Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Rep Med ; 4(6): 101082, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37343523

RESUMEN

Genetic alterations help predict the clinical behavior of diffuse gliomas, but some variability remains uncorrelated. Here, we demonstrate that haploinsufficient deletions of chromatin-bound tumor suppressor NFKB inhibitor alpha (NFKBIA) display distinct patterns of occurrence in relation to other genetic markers and are disproportionately present at recurrence. NFKBIA haploinsufficiency is associated with unfavorable patient outcomes, independent of genetic and clinicopathologic predictors. NFKBIA deletions reshape the DNA and histone methylome antipodal to the IDH mutation and induce a transcriptome landscape partly reminiscent of H3K27M mutant pediatric gliomas. In IDH mutant gliomas, NFKBIA deletions are common in tumors with a clinical course similar to that of IDH wild-type tumors. An externally validated nomogram model for estimating individual patient survival in IDH mutant gliomas confirms that NFKBIA deletions predict comparatively brief survival. Thus, NFKBIA haploinsufficiency aligns with distinct epigenome changes, portends a poor prognosis, and should be incorporated into models predicting the disease fate of diffuse gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Niño , Humanos , Neoplasias Encefálicas/genética , Epigenoma , Glioma/genética , Glioma/patología , Haploinsuficiencia/genética , Mutación/genética , Inhibidor NF-kappaB alfa/genética , Isocitrato Deshidrogenasa
2.
Pharmacoepidemiol Drug Saf ; 31(5): 534-545, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35122354

RESUMEN

PURPOSE: Current algorithms to evaluate gestational age (GA) during pregnancy rely on hospital coding at delivery and are not applicable to non-live births. We developed an algorithm using fertility procedures and fertility tests, without relying on delivery coding, to develop a novel GA algorithm in live-births and stillbirths. METHODS: Three pregnancy cohorts were identified from 16 health-plans in the Sentinel System: 1) hospital admissions for live-birth, 2) hospital admissions for stillbirth, and 3) medical chart-confirmed stillbirths. Fertility procedures and prenatal tests, recommended within specific GA windows were evaluated for inclusion in our GA algorithm. Our GA algorithm was developed against a validated delivery-based GA algorithm in live-births, implemented within a sample of chart-confirmed stillbirths, and compared to national estimates of GA at stillbirth. RESULTS: Our algorithm, including fertility procedures and 11 prenatal tests, assigned a GA at delivery to 97.9% of live-births and 92.6% of stillbirths. For live-births (n = 4 701 207), it estimated GA within 2 weeks of a reference delivery-based GA algorithm in 82.5% of pregnancies, with a mean difference of 3.7 days. In chart-confirmed stillbirths (n = 49), it estimated GA within 2 weeks of the clinically recorded GA at delivery for 80% of pregnancies, with a mean difference of 11.1 days. Implementation of the algorithm in a cohort of stillbirths (n = 40 484) had an increased percentage of deliveries after 36 weeks compared to national estimates. CONCLUSIONS: In a population of primarily commercially-insured pregnant women, fertility procedures and prenatal tests can estimate GA with sufficient sensitivity and accuracy for utility in pregnancy studies.


Asunto(s)
Nacimiento Vivo , Mortinato , Electrónica , Femenino , Fertilidad , Edad Gestacional , Humanos , Nacimiento Vivo/epidemiología , Embarazo , Mortinato/epidemiología
3.
CNS Drugs ; 35(3): 317-330, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33743151

RESUMEN

BACKGROUND: Disease-modifying therapies (DMTs) for multiple sclerosis (MS) target immunity and have the potential to increase the risk of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection and alter its clinical course. We assessed these risks in patients with MS (PwMS). OBJECTIVE: The objective of this study was to describe the overall risk of coronavirus disease 2019 (COVID-19) infection, severe disease course, and potential population-level predictors of COVID-19 infection in PwMS, and to provide a context using a cohort of patients with systemic lupus erythematosus (SLE). In addition, the association of different MS DMTs with the incidence and clinical course of COVID-19 was evaluated. Safety data from the Biogen Global Safety Database are also presented on reported cases of COVID-19 in patients treated with Biogen MS therapies. METHODS: The IBM® Explorys electronic health record database of > 72,000,000 patients from US healthcare networks identified patients with MS or SLE, with and without polymerase chain reaction-confirmed COVID-19. COVID-19 cumulative incidence, hospitalization, and deaths among DMT classes were compared using logistic regression (adjusted for age, sex, body mass index, comorbidities, and race/ethnicity). As a secondary data source to assess safety data, COVID-19 reports for Biogen MS therapies were extracted and described from Biogen's Global Safety Database. RESULTS: 30,478 PwMS with an open DMT prescription were identified within Explorys; 344 were COVID-19 positive. The most significant risk factors for acquiring COVID-19 were comorbidity score ≥ 1, body mass index ≥ 30, and Black/African ancestry. Similar risk factors were also identified for patients with SLE. Patients with MS were less likely to develop COVID-19 when treated with interferons (0.61%) and glatiramer acetate (0.51%), vs all other MS DMTs (both p < 0.001); anti-CD20 therapy was associated with the highest risk (3.45%; p < 0.0001). In the Biogen Global Safety Database, we identified 1217 patients who were COVID-19 positive treated with intramuscular interferon beta-1a, peginterferon beta-1a, natalizumab, dimethyl fumarate, diroximel fumarate, or fampridine. CONCLUSIONS: Comorbidities, obesity, and Black/African ancestry, but not age, were associated with a higher risk of SARS-CoV-2 infection in PwMS. Interferons and glatiramer acetate were associated with a reduced COVID-19 risk, whereas anti-CD20 therapies were associated with an increased risk, within the treated MS cohort. COVID-19 safety reports for patients receiving Biogen MS therapies were consistent with the Explorys database and MS literature, illustrating the replicability and power of this approach.


Asunto(s)
COVID-19/epidemiología , Hospitalización/estadística & datos numéricos , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Adolescente , Adulto , Negro o Afroamericano/estadística & datos numéricos , Anciano , Anciano de 80 o más Años , Alemtuzumab/uso terapéutico , Azatioprina/uso terapéutico , COVID-19/mortalidad , Cladribina/uso terapéutico , Comorbilidad , Crotonatos/uso terapéutico , Ciclofosfamida/uso terapéutico , Ciclosporina/uso terapéutico , Bases de Datos Factuales , Dimetilfumarato/uso terapéutico , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Hidroxibutiratos , Factores Inmunológicos/uso terapéutico , Incidencia , Interferón beta/uso terapéutico , Modelos Logísticos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/epidemiología , Masculino , Metotrexato/uso terapéutico , Persona de Mediana Edad , Mitoxantrona/uso terapéutico , Esclerosis Múltiple/epidemiología , Ácido Micofenólico/uso terapéutico , Natalizumab/uso terapéutico , Nitrilos , Obesidad/epidemiología , Factores de Riesgo , Rituximab/uso terapéutico , SARS-CoV-2 , Toluidinas/uso terapéutico , Estados Unidos/epidemiología , Población Blanca/estadística & datos numéricos , Adulto Joven
4.
PLoS One ; 15(2): e0229077, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32069320

RESUMEN

Epidermal growth factor receptor (EGFR) is a pro-tumorigenic receptor tyrosine kinase that facilitates growth for cancer cells that overexpress the receptor. Monoclonal anti-EGFR antibody Cetuximab (CTX) provides significant clinical benefit in patients with head and neck squamous cell carcinoma (HNSCC). Missense mutations in the ectodomain (ECD) of EGFR can be acquired under CTX treatment and mimic the effect of large deletions on spontaneous untethering and activation of the receptor. Little is known about the contribution of EGFR ECD mutations to EGFR activation and CTX resistance in HNSCC. We identified two concurrent non-synonymous missense mutations (G33S and N56K) mapping to domain I in or near the EGF binding pocket of the EGFR ECD in patient-derived HNSCC cells that were selected for CTX resistance through repeated exposure to the agent in an effort to mimic what may occur clinically. Structural modeling predicted that the G33S and N56K mutants would restrict adoption of a fully closed (tethered) and inactive EGFR conformation while not permitting association of EGFR with the EGF ligand or CTX. Binding studies confirmed that the mutant, untethered receptor displayed reduced affinity for both EGF and CTX but demonstrated sustained activation and presence at the cell surface with diminished internalization and sorting for endosomal degradation, leading to persistent downstream AKT signaling. Our results demonstrate that HNSCC cells can select for EGFR ECD mutations under CTX exposure that converge to trap the receptor in an open, ligand-independent, constitutively activated state. These mutants impede the receptor's competence to bind CTX possibly explaining certain cases of CTX treatment-induced or de novo resistance to CTX.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Cetuximab/farmacología , Resistencia a Antineoplásicos/genética , Neoplasias de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Antineoplásicos Inmunológicos/uso terapéutico , Cetuximab/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Humanos , Ligandos , Modelos Moleculares , Mutación Missense , Cultivo Primario de Células , Dominios Proteicos/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Células Tumorales Cultivadas
5.
Clin Infect Dis ; 70(10): 2221-2223, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31412356

RESUMEN

Concerns have been raised about progestin-containing contraceptives and the risk of human immunodeficiency virus (HIV) acquisition. Based on health insurance data from women in the United States with intrauterine device (IUD) insertions during 2011-2018, there was no increased risk of incident HIV diagnosis for levonorgestrel-releasing IUDs versus copper IUDs.


Asunto(s)
Infecciones por VIH , Dispositivos Intrauterinos de Cobre , Dispositivos Intrauterinos Medicados , Femenino , VIH , Infecciones por VIH/epidemiología , Humanos , Dispositivos Intrauterinos Medicados/efectos adversos , Levonorgestrel/efectos adversos , Estados Unidos/epidemiología
6.
Exp Cell Res ; 368(1): 67-74, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29689276

RESUMEN

Little is known about the effects of combinatorial dietary compounds on the regulation of epigenetic mechanisms involved in breast cancer prevention. The human diet consists of a multitude of components, and there is a need to elucidate how certain compounds interact in collaboration. Withaferin A (WA), found in the Indian winter cherry and documented as a DNA methyltransferase (DNMT) inhibitor, and sulforaphane (SFN), a well-known histone deacetylase (HDAC) inhibitor found in cruciferous vegetables, are two epigenetic modifying compounds that have only recently been studied in conjunction. The use of DNMT and HDAC inhibitors to reverse the malignant expression of certain genes in breast cancer has shown considerable promise. Previously, we found that SFN + WA synergistically promote breast cancer cell death. Herein, we determined that these compounds inhibit cell cycle progression from S to G2 phase in MDA-MB-231 and MCF-7 breast cancer. Furthermore, we demonstrate that this unique combination of epigenetic modifying compounds down-regulates the levels of Cyclin D1 and CDK4, and pRB; conversely, the levels of E2F mRNA and tumor suppressor p21 are increased independently of p53. We find these events coincide with an increase in unrestricted histone methylation. We propose SFN + WA-induced breast cancer cell death is attributed, in part, to epigenetic modifications that result in the modulated expression of key genes responsible for the regulation of cancer cell senescence.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Isotiocianatos/farmacología , Witanólidos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias de la Mama/tratamiento farmacológico , División Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Sulfóxidos
7.
Mol Cancer Ther ; 17(5): 921-930, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29592880

RESUMEN

HER2-targeted therapies, such as trastuzumab, have increased the survival rates of HER2+ breast cancer patients. However, despite these therapies, many tumors eventually develop resistance to these therapies. Our lab previously reported an unexpected sensitivity of HER2+ breast cancer cells to poly (ADP-ribose) polymerase inhibitors (PARPi), agents that target homologous recombination (HR)-deficient tumors, independent of a DNA repair deficiency. In this study, we investigated whether HER2+ trastuzumab-resistant (TR) breast cancer cells were susceptible to PARPi and the mechanism behind PARPi induced cytotoxicity. We demonstrate that the PARPi ABT-888 (veliparib) decreased cell survival in vitro and tumor growth in vivo of HER2+ TR breast cancer cells. PARP-1 siRNA confirmed that cytotoxicity was due, in part, to PARP-1 inhibition. Furthermore, PARP-1 silencing had variable effects on the expression of several NF-κB-regulated genes. In particular, silencing PARP-1 inhibited NF-κB activity and reduced p65 binding at the IL8 promoter, which resulted in a decrease in IL8 mRNA and protein expression. Our results provide insight in the potential mechanism by which PARPi induces cytotoxicity in HER2+ breast cancer cells and support the testing of PARPi in patients with HER2+ breast cancer resistant to trastuzumab. Mol Cancer Ther; 17(5); 921-30. ©2018 AACR.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Trastuzumab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos Inmunológicos/farmacología , Bencimidazoles/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Receptor ErbB-2/metabolismo
8.
Glia ; 66(5): 987-998, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29380422

RESUMEN

Previously, we determined microRNA-31 (miR-31) is a noncoding tumor suppressive gene frequently deleted in glioblastoma (GBM); miR-31 suppresses tumor growth, in part, by limiting the activity of NF-κB. Herein, we expand our previous studies by characterizing the role of miR-31 during neural precursor cell (NPC) to astrocyte differentiation. We demonstrate that miR-31 expression and activity is suppressed in NPCs by stem cell factors such as Lin28, c-Myc, SOX2 and Oct4. However, during astrocytogenesis, miR-31 is induced by STAT3 and SMAD1/5/8, which mediate astrocyte differentiation. We determined miR-31 is required for terminal astrocyte differentiation, and that the loss of miR-31 impairs this process and/or prevents astrocyte maturation. We demonstrate that miR-31 promotes astrocyte development, in part, by reducing the levels of Lin28, a stem cell factor implicated in NPC renewal. These data suggest that miR-31 deletions may disrupt astrocyte development and/or homeostasis.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular/fisiología , MicroARNs/metabolismo , Células-Madre Neurales/metabolismo , Animales , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Immunoblotting , Hibridación in Situ , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Xenopus laevis
9.
J Neurooncol ; 132(2): 219-229, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28181105

RESUMEN

Protein kinase CK2 is a ubiquitously expressed serine/threonine kinase composed of two catalytic subunits (α) and/or (α') and two regulatory (ß) subunits. The expression and kinase activity of CK2 is elevated in many different cancers, including glioblastoma (GBM). Brain tumor initiating cells (BTICs) are a subset of cells that are highly tumorigenic and promote the resistance of GBM to current therapies. We previously reported that CK2 activity promotes prosurvival signaling in GBM. In this study, the role of CK2 signaling in BTIC function was examined. We found that expression of CK2α was increased in CD133+ BTICs compared to CD133- cells within the same GBM xenolines. Treatment with CX-4945, an ATP-competitive inhibitor of CK2, led to reduced expression of Sox2 and Nestin, transcription factors important for the maintenance of stem cells. Similarly, inhibition of CK2 also reduced the frequency of CD133+ BTICs over the course of 7 days, indicating a role for CK2 in BTIC persistence and survival. Importantly, using an in vitro limiting dilution assay, we found that inhibition of CK2 kinase activity with CX-4945 or siRNA knockdown of the CK2 catalytic subunits reduced neurosphere formation in GBM xenolines of different molecular subtypes. Lastly, we found that inhibition of CK2 led to decreased EGFR levels in some xenolines, and combination treatment with CX-4945 and Gefitinib to inhibit CK2 and EGFR, respectively, provided optimal inhibition of viability of cells. Therefore, due to the integration of CK2 in multiple signaling pathways important for BTIC survival, CK2 is a promising target in GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Antígeno AC133/metabolismo , Animales , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Femenino , Gefitinib , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Naftiridinas/farmacología , Fenazinas , Embarazo , Quinazolinas/farmacología , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Cancer Res ; 15(3): 340-347, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28031413

RESUMEN

HER2+ breast tumors have been shown to express elevated levels of PARP1 protein. Yet, the mechanism by which PARP1 is upregulated in HER2+ breast cancer is unknown. Here, knockdown of HER2 (ERBB2) in HER2+ breast cancer cells resulted in a reduction in PARP1 protein. Conversely, ectopic overexpression of HER2 in a non-HER2-overexpressing cell line resulted in increased PARP1 protein levels. Alterations in HER2 expression had no significant effect on PARP1 transcript levels. Instead, HER2 mRNA status was inversely correlated with let-7a miRNA levels in breast cancer cells. Ectopic expression of let-7a miRNA resulted in downregulation of PARP1 protein, whereas expression of the let-7a anti-miRNA increased PARP1 protein. Furthermore, luciferase assays demonstrate that let-7a regulates PARP1 via its 3'UTR. Importantly, let-7a was significantly lower in human HER2+ breast tumors compared with HER2- breast tumors and inversely correlated with PARP1 protein levels. Finally, HER2+ breast cancer cells exhibited similar cytotoxicity to ectopic let-7a expression as the PARP inhibitor veliparib (ABT-888). Collectively, these results reveal that increased PARP1 expression in HER2+ breast cancers is regulated by the let-7a miRNA, and that let-7a is a potential strategy to suppress PARP1 activity.Implications: This study reports the novel findings that HER2 increases PARP1 protein via suppression of the let-7a miRNA, which regulates the PARP1 3'-UTR. Moreover, HER2 status correlates with high PARP1 and low let-7a in breast cancer clinical specimens. Mol Cancer Res; 15(3); 340-7. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , MicroARNs/genética , Poli(ADP-Ribosa) Polimerasa-1/biosíntesis , Receptor ErbB-2/biosíntesis , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Femenino , Humanos , MicroARNs/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , Receptor ErbB-2/genética , Transfección , Regulación hacia Arriba
11.
Food Funct ; 7(9): 4012-4019, 2016 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-27711848

RESUMEN

In the present study, anti-proliferative activities of cranberry derived flavonoids and some of their in vivo metabolites were evaluated using a panel of human bladder tumor cell lines (RT4, SCABER, and SW-780) and non-tumorigenic immortalized human uroepithelial cells (SV-HUC). Among the compounds tested, quercetin 3-O-glucoside, isorhamnetin (3'-O-methylquercetin), myricetin and quercetin showed strong concentration-dependent cell growth inhibitory activities in bladder cancer cells with IC50 values in a range of 8-92 µM. Furthermore, isorhamnetin and myricetin had very low inhibitory activity against SV-HUC even at very high concentrations (>200 µM) compared to bladder cancer cells, indicating that their cytotoxicity is selective for cancer cells. To determine whether the differential cell growth inhibitory effects of isomeric flavonoids quercetin 3-O-glucoside (active) and hyperoside (quercetin 3-O-galactoside) (inactive) are related to their metabolism by the cancer cells, SW-780 cells were incubated with these compounds and their metabolism was examined by LC-MS/MS. Compared to quercetin 3-O-glucoside, hyperoside undergoes relatively less metabolic biotransformation (methylation, glucuronidation and quinone formation). These data suggest that isorhamnetin and quercetin 3-O-glucoside may be the active forms of quercetin in prevention of bladder cancer in vivo and emphasize the importance of metabolism for the prevention of bladder cancer by diets rich in cranberries.


Asunto(s)
Flavonoides/farmacología , Inhibidores de Crecimiento/farmacología , Extractos Vegetales/farmacología , Neoplasias de la Vejiga Urinaria/fisiopatología , Vaccinium macrocarpon/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Flavonoides/química , Flavonoides/metabolismo , Frutas/química , Frutas/metabolismo , Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/metabolismo , Humanos , Estructura Molecular , Extractos Vegetales/química , Extractos Vegetales/metabolismo , Espectrometría de Masas en Tándem , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Vaccinium macrocarpon/metabolismo
12.
J Biol Chem ; 291(30): 15830-40, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27226638

RESUMEN

Inflammation and endoplasmic reticulum (ER) stress are associated with many neurological diseases. ER stress is brought on by the accumulation of misfolded proteins in the ER, which leads to activation of the unfolded protein response (UPR), a conserved pathway that transmits signals to restore homeostasis or eliminate the irreparably damaged cell. We provide evidence that inhibition or genetic haploinsufficiency of protein kinase R-like endoplasmic reticulum kinase (PERK) can selectively control inflammation brought on by ER stress without impinging on UPR-dependent survival and adaptive responses or normal immune responses. Using astrocytes lacking one or both alleles of PERK or the PERK inhibitor GSK2606414, we demonstrate that PERK haploinsufficiency or partial inhibition led to reduced ER stress-induced inflammation (IL-6, CCL2, and CCL20 expression) without compromising prosurvival responses. In contrast, complete loss of PERK blocked canonical PERK-dependent UPR genes and promoted apoptosis. Reversal of eIF2α-mediated translational repression using ISRIB potently suppressed PERK-dependent inflammatory gene expression, indicating that the selective modulation of inflammatory gene expression by PERK inhibition may be linked to attenuation of eIF2α phosphorylation and reveals a previously unknown link between translational repression and transcription of inflammatory genes. Additionally, ER-stressed astrocytes can drive an inflammatory M1-like phenotype in microglia, and this can be attenuated with inhibition of PERK. Importantly, targeting PERK neither disrupted normal cytokine signaling in astrocytes or microglia nor impaired macrophage phagocytosis or T cell polarization. Collectively, this work suggests that targeting PERK may provide a means for selective immunoregulation in the context of ER stress without disrupting normal immune function.


Asunto(s)
Astrocitos/inmunología , Estrés del Retículo Endoplásmico/inmunología , Macrófagos/inmunología , Microglía/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , eIF-2 Quinasa/inmunología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/inmunología , Indoles/farmacología , Inflamación/genética , Inflamación/inmunología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosforilación/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética
13.
J Neuroinflammation ; 12: 128, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26123499

RESUMEN

BACKGROUND: In experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, mice genetically deficient in the transcription factor signal transducer and activator of transcription 4 (STAT4) are resistant to disease. In contrast, deletion or inhibition of the Th1-associated cytokines IL-12 or IFNγ which act upstream and downstream of STAT4, respectively, does not ameliorate disease. These discordant findings imply that STAT4 may act in a non-canonical role during EAE. Recently, STAT4 has been shown to regulate GM-CSF production by CD4 T cells and this cytokine is necessary for the induction of EAE. However, it is not known if STAT4 controls GM-CSF production by both Th1 and Th17 effector CD4 T cells. METHODS: This study utilized the MOG(35-55) peptide immunization model of EAE. Intracellular cytokine staining and novel mixed bone marrow chimeric mice were used to study the CD4 T cell-intrinsic role of STAT4 during disease. STAT4 chromatin-immunoprecipitation (ChIP-PCR) experiments were performed to show STAT4 directly interacts with the Csf2 gene loci. RESULTS: Herein, we demonstrate that STAT4 controls CD4 T cell-intrinsic GM-CSF production by both Th1 and Th17 CD4 T cells during EAE as well as in vitro. Importantly, we show that STAT4 interacts with the Csf2 locus in MOG(35-55)-activated effector CD4 T cells demonstrating direct modulation of GM-CSF. CONCLUSIONS: Overall, these studies illustrate a previously unrecognized role of STAT4 to regulate GM-CSF production by not only Th1 cells, but also Th17 effector CD4 T cell subsets during EAE pathogenesis. Critically, these data highlight for the first time that STAT4 is able to modulate the effector profile of Th17 CD4 T cell subsets, which redefines our current understanding of STAT4 as a Th1-centric factor.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor de Transcripción STAT4/metabolismo , Células TH1/metabolismo , Células Th17/metabolismo , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos , Factor de Transcripción STAT4/deficiencia , Factor de Transcripción STAT4/genética , Células TH1/patología , Células Th17/patología
14.
Oncotarget ; 6(19): 17805-16, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-26164206

RESUMEN

Glioblastomas (GBMs) are deadly tumors of the central nervous system. Most GBM exhibit homozygous deletions of the CDKN2A and CDKN2B tumor suppressors at 9p21.3, although loss of CDKN2A/B alone is insufficient to drive gliomagenesis. MIR31HG, which encodes microRNA-31 (miR-31), is a novel non-coding tumor suppressor positioned adjacent to CDKN2A/B at 9p21.3. We have determined that miR-31 expression is compromised in >72% of all GBM, and for patients, this predicts significantly shortened survival times independent of CDKN2A/B status. We show that miR-31 inhibits NF-κB signaling by targeting TRADD, its upstream activator. Moreover, upon reintroduction, miR-31 significantly reduces tumor burden and lengthens survival times in animal models. As such, our work identifies loss of miR-31 as a novel non-coding tumor-driving event in GBM.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/genética , MicroARNs/genética , Transducción de Señal/genética , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Glioblastoma/metabolismo , Xenoinjertos , Humanos , Ratones , FN-kappa B/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo
15.
Mol Cell Biol ; 34(20): 3911-25, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25113558

RESUMEN

Neuroinflammation and endoplasmic reticulum (ER) stress are associated with many neurological diseases. Here, we have examined the interaction between ER stress and JAK/STAT-dependent inflammation in glial cells. We show that ER stress is present in the central nervous system (CNS) concomitant with inflammation and astrogliosis in the multiple sclerosis (MS) mouse model of experimental autoimmune encephalomyelitis (EAE). Astrocytes do not easily succumb to ER stress but rather activate an inflammatory program involving activation of STAT3 in a JAK1-dependent fashion. ER stress-induced activation of the JAK1/STAT3 axis leads to expression of interleukin 6 (IL-6) and several chemokines. Moreover, the activation of STAT3 signaling is dependent on PERK, a central component of the ER stress response, which we show is phosphorylated by JAK1. Disruption of PERK abrogates ER stress-induced activation of STAT3 and subsequent gene expression. Additionally, ER-stressed astrocytes, via paracrine signaling, can stimulate activation of microglia, leading to production of IL-6 and oncostatin M (OSM). These IL-6 cytokines can then synergize with ER stress in astrocytes to drive inflammation. Together, this work describes a new PERK/JAK1/STAT3 signaling pathway that elicits a feed-forward inflammatory loop involving astrocytes and microglia to drive neuroinflammation, which may be relevant in diseases such as MS.


Asunto(s)
Estrés del Retículo Endoplásmico , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT3/metabolismo , eIF-2 Quinasa/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , Astrocitos/fisiología , Comunicación Celular , Células Cultivadas , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Activación Enzimática , Retroalimentación Fisiológica , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , FN-kappa B/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Transducción de Señal
16.
PLoS One ; 8(11): e78728, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244348

RESUMEN

Glioblastoma (GBM) is the most aggressive, neurologically destructive and deadly tumor of the central nervous system (CNS). In GBM, the transcription factors NF-κB and STAT3 are aberrantly activated and associated with tumor cell proliferation, survival, invasion and chemoresistance. In addition, common activators of NF-κB and STAT3, including TNF-α and IL-6, respectively, are abundantly expressed in GBM tumors. Herein, we sought to elucidate the signaling crosstalk that occurs between the NF-κB and STAT3 pathways in GBM tumors. Using cultured GBM cell lines as well as primary human GBM xenografts, we elucidated the signaling crosstalk between the NF-κB and STAT3 pathways utilizing approaches that either a) reduce NF-κB p65 expression, b) inhibit NF-κB activation, c) interfere with IL-6 signaling, or d) inhibit STAT3 activation. Using the clinically relevant human GBM xenograft model, we assessed the efficacy of inhibiting NF-κB and/or STAT3 alone or in combination in mice bearing intracranial xenograft tumors in vivo. We demonstrate that TNF-α-induced activation of NF-κB is sufficient to induce IL-6 expression, activate STAT3, and elevate STAT3 target gene expression in GBM cell lines and human GBM xenografts in vitro. Moreover, the combined inhibition of NF-κB and STAT3 signaling significantly increases survival of mice bearing intracranial tumors. We propose that in GBM, the activation of NF-κB ensures subsequent STAT3 activation through the expression of IL-6. These data verify that pharmacological interventions to effectively inhibit the activity of both NF-κB and STAT3 transcription factors must be used in order to reduce glioma size and aggressiveness.


Asunto(s)
Glioblastoma/metabolismo , Interleucina-6/biosíntesis , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/terapia , Xenoinjertos , Humanos , Interleucina-6/genética , Ratones , Ratones Desnudos , FN-kappa B/genética , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Factor de Transcripción STAT3/genética
17.
Phytomedicine ; 20(1): 17-23, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23123226

RESUMEN

The incidence of type 2 diabetes and metabolic disease is rapidly increasing, but effective therapies for their prevention and treatment have been poorly tolerated or minimally effective. In this study, chronic administration of kudzu root extract (8 months, 0.2%, w/w, in diet) decreased baseline fasting plasma glucose (183±14 vs. 148±11 mg/dl) and improved glucose and insulin tolerance in C57BL/6J ob/ob mice (1.67±0.17 ng/ml [kudzu treated] vs. 2.35±0.63 ng/ml [control]), but such treatment did not alter these parameters in lean control mice. Among the mice on the kudzu supplementation, plasma levels of isoflavone metabolites were significantly higher in ob/ob versus lean control mice, and unmetabolized puerarin (11.50±5.63 ng/g) was found in adipose tissue only in the treated mice. Together, these data demonstrate that a puerarin containing kudzu diet improves glucose and insulin responsiveness in ob/ob mice, suggesting that puerarin may be a beneficial adjuvant for treating metabolic disease.


Asunto(s)
Glucemia/metabolismo , Intolerancia a la Glucosa/tratamiento farmacológico , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Obesidad/tratamiento farmacológico , Fitoterapia , Pueraria/química , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Suplementos Dietéticos , Medicamentos Herbarios Chinos/metabolismo , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Intolerancia a la Glucosa/metabolismo , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Hipolipemiantes/metabolismo , Hipolipemiantes/farmacología , Hipolipemiantes/uso terapéutico , Isoflavonas/metabolismo , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , Raíces de Plantas , Valores de Referencia
18.
J Agric Food Chem ; 59(12): 6682-8, 2011 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-21634376

RESUMEN

The glycosides of flavonoid, anthocyanins and A type proanthocyanidins in cranberry concentrate were characterized and quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Cranberry concentrate (1 g/body weight) was orally gavaged to Fischer-344 rats (n = 6), and blood and urine samples were collected over 24 h periods. Quercetin, 3'-O-methylquercetin (isorhamnetin), myricetin, kaempferol, and proanthocyanidin dimer A2, together with thirteen conjugated metabolites of quercetin and methylquercetin and intact peonidin 3-O-galactoside and cyanidin 3-O-galactoside were identified in the rat urine after cranberry treatment. Very low levels of isorhamnetin (0.48 ± 0.09 ng/mL) and proanthocyanidin dimer A2 (0.541 ± 0.10 ng/mL) were found in plasma samples after 1 h of cranberry administration. Although no quercetin was detected in plasma, MRM analysis of the methanolic extract of urinary bladder showed that chronic administration of cranberry concentrate to rats resulted in accumulation of quercetin and isorhamnetin in the bladder. These results demonstrate that cranberry components undergo rapid metabolism and elimination into the urine of rats and are present in the urinary bladder tissue potentially allowing them to inhibit urinary bladder carcinogenesis.


Asunto(s)
Frutas/química , Fenoles/análisis , Fenoles/metabolismo , Extractos Vegetales/análisis , Extractos Vegetales/metabolismo , Vaccinium macrocarpon/química , Animales , Cromatografía Liquida , Femenino , Fenoles/orina , Extractos Vegetales/orina , Ratas , Ratas Endogámicas F344 , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem , Vejiga Urinaria/química , Vejiga Urinaria/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA