Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1133883, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37266424

RESUMO

Introduction: The presence of soluble human programmed cell death-ligand 1 (shPD-L1) in the blood of patients with cancer has been reported to be negatively correlated with disease prognosis. However, little information exists about the mechanisms underlying high levels of shPD-L1 for promoting disease progression. Methods: In this study, we first analyzed the correlations between shPD-L1 and apoptosis of T cells in patients with cancer, then tested the effect of shPD-L1 on T-cell functions and the production of regulatory T cells. Results: We found that the apoptosis of human peripheral PD-1+CD4+ T cells was significantly elevated in patients with cancer compared with healthy donors and was positively correlated with circulating PD-L1 levels in patients with cancer. In vitro, monomeric shPD-L1 significantly inhibited the proliferation, cytokine secretion, and cancer cell-killing activity of peripheral blood mononuclear cells (PBMCs) activated by either agonist antibodies or HATac (high-affinity T cell activation core)-NYE (NY-ESO-1 antigen). It also promoted CD4+ T cells to express forkhead family transcription factor 3 (FoxP3) for the conversion of induced T regulatory cells, which was more significant than that mediated by soluble human PD-L1 fusion protein (shPD-L1-Fc). Discussion: These results confirm that soluble PD-L1 could be a candidate for inhibiting the functions of activated T cells, promoting peripheral tolerance to tumor cells, and implicating in system tumor immune escape in addition to the tumor microenvironment. This is an important mechanism explaining the negative correlation between peripheral blood PD-L1 levels and cancer prognosis. Therefore, understanding the roles of hPD-L1 in peripheral blood will be helpful for the development of precision immunotherapy programs in treating various tumors.


Assuntos
Antígeno B7-H1 , Neoplasias , Humanos , Antígeno B7-H1/metabolismo , Leucócitos Mononucleares/metabolismo , Ligantes , Apoptose , Microambiente Tumoral
2.
Scand J Trauma Resusc Emerg Med ; 30(1): 59, 2022 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-36397136

RESUMO

BACKGROUND: Decompressive craniectomy (DC) and intracranial pressure (ICP) monitoring are common approaches to reduce the death rate of Traumatic brain injury (TBI) patients, but the outcomes of these patients are unfavorable, particularly those who receive bilateral DC. The authors discuss their experience using ICP and other potential methods to improve the outcomes of TBI patients who receive bilateral DC. METHODS: Data from TBI patients receiving bilateral DC from Jan. 2008 to Jan. 2022 were collected via a retrospective chart review. Included patients who received unplanned contralateral DC after initial surgery were identified as unplanned secondary surgery (USS) patients. Patients' demographics and baseline medical status; pre-, intra-, and postoperative events; and follow-up visit outcome data were analyzed. RESULTS: A total of 151 TBI patients were included. Patients who underwent USS experienced more severe outcomes as assessed using the 3-month modified Rankin Scale score (P = 0.024). In bilateral DC TBI patients, USS were associated with worsen outcomes, moreover, ICP monitoring was able to lower their death rate and was associated with a lower USS incidence. In USS patients, ICP monitoring was not associated with improved outcomes but was able to lower their mortality rate (2/19, 10.5%, vs. 10/25, 40.0%; P = 0.042). CONCLUSION: The avoidance of USS may be associated with improved outcomes of TBI patients who underwent bilateral DC. ICP monitoring was a potential approach to lower USS rate in TBI patients, but its specific benefits were uncertain.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Craniectomia Descompressiva , Humanos , Craniectomia Descompressiva/métodos , Pressão Intracraniana , Estudos Retrospectivos , Resultado do Tratamento , Lesões Encefálicas Traumáticas/cirurgia
3.
J Immunother Cancer ; 8(2)2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33323464

RESUMO

BACKGROUND: In patients with hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC), virus-specific cytotoxic T lymphocytes (CTLs) fail to eliminate HCC cells expressing HBV antigens. As the expression of viral antigen in HBV-associated HCC may decrease to allow tumor to escape immune attacks, we hypothesized that an HBV surface antigen (HBsAg)-specific affinity-improved-T-cell receptor (TCR) will enable T cells to target HCC more effectively than corresponding wild-type-TCR. We also postulated that TCR promiscuity can be exploited to efficiently capture HBV variants that can hinder CTL-based therapeutics. METHODS: We applied flexi-panning to isolate affinity-improved TCRs binding to a variant antigen, the human leukocyte antigen (HLA)-A*02:01-restricted nonapeptide HBs371-379-ILSPFLPLL, from libraries constructed with a TCR cloned using the decapeptide HBs370-379-SIVSPFIPLL. The potency and safety of the affinity-improved-TCR engineered T-cells (Ai-TCR-T) were verified with potentially cross-reactive human and HBV-variant peptides, tumor and normal cells, and xenograft mouse models. RESULTS: Ai-TCR-T cells retained cognate HBV antigen specificity and recognized a wide range of HBV genotypic variants with improved sensitivity and cytotoxicity. Cell infusions produced complete elimination of HCC without recurrence in the xenograft mouse models. Elevated accumulation of CD8+ Ai-TCR-T cells in tumors correlated with tumor shrinkage. CONCLUSION: The in vitro and in vivo studies demonstrated that HBsAg-specific Ai-TCR-T cells had safety profiles similar to those of their wild-type counterparts and significantly enhanced potency. This study presents an approach to develop new therapeutic strategies for HBV-related HCC.


Assuntos
Carcinoma Hepatocelular/virologia , Vírus da Hepatite B/patogenicidade , Neoplasias Hepáticas/virologia , Linfócitos T/metabolismo , Engenharia Tecidual/métodos , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Brain Pathol ; 29(6): 782-792, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30861589

RESUMO

Giant cell glioblastoma (gcGBM) is a rare histological variant of GBM, accounting for about 1% of all GBM. The prognosis is poor generally though gcGBM does slightly better than the other IDH-wild-type GBM. Because of the rarity of the cases, there has been no comprehensive molecular analysis of gcGBM. Previously, single-gene study identified genetic changes in TP53, PTEN and TERT promoter mutation in gcGBM. In this report, we performed whole-exome sequencing (WES) to identify somatically acquired mutations and copy number variations (CNVs) in 10 gcGBM genomes. We also examined TERT promoter mutation and MGMT methylation in our cohort. On top of the reported mutations, WES revealed ATRX, PIK3R1, RB1 and SETD2 as the recurrent mutations in gcGBM. Notably, one tumor harbored a mutation in MutS homolog 6 (MSH6) that is a key mismatch repair (MMR) gene. This tumor demonstrated hypermutation phenotype and showed an increased number of somatic mutations. TERT promoter mutation and MGMT methylation were observed in 20% and 40% of our samples, respectively. In conclusion, we described relevant mutation profiling for developing future targeted therapies in gcGBM.


Assuntos
Glioblastoma/genética , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/genética , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Estudos de Coortes , Variações do Número de Cópias de DNA/genética , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Feminino , Glioblastoma/metabolismo , Histona-Lisina N-Metiltransferase/genética , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Proteínas de Ligação a Retinoblastoma/genética , Telomerase/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética , Sequenciamento do Exoma/métodos , Proteína Nuclear Ligada ao X/genética
6.
Front Med ; 13(1): 69-82, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30725257

RESUMO

Cytokine-activated T cells (CATs) can be easily expanded and are widely applied to cancer immunotherapy. However, the good efficacy of CATs is rarely reported in clinical applications because CATs have no or very low antigen specificity. The low-efficacy problem can be resolved using T cell antigen receptor-engineered CAT (TCR-CAT). Herein, we demonstrate that NY-ESO-1157-165 HLA-A*02:01-specific high-affinity TCR (HAT)-transduced CATs can specifically kill cancer cells with good efficacy. With low micromolar range dissociation equilibrium constants, HAT-transduced CATs showed good specificity with no off-target killing. Furthermore, the high-affinity TCR-CATs delivered significantly better activation and cytotoxicity than the equivalent TCR-engineered T cells (TCR-Ts) in terms of interferon-γ and granzyme B production and in vitro cancer cell killing ability. TCR-CAT may be a very good alternative to the expensive TCR-T, which is considered an effective personalized cyto-immunotherapy.


Assuntos
Citocinas/metabolismo , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Engenharia Genética , Antígeno HLA-A2/metabolismo , Humanos , Ativação Linfocitária , Receptores de Antígenos de Linfócitos T/genética
7.
Cancer Lett ; 447: 164-173, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30677447

RESUMO

Tumor cells can escape immune surveillance through the programmed cell death protein 1 (PD-1) axis suppressing T cells. However, we recently demonstrated that high-affinity variants of soluble human programmed death-ligand 1 (shPD-L1) could diminish the suppression. We propose that in comparison to the wild-type shPD-L1, the further affinity enhancement will confer the molecule with opposite characteristics that augment T-cell activation and immunotherapeutic drug potential. In this study, a new shPD-L1 variant, L3C7c, has been generated to demonstrate ∼167 fold greater affinity than wild-type hPD-L1. The L3C7c-Fc fusion protein demonstrated completely opposite effects of conventional PD-1 axis by promoting redirected T-cell proliferation, activation and cytotoxicity in vitro, as being slightly better than that of anti-PD1-Ab (Pembrolizumab). Moreover, L3C7c-Fc was more effective than Pembrolizumab in enhancing redirected T cells' ability to suppress Mel624 melanoma growth in vivo. As a downsized L3C7c-Fc variant, L3C7v-Fc improved the anti-tumor efficacy in vivo when combined with dendritic cell vaccines. In conclusion, our studies demonstrate that high-affinity hPD-L1 variants could be developed as the next generation reagents for tumor immunotherapy based on the blockade of the PD-1 axis.


Assuntos
Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Melanoma/imunologia , Melanoma/terapia , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Vacinas Anticâncer/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Imunoterapia/métodos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia
8.
World Neurosurg ; 119: e710-e716, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30092479

RESUMO

OBJECTIVE: Hematologic inflammatory markers are simple, inexpensive prognostic markers for various conditions. The prognostic significance of the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), and red blood cell distribution width (RDW) has been shown in a variety of tumors. We evaluated the prognostic value of these markers in glioma. METHODS: We performed a retrospective medical record review of 219 patients with glioma from January 2012 to January 2017, evaluating the effect of NLR, PLR, MLR, and RDW on prognosis. Correlations among these hematologic inflammatory markers were also examined. RESULTS: The patients were divided into high and low groups using the cutoff points from the receiver operating characteristic curves. High NLR was associated with a higher tumor grade (P = 0.000). Kaplan-Meier survival analyses showed that the high NLR, PLR, and MLR groups experienced inferior median overall survival (OS) compared with the low NLR, PLR, and MLR groups (11 vs. 32 months; P = 0.000; 12 vs. 21 months; P = 0.001; and 12 vs. 22 months; P = 0.006, respectively). No significant difference was found in the median OS between the high and low RDW groups (15 vs. 23 months; P = 0.184). Multivariate analysis demonstrated that NLR was an independent predictor of OS (hazard ratio, 1.758; P = 0.008). CONCLUSIONS: A high preoperative NLR, PLR, and MLR was predictive of a poor prognosis for patients with glioma. NLR was an independent prognostic factor for OS in patients with glioma.


Assuntos
Plaquetas/fisiologia , Neoplasias Encefálicas/mortalidade , Glioma/mortalidade , Leucócitos Mononucleares/fisiologia , Linfócitos/fisiologia , Idoso , Biomarcadores Tumorais , Encefalite/mortalidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neutrófilos/fisiologia , Contagem de Plaquetas , Cuidados Pré-Operatórios , Prognóstico , Estudos Retrospectivos
9.
Cancer Sci ; 109(8): 2435-2445, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29890018

RESUMO

The inhibitory checkpoint molecule programmed death (PD)-1 plays a vital role in maintaining immune homeostasis upon binding to its ligands, PD-L1 and PD-L2. Several recent studies have demonstrated that soluble PD-1 (sPD-1) can block the interaction between membrane PD-1 and PD-L1 to enhance the antitumor capability of T cells. However, the affinity of natural sPD-1 binding to PD-L1 is too low to permit therapeutic applications. Here, a PD-1 variant with approximately 3000-fold and 70-fold affinity increase to bind PD-L1 and PD-L2, respectively, was generated through directed molecular evolution and phage display technology. Structural analysis showed that mutations at amino acid positions 124 and 132 of PD-1 played major roles in enhancing the affinity of PD-1 binding to its ligands. The high-affinity PD-1 mutant could compete with the binding of antibodies specific to PD-L1 or PD-L2 on cancer cells or dendritic cells, and it could enhance the proliferation and IFN-γ release of activated lymphocytes. These features potentially qualify the high-affinity PD-1 variant as a unique candidate for the development of a new class of PD-1 immune-checkpoint blockade therapeutics.


Assuntos
Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Aminoácidos/metabolismo , Proliferação de Células/fisiologia , Técnicas de Visualização da Superfície Celular/métodos , Células Dendríticas/metabolismo , Humanos , Interferon gama/metabolismo , Ligantes , Ativação Linfocitária/fisiologia , Ligação Proteica/fisiologia , Linfócitos T/metabolismo
10.
Immunology ; 155(2): 238-250, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29791021

RESUMO

Recently, bi-functional molecules that can redirect immune effectors to tumour cells have emerged as potentially robust mediators of tumour regression in clinical trials. Two modalities in particular, bi-specific antibodies for T-cell redirection and activation (BiTe) and immune-mobilizing monoclonal T-cell receptors against cancer (ImmTAC), are being evaluated in efficacy studies as 'off-the-shelf' reagents. Optimal therapy will require an understanding and means to address regulatory mechanisms of limiting efficacy. In light of this, we evaluated the impact of induced regulatory T (iTreg) cells on the efficacy of tumour cell killing redirected by ImmTAC and demonstrated down-regulation of T-cell proliferation and expression of CD25, CD107a, Granzyme B and Perforin by ImmTAC-redirected T cells. Significant recovery of ImmTAC potency, however, could be achieved when combined with an anti-programmed cell death protein 1 monoclonal antibody. Furthermore, we found that among lung cancer patients failing to respond to ImmTAC therapy, there was a significantly higher fraction of Treg cells in the peripheral blood mononuclear cells of lung cancer patients than in healthy donors. These results provide in vitro evidence for an iTreg cell-mediated immunosuppression of ImmTAC-redirected T-cell responses. Whilst immune checkpoint blockade can reverse the Treg cell suppression, it forms a rational basis for a combination of the blockade with ImmTAC in clinical trials.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Imunológica , Terapia de Imunossupressão , Ativação Linfocitária/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Citometria de Fluxo , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
11.
Oncotarget ; 8(51): 88360-88375, 2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-29179441

RESUMO

The activated T cells can be suppressed by programed death-1 (PD-1) axis through low affinity interaction between PD-1 and PD-ligand 1 (PD-L1) in solution or on antigen presenting cells. In clinic, the concentration of soluble PD-L1 in peripheral blood negatively correlates with cancer prognosis. However, there is little information about the relation between the affinity of PD-1/PD-L1 interaction and the suppressive capacity of PD-1 axis. In this study, we analyzed inhibitory roles of high affinity soluble human PD-L1 (hPD-L1) variants, which were generated with directed molecular evolution. Resultant two clones L3C7-hPD-L1 and L3B3-hPD-L1 showed over 20 folds greater affinity than that of native hPD-L1. We found that L3B3-hPD-L1 and L3C7-hPD-L1 could compete with an anti-PD-1 antibody (EH12.1) for binding to hPD-1. More importantly, although native soluble hPD-L1 can induce suppressive effects on activated T cells, we found L3B3-hPD-L1 and L3C7-hPD-L1 attenuated the strength of PD-1 axis for suppressing the proliferation and interferon γ (IFN-γ) secretion of PBMC. In conclusion, our data provide direct evidence in which immune checkpoint receptor-ligand interactive strength can alter the the suppressive function, in particular, the suppressive capacity of PD-1 axis could be decreased with enhanced affinity of soluble PD-L1 and PD-1 interaction. Our study might provide a new direction for manipulating immune checkpoints.

12.
Neurochem Res ; 42(11): 3093-3102, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28677030

RESUMO

Dual-specificity tyrosine-regulated kinase 2 (DYRK2), a protein kinase that phosphorylates its substrates on serine/threonine, is expressed in numerous human tumors, but little is known about its role in the pathophysiology of glioma. In this study, we made an effort to explore the expression and function in human glioma. Western blot and immunohistochemistry analysis were performed to investigate the expression of DYRK2 protein in glioma tissues in 84 patients. Wound healing and transwell assay were carried out to determine the cell migration ability. We showed that the level of DYRK2 was significantly decreased in high-grade glioma tissues compared with low-grade tissues. In addition, the expression level of DYRK2 was positively correlated with glioma pathological grade and E-cadherin expression. Kaplane-Meier analysis revealed that low expression of DYRK2 was related to poor prognosis of glioma patients. Furthermore, wound healing and transwell assay revealed that DYRK2 could suppress cell migration and affect the expression levels of E-cadherin and vimentin through PI3K/AKT/GSK3ß signaling pathway. Taken together, our results implied that DYRK2 could serve as a promising prognostic biomarker as well as a potential therapeutical target of glioma.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias Encefálicas/metabolismo , Movimento Celular/fisiologia , Glioma/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Tirosina Quinases/biossíntese , Adulto , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Feminino , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores/métodos , Quinases Dyrk
13.
Cell Mol Neurobiol ; 37(8): 1501-1509, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28258514

RESUMO

p53-induced protein with a RING-H2 domain (Pirh2), also known as Rchy1, is an ubiquitin E3 ligase that regulates the turnover and functionality of several proteins involved in cell proliferation and differentiation, cell cycle checkpoints, and cell death. However, it remains unclear whether aberrant expression of Pirh2 is involved in the development of glioma, a major type of primary brain tumor in adults. Western blot and immunohistochemical analyses showed that Pirh2 was highly expressed in glioma specimens, compared with normal brain tissues. High Pirh2 expression was positively correlated with higher tumor grade, as well as Ki-67 expression. Kaplan-Meier analysis revealed that patients with high Pirh2 expression had worsened prognosis, compared with those with low Pirh2 expression. Moreover, to determine whether Pirh2 could regulate malignant behavior of glioma cells, we transfected glioma cells with interfering RNA targeting Pirh2 to specifically silence Pirh2 expression. Mechanistically, our results indicated that knockdown of Pirh2 induced the apoptosis of glioma cells. In addition, depletion of Pirh2 diminished the expression of PCNA and cyclin D1 and led to cell cycle arrest at G1 phase. Taken together, these findings for the first time suggest that Pirh2 might play an important role in the regulation of glioma proliferation and apoptosis and thus serve as a promising therapeutic target in the treatment of glioma.


Assuntos
Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Ubiquitina-Proteína Ligases/biossíntese , Adulto , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Feminino , Glioma/genética , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Taxa de Sobrevida/tendências , Ubiquitina-Proteína Ligases/genética
14.
Metab Brain Dis ; 32(2): 565-575, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28064406

RESUMO

Vps4, vacuolar protein sorting 4, belongs to ATPases Associated with diverse cellular Activities (AAA) protein family which is made up of Vps4A and Vps4B. Previous studies demonstrated that Vps4A plays vital roles in diverse aspects such as virus budding, the efficient transport of H-Ras to the PM (plasma membrane) and the involvement in the MVB (multivesiculate bodies) pathway. Interestingly, Vps4A is also expressed in the brain. However, the distribution and function of Vps4A in ICH diseases remain unclear. In this study, we show that Vps4A may be involved in neuronal apoptosis during pathophysiological processes of intracerebral hemorrhage (ICH). Based on the results of Western blot and immunohistochemistry, we found a remarkable up-regulation of Vps4A expression surrounding the hematoma after ICH. Double labeled immunofluorescence showed that Vps4A was co-expressed with NeuN but rarely with astrocytes and microglia. Morever, we detected that neuronal apoptosis marker active caspase-3 had co-localizations with Vps4A. Additionaly, Vps4A knockdown in vitro specifically leads to decreasing neuronal apoptosis coupled with increased Akt phosphorylation. All datas suggested that Vps4A was involved in promoting neuronal apoptosis via inhibiting Akt phosphorylation after ICH.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/biossíntese , Apoptose/efeitos dos fármacos , Hemorragia Cerebral/metabolismo , ATPases Vacuolares Próton-Translocadoras/biossíntese , Animais , Antígenos Nucleares/metabolismo , Comportamento Animal/efeitos dos fármacos , Caspase 3/metabolismo , Hemorragia Cerebral/patologia , Hemorragia Cerebral/psicologia , Feminino , Técnicas de Silenciamento de Genes , Masculino , Proteínas do Tecido Nervoso/metabolismo , Proteína Oncogênica v-akt/metabolismo , Fosforilação , Gravidez , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA