Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biol Reprod ; 111(1): 197-211, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38519102

RESUMEN

Heat stress induces testicular oxidative stress, impairs spermatogenesis, and increases the risk of male infertility. Recent studies have highlighted the antioxidative properties of the Sestrins family in reducing cellular oxidative damage. However, the role of Sestrins (Sestrin1, 2, and 3) in the testicular response to heat stress remains unclear. Here, we found that Sestrin2 and 3 were highly expressed in the testis relative to Sestrin1. Then, the Sestrin2-/- and Sestrin3-/- mice were generated by CRISPR/Cas9 to investigate the role of them on spermatogenesis after heat stress. Our data showed that Sestrin2-/- and Sestrin3-/- mice testes exhibited more severe damage manifested by exacerbated loss of germ cells and higher levels of oxidative stress as compared to wild-type counterparts after heat stress. Notably, Sestrin2-/- and Sestrin3-/- mice underwent a remarkable increase in heat-induced spermatocyte apoptosis than that of controls. Furthermore, the transcriptome landscape of spermatocytes and chromosome spreading showed that loss of Sestrin2 and Sestrin3 exacerbated meiotic failure by compromising DNA double-strand breaks repair after heat stress. Taken together, our work demonstrated a critical protective function of Sestrin2 and Sestrin3 in mitigating the impairments of spermatogenesis against heat stress.


Asunto(s)
Respuesta al Choque Térmico , Meiosis , Ratones Noqueados , Espermatogénesis , Animales , Masculino , Espermatogénesis/fisiología , Espermatogénesis/genética , Ratones , Meiosis/fisiología , Respuesta al Choque Térmico/fisiología , Sestrinas/genética , Sestrinas/metabolismo , Estrés Oxidativo/fisiología , Testículo/metabolismo , Espermatocitos/metabolismo , Apoptosis/fisiología
2.
CNS Neurosci Ther ; 30(3): e14664, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38516781

RESUMEN

AIMS: Neuroblastoma (NB) is the most common extracranial solid tumor in children, with a 5-year survival rate of <50% in high-risk patients. MYCN amplification is an important factor that influences the survival rate of high-risk patients. Our results indicated MYCN regulates the expression of SESN1. Therefore, this study aimed to investigate the role and mechanisms of SESN1 in NB. METHODS: siRNAs or overexpression plasmids were used to change MYCN, SESN1, or MyD88's expression. The role of SESN1 in NB cell proliferation, migration, and invasion was elucidated. Xenograft mice models were built to evaluate SESN1's effect in vivo. The correlation between SESN1 expression and clinicopathological data of patients with NB was analyzed. RNA-Seq was done to explore SESN1's downstream targets. RESULTS: SESN1 was regulated by MYCN in NB cells. Knockdown SESN1 promoted NB cell proliferation, cell migration, and cell invasion, and overexpressing SESN1 had opposite functions. Knockdown SESN1 promoted tumor growth and shortened tumor-bearing mice survival time. Low expression of SESN1 had a positive correlation with poor prognosis in patients with NB. RNA-Seq showed that Toll-like receptor (TLR) signaling pathway, and PD-L1 expression and PD-1 checkpoint pathway in cancer were potential downstream targets of SESN1. Knockdown MyD88 or TLRs inhibitor HCQ reversed the effect of knockdown SESN1 in NB cells. High expression of SESN1 was significantly associated with a higher immune score and indicated an active immune microenvironment for patients with NB. CONCLUSIONS: SESN1 functions as a new tumor suppressor gene via TLR signaling pathway in NB.


Asunto(s)
Factor 88 de Diferenciación Mieloide , Neuroblastoma , Niño , Humanos , Animales , Ratones , Proteína Proto-Oncogénica N-Myc/genética , Proteína Proto-Oncogénica N-Myc/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Factores de Transcripción/genética , Transducción de Señal/genética , Neuroblastoma/patología , Genes Supresores de Tumor , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Microambiente Tumoral , Sestrinas/genética , Sestrinas/metabolismo
3.
Autophagy ; 20(1): 15-28, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37674294

RESUMEN

Macroautophagy/autophagy is an evolutionarily highly conserved catabolic process that is important for the clearance of cytosolic contents to maintain cellular homeostasis and survival. Recent findings point toward a critical role for autophagy in brain function, not only by preserving neuronal health, but especially by controlling different aspects of neuronal development and functioning. In line with this, mutations in autophagy-related genes are linked to various key characteristics and symptoms of neurodevelopmental disorders (NDDs), including autism, micro-/macrocephaly, and epilepsy. However, the group of NDDs caused by mutations in autophagy-related genes is relatively small. A significant proportion of NDDs are associated with mutations in genes encoding epigenetic regulatory proteins that modulate gene expression, so-called chromatinopathies. Intriguingly, several of the NDD-linked chromatinopathy genes have been shown to regulate autophagy-related genes, albeit in non-neuronal contexts. From these studies it becomes evident that tight transcriptional regulation of autophagy-related genes is crucial to control autophagic activity. This opens the exciting possibility that aberrant autophagic regulation might underly nervous system impairments in NDDs with disturbed epigenetic regulation. We here summarize NDD-related chromatinopathy genes that are known to regulate transcriptional regulation of autophagy-related genes. Thereby, we want to highlight autophagy as a candidate key hub mechanism in NDD-related chromatinopathies.Abbreviations: ADNP: activity dependent neuroprotector homeobox; ASD: autism spectrum disorder; ATG: AutTophaGy related; CpG: cytosine-guanine dinucleotide; DNMT: DNA methyltransferase; EHMT: euchromatic histone lysine methyltransferase; EP300: E1A binding protein p300; EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit; H3K4me3: histone 3 lysine 4 trimethylation; H3K9me1/2/3: histone 3 lysine 9 mono-, di-, or trimethylation; H3K27me2/3: histone 3 lysine 27 di-, or trimethylation; hiPSCs: human induced pluripotent stem cells; HSP: hereditary spastic paraplegia; ID: intellectual disability; KANSL1: KAT8 regulatory NSL complex subunit 1; KAT8: lysine acetyltransferase 8; KDM1A/LSD1: lysine demethylase 1A; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NDD: neurodevelopmental disorder; PHF8: PHD finger protein 8; PHF8-XLID: PHF8-X linked intellectual disability syndrome; PTM: post-translational modification; SESN2: sestrin 2; YY1: YY1 transcription factor; YY1AP1: YY1 associated protein 1.


Asunto(s)
Trastorno del Espectro Autista , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , Humanos , Histonas/metabolismo , Epigénesis Genética , Lisina/metabolismo , Discapacidad Intelectual/genética , Trastorno del Espectro Autista/genética , Autofagia/genética , Células Madre Pluripotentes Inducidas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Sestrinas/genética , Sestrinas/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular/metabolismo , Histona Demetilasas/metabolismo
5.
Asian Pac J Cancer Prev ; 24(11): 3715-3727, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-38019229

RESUMEN

OBJECTIVE: In this research we evaluated molecular mechanism of effect of metformin in radio sensitivity of breast cancer cells. METHODS: This research was done in cellular and molecular research center of Qazvin university of Medical science in 1399 to 1401. Studied samples were two breast cancer cell lines (MCF-7 and MDA-MB-231) they are derived from primary and secondary tumors resected from a single patient. We exposed them to cumulative 50 Gy radiation and constructed radio resistant cell lines. Then resistant cell lines were treated with 50µm of metformin. Our studied groups were resistant cells treated and un treated with metformin. Then, the expression rate of miR-21-5p and SESN1 gene in resistant and control cells was checked by Quantitative Real-time PCR(qRTPCR). After the cell lines were treated with different concentrations of metformin at different intervals, the rate of cell proliferation and cell death was checked by CCK-8 assay and flow cytometry. The Western blot method was also used to confirm the expression of some genes. RESULTS: Our results showed that the expression of miR-21-5p was upregulated in radiation-resistant cancer cells (1.8±0.65) (P<0.0001) MCF-7 cell line and (1.6±0.42)(P<0.001) MBA-MD-231 cell line, while the expression of SESN1 was down regulated (0.46±0.12) (P<0.0001) MCF-7 cell line and (0.42±0.22) (P<0.001) MBA-MD-231 cell line. Metformin enhanced the radio sensitivity of breast cancer cells in a dose and time-dependent manner. Also, metformin treatment decreased the expression of miR-21-5p (0.47±0.32) (P<0.0001) MCF-7 Cell line and (0.45±0.21)(P<0.001) MBA-MD-231 cell line and increased the expression of SESN1 (1.65±0.72)(P<0.0001)MCF-7 cell line and (1.73±0.52)(P<0.0001) MBA-MD-231 cell line. The function of metformin was reversed by miR-21-5p inhibitors or the transfection of SESN1 overexpressing plasmids. CONCLUSION: In conclusion, based on this research results, metformin enhanced the radio sensitivity of breast cancer cells via modulating  the expression of miR-21-5p and SESN1.


Asunto(s)
Neoplasias de la Mama , Metformina , MicroARNs , Sestrinas , Femenino , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Células MCF-7 , Metformina/farmacología , MicroARNs/genética , Tolerancia a Radiación/genética , Factores de Transcripción , Sestrinas/genética
6.
J Dermatol Sci ; 109(2): 52-60, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36858850

RESUMEN

BACKGROUND: Treatment resistance often occurs with BRAF inhibitor (BRAFi) therapy for melanoma, bringing in a great challenge to the treatment of melanoma patients harboring mutant BRAF gene. Recent studies revealed redox vulnerability constitutes a novel opportunity to overcome BRAFi resistance. Previously we found Sestrin2 provided protection to metastatic melanoma cells by detoxifying reactive oxygen species (ROS) induced by anoikis, but its defensive role against redox stimuli elicited by BRAFi was unclear. OBJECTIVE: In-depth explored the role of Sestrin2 in BRAFi-resistant melanoma. METHODS: Vemurafenib-resistant melanoma cells were established using 451Lu and UACC62 cell lines carrying BRAFV600E mutation. Mechanistic studies were subsequently performed by transfection of lentiviral vectors encoding an shRNA against SESN2 or embedded with the coding sequences of SESN2 cDNA. RESULTS: Elevated Sestrin2 expression was found in vemurafenib-resistance melanoma cells. Further mechanistic studies revealed that BRAFi-resistant melanoma cells employ Sestrin2 to adapt to higher oxidative stress under vemurafenib exposure. It was also demonstrated that mTOR signaling was significantly activated following Sestrin2 knockdown. Given the known promoting role of active mTOR signaling in melanoma proliferation and survival, the effects of mTOR blocker and Sestrin2 ablation on BRAFi-resistant melanoma cells were further tested, and the combination was found to result in enhanced inhibition of melanoma cell growth. CONCLUSIONS: Our findings demonstrated the contribution of Sestrin2 to the development of BRAFi resistance and the fact that the combination of mTOR blocker assisted Sestrein2 ablation in eliminating BRAFi resistance of melanoma. Therefore, mTOR and Sestrin2 may be novel combinatorial therapeutic targets to overcome BRAFi resistance of melanoma.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Humanos , Vemurafenib/farmacología , Vemurafenib/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Resistencia a Antineoplásicos/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Serina-Treonina Quinasas TOR/metabolismo , Mutación , Oxidación-Reducción , Línea Celular Tumoral , Sestrinas/genética , Sestrinas/metabolismo
7.
Nature ; 608(7921): 209-216, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35859173

RESUMEN

Mechanistic target of rapamycin complex 1 (mTORC1) regulates cell growth and metabolism in response to multiple nutrients, including the essential amino acid leucine1. Recent work in cultured mammalian cells established the Sestrins as leucine-binding proteins that inhibit mTORC1 signalling during leucine deprivation2,3, but their role in the organismal response to dietary leucine remains elusive. Here we find that Sestrin-null flies (Sesn-/-) fail to inhibit mTORC1 or activate autophagy after acute leucine starvation and have impaired development and a shortened lifespan on a low-leucine diet. Knock-in flies expressing a leucine-binding-deficient Sestrin mutant (SesnL431E) have reduced, leucine-insensitive mTORC1 activity. Notably, we find that flies can discriminate between food with or without leucine, and preferentially feed and lay progeny on leucine-containing food. This preference depends on Sestrin and its capacity to bind leucine. Leucine regulates mTORC1 activity in glial cells, and knockdown of Sesn in these cells reduces the ability of flies to detect leucine-free food. Thus, nutrient sensing by mTORC1 is necessary for flies not only to adapt to, but also to detect, a diet deficient in an essential nutrient.


Asunto(s)
Adaptación Fisiológica , Dieta , Proteínas de Drosophila , Drosophila melanogaster , Leucina , Sestrinas , Adaptación Fisiológica/genética , Alimentación Animal , Animales , Autofagia , Dieta/veterinaria , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Preferencias Alimentarias , Leucina/deficiencia , Leucina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neuroglía/metabolismo , Sestrinas/deficiencia , Sestrinas/genética , Sestrinas/metabolismo , Transducción de Señal
8.
Hum Cell ; 35(4): 1100-1113, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35622213

RESUMEN

Sestrin 1 (SESN1) is a stress-inducible protein that suppresses tumors in numerous cancers. However, the function of SESN1 in head and neck squamous cell carcinoma (HNSCC) is not clear and needs to be elucidated. Here, SESN1 expression was downregulated in HNSCC tissues and cell lines, and low SESN1 expression was positively correlated with poor prognosis in patients with HNSCC. Moreover, SESN1 overexpression inhibited the proliferation, migration, and invasion of HSC-6 and CAL-33 cells. In addition, the binding relationship between miR-377-3p and SESN1 was confirmed using luciferase reporter and RNA immunoprecipitation assays. Downregulation of SESN1 expression was consistent with high levels of miR-377-3p in HNSCC tissues. Linear regression analysis of clinical HNSCC tissues revealed a negative correlation between miR-377-3p and SESN1 expression. Moreover, co-immunoprecipitation mass spectrometry analysis revealed that SESN1 interacted with SMAD3, and SMAD3 reversed the increased proliferation, migration, and invasion of HSC-6 and CAL-33 cells caused by SESN1 knockdown. In conclusion, these findings provide evidence that SESN1 functions as a tumor suppressor and reveal the miR-377-3p-SESN1-SMAD3 regulatory axis that contributes to proliferation, migration, and invasion in HNSCC development, which may represent an interventional target for HNSCC therapy.


Asunto(s)
Neoplasias de Cabeza y Cuello , MicroARNs , Sestrinas , Proteína smad3 , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , MicroARNs/genética , Sestrinas/genética , Proteína smad3/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética
9.
Lab Invest ; 102(5): 485-493, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34880414

RESUMEN

Cholestasis causes ductular reaction in the liver where the reactive cholangiocytes not only proliferate but also gain a neuroendocrine-like phenotype, leading to inflammatory cell infiltration and extracellular matrix deposition and contributing to the development and progression of cholestatic liver fibrosis. This study aims to elucidate the role of miR-200c in cholestasis-induced biliary liver fibrosis and cholangiocyte activation. We found that miR-200c was extremely abundant in cholangiocytes but was reduced by cholestasis in a bile duct ligation (BDL) mouse model; miR-200c was also decreased by bile acids in vitro. Phenotypically, loss of miR-200c exacerbated cholestatic liver injury, including periductular fibrosis, intrahepatic inflammation, and biliary hyperplasia in both the BDL model and the 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) model. We identified sestrin 1 (SESN1) as a target of miR-200c. Sesn1-/--BDL mice showed mitigation of cholestatic liver injury. On a molecular level, the pro-proliferative IL-6/AKT feedback loop was activated in Mir200c-/- livers but was inhibited in Sesn1-/- livers upon cholestasis in mice. Furthermore, rescuing expression of miR-200c by the adeno-associated virus serotype 8 ameliorated BDL-induced liver injury in Mir200c-/- mice. Taken together, this study demonstrates that miR-200c restrains the proliferative and neuroendocrine-like activation of cholangiocytes by targeting SESN1 and inhibiting the IL-6/AKT feedback loop to protect against cholestatic liver fibrosis. Our findings provide mechanistic insights regarding biliary liver fibrosis, which may help to reveal novel therapeutic targets for the treatment of cholestatic liver injury and liver fibrosis.


Asunto(s)
Colestasis , Cirrosis Hepática , MicroARNs , Sestrinas , Animales , Conductos Biliares/metabolismo , Proteínas de Ciclo Celular , Colestasis/complicaciones , Colestasis/genética , Colestasis/metabolismo , Interleucina-6/metabolismo , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sestrinas/genética
10.
Stem Cell Reports ; 16(9): 2078-2088, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34388363

RESUMEN

The health and homeostasis of skeletal muscle are preserved by a population of tissue-resident muscle stem cells (MuSCs) that maintain a state of mitotic and metabolic quiescence in adult tissues. The capacity of MuSCs to preserve the quiescent state declines with aging and metabolic insults, promoting premature activation and stem cell exhaustion. Sestrins are a class of stress-inducible proteins that act as antioxidants and inhibit the activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling complex. Despite these pivotal roles, the role of Sestrins has not been explored in adult stem cells. We show that SESTRIN1,2 loss results in hyperactivation of the mTORC1 complex, increased propensity to enter the cell cycle, and shifts in metabolic flux. Aged SESTRIN1,2 knockout mice exhibited loss of MuSCs and a reduced ability to regenerate injured muscle. These findings demonstrate that Sestrins help maintain metabolic pathways in MuSCs that protect quiescence against aging.


Asunto(s)
Metabolismo Energético , Homeostasis , Músculo Esquelético/citología , Sestrinas/genética , Células Madre/metabolismo , Factores de Edad , Animales , Biomarcadores , Técnicas de Cultivo de Célula , Separación Celular/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunohistoquímica , Inmunofenotipificación , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Sestrinas/deficiencia , Sestrinas/metabolismo , Células Madre/citología
11.
Cell Mol Gastroenterol Hepatol ; 12(3): 921-942, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33962074

RESUMEN

BACKGROUND & AIMS: Sestrin 1/2/3 (Sesn1/2/3) belong to a small family of proteins that have been implicated in the regulation of metabolic homeostasis and oxidative stress. However, the underlying mechanisms remain incompletely understood. The aim of this work was to illustrate the collective function of Sesn1/2/3 in the protection against hepatic lipotoxicity. METHODS: We used Sesn1/2/3 triple knockout (TKO) mouse and cell models to characterize oxidative stress and signal transduction under lipotoxic conditions. Biochemical, histologic, and physiological approaches were applied to illustrate the related processes. RESULTS: After feeding with a Western diet for 8 weeks, TKO mice developed remarkable metabolic associated fatty liver disease that was manifested by exacerbated hepatic steatosis, inflammation, and fibrosis compared with wild-type counterparts. Moreover, TKO mice exhibited higher levels of hepatic lipotoxicity and oxidative stress. Our biochemical data revealed a critical signaling node from sestrins to c-Jun N-terminal kinases (JNKs) in that sestrins interact with JNKs and mitogen-activated protein kinase kinase 7 and suppress the JNK phosphorylation and activity. In doing so, sestrins markedly reduced palmitate-induced lipotoxicity and oxidative stress in both mouse and human hepatocytes. CONCLUSIONS: The data from this study suggest that Sesn1/2/3 play an important role in the protection against lipotoxicity-associated oxidative stress and related pathology in the liver.


Asunto(s)
Hígado Graso/etiología , Hígado Graso/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Metabolismo de los Lípidos , Hígado/metabolismo , Estrés Oxidativo , Sestrinas/metabolismo , Animales , Biomarcadores , Citoprotección/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Hígado Graso/patología , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inflamación/complicaciones , Inflamación/etiología , Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Fosforilación , Sestrinas/genética
12.
J Endocrinol Invest ; 44(7): 1395-1405, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33048307

RESUMEN

PURPOSE: We earlier reported that Sestrin2 regulates monocyte activation and atherogenic events through AMPK-mTOR nexus under high-glucose and dyslipidemic conditions. However, the statuses of Sestrins in diabetes and dyslipidemia are not known. We report here on the status of Sestrins and their association with diabetic dyslipidemia and atherosclerosis. METHODS: Individuals with normal glucose tolerance (NGT) (n = 46), dyslipidemia (n = 42), and patients with Type 2 diabetes with (n = 41) and without dyslipidemia (n = 40) were recruited from a tertiary diabetes centre, Chennai, India to study the mRNA expression levels of Sestrins (1, 2, and 3) in monocytes by RT-qPCR. Serum levels of Sestrins were measured using ELISA. Atherogenic index of plasma was calculated as log (triglyceride/HDL). RESULTS: mRNA expressions of Sestrin1 and Sestrin3 were significantly reduced in monocytes under dyslipidemic conditions but not in diabetes condition. Interestingly, Sestrin2 mRNA expression was significantly reduced in all disease conditions including dyslipidemia, and diabetes with and without dyslipidemia. Sestrin2 mRNA levels were negatively correlated with glycemic and lipid parameters and plasma atherogenic index. Furthermore, circulatory Sestrin2 was also found to be significantly decreased in dyslipidemia (415.2 ± 44.7 pg/ml), diabetes (375 ± 45 pg/ml), and diabetes with dyslipidemia (319.2 ± 26.3 pg/ml) compared to NGT (706.3 ± 77 pg/ml) and negatively correlated with glycemic, lipid parameters, and plasma atherogenic index. CONCLUSION: We report for the first time that Sestrins levels are significantly decreased in diabetes and dyslipidemic conditions. More strikingly, Sestrin2 had a strong association with atherogenic risk factors and severity of atherogenic index and we suggest that Sestrin2 may be used as a biomarker for assessing atherogenesis.


Asunto(s)
Aterosclerosis/patología , Biomarcadores/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Dislipidemias/complicaciones , Proteínas Nucleares/metabolismo , Sestrinas/metabolismo , Adulto , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Monocitos/patología , Proteínas Nucleares/genética , Proyectos Piloto , Pronóstico , Factores de Riesgo , Sestrinas/genética , Índice de Severidad de la Enfermedad
13.
Brain Res ; 1750: 147165, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33069734

RESUMEN

Sestrin1 (Sesn1) acts as a stress-inducible protein that performs a remarkable cytoprotective function upon diverse cellular stresses. However, whether Sesn1 exerts a cytoprotective role in neurons following cerebral ischemia/reperfusion injury is unknown. The goal of this work was to evaluate the role of Sesn1 in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced neuronal injury in vitro. The induction of Sesn1 was found in neurons exposed to OGD/R treatment. The silencing of Sesn1 rendered neurons more vulnerable to OGD/R injury, while the up-regulation of Sesn1 ameliorated OGD/R-induced neuronal injury by reducing apoptosis and the generation of reactive oxygen species (ROS). Furthermore, the up-regulation of Sesn1 promoted the activity of the nuclear factor-erythroid 2-related factor 2 (Nrf2) by down-regulating the expression of the Kelchlike ECH-associated protein 1 (Keap1). The restoration of Keap1 or the suppression of Nrf2 remarkably abolished the Sesn1-induced neuroprotection effects in OGD/R-exposed neurons. In summary, our work indicates that Sesn1 is a remarkable neuroprotective protein that potentiates Nrf2 activation via Keap1 to ameliorate OGD/R-induced injury.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Glucosa/metabolismo , Hipocampo , Proteína 1 Asociada A ECH Tipo Kelch/fisiología , Ratones , Factor 2 Relacionado con NF-E2/fisiología , Neuronas , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Oxígeno/metabolismo , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Sestrinas/genética , Sestrinas/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA