Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
BMC Psychiatry ; 20(1): 41, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005204

ABSTRACT

BACKGROUND: Genome Wide Association Studies (GWAS) have been conducted to identify genes and pathways involved in development of opioid use disorder. This study extends the first GWAS of substance use disorder (SUD) patients from the United Arab Emirates (UAE) by stratifying the study group based on opioid use, which is the most common substance of use in this cohort. METHODS: The GWAS cohort consisted of 512 (262 case, 250 controls) male participants from the UAE. The samples were genotyped using the Illumina Omni5 Exome system. Data was stratified according to opioid use using PLINK. Haplotype analysis was conducted using Haploview 4.2. RESULTS: Two main associations were identified in this study. Firstly, two SNPs on chromosome 7 were associated with opioid use disorder, rs118129027 (p-value = 1.23 × 10 - 8) and rs74477937 (p-value = 1.48 × 10 - 8). This has been reported in Alblooshi et al. (Am J Med Genet B Neuropsychiatr Genet 180(1):68-79, 2019). Secondly, haplotypes on chromosome 2 which mapped to the KIAA1211L locus were identified in association with opioid use. Five SNPs in high linkage disequilibrium (LD) (rs2280142, rs6542837, rs12712037, rs10175560, rs11900524) were arranged into haplotypes. Two haplotypes GAGCG and AGTTA were associated with opioid use disorders (p-value 3.26 × 10- 8 and 7.16 × 10- 7, respectively). CONCLUSION: This is the first GWAS to identify candidate genes associated with opioid use disorder in participants from the UAE. The lack of other genetic data of Arabian descent opioid use patients has hindered replication of the findings. Nevertheless, the outcomes implicate new pathways in opioid use disorder that requires further research to assess the role of the identified genes in the development of opioid use disorder.


Subject(s)
Genome-Wide Association Study , Microfilament Proteins/genetics , Opioid-Related Disorders , Chromosomes, Human, Pair 2 , Genetic Predisposition to Disease/genetics , Genotype , Haplotypes , Humans , Male , Opioid-Related Disorders/ethnology , Opioid-Related Disorders/genetics , Polymorphism, Single Nucleotide/genetics , United Arab Emirates
2.
Respir Res ; 20(1): 248, 2019 Nov 07.
Article in English | MEDLINE | ID: mdl-31699089

ABSTRACT

BACKGROUND: Small cell lung cancer (SCLC) is a highly aggressive lung cancer subtype with poor survival and limited treatment options. Sequencing results have revealed gene mutations associated with SCLC, however, the correlation between the genomic alterations and clinical prognosis of SCLC is yet unclear. METHODS: Targeted next-generation sequencing of 62 cancer related genes was performed on 53 SCLC samples. The correlations between clinical outcomes and genomic alterations were analyzed. RESULTS: 38/62 (61.3%) candidate genes harbored some alterations, while all the SCLC samples carried at least 3 gene mutations. The most common nonsynonymous mutations included ERBB2 (95.9%), CREBBP (95.9%), and TP53 (77.6%). The median nonsynonymous tumor mutation burden (TMB) was 21.7 mutations/Mb (rang, 9.3-55.9). High TMB (> 21 mutations/Mb) was good prognostic factor in overall survival (OS) (21.7 vs. 10.4 months, P = 0.012). Multivariate analysis showed that high TMB was an independent prognostic factor. The overall survival (OS) of patients carrying KIAA1211 mutation was significantly longer than those with wild-type KIAA1211 (P < 0.001). CONCLUSIONS: The current study highlights the potential role of genomic alterations for the prognosis of SCLC. Higher TMB was associated with a better prognosis, and KIAA1211 might be a good prognostic factor in SCLC.


Subject(s)
Biomarkers, Tumor/genetics , Lung Neoplasms/genetics , Microfilament Proteins/genetics , Mutation , Small Cell Lung Carcinoma/genetics , Female , Genetic Predisposition to Disease , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Staging , Phenotype , Pregnancy , Progression-Free Survival , Risk Assessment , Risk Factors , Small Cell Lung Carcinoma/mortality , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/therapy
3.
Cell Rep ; 43(6): 114286, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38796854

ABSTRACT

Tumor cell plasticity contributes to intratumoral heterogeneity and therapy resistance. Through cell plasticity, some lung adenocarcinoma (LUAD) cells transform into neuroendocrine (NE) tumor cells. However, the mechanisms of NE cell plasticity remain unclear. CRACD (capping protein inhibiting regulator of actin dynamics), a capping protein inhibitor, is frequently inactivated in cancers. CRACD knockout (KO) is sufficient to de-repress NE-related gene expression in the pulmonary epithelium and LUAD cells. In LUAD mouse models, Cracd KO increases intratumoral heterogeneity with NE gene expression. Single-cell transcriptomic analysis showed that Cracd KO-induced NE cell plasticity is associated with cell de-differentiation and stemness-related pathway activation. The single-cell transcriptomic analysis of LUAD patient tumors recapitulates that the distinct LUAD NE cell cluster expressing NE genes is co-enriched with impaired actin remodeling. This study reveals the crucial role of CRACD in restricting NE cell plasticity that induces cell de-differentiation of LUAD.


Subject(s)
Adenocarcinoma of Lung , Cell Plasticity , Lung Neoplasms , Neuroendocrine Cells , Animals , Humans , Mice , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism
4.
bioRxiv ; 2023 May 17.
Article in English | MEDLINE | ID: mdl-36824957

ABSTRACT

The mechanisms underlying immune evasion and immunotherapy resistance in small cell lung cancer (SCLC) remain unclear. Herein, we investigate the role of CRACD tumor suppressor in SCLC. We found that CRACD is frequently inactivated in SCLC, and Cracd knockout (KO) significantly accelerates SCLC development driven by loss of Rb1, Trp53, and Rbl2. Notably, the Cracd-deficient SCLC tumors display CD8+ T cell depletion and suppression of antigen presentation pathway. Mechanistically, CRACD loss silences the MHC-I pathway through EZH2. EZH2 blockade is sufficient to restore the MHC-I pathway and inhibit CRACD loss-associated SCLC tumorigenesis. Unsupervised single-cell transcriptomic analysis identifies SCLC patient tumors with concomitant inactivation of CRACD, impairment of tumor antigen presentation, and downregulation of EZH2 target genes. Our findings define CRACD loss as a new molecular signature associated with immune evasion of SCLC cells and proposed EZH2 blockade as a viable option for CRACD-negative SCLC treatment.

5.
bioRxiv ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37131761

ABSTRACT

Tumor cell plasticity contributes to intratumoral heterogeneity and therapy resistance. Through cell plasticity, lung adenocarcinoma (LUAD) cells transform into neuroendocrinal (NE) tumor cells. However, the mechanisms of NE cell plasticity remain unclear. CRACD, a capping protein inhibitor, is frequently inactivated in cancers. CRACD knock-out (KO) de-represses NE-related gene expression in the pulmonary epithelium and LUAD cells. In LUAD mouse models, Cracd KO increases intratumoral heterogeneity with NE gene expression. Single-cell transcriptomic analysis showed that Cracd KO-induced NE plasticity is associated with cell de-differentiation and activated stemness-related pathways. The single-cell transcriptomes of LUAD patient tumors recapitulate that the distinct LUAD NE cell cluster expressing NE genes is co-enriched with SOX2, OCT4, and NANOG pathway activation, and impaired actin remodeling. This study reveals an unexpected role of CRACD in restricting NE cell plasticity that induces cell de-differentiation, providing new insights into cell plasticity of LUAD.

6.
J Cancer ; 10(26): 6747-6753, 2019.
Article in English | MEDLINE | ID: mdl-31777604

ABSTRACT

One of the main causes of cancer disease and death worldwide is lung cancer. Our study focused on the function of KIAA1211 in non-small cell lung cancer (NSCLC). According to the data about NSCLC patients that from the Cancer Genome Atlas (TCGA), we found that KIAA1211 in NSCLC (P=5.06E-06) was significantly higher than the adjacent normal. Lentivirus-mediated short hairpin RNA (shRNA) was used to knockdown BATF expression in the human A549 NSCLC cell line and assessed by RT-qPCR and Western blot. Cell proliferation was evaluated by MTT assay and Celigo imaging cytometry. Cell apoptosis were detected by Annexin V staining. The test results showed that KIAA1211-shRNA A549 and SPC-A-1 cells can inhibit cell proliferation, and the apoptosis rate of KIAA1211-shRNA group was significantly higher than that of the control group. Knockdown of KIAA1211 inhibited NSCLC progression in xenograft tumor model. In conclusion, KIAA1211 could regulate NSCLC cells proliferation and apoptosis in vitro and in vivo. KIAA1211 may serve as a potent target for the treatment of NSCLC.

SELECTION OF CITATIONS
SEARCH DETAIL