Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Oncology (Williston Park) ; 36(2): 97-106, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35180337

RESUMEN

Follicular lymphoma (FL) is the most common indolent lymphoma and is characterized by a relapsing and remitting course. In addition to significant biologic heterogeneity, the clinical trajectory for patients is variable, with some being observed for many years, and others having aggressive disease requiring multiple treatment courses. Unfortunately, FL remains incurable, and continues to cause early mortality. Improved understanding of the genetic and immune biology of FL has led to several FDA-approved therapies in the relapsed and refractory setting, including PI3K inhibitors; immunomodulatory agents; the EZH2 inhibitor, tazemetostat; and anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, axicabtagene ciloleucel. This review outlines the current approach to the diagnosis and treatment of FL with a focus on emerging investigational therapies, including targeted protein inhibitors, antibody-drug conjugates, monoclonal antibodies, bispecific antibodies, and novel combination strategies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfoma Folicular/diagnóstico , Linfoma Folicular/terapia , Recurrencia Local de Neoplasia/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Clorhidrato de Bendamustina/administración & dosificación , Benzamidas/administración & dosificación , Compuestos de Bifenilo/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Ciclofosfamida/administración & dosificación , Doxorrubicina/administración & dosificación , Pruebas Genéticas , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inmunoconjugados/administración & dosificación , Inmunoterapia Adoptiva , Lenalidomida/administración & dosificación , Linfoma Folicular/genética , Linfoma Folicular/patología , Quimioterapia de Mantención , Morfolinas/administración & dosificación , Inhibidores de las Quinasa Fosfoinosítidos-3/administración & dosificación , Prednisona/administración & dosificación , Piridonas/administración & dosificación , Rituximab/administración & dosificación , Sulfonamidas/administración & dosificación , Vincristina/administración & dosificación
2.
Clin Orthop Relat Res ; 473(10): 3212-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26139622

RESUMEN

BACKGROUND: In patients with rotator cuff dysfunction, reverse shoulder arthroplasty can restore active forward flexion, but it does not provide a solution for the lack of active external rotation because of infraspinatus and the teres minor dysfunction. A modified L'Episcopo procedure can be performed in the same setting wherein the latissimus dorsi and teres major tendons are transferred to the lateral aspect of proximal humerus in an attempt to restore active external rotation. QUESTIONS/PURPOSES: (1) Do latissimus dorsi and teres major tendon transfers with reverse shoulder arthroplasty improve external rotation function in patients with posterosuperior rotator cuff dysfunction? (2) Do patients experience less pain and have improved outcome scores after surgery? (3) What are the complications associated with reverse shoulder arthroplasty with latissimus dorsi and teres major transfer? METHODS: Between 2007 and 2010, we treated all patients undergoing shoulder arthroplasty who had a profound external rotation lag sign and advanced fatty degeneration of the posterosuperior rotator cuff (infraspinatus plus teres minor) with this approach. A total of 21 patients (mean age 66 years; range, 58-82 years) were treated this way and followed for a minimum of 2 years (range, 26-81 months); none was lost to followup, and all have been seen in the last 5 years. We compared pre- and postoperative ranges of motion, pain, and functional status; scores were drawn from chart review. We also categorized major and minor complications. RESULTS: Active forward flexion improved from 56° ± 36° to 120° ± 38° (mean difference: 64° [95% confidence interval {CI}, 45°-83°], p < 0.001). Active external rotation with the arm adducted improved from 6° ± 16° to 38° ± 14° (mean difference: 30° [95% CI, 21°-39°], p < 0.001); active external rotation with the arm abducted improved from 19° ± 25° to 74° ± 22° (mean difference: 44° [95% CI, 22°-65°], p < 0.001). Pain visual analog score improved from 8.4 ± 2.3 to 1.7 ± 2.1 (mean difference: -6.9 [95% CI, -8.7 to -5.2], p < 0.001), and Single Assessment Numeric Evaluation score improved from 28% ± 21% to 80% ± 24% (mean difference: 46% [95% CI, 28%-64%], p < 0.001). There were six major complications, five of which were treated operatively. Overall, three patients' latissimus and teres major transfer failed based on persistent lack of external rotation. CONCLUSIONS: In patients with posterior and superior cuff deficiency, reverse shoulder arthroplasty combined with latissimus dorsi and teres major transfer through a single deltopectoral incision can reliably increase active forward flexion and external rotation. Patients experience pain relief and functional improvement but have a high rate of complications; therefore, we recommend the procedure be limited to patients indicated for reverse who have profound external rotation loss and a high grade of infraspinatus/teres minor fatty atrophy. LEVEL OF EVIDENCE: Level IV, therapeutic study.


Asunto(s)
Artroplastia/métodos , Artropatías/cirugía , Manejo del Dolor , Manguito de los Rotadores , Transferencia Tendinosa , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Artropatías/fisiopatología , Masculino , Persona de Mediana Edad , Rango del Movimiento Articular , Recuperación de la Función , Estudios Retrospectivos , Manguito de los Rotadores/fisiopatología
3.
Nanomedicine ; 10(1): 149-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23891990

RESUMEN

A major obstacle to the management of malignant glioma is the inability to effectively deliver therapeutic agent to the tumor. In this study, we describe a polymeric nanoparticle vector that not only delivers viable therapeutic, but can also be tracked in vivo using MRI. Nanoparticles, produced by a non-emulsion technique, were fabricated to carry iron oxide within the shell and the chemotherapeutic agent, temozolomide (TMZ), as the payload. Nanoparticle properties were characterized and subsequently their endocytosis-mediated uptake by glioma cells was demonstrated. Convection-enhanced delivery (CED) can disperse nanoparticles through the rodent brain and their distribution is accurately visualized by MRI. Infusion of nanoparticles does not result in observable animal toxicity relative to control. CED of TMZ-bearing nanoparticles prolongs the survival of animals with intracranial xenografts compared to control. In conclusion, the described nanoparticle vector represents a unique multifunctional platform that can be used for image-guided treatment of malignant glioma. FROM THE CLINICAL EDITOR: GBM remains one of the most notoriously treatment-unresponsive cancer types. In this study, a multifunctional nanoparticle-based temozolomide delivery system was demonstrated to possess enhanced treatment efficacy in a rodent xenograft GBM model, with the added benefit of MRI-based tracking via the incorporation of iron oxide as a T2* contrast material in the nanoparticles.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Sistemas de Liberación de Medicamentos , Glioma/tratamiento farmacológico , Nanopartículas/química , Animales , Neoplasias Encefálicas/diagnóstico por imagen , Línea Celular Tumoral , Convección , Dacarbazina/administración & dosificación , Dacarbazina/química , Compuestos Férricos/química , Glioma/diagnóstico por imagen , Glioma/patología , Humanos , Imagen por Resonancia Magnética , Ratones , Nanopartículas/uso terapéutico , Polímeros/química , Polímeros/uso terapéutico , Radiografía , Ratas , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Sci Rep ; 11(1): 5665, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707466

RESUMEN

The alkylating agent, temozolomide (TMZ), is the most commonly used chemotherapeutic for the treatment of glioblastoma (GBM). The anti-glioma effect of TMZ involves a complex response that includes G2-M cell cycle arrest and cyclin-dependent kinase 1 (CDK1) activation. While CDK1 phosphorylation is a well-described consequence of TMZ treatment, we find that TMZ also robustly induces CDK1 expression. Analysis of this pathway demonstrates that CDK1 is regulated by NF-κB via a putative κB-site in its proximal promoter. CDK1 was induced in a manner dependent on mature p50 and the atypical inhibitor κB protein, BCL-3. Treatment with TMZ induced binding of NF-κB to the κB-site as assessed by gel shift analysis and chromatin immunoprecipitation. Examination of a CDK1 promoter-reporter demonstrated the functional relevance of the κB-site and underlined the requirement of p50 and BCL-3 for activation. Targeted knockdown of CDK1 or chemical inhibition with the selective CDK1 inhibitor, RO-3306, potentiated the cytotoxic effect of TMZ. These results identify CDK1 as an NF-κB target gene regulated by p50 and BCL-3 and suggest that targeting CDK1 may be a strategy to improve the efficacy of TMZ against GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteína Quinasa CDC2/metabolismo , Glioblastoma/metabolismo , FN-kappa B/metabolismo , Temozolomida/farmacología , Proteínas del Linfoma 3 de Células B/metabolismo , Secuencia de Bases , Sitios de Unión , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteína Quinasa CDC2/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Regiones Promotoras Genéticas/genética
5.
Leuk Lymphoma ; 61(4): 799-807, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31750749

RESUMEN

Relapsed or refractory non-Hodgkin B-cell lymphoma is an aggressive disease with a dismal prognosis. Chimeric antigen receptor (CAR) T-cell therapy has emerged as a viable treatment option with durable remissions observed in clinical trials. Due to the risk of toxicities such as cytokine release syndrome and neurotoxicity, careful patient selection is critical to optimize outcomes. Narrow selection criteria were used in clinical trials that led to approval, but a wider range of patients has been successfully treated in the commercial setting. Due to lack of validated pretreatment clinical factors, including risk scores or biomarkers to predict efficacy and toxicity, choosing candidates for CAR T-cell therapy currently relies on expert opinion. Because of logistical constraints and often aggressive disease, we favor referring patients early for cellular therapy at the time of first treatment failure. Herein, we discuss criteria for patient selection using a case-based approach informed by reports of real-world outcomes.


Asunto(s)
Antígenos CD19 , Inmunoterapia Adoptiva , Humanos , Selección de Paciente , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T
6.
Blood Adv ; 4(4): 599-606, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32074275

RESUMEN

In this phase 1 study, azacitidine (AZA) was given before high-dose cytarabine (HiDAC) and mitoxantrone (mito) based on the hypothesis that epigenetic priming with a hypomethylating agent before cytotoxic chemotherapy would improve response rates in patients with high-risk acute myeloid leukemia (AML), including relapsed/refractory disease. The primary objective was to establish the recommended phase 2 dose of AZA given before standard HiDAC/mito. In a dose escalation scheme, 46 patients (median age, 66 years) received AZA at 37.5, 50, or 75 mg/m2 subcutaneously or IV once daily on days 1 to 5 followed by HiDAC (3000 mg/m2) and mitoxantrone (30 mg/m2) once each on days 6 and 10 (the HiDAC/mito dose was reduced 33% in elderly subjects). Two dose-limiting toxicities occurred (both in the same patient): acute liver failure and kidney injury at the 50 mg/m2 dose. The 30-day induction death rate was 2.2% (1 of 46). The overall response rate, including complete remission and complete remission with incomplete count recovery, was 61% (28 of 46). Previously untreated patients aged ≥60 years with therapy-related AML and de novo AML were more likely to respond than untreated patients with AML progressing from an antecedent hematologic disorder (myelodysplastic syndrome and chronic myelomonocytic leukemia). Patients with favorable European Leukemia Network risk (P = .008), NPM1 mutations (P = .007), or IDH2 mutations (P = .03) were more likely to respond, and those with TP53 mutations (P = .03) were less likely to respond. The recommended phase 2 dose of AZA is 75 mg/m2 per day on days 1 to 5 followed by HiDAC (3000 mg/m2) and mitoxantrone (30 mg/m2) once each on days 6 and 10. This trial was registered at www.clinicaltrials.gov as #NCT01839240.


Asunto(s)
Leucemia Mieloide Aguda , Mitoxantrona , Anciano , Azacitidina/efectos adversos , Citarabina/efectos adversos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Nucleofosmina , Inducción de Remisión
7.
Cancer Res ; 79(10): 2536-2548, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30940658

RESUMEN

Alkylating chemotherapy is a central component of the management of glioblastoma (GBM). Among the factors that regulate the response to alkylation damage, NF-κB acts to both promote and block cytotoxicity. In this study, we used genome-wide expression analysis in U87 GBM to identify NF-κB-dependent factors altered in response to temozolomide and found the long noncoding RNA (lncRNA) MALAT1 as one of the most significantly upregulated. In addition, we demonstrated that MALAT1 expression was coregulated by p50 (p105) and p53 via novel κB- and p53-binding sites in the proximal MALAT1 coding region. Temozolomide treatment inhibited p50 recruitment to its cognate element as a function of Ser329 phosphorylation while concomitantly increasing p53 recruitment. Moreover, luciferase reporter studies demonstrated that both κB and p53 cis-elements were required for efficient transactivation in response to temozolomide. Depletion of MALAT1 sensitized patient-derived GBM cells to temozolomide cytotoxicity, and in vivo delivery of nanoparticle-encapsulated anti-MALAT1 siRNA increased the efficacy of temozolomide in mice bearing intracranial GBM xenografts. Despite these observations, in situ hybridization of GBM specimens and analysis of publicly available datasets revealed that MALAT1 expression within GBM tissue was not prognostic of overall survival. Together, these findings support MALAT1 as a target for chemosensitization of GBM and identify p50 and p52 as primary regulators of this ncRNA. SIGNIFICANCE: These findings identify NF-κB and p53 as regulators of the lncRNA MALAT1 and suggest MALAT1 as a potential target for the chemosensitization of GBM.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/metabolismo , Glioblastoma/tratamiento farmacológico , FN-kappa B/metabolismo , ARN Largo no Codificante/biosíntesis , Temozolomida/uso terapéutico , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Daño del ADN/genética , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Pronóstico , ARN Largo no Codificante/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Sci Transl Med ; 10(448)2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973405

RESUMEN

The response of patients with gliomas to alkylating chemotherapy is heterogeneous. However, there are currently no universally accepted predictors of patient response to these agents. We identify the nuclear factor κB (NF-κB) co-regulator B cell CLL/lymphoma 3 (BCL-3) as an independent predictor of response to temozolomide (TMZ) treatment. In glioma patients with tumors that have a methylated O6-methylguanine DNA methyltransferase (MGMT) promoter, high BCL-3 expression was associated with a poor response to TMZ. Mechanistically, BCL-3 promoted a more malignant phenotype by inducing an epithelial-to-mesenchymal transition in glioblastomas through promoter-specific NF-κB dimer exchange. Carbonic anhydrase II (CAII) was identified as a downstream factor promoting BCL-3-mediated resistance to chemotherapy. Experiments in glioma xenograft mouse models demonstrated that the CAII inhibitor acetazolamide enhanced survival of TMZ-treated animals. Our data suggest that BCL-3 might be a useful indicator of glioma response to alkylating chemotherapy and that acetazolamide might be repurposed as a chemosensitizer for treating TMZ-resistant gliomas.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Glioma/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Antineoplásicos Alquilantes/farmacología , Proteínas del Linfoma 3 de Células B , Anhidrasa Carbónica II/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Análisis Multivariante , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Modelos de Riesgos Proporcionales , Multimerización de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Análisis de Supervivencia , Temozolomida/farmacología , Temozolomida/uso terapéutico , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Neuro Oncol ; 18(3): 329-39, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26534766

RESUMEN

Nuclear factor-κB (NF-κB) is a ubiquitous transcription factor that regulates multiple aspects of cancer formation, growth, and treatment response. Glioblastoma (GBM), the most common primary malignant tumor of the central nervous system, is characterized by molecular heterogeneity, resistance to therapy, and high NF-κB activity. In this review, we examine the mechanisms by which oncogenic pathways active in GBM impinge on the NF-κB system, discuss the role of NF-κB signaling in regulating the phenotypic properties that promote GBM and, finally, review the components of the NF-κB pathway that have been targeted for treatment in both preclinical studies and clinical trials. While a direct role for NF-κB in gliomagenesis has not been reported, the importance of this transcription factor in the overall malignant phenotype suggests that more rational and specific targeting of NF-κB-dependent pathways can make a significant contribution to the management of GBM.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Animales , Neoplasias Encefálicas/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Glioblastoma/patología , Humanos
10.
Cancer Res ; 75(10): 2039-48, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25808868

RESUMEN

Temozolomide is used widely to treat malignant glioma, but the overall response to this agent is generally poor. Resistance to DNA-damaging drugs such as temozolomide has been related to the induction of antiapoptotic proteins. Specifically, the transcription factor NF-κB has been suggested to participate in promoting the survival of cells exposed to chemotherapy. To identify factors that modulate cytotoxicity in the setting of DNA damage, we used an unbiased strategy to examine the NF-κB-dependent expression profile induced by temozolomide. By this route, we defined the decoy receptor DcR1 as a temozolomide response gene induced by a mechanism relying upon p50/NF-κB1. A conserved NF-κB-binding sequence (κB-site) was identified in the proximal promoter and was demonstrated to be required for DcR1 induction by temozolomide. Loss-of-function and gain-of-function studies reveal that the atypical IκB protein, Bcl3, is also required for induction of DcR1 by temozolomide. Mechanistically, DcR1 attenuates temozolomide efficacy by blunting activation of the Fas receptor pathway in p53(+/+) glioma cells. Intracranial xenograft studies show that DcR1 depletion in glioma cells enhances the efficacy of temozolomide. Taken together, our results show how DcR1 upregulation mediates temozolomide resistance and provide a rationale for DcR1 targeting as a strategy to sensitize gliomas to this widely used chemotherapy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Subunidad p50 de NF-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Animales , Proteínas del Linfoma 3 de Células B , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Dacarbazina/farmacología , Resistencia a Antineoplásicos , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Masculino , Ratones Desnudos , Regiones Promotoras Genéticas , Unión Proteica , Miembro 10c de Receptores del Factor de Necrosis Tumoral , Temozolomida , Activación Transcripcional , Receptores Señuelo del Factor de Necrosis Tumoral/química , Receptores Señuelo del Factor de Necrosis Tumoral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cell Cycle ; 14(4): 566-76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25590437

RESUMEN

The apical damage kinase, ATR, is activated by replication stress (RS) both in response to DNA damage and during normal S-phase. Loss of function studies indicates that ATR acts to stabilize replication forks, block cell cycle progression and promote replication restart. Although checkpoint failure and replication fork collapse can result in cell death, no direct cytotoxic pathway downstream of ATR has previously been described. Here, we show that ATR directly reduces survival by inducing phosphorylation of the p50 (NF-κB1, p105) subunit of NF-кB and moreover, that this response is necessary for genome maintenance independent of checkpoint activity. Cell free and in vivo studies demonstrate that RS induces phosphorylation of p50 in an ATR-dependent but DNA damage-independent manner that acts to modulate NF-кB activity without affecting p50/p65 nuclear translocation. This response, evident in human and murine cells, occurs not only in response to exogenous RS but also during the unperturbed S-phase. Functionally, the p50 response results in inhibition of anti-apoptotic gene expression that acts to sensitize cells to DNA strand breaks independent of damage repair. Ultimately, loss of this pathway causes genomic instability due to the accumulation of chromosomal breaks. Together, the data indicate that during S-phase ATR acts via p50 to ensure that cells with elevated levels of replication-associated DNA damage are eliminated.


Asunto(s)
Replicación del ADN/fisiología , Inestabilidad Genómica/fisiología , Subunidad p50 de NF-kappa B/metabolismo , Fase S/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Ensayo Cometa , Cartilla de ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Inestabilidad Genómica/genética , Humanos , Immunoblotting , Inmunoprecipitación , Luciferasas , Fosforilación , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Aging (Albany NY) ; 6(11): 931-43, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25553648

RESUMEN

NF-κB is a major regulator of age-dependent gene expression and the p50/NF-κB1 subunit is an integral modulator of NF-κB signaling. Here, we examined Nfkb1-/- mice to investigate the relationship between this subunit and aging. Although Nfkb1-/- mice appear similar to littermates at six months of age, by 12 months they have a higher incidence of several observable age-related phenotypes. In addition, aged Nfkb1-/- animals have increased kyphosis, decreased cortical bone, increased brain GFAP staining and a decrease in overall lifespan compared to Nfkb1+/+. In vitro, serially passaged primary Nfkb1-/- MEFs have more senescent cells than comparable Nfkb1+/+ MEFs. Also, Nfkb1-/- MEFs have greater amounts of phospho-H2AX foci and lower levels of spontaneous apoptosis than Nfkb1+/+, findings that are mirrored in the brains of Nfkb1-/- animals compared to Nfkb1+/+. Finally, in wildtype animals a substantial decrease in p50 DNA binding is seen in aged tissue compared to young. Together, these data show that loss of Nfkb1 leads to early animal aging that is associated with reduced apoptosis and increased cellular senescence. Moreover, loss of p50 DNA binding is a prominent feature of aged mice relative to young. These findings support the strong link between the NF-κB pathway and mammalian aging.


Asunto(s)
Envejecimiento Prematuro , Envejecimiento/metabolismo , Subunidad p50 de NF-kappa B/deficiencia , Factores de Edad , Envejecimiento/genética , Envejecimiento/patología , Animales , Apoptosis , Huesos/metabolismo , Huesos/patología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Senescencia Celular , ADN/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Genotipo , Gliosis , Histonas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Cifosis/genética , Cifosis/metabolismo , Cifosis/patología , Longevidad , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p50 de NF-kappa B/genética , Fenotipo , Fosforilación , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA