Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Int J Mol Sci ; 24(17)2023 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-37686468

RESUMEN

Both FoxO transcription factors and the circadian clock act on the interface of metabolism and cell cycle regulation and are important regulators of cellular stress and stem cell homeostasis. Importantly, FoxO3 preserves the adult neural stem cell population by regulating cell cycle and cellular metabolism and has been shown to regulate circadian rhythms in the liver. However, whether FoxO3 is a regulator of circadian rhythms in neural stem cells remains unknown. Here, we show that loss of FoxO3 disrupts circadian rhythmicity in cultures of neural stem cells, an effect that is mediated via regulation of Clock transcriptional levels. Using Rev-Erbα-VNP as a reporter, we then demonstrate that loss of FoxO3 does not disrupt circadian rhythmicity at the single cell level. A meta-analysis of published data revealed dynamic co-occupancy of multiple circadian clock components within FoxO3 regulatory regions, indicating that FoxO3 is a Clock-controlled gene. Finally, we examined proliferation in the hippocampus of FoxO3-deficient mice and found that loss of FoxO3 delayed the circadian phase of hippocampal proliferation, indicating that FoxO3 regulates correct timing of NSC proliferation. Taken together, our data suggest that FoxO3 is an integral part of circadian regulation of neural stem cell homeostasis.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Proteína Forkhead Box O3 , Células-Madre Neurales , Animales , Ratones , Ciclo Celular , División Celular , Relojes Circadianos/genética , Ritmo Circadiano/genética , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/fisiología
2.
Kaohsiung J Med Sci ; 38(2): 87-96, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34529353

RESUMEN

Pyroptosis is a novel proinflammatory programmed cell death process. This study was designed to investigate the functional mechanisms of long noncoding RNA growth arrest-specific transcript 5 (lncRNA GAS5) on lipopolysaccharide (LPS)-induced human bronchial epithelial cell (HBEC) pyroptosis. LPS was used to induce pyroptosis in HBECs, followed by the detection of the expression of GAS5, forkhead box O3 (FOXO3), and nuclear factor E2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signaling pathway-related factors. Cell viability was evaluated using CCK-8 assay, lactate dehydrogenase (LDH) release was assessed by LDH assay kit and caspase-1 activity by flow cytometry. Furthermore, expression of NOD-like receptor family pyrin domain containing 3 and pyroptosis-related proteins was evaluated using Western blot analysis, while enzyme-linked immunosorbent assay was used to determine the levels of inflammatory factors. The interaction between GAS5 and FOXO3 was confirmed using bioinformatic prediction, RNA immunoprecipitation assay, RNA pull-down, and dual-luciferase reporter gene assay. Treatment of HBECs with LPS upregulated the expression of GAS5 and FOXO3, resulting in the inactivation of the Nrf2/HO-1 signaling pathway. On the other hand, inhibition of both GAS5 and FOXO3 promoted cell viability, reduced LDH release, pyroptosis, and inflammatory response in LPS-induced HBECs. Furthermore, FOXO3 could interact with GAS5, while FOXO3 overexpression reversed the inhibitory effect of GAS5 knockdown on cell pyroptosis. Thus, mechanistically, inhibition of FOXO3 activates the Nrf2/HO-1 pathway to suppress LPS-induced pyroptosis in HBECs. This study revealed that GAS5 knockdown attenuates FOXO3 expression thereby activating the Nrf2/HO-1 pathway to inhibit LPS-induced pyroptosis in HBECs. These findings may contribute to identifying novel targets that inhibit pyroptosis in HBECs.


Asunto(s)
Bronquios/citología , Células Epiteliales , Proteína Forkhead Box O3/fisiología , Piroptosis , ARN Largo no Codificante/fisiología , ARN Nucleolar Pequeño/genética , Mucosa Respiratoria/citología , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Humanos , Lipopolisacáridos/farmacología , Piroptosis/efectos de los fármacos
3.
Life Sci ; 291: 120258, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34952043

RESUMEN

AIMS: Enhancer of zeste homolog 2 (EZH2), a histone H3 lysine 27 methyltransferase, has been shown to play a role in kidney diseases. However, its role in hyperoxaluria-induced renal tubular epithelial cells (TECs) injury remains unclear. MATERIALS AND METHODS: A hyperoxaluria rat model was established by providing 0.5% ammonium chloride and drinking water containing 1% ethylene glycol. TECs were exposed to oxalate stress. The 3-DZNeP, a selective EZH2 inhibitor, was administered in vivo and in vitro. Cell viability, ROS production, and apoptosis ratio were evaluated. Crystal deposition was detected by Von Kossa staining and kidney tissue injury was detected by HE staining and TUNEL. EZH2, H3K27me3, cleaved-caspase3, IL-6, and MCP-1 were examined by western blot or immunohistochemistry. KEY FINDINGS: Inhibition of EZH2 by 3-DZNeP significantly attenuated hyperoxaluria-induced oxidative and inflammatory injury and CaOx crystal deposition in vivo. Similarly, inhibition of EZH2 using 3-DZNeP or shRNA restored cell viability, suppressed LDH release and the production of intracellular ROS in vitro. Furthermore, the MAPK signaling pathway and FoxO3a levels were activated or elevated in TECs exposed to oxalate. EZH2 inhibition using 3-DZNeP blocked these effects. CC90003 (ERK inhibitor) or SB203580 (p38 inhibitor) did not significantly affect the expression of FoxO3a in TECs treated with 3-DZNeP and oxalate; only SP600125 (JNK inhibitor) significantly decreased FoxO3a expression. SIGNIFICANCE: EZH2 inhibition protects against oxalate-induced TECs injury and reduces CaOx crystal deposition in the kidney may by modulating the JNK/FoxO3a pathway; EZH2 may be a promising therapeutic target in TECs injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Hiperoxaluria/metabolismo , Lesión Renal Aguda/fisiopatología , Animales , Apoptosis/efectos de los fármacos , China , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Células Epiteliales/metabolismo , Proteína Forkhead Box O3/fisiología , Hiperoxaluria/fisiopatología , Riñón/metabolismo , Enfermedades Renales/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
4.
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
5.
J Neuroimmunol ; 361: 577752, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34715591

RESUMEN

Neuromyelitis Optica (NMO) is an autoimmune inflammatory disease that affects the optic nerves and spinal cord. The autoantibody is generated against the abundant water channel protein of the brain, Aquaporin 4 (AQP4). Of the two isoforms of AQP4, the shorter one (M23) often exists as a supramolecular assembly known as an orthogonal array of particles (OAPs). There have been debates about the fate of these AQP4 clusters upon binding to the antibody, the exact mechanism of its turnover, and the proteins associated with the process. Recently several clinical cases of NMO were reported delineating the effect of Rituximab (RTX) therapy. Extending these reports at the cell signaling level, we developed a glioma based cellular model that mimicked antibody binding and helped us track the subsequent events including a variation of AQP4 levels, alterations in cellular morphology, and the changes in downstream signaling cascades. Our results revealed the extent of perturbations in the signaling pathways related to stress involving ERK, JNK, and AKT1 together with markers for cell death. We could also decipher the possible routes of degradation of AQP4, post-exposure to antibody. We further investigated the effect of autoantibody on AQP4 transcriptional level and involvement of FOXO3a and miRNA-145 in the regulation of transcription. This study highlights the differential outcome at the cellular level when treated with the serum of the same patient pre and post RTX therapy and for the first time mechanistically describes the effect of RTX.


Asunto(s)
Acuaporina 4/metabolismo , Autoanticuerpos/sangre , Autoantígenos/metabolismo , Inmunoglobulina G/sangre , Neuromielitis Óptica/metabolismo , Rituximab/uso terapéutico , Adulto , Acuaporina 4/genética , Acuaporina 4/inmunología , Autoantígenos/genética , Autoantígenos/inmunología , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/ultraestructura , Forma de la Célula , Extensiones de la Superficie Celular/ultraestructura , Femenino , Proteína Forkhead Box O3/fisiología , Glioblastoma , Humanos , Leupeptinas/farmacología , Masculino , MicroARNs/genética , Microscopía Confocal , Neuromielitis Óptica/tratamiento farmacológico , Neuromielitis Óptica/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Transcripción Genética , Adulto Joven
6.
Front Immunol ; 12: 645962, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34122406

RESUMEN

The failure of M. bovis BCG to induce long-term protection has been endowed to its inability to escape the phagolysosome, leading to mild activation of CD8+ mediated T cell response. Induction of apoptosis in host cells plays an important role in potentiating dendritic cells-mediated priming of CD8+ T cells, a process defined as "cross-priming." Moreover, IL-10 secretion by infected cells has been reported to hamper BCG-induced immunity against Tuberculosis (TB). Previously, we have reported that apoptosis of BCG-infected macrophages and inhibition of IL-10 secretion is FOXO3 dependent, a transcription factor negatively regulated by the pro-survival activated threonine kinase, Akt. We speculate that FOXO3-mediated induction of apoptosis and abrogation of IL-10 secretion along with M. bovis BCG immunization might enhance the protection imparted by BCG. Here, we have assessed whether co-administration of a known anti-cancer Akt inhibitor, MK-2206, enhances the protective efficacy of M. bovis BCG in mice model of infection. We observed that in vitro MK-2206 treatment resulted in FOXO3 activation, enhanced BCG-induced apoptosis of macrophages and inhibition of IL-10 secretion. Co-administration of M. bovis BCG along with MK-2206 also increased apoptosis of antigen-presenting cells in draining lymph nodes of immunized mice. Further, MK-2206 administration improved BCG-induced CD4+ and CD8+ effector T cells responses and its ability to induce both effector and central memory T cells. Finally, we show that co-administration of MK-2206 enhanced the protection imparted by M. bovis BCG against Mtb in aerosol infected mice and guinea pigs. Taken together, we provide evidence that MK-2206-mediated activation of FOXO3 potentiates BCG-induced immunity and imparts protection against Mtb through enhanced innate immune response.


Asunto(s)
Vacuna BCG/inmunología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Tuberculosis/prevención & control , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/fisiología , Apoptosis/efectos de los fármacos , Células Cultivadas , Proteína Forkhead Box O3/fisiología , Cobayas , Memoria Inmunológica/efectos de los fármacos , Macrófagos/microbiología , Ratones , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
7.
Urol Int ; 105(7-8): 687-696, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33965964

RESUMEN

BACKGROUND: Renal ischemia/reperfusion (I/R) injury (RIRI) is the main cause of acute kidney injury (AKI) in patients. We investigated the role of miR-182 after renal ischemia/reperfusion (I/R) in rat to characterize the microRNA (miRNA) network activated during development and recovery from RIRI. METHODS AND RESULTS: 12 h after lethal (45 min) renal ischemia, AKI was verified by renal histology (tubular necrosis and regeneration), blood urea nitrogen level, and renal mRNA expression in Wistar rats. We found that miR-182 markedly increased after renal I/R. In cell hypoxia/reoxygenation model, we found similar upregulation of miR-182. In function gain/loss assay, we confirmed an impaired effect of miR-182 and identified Forkhead box O3 (FoxO3) as a direct downstream target of it. By using miR-182 antagomir, the I/R injury was markedly ameliorated. CONCLUSIONS: Our results demonstrate that miR-182 promotes cell apoptosis and I/R injury through directly binding to FoxO3. The present study will provide potential therapeutic targets for renal I/R-induced AKI, and open a new avenue for AKI treatment by manipulating miRNAs levels.


Asunto(s)
Lesión Renal Aguda/etiología , Proteína Forkhead Box O3/fisiología , MicroARNs/fisiología , Daño por Reperfusión/complicaciones , Lesión Renal Aguda/genética , Animales , Masculino , Ratas , Ratas Wistar , Daño por Reperfusión/genética
8.
Mol Neurobiol ; 58(11): 5421-5436, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33983546

RESUMEN

Maintaining an intact pool of neural progenitor cells (NPCs) is crucial for generating new and functionally active neurons. Methamphetamine (METH) can exacerbate the HIV-induced deficit of adult neurogenesis; however, potential mechanisms of this influence are still poorly understood. In the present study, we present evidence that chronic exposure to METH combined with brain infection by EcoHIV results in enhanced proliferation of NPCs in the subventricular zone (SVZ) in mice. This effect was long-lasting as it was preserved ex vivo in NPCs isolated from the exposed mice over several passages in the absence of additional treatments. Increased proliferation in response to METH plus HIV was associated with dysregulation of cyclin B1 and cyclin D. Transcriptomic studies indicated that 27 out of the top 30 differentially expressed genes in response to METH plus EcoHIV were targets of the forkhead box O transcriptional factor (FOXO) and primarily FOXO3. Additional ex vivo studies and in vitro experiments using human NPCs exposed to METH and infected with HIV revealed upregulation of the CXCL12-CXCR4 axis, leading to activation of downstream pAkt and pErk, the pathways that can phosphorylate FOXO3 and force its exports from the nuclei into the cytoplasm. Indeed, nuclear expulsion of FOXO3 was demonstrated both in mice exposed to METH and infected with EcoHIV and in cell cultures of human NPCs. These results provide novel information that exposure to METH combined with HIV infection can induce aberrant proliferation of SVZ-derived NPCs and identifies CXCL12-CXCR4-Akt-1-mediated phosphorylation of FOXO3 as the mechanism responsible for this effect.


Asunto(s)
Proteína Forkhead Box O3/fisiología , VIH-1/fisiología , Metanfetamina/toxicidad , Células-Madre Neurales/efectos de los fármacos , Complejo SIDA Demencia/complicaciones , Complejo SIDA Demencia/virología , Animales , Encéfalo/patología , Encéfalo/virología , Ciclo Celular , División Celular , Células Cultivadas , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/genética , Cromonas/farmacología , Modelos Animales de Enfermedad , Regulación Viral de la Expresión Génica , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Humanos , Ventrículos Laterales/patología , Masculino , Metanfetamina/farmacología , Ratones , Ratones Endogámicos C57BL , Morfolinas/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Células-Madre Neurales/virología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Trastornos Relacionados con Sustancias/complicaciones
9.
J Orthop Surg Res ; 16(1): 259, 2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33853636

RESUMEN

BACKGROUND: Osteoarthritis (OA) is a worldwide musculoskeletal disorder. However, disease-modifying therapies for OA are not available. Here, we aimed to characterize the molecular signatures of OA and to identify novel therapeutic targets and strategies to improve the treatment of OA. METHODS: We collected genome-wide transcriptome data performed on 132 OA and 74 normal human cartilage or synovium tissues from 7 independent datasets. Differential gene expression analysis and functional enrichment were performed to identify genes and pathways that were dysregulated in OA. The computational drug repurposing method was used to uncover drugs that could be repurposed to treat OA. RESULTS: We identified several pathways associated with the development of OA, such as extracellular matrix organization, inflammation, bone development, and ossification. By protein-protein interaction (PPI) network analysis, we prioritized several hub genes, such as JUN, CDKN1A, VEGFA, and FOXO3. Moreover, we repurposed several FDA-approved drugs, such as cardiac glycosides, that could be used in the treatment of OA. CONCLUSIONS: We proposed that the hub genes we identified would play a role in cartilage homeostasis and could be important diagnostic and therapeutic targets. Drugs such as cardiac glycosides provided new possibilities for the treatment of OA.


Asunto(s)
Biología Computacional/métodos , Reposicionamiento de Medicamentos/métodos , Expresión Génica/genética , Osteoartritis/genética , Mapas de Interacción de Proteínas/genética , Proteínas Proto-Oncogénicas c-jun/fisiología , Glicósidos Cardíacos/uso terapéutico , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Femenino , Proteína Forkhead Box O3/fisiología , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Terapia Molecular Dirigida , Osteoartritis/diagnóstico , Osteoartritis/tratamiento farmacológico , Factores de Transcripción , Factor A de Crecimiento Endotelial Vascular/fisiología
10.
Life Sci ; 272: 119119, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33508296

RESUMEN

AIMS: Acute kidney injury (AKI) is a severe complication of autologous orthotopic liver transplantation (AOLT). Apoptosis has been shown to be involved in renal ischemia/reperfusion, and the PI3K/AKT signaling pathway is involved in numerous cell processes, including promoting cell survival and inhibiting apoptosis. We aimed to verify whether the PI3K/AKT signaling pathway participates in the development of post-AOLT AKI. METHODS: Male Sprague-Dawley rats underwent AOLT with or without treatment with insulin-like growth factor-1 (IGF-1, a PI3K/AKT activator) and LY294002 (a PI3K/AKT inhibitor; n = 8/group). NRK-52E cells (rat renal tubular epithelial cell line) were subjected to hypoxia-re-oxygenation to mimic renal cell I/R injury in vitro, and confirm whether silencing information regulator 1 (SIRT1) mediated the protective effects of PI3K/AKT by deacetylating forkhead protein O3a (FoxO3a). KEY FINDINGS: During the reperfusion stage, kidney injury peaked at 8 h after reperfusion, then gradually recovered, which was consistent with the dynamic changes in apoptosis and the protein expressions of Bcl-2 interacting mediator of cell death (Bim), Fas ligand (FasL), and nuclear FoxO3a AKT phosphorylation and nuclear SIRT1 protein expression were also upregulated. IGF-1 application decreased Bim, FasL, and nuclear FoxO3a protein expressions, and protected against apoptosis and AKI. In NRK-52E cells, IGF-1 upregulated nuclear SIRT1 expression, reduced FoxO3a acetylation, downregulated Bim and FasL protein expressions, and attenuated apoptosis and AKI; these effects were reversed by SIRT1 blocking. CONCLUSION: The activation of the PI3K/AKT signaling pathway not only induced FoxO3a nuclear export but also deacetylation through upregulating nuclear SIRT1 expression to attenuate post-AOLT AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteína Forkhead Box O3/metabolismo , Acetilación , Transporte Activo de Núcleo Celular , Lesión Renal Aguda/fisiopatología , Animales , Apoptosis/efectos de los fármacos , China , Proteína Forkhead Box O3/fisiología , Trasplante de Hígado/efectos adversos , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
11.
Hum Cell ; 34(1): 14-27, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32920731

RESUMEN

Mesenchymal stem cells (MSCs) are a promising regenerative medicine. The roles of miRNAs in osteogenic differentiation of bone marrow MSCs (BM-MSCs) remained less reported. Forkhead Box O3 (FOXO3) and alkaline phosphatase (ALP) levels in the BM-MSCs were measured on 3, 7, and 14 days after osteogenic differentiation. After transfection of FOXO3 overexpression plasmids or siFOXO3 into BM-MSCs, factors related to osteogenic differentiation or cell autophagy were determined. Besides, 3-methyladenine or rapamycin, as well as miR-223-3p mimic or inhibitor were applied to further determine the effect of FOXO3 in BM-MSCs. FOXO3 and ALP levels were increased in a time-dependent manner with osteogenic differentiation, supported by Alizarin Red Staining. Furthermore, up-regulated FOXO3 increased levels of ALP and factors related to osteogenic differentiation by increasing levels of autophagy-related factors. FOXO3, targeted by miR-223-3p, reversed the effects of miR-223-3p on factors related to BM-MSC autophagy and osteogenic differentiation. Down-regulated miR-223-3p expression promoted osteogenic differentiation of BM-MSCs by enhancing autophagy via targeting FOXO3, suggesting the potential of miR-223-3p as a therapeutic target for enhancing bone functions.


Asunto(s)
Autofagia/genética , Diferenciación Celular/genética , Proteína Forkhead Box O3/fisiología , Células Madre Mesenquimatosas/fisiología , MicroARNs/fisiología , Osteogénesis/genética , Autofagia/fisiología , Células Cultivadas , Humanos
12.
Oncogene ; 40(4): 777-790, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262463

RESUMEN

Metastasis remains the major obstacle to improved survival for breast cancer patients. Downregulation of FOXO3a transcription factor in breast cancer is causally associated with the development of metastasis through poorly understood mechanisms. Here, we report that FOXO3a is functionally related to the inhibition of VEGF-A/NRP1 signaling and to the consequent suppression of breast cancer metastasis. We show that FOXO3a directly induces miR-29b-2 and miR-338 expression. Ectopic expression of miR-29b-2/miR-338 significantly suppresses EMT, migration/invasion, and in vivo metastasis of breast cancer. Moreover, we demonstrate that miR-29b-2 directly targets VEGF-A while miR-338 directly targets NRP1, and show that regulation of miR-29b-2 and miR-338 mediates the ability of FOXO3a to suppress VEGF-A/NRP1 signaling and breast cancer metastasis. Clinically, our results show that the FOXO3a-miR-29b-2/miR-338-VEGF-A/NRP1 axis is dysregulated and plays a critical role in disease progression in breast cancer. Collectively, our findings propose that FOXO3a functions as a metastasis suppressor, and define a novel signaling axis of FOXO3a-miRNA-VEGF-A/NRP1 in breast cancer, which might be potential therapeutic targets for breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proteína Forkhead Box O3/fisiología , MicroARNs/fisiología , Neuropilina-1/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Adulto , Anciano , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal/fisiología
13.
Life Sci ; 267: 118942, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359247

RESUMEN

AIMS: Intracerebral hemorrhage (ICH) induces serious neuroinflammation and damage of blood-brain barrier. We aim to investigate the role of brown fat enriched lncRNA 1 (Blnc1) in the development of ICH in mice. METHODS: An ICH model was established with autologous blood injection in C57BL/6 mice, and Blnc1 siRNA was injected intracranially. Blnc1 levels were detected and brain injury was evaluated at day 3. Primary brain microvascular endothelial cells (BMVECs) were isolated from new born mice and gain- and loss-of-function experiments were performed to investigate the role of Blnc1. Then, ICH cell model was established by treating BMVECs with oxygen and glucose deprivation (OGD) plus hemin, and Blnc1 siRNA was transfected into the cells. BMVEC functions, including viability, invasion, apoptosis, permeability and secretion of inflammatory cytokines were analyzed. KEY FINDINGS: Blnc1 was upregulated in perihematomal edema, hematoma and microvessel in the brain of ICH mice. Blnc1 negatively regulated viability and migration, and facilitated apoptosis, permeability and inflammatory cytokine secretion in BMVECs. Silencing Blnc1 restrained OGD plus hemin-caused reduction of BMVEC viability and migration and the induction of apoptosis, permeability and inflammation response, and suppressed PPAR-γ/SIRT6-mediated FoxO3 activation, which could be reversed by T0070907 (PPAR-γ inhibitor). Downregulation of Blnc1 ameliorated ICH-induced nerve injury, brain edema, blood brain barrier destruction, inflammation response and hematoma. Moreover, Blnc1 levels were positively correlated with PPAR-γ levels, and Blnc1 interference suppressed PPAR-γ/SIRT6-mediated activation of FoxO3 signaling in ICH mice. SIGNIFICANCE: Silencing Blnc1 alleviated nerve injury and inflammatory response caused by ICH through activating PPAR-γ/SIRT6/FoxO3 pathway.


Asunto(s)
Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , ARN Largo no Codificante/genética , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Edema Encefálico/metabolismo , Lesiones Encefálicas/etiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteína Forkhead Box O3/fisiología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/metabolismo , PPAR gamma/fisiología , Permeabilidad/efectos de los fármacos , ARN Largo no Codificante/metabolismo , Transducción de Señal/efectos de los fármacos , Sirtuinas/metabolismo , Sirtuinas/fisiología
14.
Aging (Albany NY) ; 12(20): 20862-20879, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33065553

RESUMEN

Triggering receptor expressed on myeloid cells 2 (TREM2) has been shown with a neuroprotective function against inflammation and neuronal injury in Alzheimer's disease (AD). However, the TREM2 induced anti-inflammatory mechanism is still not well known. In this study it has been demonstrated that the expression of TREM2 was upregulated in hippocampus of 5xFAD mice, whereas TREM2 knock-out mediated by AAV significantly increased the levels of pro-inflammatory cytokines and aggravated cognitive defect. Additionally, FoxO3a, a downstream member of the PI3K/AKT pathway, could be activated by TREM2 defect via the PI3K/AKT signaling in 5xFAD mice. That suggests TREM2-induced protection is associated with the PI3K-FoxO3a axis. On the contrary, overexpression of TREM2 alleviated the LPS-induced inflammatory response and induced M2 phenotype microglia in vitro. This phenomenon can be abolished by applying the PI3K inhibitor LY294002, suggesting FoxO3a not only participates in TREM2-induced anti-inflammation response, but is also involved in regulating the phenotype of microglia. Taken together, our results show that the protective functions of TREM2, both in inflammatory response and cognitive impairment as well as in the decrease of M1 phenotype microglia, are related to PI3K/AKT/FoxO3a signaling pathway in AD mice.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Disfunción Cognitiva/etiología , Proteína Forkhead Box O3/fisiología , Inflamación/etiología , Glicoproteínas de Membrana/fisiología , Neuroprotección , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptores Inmunológicos/fisiología , Transducción de Señal , Animales , Encéfalo , Disfunción Cognitiva/prevención & control , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/fisiología
15.
Carbohydr Polym ; 247: 116740, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-32829859

RESUMEN

Pulmonary fibrosis (PF) is a lung disease with highly heterogeneous and mortality rate, but its therapeutic options are now still limited. Corona virus disease 2019 (COVID-19) has been characterized by WHO as a pandemic, and the global number of confirmed COVID-19 cases has been more than 8.0 million. It is strongly supported for that PF should be one of the major complications in COVID-19 patients by the evidences of epidemiology, viral immunology and current clinical researches. The anti-PF properties of naturally occurring polysaccharides have attracted increasing attention in last two decades, but is still lack of a comprehensively understanding. In present review, the resources, structural features, anti-PF activities, and underlying mechanisms of these polysaccharides are summarized and analyzed, which was expected to provide a scientific evidence supporting the application of polysaccharides for preventing or treating PF in COVID-19 patients.


Asunto(s)
Betacoronavirus , Productos Biológicos/uso terapéutico , Infecciones por Coronavirus/complicaciones , Pandemias , Neumonía Viral/complicaciones , Polisacáridos/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Bleomicina/toxicidad , COVID-19 , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Proteína Forkhead Box O3/fisiología , Hongos/química , Ribonucleoproteína Nuclear Heterogénea D0/fisiología , Humanos , Macrófagos/efectos de los fármacos , Medicina Tradicional China , Ratones , Neutrófilos/efectos de los fármacos , Fitoterapia , Plantas Medicinales/química , Polisacáridos/farmacología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/prevención & control , ARN Largo no Codificante/antagonistas & inhibidores , Ratas , SARS-CoV-2 , Algas Marinas/química , Transducción de Señal/efectos de los fármacos , Proteína Smad2/fisiología , Proteína smad3/fisiología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores
16.
Mar Drugs ; 18(7)2020 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-32635607

RESUMEN

In recent years, the scientific interest in natural compounds with geroprotective activities has grown exponentially. Among the various naturally derived molecules, astaxanthin (ASX) represents a highly promising candidate geroprotector. By virtue of the central polyene chain, ASX acts as a scavenger of free radicals in the internal membrane layer and simultaneously controls oxidation on the membrane surface. Moreover, several studies have highlighted ASX's ability to modulate numerous biological mechanisms at the cellular level, including the modulation of transcription factors and genes directly linked to longevity-related pathways. One of the main relevant evolutionarily-conserved transcription factors modulated by astaxanthin is the forkhead box O3 gene (FOXO3), which has been recognized as a critical controller of cell fate and function. Moreover, FOXO3 is one of only two genes shown to robustly affect human longevity. Due to its tropism in the brain, ASX has recently been studied as a putative neuroprotective molecule capable of delaying or preventing brain aging in different experimental models of brain damage or neurodegenerative diseases. Astaxanthin has been observed to slow down brain aging by increasing brain-derived neurotrophic factor (BDNF) levels in the brain, attenuating oxidative damage to lipids, protein, and DNA and protecting mitochondrial functions. Emerging data now suggest that ASX can modulate Nrf2, FOXO3, Sirt1, and Klotho proteins that are linked to longevity. Together, these mechanisms provide support for a role of ASX as a potential geroneuroprotector.


Asunto(s)
Envejecimiento/efectos de los fármacos , Encéfalo/fisiología , Proteína Forkhead Box O3/fisiología , Fármacos Neuroprotectores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Fármacos Neuroprotectores/farmacología , Xantófilas/metabolismo , Xantófilas/farmacología
17.
J Zhejiang Univ Sci B ; 21(1): 64-76, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31898443

RESUMEN

Proteasome inhibitors have shown remarkable success in the treatment of hematologic neoplasm. There has been a lot of attention to applying these drugs for solid tumor treatment. Recent preclinical study has signified the effectiveness on cell proliferation inhibition in lung adenocarcinoma treated by carfilzomib (CFZ), a second generation proteasome inhibitor. However, no insight has been gained regarding the mechanism. In this study, we have systematically investigated the CFZ functions in cell proliferation and growth, cell cycle arrest, and apoptosis in lung adenocarcinoma cells. Flow cytometry experiments showed that CFZ significantly induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma. MTS and colony formation assays revealed that CFZ substantially inhibited survival of lung adenocarcinoma cells. All results were consistently correlated to the upregulation expression of Gadd45a, which is an important gene in modulating cell cycle arrest and apoptosis in response to physiologic and environmental stresses. Here, upregulation of Gadd45a expression was observed after CFZ treatment. Knocking down Gadd45a expression suppressed G2/M arrest and apoptosis in CFZ-treated cells, and reduced cytotoxicity of this drug. The protein expression analysis has further identified that the AKT/FOXO3a pathway is involved in Gadd45a upregulation after CFZ treatment. These findings unveil a novel mechanism of proteasome inhibitor in anti-solid tumor activity, and shed light on novel preferable therapeutic strategy for lung adenocarcinoma. We believe that Gadd45a expression can be a highly promising candidate predictor in evaluating the efficacy of proteasome inhibitors in solid tumor therapy.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Proteínas de Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Oligopéptidos/farmacología , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proteína Forkhead Box O3/fisiología , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-akt/fisiología , Regulación hacia Arriba
18.
Clin Res Hepatol Gastroenterol ; 44(1): 29-37, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31208923

RESUMEN

BACKGROUND: To unravel the fundamental role of miR-498 in the context of hepatocellular carcinoma cells and understands underlying potential mechanism. METHODS: Relative viability was interrogated using MTT method and cell proliferation was determined with colony formation assay. The protein levels of cleaved Caspase-3, Bcl-2, Cyclin D, CDK4, FOXO3 and ß-actin were analyzed by western blotting. Cell invasion and migration was evaluated by transwell assay and wound healing, respectively. The relative abundance of Cyclin D, CDK4, FOXO3 and miR-498 transcripts was measured using real-time PCR. The regulatory action of miR-498 on FOXO3 expression was analyzed with luciferase reporter. RESULTS: Ectopic over-expression of miR-498 significantly inhibited viability and proliferation, suppressed cell migration and invasion, delayed cell cycle progression. We further identified FOXO3 as downstream target gene of miR-498, and positively modulated FOXO3 translation in miR-498-proficient cells consequently contributed to its anti-tumoral properties. CONCLUSIONS: Our data highlighted the tumor suppressor role of miR-498-FOXO3 signaling in hepatocellular carcinoma cells, which might hold promise for therapeutic exploitation.


Asunto(s)
Carcinoma Hepatocelular/patología , Proliferación Celular , Proteína Forkhead Box O3/fisiología , Neoplasias Hepáticas/patología , MicroARNs/fisiología , Carcinoma Hepatocelular/genética , Movimiento Celular , Humanos , Neoplasias Hepáticas/genética , Invasividad Neoplásica , Células Tumorales Cultivadas , Regulación hacia Arriba
19.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-1010516

RESUMEN

Proteasome inhibitors have shown remarkable success in the treatment of hematologic neoplasm. There has been a lot of attention to applying these drugs for solid tumor treatment. Recent preclinical study has signified the effectiveness on cell proliferation inhibition in lung adenocarcinoma treated by carfilzomib (CFZ), a second generation proteasome inhibitor. However, no insight has been gained regarding the mechanism. In this study, we have systematically investigated the CFZ functions in cell proliferation and growth, cell cycle arrest, and apoptosis in lung adenocarcinoma cells. Flow cytometry experiments showed that CFZ significantly induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma. MTS and colony formation assays revealed that CFZ substantially inhibited survival of lung adenocarcinoma cells. All results were consistently correlated to the upregulation expression of Gadd45a, which is an important gene in modulating cell cycle arrest and apoptosis in response to physiologic and environmental stresses. Here, upregulation of Gadd45a expression was observed after CFZ treatment. Knocking down Gadd45a expression suppressed G2/M arrest and apoptosis in CFZ-treated cells, and reduced cytotoxicity of this drug. The protein expression analysis has further identified that the AKT/FOXO3a pathway is involved in Gadd45a upregulation after CFZ treatment. These findings unveil a novel mechanism of proteasome inhibitor in anti-solid tumor activity, and shed light on novel preferable therapeutic strategy for lung adenocarcinoma. We believe that Gadd45a expression can be a highly promising candidate predictor in evaluating the efficacy of proteasome inhibitors in solid tumor therapy.


Asunto(s)
Humanos , Adenocarcinoma del Pulmón/patología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proteína Forkhead Box O3/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/patología , Oligopéptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/fisiología , Regulación hacia Arriba
20.
Lipids Health Dis ; 18(1): 197, 2019 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-31729980

RESUMEN

BACKGROUND: Excessive intrahepatic lipid accumulation is the major characteristic of nonalcoholic fatty liver disease (NAFLD). We sought to identify the mechanisms involved in hepatic triglyceride (TG) homeostasis. Forkhead box class O (FoxO) transcription factors have been shown to play an important role in hepatic metabolism. However, little is known about the effect of FoxO3 on hepatic TG metabolism. METHODS: Liver biopsy samples from patients with NALFD and liver tissues from high glucose and high sucrose (HFHS) fed mice, ob/ob mice and db/db mice were collected for protein and mRNA analysis. HepG2 cells were transfected with small interfering RNA to mediate FoxO3 knockdown, or adenovirus and plasmid to mediate FoxO3 overexpression. FoxO3-cDNA was delivered by adenovirus to the liver of C57BL/6 J male mice on a chow diet or on a high-fat diet, followed by determination of hepatic lipid metabolism. Sterol regulatory element-binding protein 1c (SREBP1c) luciferase reporter gene plasmid was co-transfected into HepG2 cells with FoxO3 overexpression plasmid. RESULTS: FoxO3 expression was increased in the livers of HFHS mice, ob/ob mice, db/db mice and patients with NAFLD. Knockdown of FoxO3 reduced whereas overexpression of FoxO3 increased cellular TG concentrations in HepG2 cells. FoxO3 gain-of-function caused hepatic TG deposition in C57BL/6 J mice on a chow diet and aggravated hepatic steatosis when fed a high-fat diet. Analysis of the transcripts established the increased expression of genes related to TG synthesis, including SREBP1c, SCD1, FAS, ACC1, GPAM and DGAT2 in mouse liver. Mechanistically, overexpression of FoxO3 stimulated the expression of SREBP1c, whereas knockdown of FoxO3 inhibited the expression of SREBP1c. Luciferase reporter assays showed that SREBP1c regulated the transcriptional activity of the SREBP1c promoter. CONCLUSIONS: FoxO3 promotes the transcriptional activity of the SREBP1c promoter, thus leading to increased TG synthesis and hepatic TG accumulation.


Asunto(s)
Proteína Forkhead Box O3/fisiología , Hígado/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo , Animales , Western Blotting , Hígado Graso/metabolismo , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...