Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Signal Transduct Target Ther ; 8(1): 425, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37945593

RESUMEN

Proper subcellular localization is crucial for the functioning of biomacromolecules, including proteins and RNAs. Nuclear transport is a fundamental cellular process that regulates the localization of many macromolecules within the nuclear or cytoplasmic compartments. In humans, approximately 60 proteins are involved in nuclear transport, including nucleoporins that form membrane-embedded nuclear pore complexes, karyopherins that transport cargoes through these complexes, and Ran system proteins that ensure directed and rapid transport. Many of these nuclear transport proteins play additional and essential roles in mitosis, biomolecular condensation, and gene transcription. Dysregulation of nuclear transport is linked to major human diseases such as cancer, neurodegenerative diseases, and viral infections. Selinexor (KPT-330), an inhibitor targeting the nuclear export factor XPO1 (also known as CRM1), was approved in 2019 to treat two types of blood cancers, and dozens of clinical trials of are ongoing. This review summarizes approximately three decades of research data in this field but focuses on the structure and function of individual nuclear transport proteins from recent studies, providing a cutting-edge and holistic view on the role of nuclear transport proteins in health and disease. In-depth knowledge of this rapidly evolving field has the potential to bring new insights into fundamental biology, pathogenic mechanisms, and therapeutic approaches.


Asunto(s)
Neoplasias , Receptores Citoplasmáticos y Nucleares , Humanos , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Transporte Activo de Núcleo Celular/genética , Carioferinas/genética , Carioferinas/metabolismo , Carioferinas/uso terapéutico , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Neoplasias/metabolismo , Proteína de Unión al GTP ran
2.
Blood Adv ; 7(12): 2926-2937, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-36827679

RESUMEN

Selinexor (KPT-330) is a small molecule inhibitor of XPO1, which mediates the transport of tumor suppressor proteins, oncogene messenger RNAs, and other proteins involved in governing cell growthfrom the cell nucleus to the cytoplasm. It is overexpressed in many cancer types. Because eukaryotic translation initiator factor 4E (eIF4E) plays a critical role in protein translation in cancer cells in multiple myeloma (MM), we evaluated the effectiveness of combined inhibition of protein translation and nuclear export in MM. Selinexor, an inhibitor of nuclear protein export, dose-dependently decreased eIF4E, IKZF1, and c-MYC protein levels. Using a doxycycline-inducible-pLKO-Tet-On vector, knockdown of eIF4E significantly enhanced the antiproliferative effects of selinexor, sensitized resistant MM cells to selinexor, and increased apoptosis in MM cells. Immunofluorescent analysis of MM cells showed that the combined treatment increased the localization of residual eIF4E to the nucleus compared with selinexor-only treatment. The overexpression of eIF4E at least partially rescued the effects of selinexor in MM cells by reducing G1 cell cycle arrest and increasing the selinexor-IC50 10-fold. Moreover, the combination of selinexor with pharmacologic inhibitors of protein translation showed synergistic anti-MM effects. These results suggest a synergistic anti-MM effect of selinexor combined with eIF4E inhibitors in vitro. Our work provides a better understanding of the potential mechanism of resistance to selinexor and a rationale for combining selinexor with eIF4E inhibitors for the treatment of MM.


Asunto(s)
Carioferinas , Mieloma Múltiple , Humanos , Transporte Activo de Núcleo Celular , Carioferinas/metabolismo , Carioferinas/farmacología , Carioferinas/uso terapéutico , Factor 4E Eucariótico de Iniciación/metabolismo , Apoptosis , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Biosíntesis de Proteínas
3.
J Cutan Pathol ; 47(10): 954-959, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32394425

RESUMEN

Immune checkpoint inhibitors (ICIs) for cancer treatment have revolutionized the field of medicine. However, an unintended but frequent consequence of ICI therapy is the development of cutaneous immune-related adverse events (irAEs), such as lichenoid dermatitis irAEs (LD-irAEs). The hypertrophic variant of LD-irAE may be a diagnostic challenge since it can mimic superficially invasive squamous cell carcinoma (SCC). A 79-year-old woman with metastatic melanoma who began treatment with an ICI-pembrolizumab-plus exportin-1 (XPO1) inhibitor presented after 1 month of therapy with symmetrical violaceous papules coalescing into plaques and with two nodules of the bilateral dorsal hands. Biopsy of the nodules revealed an actinic keratosis and atypical epidermal proliferation concerning for SCC. However, in the ensuing 3 weeks, the patient developed multiple new erythematous, violaceous, and scaly macules and papules, some coalescing into plaques on the extremities. Biopsies of these lesions revealed exuberant irregular epidermal hyperplasia with hypermaturation and lichenoid infiltrate concentrated at the base of the elongated, broadened rete ridges, consistent with hypertrophic LD-irAE. Treatment included topical fluocinonide ointment, intralesional triamcinolone injections and oral acitretin. Distinguishing hypertrophic LD-irAE and SCC can be challenging since both entities share histopathologic features; thus, correlation with clinical presentation is essential for diagnosis and optimal patient management.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/efectos adversos , Carioferinas/antagonistas & inhibidores , Erupciones Liquenoides/patología , Melanoma/secundario , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Acitretina/administración & dosificación , Acitretina/uso terapéutico , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma de Células Escamosas , Dermatitis/inmunología , Dermatitis/patología , Erupciones por Medicamentos/patología , Quimioterapia Combinada , Femenino , Fluocinonida/administración & dosificación , Fluocinonida/uso terapéutico , Glucocorticoides/administración & dosificación , Glucocorticoides/uso terapéutico , Humanos , Hipertrofia/patología , Carioferinas/efectos adversos , Carioferinas/uso terapéutico , Queratolíticos/administración & dosificación , Queratolíticos/uso terapéutico , Erupciones Liquenoides/inducido químicamente , Erupciones Liquenoides/inmunología , Melanoma/tratamiento farmacológico , Resultado del Tratamiento , Triamcinolona/administración & dosificación , Triamcinolona/uso terapéutico , Proteína Exportina 1
4.
Mol Cancer Ther ; 19(2): 540-551, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31594826

RESUMEN

High-grade glioma (HGG) is the leading cause of cancer-related death among children. Selinexor, an orally bioavailable, reversible inhibitor of the nuclear export protein, exportin 1, is in clinical trials for a range of cancers, including HGG. It inhibits the NF-κB pathway and strongly induces the expression of nerve growth factor receptor (NGFR) in preclinical cancer models. We hypothesized that selinexor inhibits NF-κB via upregulation of NGFR. In HGG cells, sensitivity to selinexor correlated with increased induction of cell surface NGFR expression. Knocking down NGFR in HGG cells increased proliferation, anchorage-independent growth, stemness markers, and levels of transcriptionally available nuclear NF-κB not bound to IκB-α, while decreasing apoptosis and sensitivity to selinexor. Increasing IκB-α levels in NGFR knockdown cells restored sensitivity to selinexor. Overexpression of NGFR using cDNA reduced levels of free nuclear NF-κB, decreased stemness markers, and increased markers of cellular differentiation. In all HGG lines tested, selinexor decreased phosphorylation of NF-κB at serine 536 (a site associated with increased transcription of proliferative and inflammatory genes). Because resistance to selinexor monotherapy occurred in our in vivo model, we screened selinexor with a panel of FDA-approved anticancer agents. Bortezomib, a proteasome inhibitor that inhibits the NF-κB pathway through a different mechanism than selinexor, showed synergy with selinexor against HGG in vitro Our results help elucidate selinexor's mechanism of action and identify NGFR as a potential biomarker of its effect in HGG and in addition suggest a combination therapy strategy for these challenging tumors.


Asunto(s)
Glioma/genética , Carioferinas/uso terapéutico , FN-kappa B/metabolismo , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Receptores de Factor de Crecimiento Nervioso/metabolismo , Humanos , Carioferinas/farmacología , Clasificación del Tumor , Receptores Citoplasmáticos y Nucleares/farmacología , Transfección , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...