Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Org Biomol Chem ; 18(4): 725-737, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31912858

RESUMO

Nine cytotoxic [5/7] and [6/6] benzannulated steroid spiroketals were synthesized by palladium catalyzed spiroketalization of 5α and 5ß-alkynediols derived from testosterone, diosgenin and cholesterol. The regioselectivity of the spiroketalization reaction is decisively influenced by the α or ß-orientation of the hydroxyl group at C-5. NMR and single crystal X-ray diffraction corroborated the structure of the obtained compounds. Compounds bearing a cholestane skeleton exhibited higher cytotoxicity against U251 and T47D cells, and the BrdU incorporation assay suggests that the synthesized spiroketals inhibit cell proliferation.

2.
Genes (Basel) ; 14(12)2023 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-38137011

RESUMO

BACKGROUND: Traumatic spinal cord injury (SCI) is a disabling condition that affects millions of people around the world. Currently, no clinical treatment can restore spinal cord function. Comparison of molecular responses in regenerating to non-regenerating vertebrates can shed light on neural restoration. The axolotl (Ambystoma mexicanum) is an amphibian that regenerates regions of the brain or spinal cord after damage. METHODS: In this study, we compared the transcriptomes after SCI at acute (1-2 days after SCI) and sub-acute (6-7 days post-SCI) periods through the analysis of RNA-seq public datasets from axolotl and non-regenerating rodents. RESULTS: Genes related to wound healing and immune responses were upregulated in axolotls, rats, and mice after SCI; however, the immune-related processes were more prevalent in rodents. In the acute phase of SCI in the axolotl, the molecular pathways and genes associated with early development were upregulated, while processes related to neuronal function were downregulated. Importantly, the downregulation of processes related to sensorial and motor functions was observed only in rodents. This analysis also revealed that genes related to pluripotency, cytoskeleton rearrangement, and transposable elements (e.g., Sox2, Krt5, and LOC100130764) were among the most upregulated in the axolotl. Finally, gene regulatory networks in axolotls revealed the early activation of genes related to neurogenesis, including Atf3/4 and Foxa2. CONCLUSIONS: Immune-related processes are upregulated shortly after SCI in axolotls and rodents; however, a strong immune response is more noticeable in rodents. Genes related to early development and neurogenesis are upregulated beginning in the acute stage of SCI in axolotls, while the loss of motor and sensory functions is detected only in rodents during the sub-acute period of SCI. The approach employed in this study might be useful for designing and establishing regenerative therapies after SCI in mammals, including humans.


Assuntos
Ambystoma mexicanum , Traumatismos da Medula Espinal , Humanos , Animais , Ratos , Camundongos , Ambystoma mexicanum/genética , RNA-Seq , Roedores/genética , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/metabolismo , Perfilação da Expressão Gênica , Modelos Animais
3.
Front Cell Dev Biol ; 10: 884748, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36353512

RESUMO

Neurodegenerative diseases affect millions of people worldwide and there are currently no cures. Two types of common neurodegenerative diseases are Alzheimer's (AD) and Parkinson's disease (PD). Single-cell and single-nuclei RNA sequencing (scRNA-seq and snRNA-seq) have become powerful tools to elucidate the inherent complexity and dynamics of the central nervous system at cellular resolution. This technology has allowed the identification of cell types and states, providing new insights into cellular susceptibilities and molecular mechanisms underlying neurodegenerative conditions. Exciting research using high throughput scRNA-seq and snRNA-seq technologies to study AD and PD is emerging. Herein we review the recent progress in understanding these neurodegenerative diseases using these state-of-the-art technologies. We discuss the fundamental principles and implications of single-cell sequencing of the human brain. Moreover, we review some examples of the computational and analytical tools required to interpret the extensive amount of data generated from these assays. We conclude by highlighting challenges and limitations in the application of these technologies in the study of AD and PD.

4.
Front Endocrinol (Lausanne) ; 13: 703733, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35197928

RESUMO

Glioblastomas (GBM) are the most frequent and aggressive brain tumors. 17ß-estradiol (E2) increases proliferation, migration, and invasion of human GBM cells; however underlying mechanisms are no fully understood. Zeste 2 Enhancer Homologous enzyme (EZH2) is a methyltransferase part of Polycomb 2 repressor complex (PRC2). In GBM, EZH2 is overexpressed and involved in the cell cycle, migration, and invasion processes. We studied the role of EZH2 in the pro-oncogenic actions of E2 in human GBM cells. EZH2 gene silencing and pharmacological inhibition of EZH2 blocked proliferation, migration, and invasion of GBM cells induced by E2. We identified in silico additional putative estrogen response elements (EREs) at the EZH2 promoter, but E2 did not modify EZH2 expression. In silico analysis also revealed that among human GBM samples, EZH2 expression was homogeneous; in contrast, the heterogeneous expression of estrogen receptors (ERs) allowed the classification of the samples into groups. Even in the GBM cluster with high expression of ERs and those of their target genes, the expression of PCR2 target genes did not change. Overall, our data suggest that in GBM cells, pro-oncogenic actions of E2 are mediated by EZH2, without changes in EZH2 expression and by mechanisms that appear to be unrelated to the transcriptional activity of ERs.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste , Glioblastoma , Movimento Celular/genética , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Estradiol/farmacologia , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos
5.
Toxics ; 10(9)2022 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-36136478

RESUMO

Perinatal exposure to bisphenol A (BPA) in murine models has been reported to affect social behavior and increase anxiety. However, there is little information about the effects of BPA exposure during puberty, a period in which sex hormones influence the maturation and differentiation of the brain. In this work, we evaluated the effect of BPA administration during the juvenile stage (PND 21-50) on anxiety in male and female rats. Newly weaned Wistar rats were treated with BPA (0, 50, or 500 µg/kg/day) for 30 days. To compare the intra- and inter-sex behavioral profiles, rats were evaluated using four different anxiety models: the Open field test (OFT), the Elevated plus maze (EPM), the Light-dark box test (LDBT), and the Defensive burying test (DBT). Males exhibited a clear-cut anxious profile at both doses in all four tests, while no clear behavioral effect of BPA exposure was observed in female rats. The latter showed an altered estrous cycle that initiated earlier in life and had a shorter duration, with the estrous phase predominating. Moreover, the expression of ESR1, ESR2, GABRA1, GRIN1, GR, MR, and AR genes increased in the hippocampus and hypothalamus of male rats treated with 50 µg/kg, but not in females. Our results indicate that BPA consistently induces a higher anxiety profile in male than in female rats, as evidenced predominantly by an increase in passive-coping behaviors and changes in brain gene expression, highlighting the importance of sex in peripubertal behavioral toxicology studies.

6.
Methods Mol Biol ; 2174: 193-206, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32813251

RESUMO

Beyond cell proliferation, one of the most outstanding characteristics of the cancerous cells that promotes the tumoral progression is their high capacity to migrate and invade the surrounding healthy tissue. These cellular processes (migration and invasion) are critical steps to metastasis. Metastatic progression of the tumors is often the leading cause of morbidity and mortality in cancer patients. Critical genes and cell signaling pathways involved in cell migration and invasion of tumor cells have been identified, and several clinical efforts to alleviate cancer are focused on them; however, once the tumor has metastasized, it is extremely difficult to stop the progression of very aggressive forms of cancer such as glioblastomas. Therefore, it is crucial to elucidate the specific molecular mechanisms underlying tumor progression. In this chapter, we describe some methods to study tumor progression by assessing migration and cell invasion in 2D and 3D cell culture conditions.


Assuntos
Ensaios de Migração Celular/métodos , Neoplasias/patologia , Técnicas de Cultura de Tecidos/métodos , Humanos , Células Tumorais Cultivadas
7.
Cells ; 9(4)2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32295179

RESUMO

Oligodendrocytes are the myelinating cells of the central nervous system (CNS). These cells arise during the embryonic development by the specification of the neural stem cells to oligodendroglial progenitor cells (OPC); newly formed OPC proliferate, migrate, differentiate, and mature to myelinating oligodendrocytes in the perinatal period. It is known that progesterone promotes the proliferation and differentiation of OPC in early postnatal life through the activation of the intracellular progesterone receptor (PR). Progesterone supports nerve myelination after spinal cord injury in adults. However, the role of progesterone in embryonic OPC differentiation as well as the specific PR isoform involved in progesterone actions in these cells is unknown. By using primary cultures obtained from the embryonic mouse spinal cord, we showed that embryonic OPC expresses both PR-A and PR-B isoforms. We found that progesterone increases the proliferation, differentiation, and myelination potential of embryonic OPC through its PR by upregulating the expression of oligodendroglial genes such as neuron/glia antigen 2 (NG2), sex determining region Y-box9 (SOX9), myelin basic protein (MBP), 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNP1), and NK6 homeobox 1 (NKX 6.1). These effects are likely mediated by PR-B, as they are blocked by the silencing of this isoform. The results suggest that progesterone contributes to the process of oligodendrogenesis during prenatal life through specific activation of PR-B.


Assuntos
Sistema Nervoso Central/embriologia , Neurogênese/genética , Oligodendroglia/imunologia , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/metabolismo , Animais , Diferenciação Celular , Feminino , Camundongos
8.
Horm Cancer ; 11(2): 117-127, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32077034

RESUMO

Glioblastomas (GBM) are the most frequent and aggressive primary tumor of the central nervous system. In recent years, it has been proposed that sex hormones such as progesterone play an essential role in GBM biology. Membrane progesterone receptors (mPRs) are a group of G protein-coupled receptors with a wide distribution and multiple functions in the organism. There are five mPRs subtypes described in humans: mPRα, mPRß, mPRγ, mPRδ, and mPRε. It has been reported that human-derived GBM cells express the mPRα, mPRß, and mPRγ subtypes, and that progesterone promotes GBM progression in part by mPRα specific activation; however, it is still unknown if mPRδ and mPRε are also expressed in this type of tumor cells. In this study, we characterized the expression and hormonal regulation of mPRδ and mPRε in human GBM cells. We also analyzed a set of biopsies from TCGA. We found that the expression of these receptors is dependent on the tumor's grade and that mPRδ expression is directly correlated to patients' survival while the opposite is observed for mPRε. By RT-qPCR, Western blot, and immunofluorescence, the expression of mPRδ and mPRε was detected for the first time in human GBM cells. An in silico analysis showed possible progesterone response elements in the promoter regions of mPRδ and mPRε, and progesterone treatments downregulated the expression of these receptors. Our results suggest that mPRδ and mPRε are expressed in human GBM cells and that they are relevant to GBM biology.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Masculino , Gradação de Tumores , Prognóstico , Receptores de Progesterona/biossíntese , Receptores de Progesterona/genética
9.
Front Neurosci ; 13: 503, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31156378

RESUMO

Although progesterone is a steroid hormone mainly associated with female reproductive functions, such as uterine receptivity and maintenance of pregnancy, accumulating data have shown its physiological actions to extend to several non-reproductive functions in the central nervous system (CNS) both in males and females. In fact, progesterone is de novo synthesized in specific brain regions by neurons and glial cells and is involved in the regulation of various molecular and cellular processes underlying myelination, neuroprotection, neuromodulation, learning and memory, and mood. Furthermore, progesterone has been reported to be implicated in critical developmental events, such as cell differentiation and neural circuits formation. This view is supported by the increase in progesterone synthesis observed during pregnancy in both the placenta and the fetal brain. In the present review, we will focus on progesterone actions during CNS development.

10.
Mol Cell Endocrinol ; 477: 81-89, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29894708

RESUMO

BACKGROUND AND AIMS: Glioblastoma is the most frequent and aggressive brain tumor due to its high capacity to migrate and invade normal brain tissue. The steroid hormone progesterone (P4) contributes to the progression of glioblastoma by promoting proliferation, migration, and cellular invasion through the activation of its intracellular receptor (PR). However, the use of PR antagonist RU486 partially blocks the effects of P4, suggesting the participation of signaling pathways such as those mediated by membrane receptors to P4 (mPRs). Therefore, this study aimed to investigate the effects of mPRα subtype activation on proliferation, migration, and invasion of human glioblastoma cells. METHODS: We treated human glioblastoma cell lines U87 and U251 with the specific mPRα agonist Org OD 02-0, and evaluated its effects on cell number, proliferation, migration, and invasion. Additionally, we measured the phosphorylation of the kinases Src and Akt in both cell lines upon Org OD 02-0 treatment. RESULTS: Org OD 02-0 (100 nM) augmented the number of U87 and U251 cells by increasing cell proliferation. The treatment with this agonist also increased U87 and U251 cell migration and invasion. Both proliferation and cell invasion decreased when mPRα expression was silenced. Finally, we observed that Org OD 02-0 increased the content of p-Src and p-Akt in both cell lines. CONCLUSION: Our data suggest that P4 produces its effects in human glioblastoma progression not only by PR interaction but also through cell signaling pathways activated by mPRα.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Membrana Celular/metabolismo , Movimento Celular , Glioblastoma/metabolismo , Glioblastoma/patologia , Receptores de Progesterona/metabolismo , Neoplasias Encefálicas/enzimologia , Contagem de Células , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glioblastoma/enzimologia , Humanos , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Progesterona/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Progesterona/agonistas , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa