Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Oral Dis ; 2023 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-37103670

RESUMO

OBJECTIVE: The temporomandibular joint (TMJ) disc cushions intraarticular stress during mandibular movements. While mechanical overloading is related to cartilage degeneration, the pathogenesis of TMJ disc degeneration is unclear. Here, we determined the regulatory role of mechanoinductive transient receptor potential vanilloid 4 (TRPV4) in mechanical overload-induced TMJ disc degeneration. METHODS: We explored the effect of mechanical overload on the TMJ discs in a rat occlusal interference model in vivo, and by applying sustained compressive force in vitro. TRPV4 inhibition was delivered by small interfering RNA or GSK2193874; TRPV4 activation was delivered by GSK1016790A. The protective effect of TRPV4 inhibition was validated in the rat occlusal interference model. RESULTS: Occlusal interference induced TMJ disc degeneration with enhanced extracellular matrix degradation in vivo and mechanical overload promoted inflammatory responses in the TMJ disc cells via Ca2+ influx with significantly upregulated TRPV4. TRPV4 inhibition reversed mechanical overload-induced inflammatory responses; TRPV4 activation simulated mechanical overload-induced inflammatory responses. Moreover, TRPV4 inhibition alleviated TMJ disc degeneration in the rat occlusal interference model. CONCLUSION: Our findings suggest TRPV4 plays a pivotal role in the pathogenesis of mechanical overload-induced TMJ disc degeneration and may be a promising target for the treatment of degenerative changes of the TMJ disc.

2.
Cell Mol Life Sci ; 76(19): 3861-3873, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30980112

RESUMO

The ability of cells to repair DNA double-strand breaks (DSBs) is important for maintaining genome stability and eliminating oncogenic DNA lesions. Two distinct and complementary pathways, non-homologous end joining (NHEJ) and homologous recombination (HR), are employed by mammalian cells to repair DNA DSBs. Each pathway is tightly controlled in response to increased DSBs. The Ku heterodimer has been shown to play a regulatory role in NHEJ repair. Ku80 ubiquitination contributes to the selection of a DSB repair pathway by causing the removal of Ku heterodimers from DSB sites. However, whether Ku80 deubiquitination also plays a role in regulating DSB repair is unknown. To address this question, we performed a comprehensive study of the deubiquitinase specific for Ku80, and our study showed that the deubiquitinase OTUD5 serves as an important regulator of NHEJ repair by increasing the stability of Ku80. Further studies revealed that OTUD5 depletion impaired NHEJ repair, and hence reduced overall DSB repair. Furthermore, OTUD5-depleted cells displayed excess end resection; as a result, HR repair was facilitated by OTUD5 depletion during the S/G2 phase. In summary, our study demonstrates that OTUD5 is a specific deubiquitinase for Ku80 and establishes OTUD5 as an important and positive regulator of NHEJ repair.


Assuntos
Reparo do DNA por Junção de Extremidades , Endopeptidases/metabolismo , Autoantígeno Ku/metabolismo , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Reparo do DNA , Fase G2/genética , Humanos , Estabilidade Proteica , Fase S/genética , Ubiquitinação
3.
Med Sci Monit ; 25: 8651-8659, 2019 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-31860636

RESUMO

BACKGROUND The oncogene PIM1, encoding a constitutively active serine/threonine protein kinase, is involved in the regulation of cell proliferation, survival, differentiation, and apoptosis. There is a growing body of literature on the role of PIM1-mediated cellular senescence, but the precise mechanism remains unclear. MATERIAL AND METHODS Silver staining and LC-MS/MS analysis were performed to investigate the protein interacting with PIM1. Immunofluorescence, Co-IP, and Western blot assay were used to assess the interaction of PIM1 and SND1. EdU incorporation and CCK8 assay were used to detect cell proliferation and immunohistochemistry was used to detect the level of the indicated protein. RESULTS We found that PIM1 can bind directly and phosphorylate SND1. In addition, decreased expression of SND1 leads to the upregulation of SASP. SND1 is involved in cellular senescence induced by PIM1. CONCLUSIONS We investigated the role of PIM1 in oncogene-induced normal cellular senescence. Our results promote further understanding of the mechanisms underlying OIS and suggest potential applications for preventing tumorigenesis.


Assuntos
Endonucleases/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Senescência Celular/fisiologia , Cromatografia Líquida/métodos , Células HEK293 , Humanos , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Espectrometria de Massas em Tandem/métodos
4.
Biochem Biophys Res Commun ; 503(4): 2784-2791, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30100061

RESUMO

TIMELESS protein is known to be essential for normal circadian rhythms. Aging is a deleterious process which affects all the physiological functions of complex organisms including the circadian rhythms. The circadian aging may produce disorganization among the circadian rhythms, arrhythmicity and even, disconnection from the environment, resulting in a detrimental situation to the organism. However, the role of circadian genes on the aging process is poorly understood. In present study, we found TIMELESS was down-regulated in cellular senescence, and further research indicated E2F1 bound to the promotor of TIMELESS and regulated its expression in cellular senescence. Knockdown of TIMELESS accelerated cellular senescence induced by ectopic expression of RasV12, and overexpression of TIMELESS delayed this kind onset of senescence. Meanwhile, micrococcal nuclease assays proved depletion of TIMELESS exacerbated genomic instability at the onset of senescence. Together, our data reveal that TIMELESS plays a role in OIS, which is associated with genome stability changing.


Assuntos
Proteínas de Ciclo Celular/genética , Senescência Celular/genética , Ritmo Circadiano/genética , Fator de Transcrição E2F1/genética , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Senescência Celular/efeitos dos fármacos , Fator de Transcrição E2F1/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica , Genes Reporter , Instabilidade Genômica , Células HEK293 , Humanos , Peróxido de Hidrogênio/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , beta-Galactosidase/genética , beta-Galactosidase/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
5.
Biochem Biophys Res Commun ; 501(3): 779-785, 2018 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-29763603

RESUMO

The proto-oncogene PIM1 encodes Ser/Thr kinase and regulates cell growth, differentiation and apoptosis. However, more and more studies including ours have found that PIM1 can induce senescence in normal human diploid fibroblasts and behave as a tumor suppressor. But the relevant molecular mechanism of this process is not yet clear. It has been reported that Chromobox homolog 8 (CBX8) binds directly to INK4A as a transcriptional repressor, thereby suppressing stress-induced senescence. Here we report that PIM1 can phosphorylate CBX8 to promote its degradation, thereby up-regulating p16, during PIM1-induced cell senescence. Overexpression of CBX8 can inhibit PIM1-induced cell senescence. These data suggest that to promote CBX8 degradation may be an important molecular mechanism of PIM1-induced cell senescence.


Assuntos
Senescência Celular , Fibroblastos/citologia , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Linhagem Celular , Proliferação de Células , Regulação para Baixo , Fibroblastos/metabolismo , Genes p16 , Células HEK293 , Humanos , Fosforilação , Complexo Repressor Polycomb 1/genética , Proteólise , Proto-Oncogene Mas , Regulação para Cima
6.
Mol Cell Biochem ; 446(1-2): 137-148, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29388151

RESUMO

P16 is the product of cyclin-dependent kinase 2 (CDKN2A) gene and plays multi-pronged roles in the cancer progression. Breast cancer (BC) is the most commonly diagnosed cancer type among females. In the current study, the potential function of P16 in the growth and metastasis of BC was investigated. Firstly, the expression statuses of P16 in different cancer types were investigated using Oncomine database and validated with corresponding cancer cell lines. Afterwards, the expression of P16 was knocked down in BC cell line BT-549 and the effect on the cell proliferation, sensitivity to paclitaxel (TAX), apoptosis, migration, and invasion abilities was assessed using CCK-8, Edu, flow cytometry, scratch, and transwell assays, respectively. The influence of P16 inhibition and P16 overexpression on the activity of IL-6/JAK/STAT3 signaling was explored. Additionally, the effect of P16 inhibition on the tumor growth was verified with a BC xenograft mice model. The abnormal expression of P16 was detected in BC cell line BT-549 as well as colorectal cancer and osteosarcoma cell lines. The inhibition of P16 suppressed the cell proliferation, invasion, and migration abilities while induced the apoptosis and sensitivity to TAX in BT-549 cells. At molecular level, P16 knockdown inhibited the expression of IL6ST and Survivin, and the phosphorylation of JAK2 and STAT3. However, the induced expression of P16 in P16-knockdown BT-549 cells restored the activity of IL-6/JAK2/STAT3 pathway. The results of in vitro assays were confirmed with BC xenograft models: the inhibition of P16 decreased the tumor growth rate. Findings outlined in the current study demonstrated that the inhibition of P16 decreased the growth and metastasis potential of BC cells by inhibiting IL-6/JAK2/STAT3 signaling.


Assuntos
Neoplasias da Mama/metabolismo , Movimento Celular , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Interleucina-6/metabolismo , Janus Quinase 2/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Xenoenxertos , Humanos , Interleucina-6/genética , Janus Quinase 2/genética , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Fator de Transcrição STAT3/genética
7.
Exp Cell Res ; 350(1): 1-8, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27756608

RESUMO

Androgens (AR) play an important role in initiation and progression of prostate cancer. It has been shown that AR exert their effects mainly through the androgen-activated AR which binds to androgen response elements (AREs) in the regulatory regions of target genes to regulate the transcription of androgen-responsive genes, thus, identification of AR downstream target gene is critical to understand androgen function in prostate cancer. In this study, our results showed that androgen treatment of LNCaP cells induced PTTG1 expression, which was blocked by the androgen receptor antagonist, Casodex. Bioinformatics analysis and experiments using PTTG1 promoter deletion mutants showed that the PTTG1 promoter contains a putative androgen response element (ARE), which localizes in the -851 to -836 region of the promoter. Androgen activated androgen receptor (AR) binding to this ARE was confirmed by Chromatin immunoprecipitation (ChIP) assay. Furthermore, Knockdown of PTTG1 expression using short hairpin RNA significantly reduced androgen-induced LNCaP cell growth and invasion. In addition, we showed PTTG1 is highly expressed in metastasis prostate cancer tissue. These results suggest that PTTG1 is a novel downstream target gene of androgen receptor and take part in prostate cancer proliferation and metastasis.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Próstata/metabolismo , Neoplasias da Próstata/genética , Receptores Androgênicos/metabolismo , Securina/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores Androgênicos/genética , Securina/metabolismo
8.
J Biol Chem ; 291(2): 959-67, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26507658

RESUMO

Ubiquitin modification at double strand breaks (DSB) sites is an essential regulator of signaling and repair. γH2AX extends from DSB sites and provides a platform for subsequent recruitment and amplification of DNA repair proteins and signaling factors. Here, we found that RNF8/RNF168 ubiquitylates γH2AX. We identified that USP11 is a unique deubiquitylation enzyme for γH2AX. USP11 deubiquitylates γH2AX both in vivo and in vitro but not the canonical (ub)-K119-H2A and (ub)-K120-H2B in vitro, and USP11 ablation enhances the levels of γH2AX ubiquitylation. We also found that USP11 interacts with γH2AX both in vivo and in vitro. We found that 53BP1 and ubiquitin-conjugated proteins are misregulated to be retained longer and stronger at DSB sites after knockdown of USP11. We further found that cells are hypersensitive to γ-irradiation after ablation of USP11. Together, our findings elucidate deeply and extensively the mechanism of RNF8/RNF168 and USP11 to maintain the proper status of ubiquitylation γH2AX to repair DSB.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Tioléster Hidrolases/metabolismo , Ubiquitinação , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos da radiação , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligação Proteica/efeitos da radiação , Radiação Ionizante , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina-Proteína Ligases , Ubiquitinação/efeitos da radiação
9.
J Biol Chem ; 291(19): 10307-17, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26975375

RESUMO

Parkin, a ubiquitin E3 ligase, is mutated in most cases of autosomal recessive early onset Parkinson disease. It was discovered that Parkin is also mutated in glioblastoma and other human malignancies and that it inhibits tumor cell growth. Here, we identified pyruvate kinase M2 (PKM2) as a unique substrate for parkin through biochemical purification. We found that parkin interacts with PKM2 both in vitro and in vivo, and this interaction dramatically increases during glucose starvation. Ubiquitylation of PKM2 by parkin does not affect its stability but decreases its enzymatic activity. Parkin regulates the glycolysis pathway and affects the cell metabolism. Our studies revealed the novel important roles of parkin in tumor cell metabolism and provided new insight for therapy of Parkinson disease.


Assuntos
Glucose/metabolismo , Glicólise , Doença de Parkinson/enzimologia , Piruvato Quinase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Linhagem Celular , Glucose/genética , Humanos , Doença de Parkinson/genética , Doença de Parkinson/patologia , Piruvato Quinase/química , Piruvato Quinase/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética
10.
J Biol Chem ; 290(50): 30175-92, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26491010

RESUMO

Long non-coding RNAs (lncRNAs) have recently emerged as key players in many physiologic and pathologic processes. Although many lncRNAs have been identified, few lncRNAs have been characterized functionally in aging. In this study, we used human fibroblast cells to investigate genome-wide lncRNA expression during cellular senescence. We identified 968 down-regulated lncRNAs and 899 up-regulated lncRNAs in senescent cells compared with young cells. Among these lncRNAs, we characterized a senescence-associated lncRNA (SALNR), whose expression was reduced during cellular senescence and in premalignant colon adenomas. Overexpression of SALNR delayed cellular senescence in fibroblast cells. Furthermore, we found that SALNR interacts with NF90 (nuclear factor of activated T-cells, 90 kDa), an RNA-binding protein suppressing miRNA biogenesis. We demonstrated that NF90 is a SALNR downstream target, whose inhibition led to premature senescence and enhanced expressions of senescence-associated miRNAs. Moreover, our data showed that Ras-induced stress promotes NF90 nucleolus translocation and suppresses its ability to suppress senescence-associated miRNA biogenesis, which could be rescued by SALNR overexpression. These data suggest that lncRNA SALNR modulates cellular senescence at least partly through changing NF90 activity.


Assuntos
Senescência Celular/genética , Proteínas do Fator Nuclear 90/fisiologia , Oncogenes , RNA Longo não Codificante/genética , Nucléolo Celular/metabolismo , Células Cultivadas , Estudo de Associação Genômica Ampla , Humanos , Transporte Proteico
11.
FASEB J ; 29(7): 2883-93, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25805831

RESUMO

Calcium and calcium channels are closely related to the estrogen-induced nongenomic effect of endometrial carcinoma, but the specific role of calcium channels is unknown. This study aimed to explore the expression and the biologic effect of the L-type calcium channel in endometrial carcinoma cells and to clarify the molecular mechanism of the relationship between L-type calcium channels and estrogen. The immunohistochemical results showed that Ca(2+) channel subunit α 1D (Cav1.3) expression was high in atypical hyperplasia (1.90 ± 0.35) and endometrial carcinoma tissues (2.05 ± 0.82) but weak (0.80 ± 0.15) in benign endometrial tissues (P < 0.05). Treatment with 17ß-estradiol rapidly increased Cav1.3 expression in a dose- and time-dependent manner, and 100 nM cell-impermeable ß-estradiol-6-(O-carboxymethyl)oxime:bovine serum albumin also promoted Cav1.3 expression. Transfection with small interfering RNA against G protein-coupled estrogen receptor (GPER) suppressed estrogen-induced up-regulation of Cav1.3 compared with control cells and markedly reduced the estrogen-induced phosphorylation of ERK1/2 and CREB. Knocking down the Cav1.3 significantly suppressed estrogen-stimulated Ca(2+) influx, cell proliferation, and migration in endometrial cancer cells. Taken together, Cav1.3 was overexpressed in atypical hyperplasia and endometrial carcinoma, and the estrogen-induced phosphorylation of downstream molecular ERK1/2 and CREB is the result of activation of the GPER pathway. L-type channel Cav1.3 is required for estrogen-stimulated Ca(2+) influx and contributes broadly to the development of endometrial cancer. The Cav1.3 channel may be a new target for endometrial carcinoma treatment.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Estradiol/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Idoso , Animais , Canais de Cálcio Tipo L/genética , Sinalização do Cálcio , Bovinos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases , Pessoa de Meia-Idade , RNA Interferente Pequeno/genética , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética
12.
Int J Cancer ; 136(11): 2524-34, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25382637

RESUMO

To study the mechanisms underlying the IL-6-promoted angiogenic microenvironment in EGFRvIII-positive glioblastoma, VEGF expression in EGFRvIII-positive/negative tumors was determined by optical molecular imaging. Next, the HUVEC tube formation assay, Western blot, qPCR, RNA silencing, chromatin immunoprecipitation, luciferase reporter and ELISA assays were performed to examine the role of IL-6 and C/EBPß in the formation of the angiogenic microenvironment in EGFRvIII-positive tumors. Finally, in vitro and in vivo genistein treatment experiments were conducted to challenge the interaction between the IL-6 promoter and C/EBPß. Optical imaging revealed greater VEGF expression in EGFRvIII-positive tumor-bearing mice, suggesting an angiogenic microenvironment. In vitro experiments demonstrated that C/EBPß-mediated regulation of IL-6 was indispensable for maintenance of this angiogenic microenvironment. In contrast, genistein-mediated upregulation of CHOP impeded C/EBPß interaction with the IL-6 promoter, thus disturbing the angiogenic microenvironment. This more malignant microenvironment in EGFRvIII glioblastoma is generated, at least in part, by greater VEGF, IL-6 and C/EBPß expression. Interaction of C/EBPß with the IL-6 promoter maintains this angiogenic microenvironment, while disturbance of this dynamically balanced interaction inhibits EGFRvIII tumor proliferation by reducing both VEGF and IL-6 expression.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Receptores ErbB/metabolismo , Genisteína/farmacologia , Glioblastoma/metabolismo , Interleucina-6/metabolismo , Fator de Transcrição CHOP/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Interleucina-6/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neovascularização Patológica , Regiões Promotoras Genéticas , Microambiente Tumoral
13.
Biochem Biophys Res Commun ; 450(1): 470-5, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24909689

RESUMO

DNA damage may regulate microRNA (miRNA) biosynthesis at the levels of miRNA transcription, processing and maturation. Although involvement of E2F1 in the regulation of miRNA gene activation in response to DNA damage has been documented, little is known about the role of E2F1 in miRNA processing. In this study we demonstrate that E2F1 enhances miR-630 biosynthesis under cisplatin (CIS) exposure through promoting DROSHA-mediated pri-miR-630 processing. Northern blot and RT-qPCR revealed that CIS exposure caused not only an increase in pri-miR-630 but also much more increase in pre-miR-630 and mature miR-630. The increases in pri-miR-630 and pre-miR-630 expression in unmatched proportion indicated that primary transcript processing was involved in CIS-stimulated miR-630 biosynthesis. Furthermore, combination of reporter enzyme assay with mutation and over-expression of E2F1 showed that induction of DROSHA promoted miR-630 expression, in which CIS-induced E2F1 activated DROSHA gene expression by recognizing and binding two E2F1 sites at the positions -214/-207 and -167/-160 of the DROSHA promoter. The increased binding of E2F1 to the DROSHA promoter in CIS-exposed cells was further evidenced by chromatin immunoprecipitation assay. Together, E2F1-regulated DROSHA promotes pri-miR-630 processing, thereby, contributes to CIS-stimulated miR-630 expression. The involvement of E2F1-dependent DROSHA activation in pri-miRNA processing under DNA damage stress will provide further insight into the regulation of miRNA biosynthesis. These data also give us a deeper understanding of E2F1 role in response to DNA damage.


Assuntos
Cisplatino/farmacologia , Fator de Transcrição E2F1/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , MicroRNAs/biossíntese , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Ribonuclease III/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Humanos
14.
Biol Cell ; 105(10): 488-99, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23746120

RESUMO

BACKGROUND INFORMATION: The mitogen-inducible gene-6 (Mig-6) is a non-kinase scaffolding adaptor protein. It has been shown that Mig-6 may play important roles in regulating stress response, maintaining homeostasis and functioning as a tumour suppressor. In this study, we investigated the role of Mig-6 in cellular senescence. RESULTS: Our results showed that Mig-6 is up-regulated during the senescence process. Functional analysis indicated that cells over-expressing Mig-6 have reduced DNA synthesis and showed the signs of senescence. Knockdown of Mig-6 delayed the initiation of Ras-induced cellular senescence. These results suggest that the increase of Mig-6 expression contributes to establishment of cellular senescence. Furthermore, our results showed that Mig-6 induction of senescence is related to its inhibition of EGF receptor (EGFR)/Erb B signalling. Subsequent analysis of the mechanism responsible for the up-regulation of its expression showed that FOXO3A transcriptionally up-regulates Mig-6 expression via directly binding to the FOXO response element in Mig-6 5'-flanking regulatory sequences. CONCLUSIONS: Mig-6 induces premature senescence via functioning in regulation of cellular senescence in normal diploid fibroblasts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Senescência Celular , Diploide , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Humanos , Elementos de Resposta/genética , Proteínas Supressoras de Tumor/genética , Regulação para Cima
15.
Cancer Lett ; 598: 217109, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39002692

RESUMO

Gemcitabine serves as a first-line chemotherapeutic treatment for pancreatic cancer (PC), but it is prone to rapid drug resistance. Increasing the sensitivity of PC to gemcitabine has long been a focus of research. Fasting interventions may augment the effects of chemotherapy and present new options. SIRT7 is known to link metabolism with various cellular processes through post-translational modifications. We found upregulation of SIRT7 in PC cells is associated with poor prognosis and gemcitabine resistance. Cross-analysis of RNA-seq and ATAC-seq data suggested that GLUT3 might be a downstream target gene of SIRT7. Subsequent investigations demonstrated that SIRT7 directly interacts with the enhancer region of GLUT3 to desuccinylate H3K122. Our group's another study revealed that GLUT3 can transport gemcitabine in breast cancer cells. Here, we found GLUT3 KD reduces the sensitivity of PC cells to gemcitabine, and SIRT7 KD-associated gemcitabine-sensitizing could be reversed by GLUT3 KD. While fasting mimicking induced upregulation of SIRT7 expression in PC cells, knocking down SIRT7 enhanced sensitivity to gemcitabine through upregulating GLUT3 expression. We further confirmed the effect of SIRT7 deficiency on the sensitivity of gemcitabine under fasting conditions using a mouse xenograft model. In summary, our study demonstrates that SIRT7 can regulate GLUT3 expression by binding to its enhancer and altering H3K122 succinylation levels, thus affecting gemcitabine sensitivity in PC cells. Additionally, combining SIRT7 knockdown with fasting may improve the efficacy of gemcitabine. This unveils a novel mechanism by which SIRT7 influences gemcitabine sensitivity in PC and offer innovative strategies for clinical combination therapy with gemcitabine.

16.
J Gastroenterol Hepatol ; 28(2): 365-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23339388

RESUMO

BACKGROUND AND AIM: Arginine is a nonessential amino acid for humans and mice because it can be synthesized from citrulline by argininosuccinate synthetase (ASS) and argininosuccinate lyase. Hepatocellular carcinoma (HCC) is believed to be auxotrophic for arginine through the lack of expression of ASS. However, there are also some ASS-positive HCC cells. Therefore, the aim of this article was to study the levels of arginine and the expression of ASS in patients with HCC. METHODS: Thirty patients with HCC who had undergone HCC surgery were enrolled in the study. Serum arginine levels were determined with an automatic amino acid analyzer. ASS expression was examined by Western blot and immunohistochemistry. Methylation specific polymerase chain reaction and methylation sequencing were performed to detect the methylation of DNA encoding ASS. RESULTS: There was a decrease of arginine in HCC patients compared with that of healthy control. High expression of ASS was found in the adjacent tissues by Western blot and immunohistochemistry. Little ASS expression was found in most HCC tissues, but there were also some HCC tissues that expressed low levels of ASS. Methylation of the DNA encoding ASS was obviously higher in HCC tissues than that in paired adjacent tissues. CONCLUSIONS: ASS expression is decreased significantly in HCC tissues. The downregulation of arginine and ASS expression may be a self-defense action of the body against malignant tumors, and the decreased arginine and ASS levels in HCC patients are an advantage for the arginine deiminase treatment.


Assuntos
Argininossuccinato Sintase/análise , Biomarcadores Tumorais/análise , Carcinoma Hepatocelular/enzimologia , Neoplasias Hepáticas/enzimologia , Adulto , Arginina/sangue , Western Blotting , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/cirurgia , Estudos de Casos e Controles , Metilação de DNA , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/cirurgia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Invest Ophthalmol Vis Sci ; 64(12): 39, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37750741

RESUMO

Purpose: Wet AMD (wAMD) is associated with cellular senescence. However, senescent cell-targeted therapies for wAMD have rarely been comprehensively studied. This study aimed to explore the therapeutic effects of senolytic agents on choroidal neovascularization (CNV). Methods: RNA sequencing datasets were obtained from the Gene Expression Omnibus database and used to explore the association between senescence and wAMD. We explored the effects of senescent adult RPE cell line-19 cells on the proliferation, migration, invasion, and tube formation of human umbilical vein endothelial cells. A laser-induced CNV animal model was used to study wAMD. We studied a senescent cell elimination therapy for CNV progression using two types of senolytics and a transgenic method. Results: Cells in the retinal pigment epithelium-choroid of the CNV model were enriched in senescence, inflammation, and angiogenesis gene sets. AP20187 was used to specifically eliminate senescent cells and proven to alleviate CNV progression in INK-ATTAC transgenic mice. Senescent adult RPE cell line-1 cells produced elevated levels of senescence-associated secretory phenotypes, including VEGFs; they also demonstrated increased proliferation, migration, invasion, and tube formation in human umbilical vein endothelial cells. The number of senescent cells increased in the laser-induced CNV rat model, and intravitreal injections of dasatinib with quercetin reduced the expression of p16 in CNV and alleviated neovascularization. Conclusions: Senescent RPE cells can accelerate pathological neovascularization; thus, senescent cell-targeting therapy has great clinical potential for wAMD.


Assuntos
Neovascularização de Coroide , Quercetina , Adulto , Humanos , Camundongos , Animais , Ratos , Dasatinibe/farmacologia , Quercetina/farmacologia , Quercetina/uso terapêutico , Epitélio Pigmentado da Retina , Neovascularização de Coroide/tratamento farmacológico , Neovascularização de Coroide/genética , Senescência Celular , Corioide , Células Endoteliais da Veia Umbilical Humana , Metildopa
18.
Int J Mol Sci ; 13(3): 3563-3582, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22489168

RESUMO

Autophagy is a highly regulated intracellular process involved in the turnover of most cellular constituents and in the maintenance of cellular homeostasis. In this study, we show that the activity of autophagy increases in H(2)O(2) or RasV12-induced senescent fibroblasts. Inhibiting autophagy promotes cell apoptosis in senescent cells, suggesting that autophagy activation plays a cytoprotective role. Furthermore, our data indicate that the increase of autophagy in senescent cells is linked to the activation of transcription factor FoxO3A, which blocks ATP generation by transcriptionally up-regulating the expression of PDK4, an inhibitor of pyruvate dehydrogenase complex, thus leading to AMPK activation and mTOR inhibition. These findings suggest a novel mechanism by which FoxO3A factors can activate autophagy via metabolic alteration.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/fisiologia , Senescência Celular/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Quinases Ativadas por AMP/biossíntese , Trifosfato de Adenosina/biossíntese , Apoptose , Proliferação de Células , Células Cultivadas , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/patologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Piruvato Desidrogenase Quinase de Transferência de Acetil , Complexo Piruvato Desidrogenase/antagonistas & inibidores , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Serina-Treonina Quinases TOR/antagonistas & inibidores
19.
J Funct Biomater ; 14(1)2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36662053

RESUMO

Senescent cells accumulate in aged organisms and promote the progression of age-related diseases including cataracts. Therefore, we aimed to study the therapeutic effects of senescence-targeting drugs on cataracts. In this study, a 28-day D-galactose-induced cataract rat model was used. The opacity index, a grading based on slit-lamp observations, was used to assess lens cloudiness. Furthermore, the average lens density (ALD), lens density standard deviation (LDSD), and maximum lens density (MLD) obtained from Scheimpflug images were used to assess lens transparency. Immunohistochemical stainings for p16 and γH2AX were used as hallmarks of senescence. We treated rat cataract models with the senolytic drug combination dasatinib plus quercetin (D+Q) and senescence-associated secretory phenotype (SASP) inhibitors. In comparison to control lenses, D-galactose-induced cataract lenses showed a higher opacity index, ALD, LDSD, and MLD values, as well as accumulation of senescent lens epithelial cells (LECs). After D+Q treatment, ALD, LDSD, and MLD values on day 21 were significantly lower than those of vehicle-treated model rats. The expression levels of p16 and γH2AX were also reduced after D+Q administration. In addition, the SASP inhibitor rapamycin decreased the opacity index, ALD, LDSD, and MLD values on day 21. In conclusion, D+Q alleviated D-galactose-induced cataract progression by reducing the senescent LEC burden in the early stage of cataract.

20.
Chin Med J (Engl) ; 135(19): 2326-2337, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36535010

RESUMO

BACKGROUND: Cell competition is an important feature in pancreatic cancer (PC) progression, but the underlying mechanism remains elusive. This study aims to explore the role of exosomes derived from normal pancreatic ductal epithelial cells involved in PC progression. METHODS: PC cells and pancreatic stellate cells (PSCs) were treated with exosomes isolated from pancreatic ductal epithelial cells. Cell proliferation was assessed by CCK8 assays. Cell migration and invasion were assessed by Transwell assays. PC and matched adjacent non-tumor tissue specimens were obtained from 46 patients pathologically diagnosed with PC at Peking University First Hospital from 2013 to 2017. Tissue miR-485-3p and p21-activated kinase-1 (PAK1) expression was examined by real-time polymerase chain reaction (RT-PCR), and the relationship of the two was analyzed using Pearman's product-moment correlation. The clinical significance of miR-485-3p was analyzed using the Chi-square test, Wilcoxon rank-sum test, and Fisher exact probability, respectively. The binding of miR-485-3p to PAK1 5'-untranslated region (5'-UTR) was examined by luciferase assay. PC cells were xenografted into nude mice as a PC metastasis model. RESULTS: Exosomes from pancreatic ductal epithelial cells suppressed PC cell migration and invasion as well as the secretion and migration of PSCs. MiR-485-3p was enriched in the exosomes of pancreatic ductal epithelial cells but deficient in those of PC cells and PSCs, in accordance with the lower level in PSCs and PC cells than that in pancreatic ductal cells. And the mature miR-485-3p could be delivered into these cells by the exosomes secreted by normal pancreatic duct cells, to inhibit PC cell migration and invasion. Clinical data analysis showed that miR-485-3p was significantly decreased in PC tissues (P < 0.05) and was negatively associated with lymphovascular invasion (P = 0.044). As a direct target of miR-485-3p, PAK1 was found to exert an inhibitory effect on PC cells, and there was a significantly negative correlation between the expression levels of miR-485-3p and PAK1 (r = -0.6525, P < 0.0001) in PC tissues. Moreover, miR-485-3p could suppress PC metastasis in vivo by targeting p21-activated kinase-1. CONCLUSIONS: Exosomal miR-485-3p delivered by normal pancreatic ductal epithelial cells into PC cells inhibits PC metastasis by directly targeting PAK1. The restoration of miR-485-3p by exosomes or some other vehicle might be a novel approach for PC treatment.


Assuntos
MicroRNAs , Ductos Pancreáticos , Neoplasias Pancreáticas , Quinases Ativadas por p21 , Animais , Camundongos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , MicroRNAs/metabolismo , Quinases Ativadas por p21/metabolismo , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Humanos , Exossomos , Metástase Neoplásica , Neoplasias Pancreáticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA