Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Acta Biochim Biophys Sin (Shanghai) ; 54(2): 179-186, 2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35538026

RESUMEN

Hypoxia-induced epigenetic regulation calls for more effective therapeutic targets for esophageal cancer. We used GEPIA and UALCAN databases to screen survival-related and cancer stage-associated genes. Eca109 and KYSE450 esophageal cancer cell lines were cultured under normoxia, hypoxia, or CoCl-induced hypoxia conditions, which were further transfected with plasmids expressing RB binding protein 7 (RBBP7), hypoxia-inducible factor 1 (HIF1)-α, or RBBP7 shRNA. Colony formation and MTT assays were used to detect cell proliferation. Tumor sphere formation and stemness marker detection were applied to assess cell stemness. RT-PCR and western blot analysis were used to detect the relative mRNA level and protein expression, respectively. Luciferase assay was utilized to detect the direct interaction between HIF1α and RBBP7. Up-regulated RBBP7 was identified as one of the most prominent survival-related genes, which is negatively correlated with the overall survival (OS), disease recurrence-free survival (DFS), and tumor stages. Hypoxia-induced HIF1α up-regulates RBBP7 expression, which promotes esophagus cancer cell viability, proliferation, and stemness with increased cyclin-dependent kinase 4 (CDK4) expression. Luciferase reporter assay verified that HIF1α transcriptionally regulates the expression of RBBP7. We conclude that hypoxia induces high expression of RBBP7 which is at least partially mediated by HIF1α, up-regulates the expression of downstream CDK4, and thereby promotes tumor progression in esophageal cancer cells.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina , Neoplasias Esofágicas , Proteína 7 de Unión a Retinoblastoma , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular/genética , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/genética , Progresión de la Enfermedad , Epigénesis Genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína 7 de Unión a Retinoblastoma/biosíntesis , Proteína 7 de Unión a Retinoblastoma/genética , Proteína 7 de Unión a Retinoblastoma/metabolismo
2.
JCI Insight ; 7(3)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34932500

RESUMEN

Standard radiation therapy (RT) does not reliably provide locoregional control for women with multinode-positive breast cancer and triple-negative breast cancer (TNBC). We hypothesized that CDK4/6 inhibition (CDK4/6i) would increase the radiosensitivity not only of estrogen receptor-positive (ER+) cells, but also of TNBC that expresses retinoblastoma (RB) protein. We found that CDK4/6i radiosensitized RB WT TNBC (n = 4, radiation enhancement ratio [rER]: 1.49-2.22) but failed to radiosensitize RB-null TNBC (n = 3, rER: 0.84-1.00). RB expression predicted response to CDK4/6i + RT (R2 = 0.84), and radiosensitization was lost in ER+/TNBC cells (rER: 0.88-1.13) after RB1 knockdown in isogenic and nonisogenic models. CDK4/6i suppressed homologous recombination (HR) in RB WT cells but not in RB-null cells or isogenic models of RB1 loss; HR competency was rescued with RB reexpression. Radiosensitization was independent of nonhomologous end joining and the known effects of CDK4/6i on cell cycle arrest. Mechanistically, RB and RAD51 interact in vitro to promote HR repair. CDK4/6i produced RB-dependent radiosensitization in TNBC xenografts but not in isogenic RB1-null xenografts. Our data provide the preclinical rationale for a clinical trial expanding the use of CDK4/6i + RT to difficult-to-control RB-intact breast cancers (including TNBC) and nominate RB status as a predictive biomarker of therapeutic efficacy.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales , Neoplasias de la Mama Triple Negativas/radioterapia , Animales , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones SCID , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Oxid Med Cell Longev ; 2019: 5084689, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281582

RESUMEN

The connective tissue of the umbilical cord contains stem cells called Wharton's jelly cells. These cells express core transcription factors (NANOG, OCT4, and SOX2). The protein product of the SOX2 gene controls the cell cycle by interacting with cyclin D (directly and indirectly) and cycle inhibitors-p21 and p27, as well as two E2f3 protein isoforms. The aim of the study was to analyze the effect of SOX2 on the cell cycle of stem cells of Wharton's jelly. The material for the study was the stem cells of Wharton's jelly isolated from 20 umbilical cords collected during childbirth. The stem cells collected were subjected to cytometric analysis, cell culture, and RNA isolation. cDNA was the starting material for the analysis of gene expression: SOX2, CCND1, CDK4, and CDKN1B. The studies indicate a high proliferative potential of the Wharton's jelly stem cells and the inhibitory effect of SOX2 on the expression of the CCND1 and CDK4 gene.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Gelatina de Wharton/citología , Gelatina de Wharton/metabolismo , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Ciclina D1/biosíntesis , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/genética , Expresión Génica , Humanos , Factores de Transcripción SOXB1/genética
4.
Biochim Biophys Acta Mol Cell Res ; 1866(5): 945-956, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30826331

RESUMEN

The maintenance of stem cells often requires the support of feeder cells. Primary mouse embryonic fibroblasts (MEFs) have traditionally been used as feeder cells, and although these MEF-derived feeder cells have exhibited a reasonable performance, they require repeated cell isolation, since MEFs cannot expand indefinitely. To overcome this limitation, immortalized cells, such as STO cells, have been used. However, one major disadvantage is that previously reported immortalized cells can only support stem cell cultures for a relatively short period, typically 4 to 7 days. In this study, we found that our newly established rat-derived fibroblasts immortalized by the expression of mutant cyclin-dependent kinase 4, cyclin D, and telomerase reverse transcriptase, can function as feeder cells for relatively long cell culture periods of approximately 14 days. The rat-derived immortalized cells developed in this study should be a useful source of feeder cells to support stem cell research.


Asunto(s)
Ciclina D/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Células Nutrientes/metabolismo , Fibroblastos/metabolismo , Mutación , Células Madre/metabolismo , Telomerasa/biosíntesis , Animales , Línea Celular Transformada , Técnicas de Cocultivo , Ciclina D/genética , Quinasa 4 Dependiente de la Ciclina/genética , Células Nutrientes/citología , Fibroblastos/citología , Ratones , Ratas , Células Madre/citología , Telomerasa/genética
5.
Head Neck Pathol ; 13(2): 169-176, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29748845

RESUMEN

Traumatized lipomas with degenerative change may demonstrate histopathologic features that mimic atypical lipomatous tumor (ALT). Previously reported series of ALT involving the oral cavity preceded routine use of MDM2 and CDK4 immunohistochemistry. Our aim is to evaluate MDM2 and CDK4 immunohistochemical expression in adipocytic tumors arising in this site, in conjunction with the histiocytic marker PU.1, to determine whether MDM2 and CDK4 impacts classification. 17 cases originally diagnosed as ALT were retrieved and immunohistochemical studies for MDM2, CDK4 and PU.1 were performed. FISH analysis for MDM2 amplification was performed in select cases. For this study group, the male:female ratio was 9:8 and the median age was 62 (range 41-88). All 17 cases presented as well- or predominantly well-circumscribed proliferations of variably sized, mature adipocytes exhibiting uni- or multi-vacuolation with occasional scalloped nuclei and mild nuclear atypia. Variable amounts of fibrous stroma with focal myxoid change and bland spindle cells were identified in 14/17 cases. Lipoblasts or atypical hyperchromatic stromal cells were not identified in any cases. 14 of 17 cases were negative for MDM2 and CDK4 in tumor cells and 11 of these 14 showed weak nuclear positivity for MDM2 in histiocytes. 3 of 17 cases showed weak, multifocal immunohistochemical expression of MDM2 and CDK4. PU.1 highlighted histiocytes in all 17 cases. FISH analysis for MDM2 amplification was negative in all 3 cases with weak MDM2/CDK4 expression. All cases were reclassified as lipoma with degenerative changes. ALT, in all likelihood, is less common than previously thought in this anatomic location and best diagnosed with ancillary studies. MDM2 expression in histiocytes is best interpreted in conjunction with CDK4 immunohistochemistry and confirmatory FISH for MDM2 amplification.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/biosíntesis , Lipoma/diagnóstico , Neoplasias de la Boca/diagnóstico , Proteínas Proto-Oncogénicas c-mdm2/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Quinasa 4 Dependiente de la Ciclina/análisis , Diagnóstico Diferencial , Femenino , Humanos , Liposarcoma/diagnóstico , Masculino , Uso Excesivo de los Servicios de Salud/prevención & control , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-mdm2/análisis
6.
Int J Med Microbiol ; 308(7): 819-828, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30033344

RESUMEN

Ureaplasma spp. are known to be associated with human genitourinary tract diseases and perinatal diseases and Ureaplasma spp. Lipid-associated membrane proteins (LAMPs) play important roles in their related diseases. However, the exact mechanism underlying pathogenesis of Ureaplasma spp. LAMPs is largely unknown. In this study, we explored the pathogenic mechanisms of Ureaplasma spp. LAMPs by elucidating their role in modulating the cell cycle and related signaling pathways in human monocytic cell U937, which is highly related to the inflammatory and protective effect in infectious diseases. We utilized the two ATCC reference strains (Ureaplasma parvum serovar 3 str. ATCC 27,815 (UPA3) and Ureaplasma urealyticum serovar 8 str. ATCC 27,618 (UUR8)) and nine clinical isolates which including both UPA and UUR to study the effects of Ureaplasma spp. LAMPs on U937 in vitro. We found that LAMPs derived from UUR8 and both UPA and UUR of clinical strains markedly inhibited the cell proliferation, while UPA3 LAMPs suppressed slightly. Besides, the result of flow cytometry analysis indicated all the Ureaplasma spp. LAMPs could arrest U937 cells in G1 phase. Next, we found that the cell cycle arrest was associated with increased levels of p53 and p21, and a concomitant decrease in the levels of CDK2, CDK4, CDK6 and cyclin E1 at both transcriptional and translational levels after treatment with LAMPs derived from UUR8 or clinical strains, while only cyclin E1 was down-regulated after treatment with UPA3 LAMPs. Further study showed that p53 down-regulation had almost no effect on the distribution of cell cycle and the expression of p21. In conclusion, this study demonstrated that LAMPs derived from UUR8 and clinical strains could inhibit the proliferation of U937 cells by inducing G1 cell cycle arrest through increasing the p21 expression in a p53-independent manner, while UPA3 LAMPs could induce the cell cycle arrest slightly. Our study could contribute to the understanding of Ureaplasma spp. pathogenesis, which has potential value for the treatment of Ureaplasma spp. infections.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Lipoproteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Infecciones por Ureaplasma/patología , Ureaplasma/patogenicidad , Línea Celular Tumoral , Proliferación Celular/fisiología , Ciclina E/biosíntesis , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Humanos , Proteínas Oncogénicas/biosíntesis , Células U937 , Ureaplasma/aislamiento & purificación , Enfermedades Urológicas/microbiología
7.
J Antibiot (Tokyo) ; 71(9): 808-817, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29752478

RESUMEN

Bafilomycin C1, which was isolated from Streptomyces albolongus in our previous work, exhibited strong cytotoxicity against several cancer cell lines. This study aimed to evaluate its antitumor effect on human hepatocellular cancer SMMC7721 cells and the underlying mechanism in vitro and in vivo. MTT assay revealed that bafilomycin C1 retarded SMMC7721 cell growth and proliferation. Western blot and real-time qPCR analysis revealed that bafilomycin C1 caused partial G0/G1 phase cell-cycle arrest, downregulated the expression of cyclin D3, cyclin E1, CDK2, CDK4, and CDK6 and upregulated the expression of p21. Moreover, bafilomycin C1 caused mitochondrial membrane dysfunction through oxidative stress. Furthermore, bafilomycin C1 decreased the expression of Bcl-2; increased the expression of Bax, p53, and P-p53; and increased cleavage of caspase-9 and caspase-3, thereby inducing the intrinsic caspase-dependent apoptotic pathway. In vivo experiments in mice suggested that bafilomycin C1 suppressed tumor growth with few side effects. Cell-cycle arrest and induced apoptosis in tumor tissues in a mouse model treated with bafilomycin C1 were demonstrated by histological analyses, western blot and TUNEL. These findings indicate that bafilomycin C1 may be a promising candidate for hepatic cellular cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Macrólidos/farmacología , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D3/biosíntesis , Ciclina E/biosíntesis , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo , Membranas Mitocondriales/patología , Proteínas Oncogénicas/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Streptomyces/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Med Rep ; 18(1): 421-428, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29749488

RESUMEN

The long­term survival rate in paediatric acute lymphoblastic leukaemia (ALL) exceeds 80%; however, the outcome of adult ALL remains to be poor. Glucocorticoids (GCs) are the preferred drugs in the traditional treatment of ALL patients. In the anti­leukaemia molecular mechanisms of GCs, c­Myc inhibition serves a critical role. In the present study, a c­Myc inhibitor that increased the sensitivity to GCs in NALM6 cells of the B­cell­ALL cell line and CEM cells of the T­cell­ALL cell line was investigated. The data demonstrated that 10058­F4, a c­Myc inhibitor, increased the growth inhibition, G0/G1 phase arrest and apoptosis of the NALM6 and CEM cells as induced by dexamethasone (DXM), a type of GC. Additionally, 10058­F4 reinforced the decreased expressions of c­Myc, cyclin­dependent kinase (CDK)­4 and CDK6 in the NALM6 and CEM cells treated with DXM. These findings indicated that DXM in combination with the c­Myc inhibitor 10058­F4 may be a novel, potent therapeutic strategy for the treatment of ALL.


Asunto(s)
Dexametasona/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Tiazoles/farmacología , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/genética , Dexametasona/agonistas , Sinergismo Farmacológico , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Tiazoles/agonistas
9.
Cell Death Dis ; 9(5): 446, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29670090

RESUMEN

Synovial sarcoma is a highly aggressive but rare form of soft tissue malignancy that primarily affects the extremities of the arms or legs, for which current chemotherapeutic agents have not been proven to be very effective. The cyclin-dependent kinase 4/6-retinoblastoma protein (CDK4/6-Rb) pathway of cell cycle control is known to be aberrant in a large proportion of cancers. Recently, CDK4 inhibitors have successfully been used pre-clinically for the treatment of many human cancers, and in 2015, following the success of clinical trials, the FDA approved the first selective CDK4/6 inhibitor, palbociclib, for the treatment of endocrine therapy resistant breast cancers. However, the expression and therapeutic potential of targeting CDK4 in synovial sarcoma remains unclear. In the present study, we report that CDK4 is highly expressed in human synovial sarcoma, and high CDK4 expressions are associated with poor prognosis in sarcomas patients and the clinical stage and the TNM grade in synovial sarcoma patients. Knockdown of CDK4 with specific small interference RNAs inhibits cell proliferation and enhances apoptotic effects in synovial sarcoma cells. CDK4 inhibitor palbociclib suppresses synovial sarcoma cell proliferation and growth in a dose and time-dependent manner. Palbociclib also inhibits the CDK4/6-Rb signaling pathway and promotes cell apoptosis without changing CDK4/6 protein levels, suggesting that palbociclib only represses the hyper-activation, not the expression of CDK4/6. Flow cytometry analysis reveals that palbociclib induces G1 cell-cycle arrest and apoptotic effects by targeting the CDK4/6-Rb pathway in synovial sarcoma cells. Furthermore, wound healing assays demonstrate that inhibition of the CDK4/6-Rb pathway by palbociclib significantly decreases synovial sarcoma cell migration in vitro. Our study highlights the importance of the CDK4/6-Rb pathway in human synovial sarcoma pathogenesis, and the role of the current selective CDK4/6 inhibitor, palbociclib, as a potential promising targeted therapeutic agent in the treatment of human synovial sarcoma.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Sarcoma Sinovial , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Sarcoma Sinovial/tratamiento farmacológico , Sarcoma Sinovial/enzimología
11.
Mol Cell Biochem ; 439(1-2): 45-52, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28770470

RESUMEN

Cellular senescence is a restricting factor for regenerative therapies with somatic stem cells. We showed previously that the onset of cellular senescence inhibits the osteogenic differentiation in stem cells of the dental follicle (DFCs), although the mechanism remains elusive. Two different pathways are involved in the induction of the cellular senescence, which are driven either by the cell cycle protein P21 or by the cell cycle protein P16. In this study, we investigated the expression of cell cycle proteins in DFCs after the induction of cellular senescence. The induction of cellular senescence was proved by an increased expression of ß-galactosidase and an increased population doubling time after a prolonged cell culture. Cellular senescence regulated the expression of cell cycle proteins. The expression of cell cycle protein P16 was up-regulated, which correlates with the induction of cellular senescence markers in DFCs. However, the expression of cyclin-dependent kinases (CDK)2 and 4 and the expression of the cell cycle protein P21 were successively decreased in DFCs. In conclusion, our data suggest that a P16-dependent pathway drives the induction of cellular senescence in DFCs.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Saco Dental/metabolismo , Transducción de Señal , Células Cultivadas , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Saco Dental/citología , Regulación de la Expresión Génica , Humanos
12.
Eur Rev Med Pharmacol Sci ; 21(17): 3821-3826, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28975985

RESUMEN

OBJECTIVE: In this study, we studied the expression profile of SOX12 gene in different pathological stages of hepatocellular carcinoma (HCC) and explored the possible downstream genes related to its regulation of HCC invasion and migration. MATERIALS AND METHODS: Bioinformatic data mining was performed in liver cancer cohort in TCGA. HepG2 cells were used as in vitro cell model to assess the effect of SOX12 on CDK4 and IGF2BP1 expression. The association between the expression of SOX12, CDK4 or IGF2BP1 and survival time in HCC patients was also assessed based on the data in TCGA. RESULTS: The regional lymph node metastasis (N1) and distant metastasis (M1) cases had significantly increased SOX12 expression than the lymph node negative (N0) and distant metastasis negative (M0) cases respectively. CDK4 and IGF2BP1 were among the top 20 SOX12 co-upregulated genes in HCC, which have well characterized enhancing the effect on HCC cell growth, invasion, and metastasis. In HepG2 cells, knockdown of SOX12 reduced IGF2BP1 and CDK4 expression, while enforced SOX12 expression significantly enhanced their expression. Low SOX12 or CDK4 expression was associated with significantly better 3-year survival and better 5-year survival. Low IGF2BP1 expression was associated with significantly better 3-year survival in HCC patients. CONCLUSIONS: SOX12 upregulation is associated with regional and distant metastasis of HCC. SOX12 can increase the expression of CDK4 and IGF2BP1, which confer malignant phenotypes to HCC. The expression of SOX12, IGF2BP1 or CDK4 might be potential clinical prognostic markers for HCC.


Asunto(s)
Carcinoma Hepatocelular , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Neoplasias Hepáticas , Metástasis de la Neoplasia , Proteínas de Unión al ARN/biosíntesis , Factores de Transcripción SOXC/metabolismo , Regulación hacia Arriba , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Factores de Transcripción SOXC/biosíntesis , Factores de Transcripción SOXC/genética , Análisis de Supervivencia
13.
Pancreas ; 46(10): 1347-1353, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28991877

RESUMEN

OBJECTIVES: Dysregulation of the cell cycle has been observed and implicated as an etiologic factor in a range of human malignancies, but remains relatively unstudied in neuroendocrine tumors (NETs). We evaluated expression of key proteins involved in cell cycle regulation in a large cohort of NETs. METHODS: We evaluated immunohistochemical expression of CDKN1B, CDKN1A, CDKN2A, CDK2, CDK4, CDK6, cyclin D1, cyclin E1, and phosphorylated retinoblastoma protein (phospho-RB1) in a cohort of 267 patients with NETs. We then explored associations between cell cycle protein expression, mutational status, histologic features, and overall survival. RESULTS: We found that high expression of CDK4, CDK6, CCND1, and phospho-RB1 was associated with higher proliferative index, as defined by MKI67. We additionally observed a trend toward shorter overall survival associated with low expression of CDKN1B. This association seemed strongest in SINETs (multivariate hazards ratio, 2.04; 95% confidence interval, 1.06-3.93; P = 0.03). We found no clear association between CDKN1B mutation and protein expression. CONCLUSIONS: Our results suggest that dysregulation and activation of the CDK4/CDK6-CCND1-phospho-RB1 axis is associated with higher proliferative index in NETs. Investigation of the therapeutic potential of CDK4/CDK6 inhibitors in higher grade NETs is warranted.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Ciclina D1/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Tumores Neuroendocrinos/metabolismo , Proteína de Retinoblastoma/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Índice Mitótico , Tumores Neuroendocrinos/patología , Fosforilación , Proteína de Retinoblastoma/metabolismo , Adulto Joven
14.
Virchows Arch ; 471(1): 117-122, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28477272

RESUMEN

Ischemic fasciitis is a benign myofibroblastic lesion, occurring in the sacral region or proximal thigh of elderly or bedridden individuals. The pathogenesis of ischemic fasciitis is thought to be based on ischemic condition; however, it has never been demonstrated. In this study, we examined the expression of ischemia-associated proteins in ischemic fasciitis by immunohistochemical and genetic methods. Specifically, this study aimed to reveal the expression of HIF-1α, MDM2, CDK4, p16, and gene amplification of MDM2 gene. Seven cases of ischemic fasciitis from among the soft-tissue tumors registered at our institution were retrieved. Histopathological findings were as follows: poorly demarcated nodular masses, a proliferation of spindle-shaped fibroblastic or myofibroblastic cells with oval nuclei and eosinophilic or pale cytoplasm, zonal fibrinous deposition, pseudocystic degeneration, granulation-like proliferation of capillary vessels, ganglion-like cells, myxoid or hyalinized stroma, and chronic inflammatory infiltration. Immunohistochemically, the spindle cells were positive for HIF-1α (7/7 cases), MDM2 (4/7 cases), CDK4 (4/7 cases), p16 (7/7 cases), p53 (2/7 case), cyclin D1 (7/7 cases), and alpha-smooth muscle actin (6/7 cases). Neither MDM2 gene amplification nor USP6 gene split signal was detected in any case. Overexpression of the above proteins may be associated with the pathogenic mechanism of ischemic fasciitis. It is noted that the immunohistochemical positivity of MDM2, CDK4, and p16 do not necessarily indicate malignant neoplasm such as dedifferentiated liposarcoma.


Asunto(s)
Biomarcadores/análisis , Fascitis/patología , Isquemia/patología , Transcriptoma , Anciano , Anciano de 80 o más Años , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Fascitis/metabolismo , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Hibridación Fluorescente in Situ , Isquemia/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-mdm2/biosíntesis
15.
Am J Physiol Heart Circ Physiol ; 312(5): H919-H931, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28283550

RESUMEN

ß-Adrenergic receptors (ß-ARs) and catecholamines are present in rodents as early as embryonic day (E)10.5. However, it is not known whether ß-AR signaling plays any role in the proliferation and differentiation of ventricular cells in the embryonic heart. Here, we characterized expression profiles of ß-AR subtypes and established dose-response curves for the nonselective ß-AR agonist isoproterenol (ISO) in the developing mouse ventricular cells. Furthermore, we investigated the effects of ISO on cell cycle activity and differentiation of cultured E11.5 ventricular cells. ISO treatment significantly reduced tritiated thymidine incorporation and cell proliferation rates in both cardiac progenitor cell and cardiomyocyte populations. The ISO-mediated effects on DNA synthesis could be abolished by cotreatment of E11.5 cultures with either metoprolol (a ß1-AR antagonist) or ICI-118,551 (a ß2-AR antagonist). In contrast, ISO-mediated effects on cell proliferation could be abolished only by metoprolol. Furthermore, ISO treatment significantly increased the percentage of differentiated cardiomyocytes compared with that in control cultures. Additional experiments revealed that ß-AR stimulation leads to downregulation of Erk and Akt phosphorylation followed by significant decreases in cyclin D1 and cyclin-dependent kinase 4 levels in E11.5 ventricular cells. Consistent with in vitro results, we found that chronic stimulation of recipient mice with ISO after intracardiac cell transplantation significantly decreased graft size, whereas metoprolol protected grafts from the inhibitory effects of systemic catecholamines. Collectively, these results underscore the effects of ß-AR signaling in cardiac development as well as graft expansion after cell transplantation.NEW & NOTEWORTHY ß-Adrenergic receptor (ß-AR) stimulation can decrease the proliferation of embryonic ventricular cells in vitro and reduce the graft size after intracardiac cell transplantation. In contrast, ß1-AR antagonists can abrogate the antiproliferative effects mediated by ß-AR stimulation and increase graft size. These results highlight potential interactions between adrenergic drugs and cell transplantation.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Madre Embrionarias/trasplante , Ventrículos Cardíacos/citología , Receptores Adrenérgicos beta/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Ciclina D1/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/genética , Ventrículos Cardíacos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Receptores Adrenérgicos beta/genética , Timidina/metabolismo
16.
J Cutan Pathol ; 44(3): 283-288, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27778364

RESUMEN

Liposarcoma, usually arises in deep soft tissues and pleomorphic liposarcoma (PL), is the rarest histopathologic variant. However, 15 cases of entirely dermal PL have been reported. We describe a case of a 79-year-old man who developed a rapidly growing nodule on his thorax. Excisional biopsy was performed and immunohistochemical studies were carried. The lesion was a well-circumscribed dermal nodule composed of multivacuolated pleomorphic lipoblasts and atypical mitotic figures. Neoplastic cells expressed CD10 and resulted negative S100 protein, Melan-A, MITF-1, AE1/AE3, CD4, CD68 (PGM1), retinoblastoma gene family protein, pericentrine and lysozyme. Adipophilin stain showed the lipid contents in the cytoplasm of the neoplastic cells. MDM2 and CDK4 resulted both negative. A diagnosis of primary dermal PL was made. This case shows the utility of adipophilin immunostaining to prove the lipid contents in neoplastic cells, which has the advantage of using formalin-fixed paraffin-embedded tissue and making needless frozen sections and ultrastructural studies to show these findings. Negative MDM2/CDK4 staining in our case argues against the possibility of dedifferentiated liposarcoma and further supports the diagnosis of true PL.


Asunto(s)
Biomarcadores de Tumor/análisis , Liposarcoma/diagnóstico , Perilipina-2/biosíntesis , Neoplasias Cutáneas/diagnóstico , Anciano , Quinasa 4 Dependiente de la Ciclina/análisis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Humanos , Inmunohistoquímica , Masculino , Perilipina-2/análisis , Proteínas Proto-Oncogénicas c-mdm2/análisis , Proteínas Proto-Oncogénicas c-mdm2/biosíntesis
17.
Sci Rep ; 6: 35783, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27779188

RESUMEN

Simvastatin is currently one of the most common drugs for old patients with hyperlipidemia, hypercholesterolemia and atherosclerotic diseases by reducing cholesterol level and anti-lipid properties. Importantly, simvastatin has also been reported to have anti-tumor effect, but the underlying mechanism is largely unknown. We collected several human bladder samples and performed microarray. Data analysis suggested bladder cancer (BCa) was significantly associated with fatty acid/lipid metabolism via PPAR signalling pathway. We observed simvastatin did not trigger BCa cell apoptosis, but reduced cell proliferation in a dose- and time-dependent manner, accompanied by PPARγ-activation. Moreover, flow cytometry analysis indicated that simvastatin induced cell cycle arrest at G0/G1 phase, suggested by downregulation of CDK4/6 and Cyclin D1. Furthermore, simvastatin suppressed BCa cell metastasis by inhibiting EMT and affecting AKT/GSK3ß. More importantly, we found that the cell cycle arrest at G0/G1 phase and the alterations of CDK4/6 and Cyclin D1 triggered by simvastatin could be recovered by PPARγ-antagonist (GW9662), whereas the treatment of PPARα-antagonist (GW6471) shown no significant effects on the BCa cells. Taken together, our study for the first time revealed that simvastatin inhibited bladder cancer cell proliferation and induced cell cycle arrest at G1/G0 phase via PPARγ signalling pathway.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , PPAR gamma/metabolismo , Simvastatina/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Anilidas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Ciclina D1/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Activación Enzimática/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Receptores ErbB/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Invasividad Neoplásica/patología , Oxazoles/farmacología , PPAR gamma/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tirosina/análogos & derivados , Tirosina/farmacología , Vejiga Urinaria/patología
18.
Dev Biol ; 418(1): 124-134, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27475488

RESUMEN

Cell fate specification during organogenesis is usually followed by a phase of cell proliferation to produce the required number of differentiated cells. The Caenorhabditis elegans vulva is an excellent model to study how cell fate specification and cell proliferation are coordinated. The six vulval precursor cells (VPCs) are born at the first larval stage, but they arrest in the G1 phase of the cell cycle until the beginning of the third larval stage, when their fates are specified and the three proximal VPCs proliferate to generate 22 vulval cells. An epidermal growth factor (EGF) signal from the gonadal anchor cell combined with lateral DELTA/NOTCH signaling between the VPCs determine the primary (1°) and secondary (2°) fates, respectively. The hox gene lin-39 plays a key role in integrating these spatial patterning signals and in maintaining the VPCs as polarized epithelial cells. Using a fusion-defective eff-1(lf) mutation to keep the VPCs polarized, we find that VPCs lacking lin-39 can neither activate lateral NOTCH signaling nor proliferate. LIN-39 promotes cell cycle progression through two distinct mechanisms. First, LIN-39 maintains the VPCs competent to proliferate by inducing cdk-4 cdk and cye-1 cyclinE expression via a non-canonical HOX binding motif. Second, LIN-39 activates in the adjacent VPCs the NOTCH signaling pathway, which promotes VPC proliferation independently of LIN-39. The hox gene lin-39 is therefore a central node in a regulatory network coordinating VPC differentiation and proliferation.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriología , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Proteínas de Homeodominio/metabolismo , Organogénesis/genética , Receptores Notch/metabolismo , Vulva/embriología , Animales , Tipificación del Cuerpo/genética , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/biosíntesis , Proteínas de Caenorhabditis elegans/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/biosíntesis , Células Epiteliales/citología , Femenino , Glicoproteínas de Membrana/genética , Transducción de Señal
19.
Sci Rep ; 6: 31085, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27484146

RESUMEN

Pluripotent stem cells (PSCs), including embryonic and induced pluripotent stem cells (iPSCs), show atypical cell cycle regulation characterized by a high proliferation rate and a shorter G1 phase compared with somatic cells. The mechanisms by which somatic cells remodel their cell cycle to achieve the high proliferation rate of PSCs during reprogramming are unclear. Here we identify that the Ink4 protein p18, which is expressed at high levels in somatic cells but at low levels in PSCs, is a roadblock to successful reprogramming. Mild inhibition of p18 expression enhances reprogramming efficiency, while ectopic expression of p18 completely blocks reprogramming. Mechanistic studies show that expression of wild-type p18, but not a p18(D68N) mutant which cannot inhibit Cdk4/6, down-regulates expression of Cdk4/6 target genes involved in DNA synthesis (TK, TS, DHFR, PCNA) and cell cycle regulation (CDK1 and CCNA2) and thus inhibits reprogramming. These results indicate that p18 blocks reprogramming by targeting Cdk4/6-mediated cell cycle regulation. Taken together, our results define a novel pathway that inhibits somatic cell reprogramming, and provide a new target to enhance reprogramming efficiency.


Asunto(s)
Ciclo Celular , Reprogramación Celular , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Mutación Missense , Nucleósido Difosfato Quinasas NM23/biosíntesis , Células Madre Pluripotentes/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , Humanos , Ratones , Nucleósido Difosfato Quinasas NM23/genética
20.
Genet Mol Res ; 15(2)2016 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-27323123

RESUMEN

Radiotherapy is one of the most important treatments for esophageal cancer, but radioresistance remains a major challenge. Previous studies have shown that microRNAs (miRNAs or miRs) are involved in human cancers. miR-124 has been widely reported in various cancers and it is intimately involved in proliferation, cell cycle regulation, apoptosis, migration, and invasion of cancer cells. The aim of this study was to explore the relationship between the miR-124/cyclin-dependent kinase 4 (CDK4) axis and the radiosensitivity of esophageal cancer cells. In this study, we identified the reduced expression of miR-124 in 18 paired esophageal cancer tissues compared to their matched normal tissues. In order to investigate the physiological role of miR-124 in esophageal cancer, the cell counting kit-8 (CCK-8) assay and wound healing assay were performed, and the results suggest that miR-124 overexpression decreases tumor growth and aggression. Next, we detected the effects of ectopic miR-124 expression on the apoptosis of an esophageal cancer cell line (TE-1) following radiotherapy. Using the CCK-8 assay and Hoechst 332528 stain, we found that ectopic expression of miR-124 led to a higher percentage of apoptotic cells. Finally, we identified that CDK4 is a direct target of miR-124 in TE-1 cells using target prediction algorithms and a luciferase reporter assay. Moreover, western blot assay confirmed that CDK4 was downregulated during miR-124 transfection. Taken together, we illustrate that the miR-124/CDK4 axis plays an important role in radiation sensitivity of human esophageal cancer cells by targeting CDK4.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/biosíntesis , Neoplasias Esofágicas/genética , MicroARNs/genética , Tolerancia a Radiación/genética , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/genética , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/radioterapia , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...