Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 10(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35091454

RESUMEN

BACKGROUND: Circulating monocytes are functionally heterogeneous and can be divided into classical (CMo), intermediate (IMo), and non-CMo/patrolling monocyte (PMo) subsets. CMo can differentiate into PMo through IMo. PMos have been shown to inhibit cancer metastasis but the role of IMo is unclear. To date, no strategy has been developed to inhibit cancer metastasis through enhancing PMo/IMo differentiation. METHODS: We screened multiple inflammatory cytokines/chemokines activity of modulating PMo/IMo associated cell markers expression using human monocyte in vitro culture system. We tested our candidate cytokine activity in vivo using multiple mice models. We identified critical key factors and cytokines for our candidate cytokine activity by using gene-knockout mice and neutralization antibodies. RESULTS: We identified IFN-γ as a candidate inflammatory cytokine in the regulation of human IMo/PMo marker expression. Our in vivo data demonstrated that IMo expansion was induced by short-term (3 days) IFN-γ treatment through increasing CMo-IMo differentiation and blocking IMo-PMo differentiation. The IMo induced by IFN-γ (IFN-IMo), but not IFN-γ activated CMo (IFN-CMo), inhibited cancer metastasis by 90%. Surprizing, the effect of IFN-γ is greater in PMo deficiency mice, indicating the effect of IFN-IMo is not mediated through further differentiation into PMo. We also found that IFN-IMos induced by short-term IFN-γ treatment robustly boosted NK cell expansion for threefold and promoted NK differentiation and function through IL-27 and CXCL9. Furthermore, we identified that FOXO1, a key molecule controlling cellular energy metabolism, mediated the effect of IFN-γ induced IL-27 expression, and that NR4A1, a key molecule controlling PMo differentiation and inhibiting cancer metastasis, inhibited the pro-NK cell and anti-metastasis activity of IFN-IMo by suppressing CXCL9 expression. CONCLUSIONS: We have discovered the antimetastasis and pro-NK cell activity of IFN-IMo, identified FOXO1 as a key molecule for IFN-γ driven monocyte differentiation and function, and found NR4A1 as an inhibitory molecule for IFN-IMo activity. Our study has not only shown novel mechanisms for a classical antitumor cytokine but also provided potential target for developing superior monocytic cell therapy against cancer metastasis.


Asunto(s)
Proteína Forkhead Box O1/fisiología , Interferón gamma/farmacología , Interleucina-27/fisiología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Monocitos/efectos de los fármacos , Metástasis de la Neoplasia/prevención & control , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Monocitos/fisiología , Factor de Transcripción STAT1/fisiología
2.
Chem Biol Drug Des ; 99(2): 344-361, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34862852

RESUMEN

Diabetes mellitus type 2 (T2D) is one of the metabolic disorders suffered by a global human being. Certain factors, such as lifestyle and heredity, can increase a person's tendency for T2D. Various genes and proteins play a role in the development of insulin resistance and ultimately diabetes in which one central protein that is discussed in this review is FoxO1. In this review, we regard FoxO1 activation as detrimental, promote high plasma glucose level, and induce insulin resistance. Indeed, many contrasting studies arise since FoxO1 is an important protein to alleviate oxidative stress and promote cell survival, for example, also by preventing hyperglycemic-induced cell death. Inter-relation to PPARG, another important protein in metabolism, is also discussed. Ultimately, we discussed contrasting approaches of targeting FoxO1 to combat diabetes mellitus by small molecules.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Proteína Forkhead Box O1/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteína Forkhead Box O1/efectos de los fármacos , Humanos , Resistencia a la Insulina , Estrés Oxidativo , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
3.
Endocrinology ; 163(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34971379

RESUMEN

Understanding the molecular mechanisms underlying pituitary organogenesis and function is essential for improving therapeutics and molecular diagnoses for hypopituitarism. We previously found that deletion of the forkhead factor, Foxo1, in the pituitary gland early in development delays somatotrope differentiation. While these mice grow normally, they have reduced growth hormone expression and free serum insulin-like growth factor-1 (IGF1) levels, suggesting a defect in somatotrope function. FOXO factors show functional redundancy in other tissues, so we deleted both Foxo1 and its closely related family member, Foxo3, from the primordial pituitary. We find that this results in a significant reduction in growth. Consistent with this, male and female mice in which both genes have been deleted in the pituitary gland (dKO) exhibit reduced pituitary growth hormone expression and serum IGF1 levels. Expression of the somatotrope differentiation factor, Neurod4, is reduced in these mice. This suggests a mechanism underlying proper somatotrope function is the regulation of Neurod4 expression by FOXO factors. Additionally, dKO mice have reduced Lhb expression and females also have reduced Fshb and Prl expression. These studies reveal FOXO transcription factors as important regulators of pituitary gland function.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Somatotrofos/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Femenino , Proteína Forkhead Box O1/deficiencia , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/fisiología , Proteína Forkhead Box O3/deficiencia , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/fisiología , Expresión Génica , Hormona del Crecimiento/genética , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Ratones , Ratones Noqueados , Hipófisis/química , Hipófisis/fisiología , ARN Mensajero/análisis , Somatotrofos/química
4.
Cell Rep ; 37(9): 110048, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34852224

RESUMEN

Intraluminal lymphatic valves (LVs) contribute to the prevention of lymph backflow and maintain circulatory homeostasis. Several reports have investigated the molecular mechanisms which promote LV formation; however, the way in which they are suppressed is not completely clear. We show that the forkhead transcription factor FOXO1 is a suppressor of LV formation and maintenance in lymphatic endothelial cells. Oscillatory shear stress by bidirectional flow inactivates FOXO1 via Akt phosphorylation, resulting in the upregulation of a subset of LV-specific genes mediated by downregulation of a transcriptional repressor, PRDM1. Mice with an endothelial-specific Foxo1 deletion have an increase in LVs, and overexpression of Foxo1 in mice produces a decrease in LVs. Genetic reduction of PRDM1 rescues the decrease in LV by Foxo1 overexpression. In conclusion, FOXO1 plays a critical role in lymph flow homeostasis by preventing excess LV formation. This gene might be a therapeutic target for lymphatic circulatory abnormalities.


Asunto(s)
Proteína Forkhead Box O1/fisiología , Linfangiogénesis , Vasos Linfáticos/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Animales , Femenino , Humanos , Vasos Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Transducción de Señal
5.
Theranostics ; 11(18): 8624-8639, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34522203

RESUMEN

Rationale: The rennin-angiotensin-aldosterone system (RAAS) plays a critical role in the pathogenesis of diabetic cardiomyopathy, but the role of a member of RAAS, angiotensin IV (Ang IV), in this disease and its underlying mechanism are unclear. This study was aimed to clarify the effects of Ang IV and its downstream mediator forkhead box protein O1 (FoxO1) on diabetic cardiomyopathy. Methods:In vivo, diabetic mice were treated with low-, medium- and high-dose Ang IV, AT4R antagonist divalinal, FoxO1 inhibitor AS1842856 (AS), or their combinations. In vitro, H9C2 cardiomyocytes and cardiac fibroblasts were treated with different concentrations of glucose, low-, medium- and high-dose Ang IV, divalinal, FoxO1-overexpression plasmid (FoxO1-OE), AS, or their combinations. Results: Ang IV treatment dose-dependently attenuated left ventricular dysfunction, fibrosis, and myocyte apoptosis in diabetic mice. Besides, enhanced autophagy and FoxO1 protein expression by diabetes were dose-dependently suppressed by Ang IV treatment. However, these cardioprotective effects of Ang IV were completely abolished by divalinal administration. Bioinformatics analysis revealed that the differentially expressed genes were enriched in autophagy, apoptosis, and FoxO signaling pathways among control, diabetes, and diabetes+high-dose Ang IV groups. Similar to Ang IV, AS treatment ameliorated diabetic cardiomyopathy in mice. In vitro, high glucose stimulation increased collagen expression, apoptosis, overactive autophagy flux and FoxO1 nuclear translocation in cardiomyocytes, and upregulated collagen and FoxO1 expression in cardiac fibroblasts, which were substantially attenuated by Ang IV treatment. However, these protective effects of Ang IV were completely blocked by the use of divalinal or FoxO1-OE, and these detrimental effects were reversed by the additional administration of AS. Conclusions: Ang IV treatment dose-dependently attenuated left ventricular dysfunction and remodeling in a mouse model of diabetic cardiomyopathy, and the mechanisms involved stimulation of AT4R and suppression of FoxO1-mediated fibrosis, apoptosis, and overactive autophagy.


Asunto(s)
Angiotensina II/análogos & derivados , Cardiomiopatías Diabéticas/fisiopatología , Proteína Forkhead Box O1/metabolismo , Angiotensina II/metabolismo , Angiotensina II/farmacología , Angiotensina II/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Autofagia/efectos de los fármacos , Autofagia/fisiología , Glucemia , Línea Celular , Diabetes Mellitus Experimental , Modelos Animales de Enfermedad , Fibrosis/fisiopatología , Proteína Forkhead Box O1/fisiología , Glucosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Quinolonas/farmacología , Transducción de Señal , Estreptozocina/farmacología , Disfunción Ventricular Izquierda
6.
J Clin Invest ; 131(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34428182

RESUMEN

Insulin and IGF-1 are essential for adipocyte differentiation and function. Mice lacking insulin and IGF-1 receptors in fat (FIGIR-KO, fat-specific IGF-1 receptor and insulin receptor-KO) exhibit complete loss of white and brown adipose tissue (WAT and BAT), glucose intolerance, insulin resistance, hepatosteatosis, and cold intolerance. To determine the role of FOXO transcription factors in the altered adipose phenotype, we generated FIGIR-KO mice with fat-specific KO of fat-expressed Foxos [Foxo1, Foxo3, Foxo4] (F-Quint-KO). Unlike FIGIR-KO mice, F-Quint-KO mice had normal BAT, glucose tolerance, insulin-regulated hepatic glucose production, and cold tolerance. However, loss of FOXOs only partially rescued subcutaneous WAT and hepatosteatosis, did not rescue perigonadal WAT or systemic insulin resistance, and led to even more marked hyperinsulinemia. Thus, FOXOs play different roles in insulin/IGF-1 action in different adipose depots, being most important in BAT, followed by subcutaneous WAT and then by visceral WAT. Disruption of FOXOs in fat also led to a reversal of insulin resistance in liver, but not in skeletal muscle, and an exacerbation of hyperinsulinemia. Thus, adipose FOXOs play a unique role in regulating crosstalk between adipose depots, liver, and ß cells.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Proteína Forkhead Box O1/fisiología , Insulina/farmacología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Metabolismo Energético , Glucosa/metabolismo , Insulina/sangre , Células Secretoras de Insulina/patología , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Receptor IGF Tipo 1/fisiología , Receptor de Insulina/fisiología
7.
Diabetes ; 70(7): 1536-1548, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33597203

RESUMEN

Exosomes have been implicated in diabetic kidney disease (DKD), but the regulation of exosomes in DKD is largely unknown. Here, we have verified the decrease of exosome secretion in DKD and unveiled the underlying mechanism. In Boston University mouse proximal tubule (BUMPT) cells, high-glucose (HG) treatment led to a significant decrease in exosome secretion, which was associated with specific downregulation of RAB27B, a key guanosine-5'-triphosphatase in exosome secretion. Overexpression of RAB27B restored exosome secretion in HG-treated cells, suggesting a role of RAB27B downregulation in the decrease of exosome secretion in DKD. To understand the mechanism of RAB27B downregulation, we conducted bioinformatics analysis that identified FOXO1 binding sites in the Rab27b gene promoter. Consistently, HG induced phosphorylation of FOXO1 in BUMPT cells, preventing FOXO1 accumulation and activation in the nucleus. Overexpression of nonphosphorylatable, constitutively active FOXO1 led to the upregulation of RAB27B and an increase in exosome secretion in HG-treated cells. In vivo, compared with normal mice, diabetic mice showed increased FOXO1 phosphorylation, decreased RAB27B expression, and reduced exosome secretion. Collectively, these results unveil the mechanism of exosome dysfunction in DKD where FOXO1 is phosphorylated and inactivated in DKD, resulting in RAB27B downregulation and the decrease of exosome secretion.


Asunto(s)
Nefropatías Diabéticas/etiología , Exosomas/fisiología , Proteína Forkhead Box O1/fisiología , Proteínas de Unión al GTP rab/fisiología , Animales , Células Cultivadas , Regulación hacia Abajo , Tasa de Filtración Glomerular , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación
8.
Med Sci Monit ; 27: e928232, 2021 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-33589581

RESUMEN

BACKGROUND Studies have shown that diabetes mellitus (DM) has a negative impact on male reproductive function, which may lead to changes in the testis and epididymis and a decline in semen quality. MATERIAL AND METHODS We performed animal experiments with 6 diabetic db/db mice as the model group (group B) and 6 C57BL/6J mice as the control group (group A). After adaptive feeding for 7 days, the sperm quality of each group was measured. Concurrently, the morphology of the mouse testis was observed by hematoxylin-eosin (H&E) staining. The expression of the PI3K, Akt, FoxO1, FasL, IL-6, and Stat3 proteins and mRNAs in the testicular tissue was detected by western blotting and RT-qPCR. RESULTS The number of spermatozoa and sperm motility of group A was significantly higher than that of group B (P<0.05). H&E staining of the testicular tissue showed the seminiferous tubules in group B mice were damaged to varying degrees and the seminiferous tubules were sparsely arranged. Compared with those of group A, the expression levels of PI3K, Akt, and Stat3 proteins and mRNAs in group B were significantly lower (P<0.05), while the expression levels of FoxO1, FasL, and IL-6 proteins and mRNAs in group B mice were significantly higher (P<0.05). CONCLUSIONS This study demonstrated that DM inhibited the expression of PI3K, Akt, and Stat3 proteins and mRNAs in the FoxO1 pathway and promoted the expression of FoxO1, FasL, and IL-6 proteins and mRNAs, leading to abnormal apoptosis of testicular tissue cells and functional damage, and eventually spermatogenic dysfunction.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Proteína Forkhead Box O1/metabolismo , Espermatozoides/metabolismo , Animales , Apoptosis/efectos de los fármacos , China , Diabetes Mellitus/metabolismo , Diabetes Mellitus Experimental , Proteína Forkhead Box O1/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Análisis de Semen , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Recuento de Espermatozoides , Motilidad Espermática/fisiología , Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos
10.
Front Endocrinol (Lausanne) ; 11: 572981, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133019

RESUMEN

Glucocorticoids (GCs) and the glucocorticoid receptor (GR) are important regulators of development, inflammation, stress response and metabolism, demonstrated in various diseases including Addison's disease, Cushing's syndrome and by the many side effects of prolonged clinical administration of GCs. These conditions include severe metabolic challenges in key metabolic organs like the liver. In the liver, GR is known to regulate the transcription of key enzymes in glucose and lipid metabolism and contribute to the regulation of circadian-expressed genes. Insights to the modes of GR regulation and the underlying functional mechanisms are key for understanding diseases and for the development of improved clinical uses of GCs. The activity and function of GR is regulated at numerous levels including ligand availability, interaction with heat shock protein (HSP) complexes, expression of GR isoforms and posttranslational modifications. Moreover, recent genomics studies show functional interaction with multiple transcription factors (TF) and coregulators in complex transcriptional networks controlling cell type-specific gene expression by GCs. In this review we describe the different regulatory steps important for GR activity and discuss how different TF interaction partners of GR selectively control hepatic gene transcription and metabolism.


Asunto(s)
Redes Reguladoras de Genes , Hígado/metabolismo , Receptores de Glucocorticoides/fisiología , Factores de Transcripción/fisiología , Transporte Activo de Núcleo Celular , Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos/fisiología , Proteína Forkhead Box O1/fisiología , Humanos , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Factor de Transcripción STAT5/fisiología , Transducción de Señal/fisiología
11.
Mol Cell Endocrinol ; 517: 110871, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32450284

RESUMEN

FOXOs belong to the forkhead transcription factor superfamily, several of which are suggested to be involved in the control of food intake. Previously, we proved that the neuropeptide FF (NPFF) peptide was involved in feeding regulation in spotted sea bass. In the present study, seven members of the foxo family were identified in the whole genome of spotted sea bass. The distributions of these genes in different tissues were analyzed by qRT-PCR. Variations in the foxo1a and npff expression profiles during short-term starvation showed similar expression patterns. The colocalization of foxo1a and npff in the telencephalon, hypothalamus, stomach and intestine further provided evidence that foxo1a may act directly to promote the transcription of npff. Thirteen predicted FOXO1 binding sites were found in the 5' upstream region of npff. Luciferase assay results showed that FOXO1A was able to activate npff transcriptional responses by directly binding DNA response elements, and the key regulatory areas and sites of FOXO1A on the npff promoter were confirmed by deletion and site-directed mutagenesis analyses. These findings may help to elucidate the role of FOXO1 in the regulation of feeding processes in teleosts.


Asunto(s)
Lubina/genética , Conducta Alimentaria , Proteína Forkhead Box O1/fisiología , Regulación de la Expresión Génica , Oligopéptidos/biosíntesis , Animales , Secuencia de Bases , Sitios de Unión , Química Encefálica , Células Cultivadas , Secuencia Conservada , Tracto Gastrointestinal/química , Genes Reporteros , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , Oligopéptidos/genética , Especificidad de Órganos , Filogenia , Regiones Promotoras Genéticas , Dominios Proteicos , Distribución Aleatoria , Proteínas Recombinantes/metabolismo , Inanición/genética , Transcripción Genética , Vertebrados/genética
13.
J Orthop Res ; 38(1): 43-58, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31424116

RESUMEN

Injuries to flexor tendons can be complicated by fibrotic adhesions, which severely impair the function of the hand. Plasminogen activator inhibitor 1 (PAI-1/SERPINE1), a master suppressor of fibrinolysis and protease activity, is associated with adhesions. Here, we used next-generation RNA sequencing (RNA-Seq) to assess genome-wide differences in messenger RNA expression due to PAI-1 deficiency after zone II flexor tendon injury. We used the ingenuity pathway analysis to characterize molecular pathways and biological drivers associated with differentially expressed genes (DEG). Analysis of hundreds of overlapping and DEG in PAI-1 knockout (KO) and wild-type mice (C57Bl/6J) during tendon healing revealed common and distinct biological processes. Pathway analysis identified cell proliferation, survival, and senescence, as well as chronic inflammation as potential drivers of fibrotic healing and adhesions in injured tendons. Importantly, we identified the activation of PTEN signaling and the inhibition of FOXO1-associated biological processes as unique transcriptional signatures of the healing tendon in the PAI-1/Serpine1 KO mice. Further, transcriptomic differences due to the genetic deletion of PAI-1 were mechanistically linked to PI3K/Akt/mTOR, PKC, and MAPK signaling cascades. These transcriptional observations provide novel insights into the biological roles of PAI-1 in tendon healing and could identify therapeutic targets to achieve scar-free regenerative healing of tendons. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:43-58, 2020.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/fisiología , Traumatismos de los Tendones/fisiopatología , Transcriptoma , Cicatrización de Heridas , Animales , Proteína Forkhead Box O1/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/fisiología , Proteína Quinasa C/fisiología
14.
Cell Syst ; 9(6): 580-588.e4, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31838146

RESUMEN

Cells sense and respond to signals in their local environment by activating signaling cascades that lead to phenotypic changes. Differences in these signals can be discriminated at the population level; however, single cells have been thought to be limited in their capacity to distinguish ligand doses due to signaling noise. We describe here the rational development of a genetically encoded FoxO1 sensor, which serves as a down-stream readout of insulin growth factor-phosphatidylinositol 3-kinase IGF-PI3K-AKT signaling pathway activity. With this reporter, we tracked individual cell responses to multiple IGF-I doses, pathway inhibitors, and repeated treatments. We observed that individual cells can discriminate multiple IGF-I doses, and these responses are sustained over time, are reproducible at the single-cell level, and display cell-to-cell heterogeneity. These studies imply that cell-to-cell variation in signaling responses is biologically meaningful and support the endeavor to elucidate mechanisms of cell signaling at the level of the individual cell.


Asunto(s)
Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Análisis de la Célula Individual/métodos , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/fisiología , Células HeLa , Humanos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Somatomedinas/metabolismo
15.
Sci Rep ; 9(1): 10181, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31308441

RESUMEN

Exposure to maternal diabetes during pregnancy results in diabetes in offspring, but its underlying mechanisms are unclear. Here, we investigated the phenotype and molecular defects of the offspring of poorly controlled diabetic female mice generated by streptozotocin (STZ) administration. Offspring was exposed to maternal diabetes during pregnancy and lactation. The body weight of STZ offspring was lower than that of control offspring at birth and in adulthood, and glucose tolerance was impaired in adult STZ offspring. Interestingly, the phenotype was more pronounced in male offspring. We next investigated the morphology of islets and expression of ß cell-related genes, but no significant changes were observed. However, transcriptome analysis of the liver revealed activation of the fork head box protein O1 (Foxo1) pathway in STZ male offspring. Notably, two key gluconeogenesis enzyme genes, glucose 6 phosphatase catalytic subunit (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1), were upregulated. Consistent with this finding, phosphorylation of Foxo1 was decreased in the liver of STZ male offspring. These changes were not obvious in female offspring. The activation of Foxo1 and gluconeogenesis in the liver may have contributed to the impaired glucose tolerance of STZ male offspring.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Proteína Forkhead Box O1/metabolismo , Intolerancia a la Glucosa/etiología , Animales , Glucemia/metabolismo , Diabetes Gestacional/metabolismo , Femenino , Proteína Forkhead Box O1/fisiología , Gluconeogénesis/genética , Intolerancia a la Glucosa/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Lactancia/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Embarazo , Estreptozocina/farmacología
16.
EMBO J ; 38(11)2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31015337

RESUMEN

In contrast to other B-cell antigen receptor (BCR) classes, the function of IgD BCR on mature B cells remains largely elusive as mature B cells co-express IgM, which is sufficient for development, survival, and activation of B cells. Here, we show that IgD expression is regulated by the forkhead box transcription factor FoxO1, thereby shifting the responsiveness of mature B cells towards recognition of multivalent antigen. FoxO1 is repressed by phosphoinositide 3-kinase (PI3K) signaling and requires the lipid phosphatase Pten for its activation. Consequently, Pten-deficient B cells expressing knock-ins for BCR heavy and light chain genes are unable to upregulate IgD. Furthermore, in the presence of autoantigen, Pten-deficient B cells cannot eliminate the autoreactive BCR specificity by secondary light chain gene recombination. Instead, Pten-deficient B cells downregulate BCR expression and become unresponsive to further BCR-mediated stimulation. Notably, we observed a delayed germinal center (GC) reaction by IgD-deficient B cells after immunization with trinitrophenyl-ovalbumin (TNP-Ova), a commonly used antigen for T-cell-dependent antibody responses. Together, our data suggest that the activation of IgD expression by Pten/FoxO1 results in mature B cells that are selectively responsive to multivalent antigen and are capable of initiating rapid GC reactions and T-cell-dependent antibody responses.


Asunto(s)
Linfocitos B/fisiología , Centro Germinal/fisiología , Inmunoglobulina D/genética , Fosfohidrolasa PTEN/fisiología , Receptores de Antígenos de Linfocitos B/genética , Animales , Células Cultivadas , Proteína Forkhead Box O1/fisiología , Regulación de la Expresión Génica/inmunología , Centro Germinal/metabolismo , Inmunoglobulina D/inmunología , Inmunoglobulina D/metabolismo , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología
17.
Biochem Biophys Res Commun ; 513(4): 1019-1026, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31010677

RESUMEN

Dexamethasone (DEX), a widely used glucocorticoid with strong anti-inflammatory and immunosuppressive activities, has been reported to induce apoptosis in osteoblasts, but the underlying mechanisms are still not comprehensively investigated. FOXO1 plays an important role in the regulation of cell proliferation and apoptosis. Our study aims to explore the role of FOXO1 in DEX-induced apoptosis of osteoblastic MC3T3-E1 cells through bioinformatics and experiments. We first employed bioinformatics to identify DEX-related genes and revealed their functions by GO enrichment analysis including FOXO1 associated biological processes. Expression level of FOXO1 was validated by GEO data. Then, experiments were performed to verify the hypothesis. CCK8 was used to detect cell viability and apoptosis was detected by flow cytometry. SiRNA was used to silence FOXO1 and western-blot was employed to detect protein expression. Results demonstrated DEX-related genes involved in cell proliferation, apoptosis and angiogenesis and FOXO1 was a regulator of apoptosis. DEX could up-regulate FOXO1 expression, inhibit cell viability, promote apoptosis. SiRNA-FOXO1 could attenuate DEX-induced apoptosis in MC3T3-E1. These findings suggested DEX could affect some vital biological processes of MC3T3-E1 and FOXO1 played an essential role in DEX-induced apoptosis in MC3T3-E1.


Asunto(s)
Apoptosis/efectos de los fármacos , Dexametasona/farmacología , Proteína Forkhead Box O1/fisiología , Osteoblastos/patología , Células 3T3 , Animales , Proteína Forkhead Box O1/genética , Silenciador del Gen , Ratones , Osteoblastos/metabolismo , ARN Interferente Pequeño/farmacología , Regulación hacia Arriba/efectos de los fármacos
18.
Aging (Albany NY) ; 10(9): 2338-2355, 2018 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-30222592

RESUMEN

Aging is characterized by a reduced ability to defend against stress, an inability to maintain homeostasis, and an increased risk of disease. In this study, a metabolomics approach was used to identify novel metabolic pathways that are perturbed in a mouse model of accelerated aging (SAMP1/kl-/-) and to gain new insights into the metabolic associations of the metformin derivative HL156A. Extensive inflammation and calcification were observed in the tissues of the SAMP1/kl-/- mice with premature aging. In mouse embryonic fibroblasts (MEFs) obtained from SAMP1/kl-/- mice, we observed that HL156A induced FOXO1 expression through inhibition of the IGF-1/AKT/mTOR signaling pathways. Treatment of HL156A decreased reactive oxygen species production and enhanced mitochondrial transmembrane potential in SAMP1/kl-/- MEFs. A metabolomic profile analysis showed that HL156A increased the GSH/GSSG ratio in the kidneys of SAMP1/kl-/- mice (8-12 weeks old). In addition, treating SAMP1/kl-/- mice with HL156A (30 mg/kg) for 4 weeks improved survival and decreased the significant elevation of oxidized GSH (GSSG) that was observed in SAMP1/kl-/- mice. In histological sections, HL156A administered SAMP1/kl-/- mice exhibited a decrease in excessive calcification. Based on these findings, we conclude that the new metformin derivative HL156A may inhibit oxidative damage by inducing glutathione metabolism and antioxidant pathways.


Asunto(s)
Antioxidantes/metabolismo , Glucuronidasa/fisiología , Guanidinas/farmacología , Proteínas de la Membrana/fisiología , Proteínas Nucleares/fisiología , Pirrolidinas/farmacología , Envejecimiento Prematuro , Animales , Proteína Forkhead Box O1/fisiología , Glutatión/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Riñón/metabolismo , Proteínas Klotho , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de la Membrana/genética , Metabolómica , Ratones , Proteínas Nucleares/genética , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/fisiología
19.
Arch Gynecol Obstet ; 298(5): 961-969, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206735

RESUMEN

PURPOSE: The prevalence of endometrial cancer (EC) is increasing worldwide. Progestin therapy is effective for both early stage EC patients who require preserving fertility and advanced or recurrent patients. Progestin resistance resulting from downregulation of progesterone receptor (PR) remains a major problem, and its mechanism is currently unclear. It was demonstrated that Sirtuin 1 (SIRT1), forkhead transcription factor 1 (FoxO1) and sterol regulatory element binding protein-1 (SREBP-1) may act as a pathway and play crucial roles in the development of EC in our previous studies. In the present study, we investigated the effect on the development of progestin resistance and the relationship with PR of SIRT1/FoxO1/SREBP-1. METHODS: A progestin-resistant Ishikawa cell line was established in the stimulation and selection of medroxyprogesterone acetate (MPA), and the resistance was analyzed by MTT assay, flow cytometry, and Transwell invasion assay. qRT-PCR and western blotting were conducted to detect the expression of SIRT1, FoxO1, SREBP-1 and PR. SIRT1 knockdown progestin-resistant cells were established by lentiviral transduction. RESULTS: The new progestin-resistant cell line presented sufficient resistance to MPA in aspects of proliferation, distribution of cell cycle and apoptosis compared with original Ishikawa cells. Besides, the invasion capability of progestin-resistant cells was observably increased. In both protein and mRNA levels, SIRT1 and SREBP-1 were upregulated in progestin-resistant cells, while PR and FoxO1 were downregulated. SIRT1 was knocked down by lentivirus transfection in progestin-resistant cells, resulting in upregulation of PR, FoxO1 and downregulation of SREBP-1, thereby SIRT1 knockdown cells were more sensitive to MPA compared with progestin-resistant cells. CONCLUSION: SIRT1/FoxO1/SREBP-1 act as a pathway targeting PR and involve in the development of progestin resistance in Ishikawa cells.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Neoplasias Endometriales/tratamiento farmacológico , Proteína Forkhead Box O1/fisiología , Progestinas/uso terapéutico , Sirtuina 1/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Proteína Forkhead Box O1/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Acetato de Medroxiprogesterona/uso terapéutico , Progestinas/farmacología , ARN Mensajero/análisis , Receptores de Progesterona/genética , Sirtuina 1/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
20.
Immunity ; 48(2): 313-326.e5, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29396161

RESUMEN

Positive selection of germinal center (GC) B cells is driven by B cell receptor (BCR) affinity and requires help from follicular T helper cells. The transcription factors c-Myc and Foxo1 are critical for GC B cell selection and survival. However, how different affinity-related signaling events control these transcription factors in a manner that links to selection is unknown. Here we showed that GC B cells reprogram CD40 and BCR signaling to transduce via NF-κB and Foxo1, respectively, whereas naive B cells propagate both signals downstream of either receptor. Although either BCR or CD40 ligation induced c-Myc in naive B cells, both signals were required to highly induce c-Myc, a critical mediator of GC B cell survival and cell cycle reentry. Thus, GC B cells rewire their signaling to enhance selection stringency via a requirement for both antigen receptor- and T cell-mediated signals to induce mediators of positive selection.


Asunto(s)
Antígenos CD40/fisiología , Centro Germinal/inmunología , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Receptores de Antígenos de Linfocitos B/fisiología , Transducción de Señal/fisiología , Animales , Proteína Forkhead Box O1/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Quinasa Syk/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...