Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Mol Med ; 25(9): 4195-4203, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33745198

RESUMO

To determine whether the deletion of p16 can correct tooth and mandible growth retardation caused by Bmi1 deficiency, we compared the tooth and mandible phenotypes of homozygous p16-deficient (p16-/- ) mice, homozygous Bmi1-deficient (Bmi1-/- ) mice, double homozygous Bmi1 and p16-deficient (Bmi1-/- p16-/- ) mice to those of their wild-type littermates at 4 weeks of age by radiograph, histochemistry and immunohistochemistry. Results showed that compared to Bmi1-/- mice, the dental mineral density, dental volume and dentin sialoprotein immunopositive areas were increased, whereas the ratio of the predentin area to total dentin area and that of biglycan immunopositive area to dentin area were decreased in Bmi1-/- p16-/- mice. These results indicate that the deletion of p16 can improve tooth development in Bmi1 knockout mice. Compared to Bmi1-/- mice, the mandible mineral density, cortical thickness, alveolar bone volume, osteoblast number and activity, alkaline phosphatase positive area were all increased significantly in Bmi1-/- p16-/- mice. These results indicate that the deletion of p16 can improve mandible growth in Bmi1 knockout mice. Furthermore, the protein expression levels of cyclin D, CDK4 and p53 were increased significantly in p16-/- mice compared with those from wild-type mice; the protein expression levels of cyclin D and CDK4 were decreased significantly, whereas those of p27 and p53 were increased significantly in Bmi1-/- mice; these parameters were partly rescued in Bmi1-/- p16-/- mice compared with those from Bmi1-/- mice. Therefore, our results indicate that Bmi1 plays roles in regulating tooth and mandible development by inhibiting p16 signal pathway which initiated entry into cell cycle.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/antagonistas & inibidores , Mandíbula/crescimento & desenvolvimento , Osteoblastos/citologia , Osteogênese , Complexo Repressor Polycomb 1/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Dente/crescimento & desenvolvimento , Animais , Ciclo Celular , Proliferação de Células , Camundongos , Camundongos Knockout , Transdução de Sinais
2.
Biochem Biophys Res Commun ; 482(4): 742-749, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27871857

RESUMO

The regeneration of injured tubular cell occurs primarily from intrinsic renal stem/progenitor cells (RSCs) labeled with CD24 and CD133 after acute tubular necrosis (ATN). Bmi-1 plays a crucial role in regulating self-renewal, differentiation and aging of multiple adult stem cells and progenitor cells. Bmi-1 was rapidly elevated in the induction of adult kidney regeneration by renal injury. To determine whether Bmi-1 maintained mobilization of RSCs in the protection from ATN, glycerol-rhabdomyolysis-induced ATN were performed in wild type (WT) and Bmi-1-deficient (Bmi-1-/-) mice. Their ATN phenotypes were analyzed; CD24 and CD133 double positive (CD24+CD133+) cells were measured; and the levels of serum urea nitrogen (SUN) and serum creatinine (SCr) were detected. We found that CD24+CD133+ RSCs were mobilized in WT ATN mice with the increased expression of Bmi-1; Bmi-1 deficiency led to increased tubular cast formation and necrosis, elevated levels of SUN and SCr, decreased tubular proliferation, and immobilized ratio of RSCs in ATN. These findings indicated that Bmi-1 played a critical role in the protection from ATN by maintaining mobilization of RSCs and would be a novel therapeutic target for preventing the progression of ATN.


Assuntos
Necrose Tubular Aguda/metabolismo , Rim/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células-Tronco/citologia , Antígeno AC133/metabolismo , Animais , Antígeno CD24/metabolismo , Diferenciação Celular , Creatinina/metabolismo , Progressão da Doença , Glicerol/química , Masculino , Camundongos , Regeneração , Rabdomiólise/metabolismo
3.
Exp Mol Med ; 52(1): 130-151, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31959867

RESUMO

To study whether TGF-ß1/IL-11/MEK/ERK (TIME) signaling mediates senescence-associated pulmonary fibrosis (SAPF) in Bmi-1-deficient (Bmi-1-/-) mice and determines the major downstream mediator of Bmi-1 and crosstalk between p16INK4a and reactive oxygen species that regulates SAPF, phenotypes were compared among 7-week-old p16INK4a and Bmi-1 double-knockout, N-acetylcysteine (NAC)-treated Bmi-1-/-, Bmi-1-/-, and wild-type mice. Pulmonary fibroblasts and alveolar type II epithelial (AT2) cells were used for experiments. Human pulmonary tissues were tested for type Ι collagen, α-smooth muscle actin (α-SMA), p16INK4a, p53, p21, and TIME signaling by using enzyme-linked immunosorbent assay (ELISA). Our results demonstrated that Bmi-1 deficiency resulted in a shortened lifespan, ventilatory resistance, poor ventilatory compliance, and SAPF, including cell senescence, DNA damage, a senescence-associated secretory phenotype and collagen overdeposition that was mediated by the upregulation of TIME signaling. The signaling stimulated cell senescence, senescence-related secretion of TGF-ß1 and IL-11 and production of collagen 1 by pulmonary fibroblasts and the epithelial-to-mesenchymal transition of AT2 cells. These processes were inhibited by anti-IL-11 or the MEK inhibitor PD98059. NAC treatment prolonged the lifespan and ameliorated pulmonary dysfunction and SAPF by downregulating TIME signaling more than p16INK4a deletion by inhibiting oxidative stress and DNA damage and promoting ubiquitin-proteasome degradation of p16INK4a and p53. Cytoplasmic p16INK4a accumulation upregulated MEK/ERK signaling by inhibiting the translocation of pERK1/2 (Thr202/Tyr204) from the cytoplasm to the nucleus in senescent fibroblasts. The accumulation of collagen 1 and α-SMA in human lungs accompanied by cell senescence may be mediated by TIME signaling. Thus, this signaling in aging fibroblasts or AT2 cells could be a therapeutic target for preventing SAPF.


Assuntos
Senescência Celular/fisiologia , Fibrose/metabolismo , Pulmão/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Dano ao DNA/fisiologia , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/patologia , Humanos , Interleucina-11/metabolismo , Pulmão/patologia , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Estresse Oxidativo/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Ubiquitina/metabolismo
5.
Sci Rep ; 7(1): 7502, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28790310

RESUMO

To determine whether p16 INK4a deletion ameliorated renal tubulointerstitial injury by inhibiting a senescence-associated secretory phenotype (SASP) in Bmi-1-deficient (Bmi-1 -/-) mice, renal phenotypes were compared among 5-week-old Bmi-1 and p16 INK4a double-knockout, and Bmi-1 -/- and wild-type mice. Fifth-passage renal interstitial fibroblasts (RIFs) from the three groups were analyzed for senescence and proliferation. The effect of Bmi-1 deficiency on epithelial-to-mesenchymal transition (EMT) was examined in Bmi-1-knockdown human renal proximal tubular epithelial (HK2) cells, which were treated with concentrated conditioned medium (CM) from the fifth-passage renal interstitial fibroblasts (RIFs) of above three group mice or with exogenous TGF-ß1. Our results demonstrated that p16 INK4a deletion largely rescued renal aging phenotypes caused by Bmi-1 deficiency, including impaired renal structure and function, decreased proliferation, increased apoptosis, senescence and SASP, DNA damage, NF-κB and TGF-ß1/Smad signal activation, inflammatory cell infiltration, and tubulointerstitial fibrosis and tubular atrophy. P16 INK4a deletion also promoted proliferation, reduced senescence and SASP of RIFs and subsequently inhibited EMT of Bmi-1-knockdown HK2 cells. TGF-ß1 further induced the EMT of Bmi-1-knockdown HK2 cells. Thus, p16 INK4a positive senescent cells would be a therapeutic target for preventing renal tubulointerstitial injury.


Assuntos
Injúria Renal Aguda/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Fibroblastos/metabolismo , Nefrite Intersticial/genética , Complexo Repressor Polycomb 1/genética , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Animais , Linhagem Celular Transformada , Proliferação de Células , Senescência Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Células Epiteliais/patologia , Fibroblastos/patologia , Regulação da Expressão Gênica , Humanos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Nefrite Intersticial/metabolismo , Nefrite Intersticial/patologia , Nefrite Intersticial/prevenção & controle , Complexo Repressor Polycomb 1/antagonistas & inibidores , Complexo Repressor Polycomb 1/deficiência , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
6.
Cell Death Dis ; 7(10): e2423, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27763638

RESUMO

Impaired apoptosis of fibroblast-like synoviocytes (FLSs) causes synovial hyperplasia, facilitating destruction of cartilage and bone in rheumatoid arthritis (RA). Tumor necrosis factor (TNF)-α, a dominant inflammatory mediator in RA pathogenesis, promotes progression of RA symptoms. Prevalence of 1, 25-dihydroxy-vitamin D3 (hereafter termed VD) deficiency is 30-63% in patients with RA. Whether VD leads to apoptosis or enhances TNF-α-mediated apoptosis in FLSs to ameliorate RA is unclear. To determine this, 10-week-old CYP27B1-deficient (CYP27B1-/-) mice with collagen-induced arthritis (CIA) were intraperitoneally treated with 1 µg/kg VD every other day for 9 weeks. RA phenotypes were compared between vehicle-treated CYP27B1-/- and wild-type CIA mice. Human rheumatoid FLS-MH7A cells were treated with Dulbecco's modified Eagle's medium (DMEM) without fetal bovine serum (FBS) for 24 h, then with different concentrations of VD and TNF-α, human vitamin D receptor (VDR) siRNA or the p53 pro-apoptotic inhibitor pifithrin-α. Apoptosis and p53 pro-apoptotic signaling were analyzed. The 19-week-old vehicle-treated CYP27B1-/- CIA mice had increased cumulative arthritis scores and levels of serous rheumatoid factors and C-reactive protein. They had exacerbated articular cartilage and bone destruction, joint space narrowing, joint stiffness, deformity and dysfunction, synovitis and TNF-α secretion, FLS hyperplasia with increased proliferation and decreased apoptosis compared to CIA mice. These RA phenotypes that were aggravated in CIA mice by CYP27B1 deficiency were largely rescued by VD treatment. In vitro, VD with TNF-α treatment upregulated p53 acetylation-mediated apoptosis in MH7A cells by promoting Sirt1 translocation from the nucleus to the cytoplasm. These findings indicated that VD with TNF-α protected against RA by promoting apoptosis of FLSs. The results indicated that clinical administration of VD could be a specific therapy to promote FLS apoptosis and prevent RA progression.


Assuntos
Apoptose/efeitos dos fármacos , Artrite Reumatoide/tratamento farmacológico , Substâncias Protetoras/uso terapêutico , Sirtuína 1/metabolismo , Sinoviócitos/metabolismo , Fator de Necrose Tumoral alfa/uso terapêutico , Proteína Supressora de Tumor p53/metabolismo , Vitamina D/análogos & derivados , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Acetilação/efeitos dos fármacos , Animais , Artrite Experimental/patologia , Artrite Reumatoide/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Cartilagem/efeitos dos fármacos , Cartilagem/patologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Inflamação/patologia , Camundongos Endogâmicos BALB C , Substâncias Protetoras/farmacologia , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Membrana Sinovial/patologia , Sinoviócitos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Vitamina D/sangue , Vitamina D/uso terapêutico
7.
Sci Rep ; 5: 13975, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26370922

RESUMO

To determine whether transplanted amniotic membrane mesenchymal stem cells (AMSCs) ameliorated the premature senescent phenotype of Bmi-1-deficient mice, postnatal 2-day-old Bmi-1(-/-) mice were injected intraperitoneally with the second-passage AMSCs from amniotic membranes of ß-galactosidase (ß-gal) transgenic mice or wild-type (WT) mice labeled with DiI. Three reinjections were given, once every seven days. Phenotypes of 5-week-old ß-gal(+) AMSC-transplanted or 6-week-old DiI(+) AMSC-transplanted Bmi-1(-/-) mice were compared with vehicle-transplanted Bmi-1(-/-) and WT mice. Vehicle-transplanted Bmi-1(-/-) mice displayed growth retardation and premature aging with decreased cell proliferation and increased cell apoptosis; a decreased ratio and dysmaturity of lymphocytic series; premature osteoporosis with reduced osteogenesis and increased adipogenesis; redox imbalance and DNA damage in multiple organs. Transplanted AMSCs carried Bmi-1 migrated into multiple organs, proliferated and differentiated into multiple tissue cells, promoted growth and delayed senescence in Bmi-1(-/-) transplant recipients. The dysmaturity of lymphocytic series were ameliorated, premature osteoporosis were rescued by promoting osteogenesis and inhibiting adipogenesis, the oxidative stress and DNA damage in multiple organs were inhibited by the AMSC transplantation in Bmi-1(-/-) mice. These findings indicate that AMSC transplantation ameliorated the premature senescent phenotype of Bmi-1-deficient mice and could be a novel therapy to delay aging and prevent aging-associated degenerative diseases.


Assuntos
Senilidade Prematura/genética , Âmnio/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Complexo Repressor Polycomb 1/deficiência , Proteínas Proto-Oncogênicas/deficiência , Senilidade Prematura/terapia , Animais , Antígenos de Superfície/metabolismo , Diferenciação Celular/genética , Movimento Celular/genética , Proliferação de Células , Dano ao DNA , Modelos Animais de Doenças , Imunofenotipagem , Contagem de Linfócitos , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Osteoporose/genética , Osteoporose/patologia , Oxirredução , Fenótipo
8.
Aging Cell ; 13(5): 797-809, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24915841

RESUMO

To determine whether Bmi-1 deficiency could lead to renal tubulointerstitial injury by mitochondrial dysfunction and increased oxidative stress in the kidney, 3-week-old Bmi-1(-/-) mice were treated with the antioxidant N-acetylcysteine (NAC, 1 mg mL(-1) ) in their drinking water, or pyrro-quinoline quinone (PQQ, 4 mg kg(-1) diet) in their diet for 2 weeks, and their renal phenotypes were compared with vehicle-treated Bmi1(-/-) and wild-type mice. Bmi-1 was knocked down in human renal proximal tubular epithelial (HK2) cells which were treated with 1 mm NAC for 72 or 96 h, and their phenotypes were compared with control cells. Five-week-old vehicle-treated Bmi-1(-/-) mice displayed renal interstitial fibrosis, tubular atrophy, and severe renal function impairment with decreased renal cell proliferation, increased renal cell apoptosis and senescence, and inflammatory cell infiltration. Impaired mitochondrial structure, decreased mitochondrial numbers, and increased oxidative stress occurred in Bmi-1(-/-) mice; subsequently, this caused DNA damage, the activation of TGF-ß1/Smad signaling, and the imbalance between extracellular matrix synthesis and degradation. Oxidative stress-induced epithelial-to-mesenchymal transition of renal tubular epithelial cells was enhanced in Bmi-1 knocked down HK2 cells. All phenotypic alterations caused by Bmi-1 deficiency were ameliorated by antioxidant treatment. These findings indicate that Bmi-1 plays a critical role in protection from renal tubulointerstitial injury by maintaining redox balance and will be a novel therapeutic target for preventing renal tubulointerstitial injury.


Assuntos
Nefropatias/genética , Nefropatias/prevenção & controle , Rim/patologia , Proteína Quinase 7 Ativada por Mitógeno/deficiência , Proteína Quinase 7 Ativada por Mitógeno/genética , Animais , Linhagem Celular Tumoral , Dano ao DNA , Transição Epitelial-Mesenquimal , Técnicas de Genotipagem , Humanos , Rim/metabolismo , Nefropatias/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Nefrite Intersticial , Oxirredução , Estresse Oxidativo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA