Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Paediatr Child Health ; 28(7): 399-403, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37885599

RESUMEN

Innovative therapeutic approaches are needed to alleviate the burden of life-limiting, rare, and chronic conditions affecting children, adolescents, and young adults (CAYA). This includes a need for improved access to both clinical research and to non-approved or off-label therapies, together with, ultimately, more therapies achieving regulatory approval in Canada. The single patient study (SPS), also known as an open label individual patient (OLIP) study, was introduced by Health Canada to open access to non-marketed drugs where a clinical trial is not readily available, but the drug is considered too investigational to be managed on a standard Special Access Program. SPS is designed for patients who have a serious or life-threatening condition and have exhausted available treatment options. Our report summarizes this relatively new development in the Canadian regulatory environment and highlights the opportunities and challenges as identified by regulators, pharmaceutical representatives, academic researchers, and patient/parent advocates.

2.
EMBO J ; 31(8): 1865-78, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22373579

RESUMEN

In response to DNA damage, cells initiate complex signalling cascades leading to growth arrest and DNA repair. The recruitment of 53BP1 to damaged sites requires the activation of the ubiquitination cascade controlled by the E3 ubiquitin ligases RNF8 and RNF168, and methylation of histone H4 on lysine 20. However, molecular events that regulate the accessibility of methylated histones, to allow the recruitment of 53BP1 to DNA breaks, are unclear. Here, we show that like 53BP1, the JMJD2A (also known as KDM4A) tandem tudor domain binds dimethylated histone H4K20; however, JMJD2A is degraded by the proteasome following the DNA damage in an RNF8-dependent manner. We demonstrate that JMJD2A is ubiquitinated by RNF8 and RNF168. Moreover, ectopic expression of JMJD2A abrogates 53BP1 recruitment to DNA damage sites, indicating a role in antagonizing 53BP1 for methylated histone marks. The combined knockdown of JMJD2A and JMJD2B significantly rescued the ability of RNF8- and RNF168-deficient cells to form 53BP1 foci. We propose that the RNF8-dependent degradation of JMJD2A regulates DNA repair by controlling the recruitment of 53BP1 at DNA damage sites.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Reparación del ADN , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilación , Modelos Biológicos , Datos de Secuencia Molecular , Unión Proteica , Transducción de Señal , Proteína 1 de Unión al Supresor Tumoral P53
3.
J Biol Chem ; 288(29): 21376-21388, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23744078

RESUMEN

DNA damage evokes a complex and highly coordinated DNA damage response (DDR) that is integral to the suppression of genomic instability. Double-strand breaks (DSBs) are considered the most deleterious form damage. Evidence suggests that trimethylation of histone H3 lysine 9 (H3K9me3) presents a barrier to DSB repair. Also, global levels of histone methylation are clinically predictive for several tumor types. Therefore, demethylation of H3K9 may be an important step in the repair of DSBs. The KDM4 subfamily of demethylases removes H3K9 tri- and dimethylation and contributes to the regulation of cellular differentiation and proliferation; mutation or aberrant expression of KDM4 proteins has been identified in several human tumors. We hypothesize that members of the KDM4 subfamily may be components of the DDR. We found that Kdm4b-enhanced GFP (EGFP) and KDM4D-EGFP were recruited rapidly to DNA damage induced by laser micro-irradiation. Focusing on the clinically relevant Kdm4b, we found that recruitment was dependent on poly(ADP-ribose) polymerase 1 activity as well as Kdm4b demethylase activity. The Kdm4 proteins did not measurably accumulate at γ-irradiation-induced γH2AX foci. Nevertheless, increased levels of Kdm4b were associated with decreased numbers of γH2AX foci 6 h after irradiation as well as increased cell survival. Finally, we found that levels of H3K9me2 and H3K9me3 were decreased at early time points after 2 gray of γ-irradiation. Taken together, these data demonstrate that Kdm4b is a DDR protein and that overexpression of Kdm4b may contribute to the failure of anti-cancer therapy that relies on the induction of DNA damage.


Asunto(s)
Daño del ADN , Rayos gamma , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de la radiación , Proteína Quinasa Activada por ADN/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/metabolismo , Histonas/metabolismo , Humanos , Rayos Láser , Lisina/metabolismo , Metilación/efectos de la radiación , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transporte de Proteínas/efectos de la radiación , Proteínas Recombinantes de Fusión/metabolismo
4.
Biochem Cell Biol ; 91(6): 369-77, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24219278

RESUMEN

The Jumonji D2 proteins (JMJD2/KDM4) function to demethylate di- and trimethylated (me2/3) histone 3 lysine 9 (H3K9me2/3) and H3K36me3. Knockout mouse models for Kdm4b and Kdm4d have not resulted in gross abnormalities, while mouse models for Kdm4a and Kdm4c have not been reported. However, the KDM4 subfamily of demethylases are overexpressed in several tumor types. Overexpression of KDM4 proteins alters transcription and chromatin remodeling, driving cellular proliferation, anchorage-independent growth, invasion, and migration. Increased proliferation occurs through KDM4-mediated modification of cell cycle timing, as well as through increased numbers of replication forks. Recent evidence also suggests that KDM4C overexpression contributes to the maintenance of a pluripotent state. Together these data suggest that overexpression of KDM4 proteins induces numerous oncogenic effects.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/genética , Histonas/genética , Neoplasias/genética , Animales , Ciclo Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ensamble y Desensamble de Cromatina , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Metilación , Ratones , Ratones Transgénicos , Neoplasias/enzimología , Neoplasias/patología , Transducción de Señal
5.
Am J Pathol ; 179(1): 411-21, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21703420

RESUMEN

The fusion tyrosine kinase NPM-ALK is central to the pathogenesis of ALK-positive anaplastic large cell lymphoma (ALK(+)ALCL). We recently identified that MSH2, a key DNA mismatch repair (MMR) protein integral to the suppression of tumorigenesis, is an NPM-ALK-interacting protein. In this study, we found in vitro evidence that enforced expression of NPM-ALK in HEK293 cells suppressed MMR function. Correlating with these findings, six of nine ALK(+)ALCL tumors displayed evidence of microsatellite instability, as opposed to none of the eight normal DNA control samples (P = 0.007, Student's t-test). Using co-immunoprecipitation, we found that increasing levels of NPM-ALK expression in HEK293 cells resulted in decreased levels of MSH6 bound to MSH2, whereas MSH2·NPM-ALK binding was increased. The NPM-ALK·MSH2 interaction was dependent on the activation/autophosphorylation of NPM-ALK, and the Y191 residue of NPM-ALK was a crucial site for this interaction and NPM-ALK-mediated MMR suppression. MSH2 was found to be tyrosine phosphorylated in the presence of NPM-ALK. Finally, NPM-ALK impeded the expected DNA damage-induced translocation of MSH2 out of the cytoplasm. To conclude, our data support a model in which the suppression of MMR by NPM-ALK is attributed to its ability to interfere with normal MSH2 biochemistry and function.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/metabolismo , Linfoma de Células B Grandes Difuso/patología , Linfoma Anaplásico de Células Grandes/patología , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Citoplasma/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/metabolismo , Inestabilidad de Microsatélites , Proteína 2 Homóloga a MutS/genética , Fosforilación , Multimerización de Proteína , Transporte de Proteínas , Proteínas Tirosina Quinasas/genética , Células Tumorales Cultivadas , Tirosina/metabolismo
6.
Mol Cell Proteomics ; 9(7): 1616-32, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20393185

RESUMEN

The oncogenic fusion protein nucleophosmin-anaplastic lymphoma kinase (NPM-ALK), found exclusively in a subset of ALK-positive anaplastic large cell lymphoma, promotes tumorigenesis by exerting its constitutively active tyrosine kinase activity. Thus, characterization of the NPM-ALK-induced changes in the phosphoproteome will likely provide insights into the biology of this oncoprotein. To achieve this goal, we used a strategy of combining sequential affinity purification of phosphopeptides and LC/MS. GP293 cells transfected with either NPM-ALK or an NPM-ALK mutant with decreased tyrosine kinase activity (negative control) were used. We identified 506 phosphoproteins detectable in NPM-ALK-expressing cells but not in the negative control. Bioinformatics analysis revealed that these phosphoproteins carry a wide diversity of biological functions, some of which have not been described in association with NPM-ALK, such as the tumor necrosis factor (TNF)/Fas/tumor necrosis factor-related apoptosis-induced ligand (TRAIL) signaling pathway and the ubiquitin proteasome degradation pathway. In particular, modulations of the TNF/Fas/TRAIL pathway by NPM-ALK were supported by our antibody microarray data. Further validation of the TNF/Fas/TRAIL pathway was performed in ALK(+) anaplastic large cell lymphoma (ALCL) cell lines with knockdown of NPM-ALK using short interference RNA, resulting in the loss of the tyrosine phosphorylation of tumor necrosis factor receptor-associated protein 1 (TRAP1) and receptor-interacting protein 1, two crucial TNF signaling molecules. Functional analyses revealed that knockdown of TRAP1 facilitated cell death induced by TRAIL or doxorubicin in ALK(+) ALCL cells. This suggests that down-regulation of TRAP1 in combination with TRAIL or doxorubicin might be a potential novel therapeutic strategy for ALK(+) ALCL. These findings demonstrated that our strategy allowed the identification of novel proteins downstream of NPM-ALK that contribute to the maintenance of neoplastic phenotype and holds great potential for future studies of cellular tyrosine kinases in normal states and diseases.


Asunto(s)
Linfoma Anaplásico de Células Grandes/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteoma/análisis , Transducción de Señal/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Datos de Secuencia Molecular , Fosfoproteínas/genética , Proteínas Tirosina Quinasas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
7.
Carcinogenesis ; 32(2): 146-53, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21045017

RESUMEN

It is well established that the tumorigenic potential of nucleophosmin (NPM)-anaplastic lymphoma kinase (ALK), an oncogenic tyrosine kinase, is dependent on its tyrosine phosphorylation. Using tandem affinity purification-mass spectrometry, we found evidence of phosphorylation of three serine residues of NPM-ALK (Serine¹³5, Serine¹64 and Serine497) ectopically expressed in GP293 cells. Using a specific anti-phosphoserine antibody and immunoprecipitation, we confirmed the presence of serine phosphorylation of NPM-ALK in all three NPM-ALK-expressing cell lines examined. Similar to the tyrosine phosphorylation, phosphorylation of these serine residues was dependent on the activation status of the kinase activation loop of ALK. All of these three serine residues are biologically important as mutation of any one of these residues resulted in a significant reduction in the tumorigenicity of NPM-ALK (assessed by cell viability and clonogenic assay), which correlated with a substantial reduction in the phosphorylation of extracellular signal-regulated kinase 1/2, c-jun N-terminal kinase and signal transducer and activator of transcription 6. Serine phosphorylation of NPM-ALK appears to be regulated by multiple serine kinases since it was markedly reduced by pharmacologic inhibitors for glycogen synthase kinase-3, casein kinase I or mitogen-activated protein kinases. In summary, our study is the first to identify serine phosphorylation of NPM-ALK and to provide evidence that it enhances the tumorigenic potential of this oncogenic protein.


Asunto(s)
Neoplasias/etiología , Proteínas Tirosina Quinasas/metabolismo , Serina/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , Invasividad Neoplásica , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/química , Tirosina/metabolismo
8.
J Biol Chem ; 285(1): 95-103, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19887368

RESUMEN

Previous studies have shown that the kinase activation loop (KAL) of the oncogenic fusion protein NPM-ALK regulates its overall tyrosine phosphorylation status and tumorigenicity. Using tandem affinity purification-mass spectrometry, we assessed how the KAL of NPM-ALK regulates the phosphorylation status of its individual tyrosines. Using the lysates of GP293 cells transfected with NPM-ALK, our highly reproducible results showed evidence of phosphorylation in all 3 tyrosines in KAL and 8 tyrosines outside KAL. We created 7 KAL mutants, each of which carried a Tyr-to-Phe mutation of >or=1 of the 3 tyrosines in KAL. A complete loss of the 8 phosphotyrosines outside KAL was found in 3 KAL mutants, and their oncogenicity (assessed by cell viability, colony formation, and the ability to phosphorylate effector proteins) was abrogated. A partial loss of the 8 phosphotyrosines was found in 4 KAL mutants, but their oncogenicity did not show simple correlation with the number of residual phosphotyrosines. Tyr-to-Phe mutations of each of the 8 phosphotyrosines outside KAL did not result in a significant decrease in the oncogenicity. In conclusion, we have provided details of how the KAL in NPM-ALK regulates its tyrosine phosphorylation pattern. Our results challenge some of the current concepts regarding the relationship between the tyrosine phosphorylation and oncogenicity of NPM-ALK.


Asunto(s)
Cromatografía de Afinidad/métodos , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Espectrometría de Masas en Tándem/métodos , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Línea Celular , Supervivencia Celular , Cromatografía Liquida , Activación Enzimática , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Neoplasias/patología , Fosforilación , Fosfotirosina/metabolismo , Estructura Secundaria de Proteína , Proteínas Tirosina Quinasas/aislamiento & purificación , Relación Estructura-Actividad
9.
Blood ; 114(2): 360-70, 2009 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-19423729

RESUMEN

Type I insulin-like growth factor receptor (IGF-IR) tyrosine kinase plays important roles in the pathogenesis of several malignancies. Although it promotes the growth of stimulated hematopoietic cells, a direct role of IGF-IR in malignant lymphoma has not been identified. Anaplastic lymphoma kinase-positive anaplastic large-cell lymphoma (ALK(+) ALCL) is a unique type of T-cell lymphoma. Approximately 85% of ALK(+) ALCL cases harbor the translocation t(2;5)(p23;q35), which generates the chimeric oncogene NPM-ALK. In the present study, we explored a possible role of IGF-IR in ALK(+) ALCL. Our results demonstrate that IGF-IR and IGF-I are widely expressed in ALK(+) ALCL cell lines and primary tumors. Importantly, we identified novel reciprocal functional interactions between IGF-IR and NPM-ALK. Antagonism of IGF-IR decreased the viability, induced apoptosis and cell-cycle arrest, and decreased proliferation and colony formation of ALK(+) ALCL cell lines. These effects could be explained by alterations of cell survival regulatory proteins downstream of IGF-IR signaling. Our findings improve current understanding of the biology of IGF-IR and NPM-ALK and have significant therapeutic implications as they identify IGF-IR signaling as a potential therapeutic target in ALK(+) ALCL and possibly other types of malignant lymphoma.


Asunto(s)
Linfoma Anaplásico de Células Grandes/enzimología , Linfoma Anaplásico de Células Grandes/patología , Proteínas Tirosina Quinasas/metabolismo , Receptor IGF Tipo 1/metabolismo , Linfocitos T/enzimología , Quinasa de Linfoma Anaplásico , Animales , Línea Celular , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Linfoma Anaplásico de Células Grandes/genética , Ratones , Unión Proteica , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas Receptoras , Receptor IGF Tipo 1/genética , Transducción de Señal
10.
Am J Pathol ; 174(2): 361-70, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19131589

RESUMEN

Nucleophosmin-anaplastic lymphoma kinase (NPM-ALK), an oncogenic fusion gene protein that is characteristically found in a subset of anaplastic large cell lymphomas, promotes tumorigenesis through its functional and physical interactions with various biologically important proteins. The identification of these interacting proteins has proven to be useful to further our understanding of NPM-ALK-mediated tumorigenesis. For the first time, we performed a proteome-wide identification of NPM-ALK-binding proteins using tandem affinity purification and a highly sensitive mass spectrometric technique. Tandem affinity purification is a recently developed method that carries a lower background and higher sensitivity compared with the conventional immunoprecipitation-based protein purification protocols. The NPM-ALK gene was cloned into an HB-tagged vector and expressed in GP293 cells. Three independent experiments were performed and the reproducibility of the data was 68%. The vast majority of the previously reported NPM-ALK-binding proteins were detected. We also identified proteins that are involved in various cellular processes that were not previously described in association with NPM-ALK, such as MCM6 and MSH2 (DNA repair), Nup98 and importin 8 (subcellular protein transport), Stim1 (calcium signaling), 82Fip (RNA regulation), and BAG2 (proteosome degradation). We believe that these data highlight the functional diversity of NPM-ALK and provide new research directions for the study of the biology of this oncoprotein.


Asunto(s)
Proteínas Tirosina Quinasas/metabolismo , Proteómica , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Western Blotting , Proteínas Portadoras/metabolismo , Línea Celular , Humanos , Inmunoprecipitación , Datos de Secuencia Molecular , Proteínas Tirosina Quinasas/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem , Transfección
11.
Carcinogenesis ; 28(10): 2131-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17615258

RESUMEN

DNA mismatch repair (MMR) is a highly conserved system that repairs DNA adducts acquired during replication, as well as some forms of exogenous/endogenous DNA damage. Additionally, MMR proteins bind to DNA adducts that are not removed by MMR and influence damage-response mechanisms other than repair. Hereditary non-polyposis colorectal cancer, as well as mouse models for MMR deficiency, illustrate that MMR proteins are required for maintenance of genetic stability and tumor suppression. In both humans and mice, the phenotype associated with Msh6-associated tumorigenesis is distinct from that of Msh2. In this study, we hypothesized that Msh6-/-;p53+/- mice would display earlier tumor onset than their Msh6-/- or p53+/- counterparts, indicating that concomitant loss of these two tumor suppressors contributes to tumorigenesis via mechanisms that are only partially interrelated. We generated a Msh6-/-;p53+/- mouse model which succumbed to malignant disease at an accelerated rate and with a tumor spectrum distinct from both Msh6-/- and p53+/- models. Alteration of tumor phenotype in the Msh6-/-;p53+/- mice included a marked increase in microsatellite instability that was associated with loss of heterozygosity of the remaining p53 allele. Also, genetic instability was inversely correlated with survival. This manuscript marks the first in vivo investigation into the association between Msh6 and p53, and their combined role in the suppression of spontaneous tumorigenesis, cell survival and genomic stability. Our results support the hypothesis that p53 and Msh6 are functionally interrelated and that, with concomitant mutation, these tumor suppressors act together to accelerate tumorigenesis.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Inestabilidad Cromosómica , Secuencia Conservada , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Muerte , Genes p53 , Genotipo , Neoplasias Renales/genética , Neoplasias Renales/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Pérdida de Heterocigocidad , Ratones , Ratones Noqueados , Repeticiones de Microsatélite , Neoplasias del Bazo/genética , Neoplasias del Bazo/patología , Neoplasias del Timo/genética , Neoplasias del Timo/patología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
12.
Schizophr Res ; 170(2-3): 245-51, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26792297

RESUMEN

BACKGROUND: The association between mental illness and osteoporosis and fractures is particularly pronounced in psychotic disorders. Antipsychotic use has previously been described to affect bone density. METHOD: A 52-week follow-up of patients switched to aripiprazole or with aripiprazole added on, conducting a specific analysis of markers of bone turnover: urinary NTX (a biomarker of bone resorption) and serum BSAP (a biomarker of bone formation). Baseline and serial measurements of bone markers NTX, BSAP and of hormones prolactin, oestrogen and testosterone were done at weeks 0 and 1, 2, 6, 12, 26 and 52, respectively. RESULTS: NTX concentration reduced over time but this did not reach significance in the whole group (log-NTX: ß=-0.0012, p=0.142). For BSAP the addition of or replacement with aripiprazole produced a significant reduction (log-BSAP: ß=-0.00039, p=0.002). Analysis with prolactin similarly showed a significant reduction (log-prolactin: ß=-0.0024, p<0.001); other hormones did not change significantly. Sensitivity analysis to compare the switchers to aripiprazole versus the "add-on" showed that the former group had a significant reduction in NTX. CONCLUSIONS: We found that switching to aripiprazole was associated with changes in molecular biomarkers of bone resorption, indicating a more favourable profile for bone health.


Asunto(s)
Antipsicóticos/efectos adversos , Aripiprazol/efectos adversos , Remodelación Ósea/efectos de los fármacos , Remodelación Ósea/fisiología , Adolescente , Adulto , Antipsicóticos/uso terapéutico , Aripiprazol/uso terapéutico , Biomarcadores/metabolismo , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/metabolismo , Estrógenos/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Factor de Transcripción PAX5/sangre , Prolactina/metabolismo , Estudios Prospectivos , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/metabolismo , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Sensibilidad y Especificidad , Testosterona/metabolismo , Adulto Joven
13.
DNA Repair (Amst) ; 2(4): 427-35, 2003 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-12606123

RESUMEN

DNA mismatch repair (MMR) is integral to the maintenance of genomic stability and more recently has been demonstrated to affect apoptosis and cell cycle arrest in response to a variety of adducts induced by exogenous agents. Comparing Msh2-null and wildtype mouse embryonic fibroblasts (MEFs), both primary and transformed, we show that Msh2 deficiency results in increased survival post-UVB, and that UVB-induced apoptosis is significantly reduced in Msh2-deficient cells. Furthermore, p53 phosphorylation at serine 15 is delayed or diminished in Msh2-deficient cells, suggesting that Msh2 may act upstream of p53 in a post-UVB apoptosis or growth arrest response pathway. Taken together, these data suggest that MMR heterodimers containing Msh2 may function as a sensor of UVB-induced DNA damage and influence the initiation of UVB-induced apoptosis, thus implicating MMR in protecting against UV-induced tumorigenesis.


Asunto(s)
Apoptosis/fisiología , Daño del ADN/efectos de la radiación , Reparación del ADN , Proteínas de Unión al ADN , Proteína p53 Supresora de Tumor/fisiología , Animales , Ratones , Proteína 2 Homóloga a MutS , Fosforilación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Rayos Ultravioleta/efectos adversos
14.
J Invest Dermatol ; 121(3): 435-40, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12925197

RESUMEN

In addition to their established role in repairing post-replicative DNA errors, DNA mismatch repair proteins contribute to cell cycle arrest and apoptosis in response to a wide range of exogenous DNA damage (e.g., alkylation-induced lesions). The role of DNA mismatch repair in response to ultraviolet-induced DNA damage has been historically controversial. Recent data, however, suggest that DNA mismatch repair proteins probably do not contribute to the removal of ultraviolet-induced DNA damage, but may be important in suppressing mutagenesis, effecting apoptosis, and suppressing tumorigenesis following exposure to ultraviolet radiation.


Asunto(s)
Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Rayos Ultravioleta/efectos adversos , Animales , Reparación del ADN/fisiología , Humanos
15.
J Invest Dermatol ; 121(4): 876-80, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14632208

RESUMEN

Recent data support a role for DNA mismatch repair in the cellular response to some forms of exogenous DNA damage beyond that of DNA repair; cells with defective DNA mismatch repair have partial or complete failure to undergo apoptosis and/or G2M arrest following specific types of damage. We propose that the DNA mismatch repair Msh2/Msh6 heterodimer, responsible for the detection of DNA damage, promotes apoptosis in normal cells, thus protecting mammals from ultraviolet-induced malignant transformation. Using primary mouse embryonic fibroblasts derived from Msh6+/+ and Msh6-/- mice, we compare the response of DNA-mismatch repair-proficient and -deficient cells to ultraviolet B radiation. In the wild-type mouse embryonic fibroblasts, ultraviolet-B-induced increases in Msh6 protein levels were not dependent on p53. Msh6-/- mouse embryonic fibroblasts were significantly less sensitive to the cytotoxic effects of ultraviolet B radiation. Further comparison of the Msh6+/+ and Msh6-/- mouse embryonic fibroblasts revealed that Msh6-/- mouse embryonic fibroblasts undergo significantly less apoptosis following ultraviolet B irradiation, thus indicating that ultraviolet-B-induced apoptosis is partially Msh6 dependent. These data support a role for Msh6 in protective cellular responses of primary cells to ultraviolet-B-induced mutagenesis and, hence, the prevention of skin cancer.


Asunto(s)
Apoptosis/fisiología , Reparación del ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/fisiología , Animales , Apoptosis/efectos de la radiación , Supervivencia Celular/fisiología , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Fibroblastos/citología , Fibroblastos/efectos de la radiación , Ratones , Ratones Noqueados , Neoplasias Cutáneas/prevención & control , Proteína p53 Supresora de Tumor/genética , Rayos Ultravioleta/efectos adversos
16.
Cell Cycle ; 12(16): 2675-83, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23907130

RESUMEN

Polycomb protein histone methyltransferase, enhancer of Zeste homolog 2 (EZH2), is frequently overexpressed in human malignancy and is implicated in cancer cell proliferation and invasion. However, it is largely unknown whether EZH2 has a role in modulating the DNA damage response. Here, we show that polycomb repressive complex 2 (PRC2) is recruited to sites of DNA damage. This recruitment is independent of histone 2A variant X (H2AX) and the PI-3-related kinases ATM and DNA-PKcs. We establish that PARP activity is required for retaining PRC2 at sites of DNA damage. Furthermore, depletion of EZH2 in cells decreases the efficiency of DSB repair and increases sensitivity of cells to gamma-irradiation. These data unravel a crucial role of PRC2 in determining cancer cellular sensitivity following DNA damage and suggest that therapeutic targeting of EZH2 activity might serve as a strategy for improving conventional chemotherapy in a given malignancy.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Complejo Represivo Polycomb 2/fisiología , Inmunoprecipitación de Cromatina , Ensayo de Unidades Formadoras de Colonias , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Microscopía Fluorescente , Complejo Represivo Polycomb 2/metabolismo
17.
DNA Repair (Amst) ; 9(2): 161-8, 2010 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-20022306

RESUMEN

Many studies have shown that DNA mismatch repair (MMR) has a role beyond that of repair in response to several types of DNA damage, including ultraviolet radiation (UV). We have demonstrated previously that the MMR-dependent component of UVB-induced apoptosis is integral to the suppression of UVB-induced tumorigenesis. Here we demonstrate that Msh6-dependent UVB-induced apoptotic pathway is both activated via the mitochondria and p53-independent. In addition, we have shown for the first time that caspase 2, an initiator caspase, localizes to the centrosomes in mitotic primary mouse embryonic fibroblasts, irrespective of MMR status and UVB treatment.


Asunto(s)
Apoptosis , Caspasa 2/metabolismo , Centrosoma/enzimología , Reparación de la Incompatibilidad de ADN , Animales , Apoptosis/efectos de la radiación , Caspasa 9/metabolismo , Extractos Celulares , Células Cultivadas , Centrosoma/efectos de la radiación , Citocromos c/metabolismo , Reparación de la Incompatibilidad de ADN/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de la radiación , Potencial de la Membrana Mitocondrial/efectos de la radiación , Ratones , Microscopía Confocal , Mitocondrias/enzimología , Mitocondrias/efectos de la radiación , Proteína 2 Homóloga a MutS/metabolismo , Transporte de Proteínas/efectos de la radiación , Receptores de Muerte Celular/metabolismo , Transducción de Señal/efectos de la radiación , Fracciones Subcelulares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta , Proteína X Asociada a bcl-2/metabolismo
18.
Carcinogenesis ; 25(10): 1821-7, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15166087

RESUMEN

DNA mismatch repair (MMR) proteins are integral to the maintenance of genomic stability and suppression of tumorigenesis due to their role in repair of post-replicative DNA errors. Recent data also support a role for MMR proteins in cellular responses to exogenous DNA damage that does not involve removal of DNA adducts. We have demonstrated previously that both Msh2- and Msh6-null primary mouse embryonic fibroblasts are significantly less sensitive to UVB (ultraviolet B)-induced cytotoxicity and apoptosis than wild-type control cells. In order to ascertain the physiological relevance of the data we have exposed MMR-deficient mice to acute and chronic UVB radiation. We found that MMR-deficiency was associated with reduced levels of apoptosis and increased residual UVB-induced DNA adducts in the epidermis 24-h following acute UVB exposure. Moreover, Msh2-null mice developed UVB-induced skin tumors at a lower level of cumulative UVB exposure and with a greater severity of onset than wild-type mice. The Msh2-null skin tumors did not display microsatellite instability, suggesting that these tumors develop via a different tumorigenic pathway than tumors that develop spontaneously. Therefore, we propose that dysfunctional MMR promotes UVB-induced tumorigenesis through reduced apoptotic elimination of damaged epidermal cells.


Asunto(s)
Apoptosis/efectos de la radiación , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/fisiología , Epidermis/efectos de la radiación , Neoplasias Inducidas por Radiación/patología , Proteínas Proto-Oncogénicas/fisiología , Neoplasias Cutáneas/patología , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Aductos de ADN , Proteínas de Unión al ADN/genética , Células Epidérmicas , Epidermis/fisiología , Humanos , Ratones , Ratones Noqueados , Repeticiones de Microsatélite , Proteína 2 Homóloga a MutS , Invasividad Neoplásica/patología , Neoplasias Inducidas por Radiación/metabolismo , Proteínas Proto-Oncogénicas/genética , Dímeros de Pirimidina , Neoplasias Cutáneas/metabolismo , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA