Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Patient Exp ; 8: 23743735211062392, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869849

RESUMO

The coronavirus (COVID-19) pandemic impacted healthcare systems worldwide. In this study, we conducted qualitative interviews with pregnant women in Ghana and the United States (US) to understand their antenatal care (ANC) experience. Adapting to the virtual nature of the pandemic, social media platforms Facebook and WhatsApp were used to recruit, consent, enroll, and interview women. Interviewers used a semi-structured guide with content validated by the US and Ghanaian collaborators. Audio recordings of the interviews were transcribed, coded using Dedoose (v8.0.35, Dedoose) and grounded theory, and analyzed for recurring themes. Between May and July 2020, 32 women (15 Ghanaians, 17 Americans), aged 25-40 years were interviewed. Major themes emerged: (i) apprehension about ANC services; (ii) disruptions to planned healthcare provider use; and (iii) changes in social support. Although the women strove to retain their ANC as planned, the pandemic universally caused several unanticipated changes. Given associations between higher maternal mortality and poor outcomes with inadequate ANC, specific policies and resources for telehealth education and intra- and postpartum support should be implemented to reduce disruptions to ANC imposed by COVID-19.

2.
FASEB J ; 34(9): 11395-11404, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32677118

RESUMO

Cereblon (CRBN) is a substrate recruiter element of the E3 cullin 4-RING ubiquitin ligase complex, and a binding target of immunomodulatory agents (IMiDs). CRBN is responsible for the pleiotropic effects of IMiDs, yet its function in angiogenesis and in mediating the antiangiogenic effects of IMiDs remains unclear. We investigated the role of CRBN in the angiogenic process and in propagating the antiangiogenic effects of IMiDs in vitro. siRNA-mediated CRBN knock down in human endothelial cells (HUVEC and HMVEC-L), did not affect endothelial cell proliferation, migration, or tube formation. Using CRBN-deficient mice, we further demonstrated that microvessal formation can occur independently of cereblon in the ex vivo mouse aortic ring model. The cereblon E3 ubiquitin ligase complex can recruit endothelial cell-specific factors, AGO2 (associated with angiogenesis), and SALL4 (associated with embryogenesis/angiogenesis), for ubiquitin-mediated degradation. Knockdown of CRBN caused a corresponding increase in AGO2 and SALL4 protein expression and IMiD treatment was able to rescue the siCRBN effect to increase the CRBN expression. These findings suggest one potential mechanism of action that likely involves a tightly coordinated regulation of CRBN with endothelial cell targets and highlight the need to further elucidate the mechanism(s), which could include cereblon-independent pathways, through which IMiDs exert their antiangiogenic effects.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Inibidores da Angiogênese/farmacologia , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Lenalidomida/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Interferência de RNA , Ubiquitina-Proteína Ligases/genética
3.
Cancer Biol Ther ; 21(3): 223-230, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31709896

RESUMO

Introduction: Transporters comprising the blood-brain barrier complicate delivery of many therapeutics to the central nervous system. The present study ascertained whether the natural product botryllamide G is viable for in vivo inhibition of ABCG2 using lapatinib as a probe for ABCB1 and ABCG2-mediated efflux from the brain. Methods: Wild-type and Mdr1a/Mdr1b (-/-) mice were treated with botryllamide G and lapatinib ("doublet therapy"), and while a separate cohort of wild-type mice was treated with botryllamide, tariquidar and lapatinib ("triplet therapy"). Results: Botryllamide G demonstrates biphasic elimination with a rapid distribution, decreasing below the in vitro IC50 of 6.9 µM within minutes, yet with a relatively slower terminal half-life (4.6 h). In Mdr1a/Mdr1b (-/-) mice, doublet therapy resulted in a significant increase in brain lapatinib AUC at 8 h (2058 h*ng/mL vs 4007 h*ng/mL; P = .031), but not plasma exposure (P = .15). No significant differences were observed after 24 h. Lapatinib brain exposure was greater through 1 h when wild-type mice were administered triplet therapy (298 h*pg/mg vs 120 h*pg/mg; P < .001), but the triplet decreased brain AUC through 24 h vs. mice administered lapatinib alone (2878 h*pg/mg vs 4461hr*ng/mL; P < .001) and did not alter the brain:plasma ratio. Conclusions: In summary, the ABCG2 inhibitor, botryllamide G, increases brain exposure to lapatinib in mice lacking Abcb1, although the combination of botryllamide G and tariquidar increases brain exposure in wild-type mice only briefly (1 h). Additional research is needed to find analogs of this compound that have better pharmacokinetics and pharmacodynamic effects on ABCG2 inhibition.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/fisiologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Acrilamidas/farmacologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Lapatinib/farmacocinética , Fenóis/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Lapatinib/administração & dosagem , Lapatinib/metabolismo , Masculino , Camundongos , Camundongos Knockout , Distribuição Tecidual , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
4.
Sci Rep ; 9(1): 14884, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31619706

RESUMO

Carfilzomib-lenalidomide-dexamethasone (KRd) therapy has yielded promising results in patients with newly diagnosed multiple myeloma (NDMM). Cereblon (CRBN) is the direct molecular target of lenalidomide and genetic polymorphisms in CRBN have been associated with lenalidomide efficacy. In this study, we assessed the correlation of five single nucleotide variants (SNVs) in the CRBN gene with clinical response and outcomes in patients with NDMM administered KRd therapy with lenalidomide maintenance, achieving favorable trial endpoints in a prospective Phase II study (NCT01402284). Of the observed SNVs, no associations with KRd therapy response were found in this patient cohort, although strong trends in hypoalbuminemia grade and hyperbilirubinemia grade emerged across the CRBN rs1672753 genotype (P = 0.0008) and the rs1714327 genotype (P = 0.0010), respectively. Our results do not provide conclusive support for the predictive utility of CRBN gene polymorphisms as potential biomarkers of clinical response to lenalidomide-based therapy in our patient population. However, these findings remain to be validated in prospective studies using larger patient populations.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Hiperbilirrubinemia/diagnóstico , Hipoalbuminemia/diagnóstico , Lenalidomida/administração & dosagem , Mieloma Múltiplo/tratamento farmacológico , Polimorfismo de Nucleotídeo Único , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Dexametasona/administração & dosagem , Dexametasona/efeitos adversos , Esquema de Medicação , Feminino , Expressão Gênica , Genótipo , Humanos , Hiperbilirrubinemia/induzido quimicamente , Hiperbilirrubinemia/fisiopatologia , Hipoalbuminemia/induzido quimicamente , Hipoalbuminemia/fisiopatologia , Lenalidomida/efeitos adversos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Estudos Prospectivos , Resultado do Tratamento , Ubiquitina-Proteína Ligases
6.
Mol Pharmacol ; 96(2): 158-167, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31175181

RESUMO

Mithramycin demonstrates preclinical anticancer activity, but its therapeutic dose is limited by the development of hepatotoxicity that remains poorly characterized. A pharmacogenomics characterization of mithramycin-induced transaminitis revealed that hepatotoxicity is associated with germline variants in genes involved in bile disposition: ABCB4 (multidrug resistance 3) rs2302387 and ABCB11 [bile salt export pump (BSEP)] rs4668115 reduce transporter expression (P < 0.05) and were associated with ≥grade 3 transaminitis developing 24 hours after the third infusion of mithramycin (25 mcg/kg, 6 hours/infusion, every day ×7, every 28 days; P < 0.0040). A similar relationship was observed in a pediatric cohort. We therefore undertook to characterize the mechanism of mithramycin-induced acute transaminitis. As mithramycin affects cellular response to bile acid treatment by altering the expression of multiple bile transporters (e.g., ABCB4, ABCB11, sodium/taurocholate cotransporting polypeptide, organic solute transporter α/ß) in several cell lines [Huh7, HepaRG, HepaRG BSEP (-/-)] and primary human hepatocytes, we hypothesized that mithramycin inhibited bile-mediated activation of the farnesoid X receptor (FXR). FXR was downregulated in all hepatocyte cell lines and primary human hepatocytes (P < 0.0001), and mithramycin inhibited chenodeoxycholic acid- and GW4046-induced FXR-galactose-induced gene 4 luciferase reporter activity (P < 0.001). Mithramycin promoted glycochenodeoxycholic acid-induced cytotoxicity in ABCB11 (-/-) cells and increased the overall intracellular concentration of bile acids in primary human hepatocytes grown in sandwich culture (P < 0.01). Mithramycin is a FXR expression and FXR transactivation inhibitor that inhibits bile flow and potentiates bile-induced cellular toxicity, particularly in cells with low ABCB11 function. These results suggest that mithramycin causes hepatotoxicity through derangement of bile acid disposition; results also suggest that pharmacogenomic markers may be useful to identify patients who may tolerate higher mithramycin doses. SIGNIFICANCE STATEMENT: The present study characterizes a novel mechanism of drug-induced hepatotoxicity in which mithramycin not only alters farnesoid X receptor (FXR) and small heterodimer partner gene expression but also inhibits bile acid binding to FXR, resulting in deregulation of cellular bile homeostasis. Two novel single-nucleotide polymorphisms in bile flow transporters are associated with mithramycin-induced liver function test elevations, and the present results are the rationale for a genotype-directed clinical trial using mithramycin in patients with thoracic malignancies.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Proteínas de Membrana Transportadoras/genética , Plicamicina/efeitos adversos , Neoplasias Torácicas/tratamento farmacológico , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Adulto , Idoso , Linhagem Celular Tumoral , Doença Hepática Induzida por Substâncias e Drogas/genética , Ensaios Clínicos Fase II como Assunto , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Pessoa de Meia-Idade , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Neoplasias Torácicas/genética , Neoplasias Torácicas/metabolismo
7.
Mar Drugs ; 16(7)2018 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-30029505

RESUMO

Elements of the hypoxia inducible factor (HIF) transcriptional system, a key regulator of the cellular hypoxic response, are up-regulated in a range of cancer cells. HIF is fundamentally involved in tumor angiogenesis, invasion, and energy metabolism. Inhibition of the transcriptional activity of HIF may be of therapeutic benefit to cancer patients. We recently described the identification of two marine pyrroloiminoquinone alkaloids with potent activity in inhibiting the interaction between the oncogenic transcription factor HIF-1α and the coactivator protein p300. Herein, we present further characterization data for these two screening hits: discorhabdin H (1) and discorhabdin L (2), with a specific focus on their anti-angiogenic and anti-tumor effects. We demonstrated that only discorhabdin L (2) possesses excellent anti-angiogenic activity in inhibiting endothelial cell proliferation and tube formation, as well as decreasing microvessel outgrowth in the ex vivo rat aortic ring assay. We further showed that discorhabdin L (2) significantly inhibits in vivo prostate tumor growth in a LNCaP xenograft model. In conclusion, our findings suggest that discorhabdin L (2) represents a promising HIF-1α inhibitor worthy of further drug development.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacocinética , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Neovascularização Patológica/tratamento farmacológico , Quinonas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Proteína p300 Associada a E1A/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Camundongos SCID , Neovascularização Patológica/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
8.
Cancer Biol Ther ; 19(10): 869-870, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29757697

RESUMO

Numerous growth-inducing signaling pathways have been implicated in the development of metastatic castrate-resistant prostate cancer, but their cross-talk with androgen receptor functions remains poorly understood. A recent study published in Science Signaling by Chen et al. 1 has identified a novel androgen-mediated signaling axis driven by loss of SPDEF and gain of TGFBI to facilitate metastasis, which may explain the acquisition of resistance to androgen deprivation therapy. These findings suggest that therapeutic inhibition of androgen signaling may inadvertently promote castrate resistance by inhibiting tumor suppressive functions of the androgen receptor.


Assuntos
Neoplasias Ósseas , Neoplasias da Próstata , Antagonistas de Androgênios , Androgênios , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Proteínas Proto-Oncogênicas c-ets , Receptores Androgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...