Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Oncologist ; 27(11): 940-951, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-35983951

RESUMEN

INTRODUCTION: Neuroendocrine neoplasms (NEN) are heterogeneous malignancies that can arise at almost any anatomical site and are classified as biologically distinct well-differentiated neuroendocrine tumors (NET) and poorly differentiated neuroendocrine carcinomas (NEC). Current systemic therapies for advanced disease, including targeted therapies, chemotherapy, and immunotherapy, are associated with limited duration of response. New therapeutic targets are needed. One promising target is delta-like ligand 3 (DLL3), an inhibitory ligand of the Notch receptor whose overexpression on the surface of NEN is associated with tumorigenesis. METHODS: This article is a narrative review that highlights the role of DLL3 in NEN progression and prognosis, the potential for therapeutic targeting of DLL3, and ongoing studies of DLL3-targeting therapies. Classification, incidence, pathogenesis, and current management of NEN are reviewed to provide biological context and illustrate the unmet clinical needs. DISCUSSION: DLL3 is overexpressed in many NENs, implicated in tumor progression, and is typically associated with poor clinical outcomes, particularly in patients with NEC. Targeted therapies using DLL3 as a homing beacon for cytotoxic activity mediated via several different mechanisms (eg, antibody-drug conjugates, T-cell engager molecules, CAR-Ts) have shown promising clinical activity in small-cell lung cancer (SCLC). DLL3 may be a clinically actionable target across NEN. CONCLUSIONS: Current treatment options for NEN do not provide sustained responses. DLL3 is expressed on the cell surface of many NEN types and is associated with poor clinical outcomes. Initial clinical studies targeting DLL3 therapeutically in SCLC have been promising, and additional studies are expanding this approach to the broader group of NEN.


Asunto(s)
Carcinoma Neuroendocrino , Neoplasias Pulmonares , Tumores Neuroendocrinos , Carcinoma Pulmonar de Células Pequeñas , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ligandos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Carcinoma Neuroendocrino/patología
2.
Am J Physiol Endocrinol Metab ; 307(1): E84-92, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24824656

RESUMEN

Menin, the product of the MEN1 gene, functions as a tumor suppressor and was first identified in 1997 due to its causative role in the endocrine tumor disorder multiple endocrine neoplasia, type 1 (MEN1). More recently, menin has been identified as a key player in pancreatic islet biology with the observation of an inverse relationship between menin levels and pancreatic islet proliferation. However, the factors regulating menin and the MEN1 gene in the pancreas are poorly understood. Here, we describe the regulation of menin by miR-24 and demonstrate that miR-24 directly decreases menin levels and impacts downstream cell cycle inhibitors in MIN6 insulinoma cells and in ßlox5 immortalized ß-cells. This regulation of menin impacts cell viability and proliferation in ßlox5 cells. Furthermore, our data show a feedback regulation between miR-24 and menin that is present in the pancreas, suggesting that miR-24 regulates menin levels in the pancreatic islet.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiopatología , MicroARNs/fisiología , Páncreas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Animales , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Ratones , Ratones Noqueados
3.
Front Immunol ; 15: 1357731, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38784379

RESUMEN

Long-term immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires the identification of T-cell epitopes affecting host immunogenicity. In this computational study, we explored the CD8+ epitope diversity estimated in 27 of the most common HLA-A and HLA-B alleles, representing most of the United States population. Analysis of 16 SARS-CoV-2 variants [B.1, Alpha (B.1.1.7), five Delta (AY.100, AY.25, AY.3, AY.3.1, AY.44), and nine Omicron (BA.1, BA.1.1, BA.2, BA.4, BA.5, BQ.1, BQ.1.1, XBB.1, XBB.1.5)] in analyzed MHC class I alleles revealed that SARS-CoV-2 CD8+ epitope conservation was estimated at 87.6%-96.5% in spike (S), 92.5%-99.6% in membrane (M), and 94.6%-99% in nucleocapsid (N). As the virus mutated, an increasing proportion of S epitopes experienced reduced predicted binding affinity: 70% of Omicron BQ.1-XBB.1.5 S epitopes experienced decreased predicted binding, as compared with ~3% and ~15% in the earlier strains Delta AY.100-AY.44 and Omicron BA.1-BA.5, respectively. Additionally, we identified several novel candidate HLA alleles that may be more susceptible to severe disease, notably HLA-A*32:01, HLA-A*26:01, and HLA-B*53:01, and relatively protected from disease, such as HLA-A*31:01, HLA-B*40:01, HLA-B*44:03, and HLA-B*57:01. Our findings support the hypothesis that viral genetic variation affecting CD8 T-cell epitope immunogenicity contributes to determining the clinical severity of acute COVID-19. Achieving long-term COVID-19 immunity will require an understanding of the relationship between T cells, SARS-CoV-2 variants, and host MHC class I genetics. This project is one of the first to explore the SARS-CoV-2 CD8+ epitope diversity that putatively impacts much of the United States population.


Asunto(s)
COVID-19 , Biología Computacional , Epítopos de Linfocito T , SARS-CoV-2 , Humanos , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/genética , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/inmunología , COVID-19/virología , Estados Unidos/epidemiología , Biología Computacional/métodos , Linfocitos T CD8-positivos/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Alelos , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Índice de Severidad de la Enfermedad , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética
4.
Front Oncol ; 12: 901435, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35747820

RESUMEN

Gastroenteropancreatic neuroendocrine neoplasms are a rare, diverse group of neuroendocrine tumors that form in the pancreatic and gastrointestinal tract, and often present with side effects due to hormone hypersecretion. The pathogenesis of these tumors is known to be linked to several genetic disorders, but sporadic tumors occur due to dysregulation of additional genes that regulate proliferation and metastasis, but also the epigenome. Epigenetic regulation in these tumors includes DNA methylation, chromatin remodeling and regulation by noncoding RNAs. Several large studies demonstrate the identification of epigenetic signatures that may serve as biomarkers, and others identify innovative, epigenetics-based targets that utilize both pharmacological and theranostic approaches towards the development of new treatment approaches.

5.
Prog Mol Biol Transl Sci ; 190(1): 39-56, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36008001

RESUMEN

In the past two decades since the first sequencing of the human genome, clinical genomics has undergone a renaissance with the advent of next generation sequencing (NGS) technologies. Whole genome sequencing is now increasingly available in the clinical setting at a fraction of the cost of the human genome reference sequence, and with turnaround times as short as 7-10days. Clinical genetics-based management has become more tractable with improved payer reimbursement and increased availability of targeted therapies, especially in the oncology space. Precision diagnostics in cancer allow clinicians to assess a patient's risk of developing cancers, detect and classify tumors, predict prognoses, select targeted therapies when available, and monitor their patient's disease burden longitudinally. This approach ushers in a new era of predictive and preventive oncology care that can be personalized to the genetics of the individual patient, the patient's tumor, and their clinical presentation. Within oncology, there are global collaborative efforts including the International Cancer Genome Consortium (ICGC), Pan-Cancer Analysis of Whole Genomes (PCAWG) and The Cancer Genome Atlas (TCGA) that have archived thousands of cancer genomes. In collaboration with databases including but not limited to cancer cataloging variant association with disease risk (ClinVar and others) and drug responses (PharmGKB and others), the study of somatic and germline genomic variation provides a wealth of information on cancer development, evolution, heterogeneity, and treatment response. Although many challenges still exist for defining all possible actionable genomic variants, we have unprecedented opportunity to gain mechanistic and clinical insights into human neoplastic diseases.


Asunto(s)
Neoplasias , Genoma Humano , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/terapia
6.
Exp Biol Med (Maywood) ; 247(10): 842-847, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35130743

RESUMEN

Cytokine-driven hyper inflammation has been identified as a critical factor behind poor outcomes in patients severely infected with SARS-CoV-2 virus. Notably, protein ISGylation, a protein conjugated form of Type 1 IFN-inducible ubiquitin-like protein ISG15 (Interferon-Stimulated Gene 15), induces cytokine storm (CS) and augments colonic inflammation in colitis-associated colon cancers in mouse models. However, whether ISGylation is increased and causally responsible for CS and hyper inflammation in symptomatic COVID-19 patients is unknown. Here, we measured ISGylation levels in peripheral blood mononuclear cells (PBMCs) from 10 symptomatic (SARS-CoV-2-positive with symptoms) and asymptomatic (SARS-CoV-2-positive with no symptoms) COVID-19 patients, and 4 uninfected individuals (SARS-CoV-2-negative), using WesTm assay. Strikingly, we note significant increases in protein ISGylation and MX-1 (myxovirus-resistance protein-1) protein levels, both induced by type-I IFN, in symptomatic but not in asymptomatic patients and uninfected individuals. Knowing that ISGylation augments CS and intestinal inflammation in colon cancers, we propose that increased ISGylation may be an underlying cause of CS and inflammation in symptomatic patients.


Asunto(s)
COVID-19 , Ubiquitinas , Animales , Citocinas/metabolismo , Humanos , Inflamación , Leucocitos Mononucleares/metabolismo , Ratones , SARS-CoV-2 , Ubiquitinas/metabolismo
7.
Methods Mol Biol ; 2422: 233-245, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34859410

RESUMEN

Mammosphere formation assays are a popular and convenient technique in the study of breast cancer by providing an in vitro mechanism by which to study breast cancer stem cell (BCSC) contribution to tumorigenesis, as well as more closely mimicking the three-dimensional tumor microenvironment. In these assays, BCSCs are stimulated to proliferate in low adherence tissue culture dishes and the resulting mammospheres exhibit activation of stem cell-related signaling pathways. Here we describe the process for generating and analyzing mammospheres under varying conditions.


Asunto(s)
Neoplasias de la Mama , Mama , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Células Madre Neoplásicas , Receptores de Estrógenos , Microambiente Tumoral
8.
Nat Rev Drug Discov ; 20(2): 125-144, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33293690

RESUMEN

Notch signalling is involved in many aspects of cancer biology, including angiogenesis, tumour immunity and the maintenance of cancer stem-like cells. In addition, Notch can function as an oncogene and a tumour suppressor in different cancers and in different cell populations within the same tumour. Despite promising preclinical results and early-phase clinical trials, the goal of developing safe, effective, tumour-selective Notch-targeting agents for clinical use remains elusive. However, our continually improving understanding of Notch signalling in specific cancers, individual cancer cases and different cell populations, as well as crosstalk between pathways, is aiding the discovery and development of novel investigational Notch-targeted therapeutics.


Asunto(s)
Antineoplásicos/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/metabolismo , Receptores Notch/metabolismo , Animales , Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Inmunoterapia/métodos , Inmunoterapia/tendencias , Neoplasias/inmunología , Neoplasias/terapia , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Receptores Notch/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
9.
Mol Cell Endocrinol ; 287(1-2): 40-6, 2008 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-18367319

RESUMEN

Selective estrogen receptor modulators (SERMs) have the unique potential to provide estrogenic effects in the skeletal and cardiovascular system, while minimizing/eliminating side effects on reproductive organs. However, despite the unifying characteristic of mixed estrogen receptor (ER) agonist/antagonist activity, compounds within this class are not interchangeable. In order to define and compare the effects of SERMs on different hormone-responsive tissues, we evaluated effects of bazedoxifene acetate (BZA), lasofoxifene (LAS) and raloxifene (RAL) in the mammary gland and uterus of the ovariectomized mouse. Endpoints measured included those regulated by estradiol alone (uterine wet weight, uterine G protein-coupled receptor 105 (GPR105) mRNA expression and mammary gland indoleamine-pyrrole 2,3 dioxygenase (INDO) mRNA expression) as well as others that required the combination of estradiol and progesterone (uterine serine protease inhibitor Kazal type 3 (Spink3) mRNA expression, mammary gland morphology and mammary gland defensin beta1 (Defbeta1) mRNA expression). The three SERMs tested had variable agonist and antagonist activity on these endpoints. In the uterus, the SERMs were mixed agonists/antagonists on estradiol-induced wet weight increase, whereas all three SERMs were estrogen receptor antagonists on GPR105 mRNA expression. However, in the presence of progesterone, BZA and RAL were agonists on Spink3 expression, while LAS was primarily an antagonist. In the mammary gland, BZA and RAL were predominantly agonists on the endpoint of mammary morphology and all three SERMs were clear agonists on Defbeta1 mRNA expression, an E+P-dependent marker. Finally, LAS and RAL had mixed agonist/antagonist activity on INDO mRNA expression, while BZA had only antagonist activity. These results demonstrate that compounds with small structural differences can elicit distinct biological responses, and that in general, SERMs tended to behave more as antagonists on endpoints requiring estrogen alone and agonists on endpoints requiring the combination of estrogen and progesterone.


Asunto(s)
Estradiol/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Progesterona/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Útero/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glándulas Mamarias Animales/citología , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Proteínas de Secreción Prostática/genética , Proteínas de Secreción Prostática/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y , Inhibidor de Tripsina Pancreática de Kazal , Útero/citología , beta-Defensinas/genética , beta-Defensinas/metabolismo
10.
Biomedicines ; 6(3)2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018256

RESUMEN

Breast cancer stem cells (BCSC) have been implicated in tumor initiation, progression, metastasis, recurrence, and resistance to therapy. The origins of BCSCs remain controversial due to tumor heterogeneity and the presence of such small side populations for study, but nonetheless, cell surface markers and their correlation with BCSC functionality continue to be identified. BCSCs are driven by persistent activation of developmental pathways, such as Notch, Wnt, Hippo, and Hedgehog and new treatment strategies that are aimed at these pathways are in preclinical and clinical development.

11.
Mol Cell Biol ; 23(2): 493-509, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12509449

RESUMEN

Menin is a 70-kDa protein encoded by MEN1, the tumor suppressor gene disrupted in multiple endocrine neoplasia type 1. In a yeast two-hybrid system based on reconstitution of Ras signaling, menin was found to interact with the 32-kDa subunit (RPA2) of replication protein A (RPA), a heterotrimeric protein required for DNA replication, recombination, and repair. The menin-RPA2 interaction was confirmed in a conventional yeast two-hybrid system and by direct interaction between purified proteins. Menin-RPA2 binding was inhibited by a number of menin missense mutations found in individuals with multiple endocrine neoplasia type 1, and the interacting regions were mapped to the N-terminal portion of menin and amino acids 43 to 171 of RPA2. This region of RPA2 contains a weak single-stranded DNA-binding domain, but menin had no detectable effect on RPA-DNA binding in vitro. Menin bound preferentially in vitro to free RPA2 rather than the RPA heterotrimer or a subcomplex consisting of RPA2 bound to the 14-kDa subunit (RPA3). However, the 70-kDa subunit (RPA1) was coprecipitated from HeLa cell extracts along with RPA2 by menin-specific antibodies, suggesting that menin binds to the RPA heterotrimer or a novel RPA1-RPA2-containing complex in vivo. This finding was consistent with the extensive overlap in the nuclear localization patterns of endogenous menin, RPA2, and RPA1 observed by immunofluorescence.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas , Animales , Proteínas Bacterianas/metabolismo , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Cromatografía en Gel , ADN/metabolismo , Daño del ADN , ADN Complementario/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Fase G1 , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Ratones , Microscopía Fluorescente , Neoplasia Endocrina Múltiple/genética , Mutación Missense , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Proteína de Replicación A , Fase S , Transfección , Técnicas del Sistema de Dos Híbridos
12.
Mol Cell Biol ; 23(17): 6075-85, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12917331

RESUMEN

Patients with multiple endocrine neoplasia type 1 (MEN1) develop multiple endocrine tumors, primarily affecting the parathyroid, pituitary, and endocrine pancreas, due to the inactivation of the MEN1 gene. A conditional mouse model was developed to evaluate the loss of the mouse homolog, Men1, in the pancreatic beta cell. Men1 in these mice contains exons 3 to 8 flanked by loxP sites, such that, when the mice are crossed to transgenic mice expressing cre from the rat insulin promoter (RIP-cre), exons 3 to 8 are deleted in beta cells. By 60 weeks of age, >80% of mice homozygous for the floxed Men1 gene and expressing RIP-cre develop multiple pancreatic islet adenomas. The formation of adenomas results in elevated serum insulin levels and decreased blood glucose levels. The delay in tumor appearance, even with early loss of both copies of Men1, implies that additional somatic events are required for adenoma formation in beta cells. Comparative genomic hybridization of beta cell tumor DNA from these mice reveals duplication of chromosome 11, potentially revealing regions of interest with respect to tumorigenesis.


Asunto(s)
Adenoma/genética , Insulinoma/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas , Adenoma/metabolismo , Adenoma/patología , Animales , Glucemia/metabolismo , División Celular/genética , Células Cultivadas , Supervivencia sin Enfermedad , Ingeniería Genética/métodos , Heterocigoto , Homocigoto , Hiperplasia/genética , Hibridación in Situ/métodos , Insulina/genética , Insulina/metabolismo , Insulinoma/metabolismo , Insulinoma/patología , Integrasas/genética , Islotes Pancreáticos/patología , Islotes Pancreáticos/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Hipófisis/patología , Regiones Promotoras Genéticas , Eliminación de Secuencia , Proteínas Virales/genética
13.
Maturitas ; 58(4): 366-76, 2007 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-17997058

RESUMEN

BACKGROUND: Vaginal atrophy (VA) is a prevalent disorder in postmenopausal women that is characterized by decreased epithelial thickness, reduced vaginal maturation index (VMI) and increased vaginal pH. Current medical therapy consists of local or systemic replacement of estrogens. OBJECTIVE: The goal of this study was to understand, at a molecular level, the effect of estradiol (E2) on the vaginal epithelium. METHODS: Nineteen women were treated with E2 delivered through a skin patch at a dose of 0.05mg/day for 12 weeks. The diagnosis of VA was confirmed by a VMI with < or =5% superficial cells and vaginal pH>5.0. Vaginal biopsy samples were collected at baseline and after treatment. Differentially expressed mRNA transcripts in these biopsies were determined by microarray analysis. RESULTS: All 19 subjects had increased VMI (>5%) and/or reduced pH (< or =5) following treatment. Most subjects also had increased serum E2 levels and reduced serum FSH levels. Transcriptional profiling of vaginal biopsies identified over 3000 E2-regulated genes, including those involved in several key pathways known to regulate cell growth and proliferation, barrier function and pathogen defense. CONCLUSIONS: E2 controls a plethora of cellular pathways that are concordant with its profound effect on vaginal physiology. The data presented here are a useful step toward understanding the role of E2 in vaginal tissue and the development of novel therapeutics for the treatment of VA.


Asunto(s)
Climaterio/genética , Estradiol/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Vagina/patología , Administración Cutánea , Adulto , Anciano , Atrofia , Biopsia , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Climaterio/efectos de los fármacos , Proteínas Ricas en Prolina del Estrato Córneo , Desmogleína 1/genética , Epitelio/efectos de los fármacos , Epitelio/patología , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Humanos , Concentración de Iones de Hidrógeno , Metaloproteinasa 10 de la Matriz/genética , Proteínas de la Membrana/genética , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Receptores CXCR6 , Receptores de Quimiocina , Receptores Virales , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/efectos de los fármacos , Vagina/efectos de los fármacos , Vagina/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-29255447

RESUMEN

The molecular events leading to gastroenteropancreatic neuroendocrine tumor (GEP-NET) formation are largely unknown. Over the past decades, systemic chemotherapies have been replaced by therapies directed at particular molecular targets such as the somatostatin receptors, mTOR complexes or proangiogenic molecules. These approaches have demonstrated some success in subtypes of this heterogeneous tumor group, but responses are still widely varied. This review highlights the clinical trials ongoing for neuroendocrine tumors (NETs) and includes emerging immunotherapy, which holds great promise for NETs based on successes in other tumor types. Current avenues of preclinical research, including Notch and PI3K/AKT, will lead to additional targeted therapies based on genome-wide studies that have cast a wide net in the search for driver mutations. Future preclinical and clinical investigations are required to identify those mutations predictive of therapeutic response or disease progression. Results of current clinical trials outlined here will better inform patient management with respect to agent selection, timing, duration and combination therapy in the treatment of NETs.

15.
Oncotarget ; 8(50): 87054-87072, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29152063

RESUMEN

The differentiation status of tumors is used as a prognostic indicator, with tumors comprised of less differentiated cells exhibiting higher levels of aggressiveness that correlate with a poor prognosis. Although oncogenes contribute to blocking differentiation, it is not clear how they globally alter miRNA expression during differentiation to achieve this result. The pediatric sarcoma Alveolar Rhabdomyosarcoma, which is primarily characterized by the expression of the PAX3-FOXO1 oncogenic fusion protein, consists of undifferentiated muscle cells. However, it is unclear what role PAX3-FOXO1 plays in promoting the undifferentiated state. We demonstrate that expression of PAX3-FOXO1 globally alters the expression of over 80 individual miRNA during early myogenic differentiation, resulting in three primary effects: 1) inhibition of the expression of 51 miRNA essential for promoting myogenesis, 2) promoting the aberrant expression of 43 miRNA not normally expressed during myogenesis, and 3) altering the expression pattern of 39 additional miRNA. Combined, these changes are predicted to have an overall negative effect on myogenic differentiation. This is one of the first studies describing how an oncogene globally alters miRNA expression to block differentiation and has clinical implications for the development of much needed multi-faceted tumor-specific therapeutic regimens.

16.
J Steroid Biochem Mol Biol ; 101(1): 11-21, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16920353

RESUMEN

We have identified mRNA markers of estradiol and progesterone action in the mouse mammary gland and uterus to establish an in vivo model for the evaluation of novel and potentially tissue selective estrogens and progestins. Gene chip analysis of mRNA from ovariectomized (OVX) mice treated with vehicle (V), 17beta-estradiol (E2), progesterone (P) or E2+P for 7 days identified defensinbeta1 (Defbeta1) and indoleamine-pyrrole 2,3 dioxygenase (INDO) as markers of E2 and P action in the mammary gland, and serine protease inhibitor, Kazal type 3 (Spink3) and G protein-coupled receptor 105 (GPR105) as markers in the uterus. Defbeta1 and Spink3 are both upregulated by E2+P, whereas INDO and GPR105 have a complementary profile of upregulation by E2 alone and suppression of the E2 effect by P. Quantitative RT-PCR analysis of mammary gland markers was concordant with histological changes. Using this model, medroxyprogesterone acetate (MPA) and tanaproget (TNPR), a novel nonsteroidal progesterone receptor agonist, were evaluated and found to have no marked tissue selectivity relative to progesterone. In addition, the ERalpha selective ligand propyl pyrazole triol (PPT) and the ERbeta selective ligands ERB-041 and WAY-202196 were evaluated on the mammary gland endpoints of histology and Defbeta1 mRNA expression, and showed that ERalpha stimulation is necessary and sufficient for eliciting estradiol-mediated changes in the mammary gland.


Asunto(s)
Estrógenos/fisiología , Regulación de la Expresión Génica , Marcadores Genéticos , Glándulas Mamarias Animales/metabolismo , Modelos Animales , Progesterona/fisiología , Útero/metabolismo , Animales , Secuencia de Bases , Benzoxazinas/farmacología , Cartilla de ADN , Femenino , Perfilación de la Expresión Génica , Acetato de Medroxiprogesterona/farmacología , Ratones , Ratones Endogámicos C57BL , Naftoles/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxazoles/farmacología , Fenoles , Pirazoles/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tionas/farmacología
17.
Cancer Res ; 63(22): 8022-8, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14633735

RESUMEN

The inactivation of the MEN1 tumor suppressor gene in patients leads to a constellation of changes in endocrine tissues, including parathyroid neoplasia, pituitary adenomas, pancreatic neuroendocrine tumors, and carcinoids. To study the pathophysiological consequences of the deletion of the MEN1 gene, we set out to create a mouse model of hyperparathyroidism resulting from the deletion of the Men1 gene in parathyroid tissue. We introduced a Men1 gene flanked by loxP sites into the mouse germ line and then used a parathyroid cell-specific promoter to drive the expression of Cre recombinase, resulting in the deletion of the Men1 gene. Here, we show that loss of Men1 gene function in the parathyroid glands of mice results in histological changes consistent with parathyroid neoplasia as well as systemic hypercalcemia. This model provides a means for dissecting the molecular basis of this familial cancer syndrome and may allow for the development of new strategies to treat related forms of hypercalcemia.


Asunto(s)
Hipercalcemia/genética , Hiperparatiroidismo/genética , Neoplasias de las Paratiroides/genética , Proteínas Proto-Oncogénicas/genética , Animales , Calcio/sangre , Cruzamientos Genéticos , Femenino , Eliminación de Gen , Humanos , Hipercalcemia/sangre , Hiperparatiroidismo/sangre , Integrasas/genética , Masculino , Ratones , Ratones Transgénicos , Neoplasias de las Paratiroides/sangre , Proteínas Virales/genética
18.
Front Oncol ; 6: 94, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27148486

RESUMEN

Carcinoids and neuroendocrine tumors (NETs) are a heterogeneous group of tumors that arise from the neuroendocrine cells of the GI tract, endocrine pancreas, and the respiratory system. NETs remain significantly understudied with respect to molecular mechanisms of pathogenesis, particularly the role of cell fate signaling systems such as Notch. The abundance of literature on the Notch pathway is a testament to its complexity in different cellular environments. Notch receptors can function as oncogenes in some contexts and tumor suppressors in others. The genetic heterogeneity of NETs suggests that to fully understand the roles and the potential therapeutic implications of Notch signaling in NETs, a comprehensive analysis of Notch expression patterns and potential roles across all NET subtypes is required.

19.
Oncotarget ; 7(39): 62814-62835, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27588498

RESUMEN

While many solid tumors are defined by the presence of a particular oncogene, the role that this oncogene plays in driving transformation through the acquisition of aneuploidy and overcoming growth arrest are often not known. Further, although aneuploidy is present in many solid tumors, it is not clear whether it is the cause or effect of malignant transformation. The childhood sarcoma, Alveolar Rhabdomyosarcoma (ARMS), is primarily defined by the t(2;13)(q35;q14) translocation, creating the PAX3-FOXO1 fusion protein. It is unclear what role PAX3-FOXO1 plays in the initial stages of tumor development through the acquisition and persistence of aneuploidy. In this study we demonstrate that PAX3-FOXO1 serves as a driver mutation to initiate a cascade of mRNA and miRNA changes that ultimately reprogram proliferating myoblasts to induce the formation of ARMS. We present evidence that cells containing PAX3-FOXO1 have changes in the expression of mRNA and miRNA essential for maintaining proper chromosome number and structure thereby promoting aneuploidy. Further, we demonstrate that the presence of PAX3-FOXO1 alters the expression of growth factor related mRNA and miRNA, thereby overriding aneuploid-dependent growth arrest. Finally, we present evidence that phosphorylation of PAX3-FOXO1 contributes to these changes. This is one of the first studies describing how an oncogene and post-translational modifications drive the development of a tumor through the acquisition and persistence of aneuploidy. This mechanism has implications for other solid tumors where large-scale genomics studies may elucidate how global alterations contribute to tumor phenotypes allowing the development of much needed multi-faceted tumor-specific therapeutic regimens.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Mutación , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX3/metabolismo , Rabdomiosarcoma Alveolar/genética , Aneuploidia , Animales , Ciclo Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Aberraciones Cromosómicas , Progresión de la Enfermedad , Proteína Forkhead Box O1/genética , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Mitosis , Desarrollo de Músculos , Mioblastos/metabolismo , Factor de Transcripción PAX3/genética , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Rabdomiosarcoma Alveolar/metabolismo , Translocación Genética
20.
PLoS One ; 10(12): e0145792, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26709830

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is a genetic disorder characterized by tissue-specific tumors in the endocrine pancreas, parathyroid, and pituitary glands. Although tumor development in these tissues is dependent upon genetic inactivation of the tumor suppressor Men1, loss of both alleles of this gene is not sufficient to induce these cancers. Men1 encodes menin, a nuclear protein that influences transcription. A previous ChIP on chip analysis suggested that menin binds promoter sequences of nol3, encoding ARC, which is a cell death inhibitor that has been implicated in cancer pathogenesis. We hypothesized that ARC functions as a co-factor with Men1 loss to induce the tissue-restricted distribution of tumors seen in MEN1. Using mouse models that recapitulate this syndrome, we found that biallelic deletion of Men1 results in selective induction of ARC expression in tissues that develop tumors. Specifically, loss of Men1 in all cells of the pancreas resulted in marked increases in ARC mRNA and protein in the endocrine, but not exocrine, pancreas. Similarly, ARC expression increased in the parathyroid with inactivation of Men1 in that tissue. To test if ARC contributes to MEN1 tumor development in the endocrine pancreas, we generated mice that lacked none, one, or both copies of ARC in the context of Men1 deletion. Studies in a cohort of 126 mice demonstrated that, although mice lacking Men1 developed insulinomas as expected, elimination of ARC in this context did not significantly alter tumor load. Cellular rates of proliferation and death in these tumors were also not perturbed in the absence of ARC. These results indicate that ARC is upregulated by loss Men1 in the tissue-restricted distribution of MEN1 tumors, but that ARC is not required for tumor development in this syndrome.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Musculares/biosíntesis , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Femenino , Eliminación de Gen , Genes Supresores de Tumor , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA