Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Death Dis ; 14(12): 797, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38057340
2.
Cell Death Dis ; 14(10): 702, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37891184

RESUMEN

FOXO family of proteins are transcription factors involved in many physiological and pathological processes including cellular homeostasis, stem cell maintenance, cancer, metabolic, and cardiovascular diseases. Genetic evidence has been accumulating to suggest a prominent role of FOXOs in lifespan regulation in animal systems from hydra, C elegans, Drosophila, and mice. Together with the observation that FOXO3 is the second most replicated gene associated with extreme human longevity suggests that pharmacological targeting of FOXO proteins can be a promising approach to treat cancer and other age-related diseases and extend life and health span. However, due to the broad range of cellular functions of the FOXO family members FOXO1, 3, 4, and 6, isoform-specific targeting of FOXOs might lead to greater benefits and cause fewer side effects. Therefore, a deeper understanding of the common and specific features of these proteins as well as their redundant and specific functions in our cells represents the basis of specific targeting strategies. In this review, we provide an overview of the evolution, structure, function, and disease-relevance of each of the FOXO family members.


Asunto(s)
Factores de Transcripción Forkhead , Neoplasias , Humanos , Ratones , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Caenorhabditis elegans/metabolismo , Longevidad/genética , Isoformas de Proteínas/genética , Neoplasias/patología
3.
Science ; 380(6642): eabj5559, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37079666

RESUMEN

Cells respond to mitochondrial poisons with rapid activation of the adenosine monophosphate-activated protein kinase (AMPK), causing acute metabolic changes through phosphorylation and prolonged adaptation of metabolism through transcriptional effects. Transcription factor EB (TFEB) is a major effector of AMPK that increases expression of lysosome genes in response to energetic stress, but how AMPK activates TFEB remains unresolved. We demonstrate that AMPK directly phosphorylates five conserved serine residues in folliculin-interacting protein 1 (FNIP1), suppressing the function of the folliculin (FLCN)-FNIP1 complex. FNIP1 phosphorylation is required for AMPK to induce nuclear translocation of TFEB and TFEB-dependent increases of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) and estrogen-related receptor alpha (ERRα) messenger RNAs. Thus, mitochondrial damage triggers AMPK-FNIP1-dependent nuclear translocation of TFEB, inducing sequential waves of lysosomal and mitochondrial biogenesis.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Lisosomas , Mitocondrias , Biogénesis de Organelos , Proteínas Quinasas Activadas por AMP/metabolismo , Lisosomas/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Humanos
4.
Emerg Infect Dis ; 29(3): 569-575, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36737101

RESUMEN

We estimated comparative primary and booster vaccine effectiveness (VE) of SARS-CoV-2 Omicron BA.5 and BA.2 lineages against infection and disease progression. During April-June 2022, we implemented a case-case and cohort study and classified lineages using whole-genome sequencing or spike gene target failure. For the case-case study, we estimated the adjusted odds ratios (aORs) of vaccination using a logistic regression. For the cohort study, we estimated VE against disease progression using a penalized logistic regression. We observed no reduced VE for primary (aOR 1.07 [95% CI 0.93-1.23]) or booster (aOR 0.96 [95% CI 0.84-1.09]) vaccination against BA.5 infection. Among BA.5 case-patients, booster VE against progression to hospitalization was lower than that among BA.2 case-patients (VE 77% [95% CI 49%-90%] vs. VE 93% [95% CI 86%-97%]). Although booster vaccination is less effective against BA.5 than against BA.2, it offers substantial protection against progression from BA.5 infection to severe disease.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Humanos , Portugal , Estudios de Cohortes , SARS-CoV-2 , Progresión de la Enfermedad
5.
Traffic ; 23(12): 587-599, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36353954

RESUMEN

Chromosomal region maintenance 1 (CRM1 also known as Xpo1 and exportin-1) is the receptor for the nuclear export controlling the intracellular localization and function of many cellular and viral proteins that play a crucial role in viral infections and cancer. The inhibition of CRM1 has emerged as a promising therapeutic approach to interfere with the lifecycle of many viruses, for the treatment of cancer, and to overcome therapy resistance. Recently, selinexor has been approved as the first CRM1 inhibitor for the treatment of multiple myeloma, providing proof of concept for this therapeutic option with a new mode of action. However, selinexor is associated with dose-limiting toxicity and hence, the discovery of alternative small molecule leads that could be developed as less toxic anticancer and antiviral therapeutics will have a significant impact in the clinic. Here, we report a CRM1 inhibitor discovery platform. The development of this platform includes reporter cell lines that monitor CRM1 activity by using red fluorescent protein or green fluorescent protein-labeled HIV-1 Rev protein with a strong heterologous nuclear export signal. Simultaneously, the intracellular localization of other proteins, to be interrogated for their capacity to undergo CRM1-mediated export, can be followed by co-culturing stable cell lines expressing fluorescent fusion proteins. We used this platform to interrogate the mode of nuclear export of several proteins, including PDK1, p110α, STAT5A, FOXO1, 3, 4 and TRIB2, and to screen a compound collection. We show that while p110α partially relies on CRM1-dependent nuclear export, TRIB2 is exported from the nucleus in a CRM1-independent manner. Compound screening revealed the striking activity of an organoselenium compound on the CRM1 nuclear export receptor.


Asunto(s)
VIH-1 , Transporte Activo de Núcleo Celular , VIH-1/metabolismo , Carioferinas/metabolismo , Triazoles/metabolismo , Hidrazinas/farmacología , Hidrazinas/metabolismo , Núcleo Celular/metabolismo
6.
Molecules ; 27(17)2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-36080182

RESUMEN

Forkhead box O (FOXO) proteins are transcription factors involved in cancer and aging and their pharmacological manipulation could be beneficial for the treatment of cancer and healthy aging. FOXO proteins are mainly regulated by post-translational modifications including phosphorylation, acetylation and ubiquitination. As these modifications are reversible, activation and inactivation of FOXO factors is attainable through pharmacological treatment. One major regulatory input of FOXO signaling is mediated by protein kinases. Here, we use specific inhibitors against different kinases including PI3K, mTOR, MEK and ALK, and other receptor tyrosine kinases (RTKs) to determine their effect on FOXO3 activity. While we show that inhibition of PI3K efficiently drives FOXO3 into the cell nucleus, the dual PI3K/mTOR inhibitors dactolisib and PI-103 induce nuclear FOXO translocation more potently than the PI3Kδ inhibitor idelalisib. Furthermore, specific inhibition of mTOR kinase activity affecting both mTORC1 and mTORC2 potently induced nuclear translocation of FOXO3, while rapamycin, which specifically inhibits the mTORC1, failed to affect FOXO3. Interestingly, inhibition of the MAPK pathway had no effect on the localization of FOXO3 and upstream RTK inhibition only weakly induced nuclear FOXO3. We also measured the effect of the test compounds on the phosphorylation status of AKT, FOXO3 and ERK, on FOXO-dependent transcriptional activity and on the subcellular localization of other FOXO isoforms. We conclude that mTORC2 is the most important second layer kinase negatively regulating FOXO activity.


Asunto(s)
Factores de Transcripción Forkhead , Serina-Treonina Quinasas TOR , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
7.
J Inorg Biochem ; 235: 111915, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35834898

RESUMEN

Decavanadate is a polyoxometalate (POMs) that has shown extensive biological activities, including antidiabetic and anticancer activity. Importantly, vanadium-based compounds as well as antidiabetic biguanide drugs, such as metformin, have shown to exert therapeutic effects in melanoma. A combination of these agents, the metformin-decavanadate complex, was also recognized for its antidiabetic effects and recently described as a better treatment than the monotherapy with metformin enabling lower dosage in rodent models of diabetes. Herein, we compare the effects of decavanadate and metformin-decavanadate on Ca2+-ATPase activity in sarcoplasmic reticulum vesicles from rabbit skeletal muscles and on cell signaling events and viability in human melanoma cells. We show that unlike the decavanadate-mediated non-competitive mechanism, metformin-decavanadate inhibits Ca2+-ATPase by a mixed-type competitive-non-competitive inhibition with an IC50 value about 6 times higher (87 µM) than the previously described for decavanadate (15 µM). We also found that both decavanadate and metformin-decavanadate exert antiproliferative effects on melanoma cells at 10 times lower concentrations than monomeric vanadate. Western blot analysis revealed that both, decavanadate and metformin-decavanadate increased phosphorylation of extracellular signal-regulated kinase (ERK) and serine/threonine protein kinase AKT signaling proteins upon 24 h drug exposure, suggesting that the anti-proliferative activities of these compounds act independent of growth-factor signaling pathways.


Asunto(s)
Melanoma , Metformina , Adenosina Trifosfatasas , Animales , Aniones , Humanos , Hipoglucemiantes , Melanoma/tratamiento farmacológico , Metformina/farmacología , Polielectrolitos , Conejos , Vanadatos/farmacología
8.
J Gerontol A Biol Sci Med Sci ; 77(8): 1485-1493, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34508571

RESUMEN

Several chemical compounds including natural products have been suggested as being effective against age-related diseases or as beneficial for a healthy life. On the other hand, forkhead box O (FOXO) proteins are emerging as key cellular components associated with extreme human longevity. FOXO proteins are mainly regulated by posttranslational modifications and as these modifications are reversible, activation and inactivation of FOXO are attainable through pharmacological treatment. Here, we questioned whether a panel of compounds with known health-beneficial properties has the capacity to induce the activity of FOXO factors. We show that resveratrol, a phytoalexin present in grapes and other food products, the amide alkaloid piperlongumine found in the fruit of the long pepper, and the plant-derived ß-carboline compound harmine induced nuclear translocation of FOXO3. We also show that piperlongumine and harmine but not resveratrol activate FOXO-dependent transcription. We determined the half maximal effective concentration (EC50) values for resveratrol, piperlongumine, and harmine for FOXO translocation, and analyzed their inhibitory impact on chromosomal maintenance 1 (CRM1)-mediated nuclear export and the production of reactive oxygen species (ROS). We also used chemical biology approach and Western blot analysis to explore the underlying molecular mechanisms. We show that harmine, piperlongumine, and resveratrol activate FOXO3 independently of phosphoinositide 3-kinase (PI3K)/AKT signaling and the CRM1-mediated nuclear export. The effect of harmine on FOXO3 activity is at least partially mediated through the inhibition of dual-specificity tyrosine (Y) phosphorylationregulated kinase 1A (DYRK1A) and can be reverted by the inhibition of sirtuins (SIRTs).


Asunto(s)
Proteína Forkhead Box O3 , Proteínas Proto-Oncogénicas c-akt , Dioxolanos/farmacología , Proteína Forkhead Box O3/metabolismo , Harmina/farmacología , Humanos , Carioferinas , Fosfatidilinositol 3-Quinasas , Receptores Citoplasmáticos y Nucleares , Resveratrol/farmacología , Proteína Exportina 1
9.
Cancers (Basel) ; 13(11)2021 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-34198908

RESUMEN

The Tribbles family of pseudokinases controls a wide number of processes during cancer on-set and progression. However, the exact contribution of each of the three family members is still to be defined. Their function appears to be context-dependent as they can act as oncogenes or tumor suppressor genes. They act as scaffolds modulating the activity of several signaling pathways involved in different cellular processes. In this review, we discuss the state-of-knowledge for TRIB1, TRIB2 and TRIB3 in the development and progression of colorectal cancer. We take a perspective look at the role of Tribbles proteins as potential biomarkers and therapeutic targets. Specifically, we chronologically systematized all available articles since 2003 until 2020, for which Tribbles were associated with colorectal cancer human samples or cell lines. Herein, we discuss: (1) Tribbles amplification and overexpression; (2) the clinical significance of Tribbles overexpression; (3) upstream Tribbles gene and protein expression regulation; (4) Tribbles pharmacological modulation; (5) genetic modulation of Tribbles; and (6) downstream mechanisms regulated by Tribbles; establishing a comprehensive timeline, essential to better consolidate the current knowledge of Tribbles' role in colorectal cancer.

10.
Cancers (Basel) ; 12(12)2020 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33316942

RESUMEN

Therapy resistance is responsible for most relapses in patients with cancer and is the major challenge to improving the clinical outcome. The pseudokinase Tribbles homologue 2 (TRIB2) has been characterized as an important driver of resistance to several anti-cancer drugs, including the dual ATP-competitive PI3K and mTOR inhibitor dactolisib (BEZ235). TRIB2 promotes AKT activity, leading to the inactivation of FOXO transcription factors, which are known to mediate the cell response to antitumor drugs. To characterize the downstream events of TRIB2 activity, we analyzed the gene expression profiles of isogenic cell lines with different TRIB2 statuses by RNA sequencing. Using a connectivity map-based computational approach, we identified drug-induced gene-expression profiles that invert the TRIB2-associated expression profile. In particular, the natural alkaloids harmine and piperlongumine not only produced inverse gene expression profiles but also synergistically increased BEZ235-induced cell toxicity. Importantly, both agents promote FOXO nuclear translocation without interfering with the nuclear export machinery and induce the transcription of FOXO target genes. Our results highlight the great potential of this approach for drug repurposing and suggest that harmine and piperlongumine or similar compounds might be useful in the clinic to overcome TRIB2-mediated therapy resistance in cancer patients.

11.
Front Pharmacol ; 11: 625, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32574233

RESUMEN

The transport through the nuclear pore complex is used by cancer cells to evade tumor-suppressive mechanisms. Several tumor-suppressors have been shown to be excluded from the cell nucleus in cancer cells by the nuclear export receptor CRM1 and abnormal expression of CRM1 is oncogenic. Inhibition of CRM1 has long been postulated as potential approach for the treatment of cancer and to overcome therapy resistance. Furthermore, the nuclear export of viral components mediated by the CRM1 is crucial in various stages of the viral lifecycle and assembly of many viruses from diverse families, including coronavirus. However, the first nuclear export inhibitors failed or never entered into clinical trials. More recently CRM1 reemerged as a cancer target and a successful proof of concept was achieved with the clinical approval of Selinexor. The chemical complexity of natural products is a promising perspective for the discovery of new nuclear export inhibitors with a favorable toxicity profile. Several screening campaigns have been performed and several natural product-based nuclear export inhibitors have been identified. With this review we give an overview over the role of CRM1-mediated nuclear export in cancer and the effort made to identify and develop nuclear export inhibitors in particular from natural sources.

12.
J Cell Mol Med ; 24(7): 3766-3778, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32096600

RESUMEN

The recent development of the CRISPR/Cas9 system as an efficient and accessible programmable genome-editing tool has revolutionized basic science research. CRISPR/Cas9 system-based technologies have armed researchers with new powerful tools to unveil the impact of genetics on disease development by enabling the creation of precise cellular and animal models of human diseases. The therapeutic potential of these technologies is tremendous, particularly in gene therapy, in which a patient-specific mutation is genetically corrected in order to treat human diseases that are untreatable with conventional therapies. However, the translation of CRISPR/Cas9 into the clinics will be challenging, since we still need to improve the efficiency, specificity and delivery of this technology. In this review, we focus on several in vitro, in vivo and ex vivo applications of the CRISPR/Cas9 system in human disease-focused research, explore the potential of this technology in translational medicine and discuss some of the major challenges for its future use in patients.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/tendencias , Terapia Genética/tendencias , Investigación Biomédica Traslacional/tendencias , Animales , Humanos , Modelos Animales , Mutación/genética
13.
Methods Mol Biol ; 1890: 103-113, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30414148

RESUMEN

FOXO proteins represent a subfamily of transcription factors that belong to the forkhead family. The study of FOXO target genes can be performed using Real-Time PCR (RT-PCR). The RT-PCR is a sensitive method that allows the detection and quantification of minute amounts of nucleic acids. In RT-PCR the accumulation of the amplicon is detected and measured as the reaction progresses. Here, we describe the application of RT-PCR technique to monitor the transcriptional activity of FOXO transcription factors.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Análisis de Datos , Expresión Génica , Genes Reporteros , Humanos , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Methods Mol Biol ; 1890: 163-170, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30414153

RESUMEN

Forkhead box O (FOXO) factors are tumor suppressor proteins commonly inactivated in human tumors. Furthermore, genetic variation within the FOXO3a gene is consistently associated with human longevity. FOXO proteins are usually inactivated by posttranslational modifications leading to cytoplasmic mislocalization. Therefore, the pharmacological activation by promoting nuclear localization of FOXOs is considered an attractive therapeutic approach to treat cancer and age-related diseases. We developed a cell-based imaging assay to screen for chemical agents capable of inhibiting the nuclear export and in turn trapping proteins that contain a nuclear export sequence including FOXO factors in the nucleus. The fluorescent signal of untreated assay cells localizes predominantly to the cytoplasm. Upon treatment with the nuclear export inhibitors the fluorescent-tagged reporter proteins appear as speckles in the nucleus. In a personalized medicine context, drugs capable of reactivating FOXO factors might be of enormous clinical value in human tumors in which these proteins are inactivated. Here, we describe the procedures for monitoring nuclear export which is suitable for high-throughput screening of compound collections.


Asunto(s)
Núcleo Celular/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Factores de Transcripción Forkhead/metabolismo , Imagen Molecular/métodos , Transporte de Proteínas/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Línea Celular Tumoral , Descubrimiento de Drogas/métodos , Expresión Génica , Genes Reporteros , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
15.
Curr Pharm Des ; 24(5): 549-558, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29366407

RESUMEN

Melanoma is the deadliest form of skin cancer being responsible for 80% of skin cancer deaths. Furthermore, the incidence of metastatic melanoma has increased over the past three decades with a mortality rate that continues to rise faster than most of all other cancers. The last few years have witnessed an unparalleled change in treatment options for patients with metastatic melanoma by the development of new therapeutic strategies like targeted therapies and immunotherapies that highly improved the patient's prognosis. Despite the paradigm- shifting success of these novel treatments, their effectiveness is still limited by intrinsic or acquired resistance. The objective of this review is to provide an overview of the new available treatment modalities, criteria to select patients who might benefit from a specific therapy, mechanisms of innate and acquired resistance to these treatments and to discuss strategies to overcome drug resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia , Melanoma/terapia , Neoplasias Cutáneas/terapia , Humanos , Melanoma/patología , Neoplasias Cutáneas/patología
16.
Medchemcomm ; 8(1): 53-66, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-30108690

RESUMEN

Intrinsic and acquired resistance to conventional and targeted therapeutics is a fundamental reason for treatment failure in many cancer patients. Targeted approaches to overcome chemoresistance as well as resistance to targeted approaches require in depth understanding of the underlying molecular mechanisms. The anti-cancer activity of a drug can be limited by a broad variety of molecular events at different levels of drug action in a cell-autonomous and non-cell-autonomous manner. This review summarizes recent insights into the adaptive mechanisms used by tumours to resist therapy including cellular phenotypic plasticity, dynamic alterations of the tumour microenvironment, activation of redundant signal transduction pathways, modulation of drug target expression levels, and exploitation of pro-survival responses.

17.
PLoS One ; 11(12): e0167491, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27936162

RESUMEN

FOXO factors are tumour suppressor proteins commonly inactivated in human tumours by posttranslational modifications. Furthermore, genetic variation within the FOXO3a gene is consistently associated with human longevity. Therefore, the pharmacological activation of FOXO proteins is considered as an attractive therapeutic approach to treat cancer and age-related diseases. In order to identify agents capable of activating FOXOs, we tested a collection of small chemical compounds using image-based high content screening technology. Here, we report the discovery of LOM612 (compound 1a), a newly synthesized isothiazolonaphthoquinone as a potent FOXO relocator. Compound 1a induces nuclear translocation of a FOXO3a reporter protein as well as endogenous FOXO3a and FOXO1 in U2OS cells in a dose-dependent manner. This activity does not affect the subcellular localization of other cellular proteins including NFkB or inhibit CRM1-mediated nuclear export. Furthermore, compound 1a shows a potent antiproliferative effect in human cancer cell lines.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O3/metabolismo , Naftoquinonas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Tiazoles/farmacología , Línea Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Citoplasma/metabolismo , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Células MCF-7 , Microscopía Confocal , Modelos Químicos , Estructura Molecular , Naftoquinonas/química , Transporte de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Tiazoles/química
18.
Cancer J ; 21(6): 441-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26588674

RESUMEN

All drugs have molecular targets; however, this does not mean that they are targeted therapeutics. Only by the interaction with a disease-specific molecule can the drug be classified as a targeted therapeutic. This is often not clearly defined and might refer to several different therapeutic modalities such as genomically targeted therapy, immune checkpoint therapy, or pharmacokinetic targeting. To develop a precise concept of targeted therapy, it is crucial to understand how drugs were discovered and how our rapidly expanding knowledge concerning disease mechanism is driving a fundamental conceptual change in modern drug discovery and development. In combination with the increasingly detailed analysis of disease at an individual patient level, we believe that it is very timely to consider the past and current approaches involved in the development of new medicines and to discuss the paradigm shift in and basic concepts associated with targeted therapies and personalized medicine.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Medicina de Precisión , Diseño de Fármacos , Humanos
19.
Cancer Genet ; 205(4): 138-46, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22559974

RESUMEN

The estrogen receptor (ER) is a well-known predictor of breast cancer response to endocrine therapy. ER+ progesterone receptor (PR)- breast tumors have a poorer response to endocrine therapy and a more aggressive phenotype than ER+PR+ tumors. A comparative genomic hybridization array technique was used to examine 25 ER+PR+ and 23 ER+PR- tumors. Tissue microarrays composed of 50 ER+PR+ and 50 ER+PR- tumors were developed to validate the comparative genomic hybridization array results. The genes of interest were analyzed by fluorescence in situ hybridization. The ER+PR- group had a slightly different genomic profile when compared with ER+PR+ tumors. Chromosomes 17 and 20 contained the most overlapping gains, and chromosomes 3, 8, 9, 14, 17, 21, and 22 contained the most overlapping losses when compared with the ER+PR+ group. The gained regions, 17q23.2-q23.3 and 20q13.12, and the lost regions, 3p21.32-p12.3, 9pter-p13.2, 17pter-p12, and 21pter-q21.1, occurred at different alteration frequencies and were statistically significant in the ER+PR- tumors compared with the ER+PR+ tumors. ER+PR- breast tumors have a different genomic profile compared with ER+PR+ tumors. Differentially lost regions in the ER+PR- group included genes with tumor suppressor functions and genes involved in apoptosis, mitosis, angiogenesis, and cell spreading. Differentially gained regions included genes such as MAP3K3, RPS6KB1, and ZNF217. Amplification of these genes could contribute to resistance to apoptosis, increased activation of the PI3K/Akt/mTOR pathway, and the loss of PR in at least some ER+PR- tumors.


Asunto(s)
Neoplasias de la Mama/genética , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Anciano , Apoptosis/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Variaciones en el Número de Copia de ADN , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Hibridación Fluorescente in Situ , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Estrógenos/biosíntesis , Receptores de Progesterona/biosíntesis , Eliminación de Secuencia
20.
Haematologica ; 97(4): 534-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22102704

RESUMEN

The disruption of RUNX1 function is one of the main mechanisms of disease observed in hematopoietic malignancies and the description of novel genetic events that lead to a RUNX1 loss of function has been accelerated with the development of genomic technologies. Here we describe the molecular characterization of a new t(4;21)(q21;q22) in a de novo myelodysplastic syndrome that resulted in the deletion of the RUNX1 gene. We demonstrated by quantitative real-time RT-PCR an almost complete depletion of the expression of the RUNX1 gene in our t(4;21) case compared with CD34(+) cells that was independent of mutation or DNA methylation. More importantly, we explored and confirmed the possibility that this abrogation also prevented transactivation of RUNX1 target genes, perhaps confirming the genetic origin of the thrombocytopenia and the myelodysplastic features observed in our patient, and certainly mimicking what has been observed in the presence of the RUNX1/ETO fusion protein.


Asunto(s)
Expresión Génica , Síndromes Mielodisplásicos/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Translocación Genética , Bandeo Cromosómico , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 4 , Humanos , Masculino , Persona de Mediana Edad , Proteína 1 Compañera de Translocación de RUNX1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...