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.
J Clin Neurosci ; 125: 159-166, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38815302

RESUMO

BACKGROUND: Philadelphia chromosome-negative myeloproliferative neoplasms (Ph-negative MPNs) are linked with various complications, notably ischemic stroke. The study aims to identify risk factors for ischemic stroke in Ph-negative MPNs patients. METHODS: Patients were categorized into two groups based on whether they had experienced ischemic stroke. Subsequently, an analysis of demographics, biochemical makers, and genetic mutations (JAK2V617F and CALR mutations), was conducted to identify potential associations with an elevated risk of ischemic stroke in individuals with Ph-negative MPNs. RESULTS: A total of 185 patients diagnosed with Ph-negative MPNs participated in the study, including 82 with essential thrombocythemia (ET), 78 with polycythemia vera (PV), and 25 with primary myelofibrosis (PMF). Among these, 57 patients (30.8 %) had a history of ischemic stroke. Independent risk factors associated with ischemic stroke in Ph-negative MPNs patients included hypertension (OR = 5.076) and smoking (OR = 5.426). Among ET patients, smoking (OR = 4.114) and an elevated percentage of neutrophils (OR = 1.080) were both positively correlated with ischemic stroke incidence. For PV patients, hypertension (OR = 4.647), smoking (OR = 6.065), and an increased percentage of lymphocytes (OR = 1.039) were independently associated with ischemic stroke. Regardless of the presence of the JAK2V617F mutation, hypertension was the sole positively and independently associated risk factor for ischemic stroke. The odds ratios for patients with the JAK2V617F mutation was 3.103, while for those without the mutation, it was 11.25. CONCLUSIONS: Hypertension was a more substantial factor associated with an increased incidence of ischemic stroke in Ph-negative MPNs patients.


Assuntos
AVC Isquêmico , Janus Quinase 2 , Transtornos Mieloproliferativos , Cromossomo Filadélfia , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Fatores de Risco , AVC Isquêmico/epidemiologia , AVC Isquêmico/genética , Idoso , Janus Quinase 2/genética , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/complicações , Transtornos Mieloproliferativos/epidemiologia , Adulto , Hipertensão/complicações , Hipertensão/epidemiologia , Mutação , Calreticulina/genética , Idoso de 80 Anos ou mais , Fumar/efeitos adversos , Fumar/epidemiologia
2.
iScience ; 27(2): 108852, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38303706

RESUMO

Diabetic cardiomyopathy (DCM) is a cardiovascular complication with no known cure. In this study, we evaluated the combination of ultrasound-targeted microbubble destruction (UTMD) and cationic microbubbles (CMBs) for cardiac S-adenosyl homocysteine hydrolase (SAHH) gene transfection as potential DCM therapy. Models of high glucose/fat (HG/HF)-induced H9C2 cells and streptozotocin-induced DCM rats were established. Ultrasound-mediated SAHH delivery using CMBs was a safe and noninvasive approach for spatially localized drug administration both in vitro and in vivo. Notably, SAHH overexpression increased cell viability and antioxidative stress and inhibited apoptosis of HG/HF-induced H9C2 cells. Likewise, UTMD-mediated SAHH delivery attenuated apoptosis, oxidative stress, cardiac fibrosis, and myocardial dysfunction in DCM rats. Activation of the AMPK/FOXO3/SIRT3 signaling pathway may be a key mechanism mediating the role of SAHH in regulating myocardial injury. Thus, UTMD-mediated SAHH transfection may be an important advancement in cardiac gene therapy for restoring ventricular function after DCM.

3.
Stem Cells Int ; 2022: 1252557, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35873535

RESUMO

Objective: Rapid restoration of corneal epithelium integrity after injury is particularly important for preserving corneal transparency and vision. Mesenchymal stem cells (MSCs) can be taken into account as the promising regenerative therapeutics for improvement of wound healing processes based on the variety of the effective components. The extracellular vesicles form MSCs, especially exosomes, have been considered as important paracrine mediators though transferring microRNAs into recipient cell. This study investigated the mechanism of human umbilical cord MSC-derived small extracellular vesicles (HUMSC-sEVs) on corneal epithelial wound healing. Methods: HUMSC-sEVs were identified by transmission electron microscopy, nanoparticle tracking analysis, and Western blot. Corneal fluorescein staining and histological staining were evaluated in a corneal mechanical wound model. Changes in HCEC proliferation after HUMSC-sEVs or miR-21 mimic treatment were evaluated by CCK-8 and EdU assays, while migration was assessed by in vitro scratch wound assay. Full-length transcriptome sequencing was performed to identify the differentially expressed genes associated with HUMSC-sEVs treatment, followed by validation via real-time PCR and Western blot. Results: The sEVs derived from HUMSCs can significantly promote corneal epithelial cell proliferation, migration in vitro, and corneal epithelial wound healing in vivo. Similar effects were obtained after miR-21 transfection, while the beneficial effects of HUMSC-sEVs were partially negated by miR-21 knockdown. Results also show that the benefits are associated with decreased PTEN level and activated the PI3K/Akt signaling pathway in HCECs. Conclusion: HUMSC-sEVs could enhance the recovery of corneal epithelial wounds though restraining PTEN by transferring miR-21 and may represent a promising novel therapeutic agent for corneal wound repair.

4.
Biochem Biophys Res Commun ; 568: 8-14, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34171541

RESUMO

BACKGROUND: Glioma is one of the most malignant tumors worldwide. This study was aimed to study the effect of miR-379-5p/MGST1 on cell proliferation, migration, invasion and EMT in glioma. METHODS: RT-qPCR detected the expression of miR-379-5p and MGST1 in RNA level in glioma cell lines. Bioinformatic analysis was made to explore the associations between miR-379-5p and MGST1 while survival analysis was made with regards to MGST1 expression in glioma patients. Western blot analysis was applied to measure the EMT changes. MTT examined the cell viability. Transwell was used to detect the cellular invasion and migration. The binding sites between miR-379-5p and MGST1 were validated by luciferase reporter assays. RESULTS: miR-379-5p expression was lower in glioma cells. MiR-379-5p increase inhibited the viability, migration, invasion and EMT while inhibition of miR-379-5p showed a reverse effect. MGST1 inhibition curbed the cell functions. MiR-379-5p targeted and regulated MGST1 expression. Lower MGST1 is related to higher survival rate. CONCLUSION: miR-379-5p could regulate glioma cell viability, migration, invasion and EMT through MGST1, suggesting that miR-379-5p/MGST1 axis might function in the regulation of glioma progression.


Assuntos
Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glutationa Transferase/genética , MicroRNAs/genética , Neoplasias Encefálicas/patologia , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Glioma/patologia , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia
5.
Eur J Pharmacol ; 898: 173932, 2021 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-33631180

RESUMO

We aimed to investigate the role and mechanism of sevoflurane (SEV) preconditioning in liver ischemia-reperfusion (I/R) injury. In vivo, rats were randomly divided into Sham group, I/R rat model group, I/R + SEV group and SEV group. In vitro, hypoxia-reoxygenation (H/R) cell model were established. Hematoxylin-Eosin (H&E) and TUNEL assay were used to evaluate the degree of tissue damage and detect apoptosis in rats, respectively. HO-1, nuclear Nrf2 and cytosolic Nrf2 expressions were detected by immunohistochemical staining, Western blot analysis and quantitative real-time PCR (qRT-PCR), respectively. Contents of Lactate dehydrogenase (LDH), malondialdehyde (MDA), and reactive oxygen species (ROS) were determined by corresponding kits. Inflammatory factor levels, cell viability, apoptosis were detected by enzyme-linked immunosorbent assay (ELISA), MTT assay, and flow cytometry, respectively.In the I/R group, liver damage was severe, apoptosis-positive cells were increased, HO-1 and nuclear Nrf2 expressions were increased, and cytosolic Nrf2 expression was decreased. After SEV pretreatment, the degree of liver injury and apoptosis in rats were significantly reduced, HO-1 and nuclear Nrf2 expressions were increased significantly, and cytosolic Nrf2 expression was decreased. 4% SEV had the best mitigating effect on H/R-induced liver cell damage, as evidenced by reduced contents of LDH and MDA, decreased inflammatory factors, a lowered apoptosis rate, inhibited ROS production, effectively promoted Nrf2 nucleation, and activated Nrf/HO-1 pathway. ML385 pretreatment significantly inhibited the effect of SEV on hepatocytes.Sevoflurane protects the liver from ischemia-reperfusion injury by regulating the Nrf2/HO-1 pathway.


Assuntos
Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatopatias/prevenção & controle , Fígado/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Sevoflurano/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1/genética , Hepatócitos/enzimologia , Hepatócitos/patologia , Mediadores da Inflamação/metabolismo , Fígado/enzimologia , Fígado/patologia , Hepatopatias/enzimologia , Hepatopatias/patologia , Masculino , Proteínas de Membrana/genética , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais
6.
Inflammation ; 44(4): 1288-1301, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33496895

RESUMO

Propofol (PRO) protects against hepatic ischemia/reperfusion (I/R) injury. Bnip3 is involved in the I/R-induced injury. This study investigated whether the effect of PRO on hepatic hypoxia/reoxygenation (H/R) injury was realized through regulating Bnip3. After establishing a hepatic ischemia reperfusion (I/R ) injury model in mice, the serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were determined by an automatic biochemical analyzer. The histopathology and apoptosis of liver tissues were detected by hematoxylin-eosin and TUNEL staining. After the H/R liver cells were cultured and treated with PRO, the viability, apoptosis, reactive oxygen species (ROS) production, and the levels of lactate dehydrogenase (LDH), malondialdehyde (MDA), TNF-α, and IL-6 were detected by MTT, flow cytometry, colorimetry, and ELISA. The expressions of Bnip3 and apoptosis-related factors in I/R mouse liver tissues and H/R cells were determined by immunohistochemical assay, immunofluorescence, Western blot, or RT-qPCR. PRO ameliorated the abnormal histopathology, reduced cell apoptosis and the levels of AST, ALT, Bnip3, Cleaved Caspase-3, and Bax, but upregulated the Bcl-2 level in the liver tissues of I/R mice. In H/R liver cells, PRO promoted the cell viability, downregulated the levels of LDH, MDA, TNF-α, IL-6, and reduced ROS production. Moreover, PRO promoted the downregulated expressions of cytosolic Bnip3, total Bni3p, Cleaved Caspase-3, and Bax and upregulated the Bcl-2 level. siBnip3 reversed the effect of H/R on the liver cells, and its overexpression also reversed the effect of PRO on H/R-induced liver cells. PRO protects against hepatic I/R injury via inhibiting Bnip3.


Assuntos
Fígado/metabolismo , Proteínas de Membrana/biossíntese , Proteínas Mitocondriais/biossíntese , Estresse Oxidativo/fisiologia , Propofol/uso terapêutico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular , Hipnóticos e Sedativos/farmacologia , Hipnóticos e Sedativos/uso terapêutico , Fígado/irrigação sanguínea , Fígado/efeitos dos fármacos , Fígado/lesões , Masculino , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Propofol/farmacologia
7.
Neuroreport ; 31(17): 1189-1198, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33044324

RESUMO

Neuroblastoma is a common malignancy and frequently affects children, leading to a low survival rate. Long noncoding RNAs (lncRNAs) are reported to be closely related to cancer progression. The purpose of this study was to explore a novel mechanism of lncRNA nuclear-enriched abundant transcript 1 (NEAT1) in neuroblastoma. NEAT1 was upregulated in neuroblastoma cell lines (IMR32 and SK-N-SH). Overexpression of NEAT1 increased proliferation inhibited by cisplatin and decreased apoptosis promoted by cisplatin. MicroRNA-326 (miR-326) was a target of NEAT1 and miR-326 reintroduction abolished the effects of NEAT1 overexpression on cell proliferation and apoptosis. Moreover, NEAT1 overexpression activated Janus kinase/signal transducer and activator of transcription 3 (JAK1/STAT3) signaling pathway through absorbing miR-326. Besides, NEAT1 overexpression promoted tumor growth in vivo through stimulating the expression of p-JAK1 and p-STAT3 but inhibiting miR-326 expression. NEAT1 accelerated proliferation and weakened apoptosis of neuroblastoma cells treated by cisplatin by targeting miR-326 through activating JAK1/STAT3 signaling pathway, suggesting that NEAT1 was a potential biomarker against neuroblastoma.


Assuntos
Cisplatino/administração & dosagem , Janus Quinase 1/biossíntese , MicroRNAs/biossíntese , Neuroblastoma/metabolismo , RNA Longo não Codificante/biossíntese , Fator de Transcrição STAT3/biossíntese , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Janus Quinase 1/genética , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Neuroblastoma/genética , RNA Longo não Codificante/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
8.
Am J Transl Res ; 12(6): 2711-2725, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32655803

RESUMO

BACKGROUND: Imatinib resistance is commonly associated with the activation of BCR-ABL signaling in chronic myeloid leukaemia (CML). The activation of Lyn can result in imatinib resistance by regulating the formation of BCR-ABL protein complexes. SIRT1 is a novel survival pathway activated by BCR-ABL expression in haematopoietic progenitor cells. This study aimed to investigate whether the signaling pathway of Lyn/BCR-ABL/SIRT1 could mediate imatinib resistance in CML. METHODS: The MTT assay was used to detect cell viability. Apoptosis was measured by a flow cytometry assay. Protein expression was detected by Western blotting. Knockdown CML cells were constructed by shRNA interference. The CML mouse model was used to investigate the role of SIRT1 in CML in vivo. RESULTS: Lyn was overexpressed in K562R cells. BCR-ABL phosphorylation and activation were promoted by Lyn. Imatinib suppressed BCR-ABL phosphorylation in both K562 and K562R cells. BCR-ABL positively regulated SIRT1 and Foxo1 but negatively regulated acetylated Foxo1 (Ac-Foxo1) and p53 expression. Pharmacological inhibition of SIRT1 or knockdown of SIRT1 increased apoptosis and reduced growth in vitro and in vivo. Foxo1 was downregulated by SIRT1 inhibition or knockdown, while Ac-Foxo1 and p53 were upregulated. In vivo experiments showed that imatinib and/or SIRT1 inhibition both prolonged the survival of the CML mouse model and that the effects of imatinib were enhanced in combination with SIRT1 inhibition. CONCLUSION: We proposed a novel molecular mechanism of imatinib resistance in CML in which the high expression of Lyn in imatinib-resistant cells inhibited Ac-Foxo1 and p53 expression through the BCR-ABL/SIRT1/Foxo1 signaling pathway, thus reducing apoptosis and mediating imatinib resistance.

9.
Arch Toxicol ; 94(4): 1203-1213, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32112223

RESUMO

Polymorphisms in arsenic (+ 3 oxidation state) methyltransferase (AS3MT) have been shown to be related to interindividual variations in arsenic metabolism and to influence adverse health effects in acute promyelocytic leukemia (APL) patients treated with arsenic trioxide (As2O3). The occurrence of hyperleukocytosis with As2O3 treatment seriously affects the early survival rate of APL patients, but no definite explanation for such a complication has been clearly established. To clarify the causes of this situation, AS3MT polymorphisms 14215 (rs3740390), 14458 (rs11191439), 27215 (rs11191446), and 35991 (rs10748835) and profiles of plasma arsenic metabolites were evaluated in a group of 54 newly diagnosed APL patients treated with single-agent As2O3. High-performance liquid chromatography-hydride generation-atomic fluorescence spectrometry (HPLC-HG-AFS) was used to determine the concentrations of plasma arsenic metabolites. Plasma arsenic methylation metabolism capacity was evaluated by the percentage of inorganic arsenic (iAs), monomethylarsonic acid (MMA), dimethylarsinic acid (DMA), primary methylation index (PMI, MMA/iAs), and secondary methylation index (SMI, DMA/MMA). The results showed that APL patients who developed hyperleukocytosis had a higher plasma iAs%, but a lower MMA% and PMI than those who did not develop hyperleukocytosis during As2O3 treatment. In addition, patients with the AS3MT 14215 (rs3740390) CC genotype had significantly higher plasma iAs% and incidence of hyperleukocytosis, but lower PMI than patients with the CT + TT genotype. Conversely, we did not observe statistically significant associations between the occurrence of hyperleukocytosis and AS3MT 14458 (rs11191439), 27215 (rs11191446), and 35991 (rs10748835) polymorphisms in our study subjects. These results indicated that AS3MT 14215 (rs3740390) might be used as an indicator for predicting the occurrence of hyperleukocytosis in APL patients treated with As2O3.


Assuntos
Antineoplásicos/efeitos adversos , Trióxido de Arsênio/efeitos adversos , Arsênio/metabolismo , Metiltransferases/genética , Adulto , Feminino , Genótipo , Humanos , Leucemia Promielocítica Aguda , Leucócitos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único
10.
Cancer Manag Res ; 11: 3187-3196, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31114360

RESUMO

Purpose: Assessing the possibility of finding tumor-infiltrating lymphocytes (TIL) in bone marrow of acute myeloid leukemia (AML) patients and evaluating the anti-tumor activity of these TIL against autologous AML cells. Patients and methods: TIL were immunomagnetically isolated by using anti-CD3 from bone marrow samples of 20 patients at the presentation of AML or four weeks upon completion of chemotherapy. TIL were ex vivo expanded for two weeks and immunophenotyped. Functionality in terms of cytokine secretion and cytotoxicity was assessed by γ-interferon quantitation and Elispot assay, respectively. Results: TIL were detected in bone marrow samples of 50% (10/20) of the patient cohort. They were noted to highly express CD137 and PD-1 and display a significantly higher anti-tumor reactivity compared to that of autologous peripheral blood lymphocytes. TIL could be expanded in co-cultures with irradiated feeder cells supplemented with interleukin (IL)-7 and IL-15. Conclusion: Data suggested the presence of reactive γ-interferon-secreting TIL in AML patients. They are expandable and possess anti-tumor activity, which might have a great potential in the development of adoptive cellular therapy for AML.

11.
Biomed Res Int ; 2014: 969768, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24955372

RESUMO

RNA-Seq is emerging as an increasingly important tool in biological research, and it provides the most direct evidence of the relationship between the physiological state and molecular changes in cells. A large amount of RNA-Seq data across diverse experimental conditions have been generated and deposited in public databases. However, most developed approaches for coexpression analyses focus on the coexpression pattern mining of the transcriptome, thereby ignoring the magnitude of gene differences in one pattern. Furthermore, the functional relationships of genes in one pattern, and notably among patterns, were not always recognized. In this study, we developed an integrated strategy to identify differential coexpression patterns of genes and probed the functional mechanisms of the modules. Two real datasets were used to validate the method and allow comparisons with other methods. One of the datasets was selected to illustrate the flow of a typical analysis. In summary, we present an approach to robustly detect coexpression patterns in transcriptomes and to stratify patterns according to their relative differences. Furthermore, a global relationship between patterns and biological functions was constructed. In addition, a freely accessible web toolkit "coexpression pattern mining and GO functional analysis" (COGO) was developed.


Assuntos
Mineração de Dados/métodos , Bases de Dados Genéticas , Redes Reguladoras de Genes , Análise de Sequência de RNA , Algoritmos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Transcriptoma/genética
12.
Acta Haematol ; 130(4): 260-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23867347

RESUMO

BACKGROUND/AIM: Dysregulated Hedgehog (Hh) signaling has been implicated in several human malignancies. Hh signaling inhibitors are predicted to have a minimal effect when the Smoothened receptor is mutated. Implications that Gli proteins are molecular targets of arsenic trioxide (ATO) action prompted us to investigate the expression of Hh signaling in acute promyelocytic leukemia (APL) and the influence of ATO on the Hh signaling pathway in APL. METHODS: Quantitative real-time reverse transcription polymerase chain reaction and Western blot were employed to analyze the expression of Hh pathway components and the influence of ATO on the Hh signaling pathway in APL. RESULTS: The expression of Hh pathway components was significantly upregulated in APL. In newly diagnosed APL patients, Gli2 expression was significantly positively correlated with Gli1 (R = 0.57, p < 0.001) and Smo (R = 0.56, p < 0.001) and the expression of Hh pathway components was significantly higher in the high WBC group (p < 0.05). ATO can significantly downregulate the expression of Hh pathway components in vitro and in vivo (p < 0.05). CONCLUSION: The Hh pathway is aberrantly activated in APL and associated with a bad prognostic factor. ATO can effectively inhibit the expression of the Hh pathway. The obtained data give the first clinical evidence for the application of ATO in tumors exhibiting an aberrantly activated Hh pathway.


Assuntos
Arsenicais/farmacologia , Proteínas Hedgehog/antagonistas & inibidores , Leucemia Promielocítica Aguda/fisiopatologia , Óxidos/farmacologia , Adolescente , Adulto , Idoso , Trióxido de Arsênio , Arsenicais/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/biossíntese , Leucemia Promielocítica Aguda/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/biossíntese , Óxidos/uso terapêutico , Receptores Patched , Receptores de Superfície Celular/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Receptor Smoothened , Fatores de Transcrição/biossíntese , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
13.
Brain Res ; 1376: 1-9, 2011 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-21172311

RESUMO

The human serotonin receptor 1B (HRT1B) plays an important role in regulating serotonin release. Previous research has suggested that the genetic variation of the HTR1B gene may confer susceptibility to alcoholism or some subtypes of alcohol dependence, but the evidence has been inconsistent. The aim of the present study is to examine whether polymorphic variants of the HTR1B gene are associated with alcohol dependence subtypes or drinking-related behaviors in Chinese Han population. Alcohol-dependent (AD) male patients (n=135) and controls (n=143) were genotyped for two polymorphisms: A161T in the promoter region and the synonymous variation G861C in the coding region of HTR1B. The results showed that the A161T polymorphism was associated with alcohol dependence (T vs. A allele: p=0.002; OR=2.18, 95% CI: 1.32-3.60). This association was strengthened in those with positive family history (OR=3.12, 95% CI: 1.71-5.70) and/or early onset (OR=4.53, 95% CI: 2.18-9.44) of alcohol dependence. The A161T variant was also significantly associated with age of onset of alcoholism (p=0.001). Furthermore, there was a significant difference of haplotypic frequencies between patients and controls (χ(2)=14.84, df=3, p=0.002), with one common haplotype AG of being significantly underrepresented among the patient group compared to the control group (34% vs. 47.7%, permutation p=0.0034; OR=0.56; 95% CI: 0.39-0.79). These findings confirm HTR1B as a susceptibility gene for alcohol dependence in the sample of Chinese Han population. The HTR1B A-161T polymorphism may be particularly valuable as a functional genetic marker for alcoholism and merits additional study.


Assuntos
Alcoolismo/genética , Povo Asiático/genética , Biomarcadores/análise , Predisposição Genética para Doença/genética , Polimorfismo de Nucleotídeo Único , Receptor 5-HT1B de Serotonina/genética , Adulto , Genótipo , Humanos , Masculino
14.
Biochem Biophys Res Commun ; 377(4): 1265-8, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18983974

RESUMO

Small ubiquitin-like modifier 1 (SUMO1) haploinsufficiency results in cleft lip and palate in animal models. However, no studies have linked SUMO1 to non-syndromic cleft lip with or without cleft palate (NSCLP) in humans. In the present study, we investigated the potential association between SUMO1 single nucleotide polymorphisms (SNPs) and risk for human NSCLP. From 181 patients and 162 healthy controls, we found statistically significant correlations between a 4-SNP SUMO1 haplotype and NSCLP. These data are the first to suggest a role for SUMO1 gene variation in human NSCLP development.


Assuntos
Fenda Labial/genética , Fissura Palatina/genética , Proteína SUMO-1/genética , Adolescente , Adulto , Povo Asiático/genética , Criança , Pré-Escolar , Fenda Labial/complicações , Fissura Palatina/complicações , Feminino , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA