首页 > 最新文献

Global Medical Genetics最新文献

英文 中文
Maternal Transmission of the PAX7 Single Nucleotide Polymorphisms among Indian Cleft Trios. PAX7单核苷酸多态性在印度三胞胎中的母系传播
IF 1.7 Pub Date : 2023-01-01 DOI: 10.1055/s-0042-1760383
Mahamad Irfanulla Khan, Prashanth C S, Mohammed S Mustak, Sheikh Nizamuddin

Cleft lip and/or cleft palate (CL/P) is one of the most common congenital anomalies of the human face with a complex etiology involving multiple genetic and environmental factors. Several studies have shown the association of the paired box 7 ( PAX7 ) gene with CL/P in different populations worldwide. However, the current literature reveals no reported case-parent trio studies to evaluate the association between the PAX7 gene and the risk of nonsyndromic cleft lip and/or palate (NSCL/P) in the Indian population. Hence, the purpose of this study was to assess the PAX7 gene single nucleotide polymorphisms (SNPs) in the etiology of NSCL/P among the Indian cleft trios. Forty Indian case-parent trios of NSCL/P were included. The cases and their parents' genomic DNA were extracted. The SNPs rs9439714, rs1339062, rs6695765, rs742071, and rs618941of the PAX7 gene were genotyped using the Agena Bio MassARRAY analysis. The allelic transmission disequilibrium test was performed using PLINK software while pair-wise linkage disequilibrium by the Haploview program. The SNP rs9439714 showed evidence of association ( p -value = 0.02, odds ratio = 3) with NSCL/P. Considering the parent-of-origin effects, the SNPs rs9439714 and rs618941 showed an excess maternal transmission of allele C at rs9439714 ( p -value = 0.05) and G allele at rs618941 ( p -value = 0.04). The results of the present study suggested that the SNPs rs9439714 and rs618941 showed an excess maternal transmission of alleles suggestive of the possible role of the PAX7 gene involvement in the etiology of NSCL/P in the Indian population.

唇裂和/或腭裂(CL/P)是最常见的先天性面部畸形之一,其病因复杂,涉及多种遗传和环境因素。一些研究表明,配对盒7 (PAX7)基因与全球不同人群的CL/P有关。然而,目前的文献显示没有报道的病例-父母三人研究来评估PAX7基因与印度人群非综合征性唇腭裂(NSCL/P)风险之间的关系。因此,本研究的目的是评估PAX7基因单核苷酸多态性(snp)在印度唇腭裂三胞胎中NSCL/P病因学中的作用。包括40例印度非典型scl /P病例-父母三人组。提取了这些病例及其父母的基因组DNA。PAX7基因的snp rs9439714、rs1339062、rs6695765、rs742071和rs618941采用Agena Bio MassARRAY分析进行基因分型。等位基因传递不平衡检验采用PLINK软件,配对连锁不平衡检验采用Haploview程序。SNP rs9439714与NSCL/ p相关(p值= 0.02,优势比= 3)。考虑到亲本效应,snp rs9439714和rs618941在rs9439714位点和rs618941位点的等位基因C (p值= 0.05)和G等位基因(p值= 0.04)的母本遗传过量。本研究结果表明,snp rs9439714和rs618941显示了等位基因的过量母系传播,提示PAX7基因参与印度人群nsl /P病因学的可能作用。
{"title":"Maternal Transmission of the <i>PAX7</i> Single Nucleotide Polymorphisms among Indian Cleft Trios.","authors":"Mahamad Irfanulla Khan,&nbsp;Prashanth C S,&nbsp;Mohammed S Mustak,&nbsp;Sheikh Nizamuddin","doi":"10.1055/s-0042-1760383","DOIUrl":"https://doi.org/10.1055/s-0042-1760383","url":null,"abstract":"<p><p>Cleft lip and/or cleft palate (CL/P) is one of the most common congenital anomalies of the human face with a complex etiology involving multiple genetic and environmental factors. Several studies have shown the association of the paired box 7 ( <i>PAX7</i> ) gene with CL/P in different populations worldwide. However, the current literature reveals no reported case-parent trio studies to evaluate the association between the <i>PAX7</i> gene and the risk of nonsyndromic cleft lip and/or palate (NSCL/P) in the Indian population. Hence, the purpose of this study was to assess the <i>PAX7</i> gene single nucleotide polymorphisms (SNPs) in the etiology of NSCL/P among the Indian cleft trios. Forty Indian case-parent trios of NSCL/P were included. The cases and their parents' genomic DNA were extracted. The SNPs rs9439714, rs1339062, rs6695765, rs742071, and rs618941of the <i>PAX7</i> gene were genotyped using the Agena Bio MassARRAY analysis. The allelic transmission disequilibrium test was performed using PLINK software while pair-wise linkage disequilibrium by the Haploview program. The SNP rs9439714 showed evidence of association ( <i>p</i> -value = 0.02, odds ratio = 3) with NSCL/P. Considering the parent-of-origin effects, the SNPs rs9439714 and rs618941 showed an excess maternal transmission of allele C at rs9439714 ( <i>p</i> -value = 0.05) and G allele at rs618941 ( <i>p</i> -value = 0.04). The results of the present study suggested that the SNPs rs9439714 and rs618941 showed an excess maternal transmission of alleles suggestive of the possible role of the <i>PAX7</i> gene involvement in the etiology of NSCL/P in the Indian population.</p>","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9873478/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10680947","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PMS2 Pathogenic Variant in Lynch Syndrome-Associated Colorectal Cancer with Polyps. Lynch综合征相关结直肠癌伴息肉的PMS2致病变异
IF 1.7 Pub Date : 2023-01-01 DOI: 10.1055/s-0042-1759888
Henriette Poaty, Lauria Batamba Bouya, Aimé Lumaka, Arnaud Mongo-Onkouo, Deby Gassaye

Background  Lynch syndrome (LS) is an autosomal dominant condition due to the germline mutation in the mismatch repair (MMR) genes including MLH1 , MSH2 , MSH6, and PMS2 (post-meiotic segregation increased 2). The MMR mutation carriers have high risk for cancers. Pathogenic PMS2 variants are rarely reported in LS-associated colorectal cancer (CRC) with colorectal polyps. The aim of the study was to investigate the genetic etiology of CRC in an individual with CRC with multiple colorectal polyps and a family history of cancers. Patients and Methods  The index patient was an African male affected by CRC with multiple colorectal polyps. The clinical diagnostic for LS was based on the Amsterdam II criteria and pedigree. Next-generation sequencing with inherited cancer genes panel was used to detect the pathogenic variant. Results  The patient fulfilled the Amsterdam II criteria and the pedigree revealed a family history of recurrent CRC. A deleterious PMS2 germline heterozygous mutation c.2192_2196delTAACT was detected. Conclusion  Our study supports the notion that LS may be associated with polyps and shows the predisposition of PMS2 heterozygous mutation in LS-associated CRC at young age.

Lynch综合征(LS)是一种常染色体显性遗传病,其原因是错配修复(MMR)基因(包括MLH1、MSH2、MSH6和PMS2)的种系突变(减数分裂后分离增加2)。MMR突变携带者患癌症的风险很高。致病性PMS2变异在ls相关性结直肠癌(CRC)伴结直肠息肉中很少报道。该研究的目的是调查结直肠癌合并多发性结直肠息肉和癌症家族史的个体结直肠癌的遗传病因。患者与方法指标患者为非洲男性结直肠癌合并多发结肠息肉患者。LS的临床诊断是基于阿姆斯特丹II标准和谱系。采用新一代遗传癌基因测序技术检测致病变异。结果该患者符合阿姆斯特丹II型标准,家谱显示有CRC复发家族史。检测到一个有害的PMS2种系杂合突变c.2192_2196delTAACT。结论我们的研究支持了LS可能与息肉相关的观点,并显示了PMS2杂合突变在LS相关的CRC中在年轻时的易感性。
{"title":"<i>PMS2</i> Pathogenic Variant in Lynch Syndrome-Associated Colorectal Cancer with Polyps.","authors":"Henriette Poaty,&nbsp;Lauria Batamba Bouya,&nbsp;Aimé Lumaka,&nbsp;Arnaud Mongo-Onkouo,&nbsp;Deby Gassaye","doi":"10.1055/s-0042-1759888","DOIUrl":"https://doi.org/10.1055/s-0042-1759888","url":null,"abstract":"<p><p><b>Background</b>  Lynch syndrome (LS) is an autosomal dominant condition due to the germline mutation in the mismatch repair (MMR) genes including <i>MLH1</i> , <i>MSH2</i> , <i>MSH6,</i> and <i>PMS2</i> (post-meiotic segregation increased 2). The MMR mutation carriers have high risk for cancers. Pathogenic <i>PMS2</i> variants are rarely reported in LS-associated colorectal cancer (CRC) with colorectal polyps. The aim of the study was to investigate the genetic etiology of CRC in an individual with CRC with multiple colorectal polyps and a family history of cancers. <b>Patients and Methods</b>  The index patient was an African male affected by CRC with multiple colorectal polyps. The clinical diagnostic for LS was based on the Amsterdam II criteria and pedigree. Next-generation sequencing with inherited cancer genes panel was used to detect the pathogenic variant. <b>Results</b>  The patient fulfilled the Amsterdam II criteria and the pedigree revealed a family history of recurrent CRC. A deleterious <i>PMS2</i> germline heterozygous mutation c.2192_2196delTAACT was detected. <b>Conclusion</b>  Our study supports the notion that LS may be associated with polyps and shows the predisposition of <i>PMS2</i> heterozygous mutation in LS-associated CRC at young age.</p>","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9833889/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10536395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mystery Behind Barrett's Esophagus: The Origin and Malignant Transformation of Esophageal Adenocarcinoma. 巴雷特食管背后的奥秘:食管腺癌的起源和恶性转化。
IF 1.7 Pub Date : 2022-12-21 eCollection Date: 2022-12-01 DOI: 10.1055/s-0042-1758764
Xiayao Diao
Esophageal cancer (EC) is the eighth most common cancer in the world, with an estimated 604,100 new cases in 2020, accounting for 3.1% of all cancer cases.1 Esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) are the two main histologic subtypes of EC. ESCC predominantly affects developing countries and accounts for more than 88.8% in Chinese EC patients,2,3 while EAC predominantly affects developed countries and accounts for 80.1% in EC patients from United States.4,5 Multiple risk factors, such as Barrett’s esophagus, are associated with EC development. Barrett’s esophagus is a typical metaplastic disease that begins at the gastroesophageal junctions with proximal displacement of the squamocolumnar junctions. Intestinal metaplasia increases the propensity for ECs, especially EACs, and may result from transcriptional switches within gastric cell types or products of intestinal cell types, but the exact origin is unclear. However, half of EAC patients were not observed to have Barrett’s esophagus at the time of diagnosis.6,7 Therefore, we cannot help but ask the following question: does Barrett’s esophagus increase the risk of EAC? This question can be answered by determining the origin of Barrett’s esophagus. Most scientists believe that Barrett’s esophagus originates from many sources, such as various specific cell populations in the gastroesophageal junctions and esophageal submucosal glands. Lineage tracing studies in mouse models is the primary method for exploring Barrett’s esophagus origin. However, the squamous pregastric keratinization and lack of esophageal submucosal glands make this animal model unable to fully mimic human gastroesophageal physiology. Additionally, isolation of esophageal submucosal glands from fresh human tissue is particularly difficult. All of these have become the major obstacles to lineage tracing studies. In a study recently published in Science, titled “Molecular phenotyping reveals the identity of Barrett’s esophagus and its malignant transition,” Nowicki-Osuch et al8 successfully harvested the tissue samples across the gastroesophageal junction and isolated esophageal submucosal glands from patients and healthy individuals to explore the exact source of Barrett’s esophagus. These tissue samples were analyzed by single-cell transcriptomic profiling, in silico lineage tracing of methylation, and somatic mutation/open chromatin array. The functional validation was performed in organoid models. In brief, the authors immuno-stained pan-epithelial tissues, squamous tissues, columnar tissues, and esophageal submucosal glands of fresh human esophagus tissue with cadherin 1 (CDH1), keratin 5 (KRT5), keratin 8 (KRT8), and keratin 7 (KRT7) antibodies, respectively, and then used the three-dimensional confocal microscopy to identify and isolate the ductal cells, oncocytes, mucous cells, and myoepithelial cells. Theyobserved a population of P63þKRT5þKRT7þ cells (transitional basal progenitor) in
{"title":"Mystery Behind Barrett's Esophagus: The Origin and Malignant Transformation of Esophageal Adenocarcinoma.","authors":"Xiayao Diao","doi":"10.1055/s-0042-1758764","DOIUrl":"10.1055/s-0042-1758764","url":null,"abstract":"Esophageal cancer (EC) is the eighth most common cancer in the world, with an estimated 604,100 new cases in 2020, accounting for 3.1% of all cancer cases.1 Esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) are the two main histologic subtypes of EC. ESCC predominantly affects developing countries and accounts for more than 88.8% in Chinese EC patients,2,3 while EAC predominantly affects developed countries and accounts for 80.1% in EC patients from United States.4,5 Multiple risk factors, such as Barrett’s esophagus, are associated with EC development. Barrett’s esophagus is a typical metaplastic disease that begins at the gastroesophageal junctions with proximal displacement of the squamocolumnar junctions. Intestinal metaplasia increases the propensity for ECs, especially EACs, and may result from transcriptional switches within gastric cell types or products of intestinal cell types, but the exact origin is unclear. However, half of EAC patients were not observed to have Barrett’s esophagus at the time of diagnosis.6,7 Therefore, we cannot help but ask the following question: does Barrett’s esophagus increase the risk of EAC? This question can be answered by determining the origin of Barrett’s esophagus. Most scientists believe that Barrett’s esophagus originates from many sources, such as various specific cell populations in the gastroesophageal junctions and esophageal submucosal glands. Lineage tracing studies in mouse models is the primary method for exploring Barrett’s esophagus origin. However, the squamous pregastric keratinization and lack of esophageal submucosal glands make this animal model unable to fully mimic human gastroesophageal physiology. Additionally, isolation of esophageal submucosal glands from fresh human tissue is particularly difficult. All of these have become the major obstacles to lineage tracing studies. In a study recently published in Science, titled “Molecular phenotyping reveals the identity of Barrett’s esophagus and its malignant transition,” Nowicki-Osuch et al8 successfully harvested the tissue samples across the gastroesophageal junction and isolated esophageal submucosal glands from patients and healthy individuals to explore the exact source of Barrett’s esophagus. These tissue samples were analyzed by single-cell transcriptomic profiling, in silico lineage tracing of methylation, and somatic mutation/open chromatin array. The functional validation was performed in organoid models. In brief, the authors immuno-stained pan-epithelial tissues, squamous tissues, columnar tissues, and esophageal submucosal glands of fresh human esophagus tissue with cadherin 1 (CDH1), keratin 5 (KRT5), keratin 8 (KRT8), and keratin 7 (KRT7) antibodies, respectively, and then used the three-dimensional confocal microscopy to identify and isolate the ductal cells, oncocytes, mucous cells, and myoepithelial cells. Theyobserved a population of P63þKRT5þKRT7þ cells (transitional basal progenitor) in ","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2022-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9771686/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10435826","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Most Comprehensive Study at Single-Cell Resolution: A Giant Step toward Understanding Gastric Cancer. 单细胞分辨率最全面的研究:了解胃癌的一大步。
IF 1.7 Pub Date : 2022-12-01 DOI: 10.1055/s-0042-1758763
Fei-Yu Diao
In 2020, gastric cancer is thefifthcommoncancerand thefifth leading cause of cancer death in the world.1 It has the highest incidence andmortality rate in Asian countries, such as China, Japan, and South Korea.2,3 Heterogeneity at the histologic, transcriptomic, genomic, and epigenomic levels exists between gastric cancer patients (interpatient heterogeneity) and within individual tumor mass (intertumoral heterogeneity). It leads to different cancer biological behaviors and treatment response.4 Therefore, biomarkers developed based on theheterogeneityofgastric cancer playan important role in guiding clinical treatment and improving patient prognosis.5,6 Although some current cancer genome projects, The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group, have made great progress in facilitating the molecular typing of gastric cancer, their role in improving the prognosis of gastric cancer patients is limited. Therefore, to conduct high-resolution studies at molecular level in a wide range of patients to guide the clinical treatment of gastric cancer is necessary. Previous “bulk-transcriptome” studies have found that each gastric cancer case has a unique expression profile contributed by cancer cells and resident cell types of tumor microenvironment (such as cancer-associatedfibroblasts, immune cells, and endothelial cells, etc.),7 but the underlying molecular mechanisms of how tumor microenvironment resident cells drive tumor phenotype evolution and clinical progression remain unknown. With the advances in bioinformatics, bulk sequencing data has been successfully decomposed into lineage-specific constituent programs, but this approach fails to discern rare cell populations,fine-scale tissue lineages, cell–cell interactions, and relationships between lineages.8 Single-cell RNA sequencing (scRNA-seq) is the primary tool for addressing these issues. It can detect gene expression in thousands of cells simultaneously, enabling comprehensive analysis of different cell types in tumor mass under different conditions. Indeed, scRNA-seq on gastric cancer tissues from various sources has provided unique insights of cancer biology. However, these current scRNAseq studies are limited by the number of samples and cells, aswell as the dissociation requirements for tissues, which had led to the loss of many key information, especially spatial information. Thus, digital spatial analysis, in situ sequencing, and multiplexed error-robust fluorescence in situ hybridization platforms have been developed to maximize the preservation of spatial information, and thereby allowing the indepth analysis of tumor–tumor microenvironment interactions. In a study recently published in Cancer Discovery, titled “Single-Cell Atlas of Lineage States, Tumor Microenvironment, and Subtype-Specific Expression Programs in Gastric Cancer,” Kumar et al9 delineated a comprehensive single-cell atlas of gastric cancer specimens across clinical stages and histologic subtypes by scRNA-
{"title":"The Most Comprehensive Study at Single-Cell Resolution: A Giant Step toward Understanding Gastric Cancer.","authors":"Fei-Yu Diao","doi":"10.1055/s-0042-1758763","DOIUrl":"https://doi.org/10.1055/s-0042-1758763","url":null,"abstract":"In 2020, gastric cancer is thefifthcommoncancerand thefifth leading cause of cancer death in the world.1 It has the highest incidence andmortality rate in Asian countries, such as China, Japan, and South Korea.2,3 Heterogeneity at the histologic, transcriptomic, genomic, and epigenomic levels exists between gastric cancer patients (interpatient heterogeneity) and within individual tumor mass (intertumoral heterogeneity). It leads to different cancer biological behaviors and treatment response.4 Therefore, biomarkers developed based on theheterogeneityofgastric cancer playan important role in guiding clinical treatment and improving patient prognosis.5,6 Although some current cancer genome projects, The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group, have made great progress in facilitating the molecular typing of gastric cancer, their role in improving the prognosis of gastric cancer patients is limited. Therefore, to conduct high-resolution studies at molecular level in a wide range of patients to guide the clinical treatment of gastric cancer is necessary. Previous “bulk-transcriptome” studies have found that each gastric cancer case has a unique expression profile contributed by cancer cells and resident cell types of tumor microenvironment (such as cancer-associatedfibroblasts, immune cells, and endothelial cells, etc.),7 but the underlying molecular mechanisms of how tumor microenvironment resident cells drive tumor phenotype evolution and clinical progression remain unknown. With the advances in bioinformatics, bulk sequencing data has been successfully decomposed into lineage-specific constituent programs, but this approach fails to discern rare cell populations,fine-scale tissue lineages, cell–cell interactions, and relationships between lineages.8 Single-cell RNA sequencing (scRNA-seq) is the primary tool for addressing these issues. It can detect gene expression in thousands of cells simultaneously, enabling comprehensive analysis of different cell types in tumor mass under different conditions. Indeed, scRNA-seq on gastric cancer tissues from various sources has provided unique insights of cancer biology. However, these current scRNAseq studies are limited by the number of samples and cells, aswell as the dissociation requirements for tissues, which had led to the loss of many key information, especially spatial information. Thus, digital spatial analysis, in situ sequencing, and multiplexed error-robust fluorescence in situ hybridization platforms have been developed to maximize the preservation of spatial information, and thereby allowing the indepth analysis of tumor–tumor microenvironment interactions. In a study recently published in Cancer Discovery, titled “Single-Cell Atlas of Lineage States, Tumor Microenvironment, and Subtype-Specific Expression Programs in Gastric Cancer,” Kumar et al9 delineated a comprehensive single-cell atlas of gastric cancer specimens across clinical stages and histologic subtypes by scRNA-","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9748445/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10748701","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exosomal microRNAs Targeting TP53 Gene as Promising Prognostic Markers for Head and Neck Squamous Cell Carcinoma. 靶向TP53基因的外泌体小rna作为头颈部鳞状细胞癌有希望的预后标志物。
IF 1.7 Pub Date : 2022-12-01 DOI: 10.1055/s-0042-1758204
Vijayashree Priyadharsini Jayaseelan, Paramasivam Arumugam

Statement of Problem  MicroRNAs are small non-coding RNAs that regulate an array of functions by targeting crucial genes. A significant dysregulation in the TP53 profile has been observed in the head and neck squamous cell carcinoma (HNSCC) patients. Hence, the present in silico study was designed to identify those microRNAs which target TP53 gene and demonstrate their differential expression in HNSCC cases. Materials and Methods  The study was extended further to explore their exosomal location using database such as EVmiRNA and ExoCarta. The study follows an observational in silico design. Computational tool miRDB was used identify the microRNA targets of TP53 gene. The UALCAN server was used to ascertain the expression of microRNA in HNSCC cases derived from the Cancer Gene Atlas dataset. The survival of HNSCC patients based on the differential expression microRNA markers were recorded. Further, each of the microRNA was queried for their exosomal presence using EVmiRNA. Results  About 102 microRNA targets of TP53 gene with a target score in the range of 95-50 were identified. The differential expression data for 52 microRNAs was retrieved from the UALCAN database. The microRNAs hsa-miR-421, hsa-miR-548f-5p, and hsa-let-7c-5p were found to be differentially expressed with marked influence over the survival of HNSCC patients. Furthermore, hsa-miR-421 and hsa-let-7c-5p were found to have an exosomal origin especially in body fluids such as blood and saliva. Conclusion  The results accumulated from the present study identified three microRNAs which can affect the functions of TP53 gene and bring about serious outcomes in HNSCC patients. The microRNAs of exosomal origin targeting TP53 gene in HNSCC patients can be a promising prognostic marker, which can be further used as a therapeutic lead by designing inhibitors.

microrna是一种小的非编码rna,通过靶向关键基因来调节一系列功能。在头颈部鳞状细胞癌(HNSCC)患者中观察到TP53谱的显著失调。因此,本研究旨在鉴定那些靶向TP53基因的microrna,并证明它们在HNSCC病例中的差异表达。材料与方法利用EVmiRNA和ExoCarta等数据库进一步研究其外泌体定位。该研究遵循了一种观察性的计算机设计。使用计算工具miRDB鉴定TP53基因的microRNA靶点。使用UALCAN服务器确定来自癌症基因图谱数据集的HNSCC病例中microRNA的表达。基于差异表达的microRNA标记记录HNSCC患者的生存。此外,使用EVmiRNA查询每个microRNA的外泌体存在情况。结果共鉴定出102个TP53基因的microRNA靶点,靶值在95 ~ 50之间。从UALCAN数据库中检索52个microrna的差异表达数据。发现微小rna hsa-miR-421、hsa-miR-548f-5p和hsa-let-7c-5p的差异表达对HNSCC患者的生存有显著影响。此外,发现hsa-miR-421和hsa-let-7c-5p具有外泌体起源,特别是在血液和唾液等体液中。结论本研究积累的结果确定了三种可影响HNSCC患者TP53基因功能并带来严重后果的microrna。外泌体来源的靶向HNSCC患者TP53基因的microrna可能是一个有希望的预后标志物,可以进一步用作设计抑制剂的治疗先导。
{"title":"Exosomal microRNAs Targeting <i>TP53</i> Gene as Promising Prognostic Markers for Head and Neck Squamous Cell Carcinoma.","authors":"Vijayashree Priyadharsini Jayaseelan,&nbsp;Paramasivam Arumugam","doi":"10.1055/s-0042-1758204","DOIUrl":"https://doi.org/10.1055/s-0042-1758204","url":null,"abstract":"<p><p><b>Statement of Problem</b>  MicroRNAs are small non-coding RNAs that regulate an array of functions by targeting crucial genes. A significant dysregulation in the <i>TP53</i> profile has been observed in the head and neck squamous cell carcinoma (HNSCC) patients. Hence, the present <i>in silico</i> study was designed to identify those microRNAs which target <i>TP53</i> gene and demonstrate their differential expression in HNSCC cases. <b>Materials and Methods</b>  The study was extended further to explore their exosomal location using database such as EVmiRNA and ExoCarta. The study follows an observational <i>in silico</i> design. Computational tool miRDB was used identify the microRNA targets of <i>TP53</i> gene. The UALCAN server was used to ascertain the expression of microRNA in HNSCC cases derived from the Cancer Gene Atlas dataset. The survival of HNSCC patients based on the differential expression microRNA markers were recorded. Further, each of the microRNA was queried for their exosomal presence using EVmiRNA. <b>Results</b>  About 102 microRNA targets of <i>TP53</i> gene with a target score in the range of 95-50 were identified. The differential expression data for 52 microRNAs was retrieved from the UALCAN database. The microRNAs hsa-miR-421, hsa-miR-548f-5p, and hsa-let-7c-5p were found to be differentially expressed with marked influence over the survival of HNSCC patients. Furthermore, hsa-miR-421 and hsa-let-7c-5p were found to have an exosomal origin especially in body fluids such as blood and saliva. <b>Conclusion</b>  The results accumulated from the present study identified three microRNAs which can affect the functions of <i>TP53</i> gene and bring about serious outcomes in HNSCC patients. The microRNAs of exosomal origin targeting <i>TP53</i> gene in HNSCC patients can be a promising prognostic marker, which can be further used as a therapeutic lead by designing inhibitors.</p>","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9750795/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10748705","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Advances in Organoid Culture Research. 类器官培养研究进展。
IF 1.7 Pub Date : 2022-12-01 DOI: 10.1055/s-0042-1756662
Zhiyuan Xie, Linghao Wang, Yan Zhang

Organoids are powerful systems to facilitate the study of individuals' disorders and personalized treatments because they mimic the structural and functional characteristics of organs. However, the full potential of organoids in research has remained unrealized and the clinical applications have been limited. One of the reasons is organoids are most efficient grown in reconstituted extracellular matrix hydrogels from mouse-derived, whose poorly defined, batch-to-batch variability and immunogenicity. Another reason is that organoids lack host conditions. As a component of the tumor microenvironment, microbiota and metabolites can regulate the development and treatment in several human malignancies. Here, we introduce several engineering matrix materials and review recent advances in the coculture of organoids with microbiota and their metabolites. Finally, we discuss current trends and future possibilities to build more complex cocultures.

类器官是促进个体疾病研究和个性化治疗的强大系统,因为它们模仿器官的结构和功能特征。然而,类器官在研究中的全部潜力尚未实现,临床应用受到限制。其中一个原因是类器官在小鼠来源的细胞外基质水凝胶中最有效地生长,其定义不清,批次间可变性和免疫原性。另一个原因是类器官缺乏宿主条件。作为肿瘤微环境的组成部分,微生物群和代谢物可以调节多种人类恶性肿瘤的发展和治疗。在此,我们介绍了几种工程基质材料,并对类器官与微生物群及其代谢物共培养的最新进展进行了综述。最后,我们讨论了构建更复杂的共文化的当前趋势和未来可能性。
{"title":"Advances in Organoid Culture Research.","authors":"Zhiyuan Xie,&nbsp;Linghao Wang,&nbsp;Yan Zhang","doi":"10.1055/s-0042-1756662","DOIUrl":"https://doi.org/10.1055/s-0042-1756662","url":null,"abstract":"<p><p>Organoids are powerful systems to facilitate the study of individuals' disorders and personalized treatments because they mimic the structural and functional characteristics of organs. However, the full potential of organoids in research has remained unrealized and the clinical applications have been limited. One of the reasons is organoids are most efficient grown in reconstituted extracellular matrix hydrogels from mouse-derived, whose poorly defined, batch-to-batch variability and immunogenicity. Another reason is that organoids lack host conditions. As a component of the tumor microenvironment, microbiota and metabolites can regulate the development and treatment in several human malignancies. Here, we introduce several engineering matrix materials and review recent advances in the coculture of organoids with microbiota and their metabolites. Finally, we discuss current trends and future possibilities to build more complex cocultures.</p>","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9750796/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10748706","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Differentiation and Immunological Function of MDSC-Derived Dendritic Cells. mdsc来源的树突状细胞的分化和免疫功能。
IF 1.7 Pub Date : 2022-12-01 DOI: 10.1055/s-0042-1756659
Zequn Ding, Yan Zhang

Dendritic cells (DCs) play a key role in initiating and regulating immune responses, and in addition to their roles in vivo, DCs are used as natural adjuvants for various tumor vaccines. In vitro, monocytes can be used to induce DCs, but in tumor patients, due to insufficient bone marrow hematopoiesis, extramedullary hematopoiesis and tumor-associated myeloid cells expand, and monocytes mainly exist in the form of myeloid-derived suppressor cells (MDSCs). The purpose of this experiment was to explore the differences in the differentiation and immune function of DCs induced by MDSCs in tumor patients. In a mouse model, we used normal mouse bone marrow cell-derived DCs as control cells, and in a tumor-bearing model, we induced MDSCs in the spleen to generate DCs (MDSC-DCs). Through flow cytometry, we found that the production of MDSC-DCs was significantly higher than that of control mice, and the secretion of interferon-γ of MDSC-DCs was significantly reduced. Through OVA antigen presentation experiments, we found that the antigen presentation ability of MDSC-DCs was significantly decreased. Through adoptive treatment of tumor-bearing mice cells, we found that the antitumor immune function of MDSC-DCs was significantly reduced. After that, we explored the mechanism of the decrease of immune function activity of MDSC-DCs. We determined that the surface markers of MDSC-DCs were changed by flow cytometry. Through flow sorting and RNA sequencing, we found that some pathways and key gene expression in MDSC-DCs were changed. In conclusion, this study found that the immune function of MDSC-DCs decreased and explored the mechanism of the decreased immune function activity.

树突状细胞(dc)在启动和调节免疫反应中起着关键作用,除了它们在体内的作用外,树突状细胞还被用作各种肿瘤疫苗的天然佐剂。在体外,单核细胞可用于诱导DCs,但在肿瘤患者中,由于骨髓造血功能不足,髓外造血和肿瘤相关髓样细胞扩增,单核细胞主要以髓源性抑制细胞(myeloid-derived suppressor cells, MDSCs)的形式存在。本实验旨在探讨MDSCs在肿瘤患者诱导的dc分化及免疫功能的差异。在小鼠模型中,我们使用正常小鼠骨髓细胞衍生的dc作为对照细胞,在荷瘤模型中,我们诱导脾脏中的MDSCs生成dc (MDSC-DCs)。通过流式细胞术,我们发现MDSC-DCs的产量明显高于对照小鼠,并且MDSC-DCs的干扰素-γ分泌明显减少。通过OVA抗原呈递实验,我们发现MDSC-DCs的抗原呈递能力明显降低。通过对荷瘤小鼠细胞的过继处理,我们发现MDSC-DCs的抗肿瘤免疫功能明显降低。随后,我们探讨了MDSC-DCs免疫功能活性降低的机制。我们通过流式细胞术检测MDSC-DCs的表面标记物发生了变化。通过流式分选和RNA测序,我们发现MDSC-DCs中的一些通路和关键基因表达发生了变化。综上所述,本研究发现MDSC-DCs的免疫功能下降,并探讨了免疫功能活性下降的机制。
{"title":"Differentiation and Immunological Function of MDSC-Derived Dendritic Cells.","authors":"Zequn Ding,&nbsp;Yan Zhang","doi":"10.1055/s-0042-1756659","DOIUrl":"https://doi.org/10.1055/s-0042-1756659","url":null,"abstract":"<p><p>Dendritic cells (DCs) play a key role in initiating and regulating immune responses, and in addition to their roles in vivo, DCs are used as natural adjuvants for various tumor vaccines. In vitro, monocytes can be used to induce DCs, but in tumor patients, due to insufficient bone marrow hematopoiesis, extramedullary hematopoiesis and tumor-associated myeloid cells expand, and monocytes mainly exist in the form of myeloid-derived suppressor cells (MDSCs). The purpose of this experiment was to explore the differences in the differentiation and immune function of DCs induced by MDSCs in tumor patients. In a mouse model, we used normal mouse bone marrow cell-derived DCs as control cells, and in a tumor-bearing model, we induced MDSCs in the spleen to generate DCs (MDSC-DCs). Through flow cytometry, we found that the production of MDSC-DCs was significantly higher than that of control mice, and the secretion of interferon-γ of MDSC-DCs was significantly reduced. Through OVA antigen presentation experiments, we found that the antigen presentation ability of MDSC-DCs was significantly decreased. Through adoptive treatment of tumor-bearing mice cells, we found that the antitumor immune function of MDSC-DCs was significantly reduced. After that, we explored the mechanism of the decrease of immune function activity of MDSC-DCs. We determined that the surface markers of MDSC-DCs were changed by flow cytometry. Through flow sorting and RNA sequencing, we found that some pathways and key gene expression in MDSC-DCs were changed. In conclusion, this study found that the immune function of MDSC-DCs decreased and explored the mechanism of the decreased immune function activity.</p>","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9771685/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10428187","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of FOCAD: An Important Gene in Liver Cirrhosis. 发现FOCAD:肝硬化的重要基因。
IF 1.7 Pub Date : 2022-12-01 DOI: 10.1055/s-0042-1758351
Jinjin Shao
Liver cirrhosis is the 11th most common cause of death, causing more than 1 million deaths globally each year, and together with liver cancer, it accounts for 3.5% of all deaths worldwide.1 Cirrhosis develops due to long-term chronic liver inflammation, with the replacement of healthy liver parenchyma with diffuse liver fibrosis and regenerative nodules, leading to portal hypertension and various complications and even hepatocellular carcinoma.2–4 The management of cirrhosis is mainly based on the treatment of cause and complications including comprehensive supportive care. However, there are currently no effective drug therapies to cure the disease, and liver transplantation remains the gold standard treatment for cirrhosis.5,6 Given that cirrhosis has brought heavy health and economic burden to many countries, there is an urgent need for in-depth study of the pathogenesis and key factors of cirrhosis to find feasible intervention strategies. Cirrhosis is traditionally considered a late-onset disease that appears in adults following environmental factors, such as viral infection, a high-fat diet, or chronic alcoholism.7,8 Thus, cirrhosis caused by genetic factors appears to have received less attention than environmental factors, and the etiology in infants and young children is far less understood. In a recent study published in Nature Genetics, titled “Loss of FOCAD, manipulated through the SKI messenger RNA surveillance pathway, leads to a pediatric syndrome with cirrhosis,” Traspas and colleagues uncovered the essentiality of the FOCAD gene in maintaining liver health and provided evidence that loss-of-function mutations in FOCAD may contribute to cirrhosis in children.9 The authors reported 14 children from 10 unrelated families in seven countries presenting with a multisystem syndrome characterized by severe neonatal cirrhosis. By combing genome/exome sequencing, a novel animal model of the human disease, and in vitro biological systems, the team identify that the FOCAD gene is indispensable for maintaining human liver health. Mutations in this gene cause a form of early-onset cirrhosis that has not been documented before. Using CRISPR-Cas9 technology, they established in vitro and in vivo FOCAD knockout models to further study the cellular and molecular mechanisms of pediatric cirrhosis. Phenotypic replication of human disease in FOCAD-deficient zebrafish reveals features of altered messenger RNA degradation processes in the liver. FOCAD deficiency in patient primary cells and human liver cell lines impairs the SKI mRNA surveillance pathway by reducing levels of the RNA helicase SKIC2 and its cofactor SKIC3. Compared with other cell types, hepatocytes rely heavily on this mechanism. Hepatocytes exhibited a decrease in albumin expression and overproduction of the cytokine CCL2 following FOCAD knockout, which may play a key role in the progression of cirrhosis. These findings reveal the importance of FOCAD in maintaining liver homeostasis a
{"title":"Discovery of FOCAD: An Important Gene in Liver Cirrhosis.","authors":"Jinjin Shao","doi":"10.1055/s-0042-1758351","DOIUrl":"https://doi.org/10.1055/s-0042-1758351","url":null,"abstract":"Liver cirrhosis is the 11th most common cause of death, causing more than 1 million deaths globally each year, and together with liver cancer, it accounts for 3.5% of all deaths worldwide.1 Cirrhosis develops due to long-term chronic liver inflammation, with the replacement of healthy liver parenchyma with diffuse liver fibrosis and regenerative nodules, leading to portal hypertension and various complications and even hepatocellular carcinoma.2–4 The management of cirrhosis is mainly based on the treatment of cause and complications including comprehensive supportive care. However, there are currently no effective drug therapies to cure the disease, and liver transplantation remains the gold standard treatment for cirrhosis.5,6 Given that cirrhosis has brought heavy health and economic burden to many countries, there is an urgent need for in-depth study of the pathogenesis and key factors of cirrhosis to find feasible intervention strategies. Cirrhosis is traditionally considered a late-onset disease that appears in adults following environmental factors, such as viral infection, a high-fat diet, or chronic alcoholism.7,8 Thus, cirrhosis caused by genetic factors appears to have received less attention than environmental factors, and the etiology in infants and young children is far less understood. In a recent study published in Nature Genetics, titled “Loss of FOCAD, manipulated through the SKI messenger RNA surveillance pathway, leads to a pediatric syndrome with cirrhosis,” Traspas and colleagues uncovered the essentiality of the FOCAD gene in maintaining liver health and provided evidence that loss-of-function mutations in FOCAD may contribute to cirrhosis in children.9 The authors reported 14 children from 10 unrelated families in seven countries presenting with a multisystem syndrome characterized by severe neonatal cirrhosis. By combing genome/exome sequencing, a novel animal model of the human disease, and in vitro biological systems, the team identify that the FOCAD gene is indispensable for maintaining human liver health. Mutations in this gene cause a form of early-onset cirrhosis that has not been documented before. Using CRISPR-Cas9 technology, they established in vitro and in vivo FOCAD knockout models to further study the cellular and molecular mechanisms of pediatric cirrhosis. Phenotypic replication of human disease in FOCAD-deficient zebrafish reveals features of altered messenger RNA degradation processes in the liver. FOCAD deficiency in patient primary cells and human liver cell lines impairs the SKI mRNA surveillance pathway by reducing levels of the RNA helicase SKIC2 and its cofactor SKIC3. Compared with other cell types, hepatocytes rely heavily on this mechanism. Hepatocytes exhibited a decrease in albumin expression and overproduction of the cytokine CCL2 following FOCAD knockout, which may play a key role in the progression of cirrhosis. These findings reveal the importance of FOCAD in maintaining liver homeostasis a","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9748444/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10765414","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic Alteration in Colorectal Cancer: A Biomarker for Diagnostic and Therapeutic Application. 大肠癌的表观遗传学改变:诊断和治疗应用的生物标志物
IF 1.2 Q4 GENETICS & HEREDITY Pub Date : 2022-09-29 eCollection Date: 2022-09-01 DOI: 10.1055/s-0042-1757404
Hafsa Yousif Solayman Essa, Gunay Kusaf, Ozel Yuruker, Rasime Kalkan

Colorectal cancer (CRC) is the leading cause of cancer death worldwide. A crucial process that initiates and progresses CRC is various epigenetic and genetic changes occurring in colon epithelial cells. Recently, huge progress has been made to understand cancer epigenetics, especially regarding DNA methylation changes, histone modifications, dysregulation of miRNAs and noncoding RNAs. In the "epigenome" of colon cancer, abnormal methylation of genes that cause gene alterations or expression of miRNA has been reported in nearly all CRC; these findings can be encountered in the average CRC methylome. Epigenetic changes, known as driving events, are assumed to play a dominant part in CRC. Furthermore, as epigenetic changes in CRC become properly understood, these changes are being established as clinical biomarkers for therapeutic and diagnostic purposes. Progression in this area indicates that epigenetic changes will often be utilized in the future to prevent and treat CRC.

结肠直肠癌(CRC)是全球癌症死亡的主要原因。结肠上皮细胞发生的各种表观遗传学和基因变化是导致 CRC 发生和发展的关键过程。最近,人们在了解癌症表观遗传学方面取得了巨大进展,尤其是在 DNA 甲基化变化、组蛋白修饰、miRNA 和非编码 RNA 的失调方面。在结肠癌的 "表观基因组 "中,几乎所有的 CRC 都有导致基因改变或 miRNA 表达的基因甲基化异常的报道;这些发现在一般的 CRC 甲基组中都能遇到。表观遗传学变化被称为驱动事件,被认为在 CRC 中起着主导作用。此外,随着人们对 CRC 中表观遗传变化的正确理解,这些变化正被确立为临床生物标志物,用于治疗和诊断。这一领域的进展表明,表观遗传变化在未来将经常被用来预防和治疗 CRC。
{"title":"Epigenetic Alteration in Colorectal Cancer: A Biomarker for Diagnostic and Therapeutic Application.","authors":"Hafsa Yousif Solayman Essa, Gunay Kusaf, Ozel Yuruker, Rasime Kalkan","doi":"10.1055/s-0042-1757404","DOIUrl":"10.1055/s-0042-1757404","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is the leading cause of cancer death worldwide. A crucial process that initiates and progresses CRC is various epigenetic and genetic changes occurring in colon epithelial cells. Recently, huge progress has been made to understand cancer epigenetics, especially regarding DNA methylation changes, histone modifications, dysregulation of miRNAs and noncoding RNAs. In the \"epigenome\" of colon cancer, abnormal methylation of genes that cause gene alterations or expression of miRNA has been reported in nearly all CRC; these findings can be encountered in the average CRC methylome. Epigenetic changes, known as driving events, are assumed to play a dominant part in CRC. Furthermore, as epigenetic changes in CRC become properly understood, these changes are being established as clinical biomarkers for therapeutic and diagnostic purposes. Progression in this area indicates that epigenetic changes will often be utilized in the future to prevent and treat CRC.</p>","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.2,"publicationDate":"2022-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9522482/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40390400","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Designing In-House SARS-CoV-2 RT-qPCR Assay for Variant of Concerns. 设计内部SARS-CoV-2关注变异的RT-qPCR检测方法。
IF 1.7 Pub Date : 2022-09-20 eCollection Date: 2022-09-01 DOI: 10.1055/s-0042-1756660
Mahmut Cerkez Ergoren, Gulten Tuncel, Cenk Serhan Ozverel, Tamer Sanlidag

Variants (Alfa, Gamma, Beta, and Delta) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are circulating worldwide. These variants of concerns share some common mutations but they also have distinguishing mutations. These mutations affect transmissibility of virus and cause evasion from neutralizing antibodies. Monitoring and identification of circulating variants is of great importance for public health. In this study, an in-house SARS-CoV-2 reverse transcription quantitative polymerase chain reaction (RT-qPCR) kit was designed to detect variants of concerns by the World Health Organization. Primer sets and probes were designed to target presence of virus along with mutations for identifying different variants (for N501Y, HV69-70del, K417N, and T478K). Reactions were set by using commercially available master mixes without a reference dye. The RT-qPCR conditions were optimized by using commercially available ribonucleic acid samples of wild-type, Alfa, Beta, Gamma, and Delta variants. Several samples were also analyzed by the in-house kit after optimization studies. All Alfa variant and wild-type samples were also double confirmed with a commercially available variant detection kit demonstrating a 100% consistence with the in-house kit. Beta, Gamma, and Delta variants could not be confirmed with any other commercially available kits as there is not any available one in the market. SARS-CoV-2 variants are gaining importance during the pandemic and shaping the fight against the virus. RT-qPCR kits detecting different variants would provide a significant advantage while screening the population.

严重急性呼吸综合征冠状病毒2 (SARS-CoV-2)的变体(阿尔法、伽玛、贝塔和德尔塔)正在全球传播。这些关注点的变体共享一些共同的突变,但它们也有不同的突变。这些突变影响病毒的传播性,并导致逃避中和抗体。监测和识别流行变异对公共卫生非常重要。本研究设计了一种内部SARS-CoV-2逆转录定量聚合酶链反应(RT-qPCR)试剂盒,用于检测世界卫生组织关注的变异。引物组和探针针对病毒和突变的存在而设计,用于识别不同的变异(N501Y、HV69-70del、K417N和T478K)。反应是通过使用市售的母料而不使用参考染料来设定的。利用市售的野生型、α型、β型、γ型和δ型变异的核糖核酸样本对RT-qPCR条件进行优化。在优化研究后,还使用内部试剂盒对几个样品进行了分析。所有α变异和野生型样本也用市售变异检测试剂盒进行双重确认,证明与内部试剂盒的一致性为100%。Beta、Gamma和Delta变体无法与任何其他商用套件确认,因为市场上没有任何可用的套件。SARS-CoV-2变体在大流行期间越来越重要,并影响着抗击该病毒的斗争。RT-qPCR试剂盒检测不同的变异将在筛查人群时提供显著的优势。
{"title":"Designing In-House SARS-CoV-2 RT-qPCR Assay for Variant of Concerns.","authors":"Mahmut Cerkez Ergoren,&nbsp;Gulten Tuncel,&nbsp;Cenk Serhan Ozverel,&nbsp;Tamer Sanlidag","doi":"10.1055/s-0042-1756660","DOIUrl":"https://doi.org/10.1055/s-0042-1756660","url":null,"abstract":"<p><p>Variants (Alfa, Gamma, Beta, and Delta) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are circulating worldwide. These variants of concerns share some common mutations but they also have distinguishing mutations. These mutations affect transmissibility of virus and cause evasion from neutralizing antibodies. Monitoring and identification of circulating variants is of great importance for public health. In this study, an in-house SARS-CoV-2 reverse transcription quantitative polymerase chain reaction (RT-qPCR) kit was designed to detect variants of concerns by the World Health Organization. Primer sets and probes were designed to target presence of virus along with mutations for identifying different variants (for N501Y, HV69-70del, K417N, and T478K). Reactions were set by using commercially available master mixes without a reference dye. The RT-qPCR conditions were optimized by using commercially available ribonucleic acid samples of wild-type, Alfa, Beta, Gamma, and Delta variants. Several samples were also analyzed by the in-house kit after optimization studies. All Alfa variant and wild-type samples were also double confirmed with a commercially available variant detection kit demonstrating a 100% consistence with the in-house kit. Beta, Gamma, and Delta variants could not be confirmed with any other commercially available kits as there is not any available one in the market. SARS-CoV-2 variants are gaining importance during the pandemic and shaping the fight against the virus. RT-qPCR kits detecting different variants would provide a significant advantage while screening the population.</p>","PeriodicalId":40142,"journal":{"name":"Global Medical Genetics","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2022-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9489470/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"33477133","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
期刊
Global Medical Genetics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1