Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 41(6): 672-683, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37099695

RESUMEN

Bone marrow mesenchymal stem cells (BMSCs) are indispensable cells constituting the bone marrow microenvironment that are generally recognized as being involved in the development and progression of osteosarcoma (OS). To explore whether mTORC2 signaling inhibition in BMSCs suppressed OS growth and tumor-caused bone destruction, 3-month-old littermates genotyped Rictorflox/flox or Prx1-cre; Rictorflox/flox (with same gender) were injected with K7M2 cells in the proximal tibia. After 40 days, bone destruction was alleviated in Prx1-cre; Rictorflox/flox mice, as observed on X-ray and micro-CT. This was accompanied by decreased serum N-terminal propeptide of procollagen type I (PINP) levels and reduced tumor bone formation in vivo. Interactions between K7M2 and BMSCs were studied in vitro. Rictor-deficient BMSCs, which were cultured in tumor-conditioned medium (TCM), caused reduced bone proliferation and suppressed osteogenic differentiation. Moreover, compared with the control group, K7M2 cells cultured in BCM (culture medium extracted from Rictor-deficient BMSCs) displayed less proliferation, migration, and invasion, and attenuated osteogenic activity. Forty types of cytokines were then analyzed by mouse cytokine array and decreased levels CCL2/3/5 and interleukin-16 were detected in Rictor-deficient BMSCs. These results suggested that inhibition of mTORC2 (Rictor) signaling pathway in BMSCs exerted anti-OS effects through 2 mechanisms: (1) by suppressing the proliferation and osteogenic differentiation of BMSCs induced by OS to alleviate bone destruction; (2) by reducing the secretion of cytokines by BMSCs, which are closely related to OS cell growth, migration, invasion, and tumorigenic osteogenesis.


Asunto(s)
Neoplasias Óseas , Células Madre Mesenquimatosas , Osteosarcoma , Ratones , Animales , Osteogénesis , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular , Células de la Médula Ósea , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Citocinas/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Células Cultivadas , Osteosarcoma/metabolismo , Microambiente Tumoral
2.
BMC Pregnancy Childbirth ; 23(1): 151, 2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36890528

RESUMEN

BACKGROUND: Chemical agents such as alkylating agents (AAs) that are commonly used for the treatment of cancer cause great damage to the ovaries, thereby significantly increasing the risk of premature ovarian insufficiency (POI). However, the exact molecules underlying AA-induced POI remain largely obscure. Upregulation of the p16 gene may contribute to the progression of POI. As yet, no in vivo data from p16-deficient (KO) mice are available to demonstrate a critical role of p16 in POI. In the present study, we employed p16 KO mice to investigate whether loss of p16 could protect against POI caused by AAs. METHODS: WT mice and their p16 KO littermates received a single dose of BUL + CTX to establish an AA-induced POI mouse model. One month later, oestrous cycles were monitored. Three months later, some of the mice were sacrificed to collect sera for measurements of hormone levels and ovaries for measurements of follicle counts, the proliferation and apoptosis of granulosa cells, ovarian stromal fibrosis and vessels. The remaining mice were mated with fertile males for the fertility test. RESULTS: Our results showed that treatment with BUL + CTX significantly disrupted the oestrous cycles, increased the levels of FSH and LH while decreasing the levels of E2 and AMH, decreased the counts of primordial follicles and growing follicles while increasing the counts of atretic follicles, reduced the vascularized area in the ovarian stroma, and decreased fertility. All of these results were comparable between WT and p16 KO mice treated with BUL + CTX. In addition, ovarian fibrosis was not increased significantly in WT and p16 KO mice treated with BUL + CTX. Growing follicles with normal appearance had normally proliferating granulosa cells (without apparent apoptosis). CONCLUSION: We concluded that genetic ablation of the p16 gene did not attenuate ovarian damage or help preserve the fertility of mice challenged by AAs. This study demonstrated for the first time that p16 is dispensable for AA-induced POI. Our preliminary findings suggest that targeting p16 alone may not preserve the ovarian reserve and fertility of females treated with AAs.


Asunto(s)
Alquilantes , Insuficiencia Ovárica Primaria , Masculino , Femenino , Ratones , Humanos , Animales , Alquilantes/toxicidad , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/prevención & control , Insuficiencia Ovárica Primaria/genética , Folículo Ovárico , Células de la Granulosa
3.
FASEB J ; 35(4): e21345, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33715219

RESUMEN

Obesity is common in the middle aged population and it increases the risks of diabetes, cardiovascular diseases, certain cancers, and dementia. Yet, its etiology remains incompletely understood. Here, we show that ectopic expression of HB-EGF, an important regulator of neurogenesis, in Nestin+ neuroepithelial progenitors with the Cre-LoxP system leads to development of spontaneous middle age obesity in male mice accompanied by hyperglycemia and insulin resistance. The Nestin-HB-EGF mice show decreases in food uptake, energy expenditure, and physical activity, suggesting that reduced energy expenditure underlies the pathogenesis of this obesity model. However, HB-EGF expression in appetite-controlling POMC or AgRP neurons or adipocytes fails to induce obesity. Mechanistically, HB-EGF suppresses expression of Hypocretin/Orexin, an orexigenic neuropeptide hormone, in the hypothalamus of middle aged Nestin-HB-EGF mice. Hypothalamus Orexin administration alleviates the obese and hyperglycemic phenotypes in Nestin-HB-EGF mice. This study uncovers an important role for HB-EGF in regulating Orexin expression and energy expenditure and establishes a midlife obesity model whose pathogenesis involves age-dependent changes in hypothalamus neurons.


Asunto(s)
Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Nestina/metabolismo , Células-Madre Neurales/metabolismo , Obesidad/metabolismo , Orexinas/metabolismo , Adiponectina/sangre , Envejecimiento , Animales , Composición Corporal , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Humanos , Insulina/sangre , Leptina/sangre , Ratones , Nestina/genética , Orexinas/genética
4.
J Cell Mol Med ; 25(9): 4195-4203, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33745198

RESUMEN

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.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/antagonistas & inhibidores , Mandíbula/crecimiento & desarrollo , Osteoblastos/citología , Osteogénesis , Complejo Represivo Polycomb 1/fisiología , Proteínas Proto-Oncogénicas/fisiología , Diente/crecimiento & desarrollo , Animales , Ciclo Celular , Proliferación Celular , Ratones , Ratones Noqueados , Transducción de Señal
5.
Cancer Sci ; 112(11): 4553-4569, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34418240

RESUMEN

Numerous studies have reported that a variety of long noncoding RNAs (lncRNAs) can promote the proliferation, invasion, and migration of different tumor cells. However, different lncRNAs regulate cell functions in various forms, and the exact mechanisms are not clear. Here, we investigated the effect of the lncRNA ELF3-AS1 on gastric cancer (GC) cell function and explored the exact mechanism. Quantitative real-time polymerase chain reaction was used to detect the expression of ELF3-AS1 in GC tissues and adjacent nontumor tissues. Knockdown and overexpression of ELF3-AS1 was used to detect the effect of ELF3-AS1 on cell function. Potential downstream target genes were identified using RNA transcriptome sequencing, while RNA immunoprecipitation, chromatin immunoprecipitation, and Western blotting were performed to explore the tumor promotion mechanisms of ELF3-AS1. We observed that ELF3-AS1 was highly expressed in GC tissues, and high ELF3-AS1 expression predicted poor prognosis. The knockdown of ELF3-AS1 significantly inhibited cell proliferation, migration, and epithelial-mesenchymal transition and promoted apoptosis. Mechanistic investigations revealed that ELF3-AS1 may regulate the downstream target gene, C-C motif chemokine 20, by binding with the RNA-binding protein hnRNPK. Additionally, we found that high ELF3-AS1 expression was associated with thrombocytosis. Interleukin-6 and thrombopoietin may be involved in ELF3-AS1-induced paraneoplastic thrombocytosis. Together, our results demonstrate that aberrantly expressed ELF3-AS1 in GC may play important roles in oncogenesis and progression and is expected to become a new target for the diagnosis and treatment of GC.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Neoplasias Gástricas/metabolismo , Trombocitosis/etiología , Factores de Transcripción/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quimiocina CCL20/metabolismo , Proteínas de Unión al ADN/genética , Transición Epitelial-Mesenquimal , Femenino , Silenciador del Gen , Humanos , Interleucina-6/metabolismo , Masculino , Ratones Desnudos , Persona de Mediana Edad , Pronóstico , Proteínas Proto-Oncogénicas c-ets/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Trombopoyetina/metabolismo , Factores de Transcripción/genética , Secuenciación del Exoma
6.
J Cell Mol Med ; 24(16): 8950-8961, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32583517

RESUMEN

The transcriptional repressor Bmi-1 is involved in cell-cycle regulation and cell senescence, the deficiency of which has been shown to cause oxidative stress. This study investigated whether Bmi-1 deficiency plays a role in promoting disc degeneration and the effect of treatment with antioxidant N-acetylcysteine (NAC) on intervertebral disc degeneration. Bmi-1-/- mice were treated with the antioxidant NAC, supplied in drinking water (Bmi-1-/- +NAC). For in vitro experiments, mouse intervertebral discs were cultured under low oxygen tension and serum-limiting conditions in the presence of tumour necrosis factor α and interleukin 1ß in order to mimic degenerative insult. Disc metabolism parameters in these in vitro and in vivo studies were evaluated by histopathological, immunohistochemical and molecular methods. Bmi-1-/- mice showed lower collagen Ⅱ and aggrecan levels and higher collagen Ⅹ levels than wild-type and Bmi-1-/- +NAC mice. Bmi-1-/- mice showed significantly lower superoxide dismutase (SOD)-1, SOD-2, glutathione peroxidase (GPX)-1 and GPX-3 levels than their wild-type littermates and Bmi-1-/- + NAC mice. Relative to Bmi-1-/- mice, the control and Bmi-1-/- +NAC mice showed significantly lower p16, p21, and p53 levels. These results demonstrate that Bmi-1 plays an important role in attenuating intervertebral disc degeneration in mice by inhibiting oxidative stress and cell apoptosis.


Asunto(s)
Antioxidantes/fisiología , Degeneración del Disco Intervertebral/tratamiento farmacológico , Degeneración del Disco Intervertebral/metabolismo , Estrés Oxidativo/efectos de los fármacos , Complejo Represivo Polycomb 1/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Acetilcisteína/farmacología , Agrecanos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Colágeno/metabolismo , Interleucina-1beta/metabolismo , Disco Intervertebral/efectos de los fármacos , Disco Intervertebral/metabolismo , Ratones , Técnicas de Cultivo de Órganos/métodos , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Biochem Biophys Res Commun ; 529(4): 1165-1172, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819581

RESUMEN

Renal stem or progenitor cells (RSCs), labeled with CD24 and CD133, play an important role during the repair of renal injury. Bmi-1 is a critical factor in regulating stemness of adult stem cells or progenitor cells. To investigate whether Bmi-1 determines the stemness of RSCs by inhibiting p16 and p53, and/or maintaining redox balance, RSCs were isolated, cultured and analyzed for stemness characterizations. In RSCs from Bmi-1-deficient (Bmi-1-/-) mice and wild type (WT) littermates, self-renewal, stemness, and expressions of molecules for regulating redox balance and cell cycle progression were compared. Self-renewal of RSCs from Bmi-1 and p16 double-knockout (Bmi-1-/-p16-/-), Bmi-1 and p53 double-knockout (Bmi-1-/-p53-/-) and N-acetylcysteine (NAC)-treated Bmi-1-/- mice were further analyzed for amelioration. Human renal proximal tubular epithelial cells (HK2) were also used for signaling analysis. Our results showed that third-passage RSCs from WT mice had good stemness; Bmi-1 deficiency led to the decreased stemness, and the increased apoptosis for RSCs; NAC treatment or p16/p53 deletion ameliorated the decreased self-renewal of RSCs in Bmi-1 deficiency mice by maintaining redox balance or inhibiting cell cycle arrest respectively; Oxidative stress (OS) could negatively feedback regulate the mRNA expressions of Bmi-1, p16 and p53. In conclusion, Bmi-1 determined the stemness of RSCs through maintaining redox balance and preventing cell cycle arrest. Thus, Bmi-1 signaling molecules would be novel therapeutic targets for maintaining RSCs and hampering the progression of kidney diseases to prevent renal failure.


Asunto(s)
Riñón/citología , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células Madre/metabolismo , Acetilcisteína/farmacología , Animales , Autorrenovación de las Células/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Retroalimentación Fisiológica , Eliminación de Gen , Humanos , Masculino , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Complejo Represivo Polycomb 1/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Células Madre/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
8.
Stem Cells ; 37(9): 1200-1211, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30895687

RESUMEN

We previously demonstrated that Bmi1 deficiency leads to osteoporosis phenotype by inhibiting the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells (MSCs), but it is unclear whether overexpression of Bmi1 in MSCs stimulates skeletal development and rescues Bmi1 deficiency-induced osteoporosis. To answer this question, we constructed transgenic mice (Bmi1Tg ) that overexpressed Bmi1 driven by the Prx1 gene and analyzed their skeletal phenotype differences with that of wild-type littermates. We then hybridized Bmi1Tg to Bmi1-/- mice to generate Bmi1-/- mice overexpressing Bmi1 in MSCs and compared their skeletal phenotypes with those of Bmi1-/- and wild-type mice using imaging, histopathological, immunohistochemical, histomorphometric, cellular, and molecular methods. Bmi1Tg mice exhibited enhanced bone growth and osteoblast formation, including the augmentation of bone size, cortical and trabecular volume, number of osteoblasts, alkaline phosphatase (ALP)-positive and type I collagen-positive areas, number of total colony forming unit fibroblasts (CFU-f) and ALP+ CFU-f, and osteogenic gene expression levels. Consistently, MSC overexpressing Bmi1 in the Bmi1-/- background not only largely reversed Bmi1 systemic deficiency-induced skeletal growth retardation and osteoporosis, but also partially reversed Bmi1 deficiency-induced systemic growth retardation and premature aging. To further explore the mechanism of action of MSCs overexpressing Bmi1 in antiosteoporosis and antiaging, we examined changes in oxidative stress and expression levels of p16 and p19. Our results showed that overexpression of Bmi1 in MSCs inhibited oxidative stress and downregulated p16 and p19. Taken together, the results of this study indicate that overexpression of Bmi1 in MSCs exerts antiaging and antiosteoporosis effects by inactivating p16/p19 signaling and inhibiting oxidative stress. Stem Cells 2019;37:1200-1211.


Asunto(s)
Envejecimiento/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoporosis/genética , Estrés Oxidativo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Ratones Noqueados , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/genética
9.
Calcif Tissue Int ; 107(2): 126-134, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32356017

RESUMEN

The increasing levels of bone marrow fat evident in aging and osteoporosis are associated with low bone mass and attributed to reduced osteoblastogenesis. Local lipotoxicity has been proposed as the primary mechanism driving this reduction in bone formation. However, no studies have examined the correlation between high levels of marrow fat volumes and changes in local cellularity. In this study, we hypothesize that areas of bone marrow with high fat volumes are associated with significant changes in cell number within a similar region of interest (ROI). Inbred albino Louvain (LOU) rats, originating from the Wistar strain, have been described as a model of healthy aging with the absence of obesity but expressing the typical features of age-related bone loss. We compared local changes in distal femur cellularity and structure in specific ROI of undecalcified bone sections from 4- and 20-month-old male and female LOU rats and Wistar controls. Our results confirmed that older LOU rats exhibited significantly higher fat volumes than Wistar rats (p < 0.001). These higher fat volume/total volume were associated with lower trabecular number (p < 0.05) and thickness (p < 0.05) and higher trabecular separation (p < 0.05). In addition, osteoblast and osteocyte numbers were reduced in the similar ROI containing high levels of adiposity, while osteoclast number was higher compared to control (p < 0.03). In summary, marrow ROIs with a high level of adiposity were associated with a lower bone mass and changes in cellularity explaining associated bone loss. Further studies assessing the levels of lipotoxicity in areas of high local marrow adiposity and identifying molecular actors involved in this phenomenon are still required.


Asunto(s)
Tejido Adiposo , Factores de Edad , Médula Ósea , Osteocitos , Animales , Densidad Ósea , Huesos , Recuento de Células , Femenino , Masculino , Ratas , Ratas Wistar
10.
J Cell Biochem ; 120(8): 13994-14003, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30957908

RESUMEN

Estrogen deficiency accelerates the aging process and increases the risk of developing cardiovascular disease (CVD). Apoptosis is one of the important mechanisms of aging. p27kip1 is a cyclin-dependent kinase inhibitor that can regulate cell cycle, apoptosis, and cell motility. p27kip1 overexpression can inhibit cell cycle and increase apoptosis so it has been considered as a marker of aging. In the present study, bilateral ovariectomy (OVX) was performed as a model for menopause in wild-type (WT) and p27kip1 knockout (KO) mice to assess the effects of p27kip1 loss in myocardial aging caused by estrogen deficiency. We found that myocardial fibrosis and heart weight/body weight ratio of mice in the OVX group and p27kip1 KO group were significantly increased. Echocardiography showed that the left ventricular diameter and volume of the WT OVX group increased significantly and the cardiac function decreased. However, there was no significant difference in the results of echocardiography between the two p27kip1 KO groups. The aging and apoptosis indexes in OVX group were increased significantly, However, the indexes in p27kip1 KO mice were decreased. The expression of antioxidant indexes in OVX group was decreased significantly and p27kip1 KO can improve the antioxidant ability. These results provided that estrogen deficiency increased oxidative stress and apoptosis, accelerated aging of heart. p27kip1 KO can partly delay the aging and apoptosis of heart through upregulated antioxidant enzymes.


Asunto(s)
Senescencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Estrógenos/deficiencia , Miocardio/patología , Animales , Antioxidantes/metabolismo , Apoptosis , Peso Corporal , Daño del ADN , Electrocardiografía , Pruebas de Función Cardíaca , Ratones Noqueados , Tamaño de los Órganos
11.
J Cell Biochem ; 120(9): 14745-14755, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30994964

RESUMEN

It is known that parathyroid hormone-related peptide (PTHrP) contains a nuclear localization sequence (NLS, 87-107), which, together with its C-terminus (107-139), has been shown to positively regulate vascular smooth muscle cell (VSMCs) proliferation and vascular neointima formation, and inhibit cellular apoptosis. The role of PTHrP in ischemic cardiac diseases remains unclear. In this study, we attempted to determine whether PTHrP 87 to 139 can play a role in promoting cardiac function via enhancing angiogenesis after myocardial infarction (MI) occurred. MI was reproduced in C57BL/6 mice using a coronary artery ligation method. In total, three groups (n = 11 per group) of animals were used, and they were received either PTHrP 87 to 139 (80 µg/kg, treatment group) or saline (MI and Sham group) subcutaneously once a day for 4 weeks after MI. To measure cardiac function, an echocardiography was generated and cardiac tissue was harvested for immunohistological studies 4 weeks after operation. Our results show that, after MI, the cardiac function of the experimental mice was significantly impaired. PTHrP 87 to 139 treatment attenuated cardiac dysfunction in MI mice. Besides, as indicated by decreased heart weight/body weight and lung weight/body weight ratio, PTHrP 87 to 139 attenuated pulmonary congestion and cardiac hypertrophy. Masson staining revealed that PTHrP 87 to 139 attenuated myocardial fibrosis after MI. Also, terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling staining and the expression of cleaved caspase 3 suggested that MI-induced myocytes apotosis was inhibited by PTHrP 87 to 139. In addition to the significantly increased capillary density, PTHrP 87 to 139 treatment also induced p-Akt and several angiogenic factors. In conclusion, PTHrP 87 to 139 treatment preserved cardiac function after MI, and stimulated angiogenesis via upregulating vascular endothelial growth factor and basic fibroblast growth factor (bFGF) in infarct border zone of ischemic myocardium,. These results suggest that PTHrP 87 to 139 is of therapeutic potential for MI.


Asunto(s)
Infarto del Miocardio/prevención & control , Isquemia Miocárdica/prevención & control , Neovascularización Fisiológica , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Fragmentos de Péptidos/farmacología , Remodelación Ventricular , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Isquemia Miocárdica/etiología , Isquemia Miocárdica/patología , Neovascularización Fisiológica/efectos de los fármacos
12.
Int J Cancer ; 143(2): 368-382, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29441580

RESUMEN

Human epidemiological studies suggest that 1,25(OH)2 D3 deficiency might increase cancer incidence, but no spontaneous tumors have been reported in mice lacking 1,25(OH)2 D3 or deficient in its receptor. In our study, we detected, for the first time, diverse types of spontaneous tumors in l,25(OH)2 D3 deficient mice more than 1 year of age. This was associated with increased oxidative stress, cellular senescence and senescence-associated secretory phenotype molecules, such as hepatocyte growth factor, mediated via its receptor c-Met. Furthermore, 1,25(OH)2 D3 prevented spontaneous tumor development. We also demonstrated that l,25(OH)2 D3 deficiency accelerates allograft tumor initiation and growth by increasing oxidative stress and DNA damage, activating oncogenes, inactivating tumor suppressor genes, stimulating malignant cell proliferation and inhibiting their senescence; in contrast, supplementation with exogenous l,25(OH)2 D3 or antioxidant, or knock-down of the Bmi1 or c-Met oncogene, largely rescued the phenotypes of allograft tumors. Results from our study suggest that 1,25(OH)2 D3 deficiency enhances tumorigenesis by increasing malignant cell oxidative stress and DNA damage, stimulating microenvironmental cell senescence and a senescence-associated secretory phenotype, and activating oncogenes and inactivating tumor suppressor genes, thus increasing malignant cell proliferation. Our study provides direct evidence supporting the role of vitamin D deficiency in increasing cancer incidence. Conversely, 1,25(OH)2 D3 prevented spontaneous tumor development, suggesting that this inhibitory effect prevents the initiation and progression of tumorigenesis, thus provides a mechanistic basis for 1,25(OH)2 D3 to prevent tumorigenesis in an aging organism.


Asunto(s)
Calcitriol/administración & dosificación , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias Mamarias Animales/prevención & control , Estrés Oxidativo/efectos de los fármacos , Deficiencia de Vitamina D/tratamiento farmacológico , Animales , Calcitriol/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Senescencia Celular , Daño del ADN/efectos de los fármacos , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-met/metabolismo , Deficiencia de Vitamina D/complicaciones , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Reprod Biomed Online ; 37(4): 480-488, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30236824

RESUMEN

RESEARCH QUESTION: What is the role of mitochondrial DNA (mtDNA) in the pathogenesis of non-obstructive azoospermia (NOA)? DESIGN: mtDNA genome sequencing followed by an independent population validation were performed in 628 NOA cases and 584 healthy controls. Antioxidant capacity of serum was evaluated in 54 randomly selected cases out of 536 and 49 out of 489 controls. RESULTS: In the screening stage, 13 mtDNA haplogroups (hg) were ascertained, and 10 susceptible variants were observed. In the validation stage, hg M8* in individuals was found to be associated with increased risk of NOA [odds ratio (OR) 2.61, 95% confidence interval (CI) 1.47-4.61] (P=0.001). Unexpectedly, the frequency of m.8684C>T, the defining marker for hg M8a, was also higher in NOA (OR 4.14, 95% CI 1.56-11.03) (P=0.002). Subsequently, the frequency distributions were compared among the sub-hg of hg M8* (including hg M8a, C and Z) and, intriguingly, no significance was found in hg C and Z. Additionally, the level of total antioxidant capacity was significantly decreased (P<0.05) compared with the control group. CONCLUSIONS: hg M8a background in general played an active role in the penetrance of 8684C>T in NOA, and mtDNA genetic variants (causing low antioxidant levels) might increase mtDNA damage and impair normal spermatogenesis.


Asunto(s)
Azoospermia/genética , ADN Mitocondrial , Daño del ADN , Genoma Humano , Haplotipos , Humanos , Masculino , Espermatogénesis/genética
14.
J Biol Chem ; 291(6): 2647-63, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26677226

RESUMEN

To interpret epigenetic information, chromatin readers utilize various protein domains for recognition of DNA and histone modifications. Some readers possess multidomains for modification recognition and are thus multivalent. Bromodomain- and plant homeodomain-linked finger-containing protein 3 (BRPF3) is such a chromatin reader, containing two plant homeodomain-linked fingers, one bromodomain and a PWWP domain. However, its molecular and biological functions remain to be investigated. Here, we report that endogenous BRPF3 preferentially forms a tetrameric complex with HBO1 (also known as KAT7) and two other subunits but not with related acetyltransferases such as MOZ, MORF, TIP60, and MOF (also known as KAT6A, KAT6B, KAT5, and KAT8, respectively). We have also characterized a mutant mouse strain with a lacZ reporter inserted at the Brpf3 locus. Systematic analysis of ß-galactosidase activity revealed dynamic spatiotemporal expression of Brpf3 during mouse embryogenesis and high expression in the adult brain and testis. Brpf3 disruption, however, resulted in no obvious gross phenotypes. This is in stark contrast to Brpf1 and Brpf2, whose loss causes lethality at E9.5 and E15.5, respectively. In Brpf3-null mice and embryonic fibroblasts, RT-quantitative PCR uncovered no changes in levels of Brpf1 and Brpf2 transcripts, confirming no compensation from them. These results indicate that BRPF3 forms a functional tetrameric complex with HBO1 but is not required for mouse development and survival, thereby distinguishing BRPF3 from its paralogs, BRPF1 and BRPF2.


Asunto(s)
Embrión de Mamíferos/enzimología , Desarrollo Embrionario , Histona Acetiltransferasas/metabolismo , Complejos Multienzimáticos/metabolismo , Animales , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Células HEK293 , Histona Acetiltransferasas/genética , Humanos , Ratones , Ratones Mutantes , Complejos Multienzimáticos/genética
15.
Biochem Biophys Res Commun ; 486(2): 264-269, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28286271

RESUMEN

Nonalcoholic steatohepatitis (NASH) is characterized by excess accumulation of lipids in liver, accompanied with hepatocyte injury, cell death and inflammation. Although p16 is known as tumor suppressor in multiple cancer types, it remains unclear whether p16 plays a critical role in NASH. To determine whether p16 could play a role in the pathogenesis of NASH, wild-type mice and p16-/- mice were fed on a methionine and choline-deficient (MCD) diet for 3 weeks, and liver steatosis, fibrosis, and inflammation were evaluated. Our data show that p16-/- mice fed with MCD diet displayed more significant hepatic steatosis, hepatocyte damage, increased oxidative stress and inflammatory cell infiltration compared to MCD-fed WT mice. It was also clear that the increased ROS and the accumulation of lipid in BEL-7402 cells occurred when p16 expression was depleted with siRNA. These findings indicate that p16 may play a critical role in the development of NASH by reining in ROS production and by inhabiting inflammatory response.


Asunto(s)
Deficiencia de Colina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Hepatocitos/metabolismo , Cirrosis Hepática/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Animales , Línea Celular Tumoral , Deficiencia de Colina/etiología , Deficiencia de Colina/metabolismo , Deficiencia de Colina/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Alimentos Formulados/efectos adversos , Regulación de la Expresión Génica , Hepatocitos/patología , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Metionina/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
16.
Biochem Biophys Res Commun ; 482(4): 742-749, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27871857

RESUMEN

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.


Asunto(s)
Necrosis Tubular Aguda/metabolismo , Riñón/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células Madre/citología , Antígeno AC133/metabolismo , Animales , Antígeno CD24/metabolismo , Diferenciación Celular , Creatinina/metabolismo , Progresión de la Enfermedad , Glicerol/química , Masculino , Ratones , Regeneración , Rabdomiólisis/metabolismo
17.
Proc Natl Acad Sci U S A ; 111(38): 13960-5, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25201987

RESUMEN

Neuronal necrosis induced by calcium overload causes devastating brain dysfunction in diseases such as stroke and brain trauma. It has been considered a stochastic event lacking genetic regulation, and pharmacological means to suppress neuronal necrosis are lacking. Using a Drosophila model of calcium overloading, we found JIL-1/mitogen- and stress-activated protein kinase 1/2 is a regulator of neuronal necrosis through phosphorylation of histone H3 serine 28 (H3S28ph). Further, we identified its downstream events including displacement of polycomb repressive complex 1 (PRC1) and activation of Trithorax (Trx). To test the role of JIL-1/PRC1/Trx cascade in mammals, we studied the necrosis induced by glutamate in rat cortical neuron cultures and rodent models of brain ischemia and found the cascade is activated in these conditions and inhibition of the cascade suppresses necrosis in vitro and in vivo. Together, our research demonstrates that neuronal necrosis is regulated by a chromatin-modifying cascade, and this discovery may provide potential therapeutic targets and biomarkers for neuronal necrosis.


Asunto(s)
Calcio/metabolismo , Cromatina/metabolismo , Neuronas/metabolismo , Animales , Biomarcadores/metabolismo , Cromatina/patología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Femenino , Histonas/genética , Histonas/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Necrosis , Neuronas/patología , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Am J Physiol Endocrinol Metab ; 308(1): E51-62, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25370849

RESUMEN

We used mice with targeted deletion of 25-hydroxyvitamin D-1 α-hydroxylase [1α(OH)ase(-/-)] to investigate whether 1,25(OH)2D3 deficiency results in male infertility mediated by 1,25(OH)2D3 or extracellular calcium and phosphorus. Male 1α(OH)ase(-/-) and their wild-type littermates fed either a normal diet or a rescue diet from weaning were mated at 6-14 wk of age with female wild-type mice on the same diet. The fertility efficiency of females was analyzed, and the reproductive phenotypes of males were evaluated by histopathological and molecular techniques. Hypocalcemic and hypophosphatemic male 1α(OH)ase(-/-) mice on a normal diet developed infertility characterized by hypergonadotropic hypogonadism, with downregulation of testicular calcium channels, lower intracellular calcium levels, decreased sperm count and motility, and histological abnormalities of the testes. The proliferation of spermatogenic cells was decreased with downregulation of cyclin E and CDK2 and upregulation of p53 and p21 expression, whereas apoptosis of spermatogenic cells was increased with upregulation of Bax and p-caspase 3 expression and downregulation of Bcl-xl expression. When serum calcium and phosphorus were normalized by the rescue diet, the defective reproductive phenotype in the male 1α(OH)ase(-/-) mice, including the hypergonadotropic hypogonadism, decreased sperm count and motility, histological abnormalities of testis, and defective spermatogenesis, was reversed. These results indicate that the infertility seen in male 1,25(OH)2D3-deficient mice is not a direct effect of active vitamin D deficiency on the reproductive system but is an indirect effect mediated by extracellular calcium and phosphorus.


Asunto(s)
Calcio de la Dieta/efectos adversos , Raquitismo Hipofosfatémico Familiar/complicaciones , Infertilidad Masculina , Fósforo Dietético/efectos adversos , Deficiencia de Vitamina D/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Animales , Calcitriol/sangre , Calcio/sangre , Dieta/efectos adversos , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/etiología , Femenino , Infertilidad Masculina/sangre , Infertilidad Masculina/etiología , Masculino , Ratones , Ratones Noqueados , Minerales/farmacología , Fósforo/sangre , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/complicaciones
19.
Exp Dermatol ; 24(11): 847-52, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26121068

RESUMEN

We previously reported that deficiency of the PTHrP nuclear localization sequence (NLS) and C-terminus in PTHrP knockin (PTHrP KI) mice resulted in premature ageing of skin. P27, a cyclin-dependent kinase inhibitor, was upregulated in PTHrP KI mice and acted as a downstream target of the PTHrP NLS to regulate the proliferation of vascular smooth muscle cells. To determine the effects of p27 deficiency on premature skin ageing of PTHrP KI mice, we compared the skin phenotypes of PTHrP KI mice to those of p27 knockout (p27(-/-) ) mice and to those of double homozygous p27-deficient and PTHrP KI (p27(-/-) PTHrP KI) mice at 2 weeks age. Compared with wild-type littermates, PTHrP KI mice displayed thinner skin and decreased subcutaneous fat and collagen fibres, decreased skin cell proliferation and increased apoptosis, higher expression of p27, p19 and p53 and lower expression of cyclin E and CDK2, and increased reactive oxygen species levels and decreased antioxidant capacity. Deficiency of p27 in the PTHrP KI mice at least in part corrected the skin premature ageing phenotype resulting in thicker skin and increased subcutaneous fat and collagen. These alternations were associated with higher expression of CDK2 and cyclin E, lower expression of p19 and p53, and enhanced antioxidant capacity with increased skin cell proliferation and inhibition of apoptosis. Our results indicate that the NLS and C-terminus of PTHrP play a critical role in preventing skin from premature ageing that is partially mediated by p27.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/deficiencia , Envejecimiento de la Piel/fisiología , Animales , Antioxidantes/metabolismo , Apoptosis , Proliferación Celular , Técnicas de Sustitución del Gen , Ratones Noqueados , Ratones Desnudos , Señales de Localización Nuclear , Proteína Relacionada con la Hormona Paratiroidea/genética , Especies Reactivas de Oxígeno/metabolismo , Envejecimiento de la Piel/patología , Regulación hacia Arriba
20.
Br J Nutr ; 113(6): 909-22, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25744000

RESUMEN

In the present study, we investigated whether high dietary Ca and exogenous parathyroid hormone 1-34 fragments (PTH 1-34) have synergistic effects on bone formation in adult mice, and explored the related mechanisms. Adult male mice were fed a normal diet, a high-Ca diet, a PTH-treated diet, or a high-Ca diet combined with subcutaneously injected PTH 1-34 (80 µg/kg per d) for 4 weeks. Bone mineral density, trabecular bone volume, osteoblast number, alkaline phosphatase (ALP)- and type I collagen-positive areas, and the expression levels of osteoblastic bone formation-related genes and proteins were increased significantly in mice fed the high-Ca diet, the PTH-treated diet, and, even more dramatically, the high-Ca diet combined with PTH. Osteoclast number and surface and the ratio of receptor activator for nuclear factor-κB ligand (RANKL):osteoprotegerin (OPG) were decreased in the high-Ca diet treatment group, increased in the PTH treatment group, but not in the combined treatment group. Furthermore, third-passage osteoblasts were treated with high Ca (5 mM), PTH 1-34 (10⁻8 M) or high Ca combined with PTH 1-34. Osteoblast viability and ALP activity were increased in either the high Ca-treated or PTH-treated cultures and, even more dramatically, in the cultures treated with high Ca plus PTH, with consistent up-regulation of the expression levels of osteoblast proliferation and differentiation-related genes and proteins. These results indicate that dietary Ca and PTH play synergistic roles in promoting osteoblastic bone formation by stimulating osteoblast proliferation and differentiation.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/prevención & control , Calcio de la Dieta/uso terapéutico , Interacciones Alimento-Droga , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Hormona Paratiroidea/análogos & derivados , Hormona Paratiroidea/uso terapéutico , Animales , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Canales de Calcio/genética , Canales de Calcio/metabolismo , Calcio de la Dieta/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Terapia Combinada , Masculino , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoblastos/patología , Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Receptores Sensibles al Calcio/agonistas , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Receptores de Hormona Paratiroidea/agonistas , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA