Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39005325

ABSTRACT

The phenotype of human placental extravillous trophoblast (EVT) at the end of pregnancy reflects both first trimester differentiation from villous cytotrophoblast (CTB) and later gestational changes, including loss of proliferative and invasive capacity. Invasion abnormalities are central to two major placental pathologies, preeclampsia and placenta accreta spectrum, so characterization of the corresponding normal processes is crucial. In this report, our gene expression analysis, using purified human CTB and EVT cells, highlights an epithelial- mesenchymal transition (EMT) mechanism underlying CTB-EVT differentiation and provides a trophoblast-specific EMT signature. In parallel, DNA methylation profiling shows that CTB cells, already hypomethylated relative to non-trophoblast cell lineages, show further genome- wide hypomethylation in the transition to EVT. However, a small subgroup of genes undergoes gains of methylation (GOM) in their regulatory regions or gene bodies, associated with differential mRNA expression (DE). Prominent in this GOM-DE group are genes involved in the EMT, including multiple canonical EMT markers and the EMT-linked transcription factor RUNX1 , for which we demonstrate a functional role in modulating the migratory and invasive capacities of JEG3 trophoblast cells. This analysis of DE associated with locus-specific GOM, together with functional studies of an important GOM-DE gene, highlights epigenetically regulated genes and pathways acting in human EVT differentiation and invasion, with implications for obstetric disorders in which these processes are dysregulated.

2.
BMC Plant Biol ; 24(1): 580, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38890606

ABSTRACT

BACKGROUND: Tropospheric ozone is an air pollutant that causes negative effects on vegetation, leading to significant losses in crop productivity. It is generated by chemical reactions in the presence of sunlight between primary pollutants resulting from human activity, such as nitrogen oxides and volatile organic compounds. Due to the constantly increasing emission of ozone precursors, together with the influence of a warming climate on ozone levels, crop losses may be aggravated in the future. Therefore, the search for solutions to mitigate these losses becomes a priority. Ozone-induced abiotic stress is mainly due to reactive oxygen species generated by the spontaneous decomposition of ozone once it reaches the apoplast. In this regard, compounds with antioxidant activity offer a viable option to alleviate ozone-induced damage. Using enzymatic technology, we have developed a process that enables the production of an extract with biostimulant properties from okara, an industrial soybean byproduct. The biostimulant, named as OEE (Okara Enzymatic Extract), is water-soluble and is enriched in bioactive compounds present in okara, such as isoflavones. Additionally, it contains a significant fraction of protein hydrolysates contributing to its functional effect. Given its antioxidant capacity, we aimed to investigate whether OEE could alleviate ozone-induced damage in plants. For that, pepper plants (Capsicum annuum) exposed to ozone were treated with a foliar application of OEE. RESULTS: OEE mitigated ozone-induced damage, as evidenced by the net photosynthetic rate, electron transport rate, effective quantum yield of PSII, and delayed fluorescence. This protection was confirmed by the level of expression of genes associated with photosystem II. The beneficial effect was primarily due to its antioxidant activity, as evidenced by the lipid peroxidation rate measured through malondialdehyde content. Additionally, OEE triggered a mild oxidative response, indicated by increased activities of antioxidant enzymes in leaves (catalase, superoxide dismutase, and guaiacol peroxidase) and the oxidative stress index, providing further protection against ozone-induced stress. CONCLUSIONS: The present results support that OEE protects plants from ozone exposure. Taking into consideration that the promotion of plant resistance against abiotic damage is an important goal of biostimulants, we assume that its use as a new biostimulant could be considered.


Subject(s)
Antioxidants , Glycine max , Ozone , Stress, Physiological , Ozone/pharmacology , Glycine max/drug effects , Glycine max/physiology , Glycine max/metabolism , Stress, Physiological/drug effects , Antioxidants/metabolism , Capsicum/drug effects , Capsicum/physiology , Capsicum/metabolism , Photosynthesis/drug effects , Plant Extracts/pharmacology
3.
Front Plant Sci ; 12: 749422, 2021.
Article in English | MEDLINE | ID: mdl-34868133

ABSTRACT

Ozone is a destructive pollutant, damaging crops, and decreasing crop yield. Therefore, there is great interest in finding strategies to alleviate ozone-induced crop losses. In plants, ozone enters leaves through the stomata and is immediately degraded into reactive oxygen species (ROS), producing ROS stress in plants. ROS stress can be controlled by ROS-scavenging systems that include enzymatic or non-enzymatic mechanisms. Our research group has developed a product from rice bran, a by-product of rice milling which has bioactive molecules that act as an antioxidant compound. This product is a water-soluble rice bran enzymatic extract (RBEE) which preserves all the properties and improves the solubility of proteins and the antioxidant components of rice bran. In previous works, the beneficial properties of RBEE have been demonstrated in animals. However, to date, RBEE has not been used as a protective agent against oxidative damage in agricultural fields. The main goal of this study was to investigate the ability of RBEE to be used as a biostimulant by preventing oxidative damage in plants, after ozone exposure. To perform this investigation, pepper plants (Capsicum annuum) exposed to ozone were treated with RBEE. RBEE protected the ozone-induced damage, as revealed by net photosynthetic rate and the content of photosynthetic pigments. RBEE also decreased the induction of antioxidant enzyme activities in leaves (catalase, superoxide dismutase, and ascorbate peroxidase) due to ozone exposure. ROS generation is a common consequence of diverse cellular traumas that also activate the mitogen-activated protein kinase (MAPK) cascade. Thus, it is known that the ozone damages are triggered by the MAPK cascade. To examine the involvement of the MAPK cascade in the ozone damage CaMPK6-1, CaMPK6-2, and CaMKK5 genes were analyzed by qRT-PCR. The results showed the involvement of the MAPK pathway in both, not only in ozone damage but especially in its protection by RBEE. Taken together, these results support that RBEE protects plants against ozone exposure and its use as a new biostimulant could be proposed.

4.
Gac Med Mex ; 157(1): 102-106, 2021.
Article in English | MEDLINE | ID: mdl-34125805

ABSTRACT

INTRODUCTION: In the context of the COVID-19 pandemic, there is concern regarding the impact of the influenza season. OBJECTIVE: To analyze the impact of influenza immunization history on patients with SARS-CoV-2 infection. METHODS: Patients older than 18 years with COVID-19, registered between March and August 2020, were included. Data were analyzed using Fisher's exact test and Student's t-test. To evaluate the impact on mortality, a logistic regression model was used; the relationship between the percentage of patients who received the influenza vaccine and mortality was determined with Pearson's correlation coefficient. RESULTS: 16,879 participants were included; 17 % had a history of influenza vaccination. Mortality was lower in the group with a history of vaccination (3.5 % vs. 7 %, p < 0.0001). The vaccination rate had an inverse relationship with the mortality rate (Pearson's r: -0.922, p = 0.026). CONCLUSIONS: Previous influenza immunization was an independent protective factor for mortality in patients with COVID-19. Although further studies are needed to determine a causal relationship, it would be reasonable to increase influenza immunization in the general population.


INTRODUCCIÓN: En el contexto de la pandemia de COVID-19 existe inquietud en cuanto al impacto de la temporada de influenza. OBJETIVO: Analizar el impacto del antecedente de inmunización contra influenza en pacientes con infección por SARS-CoV-2. MÉTODOS: Se incluyeron pacientes mayores de 18 años con COVID-19, registrados entre marzo y agosto de 2020. Los datos fueron analizados mediante las pruebas exacta de Fisher y t de Student. Para evaluar el impacto en la mortalidad se utilizó un modelo de regresión logística; la relación entre el porcentaje de pacientes a quienes se aplicó la vacuna contra la influenza y la mortalidad fue determinada con el coeficiente de correlación de Pearson. RESULTADOS: Se incluyeron 16 879 participantes; 17 % tuvo antecedente de vacunación contra influenza. La mortalidad fue menor en el grupo con historia de vacunación (3.5 % versus 7 %, p < 0.0001). El porcentaje de vacunación presentó una relación inversa con el porcentaje de mortalidad (r de Pearson ­ 0.922, p = 0.026). CONCLUSIONES: La inmunización contra la influenza fue un factor protector independiente de mortalidad en pacientes con COVID-19. Aunque son necesarios más estudios para determinar la relación causal, será razonable incrementar la inmunización contra influenza en la población general.


Subject(s)
COVID-19/mortality , Influenza Vaccines , Vaccination/statistics & numerical data , Adolescent , Adult , COVID-19/prevention & control , Female , Humans , Male , Middle Aged , Retrospective Studies , Young Adult
5.
Gac. méd. Méx ; 157(1): 108-112, ene.-feb. 2021. tab, graf
Article in Spanish | LILACS | ID: biblio-1279083

ABSTRACT

Resumen Introducción: En el contexto de la pandemia de COVID-19 existe inquietud en cuanto al impacto de la temporada de influenza. Objetivo: Analizar el impacto del antecedente de inmunización contra influenza en pacientes con infección por SARS-CoV-2. Métodos: Se incluyeron pacientes mayores de 18 años con COVID-19, registrados entre marzo y agosto de 2020. Los datos fueron analizados mediante las pruebas exacta de Fisher y t de Student. Para evaluar el impacto en la mortalidad se utilizó un modelo de regresión logística; la relación entre el porcentaje de pacientes a quienes se aplicó la vacuna contra la influenza y la mortalidad fue determinada con el coeficiente de correlación de Pearson. Resultados: Se incluyeron 16 879 participantes; 17 % tuvo antecedente de vacunación contra influenza. La mortalidad fue menor en el grupo con historia de vacunación (3.5 % versus 7 %, p < 0.0001). El porcentaje de vacunación presentó una relación inversa con el porcentaje de mortalidad (r de Pearson –0.922, p = 0.026). Conclusiones: La inmunización contra la influenza fue un factor protector independiente de mortalidad en pacientes con COVID-19. Aunque son necesarios más estudios para determinar la relación causal, será razonable incrementar la inmunización contra influenza en la población general.


Abstract Introduction: In the context of the COVID-19 pandemic, there is concern regarding the impact of the influenza season. Objective: To analyze the impact of influenza immunization history on patients with SARS-CoV-2 infection. Methods: Patients older than 18 years with COVID-19, registered between March and August 2020, were included. Data were analyzed using Fisher’s exact test and Student’s t-test. To evaluate the impact on mortality, a logistic regression model was used; the relationship between the percentage of patients who received the influenza vaccine and mortality was determined with Pearson’s correlation coefficient. Results: 16,879 participants were included; 17 % had a history of influenza vaccination. Mortality was lower in the group with a history of vaccination (3.5 % vs. 7 %, p < 0.0001). The vaccination rate had an inverse relationship with the mortality rate (Pearson’s r: -0.922, p = 0.026). Conclusions: Previous influenza immunization was an independent protective factor for mortality in patients with COVID-19. Although further studies are needed to determine a causal relationship, it would be reasonable to increase influenza immunization in the general population.


Subject(s)
Humans , Male , Female , Adolescent , Adult , Middle Aged , Young Adult , Influenza Vaccines , Vaccination/statistics & numerical data , COVID-19/mortality , Retrospective Studies , COVID-19/prevention & control
6.
Genome Biol ; 21(1): 153, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32594908

ABSTRACT

BACKGROUND: Mapping of allele-specific DNA methylation (ASM) can be a post-GWAS strategy for localizing regulatory sequence polymorphisms (rSNPs). The advantages of this approach, and the mechanisms underlying ASM in normal and neoplastic cells, remain to be clarified. RESULTS: We perform whole genome methyl-seq on diverse normal cells and tissues and three cancer types. After excluding imprinting, the data pinpoint 15,112 high-confidence ASM differentially methylated regions, of which 1838 contain SNPs in strong linkage disequilibrium or coinciding with GWAS peaks. ASM frequencies are increased in cancers versus matched normal tissues, due to widespread allele-specific hypomethylation and focal allele-specific hypermethylation in poised chromatin. Cancer cells show increased allele switching at ASM loci, but disruptive SNPs in specific classes of CTCF and transcription factor binding motifs are similarly correlated with ASM in cancer and non-cancer. Rare somatic mutations affecting these same motif classes track with de novo ASM. Allele-specific transcription factor binding from ChIP-seq is enriched among ASM loci, but most ASM differentially methylated regions lack such annotations, and some are found in otherwise uninformative "chromatin deserts." CONCLUSIONS: ASM is increased in cancers but occurs by a shared mechanism involving disruptive SNPs in CTCF and transcription factor binding sites in both normal and neoplastic cells. Dense ASM mapping in normal plus cancer samples reveals candidate rSNPs that are difficult to find by other approaches. Together with GWAS data, these rSNPs can nominate specific transcriptional pathways in susceptibility to autoimmune, cardiometabolic, neuropsychiatric, and neoplastic diseases.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA Methylation , Neoplasms/metabolism , Transcription Factors/metabolism , Alleles , CpG Islands , Genomic Imprinting , Humans , Linkage Disequilibrium , Neoplasms/genetics , Polymorphism, Single Nucleotide , Whole Genome Sequencing
7.
Heliyon ; 5(6): e01958, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31294110

ABSTRACT

This work presents a new bioprocess process for the extraction of bioactive components from soy pulp by-product (okara) using an enzymatic technology that was compared to a conventional water extraction. Okara is rich in fiber, fat, protein, and bioactive compounds such as isoflavones but its low solubility hampers the use in food and fertilizer industry. After the enzymatic attack with endoproteases half of the original insoluble proteins were converted into soluble peptides. Linked to this process occured the solubilization of isoflavones trapped in the insoluble protein matrix. We were able to extract up to 62.5% of the total isoflavones content, specially aglycones, the more bioactive isoflavone forms, whose values rose 9.12 times. This was probably due to the increased solubilization and interconversion from the original isoflavones. In conclusion, our process resulted in the formulation of a new functional product rich in aglycones and bioactive peptides with higher antioxidant potency than the original source. Therefore, we propose that the enzymatic extraction of okara bioactive compounds is an advantageous tool to replace conventional extraction.

8.
Nature ; 553(7687): 222-227, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323298

ABSTRACT

Chromosomal translocations that generate in-frame oncogenic gene fusions are notable examples of the success of targeted cancer therapies. We have previously described gene fusions of FGFR3-TACC3 (F3-T3) in 3% of human glioblastoma cases. Subsequent studies have reported similar frequencies of F3-T3 in many other cancers, indicating that F3-T3 is a commonly occuring fusion across all tumour types. F3-T3 fusions are potent oncogenes that confer sensitivity to FGFR inhibitors, but the downstream oncogenic signalling pathways remain unknown. Here we show that human tumours with F3-T3 fusions cluster within transcriptional subgroups that are characterized by the activation of mitochondrial functions. F3-T3 activates oxidative phosphorylation and mitochondrial biogenesis and induces sensitivity to inhibitors of oxidative metabolism. Phosphorylation of the phosphopeptide PIN4 is an intermediate step in the signalling pathway of the activation of mitochondrial metabolism. The F3-T3-PIN4 axis triggers the biogenesis of peroxisomes and the synthesis of new proteins. The anabolic response converges on the PGC1α coactivator through the production of intracellular reactive oxygen species, which enables mitochondrial respiration and tumour growth. These data illustrate the oncogenic circuit engaged by F3-T3 and show that F3-T3-positive tumours rely on mitochondrial respiration, highlighting this pathway as a therapeutic opportunity for the treatment of tumours with F3-T3 fusions. We also provide insights into the genetic alterations that initiate the chain of metabolic responses that drive mitochondrial metabolism in cancer.


Subject(s)
Cell Respiration , Microtubule-Associated Proteins/genetics , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins, Fusion/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cell Respiration/drug effects , Cell Transformation, Neoplastic/drug effects , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mitochondria/drug effects , Mitochondria/genetics , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Organelle Biogenesis , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisomes/drug effects , Peroxisomes/metabolism , Phosphorylation , Protein Biosynthesis , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Transcription, Genetic , Xenograft Model Antitumor Assays
9.
Sci Rep ; 7(1): 8100, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28808322

ABSTRACT

Proteostasis alteration and neuroinflammation are typical features of normal aging. We have previously shown that neuroinflammation alters cellular proteostasis through immunoproteasome induction, leading to a transient decrease of proteasome activity. Here, we further investigated the role of acute lipopolysaccharide (LPS)-induced hippocampal neuroinflammation in cellular proteostasis. In particular, we focused on macroautophagy (hereinafter called autophagy) and endoplasmic reticulum-associated protein degradation (ERAD). We demonstrate that LPS injection induced autophagy activation that was dependent, at least in part, on glycogen synthase kinase (GSK)-3ß activity but independent of mammalian target of rapamycin (mTOR) inhibition. Neuroinflammation also produced endoplasmic reticulum (ER) stress leading to canonical unfolded protein response (UPR) activation with a rapid activating transcription factor (ATF) 6α attenuation that resulted in a time-dependent down-regulation of ERAD markers. In this regard, the time-dependent accumulation of unspliced X-box binding protein (XBP) 1, likely because of decreased inositol-requiring enzyme (IRE) 1α-mediated splicing activity, might underlie in vivo ATF6α attenuation. Importantly, lactacystin-induced activation of ERAD was abolished in both the acute neuroinflammation model and in aged rats. Therefore, we provide a cellular pathway through which neuroinflammation might sensitize cells to neurodegeneration under stress situations, being relevant in normal aging and other disorders where neuroinflammation is a characteristic feature.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum-Associated Degradation/physiology , Inflammation/physiopathology , Proteostasis/physiology , Activating Transcription Factor 6/metabolism , Animals , Cell Line , Down-Regulation/physiology , Endoribonucleases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation/metabolism , Male , Mice , Rats , Rats, Wistar , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology , X-Box Binding Protein 1/metabolism
10.
Nature ; 529(7585): 172-7, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26735018

ABSTRACT

Mechanisms that maintain cancer stem cells are crucial to tumour progression. The ID2 protein supports cancer hallmarks including the cancer stem cell state. HIFα transcription factors, most notably HIF2α (also known as EPAS1), are expressed in and required for maintenance of cancer stem cells (CSCs). However, the pathways that are engaged by ID2 or drive HIF2α accumulation in CSCs have remained unclear. Here we report that DYRK1A and DYRK1B kinases phosphorylate ID2 on threonine 27 (Thr27). Hypoxia downregulates this phosphorylation via inactivation of DYRK1A and DYRK1B. The activity of these kinases is stimulated in normoxia by the oxygen-sensing prolyl hydroxylase PHD1 (also known as EGLN2). ID2 binds to the VHL ubiquitin ligase complex, displaces VHL-associated Cullin 2, and impairs HIF2α ubiquitylation and degradation. Phosphorylation of Thr27 of ID2 by DYRK1 blocks ID2-VHL interaction and preserves HIF2α ubiquitylation. In glioblastoma, ID2 positively modulates HIF2α activity. Conversely, elevated expression of DYRK1 phosphorylates Thr27 of ID2, leading to HIF2α destabilization, loss of glioma stemness, inhibition of tumour growth, and a more favourable outcome for patients with glioblastoma.


Subject(s)
Glioblastoma/metabolism , Glioblastoma/pathology , Inhibitor of Differentiation Protein 2/metabolism , Neoplastic Stem Cells/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Cell Line, Tumor , Cullin Proteins/metabolism , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Male , Mice , Neoplastic Stem Cells/pathology , Oxygen/metabolism , Phosphorylation , Phosphothreonine/metabolism , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Xenograft Model Antitumor Assays , Dyrk Kinases
11.
Neurobiol Aging ; 36(5): 1953-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25817083

ABSTRACT

Autophagy plays a key role in the maintenance of cellular homeostasis, and autophagy deregulation gives rise to severe disorders. Many of the signaling pathways regulating autophagy under stress conditions are still poorly understood. Using a model of proteasome stress in rat hippocampus, we have characterized the functional crosstalk between the ubiquitin proteasome system and the autophagy-lysosome pathway, identifying also age-related modifications in the crosstalk between both proteolytic systems. Under proteasome inhibition, both autophagy activation and resolution were efficiently induced in young but not in aged rats, leading to restoration of protein homeostasis only in young pyramidal neurons. Importantly, proteasome stress inhibited glycogen synthase kinase-3ß in young but activated in aged rats. This age-related difference could be because of a dysfunction in the signaling pathway of the insulin growth factor-1 under stress situations. Present data highlight the potential role of glycogen synthase kinase-3ß in the coordination of both proteolytic systems under stress situation, representing a key molecular target to sort out this deleterious effect.


Subject(s)
Aging/metabolism , Aging/physiology , Autophagy/physiology , Glycogen Synthase Kinase 3/physiology , Hippocampus/physiology , Lysosomes/physiology , Proteasome Endopeptidase Complex/physiology , Pyramidal Cells/metabolism , Signal Transduction/physiology , Animals , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Homeostasis , Insulin-Like Growth Factor I/metabolism , Male , Neurodegenerative Diseases/genetics , Proteasome Inhibitors , Proteins/metabolism , Proteolysis , Pyramidal Cells/physiology , Rats, Wistar , Ubiquitin/physiology
12.
Nutrition ; 30(4): 466-72, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24607304

ABSTRACT

OBJECTIVE: Chronic low-grade inflammation in obesity is characterized by macrophage accumulation in white adipose tissue and adipokine production deregulation. Obesity also is characterized by oxidative stress related to inflammatory signaling. The aim of this study was to analyze whether dietary supplementation with a rice bran enzymatic extract (RBEE), rich in bioactive compounds with antioxidant and hypocholesterolemic properties, would ameliorate the inflammatory state existing in visceral adipose tissue of obese Zucker rats. METHODS: Obese Zucker rats and their littermate controls, lean Zucker rats ages 8 wk, were daily fed an enriched diet with either 1% or 5% RBEE supplementation over 20 wk. Measurement of adipocyte size and mRNA expression of proinflammatory molecules from visceral abdominal/epididymal tissue was performed. RESULTS: An RBEE-supplemented diet decreased the overproduction of tumor necrosis factor-α, interleukin (IL)-6, IL-1 ß, and inducible nitric oxide synthase (iNOS), as well as the overproduction of IL-6 and iNOs in visceral abdominal adipose tissue and visceral epididymal adipose tissue, respectively. An RBEE-supplemented diet modified the adipocyte-size distribution pattern in both abdominal and epididymal adipose tissue, shifting it toward smaller cell sizes. CONCLUSIONS: Chronic administration of a novel water-soluble RBEE, rich in polyphenols, tocotrienols and γ-oryzanol, could be a suitable treatment to ameliorate the obesity-associated proinflammatory response.


Subject(s)
Adipose Tissue/drug effects , Dietary Supplements , Inflammation/drug therapy , Obesity/complications , Oryza/chemistry , Phytotherapy , Plant Extracts/therapeutic use , Adipocytes/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Biomarkers/metabolism , Cytokines/metabolism , Epididymis/drug effects , Inflammation/metabolism , Intra-Abdominal Fat/metabolism , Male , Nitric Oxide Synthase Type II/metabolism , Obesity/metabolism , Plant Extracts/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Rats, Zucker , Seeds/chemistry
13.
Nat Genet ; 45(10): 1141-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23917401

ABSTRACT

Glioblastoma is one of the most challenging forms of cancer to treat. Here we describe a computational platform that integrates the analysis of copy number variations and somatic mutations and unravels the landscape of in-frame gene fusions in glioblastoma. We found mutations with loss of heterozygosity in LZTR1, encoding an adaptor of CUL3-containing E3 ligase complexes. Mutations and deletions disrupt LZTR1 function, which restrains the self renewal and growth of glioma spheres that retain stem cell features. Loss-of-function mutations in CTNND2 target a neural-specific gene and are associated with the transformation of glioma cells along the very aggressive mesenchymal phenotype. We also report recurrent translocations that fuse the coding sequence of EGFR to several partners, with EGFR-SEPT14 being the most frequent functional gene fusion in human glioblastoma. EGFR-SEPT14 fusions activate STAT3 signaling and confer mitogen independence and sensitivity to EGFR inhibition. These results provide insights into the pathogenesis of glioblastoma and highlight new targets for therapeutic intervention.


Subject(s)
Brain Neoplasms/genetics , Genomics , Glioblastoma/genetics , Catenins/genetics , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Mutation , Transcription Factors/genetics , Delta Catenin
14.
J Clin Invest ; 123(1): 405-17, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23241957

ABSTRACT

High-grade gliomas (HGGs) are incurable brain tumors that are characterized by the presence of glioma-initiating cells (GICs). GICs are essential to tumor aggressiveness and retain the capacity for self-renewal and multilineage differentiation as long as they reside in the perivascular niche. ID proteins are master regulators of stemness and anchorage to the extracellular niche microenvironment, suggesting that they may play a role in maintaining GICs. Here, we modeled the probable therapeutic impact of ID inactivation in HGG by selective ablation of Id in tumor cells and after tumor initiation in a new mouse model of human mesenchymal HGG. Deletion of 3 Id genes induced rapid release of GICs from the perivascular niche, followed by tumor regression. GIC displacement was mediated by derepression of Rap1gap and subsequent inhibition of RAP1, a master regulator of cell adhesion. We identified a signature module of 5 genes in the ID pathway, including RAP1GAP, which segregated 2 subgroups of glioma patients with markedly different clinical outcomes. The model-informed survival analysis together with genetic and functional studies establish that ID activity is required for the maintenance of mesenchymal HGG and suggest that pharmacological inactivation of ID proteins could serve as a therapeutic strategy.


Subject(s)
Glioma/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Models, Biological , Neoplasm Proteins/metabolism , Telomere-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Disease-Free Survival , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Deletion , Glioma/genetics , Glioma/mortality , Glioma/therapy , HEK293 Cells , Humans , Inhibitor of Differentiation Protein 1/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Shelterin Complex , Survival Rate , Telomere-Binding Proteins/genetics
15.
J Agric Food Chem ; 60(50): 12304-11, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23176387

ABSTRACT

Neuroinflammation is an important contributor to pathogenesis of age-related neurodegenerative disorders such as Alzheimer's or Parkinson's disease. Accumulating evidence indicates that inhibition of microglia-mediated neuroinflammation may become a reliable protective strategy for neurodegenerative processes. Flavonoids, widely distributed in the vegetable kingdom and in foods such as honey, have been suggested as novel therapeutic agents for the reduction of the deleterious effects of neuroinflammation. The present study investigated the potential protective effect of a honey flavonoid extract (HFE) on the production of pro-inflammatory mediators by lipopolysaccharide-stimulated N13 microglia. The results show that HFE significantly inhibited the release of pro-inflammatory cytokines such as TNF-α and IL-1ß. The expressions of iNOS and the production of reactive oxygen intermediates (ROS) were also significantly inhibited. Accordingly, the present study demonstrates that HFE is a potent inhibitor of microglial activation and thus a potential preventive-therapeutic agent for neurodegenerative diseases involving neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Flavonoids/pharmacology , Honey/analysis , Lipopolysaccharides/pharmacology , Microglia/drug effects , Animals , Cell Line , Chromatography, High Pressure Liquid , Cytokines/biosynthesis , Cytokines/metabolism , Flavonoids/isolation & purification , Mice , Microglia/metabolism , Reactive Oxygen Species/metabolism , Tandem Mass Spectrometry
16.
J Neurochem ; 123(4): 635-44, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22913583

ABSTRACT

Regulation of proteasome abundance to meet cell needs under stress conditions is critical for maintaining cellular homeostasis. However, the effects of aging on this homeostatic response remain unknown. In this report, we analyzed in young and aged rat hippocampus, the dynamics of proteasome recovery induced by proteasome stress. Proteasome inhibition in young rats leads to an early and coordinate transcriptional and translational up-regulation of both the catalytic subunits of constitutive proteasome and the proteasome maturation protein. By contrast, aged rats up-regulated the inducible catalytic subunits and showed a lower and shorter expression of proteasome maturation protein. This resulted in a faster recovery of proteasome activity in young rats. Importantly, proteasome inhibition highly affected pyramidal cells, leading to the accumulation of ubiquitinated proteins in perinuclear regions of aged, but not young pyramidal neurons. These data strongly suggest that age-dependent differences in proteasome level and composition could contribute to neurodegeneration induced by proteasome dysfunction in normal and pathological aging.


Subject(s)
Aging , Hippocampus/metabolism , Proteasome Endopeptidase Complex/metabolism , Up-Regulation/physiology , Age Factors , Animals , Catalytic Domain/physiology , Cell Nucleolus/metabolism , Hippocampus/cytology , Immunoproteins/metabolism , Male , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar
17.
Science ; 337(6099): 1231-5, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22837387

ABSTRACT

The brain tumor glioblastoma multiforme (GBM) is among the most lethal forms of human cancer. Here, we report that a small subset of GBMs (3.1%; 3 of 97 tumors examined) harbors oncogenic chromosomal translocations that fuse in-frame the tyrosine kinase coding domains of fibroblast growth factor receptor (FGFR) genes (FGFR1 or FGFR3) to the transforming acidic coiled-coil (TACC) coding domains of TACC1 or TACC3, respectively. The FGFR-TACC fusion protein displays oncogenic activity when introduced into astrocytes or stereotactically transduced in the mouse brain. The fusion protein, which localizes to mitotic spindle poles, has constitutive kinase activity and induces mitotic and chromosomal segregation defects and triggers aneuploidy. Inhibition of FGFR kinase corrects the aneuploidy, and oral administration of an FGFR inhibitor prolongs survival of mice harboring intracranial FGFR3-TACC3-initiated glioma. FGFR-TACC fusions could potentially identify a subset of GBM patients who would benefit from targeted FGFR kinase inhibition.


Subject(s)
Cell Transformation, Neoplastic , Fetal Proteins/genetics , Glioblastoma/genetics , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Aneuploidy , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Chromosomal Instability , Enzyme Inhibitors/pharmacology , Fetal Proteins/chemistry , Fetal Proteins/metabolism , Glioblastoma/metabolism , Humans , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Mitosis , Neoplasm Transplantation , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Oncogene Fusion , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Piperazines/pharmacology , Protein Structure, Tertiary , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/chemistry , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Spindle Apparatus/metabolism , Translocation, Genetic , Xenograft Model Antitumor Assays
18.
J Neuroinflammation ; 9: 87, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22559833

ABSTRACT

BACKGROUND: Neuroinflammation and protein accumulation are characteristic hallmarks of both normal aging and age-related neurodegenerative diseases. However, the relationship between these factors in neurodegenerative processes is poorly understood. We have previously shown that proteasome inhibition produced higher neurodegeneration in aged than in young rats, suggesting that other additional age-related events could be involved in neurodegeneration. We evaluated the role of lipopolysaccharide (LPS)-induced neuroinflammation as a potential synergic risk factor for hippocampal neurodegeneration induced by proteasome inhibition. METHODS: Young male Wistar rats were injected with 1 µL of saline or LPS (5 mg/mL) into the hippocampus to evaluate the effect of LPS-induced neuroinflammation on protein homeostasis. The synergic effect of LPS and proteasome inhibition was analyzed in young rats that first received 1 µL of LPS and 24 h later 1 µL (5 mg/mL) of the proteasome inhibitor lactacystin. Animals were sacrificed at different times post-injection and hippocampi isolated and processed for gene expression analysis by real-time polymerase chain reaction; protein expression analysis by western blots; proteasome activity by fluorescence spectroscopy; immunofluorescence analysis by confocal microscopy; and degeneration assay by Fluoro-Jade B staining. RESULTS: LPS injection produced the accumulation of ubiquitinated proteins in hippocampal neurons, increased expression of the E2 ubiquitin-conjugating enzyme UB2L6, decreased proteasome activity and increased immunoproteasome content. However, LPS injection was not sufficient to produce neurodegeneration. The combination of neuroinflammation and proteasome inhibition leads to higher neuronal accumulation of ubiquitinated proteins, predominant expression of pro-apoptotic markers and increased neurodegeneration, when compared with LPS or lactacystin (LT) injection alone. CONCLUSIONS: Our results identify neuroinflammation as a risk factor that increases susceptibility to neurodegeneration induced by proteasome inhibition. These results highlight the modulation of neuroinflammation as a mechanism for neuronal protection that could be relevant in situations where both factors are present, such as aging and neurodegenerative diseases.


Subject(s)
Hippocampus/drug effects , Lipopolysaccharides/toxicity , Nerve Degeneration/chemically induced , Proteasome Inhibitors/toxicity , Acetylcysteine/analogs & derivatives , Acetylcysteine/toxicity , Aging/drug effects , Aging/physiology , Animals , Drug Synergism , Hippocampus/enzymology , Hippocampus/pathology , Inflammation/chemically induced , Inflammation/epidemiology , Inflammation/pathology , Male , Nerve Degeneration/epidemiology , Nerve Degeneration/pathology , Proteasome Endopeptidase Complex/metabolism , Rats , Rats, Wistar , Ubiquitination/drug effects , Ubiquitination/physiology
19.
Nat Cell Biol ; 14(5): 477-87, 2012 Apr 22.
Article in English | MEDLINE | ID: mdl-22522171

ABSTRACT

Stem-cell functions require activation of stem-cell-intrinsic transcriptional programs and extracellular interaction with a niche microenvironment. How the transcriptional machinery controls residency of stem cells in the niche is unknown. Here we show that Id proteins coordinate stem-cell activities with anchorage of neural stem cells (NSCs) to the niche. Conditional inactivation of three Id genes in NSCs triggered detachment of embryonic and postnatal NSCs from the ventricular and vascular niche, respectively. The interrogation of the gene modules directly targeted by Id deletion in NSCs revealed that Id proteins repress bHLH-mediated activation of Rap1GAP, thus serving to maintain the GTPase activity of RAP1, a key mediator of cell adhesion. Preventing the elevation of the Rap1GAP level countered the consequences of Id loss on NSC-niche interaction and stem-cell identity. Thus, by preserving anchorage of NSCs to the extracellular environment, Id activity synchronizes NSC functions to residency in the specialized niche.


Subject(s)
Antigens, Neoplasm/physiology , Cell Adhesion/physiology , Neural Stem Cells/cytology , Animals , Antigens, Neoplasm/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , GTPase-Activating Proteins/genetics , Mice
20.
J Neuroimmunol ; 238(1-2): 44-51, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21803430

ABSTRACT

To elucidate whether density of cells could contribute to the extent of microglial activation, we performed in vitro assays using three different densities of N13 microglia stimulated with LPS. Our results showed that induction of pro-inflammatory factors as TNF-α and iNOS was directly related to cell density, meanwhile the induction of the anti-inflammatory IL-10 was inversely related to cell density. Accordingly, in vivo assays showed that after LPS-injection, iNOS expression was more intense in substantia nigra, a brain area showing specific susceptibility to neurodegeneration after microglia activation, whereas IL-10 expression was more sustained in striatum, an area resistant to damage. These results support that microglia density is pivotal to control the balance between pro- and anti-inflammatory factors release.


Subject(s)
Cytokines/metabolism , Encephalitis/chemically induced , Encephalitis/pathology , Lipopolysaccharides/toxicity , Microglia/drug effects , Nitric Oxide Synthase Type II/metabolism , Analysis of Variance , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Count , Cell Line, Transformed , Cytokines/genetics , Disease Models, Animal , Encephalitis/drug therapy , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation , Imidazoles/therapeutic use , Interleukin-10/genetics , Interleukin-10/metabolism , Isothiuronium/analogs & derivatives , Isothiuronium/therapeutic use , Male , Mice , Microglia/pathology , Nitric Oxide Synthase Type II/genetics , Pyridines/therapeutic use , RNA, Messenger/metabolism , Rats , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...