Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Gastroenterol Hepatol ; 33(4): 887-893, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29063720

RESUMEN

BACKGROUND AND AIM: CDKN2A/CDKN2B locus on 9p21 is reported to be associated with various diseases, including cancer and cardiovascular and inflammatory diseases. Significant downregulation of CDKN2B-AS1 in inflamed colon tissue of inflammatory bowel disease (IBD) cases was reported in Europeans. This study aimed to confirm the suggestive association of CDKN2A/CDKN2B with IBD identified in our recent genome-wide association study (GWAS). METHODS: We examined the association of CDKN2A/CDKN2B locus with IBD in an additional sample of 574 IBD cases and 542 controls, totaling 4068 cases and 8074 controls. In silico study was performed at various levels for functional annotation of the causal variant. Co-localization of the GWAS association signals and the corresponding expression quantitative trait loci in IBD-related tissues was evaluated using eCAVIAR. RESULTS: An expanded GWAS showed genome-wide significant association of rs3731257 at 9p21 with IBD (odds ratio = 1.17, 95% confidence interval = 1.12-1.22, Pcombined  = 5.68 × 10-9 ) and Crohn's disease (odds ratio = 1.22, 95% confidence interval = 1.15-1.28, Pcombined  = 8.85 × 10-9 ) in the Korean population. Co-localization study suggested that both CDKN2B-AS1 and CDKN2A might be functionally associated with the locus in the small intestine. CONCLUSIONS: rs3731257 in CDKN2A/CDKN2B is an IBD-susceptible locus in Koreans, with a suggestive role for small intestine-specific gene regulation. Our findings suggested that alterations of the CDKN2A/CDKN2B locus could affect the pathophysiology of IBD.


Asunto(s)
Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Estudio de Asociación del Genoma Completo , Enfermedades Inflamatorias del Intestino/genética , Pueblo Asiatico/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Regulación de la Expresión Génica/genética , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Sitios de Carácter Cuantitativo/genética
2.
J Immunol ; 189(6): 2931-40, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22896639

RESUMEN

The lupus-prone NZM2410 mice present an expanded B1a cell population that we have mapped to the Sle2c1 lupus susceptibility locus. The expression of Cdkn2c, a gene encoding for cyclin-dependent kinase inhibitor p18(Ink4c) and located within Sle2c1, is significantly lower in B6.Sle2c1 B cells than in B6 B cells. To test the hypothesis that the B1a cell expansion in B6.Sle2c1 mice was due to a defective p18 expression, we analyzed the B1a cell phenotypes of p18-deficient C57BL/6 mice. We found a dose-dependent negative correlation between the number of B1a cells and p18 expression in B cells, with p18-deficient mice showing an early expansion of the peritoneal B1a cell pool. p18 deficiency enhanced the homeostatic expansion of B1a cells but not of splenic conventional B cells, and the elevated number of B6.Sle2c1 B1a cells was normalized by cyclin D2 deficiency. These data demonstrated that p18 is a key regulator of the size of the B1a cell pool. B6.p18(-/-) mice produced significant amounts of anti-DNA IgM and IgG, indicating that p18 deficiency contributes to humoral autoimmunity. Finally, we have shown that Sle2c1 increases lpr-associated lymphadenopathy and T cell-mediated pathology. B6.p18(-/-).lpr mice showed a greater lymphadenopathy than B6.Sle2c1.lpr mice, but their renal pathology was intermediate between that of B6.lpr and B6.Sle2c1.lpr mice. This indicated that p18-deficiency synergizes, at least partially, with lpr-mediated pathology. These results show that Cdkn2c contributes to lupus susceptibility by regulating the size of the B1a cell compartment and hence their contribution to autoimmunity.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Diferenciación Celular/inmunología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Animales , Subgrupos de Linfocitos B/enzimología , Diferenciación Celular/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Modelos Animales de Enfermedad , Inmunofenotipificación , Lupus Eritematoso Sistémico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Endogámicos NZB , Ratones Noqueados , Ratones Transgénicos
3.
Blood ; 117(7): 2179-88, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21163929

RESUMEN

Inhibition of Cdk4/Cdk6 by p18(INK4c) (p18) is pivotal for generation of noncycling immunoglobulin (Ig)-secreting plasma cells (PCs). In the absence of p18, CD138(+) plasmacytoid cells continue to cycle and turnover rapidly, suggesting that p18 controls PC homeostasis. We now show that p18 selectively acts in a rare population of rapidly cycling CD138(hi)/B220(hi) intermediate PCs (iPCs). While retaining certain B-cell signatures, iPCs are poised to differentiate to end-stage PCs although the majority undergo apoptosis. p18 is dispensable for the development of the PC transcriptional circuitry, and Blimp-1 and Bcl-6 are expressed fully and mutually exclusively in individual iPCs. However, a minor proportion of iPCs express both, and they are preferentially protected by p18 or Bcl-xL overexpression, consistent with expansion of the iPC pool by Bcl-xL overexpression, or loss of proapoptotic Bim or Noxa. Expression of Noxa is induced during B-cell activation, peaks in iPCs, and selectively repressed by p18. It is required to promote apoptosis of cycling B cells, especially in the absence of p18. These findings define the first physiologic function for Noxa and suggest that by repressing Noxa, induction of G1 arrest by p18 bypasses a homeostatic cell-cycle checkpoint in iPCs for PC differentiation.


Asunto(s)
Linfocitos B/citología , Linfocitos B/fisiología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Células Plasmáticas/citología , Células Plasmáticas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Animales , Apoptosis/genética , Apoptosis/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Células HEK293 , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Homeostasis/genética , Homeostasis/fisiología , Humanos , Inmunoglobulina G/biosíntesis , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/deficiencia , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/fisiología , ARN Interferente Pequeño/genética , Sindecano-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Proteína bcl-X/genética , Proteína bcl-X/fisiología
4.
J Immunol ; 186(12): 6673-82, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21543644

RESUMEN

Sle2c1 is an NZM2410- and NZB-derived lupus susceptibility locus that induces an expansion of the B1a cell compartment. B1a cells have a repertoire enriched for autoreactivity, and an expansion of this B cell subset occurs in several mouse models of lupus. A combination of genetic mapping and candidate gene analysis presents Cdkn2c, a gene encoding for cyclin-dependent kinase inhibitor p18(INK4c) (p18), as the top candidate gene for inducing the Slec2c1-associated expansion of B1a cells. A novel single nucleotide polymorphism in the NZB allele of the Cdkn2c promoter is associated with a significantly reduced Cdkn2c expression in the splenic B cells and peritoneal cavity B1a cells from Sle2c1-carrying mice, which leads to a defective G1 cell cycle arrest in splenic B cells and increased proliferation of peritoneal cavity B1a cells. As the cell cycle is differentially regulated in B1a and B2 cells, these results suggest that Cdkn2c plays a critical role in B1a cell self-renewal and that its impaired expression leads to an accumulation of these cells with high autoreactive potential.


Asunto(s)
Linfocitos B/patología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Predisposición Genética a la Enfermedad/genética , Homeostasis , Lupus Eritematoso Sistémico/patología , Animales , Autoinmunidad/genética , Subgrupos de Linfocitos B/patología , Linfocitos B/citología , Linfocitos B/fisiología , Ciclo Celular , Proliferación Celular , Mapeo Cromosómico , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Sitios Genéticos/genética , Lupus Eritematoso Sistémico/genética , Recuento de Linfocitos , Ratones , Polimorfismo de Nucleótido Simple
5.
Int J Cancer ; 124(2): 339-45, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18942719

RESUMEN

In multiple endocrine neoplasia syndrome Type 2 (MEN2), medullary thyroid carcinoma (MTC) and pheochromocytoma (PC) are associated with hereditary activating germ-line mutations in the RET proto-oncogene. Also in a large percentage of sporadic MTCs and PCs, somatic RET mutations appear to be involved in tumor formation. In one single MEN2 family an extensive variety in disease expression may be observed, indicating that additional genetic events are responsible for progression of the disease towards a more aggressive phenotype. However, these additional mutations in both hereditary and sporadic MTC and PC development are largely unknown. Here, we show for the first time the presence of somatic mutations in the cell cycle regulator P18 in human RET-associated MTCs and PCs. Each of these mutations causes an amino acid substitution in the cyclin dependent kinase-interacting region of P18(INK4C). Since these mutations partly inhibited P18(INK4C) function and reduced its stability, our findings implicate P18 as a tumor suppressor gene involved in human MTC and PC development.


Asunto(s)
Carcinoma Medular/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Feocromocitoma/metabolismo , Neoplasias de la Tiroides/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Análisis Mutacional de ADN , Progresión de la Enfermedad , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Proto-Oncogenes Mas , Homología de Secuencia de Aminoácido
6.
Mol Cell Biol ; 26(12): 4564-76, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16738322

RESUMEN

Inactivation of the Rb-mediated G1 control pathway is a common event found in many types of human tumors. To test how the Rb pathway interacts with other pathways in tumor suppression, we characterized mice with mutations in both the cyclin-dependent kinase (CDK) inhibitor p18 Ink4c and the lipid phosphatase Pten, which regulates cell growth. The double mutant mice develop a wider spectrum of tumors, including prostate cancer in the anterior and dorsolateral lobes, with nearly complete penetrance and at an accelerated rate. The remaining wild-type allele of Pten was lost at a high frequency in Pten+/- cells but not in p18+/- Pten+/- or p18-/- Pten+/- prostate tumor cells, nor in other Pten+/- tumor cells, suggesting a tissue- and genetic background-dependent haploinsufficiency of Pten in tumor suppression. p18 deletion, CDK4 overexpression, or oncoviral inactivation of Rb family proteins caused activation of Akt/PKB that was recessive to the reduction of PTEN activity. We suggest that p18 and Pten cooperate in tumor suppression by constraining a positive regulatory loop between cell growth and cell cycle control pathways.


Asunto(s)
Ciclo Celular/fisiología , Proliferación Celular , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Fosfohidrolasa PTEN/fisiología , Neoplasias de las Glándulas Suprarrenales/etiología , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/patología , Animales , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Fosfohidrolasa PTEN/genética , Neoplasias Hipofisarias/etiología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología , Transducción de Señal , Neoplasias de la Tiroides/etiología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
7.
Oncogene ; 26(20): 2833-9, 2007 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-17099725

RESUMEN

p18(Ink4c) functions as a dedicated inhibitor of cyclin-D-dependent kinases. Loss of Ink4c predisposes mice to tumor development and, in a dose-dependent manner, complements the tumor-promoting effects of various oncogenes. We have now addressed whether Ink4c loss impacts B-cell tumor development in the Emu-Myc transgenic mouse, a model of human Burkitt lymphoma. Loss of one or both alleles did not influence the onset of lymphoma in Emu-Myc transgenics, and did not appreciably affect Myc's proliferative or apoptotic responses in precancerous B cells. Nevertheless, Ink4c loss modulated the effects of Myc-induced transformation by decreasing the frequency of Arf loss, an ordinarily common event in Emu-Myc-induced lymphomas.


Asunto(s)
Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Linfoma de Células B/genética , Linfoma de Células B/prevención & control , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , Apoptosis/genética , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Linfoma de Células B/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/fisiología , Proteínas Proto-Oncogénicas c-myc/genética
8.
Mol Cells ; 41(5): 381-389, 2018 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-29665672

RESUMEN

ARF is a tumor suppressor protein that has a pivotal role in the prevention of cancer development through regulating cell proliferation, senescence, and apoptosis. As a factor that induces senescence, the role of ARF as a tumor suppressor is closely linked to the p53-MDM2 axis, which is a key process that restrains tumor formation. Thus, many cancer cells either lack a functional ARF or p53, which enables them to evade cell oncogenic stress-mediated cycle arrest, senescence, or apoptosis. In particular, the ARF gene is a frequent target of genetic and epigenetic alterations including promoter hyper-methylation or gene deletion. However, as many cancer cells still express ARF, pathways that negatively modulate transcriptional or post-translational regulation of ARF could be potentially important means for cancer cells to induce cellular proliferation. These recent findings of regulators affecting ARF protein stability along with its low levels in numerous human cancers indicate the significance of an ARF post-translational mechanism in cancers. Novel findings of regulators stimulating or suppressing ARF function would provide new therapeutic targets to manage cancer- and senescence-related diseases. In this review, we present the current knowledge on the regulation and alterations of ARF expression in human cancers, and indicate the importance of regulators of ARF as a prognostic marker and in potential therapeutic strategies.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas de Neoplasias/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Apoptosis , División Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes p16 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteína p14ARF Supresora de Tumor/deficiencia , Proteína p14ARF Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología , Ubiquitinación
9.
Islets ; 5(4): 156-69, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23896637

RESUMEN

Adult human pancreatic ß-cells are primarily quiescent (G0) yet the mechanisms controlling their quiescence are poorly understood. Here, we demonstrate, by immunofluorescence and confocal microscopy, abundant levels of the critical negative cell cycle regulators, p27(Kip1) and p18(Ink4c), 2 key members of cyclin-dependent kinase (CDK) inhibitor family, and glycogen synthase kinase-3 (GSK-3), a serine-threonine protein kinase, in islet ß-cells of adult human pancreatic tissue. Our data show that p27(Kip1) localizes primarily in ß-cell nuclei, whereas, p18(Ink4c) is mostly present in ß-cell cytosol. Additionally, p-p27(S10), a phosphorylated form of p27(Kip1), which was shown to interact with and to sequester cyclinD-CDK4/6 in the cytoplasm, is present in substantial amounts in ß-cell cytosol. Our immunofluorescence analysis displays similar distribution pattern of p27(Kip1), p-p27(S10), p18(Ink4c) and GSK-3 in islet ß-cells of adult mouse pancreatic tissue. We demonstrate marked interaction of p27(Kip1) with cyclin D3, an abundant D-type cyclin in adult human islets, and vice versa as well as with its cognate kinase partners, CDK4 and CDK6. Likewise, we show marked interaction of p18(Ink4c) with CDK4. The data collectively suggest that inhibition of CDK function by p27(Kip1) and p18(Ink4c) contributes to human ß-cell quiescence. Consistent with this, we have found by BrdU incorporation assay that combined treatments of small molecule GSK-3 inhibitor and mitogen/s lead to elevated proliferation of human ß-cells, which is caused partly due to p27(Kip1) downregulation. The results altogether suggest that ex vivo expansion of human ß-cells is achievable via increased proliferation for ß-cell replacement therapy in diabetes.


Asunto(s)
Ciclo Celular/genética , Proliferación Celular , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Células Secretoras de Insulina/fisiología , Adulto , Anciano , Animales , Recuento de Células , Células Cultivadas , Quinasas Ciclina-Dependientes/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Células Secretoras de Insulina/citología , Ratones , Persona de Mediana Edad
10.
Cancer Res ; 72(1): 165-75, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22080569

RESUMEN

The cyclin-dependent kinase (CDK) inhibitors, p18(INK4c) and p16(INK4a), both have the credentials of tumor suppressors in human cancers and mouse models. For p16(INK4a), the underlying rationale is its role in senescence, but the selective force for inactivation of p18(INK4c) in incipient cancer cells is less clear. Here, we show that in human fibroblasts undergoing replicative or oncogene-induced senescence, there is a marked decline in the levels of p18(INK4c) protein and RNA, which mirrors the accumulation of p16(INK4a). Downregulation of INK4c is not dependent on p16(INK4a), and RAS can promote the loss of INK4c without cell-cycle arrest. Downregulation of p18(INK4c) correlates with reduced expression of menin and E2F1 but is unaffected by acute cell-cycle arrest or inactivation of the retinoblastoma protein (pRb). Collectively, our data question the idea that p18(INK4c) acts as a backup for loss of p16(INK4a) and suggest that the apparent activation of p18(INK4c) in some settings represents delayed senescence rather than increased expression. We propose that the contrasting behavior of the two very similar INK4 proteins could reflect their respective roles in senescence versus differentiation.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Genes Supresores de Tumor , Oncogenes , Secuencia de Bases , Regulación hacia Abajo , Humanos , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Diabetes ; 61(3): 632-41, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22338094

RESUMEN

Pancreatic ß-cell proliferation is infrequent in adult humans and is not increased in type 2 diabetes despite obesity and insulin resistance, suggesting the existence of inhibitory factors. Free fatty acids (FFAs) may influence proliferation. In order to test whether FFAs restrict ß-cell proliferation in vivo, mice were intravenously infused with saline, Liposyn II, glucose, or both, continuously for 4 days. Lipid infusion did not alter basal ß-cell proliferation, but blocked glucose-stimulated proliferation, without inducing excess ß-cell death. In vitro exposure to FFAs inhibited proliferation in both primary mouse ß-cells and in rat insulinoma (INS-1) cells, indicating a direct effect on ß-cells. Two of the fatty acids present in Liposyn II, linoleic acid and palmitic acid, both reduced proliferation. FFAs did not interfere with cyclin D2 induction or nuclear localization by glucose, but increased expression of inhibitor of cyclin dependent kinase 4 (INK4) family cell cycle inhibitors p16 and p18. Knockdown of either p16 or p18 rescued the antiproliferative effect of FFAs. These data provide evidence for a novel antiproliferative form of ß-cell glucolipotoxicity: FFAs restrain glucose-stimulated ß-cell proliferation in vivo and in vitro through cell cycle inhibitors p16 and p18. If FFAs reduce proliferation induced by obesity and insulin resistance, targeting this pathway may lead to new treatment approaches to prevent diabetes.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Ácidos Grasos no Esterificados/fisiología , Glucosa/farmacología , Células Secretoras de Insulina/fisiología , Animales , Ciclina D2/biosíntesis , Emulsiones/farmacología , Ácidos Grasos no Esterificados/sangre , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfolípidos/farmacología , Ratas , Aceite de Cártamo/farmacología , Aceite de Soja/farmacología
13.
Ment Retard Dev Disabil Res Rev ; 12(2): 85-96, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16807909

RESUMEN

In white matter disorders such as leukodystrophies (LD), periventricular leucomalacia (PVL), or multiple sclerosis (MS), the hypomyelination or the remyelination failure by oligodendrocyte progenitor cells involves errors in the sequence of events that normally occur during development when progenitors proliferate, migrate through the white matter, contact the axon, and differentiate into myelin-forming oligodendrocytes. Multiple mechanisms underlie the eventual progressive deterioration that typifies the natural history of developmental demyelination in LD and PVL and of adult-onset demyelination in MS. Over the past few years, pathophysiological studies have mostly focused on seeking abnormalities that impede oligodendroglial maturation at the level of migration, myelination, and survival. In contrast, there has been a strikingly lower interest for early proliferative and differentiation events that are likely to be equally critical for white matter development and myelin repair. This review highlights the Yin and Yang principles of interactions between intrinsic factors that coordinately regulate progenitor cell division and the onset of differentiation, i.e. the initial steps of oligodendrocyte lineage progression that are obviously crucial in health and diseases.


Asunto(s)
Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Oligodendroglía/patología , Adulto , Ciclina E/fisiología , Quinasa 2 Dependiente de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Humanos , Recién Nacido , Leucomalacia Periventricular/genética , Leucomalacia Periventricular/patología , Esclerosis Múltiple/patología , Vaina de Mielina/genética , Vaina de Mielina/patología , Fosforilación , Células Madre/patología
14.
Arthritis Rheum ; 54(7): 2074-83, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16802342

RESUMEN

OBJECTIVE: It is known that the cyclin-dependent kinase inhibitor (CDKI) gene p21(Cip1) suppresses rheumatoid inflammation by down-modulating type I interleukin-1 receptor (IL-1RI) expression and inhibiting JNK activity. The purpose of this study was to determine whether CDK activity directly modulates the production of inflammatory molecules in patients with rheumatoid arthritis (RA). METHODS: Genes for the CDKIs p16(INK4a) and p18(INK4c), a constitutively active form of retinoblastoma (RB) gene product, cyclin D1, and CDK-4, were transferred into RA synovial fibroblasts (RASFs). RASFs were also treated with a synthetic CDK-4/6 inhibitor (CDK4I). Levels of matrix metalloproteinase 3 (MMP-3), monocyte chemoattractant protein 1 (MCP-1), and IL-1RI expression were determined by Northern blotting, real-time polymerase chain reaction analysis, and enzyme-linked immunosorbent assay. CDKIs were immunoprecipitated to reveal their association with JNK. RESULTS: Transfer of the p16(INK4a) and p18(INK4c) genes and CDK4I suppressed the production of MMP-3 and MCP-1. Unlike p21(Cip1), neither CDKI gene inhibited IL-1RI or JNK. The expression of MMP-3 was up-regulated when CDK-4 activity was augmented. This regulation functioned at the messenger RNA (mRNA) level in MMP-3, but not in MCP-1. Transfer of active RB suppressed the production of MMP-3 and MCP-1 without changing their mRNA levels. CONCLUSION: CDK-4/6 modulated the production of MMP-3 and MCP-1. MMP-3 production was regulated primarily at the mRNA level in an RB-independent manner, whereas MCP-1 production was controlled posttranscriptionally by RB. These results show that cell cycle proteins are associated with control of mediators of inflammation through multiple pathways.


Asunto(s)
Artritis Reumatoide/genética , Artritis Reumatoide/fisiopatología , Quimiocina CCL2/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Proteína de Retinoblastoma/fisiología , Artritis Reumatoide/patología , Proliferación Celular , Células Cultivadas , Quimiocina CCL2/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Metaloproteinasa 3 de la Matriz/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Receptores Tipo I de Interleucina-1 , Proteína de Retinoblastoma/genética , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
15.
Genes Dev ; 19(22): 2656-67, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16260494

RESUMEN

Recurrent genetic alterations in human medulloblastoma (MB) include mutations in the sonic hedgehog (SHH) signaling pathway and TP53 inactivation (approximately 25% and 10% of cases, respectively). However, mouse models of MB, regardless of their initiating lesions, generally depend upon p53 inactivation for rapid onset and high penetrance. The gene encoding the cyclin-dependent kinase inhibitor p18(Ink4c) is transiently expressed in mouse cerebellar granule neuronal precursor cells (GNPs) as they exit the cell division cycle and differentiate. Coinactivation of Ink4c and p53 provided cultured GNPs with an additive proliferative advantage, either in the presence or absence of Shh, and induced MB with low penetrance but with greatly increased incidence following postnatal irradiation. In contrast, mice lacking one or two functional Ink4c alleles and one copy of Patched (Ptc1) encoding the Shh receptor rapidly developed MBs that retained wild-type p53. In tumor cells purified from double heterozygotes, the wild-type Ptc1 allele, but not Ink4c, was inactivated. Therefore, when combined with Ptc1 mutation, Ink4c is haploinsufficient for tumor suppression. Methylation of INK4C (CDKN2C) was observed in four of 23 human MBs, and p18(INK4C) protein expression was extinguished in 14 of 73 cases. Hence, p18(INK4C) loss may contribute to MB formation in children.


Asunto(s)
Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Meduloblastoma/metabolismo , Receptores de Superficie Celular/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Proliferación Celular , Células Cultivadas , Cerebelo/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Proteínas Hedgehog , Humanos , Meduloblastoma/etiología , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Noqueados , Receptores Patched , Receptor Patched-1 , Transducción de Señal/fisiología , Transactivadores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA