activating it. Modeling suggests that the high affinity interaction between the phosphorylated CaM tyrosine motif and PI3Kα, can promote full PI3Kα activation by oncogenic K-Ras. Our up-to-date review discusses CaM's role in PI3K signaling at the membrane in KRAS-driven cancers. This is significant since it may help development of K-Ras-specific pharmacology.


Asunto(s)
Calmodulina/genética , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Calmodulina/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Transducción de Señal/genética , Dominios Homologos src/genética
14.
Nucleic Acids Res ; 44(D1): D527-35, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26365237

RESUMEN

Allosteric regulation, the most direct and efficient way of regulating protein function, is induced by the binding of a ligand at one site that is topographically distinct from an orthosteric site. Allosteric Database (ASD, available online at http://mdl.shsmu.edu.cn/ASD) has been developed to provide comprehensive information featuring allosteric regulation. With increasing data, fundamental questions pertaining to allostery are currently receiving more attention from the mechanism of allosteric changes in an individual protein to the entire effect of the changes in the interconnected network in the cell. Thus, the following novel features were added to this updated version: (i) structural mechanisms of more than 1600 allosteric actions were elucidated by a comparison of site structures before and after the binding of an modulator; (ii) 261 allosteric networks were identified to unveil how the allosteric action in a single protein would propagate to affect downstream proteins; (iii) two of the largest human allosteromes, protein kinases and GPCRs, were thoroughly constructed; and (iv) web interface and data organization were completely redesigned for efficient access. In addition, allosteric data have largely expanded in this update. These updates are useful for facilitating the investigation of allosteric mechanisms, dynamic networks and drug discoveries.


Asunto(s)
Regulación Alostérica , Bases de Datos de Proteínas , Descubrimiento de Drogas , Humanos , Internet , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Proteínas/química , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
15.
Cell Signal ; 27(7): 1369-79, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25843777

RESUMEN

Carbonic anhydrase IX(CA9)is a member of the carbonic anhydrase family that catalyzes the reversible hydration of carbon dioxide, and plays a key role in the regulation of pH. Although a large number of studies have shown that CA9 is strongly up-regulated by HIF1-α, little is known about the negative regulation mechanism of CA9 in cancer cells. Here we find that CREB is a key negative regulator of CA9 in gastric cancer. Over-expression of CREB can significantly repress the expression of CA9. Treating with anisomycin (ANS), an activator of p38, the phosphorylation and nuclear translocation of CREB are both promoted, while the transcription of CA9 is repressed. Besides, our results firstly identify that CREB can recruit SIRT1 (class III HDACS) by adaptor protein p300, then repress the expression of CA9. These findings may contribute to understand the negative regulation mechanisms of CA9 in gastric cancer.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteína de Unión a CREB/metabolismo , Anhidrasas Carbónicas/metabolismo , Neoplasias Gástricas/enzimología , Animales , Anisomicina/farmacología , Antígenos de Neoplasias/genética , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/genética , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/metabolismo , Sirtuina 1/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Transcripción Genética/efectos de los fármacos , Trasplante Heterólogo , Factores de Transcripción p300-CBP/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Bioinformatics ; 31(15): 2598-600, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810427

RESUMEN

Allostery allows for the fine-tuning of protein function. Targeting allosteric sites is gaining increasing recognition as a novel strategy in drug design. The key challenge in the discovery of allosteric sites has strongly motivated the development of computational methods and thus high-quality, publicly accessible standard data have become indispensable. Here, we report benchmarking data for experimentally determined allosteric sites through a complex process, including a 'Core set' with 235 unique allosteric sites and a 'Core-Diversity set' with 147 structurally diverse allosteric sites. These benchmarking sets can be exploited to develop efficient computational methods to predict unknown allosteric sites in proteins and reveal unique allosteric ligand-protein interactions to guide allosteric drug design.


Asunto(s)
Sitio Alostérico , Benchmarking , Diseño de Fármacos , Glucógeno Fosforilasa/metabolismo , Programas Informáticos , Regulación Alostérica , Humanos , Ligandos
17.
Cell Signal ; 26(12): 2667-73, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25152372

RESUMEN

Cell division cycle 42 (CDC42), an important member of the Ras homolog (Rho) family, plays a key role in regulating multiple cellular processes such as cell cycle progression, migration, cell cytoskeleton organization, cell fate determination and differentiation. Among the downstream effectors of CDC42, P21-activated kinases (PAKs) obtain the most attention. Although a large body of evidence indicates that CDC42/PAKs pathway plays important role in tumor growth, invasion and metastasis, the mechanism of their negative regulation remains unclear. Here, we identified CDC42, a PAKs activating factor, was a target of miR-133. Ectopic overexpression of miRNAs not only downregulated CDC42 expression and PAKs activation, but also inhibited cancer cell proliferation and migration. We also found that miR-133 was down-regulated in 180 pairs gastric cancer tissues. miR-133 expression was negatively associated with tumor size, invasion depth and peripheral organ metastasis. Besides, dysfunction of miR-133 was an independent prognosis factor for overall survival. Our findings could provide new insights into the molecular mechanisms of gastric carcinogenesis, and may help facilitating development of CDC42/PAK-based therapies for human cancer.


Asunto(s)
MicroARNs/genética , Transducción de Señal/genética , Neoplasias Gástricas/genética , Proteína de Unión al GTP cdc42/genética , Quinasas p21 Activadas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos
18.
Nucleic Acids Res ; 42(Database issue): D510-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24293647

RESUMEN

Allostery is the most direct and efficient way for regulation of biological macromolecule function and is induced by the binding of a ligand at an allosteric site topographically distinct from the orthosteric site. AlloSteric Database (ASD, http://mdl.shsmu.edu.cn/ASD) has been developed to provide comprehensive information on allostery. Owing to the inherent high receptor selectivity and lower target-based toxicity, allosteric regulation is expected to assume a more prominent role in drug discovery and bioengineering, leading to the rapid growth of allosteric findings. In this updated version, ASD v2.0 has expanded to 1286 allosteric proteins, 565 allosteric diseases and 22 008 allosteric modulators. A total of 907 allosteric site-modulator structural complexes and >200 structural pairs of orthosteric/allosteric sites in the allosteric proteins were constructed for researchers to develop allosteric site and pathway tools in response to community demands. Up-to-date allosteric pathways were manually curated in the updated version. In addition, both the front-end and the back-end of ASD have been redesigned and enhanced to allow more efficient access. Taken together, these updates are useful for facilitating the investigation of allosteric mechanisms, allosteric target identification and allosteric drug discovery.


Asunto(s)
Regulación Alostérica , Bases de Datos de Proteínas , Sitio Alostérico , Descubrimiento de Drogas , Internet , Proteínas/química , Proteínas/metabolismo
19.
Anat Rec (Hoboken) ; 296(10): 1561-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23873832

RESUMEN

The p21-activated kinases have been implicated in the control of cell cycle progression. However, the biological mechanism underlying the role of p21-activated kinase 4 (PAK4) in cell cycle control remains unknown. Here, by using quantitative RT-PCR and immunoblot analyses, we discovered that over-expression of PAK4 could suppress cyclin-dependent kinase inhibitor 1C (p57(Kip2) ) expression in the MCF-7 human breast cancer cell line, whereas lentiviral vector-mediated small interfering RNA (siRNA) knockdown of PAK4 markedly promoted p57(Kip2) expression in MCF-7 cells. Furthermore, PAK4-mediated down-regulation of p57(Kip2) was reversed by MG132, a specific proteasome inhibitor. The ubiquitination assay confirmed that the activity of PAK4 attenuated p57(Kip2) protein stability through the ubiquitin-proteasome pathway in MCF-7 cells. Moreover, a significant inverse correlation between PAK4 and p57(Kip2) protein levels was observed in breast cancer tissues by immunohistochemical analysis. Taken together, our data demonstrate a novel function for PAK4 in regulating the stability of p57(Kip2) , possibly through the ubiquitin-proteasome pathway, leading to increased proliferation of breast cancer cells. Thus, PAK4 may be used as a potential diagnostic and therapeutic target for human breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Quinasas p21 Activadas/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ubiquitinación , Quinasas p21 Activadas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...