Human MutationPub Date : 2025-08-29DOI: 10.1155/humu/6824122
Siying Zhu, Hongxu Tao, Robert A. Cornell, Huan Liu
{"title":"Functional Validation of Noncoding Variants Associated With Nonsyndromic Orofacial Cleft","authors":"Siying Zhu, Hongxu Tao, Robert A. Cornell, Huan Liu","doi":"10.1155/humu/6824122","DOIUrl":"https://doi.org/10.1155/humu/6824122","url":null,"abstract":"<p>Over the past decade, genome-wide association studies (GWASs) have found genetic variants associated with elevated risk for nonsyndromic orofacial cleft (NSOFC). In the post-GWAS era of NSOFC genetic research, an important aim is to identify the pathogenic variants that influence craniofacial development processes, towards understanding how they lead to disease manifestation. However, two major challenges hinder the translation of GWAS results into a mechanistic understanding. Firstly, it is uncertain whether the variants pinpointed by GWAS represent the underlying pathogenic variants; secondly, the bulk of genetic variants identified through GWAS are situated in noncoding regions of the genome, complicating their biological interpretation. Presently, research on noncoding genetic variants associated with NSOFC predominantly centers on variants located in transcriptional regulatory elements. These variants modulate transcription, subsequently altering the expression of downstream target genes and disrupting gene regulatory networks. We provide a systematic summary of the recent NSOFC-associated GWAS findings for the first time. With a particular focus on variants located in noncoding regions, we delve into current statistical methods and functional approaches for identifying and validating causal variants, aiming to bridge the gap between genetic variants identified by GWAS and their underlying pathogenic mechanism responsible for NSOFC. Deciphering causal variants underlying NSOFC offers valuable clinical insights that may advance early diagnosis, enhance risk stratification, and facilitate the discovery of novel therapeutic targets.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/6824122","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144914858","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Optical Genomic Mapping and Next-Generation Sequencing Identified Retrotransposon Insertion and Missense Variant Disrupting PARN Gene in Dyskeratosis Congenita","authors":"Qiaoyu Cao, Anqi Zhao, Zhoukai Long, Xinyi Wang, Chaolan Pan, Yumeng Wang, Wei He, Haisheng Huang, Fuying Chen, Chenfei Wang, Xiaoxiao Wang, Luming Sun, Jingjun Zhao, Ming Li","doi":"10.1155/humu/9290736","DOIUrl":"https://doi.org/10.1155/humu/9290736","url":null,"abstract":"<p>Dyskeratosis congenita (DC) is an inherited bone marrow failure syndrome characterized by defects in telomere biology and clinical manifestations such as nail dystrophy, skin pigmentation abnormalities, and mucosal leukoplakia. Here, using whole exome sequencing (WES), whole genome sequencing (WGS), optical mapping sequencing (OGM), third-generation sequencing, and mRNA sequencing, we diagnosed a participant with <i>PARN</i> gene complex compound heterozygous variants. In addition, protein structure simulation, immunohistochemistry, and western blot were conducted to investigate the structure and expression level of the PARN protein. WES revealed a maternal <i>PARN</i> variant, c.204G>T (p.Gln68His) (NM_002582.3). An insertion variant in the <i>PARN</i> gene from the father was identified by OGM and mRNA sequencing. Third-generation sequencing results determined the insertion position of the SINE-VNTR-Alu (SVA) transposon and its size (2537 bp), which was found to lead to a premature stop codon (p.Gly469delinsGlu∗). The PARN protein level of the parents was reduced due to complex heterozygous variants. Overall, OGM diagnosed the structural variants of the participant with DC, supplementing the disease variant spectrum of DC. This case highlights a novel disease-causing structural variant and the importance of transposon analysis in a clinical diagnostic setting.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/9290736","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144888148","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Exploring the Molecular Functions and Immune Relevance of Macrophage-Associated Genes in Atherosclerosis","authors":"Chenchen Yu, Haoran Wang, Huiting Xu, Peipei Kang, Jingjing Shao, Hui Zhang","doi":"10.1155/humu/9034896","DOIUrl":"https://doi.org/10.1155/humu/9034896","url":null,"abstract":"<p>Atherosclerosis is a common and significant cardiovascular condition that frequently goes undiagnosed by conventional diagnostic and treatment techniques until it reaches a more advanced stage. This challenge impedes the capacity to apply early detection and intervention measures. As a result, the creation of innovative and more accurate biomarkers is critically important. The study first recognizes genes associated with macrophages through single-cell analysis, investigating their functions. Subsequently, various machine learning approaches are utilized to identify significant regulatory genes related to macrophages. In addition, molecular docking studies are performed to evaluate the binding affinity of these crucial markers with therapeutics targeting atherosclerosis. The ImmuCellAI platform is also utilized to assess immune cell scores in atherosclerotic samples, aiding in the examination of connections between vital diagnostic markers and immune cells. Finally, the expression changes of the selected key genes are confirmed using qRT-PCR and Western blot methods. Through analyses at the single-cell level and differential assessments, we discovered 58 genes related to macrophages that exhibited differential expression. Functional evaluations indicated a strong correlation between these genes and the immune microenvironment. By conducting cluster analysis, we assessed how different subgroups of patients with atherosclerosis respond to immunotherapy. Utilizing techniques such as XGBoost, random forest, and the GOsemsim algorithm, we pinpointed five crucial diagnostic markers. Studies on molecular docking validated that these important markers could act as potential drug targets for atherosclerosis. Finally, our experimental analysis revealed a significant overexpression of these five diagnostic markers in tissues affected by atherosclerosis. This research introduces novel diagnostic indicators associated with macrophages in atherosclerosis and emphasizes their potential as targets for therapies related to the immune system.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/9034896","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144881270","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Key Genes Associated With Functional Specialization of Neonatal Peripheral Monocytes","authors":"Tingyan Xie, Zicheng Huang, Xian Chen, Zhenchao Jin, Bing Yang, Quan Tang","doi":"10.1155/humu/3009253","DOIUrl":"https://doi.org/10.1155/humu/3009253","url":null,"abstract":"<p><b>Purpose:</b> The purpose of this study is to identify genes and transcription factors underlying functional differences in neonatal versus adult peripheral blood monocytes, elucidating mechanisms of severe Group B streptococcus (GBS) infection in neonates.</p><p><b>Methods:</b> Differentially expressed genes (DEGs) in neonatal and adult peripheral blood monocytes were detected via RNA sequencing (RNA-seq), followed by assay for transposase-accessible chromatin sequencing (ATAC-seq) to characterize differentially accessible region (DAR)–associated genes. Integrated analyses of RNA-seq and ATAC-seq pinpointed candidate genes and transcription factors. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) validated the mRNA expression of common genes and transcription factors.</p><p><b>Results:</b> RNA-seq profiling of neonatal and adult peripheral monocytes identified 669 overexpressed and 440 underexpressed genes in neonates, with overexpressed genes enriched in bacterial response pathways and underexpressed genes in cytokine production and cell killing pathways. Chromatin accessibility analysis revealed 36,782 differential peaks (21,192 gained, 15,590 lost) in neonatal peripheral monocytes. Integrated RNA-seq and ATAC-seq analysis pinpointed 30 overlapping genes among DEGs, DAR-associated genes, and immunologically relevant genes (IRGs). qRT-PCR validated higher expression of <i>CEBPB</i>, <i>JUN</i>, <i>BATF</i>, <i>PTK2B</i>, and <i>ITGAV</i> and lower <i>ADA2</i> and <i>RORA</i> expression in neonatal peripheral monocytes compared to that in adults.</p><p><b>Conclusions:</b> The study revealed distinct differences in the transcriptome and chromatin accessibility between neonatal and adult peripheral monocytes, identifying potential genes linked to GBS infection vulnerability of neonates. These findings advance our understanding of neonatal immune dysfunction in severe GBS disease, informing future therapeutic targets.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/3009253","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144869299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human MutationPub Date : 2025-08-17DOI: 10.1155/humu/1743829
Ting Gong, Bin Jia, Hui Lv, Lili Zeng, Diansheng Zhong
{"title":"Computational Analyses Identified Three Diagnostic Biomarkers Associated With Programmed Cell Death for Lung Adenocarcinoma","authors":"Ting Gong, Bin Jia, Hui Lv, Lili Zeng, Diansheng Zhong","doi":"10.1155/humu/1743829","DOIUrl":"https://doi.org/10.1155/humu/1743829","url":null,"abstract":"<p><b>Background:</b> The high morbidity and mortality of lung adenocarcinoma (LUAD) are partly caused by a lack of sensitive and reliable molecular markers for early diagnosis. Programmed cell death (PCD) is a crucial process involved in tumorigenesis and immune regulation, and identifying PCD-correlated genes may contribute to the precision diagnosis and targeted therapy of LUAD.</p><p><b>Methods:</b> LUAD samples were acquired from UCSC Xena and Gene Expression Omnibus (GEO) database. PCD-correlated module genes were identified by WGCNA. “Limma” package was employed for screening differentially expressed genes (DEGs) between LUAD and control samples, followed by conducting functional enrichment analysis with “ClusterProfiler” package. Hub genes were selected through machine learning algorithms. Biomarkers for LUAD were screened and further validated by receiver operating characteristic (ROC) analysis. The robustness of the diagnostic model was verified by the confusion matrix. Immune cell infiltration was assessed employing “ESTIMATE” and “GSVA” packages. HALLMARK pathway score was calculated with the “GSVA” package. Transcription factor (TF)–biomarker–chemical network was established using NetworkAnalyst and Cytoscape software. The expressions of the biomarkers in LUAD cells were detected by in vitro experiments. The viability, migration, and invasion of the LUAD cells were measured by CCK-8, wound healing, and Transwell assays.</p><p><b>Results:</b> We identified 160 module genes and 5934 DEGs. Then, eight hub genes were selected applying LASSO and support vector machine–recursive feature elimination (SVM-RFE) analyses. Further, <i>FGR</i>, <i>TLR4</i>, and <i>NLRC4</i>, which showed an area under the ROC curve (AUC) > 0.7, were determined as the diagnostic biomarkers for LUAD. Interestingly, they were all low expressed in LUAD samples. We developed a diagnostic model that demonstrated robust performance in distinguishing LUAD samples from normal controls. The three biomarkers showed positive correlation to the infiltration of most immune cells and enrichment in HALLMARK pathways associated with inflammation, immune regulation, and cytokine signaling. Moreover, nine TFs and nine small-molecule compounds targeting the three biomarkers were predicted to construct a TF–biomarker–chemical network. Functional validation revealed that all the three biomarkers were significantly downregulated in LUAD cells. Notably, <i>FGR</i> overexpression markedly suppressed LUAD cell proliferation, migration, and invasion.</p><p><b>Conclusion:</b> This study identified three PCD-related biomarkers for LUAD diagnosis, providing new potential therapeutic targets.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/1743829","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144861626","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human MutationPub Date : 2025-08-15DOI: 10.1155/humu/5530710
Tahir N. Khan, Chunyu Liu, Kai Lee Yap, Humayoon Shafique Satti, Ayaz Khan, Muhammad Safeer, Sheraz Khan, Naveed Altaf Malik, Feng Zhang, Muhammad Tariq, Erica E. Davis
{"title":"Genetic Investigation and Transcriptome Profiling in a Nuclear Family With Peutz–Jeghers Syndrome","authors":"Tahir N. Khan, Chunyu Liu, Kai Lee Yap, Humayoon Shafique Satti, Ayaz Khan, Muhammad Safeer, Sheraz Khan, Naveed Altaf Malik, Feng Zhang, Muhammad Tariq, Erica E. Davis","doi":"10.1155/humu/5530710","DOIUrl":"https://doi.org/10.1155/humu/5530710","url":null,"abstract":"<p>Peutz–Jeghers syndrome (PJS) is a rare autosomal dominant disorder hallmarked by mucocutaneous melanocytic macules and gastrointestinal hamartomatous polyposis associated with germline/somatic pathogenic variants in the tumor suppressor <i>STK11</i>. PJS is clinically heterogeneous; however, the relationship between clinical phenotype and genotype remains elusive. Here, we report a family with PJS that harbors a heterozygous <i>STK11</i> whole gene deletion combined with a heterozygous variant in <i>TP53AIP1</i> that segregates with mucocutaneous pigmentation in the family. RNA-seq analysis followed by qRT-PCR confirmed that the expression of <i>STK11</i>, <i>TP53</i>, and <i>TP53AIP1</i> and a large fraction of p53 signaling pathway components are significantly reduced, while Wnt signaling pathway effectors are upregulated in cells from an affected individual. Our findings shed light on transcriptome-level pathway dysregulation in PJS with germline deletion of <i>STK11</i>. Further evaluation of mutational burden across relevant signaling pathways can likely inform disease prognosis.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/5530710","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144853830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human MutationPub Date : 2025-08-08eCollection Date: 2025-01-01DOI: 10.1155/humu/8771129
Lu Yang, Chunping Bo, Meiqi Chen, Bozhen Chen, Rui Zeng, Yingyan Zhou, Haifang Du, Xiaohong He
{"title":"Multiomics Identifies Potential Biomarkers in Ankylosing Spondylitis Bone Formation.","authors":"Lu Yang, Chunping Bo, Meiqi Chen, Bozhen Chen, Rui Zeng, Yingyan Zhou, Haifang Du, Xiaohong He","doi":"10.1155/humu/8771129","DOIUrl":"10.1155/humu/8771129","url":null,"abstract":"<p><p><b>Objective:</b> Ankylosing spondylitis (AS) is a long-term inflammatory condition characterized by intricate pathogenesis and significant genetic predisposition. Current treatment methods cannot completely halt the progression of the disease. The purpose of this research is to discover possible therapeutic targets for AS by integrating Mendelian Randomization (MR), transcriptomics analysis, and machine learning, providing new options for the clinical treatment of AS. <b>Methods:</b> In this study, we initially pinpointed differentially expressed genes (DEGs) linked to AS from the GEO database and acquired cis-eQTL data for these genes from the eQTLGen Consortium. Using MR and summary data-based Mendelian randomization (SMR) analyses, we screened for DEGs with causal relationships to AS. Subsequently, we analyzed the correlation between these causal genes and immune cell expression, constructed a risk prediction model, and identified key feature genes for AS. Next, we conducted phenome-wide association studies (PheWASs) on the identified AS feature genes to predict their potential adverse effects as therapeutic targets. We obtained AS-related therapeutic drugs from the DrugBank database and performed molecular docking analysis with AS feature genes. We used the CAIA collagen-induced AS mouse model; we measured joint swelling and employed microCT, H&E, and Safranin O-Fast Green staining to assess pathological changes in bone tissue. Additionally, we employed Western blot and RT-qPCR to analyze the expression levels of genes associated with bone mineralization and AS feature genes in joint tissues. <b>Results:</b> A total of 1607 DEGs were obtained from the GEO database. After MR analysis and correction, 33 positive DEGs that have a causal relationship with AS were determined. Through the correlation analysis between these genes and the expressions of immune cells, it was found that 28 genes had significant regulatory relationships with 19 kinds of immune cells, with 55 pairs of negative regulatory relationships and 49 pairs of positive regulatory relationships, respectively. Four machine learning model algorithms determined the Top 5 genes (RIOK1, FUCA2, COL9A2, USP16, and TTC16) with the highest importance scores and constructed a nomogram to evaluate the risk probability. The results of the PheWAS showed that the five characteristic genes of AS had harmful or beneficial effects on numerous disease phenotypes of multiple types of diseases. Molecular docking indicated that 14 known AS treatment drugs had potential interactions with related genes. Using RT-qPCR, we evaluated the expression levels of five key genes in the joint tissue of the CAIA collagen-induced AS mouse model. Compared to the normal control group, we found that the levels of <i>FUCA2</i> and <i>USP16</i> were significantly elevated, while the levels of <i>TTC16</i> were significantly reduced. In contrast, the expression of <i>COL9A2</i> and <i>RIOK1</i> mRNA showed no signi","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 ","pages":"8771129"},"PeriodicalIF":3.7,"publicationDate":"2025-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12356670/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144872996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Single-Cell RNA Sequencing Reveals LEF1 as a Prognostic Biomarker for Poor Outcomes in Oxaliplatin-Resistant Colorectal Cancer","authors":"Pin Huang, Ke Guo, Jiancheng Tu, Jian Fang, Liang Zhou, Xiagang Luo, Hubin Xu","doi":"10.1155/humu/6705599","DOIUrl":"https://doi.org/10.1155/humu/6705599","url":null,"abstract":"<p>Colorectal cancer (CRC) is a leading cause of cancer-related morbidity and mortality worldwide. Despite the efficacy of oxaliplatin-based chemotherapy (CT) in CRC treatment, CT resistance remains a major obstacle to successful patient outcomes. Epithelial–mesenchymal transition (EMT), a key cellular process in cancer metastasis, plays a pivotal role in resistance to CT. The tumor microenvironment (TME), particularly cancer-associated fibroblasts (CAFs), is known to contribute to EMT and therapy resistance. Here, we employ single-cell RNA sequencing (scRNA-seq) to analyze primary CRC tumor samples from patients undergoing CT and nonchemotherapy (nCT) treatments. Our study identifies specific epithelial cell clusters resistant to oxaliplatin, elucidating the molecular pathways involved in EMT and resistance. Furthermore, we explore the role of CAF subpopulations in promoting resistance within the TME. Our findings highlight the importance of functional immune profiling and genomic analyses in identifying potential biomarkers for predicting CT responses and improving personalized treatment strategies. This work provides new insights into the molecular mechanisms of oxaliplatin resistance in CRC and supports the development of novel immune-based therapeutic approaches to enhance patient outcomes.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/6705599","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144782385","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Integration of Immune Cell Signatures and Diagnostic Gene Markers in Pancreatitis: A Comprehensive Study on Therapeutic Targets and Predictive Diagnosis","authors":"Qianyu Xie, Birong Liu, Xiao Yu, Xiang Wei, Qiangsheng Xiao","doi":"10.1155/humu/7694723","DOIUrl":"https://doi.org/10.1155/humu/7694723","url":null,"abstract":"<p>Pancreatitis is a severe and increasingly prevalent disease that affects the digestive system. Early detection and accurate diagnosis of this condition are crucial for reducing mortality rates and improving patient outcomes. Therefore, the development of novel diagnostic markers is essential for enhancing clinical management and advancing the understanding of pancreatitis. The initial phase involved applying the ssGSEA method to extract hypoxia scores from these samples. Subsequently, a thorough differential expression analysis was performed, complemented by functional assessments and various machine learning techniques designed to pinpoint critical diagnostic genes relevant to pancreatitis. From this, a robust diagnostic model was constructed and validated using a series of machine learning strategies. To further validate our results, molecular docking studies were conducted to determine the binding affinities between the identified markers and standard medications such as omeprazole and lansoprazole. Additionally, the ssGSEA methodology was leveraged to compute immune cell scores within the pancreatitis samples, thus enriching the analysis of the relationships between significant diagnostic genes and various immune cell types. Finally, the experiments of ELISA and qRT-PCR were used to verify the expression of key target genes. Through WGCNA, we identified a total of 50 genes associated with hypoxic conditions within the pancreatitis samples. Further investigations, including differential expression analysis and machine learning techniques, revealed six significant diagnostic markers for pancreatitis: RAP1GDS1, TOP2A, ADK, POLL, CD44, and CD4. The diagnostic model we developed exhibited a high accuracy level in predicting pancreatitis onset, while molecular docking analyses indicated that these six key diagnostic genes hold promise as drug targets. Moreover, the ssGSEA algorithm confirmed the relationships between these diagnostic markers and a range of immune cell populations. Ultimately, the expression levels of the identified key genes were rigorously validated through experimental techniques, reinforcing the credibility of our findings.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/7694723","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144758607","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human MutationPub Date : 2025-07-23DOI: 10.1155/humu/3468799
Mikiko Soejima, Yoshiro Koda
{"title":"Simultaneous Genotyping of Three Nonsynonymous SNVs, rs1042602, rs1426654, and rs16891982 Involved in Skin Pigmentation by Fluorescent Probe-Based Melting Curve Analysis","authors":"Mikiko Soejima, Yoshiro Koda","doi":"10.1155/humu/3468799","DOIUrl":"https://doi.org/10.1155/humu/3468799","url":null,"abstract":"<p>Three nonsynonymous single nucleotide variations (SNVs), rs1042602 in TYR (p.S192Y), rs1426654 in SLC24A5 (p.A111T), and rs16891982 in SLC45A2 (p.L374F), were associated with human skin pigmentation variation and may have recently undergone positive natural selection. Furthermore, these three SNVs have been reported to correlate with the risk and prognosis of melanoma. To simultaneously determine these three SNVs, a triplex fluorescent probe-based melting curve assay (FMCA) was developed. The method was validated by analyzing genomic DNA from subjects with known genotypes. For rs16891982, triplex FMCA did not allow good separation of genotypes with the initial polymerase enzyme mix used, but by changing the enzyme mix used, the three genotypes could be clearly distinguished. Using this method, we definitively genotyped these three SNVs in 93 European, 58 Tamil, 54 Sinhalese, and 52 Bangladeshi subjects. This method allows genotyping of rs1042602, rs1426654, and rs16891982 in a relatively large number of samples to perform association studies on skin pigmentation variation or melanoma risk.</p>","PeriodicalId":13061,"journal":{"name":"Human Mutation","volume":"2025 1","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/humu/3468799","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144688062","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}