Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 34(2): 2126-2146, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31909533

RESUMEN

Bladder dysfunction is associated with the overexpression of the intermediate filament (IF) proteins desmin and vimentin in obstructed bladder smooth muscle (BSM). However, the mechanisms by which these proteins contribute to BSM dysfunction are not known. Previous studies have shown that desmin and vimentin directly participate in signal transduction. In this study, we hypothesized that BSM dysfunction associated with overexpression of desmin or vimentin is mediated via c-Jun N-terminal kinase (JNK). We employed a model of murine BSM tissue in which increased expression of desmin or vimentin was induced by adenoviral transduction to examine the sufficiency of increased IF protein expression to reduce BSM contraction. Murine BSM strips overexpressing desmin or vimentin generated less force in response to KCl and carbachol relative to the levels in control murine BSM strips, an effect associated with increased JNK2 phosphorylation and reduced myosin light chain (MLC20 ) phosphorylation. Furthermore, desmin and vimentin overexpressions did not alter BSM contractility and MLC20 phosphorylation in strips isolated from JNK2 knockout mice. Pharmacological JNK2 inhibition produced results qualitatively similar to those caused by JNK2 knockout. These findings suggest that inhibition of JNK2 may improve diminished BSM contractility associated with obstructive bladder disease.


Asunto(s)
Desmina/biosíntesis , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Contracción Muscular , Músculo Liso/metabolismo , Vejiga Urinaria/metabolismo , Vimentina/biosíntesis , Animales , Desmina/genética , Ratones , Ratones Noqueados , Proteína Quinasa 9 Activada por Mitógenos/genética , Músculo Liso/citología , Vejiga Urinaria/citología , Vimentina/genética
2.
Int J Mol Sci ; 22(18)2021 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-34576287

RESUMEN

Advancements in cancer therapy increased the cancer free survival rates and reduced the malignant related deaths. Therapeutic options for patients with thoracic cancers include surgical intervention and the application of chemotherapy with ionizing radiation. Despite these advances, cancer therapy-related cardiopulmonary dysfunction (CTRCPD) is one of the most undesirable side effects of cancer therapy and leads to limitations to cancer treatment. Chemoradiation therapy or immunotherapy promote acute and chronic cardiopulmonary damage by inducing reactive oxygen species, DNA damage, inflammation, fibrosis, deregulation of cellular immunity, cardiopulmonary failure, and non-malignant related deaths among cancer-free patients who received cancer therapy. CTRCPD is a complex entity with multiple factors involved in this pathogenesis. Although the mechanisms of cancer therapy-induced toxicities are multifactorial, damage to the cardiac and pulmonary tissue as well as subsequent fibrosis and organ failure seem to be the underlying events. The available biomarkers and treatment options are not sufficient and efficient to detect cancer therapy-induced early asymptomatic cell fate cardiopulmonary toxicity. Therefore, application of cutting-edge multi-omics technology, such us whole-exome sequencing, DNA methylation, whole-genome sequencing, metabolomics, protein mass spectrometry and single cell transcriptomics, and 10 X spatial genomics, are warranted to identify early and late toxicity, inflammation-induced carcinogenesis response biomarkers, and cancer relapse response biomarkers. In this review, we summarize the current state of knowledge on cancer therapy-induced cardiopulmonary complications and our current understanding of the pathological and molecular consequences of cancer therapy-induced cardiopulmonary fibrosis, inflammation, immune suppression, and tumor recurrence, and possible treatment options for cancer therapy-induced cardiopulmonary toxicity.


Asunto(s)
Inmunomodulación , Inflamación/etiología , Pulmón/patología , Miocardio/patología , Neoplasias/terapia , Animales , Fibrosis , Humanos
3.
Am J Pathol ; 189(4): 847-867, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30707892

RESUMEN

Caveolins (CAVs) are structural proteins of caveolae that function as signaling platforms to regulate smooth muscle contraction. Loss of CAV protein expression is associated with impaired contraction in obstruction-induced bladder smooth muscle (BSM) hypertrophy. In this study, microarray analysis of bladder RNA revealed down-regulation of CAV1, CAV2, and CAV3 gene transcription in BSM from models of obstructive bladder disease in mice and humans. We identified and characterized regulatory regions responsible for CAV1, CAV2, and CAV3 gene expression in mice with obstruction-induced BSM hypertrophy, and in men with benign prostatic hyperplasia. DNA affinity chromatography and chromatin immunoprecipitation assays revealed a greater increase in binding of GATA-binding factor 6 (GATA-6) and NF-κB to their cognate binding motifs on CAV1, CAV2, and CAV3 promoters in obstructed BSM relative to that observed in control BSM. Knockout of NF-κB subunits, shRNA-mediated knockdown of GATA-6, or pharmacologic inhibition of GATA-6 and NF-κB in BSM increased CAV1, CAV2, and CAV3 transcription and promoter activity. Conversely, overexpression of GATA-6 decreased CAV2 and CAV3 transcription and promoter activity. Collectively, these data provide new insight into the mechanisms by which CAV gene expression is repressed in hypertrophied BSM in obstructive bladder disease.


Asunto(s)
Caveolinas/antagonistas & inhibidores , Factor de Transcripción GATA6/metabolismo , Hipertrofia/patología , Músculo Liso/patología , FN-kappa B/metabolismo , Transcripción Genética , Obstrucción del Cuello de la Vejiga Urinaria/complicaciones , Anciano , Animales , Biomarcadores/análisis , Caveolinas/genética , Caveolinas/metabolismo , Factor de Transcripción GATA6/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Hipertrofia/etiología , Hipertrofia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Contracción Muscular , Músculo Liso/metabolismo , FN-kappa B/genética , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Obstrucción del Cuello de la Vejiga Urinaria/cirugía
4.
Mol Cell ; 43(4): 663-72, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21855804

RESUMEN

The LXCXE peptide motif facilitates interaction between the RB tumor suppressor and a large number of cellular proteins that are expected to impinge on diverse biological processes. In vitro and in vivo analyses demonstrated that LXCXE binding function is dispensable for RB promoter association and control of basal gene expression. Dependence on this function of RB is unmasked after DNA damage, wherein LXCXE binding is essential for exerting control over E2F3 and suppressing cell-cycle progression in the presence of genotoxic stress. Gene expression profiling revealed that the transcriptional program coordinated by this specific aspect of RB is associated with progression of human hepatocellular carcinoma and poor disease outcome. Consistent with these findings, biological challenge revealed a requirement for LXCXE binding in suppression of genotoxin-initiated hepatocellular carcinoma in vivo. Together, these studies establish an essential role of the LXCXE binding motif for RB-mediated transcriptional control, response to genotoxic insult, and tumor suppression.


Asunto(s)
Daño del ADN , Regulación de la Expresión Génica , Proteína de Retinoblastoma/fisiología , Transcripción Genética , Secuencias de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Cromatina/metabolismo , Humanos , Ratones , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/metabolismo
5.
Mol Pharm ; 15(5): 1778-1790, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29616555

RESUMEN

Second generation antiandrogens have improved overall survival for men with metastatic castrate resistant prostate cancer; however, the antiandrogens result in suppression of androgen receptor (AR) activity in all tissues resulting in dose limiting toxicity. We sought to overcome this limitation through encapsulation in a prostate specific membrane antigen (PSMA)-conjugated nanoparticle. We designed and characterized a novel nanoparticle containing an antiandrogen, enzalutamide. Selectivity and enhanced efficacy was achieved through coating the particle with PSMA. The PSMA-conjugated nanoparticle was internalized selectively in AR expressing prostate cancer cells. It did not elicit an inflammatory effect. The efficacy of enzalutamide was not compromised through insertion into the nanoparticle; in fact, lower systemic drug concentrations of enzalutamide resulted in comparable clinical activity. Normal muscle cells were not impacted by the PSMA-conjugated containing antiandrogen. This approach represents a novel strategy to increase the specificity and effectiveness of antiandrogen treatment for men with castrate resistant prostate cancer. The ability to deliver higher drug concentrations in prostate cancer cells may translate into improved clinical end points including overall survival.


Asunto(s)
Antagonistas de Andrógenos/química , Antagonistas de Andrógenos/farmacología , Nanopartículas/química , Antígeno Prostático Específico/metabolismo , Receptores Androgénicos/metabolismo , Antígenos de Superficie/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Células MCF-7 , Masculino , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 310(7): G449-59, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26822916

RESUMEN

MicroRNAs (miRNAs) belong to a group of short noncoding RNA molecules with important roles in cellular biology. miRNAs regulate gene expression by repressing translation or degrading the target mRNA. Recently, a growing body of evidence suggests that miRNAs are implicated in many diseases and could be potential biomarkers. Fibrosis and/smooth muscle (SM) dysfunction contributes to the morbidity and mortality associated with several diseases of the gastrointestinal tract (GIT). Currently available therapeutic modalities are unsuccessful in efficiently blocking or reversing fibrosis and/or SM dysfunction. Recent understanding of the role of miRNAs in signaling pathway of fibrogenesis and SM phenotype switch has provided a new insight into translational research. However, much is still unknown about the molecular targets and therapeutic potential of miRNAs in the GIT. This review discusses miRNA biology, pathophysiology of fibrosis, and aging- associated SM dysfunction in relation to the deregulation of miRNAs in the GIT. We also highlight the role of selected miRNAs associated with fibrosis and SM dysfunction-related diseases of the GIT.


Asunto(s)
Enfermedades Gastrointestinales/metabolismo , Tracto Gastrointestinal/metabolismo , MicroARNs/metabolismo , Músculo Liso/metabolismo , Factores de Edad , Animales , Epigénesis Genética , Fibrosis , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/fisiopatología , Enfermedades Gastrointestinales/terapia , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/fisiopatología , Regulación de la Expresión Génica , Marcadores Genéticos , Terapia Genética/métodos , Humanos , MicroARNs/genética , MicroARNs/uso terapéutico , Músculo Liso/patología , Músculo Liso/fisiopatología , Estrés Oxidativo , Fenotipo , Transducción de Señal
7.
Mol Carcinog ; 55(5): 743-56, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25968914

RESUMEN

Triple negative breast cancer (TNBC) is characterized by a lack in estrogen, progesterone, and epidermal growth factor 2 receptors. TNBC exhibits most of the characteristics of basal-like and claudin-low breast cancer subtypes. The main contributor in the mortality of TNBC is due to the higher invasive and migratory ability of these tumor cells. Some plant flavonoids inhibit the epithelial mesenchymal transition (EMT) of tumor cells and suppress cancer metastasis. In this study, we aimed to determine whether the flavonoid quercetin is effective in modulating the molecular signaling associated with EMT in TNBC. Our data indicated that quercetin can induce the expression of E-cadherin and also downregulate vimentin levels in TNBC. The ability of quercetin to modulate these EMT markers resulted in a mesenchymal-to-epithelial transition (MET). Quercetin-induced MET was linked with the alteration of nuclear localization of ß-catenin and modulation of ß-catenin target genes such as cyclin D1 and c-Myc. Furthermore, we observed that quercetin induced the anti-tumor activity of doxorubicin by inhibiting the migratory ability of TNBC cells. These results suggested that quercetin may inhibit TNBC metastasis and also improve the therapeutic efficacy of existing chemotherapeutic drugs.


Asunto(s)
Antioxidantes/farmacología , Quercetina/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , beta Catenina/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología
8.
J Nanobiotechnology ; 13: 61, 2015 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-26410728

RESUMEN

BACKGROUND: siRNAs have a high potential for silencing critical molecular pathways that are pathogenic. Nevertheless, their clinical application has been limited by a lack of effective and safe nanotechnology-based delivery system that allows a controlled and safe transfection to cytosol of targeted cells without the associated adverse effects. Our group recently reported a very effective and safe hybrid nanoparticle delivery system composing human IgG and poloxamer-188 for siRNA delivery to cancer cells. However, these nanoparticles need to be optimized in terms of particle size, loading capacity and encapsulation efficiency. In the present study, we explored the effects of certain production parameters on particle size, loading capacity and encapsulation efficiency. Further, to make these nanoparticles more specific in their delivery of siRNA, we conjugated anti-NTSR1-mAb to the surface of these nanoparticles to target NTSR1-overexpressing cancer cells. The mechanism of siRNA release from these antiNTSR1-mAb functionalized nanoparticles was also elucidated. RESULTS: It was demonstrated that the concentration of human IgG in the starting nanoprecipitation medium and the rotation speed of the magnetic stirrer influenced the encapsulation efficiency, loading capacity and the size of the nanoparticles produced. We also successfully transformed these nanoparticles into actively targeted nanoparticles by functionalizing with anti-NTSR1-mAb to specifically target NTSR1-overexpressing cancer cells, hence able to avoid undesired accumulation in normal cells. The mechanism of siRNA release from these nanoparticles was elucidated to be by Fickian diffusion. Using flow cytometry and fluorescence microscopy, we were able to confirm the active involvement of NTSR1 in the uptake of these anti-NTSR1-mAb functionalized hybrid nanoparticles by lung adenocarcinoma cells. CONCLUSIONS: This hybrid nanoparticle delivery system can be used as a platform technology for intracellular delivery of siRNAs to NTSR1-overexpressing tumor cells.


Asunto(s)
Nanopartículas/química , ARN Interferente Pequeño/metabolismo , Anticuerpos Monoclonales/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Endocitosis , Humanos , Cinética , Neoplasias Pulmonares/patología , Microscopía Fluorescente , Nanopartículas/ultraestructura , Tamaño de la Partícula , Receptores de Neurotensina/inmunología , Espectroscopía Infrarroja por Transformada de Fourier , Electricidad Estática
9.
Cancers (Basel) ; 15(15)2023 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-37568788

RESUMEN

Radiation therapy (RT) remains an integral component of modern oncology care, with most cancer patients receiving radiation as a part of their treatment plan. The main goal of ionizing RT is to control the local tumor burden by inducing DNA damage and apoptosis within the tumor cells. The advancement in RT, including intensity-modulated RT (IMRT), stereotactic body RT (SBRT), image-guided RT, and proton therapy, have increased the efficacy of RT, equipping clinicians with techniques to ensure precise and safe administration of radiation doses to tumor cells. In this review, we present the technological advancement in various types of RT methods and highlight their clinical utility and associated limitations. This review provides insights into how RT modulates innate immune signaling and the key players involved in modulating innate immune responses, which have not been well documented earlier. Apoptosis of cancer cells following RT triggers immune systems that contribute to the eradication of tumors through innate and adoptive immunity. The innate immune system consists of various cell types, including macrophages, dendritic cells, and natural killer cells, which serve as key mediators of innate immunity in response to RT. This review will concentrate on the significance of the innate myeloid and lymphoid lineages in anti-tumorigenic processes triggered by RT. Furthermore, we will explore essential strategies to enhance RT efficacy. This review can serve as a platform for researchers to comprehend the clinical application and limitations of various RT methods and provides insights into how RT modulates innate immune signaling.

10.
Colloids Surf B Biointerfaces ; 209(Pt 1): 112162, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34752986

RESUMEN

The use of multifunctional nanomedicines for image-guided drug delivery is currently being universally evaluated as a means of efficiently managing cancers and other diseases. In this study we evaluated the potential of an indocyanine green (ICG) and paclitaxel (PTX) loaded human serum albumin (HSA) nanoparticles that was conjugated with hyaluronic acid for use in image-guided drug delivery targeted to CD44-positive non-small cell lung cancer (NSCLC). Series of NSCLC cell lines were evaluated for the expression of CD44 using both western blot analysis and qRT-PCR and compared to a normal lung fibroblast cell line (MRC-5). Using Fluorescence microscopy and photoacoustic imaging (PA), we explored the ability of these targeted nanoparticles to selectively accumulate in NSCLC cell lines in comparison to MRC-5 and their potential for biomedical imaging towards their use for theranostic application. Results obtained suggest that these targeted nanoparticles have potential for application in both imaging and treatment of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Nanopartículas , Preparaciones Farmacéuticas , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Humanos , Receptores de Hialuranos , Verde de Indocianina , Neoplasias Pulmonares/tratamiento farmacológico , Paclitaxel/farmacología , Albúmina Sérica Humana
11.
Cancers (Basel) ; 14(17)2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36077840

RESUMEN

Prostate cancer (PCa) is the second-leading cause of cancer-related deaths in men. PCa cells require androgen receptor (AR) signaling for their growth and survival. Androgen deprivation therapy (ADT) is the preferred treatment for patients with locally advanced and metastatic PCa disease. Despite their initial response to androgen blockade, most patients eventually will develop metastatic castration-resistant prostate cancer (mCRPC). Bone metastases are common in men with mCRPC, occurring in 30% of patients within 2 years of castration resistance and in >90% of patients over the course of the disease. Patients with mCRPC-induced bone metastasis develop lesions throughout their skeleton; the 5-year survival rate for these patients is 47%. Bone-metastasis-induced early changes in the bone that proceed the osteoblastic response in the bone matrix are monitored and detected via modern magnetic resonance and PET/CT imaging technologies. Various treatment options, such as targeting osteolytic metastasis with bisphosphonates, prednisone, dexamethasone, denosumab, immunotherapy, external beam radiation therapy, radiopharmaceuticals, surgery, and pain medications are employed to treat prostate-cancer-induced bone metastasis and manage bone health. However, these diagnostics and treatment options are not very accurate nor efficient enough to treat bone metastases and manage bone health. In this review, we present the pathogenesis of PCa-induced bone metastasis, its deleterious impacts on vital organs, the impact of metastatic PCa on bone health, treatment interventions for bone metastasis and management of bone- and skeletal-related events, and possible current and future therapeutic options for bone management in the continuum of prostate cancer disease.

12.
Gastroenterology ; 138(5): 1920-30, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20100483

RESUMEN

BACKGROUND & AIMS: Hepatocellular carcinoma is the third leading cause of cancer mortality worldwide; current chemotherapeutic interventions for this disease are largely ineffective. The retinoblastoma tumor suppressor (RB) is functionally inactivated at relatively high frequency in hepatocellular carcinoma and hepatoma cell lines. Here, we analyzed the ability of CDK4/6 inhibition to inhibit hepatocyte proliferation and the effect of RB status on this process. METHODS: Hepatoma cell lines and xenograft models harboring RB knockdown and mice harboring liver-specific Rb deletion were used to define the role of RB function in response to CDK4/6 inhibition. RESULTS: Our study shows that CDK4/6-dependent cell cycle progression in hepatoma cells was readily arrested by inhibition of CDK4/6 by PD-0332991 or p16ink4a irrespective of RB status. Interestingly, upon CDK4/6 inhibition, p107 protein stability was dramatically increased as a function of RB loss. This engagement of compensatory mechanisms was critical for cell cycle inhibition in the absence of RB, because both the E1A oncoprotein and overexpression of E2F proteins were capable of overcoming the effect of CDK4/6 inhibition. These findings were recapitulated in xenograft models. Furthermore, to determine how these findings relate to hepatocyte proliferation in vivo, mice were exposed to carbon tetrachloride to induce liver regeneration followed by treatment with PD-0332991. This treatment significantly inhibited hepatocyte proliferation. Strikingly, this facet of PD-0332991 function was retained even in RB-deficient livers. CONCLUSIONS: These data show that CDK4/6 inhibition is a potent mediator of cytostasis and that RB loss can be readily compensated for in the context of both hepatoma cell lines and liver tissue.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Proliferación Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Hepatocitos/efectos de los fármacos , Neoplasias Hepáticas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteína de Retinoblastoma/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Animales , Tetracloruro de Carbono , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción E2F/metabolismo , Técnicas de Silenciamiento del Gen , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Piperazinas/farmacología , Purinas/farmacología , Piridinas/farmacología , Interferencia de ARN , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Roscovitina , Factores de Tiempo , Transfección
13.
Cancer Discov ; 11(9): 2334-2353, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33879449

RESUMEN

Loss of the retinoblastoma (RB) tumor suppressor protein is a critical step in reprogramming biological networks that drive cancer progression, although mechanistic insight has been largely limited to the impact of RB loss on cell-cycle regulation. Here, isogenic modeling of RB loss identified disease stage-specific rewiring of E2F1 function, providing the first-in-field mapping of the E2F1 cistrome and transcriptome after RB loss across disease progression. Biochemical and functional assessment using both in vitro and in vivo models identified an unexpected, prominent role for E2F1 in regulation of redox metabolism after RB loss, driving an increase in the synthesis of the antioxidant glutathione, specific to advanced disease. These E2F1-dependent events resulted in protection from reactive oxygen species in response to therapeutic intervention. On balance, these findings reveal novel pathways through which RB loss promotes cancer progression and highlight potentially new nodes of intervention for treating RB-deficient cancers. SIGNIFICANCE: This study identifies stage-specific consequences of RB loss across cancer progression that have a direct impact on tumor response to clinically utilized therapeutics. The study herein is the first to investigate the effect of RB loss on global metabolic regulation and link RB/E2F1 to redox control in multiple advanced diseases.This article is highlighted in the In This Issue feature, p. 2113.


Asunto(s)
Factor de Transcripción E2F1/genética , Neoplasias de la Retina/genética , Proteína de Retinoblastoma/genética , Retinoblastoma/genética , Animales , Línea Celular Tumoral , Humanos , Ratones , Metástasis de la Neoplasia , Neoplasias de la Retina/patología , Retinoblastoma/secundario , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancers (Basel) ; 13(1)2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-33396766

RESUMEN

Immunotherapy is one of the most effective therapeutic options for cancer patients. Five specific classes of immunotherapies, which includes cell-based chimeric antigenic receptor T-cells, checkpoint inhibitors, cancer vaccines, antibody-based targeted therapies, and oncolytic viruses. Immunotherapies can improve survival rates among cancer patients. At the same time, however, they can cause inflammation and promote adverse cardiac immune modulation and cardiac failure among some cancer patients as late as five to ten years following immunotherapy. In this review, we discuss cardiotoxicity associated with immunotherapy. We also propose using human-induced pluripotent stem cell-derived cardiomyocytes/ cardiac-stromal progenitor cells and cardiac organoid cultures as innovative experimental model systems to (1) mimic clinical treatment, resulting in reproducible data, and (2) promote the identification of immunotherapy-induced biomarkers of both early and late cardiotoxicity. Finally, we introduce the integration of omics-derived high-volume data and cardiac biology as a pathway toward the discovery of new and efficient non-toxic immunotherapy.

15.
Drug Metab Dispos ; 36(9): 1884-95, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18559485

RESUMEN

Because of its myriad physiologic functions, it is not surprising that the actions of growth hormone (GH) are mediated by recruiting/activating dozens of signaling molecules involved in numerous transduction pathways. The particular signal transduction pathway activated by the hormone is determined by the affected target cell, the sexually dimorphic secretory GH profile (masculine episodic or feminine continuous) to which the cell is exposed, and the individual's sex. In this regard, expression of female-specific CYP2C12, the most abundant cytochrome P450 in female rat liver, is solely regulated by the feminine GH profile. Sex is a modulating factor in this response in that males are considerably less responsive than females to the CYP2C12-induction effects of continuous GH. Using primary hepatocytes derived from male and female hypophysectomized rats, we have identified several factors in a transduction pathway activated by the feminine GH regime and associated with the induction of hepatic CYP2C12. Elements in the proposed pathway, in their likely order of activation, are the growth hormone receptor, extracellular signal-regulated kinases, the cAMP-response element-binding protein, and hepatocyte nuclear factors 4alpha and 6, which subsequently bind and activate the CYP2C12 promoter. Recruitment and/or activation levels of all of the component factors in the pathway were highly suppressed in male hepatocytes, possibly explaining the dramatically lower induction levels of CYP2C12 in males exposed to the same continuous GH profile as females.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Hormona del Crecimiento/farmacología , Hígado/enzimología , Caracteres Sexuales , Transducción de Señal/efectos de los fármacos , Esteroide Hidroxilasas/metabolismo , Acetilación , Animales , Secuencia de Bases , Western Blotting , Cartilla de ADN , Femenino , Hormona del Crecimiento/administración & dosificación , Inmunoprecipitación , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Ratas
16.
Mol Endocrinol ; 21(7): 1537-51, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17488973

RESUMEN

GH receptor (GHR) is a cytokine receptor family member that responds to GH by activation of the receptor-associated tyrosine kinase, JAK2 (Janus family of tyrosine kinase 2). We previously showed that JAK2, in addition to being a signal transducer, dramatically increases the half-life of mature GHR, partly by preventing constitutive GHR down-regulation. Herein we explored GHR and JAK2 determinants for both constitutive and GH-induced GHR down-regulation, exploiting the previously characterized GHR- and JAK2-deficient gamma2A reconstitution system. We found that JAK2's ability to protect mature GHR from rapid degradation measured in the presence of the protein synthesis inhibitor, cycloheximide, depended on the presence of GHR's Box 1 element and the intact JAK2 FERM (band 4.1/Ezrin/Radixin/Moesin); domain, but not the kinase-like or kinase domains of JAK2. Thus, GHR-JAK2 association, but not JAK2 kinase activity, is required for JAK2 to inhibit constitutive GHR down-regulation and enhance GHR half-life. In cells that expressed JAK2, but not cells lacking JAK2, GH markedly enhanced GHR degradation. Like JAK2-induced protection from constitutive down-regulation, GH-induced GHR down-regulation required the GHR Box 1 element and an intact JAK2 FERM domain. However, a JAK2 mutant lacking the kinase-like and kinase domains did not mediate GH-induced GHR down-regulation. Likewise, a kinase-deficient JAK2 was insufficient for this purpose, indicating that kinase activity is required. Both lactacystin (a proteasome inhibitor) and chloroquine (a lysosome inhibitor) blocked GH-induced GHR loss. Interestingly, GH-induced GHR ubiquitination, like down-regulation, was prevented in cells expressing a kinase-deficient JAK2 protein. Further, a GHR mutant, of which all the cytoplasmic tyrosine residues were changed to phenylalanines, was resistant to GH-induced GHR ubiquitination and down-regulation. Collectively, our data suggest that determinants required for JAK2 to protect mature GHR from constitutive degradation differ from those that drive GH-induced GHR down-regulation. The latter requires GH-induced JAK2 activation and GHR tyrosine phosphorylation and is correlated to GHR ubiquitination in our reconstitution system.


Asunto(s)
Receptores de Somatotropina/genética , Receptores de Somatotropina/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Línea Celular , Cloroquina/farmacología , Cicloheximida/farmacología , Regulación hacia Abajo/efectos de los fármacos , Hormona de Crecimiento Humana/farmacología , Humanos , Janus Quinasa 2/química , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Lisosomas/efectos de los fármacos , Ratones , Mutación , Fosforilación , Inhibidores de Proteasoma , Estructura Terciaria de Proteína , Inhibidores de la Síntesis de la Proteína/farmacología , Conejos , Receptores de Somatotropina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transfección , Tirosina/química
17.
Clin Cancer Res ; 24(6): 1402-1414, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29311118

RESUMEN

Purpose: The retinoblastoma tumor suppressor (RB), a key regulator of cell-cycle progression and proliferation, is functionally suppressed in up to 50% of non-small cell lung cancer (NSCLC). RB function is exquisitely controlled by a series of proteins, including the CyclinD-CDK4/6 complex. In this study, we interrogated the capacity of a CDK4/6 inhibitor, palbociclib, to activate RB function.Experimental Design and Results: We employed multiple isogenic RB-proficient and -deficient NSCLC lines to interrogate the cytostatic and cytotoxic capacity of CDK 4/6 inhibition in vitro and in vivo We demonstrate that while short-term exposure to palbociclib induces cellular senescence, prolonged exposure results in inhibition of tumor growth. Mechanistically, CDK 4/6 inhibition induces a proapoptotic transcriptional program through suppression of IAPs FOXM1 and Survivin, while simultaneously augmenting expression of SMAC and caspase-3 in an RB-dependent manner.Conclusions: This study uncovers a novel function of RB activation to induce cellular apoptosis through therapeutic administration of a palbociclib and provides a rationale for the clinical evaluation of CDK 4/6 inhibitors in the treatment of patients with NSCLC. Clin Cancer Res; 24(6); 1402-14. ©2018 AACR.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Mitocondriales/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteína de Retinoblastoma/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Biomarcadores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Ratones , Especies Reactivas de Oxígeno/metabolismo , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Endocrinology ; 148(6): 2894-903, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17332060

RESUMEN

Once reserved solely for the treatment of short stature, the now readily available recombinant GH has expanded the use of the hormone to include the treatment of cardiovascular, renal, muscular, skeletal, immunological, psychosocial, and metabolic abnormalities associated with GH deficiency. There are also proposals for the widespread use of the hormone to ameliorate or reverse aging. However, this extensive use of GH has revealed intrinsic sexual dimorphisms in which females are considerably less responsive to the therapeutic regimen than are males. Dynamic changes in the Janus kinase-2 (Jak2)/signal transducers and activators of transcription (Stat5B) signaling pathway [as determined by transducer activation, Stat5B binding to the GH-responsive promoter of the CYP2C11 gene, and expression levels of the suppressors of cytokine signaling family (Socs2, Socs3, and Cis)] were examined in male and female rat-derived primary hepatocyte cultures exposed to the masculine-like episodic GH profile. We report that the cellular actions of GH normally mediated by activation of the Jak2/Stat5B pathway are suppressed in female cells possibly due to an inherent overexpression of Cis, a member of the suppressors of cytokine signaling family that normally down-regulates the Jak2/Stat5B pathway.


Asunto(s)
Hormona del Crecimiento/fisiología , Caracteres Sexuales , Transducción de Señal/genética , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Familia 2 del Citocromo P450 , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hormona del Crecimiento/farmacología , Hipofisectomía , Janus Quinasa 2/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Regiones Promotoras Genéticas , Unión Proteica , Ratas , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroide 16-alfa-Hidroxilasa/genética , Esteroide 16-alfa-Hidroxilasa/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética
19.
Biochem Pharmacol ; 74(10): 1476-84, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17868651

RESUMEN

CYP2C11, the most commonly expressed isoform of cytochrome P450 in male rat liver, was measured in spleen, thymus and bone marrow by quantitative real-time PCR and enhanced Western blotting. CYP2C11 concentrations in the lymphoid tissues were a fraction of that observed in liver, but like the liver, were sexually dimorphic (M>F) with mRNA and protein levels in agreement. Although the response to hypophysectomy varied according to tissue and sex, expression levels of CYP2C11 in all measured tissues remained greater in males. Further differences in CYP2C11 expression between liver and lymphoid tissue were observed following restoration of the circulating masculine growth hormone profile in hypophysectomized rats. In contrast to the liver where the renaturalized growth hormone profile elevated CYP2C11 expression in both sexes, the response was opposite in spleen and thymus with isoform concentrations declining in both sexes. Lastly, the divergent response of CYP2C11 between the liver and immune system was examined in cultured splenocytes exposed to different mitogens. In contrast to the dramatic depletion of CYP2C11 reported in proliferating hepatocytes, mitogen-stimulation resulted in a significant elevation in splenocyte CYP2C11 expression. In summary, we report for the first time that thymus, spleen and bone marrow express, albeit nominal, sex-dependent levels of CYP2C11 (M>F) whose regulation appears to be under some hormonal control, but very different from that of the hepatic isoform.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Médula Ósea/enzimología , Hormona del Crecimiento/farmacología , Bazo/enzimología , Esteroide 16-alfa-Hidroxilasa/metabolismo , Timo/enzimología , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Familia 2 del Citocromo P450 , Femenino , Hipofisectomía , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Mitógenos/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Bazo/efectos de los fármacos , Esteroide 16-alfa-Hidroxilasa/genética , Timo/efectos de los fármacos
20.
Mol Ther Nucleic Acids ; 6: 259-268, 2017 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-28325292

RESUMEN

There is an unmet need in the development of an effective therapy for mutant K-ras-expressing non-small-cell lung cancer (NSCLC). Although various small molecules have been evaluated, an effective therapy remains a dream. siRNAs have the potential to downregulate mutant K-ras both at the protein and mRNA levels. However, a safe and effective delivery of siRNAs to tumors remains a limitation to their translational application in the treatment of this highly debilitating disease. Here we developed a novel hybrid nanoparticle carrier for effective delivery of anti-mutant K-ras to NSCLC (AKSLHN). The ability of this treatment modality to regress lung tumors in mouse models was evaluated as a monotherapy or as a combination treatment with erlotinib. Further, the toxicity of this treatment modality to healthy tissues was evaluated, along with its ability to elicit immune/inflammatory reactions. The results suggest that this treatment modality is a promising prospect for the treatment of mutant K-ras-expressing NSCLC without any accompanying toxicity. However, further understanding of the cellular-level interaction between AHSLHN and erlotinib needs to be attained before this promising treatment modality can be brought to the bedside.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA