Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Pharm Pract ; 35(5): 747-751, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33813934

RESUMEN

BACKGROUND: Although strategies for optimization of pharmacologic therapy in patients with heart failure with reduced ejection fraction (HFrEF) are scripted by guidelines, data from HF registries suggests that guideline-directed medical therapies (GDMT) are underutilized among eligible patients. Whether this discrepancy reflects medication intolerance, contraindications, or a quality of care issue remains unclear. OBJECTIVE: The objective of this initiative was to identify reasons for underutilization and under-dosing of HFrEF therapy in patients at a large, academic medical center. METHODS: Among 500 patients with HFrEF enrolled in a quality improvement project at a tertiary center, we evaluated usage and dosing of 4 categories of GDMT: ACE inhibitors/Angiotensin Receptor Blockers (ACE-i/ARB), Angiotensin Receptor-Neprilysin Inhibitors (ARNi), beta blockers, and Mineralocorticoid Receptor Antagonists (MRA). Reasons for nonprescription and usage of suboptimal doses were abstracted from notes in the chart and from telephone review of previous medication trials with the patient. RESULTS: Of 500 patients identified, 472 subjects had complete data for analysis. Among eligible patients, ACE-i/ARB were prescribed in 81.4% (293 of 360) and beta blockers in 94.4% (442 of 468). Of these patients, 10.6% were prescribed target doses of ACE-i/ARB and 12.4% were prescribed target doses of beta blockers. Utilization of other categories of GDMT was lower, with 54% of eligible patients prescribed MRAs and 27% prescribed an ARNi. In most cases, the reasons for nonprescription or under-dosing of GDMT were not apparent on review of the health record or discussion with the patient. CONCLUSION: Clear rationale for nonprescription and under-dosing of GDMT often cannot be ascertained from detailed review and is only rarely related to documented medication intolerance or contraindications, suggesting an opportunity for quality improvement.


Asunto(s)
Insuficiencia Cardíaca , Antagonistas Adrenérgicos beta/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Neprilisina/farmacología , Neprilisina/uso terapéutico , Receptores de Angiotensina/uso terapéutico , Volumen Sistólico
2.
Clin Cardiol ; 43(1): 4-13, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31725920

RESUMEN

Although optimal pharmacological therapy for heart failure with reduced ejection fraction (HFrEF) is carefully scripted by treatment guidelines, many eligible patients are not treated with guideline-directed medical therapy (GDMT) in clinical practice. We designed a strategy for remote optimization of GDMT on a population scale in patients with HFrEF leveraging nonphysician providers. An electronic health record-based algorithm was used to identify a cohort of patients with a diagnosis of heart failure (HF) and ejection fraction (EF) ≤ 40% receiving longitudinal follow-up at our center. Those with end-stage HF requiring inotropic support, mechanical circulatory support, or transplantation and those enrolled in hospice or palliative care were excluded. Treating providers were approached for consent to adjust medical therapy according to a sequential, stepped titration algorithm modeled on the current American College of Cardiology (ACC)/American Heart Association (AHA) HF Guidelines within a collaborative care agreement. The program was approved by the institutional review board at Brigham and Women's Hospital with a waiver of written informed consent. All patients provided verbal consent to participate. A navigator then facilitated medication adjustments by telephone and conducted longitudinal surveillance of laboratories, blood pressure, and symptoms. Each titration step was reviewed by a pharmacist with supervision as needed from a nurse practitioner and HF cardiologist. Patients were discharged from the program to their primary cardiologist after achievement of an optimal or maximally tolerated regimen. A navigator-led remote management strategy for optimization of GDMT may represent a scalable population-level strategy for closing the gap between guidelines and clinical practice in patients with HFrEF.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Navegación de Pacientes/métodos , Telemedicina/métodos , Anciano , Algoritmos , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Guías de Práctica Clínica como Asunto , Proyectos de Investigación , Volumen Sistólico
3.
PLoS One ; 11(3): e0151089, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26986722

RESUMEN

The majority of ovarian tumors eventually recur in a drug resistant form. Using cisplatin sensitive and resistant cell lines assembled into 3D spheroids we profiled gene expression and identified candidate mechanisms and biological pathways associated with cisplatin resistance. OVCAR-8 human ovarian carcinoma cells were exposed to sub-lethal concentrations of cisplatin to create a matched cisplatin-resistant cell line, OVCAR-8R. Genome-wide gene expression profiling of sensitive and resistant ovarian cancer spheroids identified 3,331 significantly differentially expressed probesets coding for 3,139 distinct protein-coding genes (Fc >2, FDR < 0.05) (S2 Table). Despite significant expression changes in some transporters including MDR1, cisplatin resistance was not associated with differences in intracellular cisplatin concentration. Cisplatin resistant cells were significantly enriched for a mesenchymal gene expression signature. OVCAR-8R resistance derived gene sets were significantly more biased to patients with shorter survival. From the most differentially expressed genes, we derived a 17-gene expression signature that identifies ovarian cancer patients with shorter overall survival in three independent datasets. We propose that the use of cisplatin resistant cell lines in 3D spheroid models is a viable approach to gain insight into resistance mechanisms relevant to ovarian tumors in patients. Our data support the emerging concept that ovarian cancers can acquire drug resistance through an epithelial-to-mesenchymal transition.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Ovario/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Recurrencia Local de Neoplasia , Ovario/metabolismo , Esferoides Celulares , Células Tumorales Cultivadas
4.
J Biochem Mol Toxicol ; 29(1): 1-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25155036

RESUMEN

Arsenic, a ubiquitous environmental toxicant, can affect lipid metabolism through mechanisms that are not well understood. We studied the effect of arsenic on serum lipids, lipid-regulating genes, and transcriptional regulator sterol regulatory element binding protein 1c (SREBP-1c). C57BL/6 mice were administered 0 or 100 ppb sodium arsenite in drinking water for 5 weeks. Arsenic exposure was associated with decreased liver weight but no change in body weight. Serum triglycerides level fell in arsenic-exposed animals, but not in fed animals, after short-term fasting. Hepatic expression of SREBP-1c was reduced in arsenic-exposed fed animals, with a 16-fold change in reduction. Similar effects were seen for SREBP-1c in white adipose tissue. However, fasting resulted in dissociation of the expression of SREBP-1c and its targets, and SREBP-1c protein content could not be shown to correlate with its mRNA expression. We conclude that arsenic modulates hepatic expression of genes involved in lipid regulation through mechanisms that are independent of SREBP-1c expression.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Arsenitos/farmacología , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Hígado/metabolismo , Compuestos de Sodio/farmacología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/biosíntesis , Animales , Arsénico/farmacología , Masculino , Ratones , Triglicéridos/biosíntesis
5.
J Appl Toxicol ; 34(5): 498-505, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23765520

RESUMEN

Arsenic (As) is considered a top environmental chemical of human health because it has been linked to adverse health effects including cancer, diabetes, cardiovascular disease, and reproductive and developmental problems. In several cell culture and animal models, As acts as an endocrine disruptor, which may underlie many of its health effects. Previous work showed that steroid receptor (SR)-driven gene expression is disrupted in cells treated with inorganic As (arsenite, iAs(+3)). In those studies, low iAs(+3) concentrations (0.1-0.7 µM) stimulated hormone-inducible transcription, whereas somewhat higher but still non-cytotoxic levels (1-3 µM) inhibited transcription. This investigation focuses on the mechanisms underlying these inhibitory effects and evaluates the role of methylated trivalent As metabolites on SR function. Recent evidence suggests that, compared with iAs, methylated forms may have distinct biochemical effects. Here, fluorescence polarization (FP) experiments utilizing purified, hormone-bound human glucocorticoid (GR) and progesterone receptor (PR) have demonstrated that neither inorganic (iAs(+3)) nor dimethylated (DMA(+3)) species of trivalent As affect receptor interactions with glucocorticoid DNA response elements (GREs). However, monomethylated forms (monomethylarsenite, MMA(+3) and monomethylarsonic diglutathione, MADG) strongly inhibit GR-GRE and PR-GRE binding. Additionally, speciation studies of iAs(+3)-treated H4IIE rat hepatoma cells show that, under treatment conditions that cause inhibition of hormone-inducible gene transcription, the intracellular concentration of MADG is sufficient to inhibit GR-GRE and PR-GRE interactions in vivo. These results indicate that arsenic's inhibitory endocrine disruption effects are probably caused in part by methylated metabolites' disruption of SR ability to bind DNA response elements that are crucial to hormone-driven gene transcription.


Asunto(s)
Arsenitos/toxicidad , ADN/genética , Disruptores Endocrinos/toxicidad , Receptores de Esteroides/genética , Elementos de Respuesta/genética , Transcripción Genética/efectos de los fármacos , Animales , Arsenitos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Disruptores Endocrinos/metabolismo , Polarización de Fluorescencia , Metilación , Ratas , Relación Estructura-Actividad
6.
Neurotoxicology ; 38: 91-100, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23845766

RESUMEN

Although many previous investigations have studied how mercury compounds cause cell death, sub-cytotoxic levels may affect mechanisms essential for the proper development of the nervous system. The present study investigates whether low doses of methylmercury (MeHg) and mercury chloride (HgCl2) can modulate the activity of JAK/STAT signaling, a pathway that promotes gliogenesis. We report that sub-cytotoxic doses of MeHg enhance ciliary neurotrophic factor (CNTF) evoked STAT3 phosphorylation in human SH-SY5Y neuroblastoma and mouse cortical neural progenitor cells (NPCs). This effect is specific for MeHg, since HgCl2 fails to enhance JAK/STAT signaling. Exposing NPCs to these low doses of MeHg (30-300nM) enhances CNTF-induced expression of STAT3-target genes such as glial fibrillary acidic protein (GFAP) and suppressors of cytokine signaling 3 (SOCS3), and increases the proportion of cells expressing GFAP following 2 days of differentiation. Higher, near-cytotoxic concentrations of MeHg and HgCl2 inhibit STAT3 phosphorylation and lead to increased production of superoxide. Lower concentrations of MeHg effective in enhancing JAK/STAT signaling (30nM) do not result in a detectable increase in superoxide nor increased expression of the oxidant-responsive genes, heme oxygenase 1, heat shock protein A5 and sirtuin 1. These findings suggest that low concentrations of MeHg inappropriately enhance STAT3 phosphorylation and glial differentiation, and that the mechanism causing this enhancement is distinct from the reactive oxygen species-associated cell death observed at higher concentrations of MeHg and HgCl2.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor Neurotrófico Ciliar/farmacología , Compuestos de Metilmercurio/farmacología , Factor de Transcripción STAT3/metabolismo , Células Madre/citología , Animales , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/biosíntesis , Humanos , Cloruro de Mercurio/farmacología , Ratones , Fosforilación/efectos de los fármacos , Células Madre/efectos de los fármacos , Superóxidos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Células Tumorales Cultivadas
7.
PLoS One ; 7(12): e50221, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226513

RESUMEN

Lymphocytes are a key component of the immune system and their differentiation and function are directly influenced by cancer. We examined peripheral blood lymphocyte (PBL) gene expression as a biomarker of illness and treatment effect using the Affymetrix Human Gene ST1 platform in patients with metastatic renal cell carcinoma (mRCC) who received combined treatment with IL-2, interferon-?-2a and dendritic cell vaccine. We examined gene expression, cytokine levels in patient serum and lymphocyte subsets as determined by flow cytometry (FCM). Pre-treatment PBLs from patients with mRCC exhibit a gene expression profile and serum cytokine profile consistent with inflammation and proliferation not found in healthy donors (HD). PBL gene expression from patients with mRCC showed increased mRNA of genes involved with T-cell and T(REG)-cell activation pathways, which was also reflected in lymphocyte subset distribution. Overall, PBL gene expression post-treatment (POST) was not significantly different than pre-treatment (PRE). Nevertheless, treatment related changes in gene expression (post-treatment minus pre-treatment) revealed an increased expression of T-cell and B-cell receptor signaling pathways in responding (R) patients compared to non-responding (NR) patients. In addition, we observed down-regulation of T(REG)-cell pathways post-treatment in R vs. NR patients. While exploratory in nature, this study supports the hypothesis that enhanced inflammatory cytotoxic pathways coupled with blunting of the regulatory pathways is necessary for effective anti-cancer activity associated with immune therapy. This type of analysis can potentially identify additional immune therapeutic targets in patients with mRCC.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Renales/genética , Células Dendríticas/inmunología , Perfilación de la Expresión Génica , Interferón-alfa/uso terapéutico , Interleucina-2/uso terapéutico , Neoplasias Renales/genética , Linfocitos/metabolismo , Carcinoma de Células Renales/sangre , Carcinoma de Células Renales/terapia , Análisis por Conglomerados , Citocinas/sangre , Femenino , Citometría de Flujo , Humanos , Neoplasias Renales/sangre , Neoplasias Renales/terapia , Subgrupos Linfocitarios , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
PLoS One ; 7(5): e38249, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693606

RESUMEN

BACKGROUND: Arsenic (As) exposure is a significant worldwide environmental health concern. Chronic exposure via contaminated drinking water has been associated with an increased incidence of a number of diseases, including reproductive and developmental effects. The goal of this study was to identify adverse outcomes in a mouse model of early life exposure to low-dose drinking water As (10 ppb, current U.S. EPA Maximum Contaminant Level). METHODOLOGY AND FINDINGS: C57B6/J pups were exposed to 10 ppb As, via the dam in her drinking water, either in utero and/or during the postnatal period. Birth outcomes, the growth of the F1 offspring, and health of the dams were assessed by a variety of measurements. Birth outcomes including litter weight, number of pups, and gestational length were unaffected. However, exposure during the in utero and postnatal period resulted in significant growth deficits in the offspring after birth, which was principally a result of decreased nutrients in the dam's breast milk. Cross-fostering of the pups reversed the growth deficit. Arsenic exposed dams displayed altered liver and breast milk triglyceride levels and serum profiles during pregnancy and lactation. The growth deficits in the F1 offspring resolved following separation from the dam and cessation of exposure in male mice, but did not resolve in female mice up to six weeks of age. CONCLUSIONS/SIGNIFICANCE: Exposure to As at the current U.S. drinking water standard during critical windows of development induces a number of adverse health outcomes for both the dam and offspring. Such effects may contribute to the increased disease risks observed in human populations.


Asunto(s)
Arsénico/farmacología , Agua Potable/química , Feto/efectos de los fármacos , Crecimiento y Desarrollo/efectos de los fármacos , Contaminantes Químicos del Agua/farmacología , Crianza de Animales Domésticos , Animales , Arsénico/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Lactancia/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Leche Humana/efectos de los fármacos , Leche Humana/metabolismo , Embarazo , Triglicéridos/metabolismo
9.
Cancer Res ; 70(5): 1981-8, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20179202

RESUMEN

Exposure to the environmental toxicant arsenic, through both contaminated water and food, contributes to significant health problems worldwide. In particular, arsenic exposure is thought to function as a carcinogen for lung, skin, and bladder cancer via mechanisms that remain largely unknown. More recently, the Hedgehog signaling pathway has also been implicated in the progression and maintenance of these same cancers. Based on these similarities, we tested the hypothesis that arsenic may act in part through activating Hedgehog signaling. Here, we show that arsenic is able to activate Hedgehog signaling in several primary and established tissue culture cells as well as in vivo. Arsenic activates Hedgehog signaling by decreasing the stability of the repressor form of GLI3, one of the transcription factors that ultimately regulate Hedgehog activity. We also show, using tumor samples from a cohort of bladder cancer patients, that high levels of arsenic exposure are associated with high levels of Hedgehog activity. Given the important role Hedgehog signaling plays in the maintenance and progression of a variety of tumors, including bladder cancer, these results suggest that arsenic exposure may in part promote cancer through the activation of Hedgehog signaling. Thus, we provide an important insight into the etiology of arsenic-induced human carcinogenesis, which may be relevant to millions of people exposed to high levels of arsenic worldwide.


Asunto(s)
Arsenitos/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Sustancias Peligrosas/toxicidad , Proteínas Hedgehog/metabolismo , Compuestos de Sodio/toxicidad , Neoplasias de la Vejiga Urinaria/inducido químicamente , Animales , Arsenitos/envenenamiento , Bovinos , Células Endoteliales/efectos de los fármacos , Sustancias Peligrosas/envenenamiento , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Transducción de Señal/efectos de los fármacos , Compuestos de Sodio/envenenamiento
10.
Environ Health Perspect ; 117(9): 1441-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19750111

RESUMEN

BACKGROUND: Arsenic exposure is a significant worldwide environmental health concern. We recently reported that 5-week exposure to environmentally relevant levels (10 and 100 ppb) of As in drinking water significantly altered components of the innate immune response in mouse lung, which we hypothesize is an important contributor to the increased risk of lung disease in exposed human populations. OBJECTIVES: We investigated the effects of As exposure on respiratory influenza A (H1N1) virus infection, a common and potentially fatal disease. METHODS: In this study, we exposed C57BL/6J mice to 100 ppb As in drinking water for 5 weeks, followed by intranasal inoculation with a sub lethal dose of influenza A/PuertoRico/8/34 (H1N1) virus. Multiple end points were assessed postinfection. RESULTS: Arsenic was associated with a number of significant changes in response to influenza, including an increase in morbidity and higher pulmonary influenza virus titers on day 7 post-infection. We also found many alterations in the immune response relative to As-unexposed controls, including a decrease in the number of dendritic cells in the mediastinal lymph nodes early in the course of infection. CONCLUSIONS: Our data indicate that chronic As exposure significantly compromises the immune response to infection. Alterations in response to repeated lung infection may also contribute to other chronic illnesses, such as bronchiectasis, which is elevated by As exposure in epidemiology studies.


Asunto(s)
Arsénico/toxicidad , Virus de la Influenza A/aislamiento & purificación , Infecciones por Orthomyxoviridae/inmunología , Animales , Líquido del Lavado Bronquioalveolar , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Environ Health Perspect ; 117(7): 1108-15, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19654921

RESUMEN

BACKGROUND: Chronic exposure to drinking water arsenic is a significant worldwide environmental health concern. Exposure to As is associated with an increased risk of lung disease, which may make it a unique toxicant, because lung toxicity is usually associated with inhalation rather than ingestion. OBJECTIVES: The goal of this study was to examine mRNA and protein expression changes in the lungs of mice exposed chronically to environmentally relevant concentrations of As in the food or drinking water, specifically examining the hypothesis that As may preferentially affect gene and protein expression related to immune function as part of its mechanism of toxicant action. METHODS: C57BL/6J mice fed a casein-based AIN-76A defined diet were exposed to 10 or 100 ppb As in drinking water or food for 5-6 weeks. RESULTS: Whole genome transcriptome profiling of animal lungs revealed significant alterations in the expression of many genes with functions in cell adhesion and migration, channels, receptors, differentiation and proliferation, and, most strikingly, aspects of the innate immune response. Confirmation of mRNA and protein expression changes in key genes of this response revealed that genes for interleukin 1beta, interleukin 1 receptor, a number of toll-like receptors, and several cytokines and cytokine receptors were significantly altered in the lungs of As-exposed mice. CONCLUSIONS: These findings indicate that chronic low-dose As exposure at the current U.S. drinking-water standard can elicit effects on the regulation of innate immunity, which may contribute to altered disease risk, particularly in lung.


Asunto(s)
Arsénico/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Abastecimiento de Agua , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Perfilación de la Expresión Génica , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
PLoS One ; 4(8): e6766, 2009 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-19707557

RESUMEN

Chronic exposure to inorganic arsenic (iAs) found in the environment is one of the most significant and widespread environmental health risks in the U.S. and throughout the world. It is associated with a broad range of health effects from cancer to diabetes as well as reproductive and developmental anomalies. This diversity of diseases can also result from disruption of metabolic and other cellular processes regulated by steroid hormone receptors via aberrant transcriptional regulation. Significantly, exposure to iAs inhibits steroid hormone-mediated gene activation. iAs exposure is associated with disease, but is also used therapeutically to treat specific cancers complicating an understanding of iAs action. Transcriptional activation by steroid hormone receptors is accompanied by changes in histone and non-histone protein post-translational modification (PTM) that result from the enzymatic activity of coactivator and corepressor proteins such as GRIP1 and CARM1. This study addresses how iAs represses steroid receptor-regulated gene transcription. PTMs on histones H3 and H4 at the glucocorticoid receptor (GR)-activated mouse mammary tumor virus (MMTV) promoter were identified by chromatin immunoprecipitation analysis following exposure to steroid hormone+/-iAs. Histone H3K18 and H3R17 amino acid residues had significantly different patterns of PTMs after treatment with iAs. Promoter interaction of the coactivator CARM1 was disrupted, but the interaction of GRIP1, a p160 coactivator through which CARM1 interacts with a promoter, was intact. Over-expression of CARM1 was able to fully restore and GRIP1 partially restored iAs-repressed transcription indicating that these coactivators are functionally associated with iAs-mediated transcriptional repression. Both are essential for robust transcription at steroid hormone regulated genes and both are associated with disease when inappropriately expressed. We postulate that iAs effects on CARM1 and GRIP1 may underlie some of its therapeutic effects and as well be associated with its toxic effects.


Asunto(s)
Arsénico/farmacología , Histonas/metabolismo , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/metabolismo , Receptores de Glucocorticoides/fisiología , Transcripción Genética/efectos de los fármacos , Animales , Línea Celular Tumoral , Cloranfenicol O-Acetiltransferasa/genética , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Ratones , Reacción en Cadena de la Polimerasa
13.
Toxicol Pathol ; 36(6): 805-17, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18812580

RESUMEN

Chronic ingestion of arsenic is associated with increased incidence of respiratory and cardiovascular diseases. To investigate the role of arsenic in early events in vascular pathology, C57BL/6 mice ingested drinking water with or without 50 ppb sodium arsenite (AsIII) for four, five, or eight weeks. At five and eight weeks, RNA from the lungs of control and AsIII-exposed animals was processed for microarray. Sixty-five genes were significantly and differentially expressed. Differential expression of extracellular matrix (ECM) gene transcripts was particularly compelling, as 91% of genes in this category, including elastin and collagen, were significantly decreased. In additional experiments, real-time RT-PCR showed an AsIII-induced decrease in many of these ECM gene transcripts in the heart and NIH3T3 fibroblast cells. Histological stains for collagen and elastin show a distinct disruption in the ECM surrounding small arteries in the heart and lung of AsIII-exposed mice. Immunohistochemical detection of alpha-smooth muscle actin in blood vessel walls was decreased in the AsIII-exposed animals. These data reveal a functional link between AsIII exposure and disruption in the vascular ECM. These AsIII-induced early pathological events may predispose humans to respiratory and cardiovascular diseases linked to chronic low-dose AsIII exposure.


Asunto(s)
Arsenitos/toxicidad , Vasos Sanguíneos/efectos de los fármacos , Proteínas de la Matriz Extracelular/genética , Expresión Génica/efectos de los fármacos , Miocardio/patología , Compuestos de Sodio/toxicidad , Animales , Arsénico/toxicidad , Arsenitos/administración & dosificación , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Colágeno/genética , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/patología , Elastina/genética , Proteínas de Choque Térmico/genética , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Compuestos de Sodio/administración & dosificación
14.
Am J Physiol Cell Physiol ; 295(3): C807-18, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18650266

RESUMEN

P-glycoprotein (Pgp), a member of the adenosine triphosphate-binding cassette (ABC) transporter superfamily, is a major drug efflux pump expressed in normal tissues, and is overexpressed in many human cancers. Overexpression of Pgp results in reduced intracellular drug concentration and cytotoxicity of chemotherapeutic drugs and is thought to contribute to multidrug resistance of cancer cells. The involvement of Pgp in clinical drug resistance has led to a search for molecules that block Pgp transporter activity to improve the efficacy and pharmacokinetics of therapeutic agents. We have recently identified and characterized a secreted toxin from Pseudomonas aeruginosa, designated cystic fibrosis transmembrane conductance regulator (CFTR) inhibitory factor (Cif). Cif reduces the apical membrane abundance of CFTR, also an ABC transporter, and inhibits the CFTR-mediated chloride ion secretion by human airway and kidney epithelial cells. We report presently that Cif also inhibits the apical membrane abundance of Pgp in kidney, airway, and intestinal epithelial cells but has no effect on plasma membrane abundance of multidrug resistance protein 1 or 2. Cif increased the drug sensitivity to doxorubicin in kidney cells expressing Pgp by 10-fold and increased the cellular accumulation of daunorubicin by 2-fold. Thus our studies show that Cif increases the sensitivity of Pgp-overexpressing cells to doxorubicin, consistent with the hypothesis that Cif affects Pgp functional expression. These results suggest that Cif may be useful to develop a new class of specific inhibitors of Pgp aimed at increasing the sensitivity of tumors to chemotherapeutic drugs, and at improving the bioavailability of Pgp transport substrates.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Antibióticos Antineoplásicos/farmacología , Proteínas Bacterianas/farmacología , Membrana Celular/efectos de los fármacos , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Epóxido Hidrolasas/farmacología , Leucocidinas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antibióticos Antineoplásicos/metabolismo , Proteínas Bacterianas/metabolismo , Células CACO-2 , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Perros , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Doxorrubicina/metabolismo , Epóxido Hidrolasas/metabolismo , Humanos , Concentración 50 Inhibidora , Leucocidinas/metabolismo , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Factores de Tiempo , Transfección
15.
Chem Biol Interact ; 173(2): 129-40, 2008 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-18396267

RESUMEN

Nutritional studies in laboratory animals have long shown that various dietary components can contribute to altered gene expression and metabolism, but diet alone has not been considered in whole animal genomic studies. In this study, global gene expression changes in mice fed either a non-purified chow or a purified diet were investigated and background metal levels in the two diets were measured by ICP-MS. C57BL/6J mice were raised for 5 weeks on either the cereal-based, non-purified LRD-5001 diet or the purified, casein-based AIN-76A diet, as part of a larger study examining the effects of low dose arsenic (As) in the diet or drinking water. Affymetrix Mouse Whole Genome 430 2.0 microarrays were used to assess gene expression changes in the liver and lung. Microarray analysis revealed that animals fed the LRD-5001 diet displayed a significantly higher hepatic expression of Phase I and II metabolism genes as well as other metabolic genes. The LRD-5001 diet masked the As-induced gene expression changes that were clearly seen in the animals fed the AIN-76A diet when each dietary group was exposed to 100 ppb As in drinking water. Trace metal analysis revealed that the LRD-5001 diet contained a mixture of inorganic and organic As at a total concentration of 390 ppb, while the AIN-76A diet contained approximately 20 ppb. These findings indicate that the use of non-purified diets may profoundly alter observable patterns of change induced by arsenic and, likely, by other experimental treatments, particularly, altering gene and protein expression.


Asunto(s)
Alimentación Animal/efectos adversos , Expresión Génica/efectos de los fármacos , Hígado/fisiología , Pulmón/fisiología , Alimentación Animal/análisis , Animales , Animales de Laboratorio , Arsénico/farmacología , Contaminación de Alimentos , Perfilación de la Expresión Génica/métodos , Genoma , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Espectrometría de Masas , Metales/análisis , Metales/farmacología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Environ Health Perspect ; 116(2): 165-72, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18288313

RESUMEN

BACKGROUND: Chronic exposure to excess arsenic in drinking water has been strongly associated with increased risks of multiple cancers, diabetes, heart disease, and reproductive and developmental problems in humans. We previously demonstrated that As, a potent endocrine disruptor at low, environmentally relevant levels, alters steroid signaling at the level of receptor-mediated gene regulation for all five steroid receptors. OBJECTIVES: The goal of this study was to determine whether As can also disrupt gene regulation via the retinoic acid (RA) receptor (RAR) and/or the thyroid hormone (TH) receptor (TR) and whether these effects are similar to previously observed effects on steroid regulation. METHODS AND RESULTS: Human embryonic NT2 or rat pituitary GH3 cells were treated with 0.01-5 microM sodium arsenite for 24 hr, with or without RA or TH, respectively, to examine effects of As on receptor-mediated gene transcription. At low, noncytotoxic doses, As significantly altered RAR-dependent gene transcription of a transfected RAR response element-luciferase construct and the native RA-inducible cytochrome P450 CYP26A gene in NT2 cells. Likewise, low-dose As significantly altered expression of a transfected TR response element-luciferase construct and the endogenous TR-regulated type I deiodinase (DIO1) gene in a similar manner in GH3 cells. An amphibian ex vivo tail metamorphosis assay was used to examine whether endocrine disruption by low-dose As could have specific pathophysiologic consequences, because tail metamorphosis is tightly controlled by TH through TR. TH-dependent tail shrinkage was inhibited in a dose-dependent manner by 0.1- 4.0 microM As. CONCLUSIONS: As had similar effects on RAR- and TR-mediated gene regulation as those previously observed for the steroid receptors, suggesting a common mechanism or action. Arsenic also profoundly affected a TR-dependent developmental process in a model animal system at very low concentrations. Because RAR and TH are critical for both normal human development and adult function and their dysregulation is associated with many disease processes, disruption of these hormone receptor-dependent processes by As is also potentially relevant to human developmental problems and disease risk.


Asunto(s)
Arsénico/toxicidad , Disruptores Endocrinos/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Metamorfosis Biológica/efectos de los fármacos , Receptores de Ácido Retinoico/efectos de los fármacos , Receptores de Hormona Tiroidea/efectos de los fármacos , Hormonas Tiroideas/fisiología , Animales , Línea Celular Tumoral , Humanos , Metamorfosis Biológica/fisiología , Receptores de Ácido Retinoico/genética , Receptores de Hormona Tiroidea/genética , Transcripción Genética , Xenopus laevis/crecimiento & desarrollo
17.
BMC Genomics ; 8: 477, 2007 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-18154678

RESUMEN

BACKGROUND: Genomic research tools such as microarrays are proving to be important resources to study the complex regulation of genes that respond to environmental perturbations. A first generation cDNA microarray was developed for the environmental indicator species Daphnia pulex, to identify genes whose regulation is modulated following exposure to the metal stressor cadmium. Our experiments revealed interesting changes in gene transcription that suggest their biological roles and their potentially toxicological features in responding to this important environmental contaminant. RESULTS: Our microarray identified genes reported in the literature to be regulated in response to cadmium exposure, suggested functional attributes for genes that share no sequence similarity to proteins in the public databases, and pointed to genes that are likely members of expanded gene families in the Daphnia genome. Genes identified on the microarray also were associated with cadmium induced phenotypes and population-level outcomes that we experimentally determined. A subset of genes regulated in response to cadmium exposure was independently validated using quantitative-realtime (Q-RT)-PCR. These microarray studies led to the discovery of three genes coding for the metal detoxication protein metallothionein (MT). The gene structures and predicted translated sequences of D. pulex MTs clearly place them in this gene family. Yet, they share little homology with previously characterized MTs. CONCLUSION: The genomic information obtained from this study represents an important first step in characterizing microarray patterns that may be diagnostic to specific environmental contaminants and give insights into their toxicological mechanisms, while also providing a practical tool for evolutionary, ecological, and toxicological functional gene discovery studies. Advances in Daphnia genomics will enable the further development of this species as a model organism for the environmental sciences.


Asunto(s)
Cadmio/toxicidad , Daphnia/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Metalotioneína/genética , Contaminantes Químicos del Agua/toxicidad , Animales , Biomarcadores , ADN Complementario , Daphnia/metabolismo , Bases de Datos Genéticas , Relación Dosis-Respuesta a Droga , Monitoreo del Ambiente , Genoma/efectos de los fármacos , Metalotioneína/metabolismo , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Environ Toxicol Chem ; 26(12): 2704-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18020683

RESUMEN

Exposure to arsenic is known to cause adverse effects in aquatic biota and wildlife and is of major concern to human health. Although numerous studies have investigated the toxicity of arsenic, little is known about the effects of acquired tolerance on arsenic accumulation and toxicity outside of cell culture models. Accordingly, studies were conducted on the estuarine fish, Fundulus heteroclitus, that were preexposed to nontoxic concentrations of arsenic (as sodium arsenite; 0.7 and 106 micromol As/L) for 96 h or naïve to elevated arsenic to determine the effects of acclimation on arsenic toxicity and accumulation. Tolerance to arsenic was rapidly (96 h) acquired in killifish that were preexposed. In toxicity tests with arsenic-acclimated killifish, preexposure to 106 micromol As/L resulted in a reduction in toxicity when compared to naïve animals. Toxicity in arsenic-acclimated fish also was distinguished by a delayed onset of mortality that manifested in dose-dependent fashion and was significant even for the lower acclimation concentration (0.7 micromol As/L). The increase tolerance acquired following preexposure to 106 micromol As/L for 96 h was associated with lower concentrations of arsenic in all monitored tissues (e.g., gill, liver, kidney) and the whole body when fish were exposed to 240 micromol As/L for an additional 96 h. In accordance with these observations, expression of the multidrug resistance- associated protein (MRP)-2 gene, which is responsible for transporting arsenic conjugated to glutathione out of cells, was increased in the liver of arsenic-acclimated fish.


Asunto(s)
Arsénico/metabolismo , Arsénico/toxicidad , Fundulidae/metabolismo , Animales , Arsénico/análisis , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas de Transporte de Membrana/efectos de los fármacos , Proteínas de Transporte de Membrana/genética , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/efectos de los fármacos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , ARN/efectos de los fármacos , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo , Pruebas de Toxicidad Aguda
19.
Cell Physiol Biochem ; 20(5): 465-72, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17762173

RESUMEN

Cystic fibrosis (CF) is caused by mutations to the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The most common of these mutations is deletion of a phenylalanine residue at position 508 (Delta F508), which accounts for approximately 70% of all CF alleles. This mutation interferes with the biogenesis and maturation of Delta F508-CFTR to the plasma membrane. However, Delta F508-CFTR can partially function upon proper localization. Thus, pharmacological correction of Delta F508-CFTR maturation holds promise in CF therapy. Our previous studies indicate that a single non-cytotoxic dose of the anthracycline doxorubicin (Dox) significantly increase Delta F508-CFTR-associated chloride secretion in MDCK cells by increasing the expression of this protein at the apical plasma membrane. We report here that Dox alters the biogenesis of Delta F508-CFTR. Treatment with Dox increases the resistance of Delta F508-CFTR to trypsin digestion, possibly by expediting protein folding. Further, treatment with Dox reduces the amount of polyubiquitinated Delta F508-CFTR in cells and prolongs the half-life of this protein. Concomitantly, treatment with Dox decreases the association of Delta F508-CFTR with HSP70 but does not alter the expression of major HSP70 family members. Based on these results, we propose that Dox expedites the folding and maturation of Delta F508-CFTR by acting as a pharmacological chaperone, which consequently promotes the functional expression of this protein in MDCK cells.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/biosíntesis , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Doxorrubicina/farmacología , Fenilalanina/genética , Eliminación de Secuencia/genética , Animales , Línea Celular , Perros , Proteínas HSP70 de Choque Térmico/metabolismo , Péptido Hidrolasas/metabolismo , Fenilalanina/metabolismo , Unión Proteica , Ubiquitina/metabolismo
20.
Toxicol Sci ; 100(1): 75-87, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17682005

RESUMEN

The mechanisms of action of drinking water arsenic in the lung and the threshold for biologic effects remain controversial. Our study utilizes Affymetrix 22,690 transcript oligonucleotide microarrays to assess the long-term effects of increasing doses of drinking water arsenic on expression levels in the mouse lung. Mice were exposed at levels commonly found in contaminated drinking water wells in the United States (0, 0.1, 1 ppb), as well as the 50 ppb former maximum contaminant level, for 5 weeks. The expression profiles revealed modification of a number of important signaling pathways, many with corroborating evidence of arsenic responsiveness. We observed statistically significant expression changes for transcripts involved in angiogenesis, lipid metabolism, oxygen transport, apoptosis, cell cycle, and immune response. Validation by reverse transcription-PCR and immunoblot assays confirmed expression changes for a subset of transcripts. These data identify arsenic-modified signaling pathways that will help guide investigations into mechanisms of arsenic's health effects and clarify the threshold for biologic effects and potential disease risk.


Asunto(s)
Arsenitos/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Pulmón/efectos de los fármacos , Compuestos de Sodio/toxicidad , Contaminantes Químicos del Agua/toxicidad , Abastecimiento de Agua , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Western Blotting , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Análisis por Conglomerados , Biología Computacional , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica/métodos , Inmunidad/efectos de los fármacos , Inmunidad/genética , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Medición de Riesgo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...