Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 1840, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-37019904

RESUMO

Cellular senescence contributes to tissue homeostasis and age-related pathologies. However, how senescence is initiated in stressed cells remains vague. Here, we discover that exposure to irradiation, oxidative or inflammatory stressors induces transient biogenesis of primary cilia, which are then used by stressed cells to communicate with the promyelocytic leukemia nuclear bodies (PML-NBs) to initiate senescence responses in human cells. Mechanistically, a ciliary ARL13B-ARL3 GTPase cascade negatively regulates the association of transition fiber protein FBF1 and SUMO-conjugating enzyme UBC9. Irreparable stresses downregulate the ciliary ARLs and release UBC9 to SUMOylate FBF1 at the ciliary base. SUMOylated FBF1 then translocates to PML-NBs to promote PML-NB biogenesis and PML-NB-dependent senescence initiation. Remarkably, Fbf1 ablation effectively subdues global senescence burden and prevents associated health decline in irradiation-treated mice. Collectively, our findings assign the primary cilium a key role in senescence induction in mammalian cells and, also, a promising target in future senotherapy strategies.


Assuntos
Cílios , Proteínas Nucleares , Humanos , Animais , Camundongos , Proteína da Leucemia Promielocítica/metabolismo , Proteínas Nucleares/metabolismo , Cílios/metabolismo , Corpos Nucleares da Leucemia Promielocítica , Sumoilação , Núcleo Celular/metabolismo , Mamíferos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
2.
Cell Rep ; 36(5): 109481, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348145

RESUMO

Preadipocytes dynamically produce sensory cilia. However, the role of primary cilia in preadipocyte differentiation and adipose homeostasis remains poorly understood. We previously identified transition fiber component FBF1 as an essential player in controlling selective cilia import. Here, we establish Fbf1tm1a/tm1a mice and discover that Fbf1tm1a/tm1a mice develop severe obesity, but surprisingly, are not predisposed to adverse metabolic complications. Obese Fbf1tm1a/tm1a mice possess unexpectedly healthy white fat tissue characterized by spontaneous upregulated beiging, hyperplasia but not hypertrophy, and low inflammation along the lifetime. Mechanistically, FBF1 governs preadipocyte differentiation by constraining the beiging program through an AKAP9-dependent, cilia-regulated PKA signaling, while recruiting the BBS chaperonin to transition fibers to suppress the hedgehog signaling-dependent adipogenic program. Remarkably, obese Fbf1tm1a/tm1a mice further fed a high-fat diet are protected from diabetes and premature death. We reveal a central role for primary cilia in the fate determination of preadipocytes and the generation of metabolically healthy fat tissue.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Tecido Adiposo Bege/metabolismo , Tecido Adiposo Branco/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adipócitos/metabolismo , Adipogenia , Animais , Respiração Celular , Cílios/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Homozigoto , Humanos , Hiperfagia/complicações , Hiperfagia/patologia , Hiperplasia , Inflamação/patologia , Masculino , Síndrome Metabólica/complicações , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Obesidade/complicações , Transdução de Sinais , Fatores de Transcrição/metabolismo
3.
J Cell Mol Med ; 25(6): 2861-2871, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33512774

RESUMO

Cisplatin nephrotoxicity has been considered as serious side effect caused by cisplatin-based chemotherapy. Recent evidence indicates that renal tubular cell apoptosis and inflammation contribute to the progression of cisplatin-induced acute kidney injury (AKI). Hepatocyte nuclear factor 1ß (HNF1ß) has been reported to regulate the development of kidney cystogenesis, diabetic nephrotoxicity, etc However, the regulatory mechanism of HNF1ß in cisplatin nephrotoxicity is largely unknown. In the present study, we examined the effects of HNF1ß deficiency on the development of cisplatin-induced AKI in vitro and in vivo. HNF1ß down-regulation exacerbated cisplatin-induced RPTC apoptosis by indirectly inducing NF-κB p65 phosphorylation and nuclear translocation. HNF1ß knockdown C57BL/6 mice were constructed by injecting intravenously with HNF1ß-interfering shRNA and PEI. The HNF1ß scramble and knockdown mice were treated with 30 mg/kg cisplatin for 3 days to induce acute kidney injury. Cisplatin treatment caused increased caspase 3 cleavage and p65 phosphorylation, elevated serum urea nitrogen and creatinine, and obvious histological damage of kidney such as fractured tubules in control mice, which were enhanced in HNF1ß knockdown mice. These results suggest that HNF1ß may ameliorate cisplatin nephrotoxicity in vitro and in vivo, probably through regulating NF-κB signalling pathway.


Assuntos
Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Fator 1-beta Nuclear de Hepatócito/genética , NF-kappa B/metabolismo , Néfrons/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Modelos Animais de Doenças , Fator 1-beta Nuclear de Hepatócito/metabolismo , Túbulos Renais/efeitos dos fármacos , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Ratos , Fator de Transcrição RelA/metabolismo
4.
Mol Med Rep ; 20(5): 4323-4330, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31545442

RESUMO

Cell migration is important for renal recovery from tubular cell injury. Heat shock transcription factor 1 (HSF1) is a well­studied regulatory factor that is active during acute kidney injury. HSF1 is also involved in the migration process during tumor metastasis. Therefore, we hypothesized that HSF1 may promote the recovery of renal function by affecting kidney tubular cell migration. A wound healing assay was used to examine the cell migration rate. The results demonstrated that the migration of rat kidney proximal tubular cells (RPTCs) was increased following knockdown of HSF1. In addition, the invasion ability of HSF1 knockdown RPTCs was also significantly upregulated. The present study also identified that transforming growth factor­ß1 (TGF­ß1) was highly expressed at the edge of the wound in control cells, and its expression was further increased upon knockdown of HSF1. Inhibition of TGF­ß1 signaling prevented RPTC HSF1 knockdown cell migration, suggesting that HSF1­regulated RPTC cell migration was dependent on the TGF­ß1 signaling pathway. Furthermore, phosphorylation of TGF­ß1 and Smad2/3 was induced in HSF1 knockdown cells. Together, these results suggest that HSF1 may suppress RPTC migration by inhibiting the activation of the TGF­ß1­Smad2/3 signaling pathway.


Assuntos
Células Epiteliais/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Túbulos Renais/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Movimento Celular/genética , Técnicas de Silenciamento de Genes , Fatores de Transcrição de Choque Térmico/genética
5.
Onco Targets Ther ; 12: 5143-5152, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31303767

RESUMO

Background: Metastasis suppressor 1 (MTSS1), a potential metastasis suppressor gene associated with tumor progression, may play an important role in cancer development. Our previous study demonstrated that MTSS1 was downregulated significantly when gastric cancer (GC) progressed and metastasized, suggesting that MTSS1 may be involved in the physiopathologic mechanism of GC. Purpose: The objective of this study was to evaluate the effect of MTSS1 expression on the biological behavior of gastric cancer cell both in vitro and in vivo. Materials and methods: The gain-and-loss function of MTSS1 in GC cells were analyzed after transfection with pEGFP-N1-MTSS1 and ShRNA431. Proliferation and invasion abilities were measured by means of plate clone formation assay and transwell assay. To further explore the underlying mechanism of MTSS1-induced tumor restrain, cell cycle distribution was analyzed using flow cytometry. Results: The results revealed that overexpression of MTSS1 significantly reduced proliferation, migration and invasion of GC cells in vivo and in vitro, while downregulation of MTSS1 had the opposite biological manifestations. Moreover, overexpression of MTSS1 induced accumulation of GC cells in G2/M phase, increased phosphorylated Cdc2 expression and decreased Cdc25C and cyclinB1 levels, suggesting MTSS1 could cause G2/M cell cycle arrest. Conclusion: Our data provided insight into an important role for MTSS1 in suppressing tumor cell proliferation, invasion and migration, indicating that MTSS, as a functional tumor suppressor in GC, could be a potential therapeutic target to prevent GC metastasis.

6.
Sci Rep ; 7: 44892, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28317867

RESUMO

Interstitial fibrosis, a common pathological feature of chronic kidney diseases, is often associated with apoptosis in renal tissues. To determine the associated apoptotic pathway and its role in renal interstitial fibrosis, we established a mouse model in which Bax and Bak, two critical genes in the intrinsic pathway of apoptosis, were deleted specifically from kidney proximal tubules and used this model to examine renal apoptosis and interstitial fibrosis following unilateral urethral obstruction (UUO). It was shown that double knockout of Bax and Bak from proximal tubules attenuated renal tubular cell apoptosis and suppressed renal interstitial fibrosis in UUO. The results indicate that the intrinsic pathway of apoptosis contributes significantly to the tubular apoptosis and renal interstitial fibrosis in kidney diseases.


Assuntos
Apoptose/genética , Túbulos Renais Proximais/metabolismo , Obstrução Uretral/genética , Obstrução Uretral/patologia , Proteína Killer-Antagonista Homóloga a bcl-2/deficiência , Proteína X Associada a bcl-2/deficiência , Animais , Colágeno/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Fibrose , Camundongos , Camundongos Knockout
7.
J Biol Chem ; 292(11): 4571-4582, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28119452

RESUMO

Nephrotoxicity is a major adverse effect of cisplatin-mediated chemotherapy in cancer patients. The pathogenesis of cisplatin-induced nephrotoxicity remains largely unclear, making it difficult to design effective renoprotective approaches. Here, we have examined the role of microRNAs (miRNAs) in cisplatin-induced nephrotoxicity. We show that cisplatin nephrotoxicity was not affected by overall depletion of both beneficial and detrimental miRNAs from kidney proximal tubular cells in mice in which the miRNA-generating enzyme Dicer had been conditionally knocked out. To identify miRNAs involved in cisplatin nephrotoxicity, we used microarray analysis to profile miRNA expression and identified 47 up-regulated microRNAs and 20 down-regulated microRNAs in kidney cortical tissues. One up-regulated miRNA was miR-375, whose expression was also induced in cisplatin-treated renal tubular cells. Interestingly, inhibition of miR-375 decreased cisplatin-induced apoptosis, suggesting that miR-375 is a cell-damaging or pro-apoptotic agent. Blockade of P53 or NF-κB attenuated cisplatin-induced miR-375 expression, supporting a role of P53 and NF-κB in miR-375 induction. We also identified hepatocyte nuclear factor 1 homeobox B (HNF-1ß) as a key downstream target of miR-375. Of note, we further demonstrated that HNF-1ß protected renal cells against cisplatin-induced apoptosis. Together, these results suggest that upon cisplatin exposure, P53 and NF-κB collaboratively induce miR-375 expression, which, in turn, represses HNF-1ß activity, resulting in renal tubular cell apoptosis and nephrotoxicity.


Assuntos
Antineoplásicos/toxicidade , Cisplatino/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 1-beta Nuclear de Hepatócito/genética , Rim/efeitos dos fármacos , MicroRNAs/genética , Animais , Apoptose/efeitos dos fármacos , Rim/metabolismo , Camundongos , Camundongos Knockout , Ribonuclease III/genética
8.
Kidney Int ; 91(1): 106-118, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27622990

RESUMO

Renal ischemia-reperfusion injury is a leading cause of acute kidney injury; the pathogenesis of which remains poorly understood and effective therapies are still lacking. Here we tested whether microRNAs, identified as critical regulators of cell health and disease, are involved in this process. We found that miR-17-5p was significantly up-regulated during renal ischemia-reperfusion injury in mice and during hypoxia in cultured renal tubular cells. In cultured cells, miR-17-5p directly inhibited the expression of death receptor 6 (DR6) and attenuated apoptosis during hypoxia. Blockade of miR-17-5p abolished the suppression of DR6 and facilitated caspase activation and apoptosis. In vivo, an miR-17-5p mimic suppressed DR6 expression and protected against renal ischemia-reperfusion injury. We further verified that miR-17-5p induction during renal ischemia-reperfusion injury was dependent on p53. Inhibition of p53 with pifithrin-α or a dominant-negative mutant led to the repression of miR-17-5p expression under hypoxia in vitro. Moreover, miR-17-5p induction during renal ischemia-reperfusion injury was attenuated in proximal tubule p53 knockout mice, supporting the role of p53 in miR-17-5p induction in vivo. Thus, p53/miR-17-5p/DR6 is a new protective pathway in renal ischemia-reperfusion injury and may be targeted for the prevention and treatment of ischemic acute kidney injury.


Assuntos
Injúria Renal Aguda/metabolismo , MicroRNAs/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Traumatismo por Reperfusão/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Injúria Renal Aguda/etiologia , Animais , Apoptose , Benzotiazóis/farmacologia , Caspases/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Túbulos Renais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Traumatismo por Reperfusão/complicações , Transdução de Sinais , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Regulação para Cima
9.
J Am Soc Nephrol ; 27(9): 2784-96, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26975439

RESUMO

MicroRNAs have been implicated in ischemic AKI. However, the specific microRNA species that regulates ischemic kidney injury remains unidentified. Our previous microarray analysis revealed microRNA-489 induction in kidneys of mice subjected to renal ischemia-reperfusion. In this study, we verified the induction of microRNA-489 during ischemic AKI in mice and further examined the underlying mechanisms. Hypoxia-inducible factor-1α deficiency associated with diminished microRNA-489 induction in cultured rat proximal tubular cells subjected to hypoxia and kidney tissues of mice after renal ischemia-reperfusion injury. Moreover, genomic analysis revealed that microRNA-489 is intronic in the calcitonin receptor gene, and chromatin immunoprecipitation assays showed increased binding of hypoxia-inducible factor-1 to a specific site in the calcitonin receptor gene promoter after hypoxia. Inhibition of microRNA-489 increased apoptosis in renal tubular cells after ATP depletion injury in vitro, whereas microRNA-489 mimics mediated protection. In mice, inhibition of microRNA-489 enhanced tubular cell death and ischemic AKI without significantly affecting tubular cell proliferation. Deep sequencing identified 417 mRNAs that were recruited to the RNA-induced silencing complex by microRNA-489. Of the identified mRNAs, 127 contain microRNA-489 targeting sites, and of those, 18 are involved in the cellular stress response, including the poly(ADP-ribose) polymerase 1 gene implicated in ischemic kidney injury. Sequence analysis and in vitro studies validated poly(ADP-ribose) polymerase 1 as a microRNA-489 target. Together, these results suggest that microRNA-489 is induced via hypoxia-inducible factor-1 during ischemic AKI to protect kidneys by targeting relevant genes.


Assuntos
Injúria Renal Aguda/etiologia , Fator 1 Induzível por Hipóxia/fisiologia , MicroRNAs/fisiologia , Animais , Células Cultivadas , Túbulos Renais/citologia , Camundongos , Ratos
10.
Sci Rep ; 6: 22171, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26916346

RESUMO

Endotoxemia in sepsis, characterized by systemic inflammation, is a major cause of acute kidney injury (AKI) in hospitalized patients, especially in intensive care unit; however the underlying pathogenesis is poorly understood. Autophagy is a conserved, cellular catabolic pathway that plays crucial roles in cellular homeostasis including the maintenance of cellular function and viability. The regulation and role of autophagy in septic or endotoxic AKI remains unclear. Here we show that autophagy was induced in kidney tubular cells in mice by the endotoxin lipopolysaccharide (LPS). Pharmacological inhibition of autophagy with chloroquine enhanced LPS-induced AKI. Moreover, specific ablation of autophagy gene 7 (Atg7) from kidney proximal tubules worsened LPS-induced AKI. Together, the results demonstrate convincing evidence of autophagy activation in endotoxic kidney injury and support a renoprotective role of autophagy in kidney tubules.


Assuntos
Injúria Renal Aguda/patologia , Proteína 7 Relacionada à Autofagia/genética , Autofagia/fisiologia , Cloroquina/uso terapêutico , Endotoxemia/patologia , Túbulos Renais Proximais/patologia , Lipopolissacarídeos/toxicidade , Injúria Renal Aguda/prevenção & controle , Animais , Autofagia/efeitos dos fármacos , Cisplatino/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão
11.
Protein Cell ; 4(11): 813-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24214874

RESUMO

Acute kidney injury (AKI), associated with significant morbidity and mortality, is widely known to involve epithelial apoptosis, excessive inflammation, and fibrosis in response to ischemia or reperfusion injury, which results in either chronic pathological changes or death. Therefore, it is imperative that investigations are conducted in order to find effective, early diagnoses, and therapeutic targets needed to help prevent and treat AKI. However, the mechanisms modulating the pathogenesis of AKI still remain largely undetermined. MicroRNAs (miRNAs), small non-coding RNA molecules, play an important role in several fundamental biological and pathological processes by a post transcriptional regulatory function of gene expression. MicroRNA-21 (miR-21) is a recently identified, typical miRNA that is functional as a regulator known to be involved in apoptosis as well as inflammatory and fibrotic signaling pathways in AKI. As a result, miR-21 is now considered a novel biomarker when diagnosing and treating AKI. This article reviews the correlative literature and research progress regarding the roles of miR-21 in AKI.


Assuntos
Injúria Renal Aguda/genética , MicroRNAs , Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/patologia , Animais , Apoptose , Biomarcadores/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Terapia de Alvo Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...