Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Curr Issues Mol Biol ; 46(5): 3990-4003, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38785514

RESUMEN

Retinoic acid (RA) regulates stemness and differentiation in human embryonic stem cells (ESCs). Ewing sarcoma (ES) is a pediatric tumor that may arise from the abnormal development of ESCs. Here we show that RA impairs the viability of SK-ES-1 ES cells and affects the cell cycle. Cells treated with RA showed increased levels of p21 and its encoding gene, CDKN1A. RA reduced mRNA and protein levels of SRY-box transcription factor 2 (SOX2) as well as mRNA levels of beta III Tubulin (TUBB3), whereas the levels of CD99 increased. Exposure to RA reduced the capability of SK-ES-1 to form tumorspheres with high expression of SOX2 and Nestin. Gene expression of CD99 and CDKN1A was reduced in ES tumors compared to non-tumoral tissue, whereas transcript levels of SOX2 were significantly higher in tumors. For NES and TUBB3, differences between tumors and control tissue did not reach statistical significance. Low expression of CD99 and NES, and high expression of SOX2, were significantly associated with a poorer patient prognosis indicated by shorter overall survival (OS). Our results indicate that RA may display rather complex modulatory effects on multiple target genes associated with the maintenance of stem cell's features versus their differentiation, cell cycle regulation, and patient prognosis in ES.

2.
Mol Cell Biochem ; 478(10): 2241-2255, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36637615

RESUMEN

Medulloblastoma, neuroblastoma, and pediatric glioma account for almost 30% of all cases of pediatric cancers. Recent evidence indicates that pediatric nervous system tumors originate from stem or progenitor cells and present a subpopulation of cells with highly tumorigenic and stem cell-like features. These cancer stem cells play a role in initiation, progression, and resistance to treatment of pediatric nervous system tumors. Histone modification, DNA methylation, chromatin remodeling, and microRNA regulation display a range of regulatory activities involved in cancer origin and progression, and cellular identity, especially those associated with stem cell features, such as self-renewal and pluripotent differentiation potential. Here, we review the contribution of different epigenetic mechanisms in pediatric nervous system tumor cancer stem cells. The choice between a differentiated and undifferentiated state can be modulated by alterations in the epigenome through the regulation of stemness genes such as CD133, SOX2, and BMI1 and the activation neuronal of differentiation markers, RBFOX3, GFAP, and S100B. Additionally, we highlighted the stage of development of epigenetic drugs and the clinical benefits and efficacy of epigenetic modulators in pediatric nervous system tumors.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias del Sistema Nervioso , Humanos , Niño , Epigenoma , Glioma/genética , Glioma/patología , Neoplasias Encefálicas/patología , Células Madre Neoplásicas/patología , Neoplasias del Sistema Nervioso/genética , Neoplasias del Sistema Nervioso/patología
3.
Int J Mol Sci ; 24(20)2023 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-37894922

RESUMEN

Changes in epigenetic programming have been proposed as being key events in the initiation and progression of childhood cancers. HMT euchromatic histone lysine methyltransferase 2 (G9a, EHMT2), which is encoded by the G9a (Ehmt2) gene, as well as its related protein GLP, which is encoded by the GLP/Ehmt1 gene, participate in epigenetic regulation by contributing to a transcriptionally repressed chromatin state. G9a/GLP activation has been reported in several cancer types. Herein, we evaluated the role of G9a in two solid pediatric tumors: neuroblastoma (NB) and Ewing sarcoma (ES). Our results show that G9a/Ehmt2 and GLP/Ehmt1 expression is higher in tumors with poorer prognosis, including St4 International Neuroblastoma Staging System (INSS) stage, MYCN amplified NB, and metastatic ES. Importantly, higher G9a and GLP levels were associated with shorter patient overall survival (OS) in both NB and ES. Moreover, pharmacological inhibition of G9a/GLP reduced cell viability in NB and ES cells. These findings suggest that G9a and GLP are associated with more aggressive NB and ES tumors and should be further investigated as being epigenetic targets in pediatric solid cancers.


Asunto(s)
Neuroblastoma , Sarcoma de Ewing , Niño , Humanos , Supervivencia Celular/genética , Epigénesis Genética , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad/metabolismo , Histona Metiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Neuroblastoma/genética , Sarcoma de Ewing/genética
4.
Mol Cell Biochem ; 476(11): 4107-4116, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34292482

RESUMEN

The transcription factor Zinc finger E-box binding 1 (ZEB1) displays a range of regulatory activities in cell function and embryonic development, including driving epithelial-mesenchymal transition. Several aspects of ZEB1 function can be regulated by its functional interactions with noncoding RNA types, namely microRNAs (miRNAs) and long noncoding RNAs (lncRNAs). Increasing evidence indicates that ZEB1 importantly influences cancer initiation, tumor progression, metastasis, and resistance to treatment. Cancer is the main disease-related cause of death in children and adolescents. Although the role of ZEB1 in pediatric cancer is still poorly understood, emerging findings have shown that it is expressed and regulates childhood solid tumors including osteosarcoma, retinoblastoma, neuroblastoma, and central nervous system tumors. Here, we review the evidence supporting a role for ZEB1, and its interplays with miRNAs and lncRNAs, in pediatric cancers.


Asunto(s)
MicroARNs/genética , Neoplasias/genética , Neoplasias/metabolismo , ARN Largo no Codificante/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Carcinogénesis , Niño , Transición Epitelial-Mesenquimal , Humanos , Neoplasias/patología , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Neoplasias de la Retina/genética , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
5.
Pediatr Blood Cancer ; 68(3): e28779, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33118699

RESUMEN

PURPOSE: The purpose of this study was to analyze the effect of hospital care volume on the overall survival of children with cancer in Southern Brazil. PATIENTS AND METHODS: We performed a retrospective cohort study of 1378 cancer patients aged 0-19 years, diagnosed with cancer between August 1, 2009 and December 31, 2015 in Rio Grande do Sul, who received hospital treatment in institutions affiliated with the Universal Health Care System (Sistema Único de Saúde [SUS]). RESULTS: Most children and adolescents were male (56.9%) and White (75.8%). The most common types of cancer in our cohort were acute leukemia (40.7%), followed by lymphoma (15.9%) and central nervous system tumors (8.8%). Ninety-five percent of the patients were treated in specialized pediatric oncology centers. The cumulative probability of survival at 5 years for all patients was 73.8% (95% confidence interval [CI] 71.4-76.0%). Survival was significantly higher for patients younger than 4 years of age (P = .012) compared to all other age groups. Patients treated in institutions with a pediatric oncology patient volume of less than 15 patients/year were 41% more likely to die than patients treated in institutions with a volume of 60 patients/year or more (P = .029). CONCLUSION: Cancer is the leading cause of death by natural causes in all age groups in Brazil, but, even so, childhood tumors are rare. This complexity makes childhood cancer care a challenge. In this study, we reiterate that pediatric cancer patients demonstrate better overall survival when treated in high-volume hospitals.


Asunto(s)
Hospitalización/estadística & datos numéricos , Hospitales de Alto Volumen/estadística & datos numéricos , Hospitales de Bajo Volumen/estadística & datos numéricos , Neoplasias/mortalidad , Adolescente , Adulto , Brasil/epidemiología , Niño , Preescolar , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Neoplasias/epidemiología , Neoplasias/patología , Neoplasias/terapia , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
6.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34681949

RESUMEN

Epigenetic mechanisms, including post-translational modifications of DNA and histones that influence chromatin structure, regulate gene expression during normal development and are also involved in carcinogenesis and cancer progression. The histone methyltransferase G9a (euchromatic histone lysine methyltransferase 2, EHMT2), which mostly mediates mono- and dimethylation by histone H3 lysine 9 (H3K9), influences gene expression involved in embryonic development and tissue differentiation. Overexpression of G9a has been observed in several cancer types, and different classes of G9a inhibitors have been developed as potential anticancer agents. Here, we review the emerging evidence suggesting the involvement of changes in G9a activity in brain tumors, namely glioblastoma (GBM), the main type of primary malignant brain cancer in adults, and medulloblastoma (MB), the most common type of malignant brain cancer in children. We also discuss the role of G9a in neuroblastoma (NB) and the drug development of G9a inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Antígenos de Histocompatibilidad , Humanos
7.
Mol Biol Rep ; 47(9): 6817-6828, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32862352

RESUMEN

A member of the Trk family of neurotrophin receptors, tropomyosin receptor kinase B (TrkB, encoded by the NTRK2 gene) is an increasingly important target in various cancer types, including glioblastoma (GBM). EGFR is among the most frequently altered oncogenes in GBM, and EGFR inhibition has been tested as an experimental therapy. Functional interactions between EGFR and TrkB have been demonstrated. In the present study, we investigated the role of TrkB and EGFR, and their interactions, in GBM. Analyses of NTRK2 and EGFR gene expression from The Cancer Genome Atlas (TCGA) datasets showed an increase in NTRK2 expression in the proneural subtype of GBM, and a strong correlation between NTRK2 and EGFR expression in glioma CpG island methylator phenotype (G-CIMP+) samples. We showed that when TrkB and EGFR inhibitors were combined, the inhibitory effect on A172 human GBM cells was more pronounced than when either inhibitor was given alone. When U87MG GBM cells were xenografted into the flank of nude mice, tumor growth was delayed by treatment with TrkB and EGFR inhibitors, given alone or combined, only at specific time points. Intracranial GBM growth in mice was not significantly affected by drug treatments. Our findings indicate that correlations between NTRK2 and EGFR expression occur in specific GBM subgroups. Also, our results using cultured cells suggest for the first time the potential of combining TrkB and EGFR inhibition for the treatment of GBM.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Inhibidores Enzimáticos/farmacología , Glioblastoma/metabolismo , Glicoproteínas de Membrana/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Receptor trkB/metabolismo , Animales , Azepinas/farmacología , Benzamidas/farmacología , Neoplasias Encefálicas/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/genética , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Clasificación del Tumor , Quinazolinas/farmacología , Receptor trkB/antagonistas & inhibidores , Receptor trkB/genética , Tirfostinos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Pediatr Hematol Oncol ; 37(2): 170-175, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31826690

RESUMEN

Histone deacetylase inhibitors (HDACis) are epigenetic agents that display antitumor activities in experimental medulloblastoma (MB). Fingolimod (FTY720), an immunosuppressant agent currently used in the treatment of multiple sclerosis, also has anticancer actions and can act as an HDACi. Here we examined whether fingolimod can inhibit human MB cell viability and survival, and if the effects are accompanied by increased histone acetylation. D283 and DAOY MB cells were treated with different doses of fingolimod. Cell viability was assessed by cell counting in a hemocytometer, and cell survival was analyzed with a colony formation assay. Histone H3 acetylation was measured with an enzyme-linked immunosorbent assay (ELISA). Fingolimod at 7.5 or 10 µM, but not at 5 µM, induced a significant reduction in cell viability in D283 and DAOY cultures, and similar results were observed for inhibition of cell survival. In both cell lines, fingolimod also led to a significant increase in the levels of acetylated H3. These findings provide preliminary evidence indicating that fingolimod induces antitumor activities in MB, possibly through a mechanism which increases H3 histone acetylation.


Asunto(s)
Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Meduloblastoma/tratamiento farmacológico , Acetilación , Clorhidrato de Fingolimod/farmacología , Humanos , Inmunosupresores/farmacología
9.
Oncology ; 97(1): 26-37, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31071716

RESUMEN

OBJECTIVE: Normally, activation of tropomyosin-related kinase (TRK) receptors by neurotrophins (NTs) stimulates intracellular pathways involved in cell survival and proliferation. Dysregulation of NT/TRK signaling may affect neoplasm prognosis. Data on NT and TRK expression in melanomas are limited, and it is unclear whether NT/TRK signaling pathways are involved in the origin and progression of this neoplasm. METHODS: We examined whether NT/TRK expression differs across different cutaneous melanoma grades and subtypes, and whether it is associated with melanoma prognosis and survival. A cross-sectional study was performed in which the expression of TrkA, TrkB, nerve growth factor (NGF), and brain-derived neurotrophic factor (BDNF) was analyzed by immunohistochemistry of 154 melanoma samples. We investigated NT/TRK expression associations with prognostic factors for melanoma, relapse-free survival (RFS), and overall survival (OS). RESULTS: Of the 154 melanoma samples, 77 (55.4%) were TrkA immunopositive, 81 (58.3%) were TrkB immunopositive, 113 (81.3%) were BDNF immunopositive, and 104 (75.4%) were NGF immunopositive. We found NT/TRK expression associated strongly with several clinical prognostic factors, including the tumor-node-metastasis stage (p < 0.001), histological subtype (p < 0.001), and Clark level (p < 0.05), as well as with a worse OS (p < 0.05 for all, except TrkB) and RFS (p < 0.05 for all). CONCLUSIONS: Our results show strong associations of NT/TRK expression with melanoma stage progression and a poor prognosis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Melanoma/genética , Glicoproteínas de Membrana/genética , Factores de Crecimiento Nervioso/genética , Receptor trkA/genética , Receptor trkB/genética , Neoplasias Cutáneas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanoma/inmunología , Melanoma/patología , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Factor de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/inmunología , Pronóstico , Receptor trkA/inmunología , Receptor trkB/inmunología , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Adulto Joven , Melanoma Cutáneo Maligno
10.
Oral Dis ; 25(8): 1925-1936, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31498938

RESUMEN

OBJECTIVES: To evaluate the expression of brain-derived neurotrophic factor (BDNF), its tyrosine kinase receptor B (TrkB), and two downstream targets of this pathway, Akt and ribosomal protein S6 (RPS6), in normal oral mucosa (NOM), oral leukoplakia (OL), and oral squamous cell carcinoma (OSCC) and correlate this expression with OSCC patients' outcomes, cell senescence, and "stemness" profile. MATERIALS AND METHODS: Ten cases of NOM, 32 OL, and 72 primary OSCC were included. Immunohistochemical analysis for BDNF, TrkB, p-TrkB, p-Akt, and p-RPS6 was performed. Cell senescence and stemness profile of OSCC were evaluated through p16 and BMI-1 immunohistochemical expression, respectively. The slides were scanned into high-resolution images and quantified through digital analysis. RESULTS: Oral squamous cell carcinoma presented increased expression of BDNF/TrkB/Akt pathway compared to NOM and OL. OSCC diagnosed in advanced clinical stages presented an upregulation of BDNF and p-TrkB. BDNF and p-Akt were identified as predictors of poor disease-specific survival. The increase in stemness profile was correlated with a decrease in p-TrkB and p-Akt expression. CONCLUSIONS: BDNF/TrkB/Akt pathway is significantly increased in malignant cells from OSCC. Moreover, BDNF and Akt represent biomarkers capable to predict a poor prognosis of OSCC patients.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias de la Boca/patología , Receptor trkB/metabolismo , Carcinoma de Células Escamosas/metabolismo , Humanos , Neoplasias de la Boca/mortalidad , Valor Predictivo de las Pruebas , Proteínas Proto-Oncogénicas c-akt , Receptor trkB/genética
11.
J Oral Pathol Med ; 47(3): 221-227, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28650560

RESUMEN

Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family of growth factors that was first known as responsible for sustain the growth, function, and plasticity of neural cells. BDNF exerts its effects by binding to the tyrosine kinase receptor B (TrkB). The BDNF/TrkB axis has been reported to be overexpressed in several neurogenic and non-neurogenic tumors. Its higher expression was associated with a poor prognosis to patients affected by different human malignancies, tumor growth, invasion, and metastasis; epithelial-mesenchymal transition and resistance to chemotherapy. BDNF/TrkB represent promising targets to the development of novel anticancer therapies. Some clinical trials are currently evaluating the efficacy of Trk protein-target drugs in different types of solid tumors. To date, few groups have evaluated the DNF/TrkB pathway in head and neck malignancies. The aims of this study were to review the literature concerning the role of BDNF/TrkB activation in head and neck squamous cell carcinoma and malignant salivary gland tumors and to discuss future perspectives of BDNF/TrkB-target therapy.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Receptor trkB/metabolismo , Antineoplásicos/farmacología , Carcinoma Adenoide Quístico/tratamiento farmacológico , Carcinoma Adenoide Quístico/metabolismo , Carcinoma Adenoide Quístico/patología , Carcinoma Mucoepidermoide/tratamiento farmacológico , Carcinoma Mucoepidermoide/metabolismo , Carcinoma Mucoepidermoide/patología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Cisplatino/farmacología , Resistencia a Antineoplásicos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Humanos , Invasividad Neoplásica , Pronóstico
12.
J Neural Transm (Vienna) ; 124(11): 1331-1339, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28864864

RESUMEN

Over two-thirds of women with breast cancer have positive tumors for hormone receptors, and these patients undergo treatment with endocrine therapy, tamoxifen being the most widely used agent. Despite being very effective in breast cancer treatment, tamoxifen is associated with side effects that include cognitive impairments. However, the specific aspects and mechanisms underlying these impairments remain to be characterized. Here, we have investigated the effects of tamoxifen and interaction with estrogen receptors on formation of memory for inhibitory avoidance conditioning in female rats. In the first experiment, Wistar female rats received a single oral dose of tamoxifen (1, 3, or 10 mg/kg) or saline by gavage immediately after training and were tested for memory consolidation 24 h after training. In the second experiment, rats received a single dose of 1 mg/kg tamoxifen or saline by gavage 3 h after training and were tested 24 h after training for memory consolidation. In the third experiment, rats received a subcutaneous injection with estrogen receptor α agonist or estrogen receptor beta agonist 30 min before the training. After training, rats received a single oral dose of tamoxifen 1 mg/kg or saline and were tested 24 h after training. In the fourth experiment, rats were trained and tested 24 h later. Immediately after test, rats received a single dose of tamoxifen (1 mg/kg) or saline by gavage and were given four additional daily test trials followed by a re-instatement. Tamoxifen at 1 mg/kg impaired memory consolidation when given immediately after training and the estrogen receptor alpha agonist improved the tamoxifen-related memory impairment. Moreover, tamoxifen impairs memory consolidation of the test. These findings indicate that estrogen receptors regulate the early phase of memory consolidation and the effects of tamoxifen on memory consolidation.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Antagonistas del Receptor de Estrógeno/farmacología , Receptor alfa de Estrógeno/metabolismo , Trastornos de la Memoria/inducido químicamente , Tamoxifeno/farmacología , Animales , Relación Dosis-Respuesta a Droga , Estrógenos/uso terapéutico , Extinción Psicológica/efectos de los fármacos , Femenino , Consolidación de la Memoria/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Nitrilos/uso terapéutico , Fenoles/uso terapéutico , Propionatos/uso terapéutico , Pirazoles/uso terapéutico , Ratas , Ratas Wistar , Estadísticas no Paramétricas
13.
Invest New Drugs ; 32(5): 783-94, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24838627

RESUMEN

TP53 mutation is a common event in many cancers, including pancreatic adenocarcinoma, where it occurs in 50-70 % of cases. In an effort to reactivate mutant p53 protein, several new drugs are being developed, including PRIMA-1 and PRIMA-1(Met)/APR-246 (p53 reactivation and induction of massive apoptosis). PRIMA-1 has been shown to induce apoptosis in tumor cells by reactivating p53 mutants, but its effect in pancreatic cancer remains unclear. Here we investigated the effects of PRIMA-1 on cell viability, cell cycle and expression of p53-regulated proteins in PANC-1 and BxPC-3 (mutant TP53), and CAPAN-2 (wild-type TP53) pancreatic cell lines. Treatment with PRIMA-1 selectively induced apoptosis and cell cycle arrest in p53 mutant cells compared to CAPAN-2 cells. The growth suppressive effect of PRIMA-1 was markedly reduced in p53 mutant cell lines transfected with p53 siRNA, supporting the role of mutant p53 in PRIMA-1 induced cell death. Moreover, treatment with the thiol group donor N-acetylcysteine completely blocked PRIMA-1-induced apoptosis and reinforced the hypothesis that thiol modifications are important for PRIMA-1 biological activity. In combination treatments, PRIMA-1 enhanced the anti-tumor activity of several chemotherapic drugs against pancreatic cancer cells and also exhibited a pronounced synergistic effect in association with the Mdm2 inhibitor Nutlin-3. Taken together, our data indicate that PRIMA-1 induces apoptosis in p53 mutant pancreatic cancer cells by promoting the re-activation of p53 and inducing proapoptotic signaling pathways, providing in vitro evidence for a potential therapeutic approach in pancreatic cancer.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Aza/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neoplasias Pancreáticas/metabolismo , Proteína p53 Supresora de Tumor/genética , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Bortezomib , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Clorhidrato de Erlotinib , Humanos , Imidazoles/farmacología , Mutación , Piperazinas/farmacología , Pirazinas/farmacología , Quinazolinas/farmacología , ARN Interferente Pequeño/genética , Proteína p53 Supresora de Tumor/metabolismo , Gemcitabina
14.
Genes (Basel) ; 15(4)2024 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-38674359

RESUMEN

Rare sarcomas present significant treatment challenges compared to more prevalent soft tissue sarcomas due to limited treatment options and a poor understanding of their biology. This study investigates a unique case of penile sarcoma, providing a comprehensive morphological and molecular analysis. Through the creation of experimental patient-derived models-including patient-derived xenograft (PDX), 3D, and monolayer primary cultures-we successfully replicated crucial molecular traits observed in the patient's tumor, such as smooth muscle actin and CD99 expression, along with specific mutations in genes like TSC2 and FGFR4. These models are helpful in assessing the potential for an in-depth exploration of this tumor's biology. This comprehensive approach holds promise in identifying potential therapeutic avenues for managing this exceedingly rare soft tissue sarcoma.


Asunto(s)
Sarcoma , Animales , Humanos , Masculino , Ratones , Mutación , Neoplasias del Pene/genética , Neoplasias del Pene/patología , Sarcoma/genética , Sarcoma/patología , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Persona de Mediana Edad
15.
Mol Biol Rep ; 40(8): 4929-34, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23661019

RESUMEN

The Ewing Sarcoma is an important tumor of bone and soft tissue. The SNPs Arg72Pro of TP53 and T309G of MDM2 have been associated with many cancer types and have been differently distributed among populations worldwide. Based on a case-control design, this study aimed to assess the role of these SNPs in 24 Ewing Sarcoma patients, compared to 91 control individuals. DNA samples were extracted from blood and genotyped for both SNPs by PCR-RFLP and confirmed by DNA sequencing. The results showed an association between the G allele of the T309G and Ewing Sarcoma (P=0.02). Comparing to the TT carriers, the risk of G allele carriers was 3.35 (95% CI=1.22-9.21) with P=0.02. At the genotypic level, an association of the TT genotype with the control group (P=0.03) was found. Comparing to the TT genotype, the risk of TG and GG was 2.97 (95% CI=1.03-8.58) with P=0.04 and 5.00 (95% CI=1.23-20.34) with P=0.02, respectively. No associations regarding the Arg72Pro SNP were found. Considering that the T309G has been associated with several types of cancer, including sarcomas, our results indicate that this SNP may also be important to Ewing Sarcoma predisposition.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Sarcoma de Ewing/genética , Proteína p53 Supresora de Tumor/genética , Sustitución de Aminoácidos/genética , Brasil , Estudios de Casos y Controles , Estudios de Asociación Genética , Genotipo , Humanos , Modelos Logísticos , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Análisis de Secuencia de ADN
16.
Childs Nerv Syst ; 29(12): 2145-50, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24092425

RESUMEN

PURPOSE: Medulloblastoma is the most common malignant childhood brain tumor for which the development of new molecularly targeted therapies is needed. Novel therapeutic targets under investigation include growth factor receptors. Here, we show that the combined inhibition of the epidermal growth factor receptor (EGFR) and neuromedin B receptor (NMBR, BB1) results in increased cell death in human medulloblastoma cell lines. METHODS: DAOY and D283 human medulloblastoma cells were treated with human recombinant neuromedin B (NMB, an NMBR agonist), the NMBR antagonist BIM-23127, the anti-EGFR monoclonal antibody cetuximab, or BIM-23127 combined with cetuximab. Cell death was examined with trypan blue cell counting. RESULTS: Both cell lines expressed mRNA for EGFR, NMB, and NMBR detected by reverse transcriptase polymerase chain reaction. Cetuximab at 10 µg/ml significantly reduced the number of DAOY cells, but did not affect D283 cells. NMB and BIM-23127 did not change cell number when used alone. However, cetuximab, at a dose that did not have an effect by itself, was able to reduce the number of DAOY cells when combined with BIM-23127. CONCLUSION: These results provide preliminary evidence that NMBR blockade can potentiate the antitumor effect of anti-EGFR therapy in medulloblastoma.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias Cerebelosas/patología , Receptores ErbB/antagonistas & inhibidores , Meduloblastoma/patología , Péptidos Cíclicos/administración & dosificación , Receptores de Bombesina/antagonistas & inhibidores , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cetuximab , Sinergismo Farmacológico , Humanos , Meduloblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Curr Probl Cancer ; 47(4): 100996, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37467541

RESUMEN

Gynecological and breast cancers affect women's health worldwide. Although chemotherapy is one of the principal treatments for cancer, it also has limitations owing to toxicity and tumor resistance to the drugs used. Thus, individualized treatment based on personal tumor characteristics is essential for improving therapeutic outcomes and patient survival. Chemoresistance and chemosensitivity tests can be useful for predicting tumor response and guiding chemotherapy choices. This methodology has already been applied to breast, ovarian, cervical, and endometrial cancers, identifying successfully which drugs cause resistance and sensitivity responses for each individual person, influencing their progression-free survival and overall response. In addition, more recent techniques, such as organoids and patient-derived xenografts, can also recapitulate patients' tumor characteristics and contribute to chemo response evaluation. Therefore, this review compiles information on chemoresistance and chemosensitivity tests performed in gynecologic and breast cancers and their main results for women's health improvement.


Asunto(s)
Neoplasias de la Mama , Neoplasias Endometriales , Ginecología , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Mama
18.
Curr Stem Cell Res Ther ; 18(7): 926-936, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35761483

RESUMEN

Resistance to chemotherapy poses a major challenge for cancer treatment. Reactivating a stem cell program resembling that seen in embryonic development can lead cancer cells to acquire a stem-cell phenotype characterized by expression of stemness genes, pluripotency, high self-renewal ability, and tumor-initiating capability. These cancer stem cells (CSCs) are usually resistant to anticancer drugs and are likely involved in treatment failure in many cancer types. Ewing sarcoma (ES) is a pediatric cancer type typically resulting from a typical genetic alteration affecting bone or soft tissues. Despite advances in treatment, survival prognostic remains poor for patients with refractory or recurrent disease. Here, we review the increasing evidence indicating that ES tumors contain a CSC subpopulation expressing stem cell genes, including BM1, OCT3/4, NANOG, and SOX2, that plays a role in resistance to drug treatment, and current experimental strategies that successfully counteract chemoresistance mediated by CSCs in ES.


Asunto(s)
Antineoplásicos , Sarcoma de Ewing , Humanos , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Células Madre Neoplásicas/metabolismo
19.
Neuromolecular Med ; 25(1): 64-74, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35716340

RESUMEN

Medulloblastoma (MB) is a malignant brain tumor that afflicts mostly children and adolescents and presents four distinct molecular subgroups, known as WNT, SHH, Group 3, and Group 4. ZEB1 is a transcription factor that promotes the expression of mesenchymal markers while restraining expression of epithelial and polarity genes. Because of ZEB1 involvement in cerebellum development, here we investigated the role of ZEB1 in MB. We found increased expression of ZEB1 in MB tumor samples compared to normal cerebellar tissue. Expression was higher in the SHH subgroup when compared to all other MB molecular subgroups. High ZEB1 expression was associated with poor prognosis in Group 3 and Group 4, whereas in patients with WNT tumors poorer prognosis were related to lower ZEB1 expression. There was a moderate correlation between ZEB1 and MYC expression in Group 3 and Group 4 MB. Treatment with the immunomodulator and histone deacetylase (HDAC) inhibitor fingolimod (FTY720) reduced ZEB1 expression specifically in D283 cells, which are representative of Group 3 and Group 4 MB. These findings reveal novel subgroup-specific associations of ZEB1 expression with survival in patients with MB and suggest that ZEB1 expression can be reduced by pharmacological agents that target HDAC activity.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Cerebelosas , Meduloblastoma , Niño , Adolescente , Humanos , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/genética , Cerebelo , Inhibidores de Histona Desacetilasas/uso terapéutico , Clorhidrato de Fingolimod/uso terapéutico , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
20.
Biochem Biophys Res Commun ; 425(2): 328-32, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22842573

RESUMEN

The clinical success of targeted treatment of colorectal cancer (CRC) is often limited by resistance to anti-epidermal growth factor receptor (EGFR) therapy. The neurotrophin brain-derived neurotrophic factor (BDNF) and its receptor TrkB have recently emerged as anticancer targets, and we have previously shown increased BDNF levels in CRC tumor samples. Here we report the findings from in vitro experiments suggesting that BDNF/TrkB signaling can protect CRC cells from the antitumor effects of EGFR blockade. The anti-EGFR monoclonal antibody cetuximab reduced both cell proliferation and the mRNA expression of BDNF and TrkB in human HT-29 CRC cells. The inhibitory effect of cetuximab on cell proliferation and survival was counteracted by the addition of human recombinant BDNF. Finally, the Trk inhibitor K252a synergistically enhanced the effect of cetuximab on cell proliferation, and this effect was blocked by BDNF. These results provide the first evidence that increased BDNF/TrkB signaling might play a role in resistance to EGFR blockade. Moreover, it is possible that targeting TrkB could potentiate the anticancer effects of anti-EGFR therapy.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Receptor trkB/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cetuximab , Células HT29 , Humanos , Receptor trkB/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA