Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Eur J Neurosci ; 59(1): 154-173, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38057955

RESUMO

Autism spectrum disorder (ASD) is a diverse group of neurodevelopmental conditions with complex origins. Individuals with ASD present various neurobiological abnormalities, including an altered immune response in the central nervous system and other tissues. Animal models like the C58/J inbred mouse strain are used to study biological characteristics of ASD. This strain is considered an idiopathic autism model because of its demonstrated reduced social preference and repetitive behaviours. Notably, C58/J mice exhibit alterations in dendritic arbour complexity, density and dendritic spines maturation in the hippocampus and prefrontal cortex (PFC), but inflammatory-related changes have not been explored in these mice. In this study, we investigated proinflammatory markers in the hippocampus and PFC of adult male C58/J mice. We discovered elevated levels of interferon gamma (IFN-γ) and monocyte chemoattractant protein 1 (MCP-1) in the hippocampus, suggesting increased inflammation, alongside a reduction in the anti-inflammatory enzyme arginase 1 (ARG1). Conversely, the PFC displayed reduced levels of TNF-α and MCP-1. Microglial analysis revealed higher levels of transmembrane protein 119 (TMEM119) and increased microglial density in a region-specific manner of the autistic-like mice, particularly in the PFC and hippocampus. Additionally, an augmented expression of the fractalkine receptor CX3CR1 was observed in the hippocampus and PFC of C58/J mice. Microglial morphological analysis shows no evident changes in the hippocampus of mice with autistic-like behaviours versus wild-type strain. These region-specific changes can contribute to modulate processes like inflammation or synaptic pruning in the C58/J mouse model of idiopathic autism.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Camundongos , Masculino , Animais , Transtorno Autístico/metabolismo , Transtorno do Espectro Autista/metabolismo , Microglia/metabolismo , Camundongos Endogâmicos , Córtex Pré-Frontal/metabolismo , Hipocampo/metabolismo , Inflamação/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
2.
Horm Behav ; 164: 105593, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38909429

RESUMO

Autism Spectrum Disorder (ASD) is characterized by differences in social communication and interaction, as well as areas of focused interests and/or repetitive behaviors. Recent studies have highlighted a higher prevalence of endocrine and reproductive disturbances among females on the autism spectrum, hinting at potential disruptions within the hypothalamus-pituitary-ovary (HPO) axis. This research aims to explore the reproductive health disparities in ASD using an animal model of autism, the C58/J inbred mouse strain, with a focus on reproductive performance and hormonal profiles compared to the C57BL/6J control strain. Our findings revealed that the estrous cycle in C58/J females is disrupted, as evidenced by a lower frequency of complete cycles and a lack of cyclical release of estradiol and progesterone compared to control mice. C58/J females also exhibited poor performance in several reproductive parameters, including reproductive lifespan and fertility index. Furthermore, estrogen receptor alpha content showed a marked decrease in the hypothalamus of C58/J mice. These alterations in the estrous cycle, hormonal imbalances, and reduced reproductive function imply dysregulation in the HPO axis. Additionally, our in-silico study identified a group of genes involved in infertility carrying single-nucleotide polymorphisms (SNPs) in the C58/J strain, which also have human orthologs associated with autism. These findings could offer valuable insights into the molecular underpinnings of neuroendocrine axis disruption and reproductive issues observed in ASD.

3.
J Neurooncol ; 161(1): 165-174, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36525166

RESUMO

PURPOSE: Astrocytomas are a type of malignant brain tumor with an unfavorable clinical course. The impact of AGT and MGMT somatic variants in the prognosis of astrocytoma is unknown, and it is controversial for TP53. Moreover, there is a lack of knowledge regarding the molecular characteristics of astrocytomas in Mexican patients. METHODS: We studied 48 Mexican patients, men and women, with astrocytoma (discovery cohort). We performed DNA deep sequencing in tumor samples, targeting AGT, MGMT and TP53, and we studied MGMT gene promoter methylation status. Then we compared our findings to a cohort which included data from patients with astrocytoma from The Cancer Genome Atlas (validation cohort). RESULTS: In the discovery cohort, we found a higher number of somatic variants in AGT and MGMT than in the validation cohort (10.4% vs < 1%, p < 0.001), and, in both cohorts, we observed only women carried variants AGT variants. We also found that the presence of either MGMT variant or promoter methylation was associated to better survival and response to chemotherapy, and, in conjunction with TP53 variants, to progression-free survival. CONCLUSIONS: The occurrence of AGT variants only in women expands our knowledge about the molecular differences in astrocytoma between men and women. The increased prevalence of AGT and MGMT variants in the discovery cohort also points towards possible distinctions in the molecular landscape of astrocytoma among populations. Our findings warrant further study.


Assuntos
Astrocitoma , Neoplasias Encefálicas , Feminino , Humanos , Masculino , Astrocitoma/patologia , Biomarcadores , Neoplasias Encefálicas/patologia , DNA/uso terapêutico , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Mutação , Prognóstico , Análise de Sequência de DNA , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
4.
Nutr Neurosci ; 25(2): 387-410, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32338174

RESUMO

Objective: This work aimed to compile information about the neuronal processes in which polyunsaturated fatty acids (PUFAs) could modulate brain plasticity, in order to analyze the role of nutritional intervention with the ω-3 and ω-6 fatty acids as a therapeutic strategy for the Autism Spectrum Disorder (ASD)-related signs and symptoms.Methods: We reviewed different articles reporting the effect of PUFAS on neurite elongation, membrane expansion, cytoskeleton rearrangement and neurotransmission, considering the ASD-related abnormalities in these processes.Results: In accordance to the reviewed studies, it is clear that ASD is one of the neurological conditions associated with an impairment in neuronal plasticity; therefore, PUFAs-rich diet improvements on cognition and behavioral deficits in individuals with autism, could be involved with the regulation of neuronal processes implicated in the atypical brain plasticity related with this neurodevelopmental disorder.Discussion: The behavioral and cognitive improvement observed in individuals with ASD after PUFAs treatment might underlie, at least in part, in the ability of ω-3 and ω-6 fatty acids to induce neurite outgrowth, probably, through the dynamic regulation of the neuronal cytoskeleton along with the expansion of neuronal membranes. Furthermore, it might also be associated with an enhancement of the efficacy of synaptic transmission and the modulation of neurotransmitters release.


Assuntos
Transtorno do Espectro Autista , Ácidos Graxos Ômega-3 , Transtorno do Espectro Autista/tratamento farmacológico , Cognição , Ácidos Graxos Ômega-3/uso terapêutico , Ácidos Graxos Insaturados , Humanos , Plasticidade Neuronal
5.
Mediators Inflamm ; 2017: 9173090, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29430083

RESUMO

Lysophosphatidic acid (LPA) is a ubiquitous lysophospholipid and one of the main membrane-derived lipid signaling molecules. LPA acts as an autocrine/paracrine messenger through at least six G protein-coupled receptors (GPCRs), known as LPA1-6, to induce various cellular processes including wound healing, differentiation, proliferation, migration, and survival. LPA receptors and autotaxin (ATX), a secreted phosphodiesterase that produces this phospholipid, are overexpressed in many cancers and impact several features of the disease, including cancer-related inflammation, development, and progression. Many ongoing studies aim to understand ATX-LPA axis signaling in cancer and its potential as a therapeutic target. In this review, we discuss the evidence linking LPA signaling to cancer-related inflammation and its impact on cancer progression.


Assuntos
Inflamação/etiologia , Lisofosfolipídeos/fisiologia , Neoplasias/etiologia , Diester Fosfórico Hidrolases/fisiologia , Humanos , Lisofosfolipídeos/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Receptores de Ácidos Lisofosfatídicos/fisiologia , Transdução de Sinais/fisiologia
6.
Sci Rep ; 14(1): 4608, 2024 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409172

RESUMO

Individuals with autism spectrum disorder (ASD) often exhibit atypical hippocampal anatomy and connectivity throughout their lifespan, potentially linked to alterations in the neurogenic process within the hippocampus. In this study, we performed an in-silico analysis to identify single-nucleotide polymorphisms (SNPs) in genes relevant to adult neurogenesis in the C58/J model of idiopathic autism. We found coding non-synonymous (Cn) SNPs in 33 genes involved in the adult neurogenic process, as well as in 142 genes associated with the signature genetic profile of neural stem cells (NSC) and neural progenitors. Based on the potential alterations in adult neurogenesis predicted by the in-silico analysis, we evaluated the number and distribution of newborn neurons in the dentate gyrus (DG) of young adult C58/J mice. We found a reduced number of newborn cells in the whole DG, a higher proportion of early neuroblasts in the subgranular layer (SGZ), and a lower proportion of neuroblasts with morphological maturation signs in the granule cell layer (GCL) of the DG compared to C57BL/6J mice. The observed changes may be associated with a delay in the maturation trajectory of newborn neurons in the C58/J strain, linked to the Cn SNPs in genes involved in adult hippocampal neurogenesis.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Camundongos , Animais , Transtorno Autístico/genética , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Hipocampo/fisiologia , Neurogênese/genética , Polimorfismo Genético , Giro Denteado/fisiologia
7.
Biochim Biophys Acta ; 1823(2): 379-86, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22142990

RESUMO

Estradiol (E2) regulates several cellular functions through the interaction with estrogen receptor subtypes, ERα and ERß, which present different functional and regulation properties. ER subtypes have been identified in human astrocytomas, the most common and aggressive primary brain tumors. We studied the role of ER subtypes in cell growth of two human astrocytoma cell lines derived from tumors of different evolution grades: U373 and D54 (grades III and IV, respectively). E2 significantly increased the number of cells in both lines and the co-administration with an ER antagonist (ICI 182, 780) significantly blocked E2 effects. ERα was the predominant subtype in both cell lines. E2 and ICI 182, 780 down-regulated ERα expression. The number of U373 and D54 cells significantly increased after PPT (ERα agonist) treatment but not after DPN (ERß agonist) one. To determine the role of SRC-1 and SRC-3 coactivators in ERα induced cell growth, we silenced them with RNA interference. Coactivator silencing blocked the increase in cell number induced by PPT. The content of proteins involved in proliferation and metastasis was also determined after PPT treatment. Western blot analysis showed that in U373 cells the content of PR isoforms (PR-A and PR-B), EGFR, VEGF and cyclin D1 increased after PPT treatment while in D54 cells only the content of EGFR was increased. Our results demonstrate that E2 induces cell growth of human astrocytoma cell lines through ERα and its interaction with SRC-1 and SRC-3 and also suggest differential roles of ERα on cell growth depending on astrocytoma grade.


Assuntos
Astrocitoma/fisiopatologia , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Coativador 1 de Receptor Nuclear/metabolismo , Coativador 3 de Receptor Nuclear/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/fisiologia , Ciclina D1/genética , Ciclina D1/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Receptor alfa de Estrogênio/genética , Humanos , Coativador 1 de Receptor Nuclear/genética , Coativador 3 de Receptor Nuclear/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Gen Comp Endocrinol ; 189: 51-8, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23660445

RESUMO

Sex steroids participate in the regulation of reproduction in female chickens. In this work, we determined the content of androgen receptor (AR), intracellular progesterone receptor isoforms (PR-A and PR-B), membrane progesterone receptor γ (mPRγ) and estrogen receptor α (ER-α) in the left growing and right regressing ovaries of Gallus domesticus from 13-day-old chicken embryos to 1-month-old chickens by western blot analysis. A marked difference in the morphological characteristics of the left and the right ovaries during development was observed. Results show a higher content of AR in the left ovary than in the right one in all ages. In the left ovary, the highest content of AR was observed on day 13 of embryonic development, and diminished with age. In the right ovary, AR was expressed from day 13 of embryonic development to 1-day-old, and became undetectable at 1-week and 1-month-old. In the left ovary, PR isoforms were not detected on day 13 of embryonic development, but they presented a marked expression after hatching. In the right ovary, the highest expression of both PR isoforms was found on 1-day-old, and significantly decreased with age. PR-B was the predominant isoform on 1-day and 1-month old in the left ovary, whereas PR-A was the predominant one on day 13 of embryonic development in the right ovary. Interestingly, mPRγ was detected at 1-week and 1-month-old in the left ovary meanwhile in the right ovary, it was detected from day 13 of embryonic development to 1-day-old. ER-α was only detected in the left ovary from day 13 to 1-week-old, while in 1-month-old chickens, it was expressed in both ovaries. In the left ovary, ER-α content was lower from 1-day to 1-month-old as compared with day 13 of embryonic development. Our results demonstrate a differential expression of sex steroid hormone receptors between the left growing and the right regressing ovary, and throughout chickens' age; and this is the first report about mPR expression in birds.


Assuntos
Ovário/metabolismo , Animais , Embrião de Galinha , Galinhas , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores Androgênicos/metabolismo , Receptores de Progesterona/metabolismo
9.
Cell Signal ; 92: 110246, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35033667

RESUMO

Medulloblastoma (MB) is the most common and aggressive pediatric intracranial tumor. Estrogen receptor ß (ERß) expression correlates with MB development and its phosphorylation modifies its transcriptional activity in a ligand-dependent or independent manner. Using in silico tools, we have identified several residues in ERß protein as potential targets of protein kinases C (PKCs) α and δ. Using Daoy cells, we observed that PKCα and PKCδ associate with ERß and induce its phosphorylation. The activation of ERß promotes MB cells proliferation and invasion, and PKCs downregulation dysregulates these steroid receptor mediated processes. Our data suggest that these kinases may play a crucial role in the regulation of the ERß transcriptional activity. Overexpression of both PKCα and PKCδ in MB biopsies samples supports their relevance in MB progression.


Assuntos
Neoplasias Cerebelares , Receptor beta de Estrogênio , Meduloblastoma , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/metabolismo , Proteína Quinase C , Linhagem Celular Tumoral , Proliferação de Células , Criança , Receptor alfa de Estrogênio , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo
10.
Horm Behav ; 59(1): 37-43, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20950620

RESUMO

We studied the role of progesterone receptor (PR) in the display of female sexual behavior during postpartum estrus in rats. Adult female rats were treated with the PR antagonist, RU486 (1.25 and 5 mg), 3 h after parturition and sexual behavior was evaluated throughout the first postpartum day. Estradiol and progesterone serum levels changed during the first 24 h postpartum. The highest estradiol and progesterone levels were found at 9 and 12 h postpartum, respectively. The predominant PR isoform in the hypothalamus and the preoptic area was PR-A during postpartum day. The content of PR-A increased at 6 h postpartum in the hypothalamus and the preoptic area, and decreased in both regions at 9 h. PR-B content only increased in the preoptic area at 12 h postpartum. The highest display of lordotic and proceptive behaviors were found at 12 h postpartum. The treatment with 1.25 and 5 mg of RU486 respectively reduced lordosis by 61% and 92% at 12 h postpartum. These results suggest that PR is essential in the display of postpartum estrus in rats.


Assuntos
Estro/metabolismo , Hipotálamo/metabolismo , Período Pós-Parto/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/metabolismo , Comportamento Sexual Animal/fisiologia , Análise de Variância , Animais , Western Blotting , Estradiol/sangue , Estro/efeitos dos fármacos , Feminino , Hipotálamo/efeitos dos fármacos , Mifepristona/farmacologia , Período Pós-Parto/efeitos dos fármacos , Postura/fisiologia , Progesterona/sangue , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Comportamento Sexual Animal/efeitos dos fármacos , Fatores de Tempo
11.
Front Endocrinol (Lausanne) ; 12: 640298, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841333

RESUMO

Glioblastomas are the most common and aggressive primary brain tumors in adults, and patients with glioblastoma have a median survival of 15 months. Some alternative therapies, such as Src family kinase inhibitors, have failed presumably because other signaling pathways compensate for their effects. In the last ten years, it has been proven that sex hormones such as progesterone (P4) can induce growth, migration, and invasion of glioblastoma cells through its intracellular progesterone receptor (PR), which is mostly known for its role as a transcription factor, but it can also induce non-genomic actions. These non-classic actions are, in part, a consequence of its interaction with cSrc, which plays a significant role in the progression of glioblastomas. We studied the relation between PR and cSrc, and its effects in human glioblastoma cells. Our results showed that P4 and R5020 (specific PR agonist) activated cSrc protein since both progestins increased the p-cSrc (Y416)/cSrc ratio in U251 and U87 human glioblastoma derived cell lines. When siRNA against the PR gene was used, the activation of cSrc by P4 was abolished. The co-immunoprecipitation assay showed that cSrc and PR interact in U251 cells. P4 treatment also promoted the increase in the p-Fak (Y397) (Y576/577)/Fak and the decrease in p-Paxillin (Y118)/Paxillin ratio, which are significant components of the focal adhesion complex and essential for migration and invasion processes. A siRNA against cSrc gene blocked the increase in the p-Fak (Y576/Y577)/Fak ratio and the migration induced by P4, but not the decrease in p-Paxillin (Y118)/Paxillin ratio. We analyzed the potential role of cSrc over PR phosphorylation in three databases, and one putative tyrosine residue in the amino acid 87 of PR was found. Our results showed that P4 induces the activation of cSrc protein through its PR. The latter and cSrc could interact in a bidirectional mode for regulating the activity of proteins involved in migration and invasion of glioblastomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Proteína Tirosina Quinase CSK/metabolismo , Glioblastoma/metabolismo , Receptores de Progesterona/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Humanos , Invasividade Neoplásica , Paxilina/metabolismo , Fosforilação , Progesterona/metabolismo , Proteínas Tirosina Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirosina/química
12.
Front Cell Neurosci ; 15: 726501, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34616277

RESUMO

Autism spectrum disorder (ASD) has a broad range of neurobiological characteristics, including alterations in dendritic spines, where approximately 90% of excitatory synapses occur. Therefore, changes in their number or morphology would be related to atypical brain communication. The C58/J inbred mouse strain displays low sociability, impaired communication, and stereotyped behavior; hence, it is considered among the animal models suitable for the study of idiopathic autism. Thus, this study aimed to evaluate the dendritic spine differences in the hippocampus and the prefrontal cortex of C58/J mice. We found changes in the number of spines and morphology in a brain region-dependent manner: a subtle decrease in spine density in the prefrontal cortex, higher frequency of immature phenotype spines characterized by filopodia-like length or small morphology, and a lower number of mature phenotype spines with mushroom-like or wide heads in the hippocampus. Moreover, an in silico analysis showed single nucleotide polymorphisms (SNPs) at genes collectively involved in regulating structural plasticity with a likely association with ASD, including MAP1A (Microtubule-Associated Protein 1A), GRM7 (Metabotropic Glutamate Receptor, 7), ANKRD11 (Ankyrin Repeat Domain 11), and SLC6A4 (Solute Carrier Family 6, member 4), which might support the relationship between the C58/J strain genome, an autistic-like behavior, and the observed anomalies in the dendritic spines.

13.
Onco Targets Ther ; 14: 3757-3768, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34168461

RESUMO

INTRODUCTION: Astrocytomas are the most common and aggressive primary brain tumors, and they are classified according to the degree of malignancy on a scale of I to IV, in which grade I is the least malignant and grade IV the highest. Many factors are related to astrocytomas progression as progesterone receptor (PR), whose transcriptional activity could be regulated by phosphorylation by protein kinase C alpha (PKCα) at the residue Ser400. Our aim was to investigate if PR phosphorylation together with PKCα expression could be used as a prognostic factor for astrocytomas malignancy. METHODS: By immunofluorescence, we detected the content of PKCα, PR and its phosphorylation at Ser400 in 46 biopsies from Mexican patients with different astrocytoma malignancy grades; by bioinformatic tools using TCGA data, we evaluated the expression of PR and PKCα mRNA according to astrocytoma malignancy grades. For all statistical analyses, significance was p<0.05. RESULTS: We detected a positive correlation between the tumor grade and the content of PKCα, PR and its phosphorylation at Ser400, as well as the intracellular colocalization of these proteins. Interestingly, using an in silico assay, we found that the PR and PKCα expression at mRNA level has an inverse ratio with astrocytomas tumor grade. DISCUSSION: These results indicate that PR and its phosphorylation at Ser400 site, as well as PKCα and their colocalization, could be considered as possible malignancy biomarkers for astrocytomas grades I-IV.

14.
Cells ; 10(4)2021 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-33916643

RESUMO

Lysophosphatidic acid (LPA) induces a wide range of cellular processes and its signaling is increased in several cancers including glioblastoma (GBM), a high-grade astrocytoma, which is the most common malignant brain tumor. LPA1 receptor is expressed in GBM cells and its signaling pathways activate protein kinases C (PKCs). A downstream target of PKC, involved in GBM progression, is the intracellular progesterone receptor (PR), which can be phosphorylated by this enzyme, increasing its transcriptional activity. Interestingly, in GBM cells, PKCα isotype translocates to the nucleus after LPA stimulation, resulting in an increase in PR phosphorylation. In this study, we determined that LPA1 receptor activation induces protein-protein interaction between PKCα and PR in human GBM cells; this interaction increased PR phosphorylation in serine400. Moreover, LPA treatment augmented VEGF transcription, a known PR target. This effect was blocked by the PR selective modulator RU486; also, the activation of LPA1/PR signaling promoted migration of GBM cells. Interestingly, using TCGA data base, we found that mRNA expression of LPAR1 increases according to tumor malignancy and correlates with a lower survival in grade III astrocytomas. These results suggest that LPA1/PR pathway regulates GBM progression.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteína Quinase C-alfa/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Lisofosfolipídeos/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Cells ; 9(9)2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32825553

RESUMO

The mesenchymal phenotype of glioblastoma multiforme (GBM), the most frequent and malignant brain tumor, is associated with the worst prognosis. The epithelial-mesenchymal transition (EMT) is a cell plasticity mechanism involved in GBM malignancy. In this study, we determined 17ß-estradiol (E2)-induced EMT by changes in cell morphology, expression of EMT markers, and cell migration and invasion assays in human GBM-derived cell lines. E2 (10 nM) modified the shape and size of GBM cells due to a reorganization of actin filaments. We evaluated EMT markers expression by RT-qPCR, Western blot, and immunofluorescence.We found that E2 upregulated the expression of the mesenchymal markers, vimentin, and N-cadherin. Scratch and transwell assays showed that E2 increased migration and invasion of GBM cells. The estrogen receptor-α (ER-α)-selective agonist 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT, 10 nM) affected similarly to E2 in terms of the expression of EMT markers and cell migration, and the treatment with the ER-α antagonist methyl-piperidino-pyrazole (MPP, 1 µM) blocked E2 and PPT effects. ER-ß-selective agonist diarylpropionitrile (DNP, 10 nM) and antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazole[1,5-a]pyrimidin-3-yl]phenol (PHTPP, 1 µM) showed no effects on EMT marker expression. These data suggest that E2 induces EMT activation through ER-α in human GBM-derived cells.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Estradiol/uso terapêutico , Estrogênios/uso terapêutico , Glioblastoma/tratamento farmacológico , Estradiol/farmacologia , Estrogênios/farmacologia , Glioblastoma/patologia , Humanos
16.
Drug Des Devel Ther ; 14: 1799-1811, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32494122

RESUMO

INTRODUCTION: Cancer stem cells (CSCs) drive the initiation, maintenance, and therapy response of breast tumors. CD49f is expressed in breast CSCs and functions in the maintenance of stemness. Thus, blockade of CD49f is a potential therapeutic approach for targeting breast CSCs. In the present study, we aimed to repurpose drugs as CD49f antagonists. MATERIALS AND METHODS: We performed consensus molecular docking using a subdomain of CD49f that is critical for heterodimerization and a collection of pharmochemicals clinically tested. Molecular dynamics simulations were employed to further characterize drug-target binding. Using MDA-MB-231 cells, we evaluated the effects of potential CD49f antagonists on 1) cell adhesion to laminin; 2) mammosphere formation; and 3) cell viability. We analyzed the effects of the drug with better CSC-selectivity on the activation of CD49f-downstream signaling by Western blot (WB) and co-immunoprecipitation. Expressions of the stem cell markers CD44 and SOX2 were analyzed by flow cytometry and WB, respectively. Transactivation of SOX2 promoter was evaluated by luciferase reporter assays. Changes in the number of CSCs were assessed by limiting-dilution xenotransplantation. RESULTS: Pranlukast, a drug used to treat asthma, bound to CD49f in silico and inhibited the adhesion of CD49f+ MDA-MB-231 cells to laminin, indicating that it antagonizes CD49f-containing integrins. Molecular dynamics analysis showed that pranlukast binding induces conformational changes in CD49f that affect its interaction with ß1-integrin subunit and constrained the conformational dynamics of the heterodimer. Pranlukast decreased the clonogenicity of breast cancer cells on mammosphere formation assay but had no impact on the viability of bulk tumor cells. Brief exposure of MDA-MB-231 cells to pranlukast altered CD49f-dependent signaling, reducing focal adhesion kinase (FAK) and phosphatidylinositol 3-kinase (PI3K) activation. Further, pranlukast-treated cells showed decreased CD44 and SOX2 expression, SOX2 promoter transactivation, and in vivo tumorigenicity, supporting that this drug reduces the frequency of CSC. CONCLUSION: Our results support the function of pranlukast as a CD49f antagonist that reduces the CSC population in triple-negative breast cancer cells. The pharmacokinetics and toxicology of this drug have already been established, rendering a potential adjuvant therapy for breast cancer patients.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Cromonas/farmacologia , Integrina alfa6/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Antineoplásicos/química , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Cromonas/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Células Tumorais Cultivadas
17.
Biochim Biophys Acta ; 1783(2): 253-62, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18166159

RESUMO

17beta-Estradiol induced LPA(1) receptor desensitization in C9 cells stably expressing LPA(1) receptors and transiently expressing estrogen receptor alpha. Such desensitization was evidenced by a reduction in lysophosphatidic acid-mediated Ca(2+)mobilization and it was associated to receptor phosphorylation and internalization. These effects of 17beta-estradiol were rapid (taking place over 5 min) and were blocked by the estrogen receptor antagonist ICI 182780. Similarly, inhibitors of phosphoinositide 3-kinase (wortmannin and LY294002) and of protein kinase C (staurosporine and Gö 6976) blocked 17beta-estradiol-induced LPA(1) receptor desensitization and phosphorylation. Confocal microscopy evidenced LPA(1) receptor internalization in response to 17beta-estradiol treatment. Association between LPA(1) receptors and protein kinase C alpha was suggested by co-immunoprecipitation assays. Protein kinase C alpha was associated with LPA(1) receptors in the absence of stimulus and such association further increased in a dynamic fashion in response to 17beta-estradiol. The results demonstrated that in C9 cells estrogens modulate LPA(1) action through estrogen receptor alpha with the participation of protein kinase C alpha and phosphoinositide 3-kinase.


Assuntos
Estrogênios/farmacologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Endocitose/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Humanos , Imunoprecipitação , Lisofosfolipídeos/farmacologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteína Quinase C/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares/efeitos dos fármacos , Transfecção
18.
J Neurosci Res ; 87(2): 470-81, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18752305

RESUMO

Several growth factors, such as vascular endothelial growth factor, brain-derived neurotrophic factor, and insulin-like growth factor-I are involved in the actions of progesterone in the central nervous system. Previous studies in neuronal and glial cultures have shown that progesterone may regulate growth factor signaling, increasing the phosphorylation of extracellular-signal regulated kinase (ERK) and the phosphorylation of Akt, components of the mitogen-activated protein kinase (MAPK) and the phosphoinositide-3 kinase (PI3K) signaling pathways, respectively. In this study, we have evaluated whether progesterone and its reduced metabolites, dihydroprogesterone and tetrahydroprogesterone, regulate PI3K and MAPK signaling in the brain of ovariectomized rats in vivo. Significant increases in the phosphorylation of ERK, in the expression of the catalytic (p110) and the regulatory (p85) subunits of PI3K and in the phosphorylation of Akt were observed in the hypothalamus, the hippocampus, and the cerebellum 24 hr after progesterone administration. Progesterone metabolites partially mimicked the effect of progesterone and had a stronger effect on MAPK and PI3K signaling in the hypothalamus than in the other brain regions. These findings suggest that progesterone regulates MAPK and PI3K signaling pathways in the central nervous system in vivo by direct hormonal actions and by mechanisms involving progesterone metabolites.


Assuntos
Encéfalo/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Progesterona/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Feminino , Ovariectomia , Ratos , Ratos Wistar
19.
J Steroid Biochem Mol Biol ; 113(1-2): 80-4, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19095059

RESUMO

Progesterone regulates several functions through the interaction with its intracellular receptor (PR) which expresses two isoforms with different functions and regulation: PR-A and PR-B. Both PR isoforms have been detected in human astrocytomas, the most common and aggressive primary brain tumours, but their regulation and function are unknown. We studied the effects of estradiol, progesterone and their receptor antagonists (ICI 182,780 and RU 486) on PR isoforms content in U373 and D54 human astrocytoma cell lines, respectively derived from grades III and IV astrocytomas, by Western blot analysis. In U373 cells we also evaluated the effects of PR-A overexpression on cell growth. We observed that in U373 cells estradiol increased the content of both PR isoforms whereas in D54 cells it had no effects. Estradiol effects were blocked by ICI 182,780. In both cell lines, PR isoforms content was down-regulated by progesterone after estradiol treatment. This effect was blocked by RU 486. We observed that overexpression of PR-A significantly diminished the increase in U373 cells number produced after progesterone treatment. Our results suggest a differential PR isoforms regulation depending on the evolution grade of human astrocytoma cells, and an inhibitory role of PR-A on progesterone effects on astrocytomas cell growth.


Assuntos
Astrocitoma/metabolismo , Receptores de Progesterona/metabolismo , Astrocitoma/patologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Isoformas de Proteínas/metabolismo , Transfecção
20.
Int J Biochem Cell Biol ; 110: 91-102, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30849522

RESUMO

Lysophosphatidic acid (LPA) is a ubiquitous lysophospholipid that induces a wide range of cellular processes such as wound healing, differentiation, proliferation, migration, and survival. LPA signaling is increased in a number of cancers. In Glioblastoma (GBM), the most aggressive brain tumor, autotaxin the enzyme that produces LPA and its receptor LPA1 are overexpressed. LPA1 is preferentially couple to Gαq proteins in these tumors that in turn activates PKCs. PKCs are involved in many cellular processes including proliferation and metastasis. In this study, we aimed to determine if a classical PKC (α isozyme), could be activated through LPA1 in GBM cell lines and if this activation impacts on cell number. We found that LPA1 induces PKCα translocation to the nucleus, but not to the cell membrane after LPA treatment and the cell number diminished when LPA1/PKCα signaling was blocked, suggesting a relevant role of LPA1 and PKCα in GBM growth.


Assuntos
Núcleo Celular/metabolismo , Glioblastoma/patologia , Proteína Quinase C-alfa/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transporte Ativo do Núcleo Celular , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Diester Fosfórico Hidrolases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA