Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
JAAPA ; 35(10): 48-52, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36165549

RESUMEN

ABSTRACT: The field of cancer genetic testing has made great advances in correctly identifying patients at risk for hereditary cancer syndromes. These tests, used during a genetic consultation, promote earlier detection of cancer and potentially increase survival. Despite these advancements, a large number of persons at risk for BRCA1/2 mutations remain untested due to a shortage of genetic counselors and lack of clinician knowledge on how to properly screen, identify, and refer patients to genetic counseling. We suggest team-based practices that physician associates/assistants (PAs) and NPs can use in collaboration with genetic counselors. We also explore how PAs and NPs can alleviate the burden on genetic counselors by taking a comprehensive family history, providing elements of counseling, and ordering appropriate genetic tests. This approach maximizes the amount of time the patient spends receiving actual genetic counseling. By creating this collaborative relationship, PAs and NPs can help increase the number of qualifying patients who receive genetic testing and counseling for hereditary breast and ovarian cancer syndromes.


Asunto(s)
Neoplasias de la Mama , Síndromes Neoplásicos Hereditarios , Neoplasias Ováricas , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Consejo , Femenino , Asesoramiento Genético , Pruebas Genéticas , Humanos , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/genética , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/genética
2.
Sci Rep ; 11(1): 291, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431926

RESUMEN

The CLOVES syndrome is an overgrowth disease arising from mosaic activating somatic mutations in the PIK3CA gene. These mutations occur during fetal development producing malformation and overgrowth of a variety of tissues. It has recently been shown that treatment with low doses of a selective inhibitor of Class I PI3K catalytic subunit p110α, the protein product of the PIK3CA gene, can yield dramatic therapeutic benefits for patients with CLOVES and PROS (a spectrum of PIK3CA-related overgrowth syndromes). To assess the long-term effects of moderate loses of p110α activity, we followed development and growth of mice with heterozygous loss of p110α (Pik3ca+/-) over their entire lifetimes, paying particular attention to effects on the brain. While homozygous deletion of the Pik3ca gene is known to result in early embryonic lethality, these Pik3ca+/- mice displayed a longer lifespan compared to their wild-type littermates. These mice appeared normal, exhibited no obvious behavioral abnormalities, and no body weight changes. However, their brains showed a significant reduction in size and weight. Notably, mice featuring deletion of one allele of Pik3ca only in the brain also showed gradually reduced brain size and weight. Mechanistically, either deletion of p110α or pharmacological inhibition of p110α activity reduced neurosphere size, but not numbers, in vitro, suggesting that p110α activity is critical for neuronal stem cells. The phenotypes observed in our two genetically engineered mouse models suggest that the sustained pharmacological inhibition of the PIK3CA activity in human patients might have both beneficial and harmful effects, and future treatments may need to be deployed in a way to avoid or minimize adverse effects.


Asunto(s)
Envejecimiento/genética , Encéfalo/crecimiento & desarrollo , Fosfatidilinositol 3-Quinasa Clase I/genética , Animales , Ratones , Mutación , Fenotipo
3.
Stem Cell Reports ; 13(3): 545-558, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31257132

RESUMEN

Bone marrow stromal cells (BMSCs) that express high levels of stem cell factor (SCF) and CXC chemokine ligand 12 (CXCL12) are one crucial component of the hematopoietic stem cell (HSC) niche. While the secreted factors produced by BMSCs to support HSCs have been well described, little is known regarding the transcriptional regulators controlling the cell fate of BMSCs and thus indirectly maintaining HSCs. BMI1 is a polycomb group protein that regulates HSCs both cell intrinsically and extrinsically, but it is unknown in which cell type and how BMI1 functions to maintain HSCs extrinsically. Here we show that Bmi1 maintains HSCs by preventing adipogenic differentiation of BMSCs. Bmi1 is highly expressed in BMSCs but becomes downregulated upon adipogenic differentiation and during aging. Deleting Bmi1 from BMSCs increased marrow adipocytes, induced HSC quiescence and depletion, and impaired hematopoiesis. We found that BMI1 repressed multiple developmental programs in BMSCs by safeguarding the repressive epigenetic marks histone H2A ubiquitylation and H3 lysine 27 trimethylation. We identified a novel adipogenic program governed by Pax3, which BMI1 repressed in BMSCs. Our results establish Bmi1 as a critical regulator of BMSC cell fate that suppresses marrow adipogenesis to create a supportive niche for HSCs.


Asunto(s)
Adipocitos/metabolismo , Células Madre Mesenquimatosas/citología , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Adipocitos/citología , Adipogénesis , Animales , Diferenciación Celular , Senescencia Celular , Femenino , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Metilación , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/metabolismo , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Nicho de Células Madre
4.
Exp Hematol ; 64: 33-44.e5, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29751067

RESUMEN

Recent advances in next-generation sequencing have identified novel mutations and revealed complex genetic architectures in human hematological malignancies. Moving forward, new methods to quickly generate animal models that recapitulate the complex genetics of human hematological disorders are needed to transform the genetic information to new therapies. Here, we used a ribonucleoprotein-based CRISPR/Cas9 system to model human clonal hematopoiesis of indeterminate potential and acute myeloid leukemia (AML). We edited multiple genes recurrently mutated in hematological disorders, including those encoding epigenetic regulators, transcriptional regulators, and signaling components in murine hematopoietic stem/progenitor cells. Tracking the clonal dynamics by sequencing the indels induced by CRISPR/Cas9 revealed clonal expansion in some recipient mice that progressed to AML initiated by leukemia-initiating cells. Our results establish that the CRISPR/Cas9-mediated multiplex mutagenesis can be used to engineer a variety of murine models of hematological malignancies with complex genetic architectures seen in human disease.


Asunto(s)
Sistemas CRISPR-Cas , Células Clonales/patología , Edición Génica/métodos , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Animales , Trasplante de Médula Ósea , Proteína 9 Asociada a CRISPR , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Genes Relacionados con las Neoplasias , Humanos , Mutación INDEL , Leucemia Mieloide Aguda/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Preleucemia/genética , Preleucemia/patología , Ribonucleoproteínas/genética , Organismos Libres de Patógenos Específicos
5.
Elife ; 72018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29451493

RESUMEN

Activation of the unfolded protein response (UPR) sustains protein homeostasis (proteostasis) and plays a fundamental role in tissue maintenance and longevity of organisms. Long-range control of UPR activation has been demonstrated in invertebrates, but such mechanisms in mammals remain elusive. Here, we show that the female sex hormone estrogen regulates the UPR in hematopoietic stem cells (HSCs). Estrogen treatment increases the capacity of HSCs to regenerate the hematopoietic system upon transplantation and accelerates regeneration after irradiation. We found that estrogen signals through estrogen receptor α (ERα) expressed in hematopoietic cells to activate the protective Ire1α-Xbp1 branch of the UPR. Further, ERα-mediated activation of the Ire1α-Xbp1 pathway confers HSCs with resistance against proteotoxic stress and promotes regeneration. Our findings reveal a systemic mechanism through which HSC function is augmented for hematopoietic regeneration.


Asunto(s)
Endorribonucleasas/metabolismo , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Células Madre Hematopoyéticas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Respuesta de Proteína Desplegada , Animales , Células Cultivadas , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Transducción de Señal
6.
Neuro Oncol ; 19(1): 22-30, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27402815

RESUMEN

BACKGROUND: Astrocytomas are the most common primary human brain tumors. Receptor tyrosine kinases (RTKs), including tyrosine receptor kinase B (TrkB, also known as tropomyosin-related kinase B; encoded by neurotrophic tyrosine kinase receptor type 2 [NTRK2]), are frequently mutated by rearrangement/fusion in high-grade and low-grade astrocytomas. We found that activated TrkB can contribute to the development of astrocytoma and might serve as a therapeutic target in this tumor type. METHODS: To identify RTKs capable of inducing astrocytoma formation, a library of human tyrosine kinases was screened for the ability to transform murine Ink4a-/-/Arf-/- astrocytes. Orthotopic allograft studies were conducted to evaluate the effects of RTKs on the development of astrocytoma. Since TrkB was identified as a driver of astrocytoma formation, the effect of the Trk inhibitors AZD1480 and RXDX-101 was assessed in astrocytoma cells expressing activated TrkB. RNA sequencing, real-time PCR, western blotting, and enzyme-linked immunosorbent assays were conducted to characterize NTRK2 in astrocytomas. RESULTS: Activated TrkB cooperated with Ink4a/Arf loss to induce the formation of astrocytomas through a mechanism mediated by activation of signal transducer and activator of transcription 3 (STAT3). TrkB activation positively correlated with Ccl2 expression. TrkB-induced astrocytomas remained dependent on TrkB signaling for survival, highlighting a role of NTRK2 as an addictive oncogene. Furthermore, the QKI-NTRK2 fusion associated with human astrocytoma transformed Ink4a-/-/Arf-/- astrocytes, and this process was also mediated via STAT3 signaling. CONCLUSIONS: Our findings provide evidence that constitutively activated NTRK2 alleles, notably the human tumor-associated QKI-NTRK2 fusion, can cooperate with Ink4a/Arf loss to drive astrocytoma formation. Therefore, we propose NTRK2 as a potential therapeutic target in the subset of astrocytoma patients defined by QKI-NTRK2 fusion.


Asunto(s)
Factor 1 de Ribosilacion-ADP/fisiología , Astrocitos/patología , Astrocitoma/patología , Glicoproteínas de Membrana/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptor trkB/metabolismo , Animales , Astrocitos/enzimología , Astrocitoma/enzimología , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Ratones Noqueados , Transducción de Señal
7.
Nat Med ; 22(7): 723-6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27270588

RESUMEN

Brain metastases represent the greatest clinical challenge in treating HER2-positive breast cancer. We report the development of orthotopic patient-derived xenografts (PDXs) of HER2-expressing breast cancer brain metastases (BCBM), and their use for the identification of targeted combination therapies. Combined inhibition of PI3K and mTOR resulted in durable tumor regressions in three of five PDXs, and therapeutic response was correlated with a reduction in the phosphorylation of 4EBP1, an mTORC1 effector. The two nonresponding PDXs showed hypermutated genomes with enrichment of mutations in DNA-repair genes, which suggests an association of genomic instability with therapeutic resistance. These findings suggest that a biomarker-driven clinical trial of PI3K inhibitor in combination with an mTOR inhibitor should be conducted for patients with HER2-positive BCBM.


Asunto(s)
Aminopiridinas/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Everolimus/farmacología , Morfolinas/farmacología , Complejos Multiproteicos/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Proteínas de Ciclo Celular , Reparación del ADN/genética , Resistencia a Antineoplásicos/genética , Quimioterapia Combinada , Factores Eucarióticos de Iniciación , Femenino , Perfilación de la Expresión Génica , Inestabilidad Genómica , Humanos , Inmunohistoquímica , Antígeno Ki-67/efectos de los fármacos , Antígeno Ki-67/metabolismo , Imagen por Resonancia Magnética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones SCID , Terapia Molecular Dirigida , Trasplante de Neoplasias , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación , Receptor ErbB-2/metabolismo , Inducción de Remisión , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Endocrinology ; 155(11): 4461-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25051445

RESUMEN

The steroid hormone aldosterone (aldo) contributes to cardiovascular disease in animal models and in humans. Aldo activates the mineralocorticoid receptor (MR), a hormone-activated transcription factor, and indeed, pharmacological MR inhibition improves cardiovascular outcomes. Because the incidence of cardiovascular disease is lower in premenopausal women, we hypothesized that estrogen (E2) signaling through the estrogen receptor (ER) may protect the vasculature by inhibiting the detrimental effects of aldo signaling through the MR. We demonstrate that E2-activated ER inhibits MR-mediated gene transcription from the mouse mammary tumor virus reporter in human embryonic kidney-293 cells. In contrast, aldo-activated MR does not affect ER-mediated gene transcription. The ERα N terminus (amino acids 1-253) containing part of the DNA-binding domain is sufficient to inhibit MR genomic function, although point mutations reveal that DNA binding, ligand-independent activation, and rapid nongenomic ERα signaling are not required for this effect. Furthermore, ERα and MR are part of a complex in cell lysates, with amino acids 1-233 of the ERα N terminus being sufficient to complex with the MR. Overall, the ability of ERα to inhibit MR-mediated gene transcription correlates with the ability of ERα segments to both localize to the nucleus and complex with the MR. In cultured vascular endothelial cells expressing ERα, E2 inhibits aldo induction of the vascular MR target gene intercellular adhesion molecule-1 (ICAM-1). ICAM-1 induction by endothelial MR is known to promote vascular inflammation that could contribute to the mechanism of aldo-induced atherosclerosis. E2 also inhibits aldo induction of ICAM-1 protein and prevents aldo-enhanced leukocyte adhesion to endothelial cells. These studies support a new model in which E2-activated ER in endothelial cells forms a complex with MR in the nucleus to modulate MR regulation of the proinflammatory gene ICAM-1. Estrogen inhibition of MR regulation of genes that contribute to cardiovascular disease may be a new mechanism by which premenopausal women are protected from cardiovascular disease.


Asunto(s)
Estrógenos/farmacología , Regulación de la Expresión Génica , Antagonistas de Receptores de Mineralocorticoides/farmacología , Receptores de Estrógenos/fisiología , Receptores de Mineralocorticoides/fisiología , Animales , Células Cultivadas , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Estrógenos/química , Receptores de Mineralocorticoides/química , Células U937 , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA