1
|
Chopra C, Kukkar D, Bhatt P, Rajesh P, Kim KH. A review of deoxyribonucleic acid-based single-nucleotide polymorphisms in diabetic kidney disease among Asian populations: Challenges and future directions. Int J Biol Macromol 2025; 316:144407. [PMID: 40403785 DOI: 10.1016/j.ijbiomac.2025.144407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 05/15/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Diabetic kidney disease (DKD) is a persistent disorder that occurs as a result of long-term diabetes mellitus with genetic and environmental risk factors. The identification of DKD associated single-nucleotide polymorphisms (SNPs) is pivotal for patient screening. This manuscript briefly outlines the pathophysiology and the role of genetic factors in DKD expansion. It further discusses the utility of bioinformatic tools and laboratory techniques requried for the identification of DKD-specific SNPs along with integrated data analysis pipelines valuable to enhance the accuracy of genetic interpretation. A comparative analysis of various SNPs has also been made across diverse Asian populations in conjunction with environmental and lifestyle factors. The clinical relevance of SNPs in predicting DKD progression and stratifying patient risk is highlighted, with focus on gene-specific pathways and associated functional outcomes. The advances in genetic screening, gene-specific therapies, and microbiome-based therapy should help expand the utility of SNPs-based identification of DKD under diverse clinical settings. A structured clinical decision-making framework is proposed to support customized treatment based on SNP profiles. However, this domain has yet to gain widespread recognition with regard to variability in the effects of SNPs across diverse demographies, challenges in clinical translation, and ethical considerations in genetic testing.
Collapse
Affiliation(s)
- Chahat Chopra
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Preeti Rajesh
- Department of Biotechnology, Brainware University, Ramkrishnapur Road, Barasat, Near Jagadighata Market, Kolkata, West Bengal 700125, India
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, Republic of Korea.
| |
Collapse
|
2
|
Santos R, Lee H, Williams A, Baffour-Kyei A, Lee SH, Troakes C, Al-Chalabi A, Breen G, Iacoangeli A. Investigating the Performance of Oxford Nanopore Long-Read Sequencing with Respect to Illumina Microarrays and Short-Read Sequencing. Int J Mol Sci 2025; 26:4492. [PMID: 40429637 PMCID: PMC12111203 DOI: 10.3390/ijms26104492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Oxford Nanopore Technologies (ONT) long-read sequencing (LRS) has emerged as a promising genomic analysis tool, yet comprehensive benchmarks with established platforms across diverse datasets remain limited. This study aimed to benchmark LRS performance against Illumina short-read sequencing (SRS) and microarrays for variant detection across different genomic contexts and to evaluate the impact of experimental factors. We sequenced 14 human genomes using the three platforms and evaluated single nucleotide variants (SNVs), insertions/deletions (indels), and structural variants (SVs) detection, stratifying by high-complexity, low-complexity, and dark genome regions while assessing effects of multiplexing, depth, and read length. LRS SNV accuracy was slightly lower than that of SRS in high-complexity regions (F-measure: 0.954 vs. 0.967) but showed comparable sensitivity in low-complexity regions. LRS showed robust performance for small (1-5 bp) indels in high-complexity regions (F-measure: 0.869), but SRS agreement decreased significantly in low-complexity regions and for larger indel sizes. Within dark regions, LRS identified more indels than SRS, but showed lower base-level accuracy. LRS identified 2.86 times more SVs than SRS, excelling at detecting large variants (>6 kb), with SV detection improving with sequencing depth. Sequencing depth strongly influenced variant calling performance, whereas multiplexing effects were minimal. Our findings provide valuable insights for optimising LRS applications in genomic research and diagnostics.
Collapse
Affiliation(s)
- Renato Santos
- Department of Biostatistics & Health Informatics, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, UK;
| | - Hyunah Lee
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, UK; (H.L.); (A.B.-K.); (S.-H.L.); (G.B.)
| | - Alexander Williams
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, UK; (H.L.); (A.B.-K.); (S.-H.L.); (G.B.)
| | - Anastasia Baffour-Kyei
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, UK; (H.L.); (A.B.-K.); (S.-H.L.); (G.B.)
| | - Sang-Hyuck Lee
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, UK; (H.L.); (A.B.-K.); (S.-H.L.); (G.B.)
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 5 Cutcombe Rd, London SE5 9RX, UK; (C.T.); (A.A.-C.)
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 5 Cutcombe Rd, London SE5 9RX, UK; (C.T.); (A.A.-C.)
| | - Gerome Breen
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, UK; (H.L.); (A.B.-K.); (S.-H.L.); (G.B.)
| | - Alfredo Iacoangeli
- Department of Biostatistics & Health Informatics, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, UK;
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 5 Cutcombe Rd, London SE5 9RX, UK; (C.T.); (A.A.-C.)
- Perron Institute for Neurological and Translational Science, Ground RR Block QE II Medical Centre Ralph & Patricia Sarich Neuroscience Building, 8 Verdun St, Nedlands, WA 6009, Australia
- NIHR Maudsley Biomedical Research Centre (BRC), South London and Maudsley NHS Foundation Trust, 16 De Crespigny Park, London SE5 8AF, UK
| |
Collapse
|
3
|
Zheng W, Ma W, Chen Z, Wang C, Sun T, Dong W, Zhang W, Zhang S, Tang Z, Li K, Zhao Y, Liu Y. DPImpute: A Genotype Imputation Framework for Ultra-Low Coverage Whole-Genome Sequencing and its Application in Genomic Selection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412482. [PMID: 40013759 PMCID: PMC12021046 DOI: 10.1002/advs.202412482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/05/2025] [Indexed: 02/28/2025]
Abstract
Whole-genome sequencing is pivotal for elucidating the complex relationships between genotype and phenotype. However, its widespread application is hindered by the high sequencing depth and large sample sizes required, especially for genomic selection (GS) reliant on precise phenotype prediction from high-density genotype data. To address this, DPImpute (Dual-Phase Impute) is developed, an two-step imputation pipeline enabling accurate whole-genome SNP genotyping under ultra-low coverage whole-genome sequencing (ulcWGS) depths, small testing sample sizes, and limited reference populations. DPImpute achieved 98.06% SNP imputation accuracy with minimal testing samples (≤10), reference samples (≤100), and an ultra-low sequencing depth of 0.3X, surpassing the accuracy of existing imputation methods. Moreover, this high accuracy is maintained across multi-ancestry human populations. Remarkably, DPImpute demonstrated accurate SNP imputation from low-coverage sequencing data from single blood cells and single blastocyst cells, highlighting its potential in embryo GS. To enhance the accessibility of DPImpute, a user-friendly web server (https://agdb.ecenr.com/DPImpute/home) is developed and a Docker container for seamless implementation. In summary, DPImpute can significantly expedite breeding programs through precise and cost-effective genotyping and serve as a valuable tool for diverse population genotyping, encompassing both human and animal studies.
Collapse
Affiliation(s)
- Weigang Zheng
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Wenlong Ma
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Zhilong Chen
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Chao Wang
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Tao Sun
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Wenjun Dong
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Wenjing Zhang
- State Key Laboratory of Swine and Poultry Breeding IndustryNational Engineering Research Center for Breeding Swine IndustryGuangdong Provincial Key Lab of Agro‐Animal Genomics and Molecular BreedingCollege of Animal ScienceSouth China Agricultural UniversityGuangzhou510642China
| | - Song Zhang
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Zhonglin Tang
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Kunpeng Institute of Modern Agriculture at FoshanChinese Academy of Agricultural SciencesFoshan528226China
| | - Kui Li
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Yunxiang Zhao
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and TechnologyGuangxi UniversityNanning530004China
| | - Yuwen Liu
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Kunpeng Institute of Modern Agriculture at FoshanChinese Academy of Agricultural SciencesFoshan528226China
| |
Collapse
|
4
|
Gundappa MK, Robledo D, Hamilton A, Houston RD, Prendergast JGD, Macqueen DJ. High performance imputation of structural and single nucleotide variants using low-coverage whole genome sequencing. Genet Sel Evol 2025; 57:16. [PMID: 40155798 PMCID: PMC11951665 DOI: 10.1186/s12711-025-00962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/01/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Whole genome sequencing (WGS), despite its advantages, is yet to replace methods for genotyping single nucleotide variants (SNVs) such as SNP arrays and targeted genotyping assays. Structural variants (SVs) have larger effects on traits than SNVs, but are more challenging to accurately genotype. Using low-coverage WGS with genotype imputation offers a cost-effective strategy to achieve genome-wide variant coverage, but is yet to be tested for SVs. METHODS Here, we investigate combined SNV and SV imputation with low-coverage WGS data in Atlantic salmon (Salmo salar). As the reference panel, we used genotypes for high-confidence SVs and SNVs for n = 365 wild individuals sampled from diverse populations. We also generated 15 × WGS data (n = 20 samples) for a commercial population external to the reference panel, and called SVs and SNVs with gold-standard approaches. An imputation method selected for its established performance using low-coverage sequencing data (GLIMPSE) was tested at WGS depths of 1 × , 2 × , 3 × , and 4 × for samples within and external to the reference panel. RESULTS SNVs were imputed with high accuracy and recall across all WGS depths, including for samples out-with the reference panel. For SVs, we compared imputation based purely on linkage disequilibrium (LD) with SNVs, to that supplemented with SV genotype likelihoods (GLs) from low-coverage WGS. Including SV GLs increased imputation accuracy, but as a trade-off with recall, requiring 3-4 × depth for best performance. Combining strategies allowed us to capture 84% of the reference panel deletions with 87% accuracy at 1 × depth. We also show that SV length affects imputation performance, with provision of SV GLs greatly enhancing accuracy for the longest SVs in the dataset. CONCLUSIONS This study highlights the promise of reference panel imputation using low-coverage WGS, including novel opportunities to enhance the resolution of genome-wide association studies by capturing SVs.
Collapse
Affiliation(s)
- Manu Kumar Gundappa
- Animal Breeding and Genomics, Wageningen University & Research, P.O. Box 338, 6700 AH, Wageningen, The Netherlands.
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK.
| | - Diego Robledo
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - Alastair Hamilton
- Hendrix Genetics, Villa 'de Körver', Spoorstraat, 695831 CK, Boxmeer, The Netherlands
| | - Ross D Houston
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
- Benchmark Genetics, Pioneer House, Edinburgh Technopole, Penicuik, EH26 0BB, UK
| | - James G D Prendergast
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - Daniel J Macqueen
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| |
Collapse
|
5
|
Celus CS, Ahmad SF, Gangwar M, Kumar S, Kumar A. Deciphering new insights into copy number variations as drivers of genomic diversity and adaptation in farm animal species. Gene 2025; 939:149159. [PMID: 39672215 DOI: 10.1016/j.gene.2024.149159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
The basis of all improvement in (re)production performance of animals and plants lies in the genetic variation. The underlying genetic variation can be further explored through investigations using molecular markers including single nucleotide polymorphism (SNP) and microsatellite, and more recently structural variants like copy number variations (CNVs). Unlike SNPs, CNVs affect a larger proportion of the genome, making them more impactful vis-à-vis variation at the phenotype level. They significantly contribute to genetic variation and provide raw material for natural and artificial selection for improved performance. CNVs are characterized as unbalanced structural variations that arise from four major mechanisms viz., non-homologous end joining (NHEJ), non-allelic homologous recombination (NAHR), fork stalling and template switching (FoSTeS), and retrotransposition. Various detection methods have been developed to identify CNVs, including molecular techniques and massively parallel sequencing. Next-generation sequencing (NGS)/high-throughput sequencing offers higher resolution and sensitivity, but challenges remain in delineating CNVs in regions with repetitive sequences or high GC content. High-throughput sequencing technologies utilize different methods based on read-pair, split-read, read depth, and assembly approaches (or their combination) to detect CNVs. Read-pair based methods work by mapping discordant reads, while the read-depth approach works on detecting the correlation between read depth and copy number of genetic segments or a gene. Split-read methods involve mapping segments of reads to different locations on the genome, while assembly methods involve comparing contigs to a reference or de novo sequencing. Similar to other marker-trait association studies, CNV-association studies are not uncommon in humans and farm animals. Soon, extensive studies will be needed to deduce the unique evolutionary trajectories and underlying molecular mechanisms for targeted genetic improvements in different farm animal species. The present review delineates the importance of CNVs in genetic studies, their generation along with programs and principles to efficiently identify them, and finally throw light on the existing literature on studies in farm animal species vis-à-vis CNVs.
Collapse
Affiliation(s)
- C S Celus
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Sheikh Firdous Ahmad
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India.
| | - Munish Gangwar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Subodh Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Amit Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| |
Collapse
|
6
|
Yang L, Tang C, Cui Y, Zhang J. A High-Throughput Screening Strategy for Bacillus subtilis Producing Menaquinone-7 Based on Fluorescence-Activated Cell Sorting. Microorganisms 2025; 13:536. [PMID: 40142429 PMCID: PMC11946230 DOI: 10.3390/microorganisms13030536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Menaquinone-7 (MK-7) is recognized for its important biological activity, and Bacillus subtilis is the preferred strain for its fermentative production. However, the limited phenotypic diversity among high-yielding strains complicates the development of rapid screening methods. To address this, we utilized the effect of MK-7 on transmembrane potential to develop a high-throughput screening (HTS) strategy for efficiently identifying strains with improved MK-7 production. Among various membrane potential fluorescent dyes tested, Rhodamine 123 was selected for quantifying intracellular MK-7 levels due to its effective staining and minimal impact on cell growth. By optimizing pretreatment protocols and staining conditions, we established an HTS protocol that combines fluorescence-activated cell sorting with HPLC to identify strains with increased MK-7 production. A linear correlation was observed between mean MK-7 content and average fluorescence intensity (R2 = 0.9646). This approach was applied to mutant libraries generated through atmospheric room temperature plasma mutagenesis. After three cycles of mutagenesis and screening, the mutant AR03-27 was identified, showing an 85.65% increase in MK-7 yield compared to the original SJTU2 strain. Resequencing analysis revealed that the top three mutants contained mutations in genes related to membrane transport, suggesting their potential role in enhancing MK-7 yield.
Collapse
Affiliation(s)
- Lina Yang
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
| | - Can Tang
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
| | - Yan Cui
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
| | - Jianhua Zhang
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
- NMPA Key Laboratory for Testing Technology of Pharmaceutical, Shanghai Institute for Food and Drug Control, Shanghai 201203, China
| |
Collapse
|
7
|
Fabiani M, Margiotti K, Libotte F, Alì C, Zangheri C, Barone MA, Viola A, Cima A, Buonomo G, Cristofano D, Lippa A, Pasquale C, Montanino Oliva M, Mesoraca A, Giorlandino C. Uniparental disomy (UPD) exclusion in embryos following Preimplantation Genetic Testing for Structural Rearrangements (PGT-SR). J Assist Reprod Genet 2025; 42:265-273. [PMID: 39693035 PMCID: PMC11805737 DOI: 10.1007/s10815-024-03352-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024] Open
Abstract
PURPOSE Uniparental disomy (UPD) is a genetic condition which both copies of a chromosome are inherited from a single parent, potentially leading to imprinting disorders. This study aimed to assess the integration of Short Tandem Repeat (STR) analysis into Preimplantation Genetic Testing for Structural Rearrangements (PGT-SR) to assess UPD risk and its impact on selecting euploid embryos for embryo transfer in couples with chromosomal translocations involving imprinted chromosomes. METHODS This study evaluated three couples carrying balanced chromosomal translocations: 45,XX,der(13;14)(q10;q10), 46,XX,t(10;11)(q22;q13), and 45,XY,der(14;15)(q10;q10). STR analysis was performed on trophectoderm (TE) biopsies after Whole Genome Amplification (WGA) after PGT-SR analysis using parental blood samples to assess UPD risk in euploid embryos. Haplotyping was conducted with five to six STR markers specific to each rearranged chromosome to detect UPD in euploid embryos. RESULTS Of the four embryos analyzed across the three families, two couples had euploid embryos that tested negative for UPD. These embryos were successfully transferred, resulting in the birth of two healthy children. In the third family, the euploid embryo also tested negative for UPD but failed to implant after transfer, resulting in no pregnancy. DISCUSSION Despite its rarity, UPD involving imprinted chromosomes poses significant clinical risks, as seen in disorders such as Prader-Willi syndrome and Angelman syndrome. This study highlights the importance of integrating UPD screening into PGT-SR protocols, to detect both heterodisomic and isodisomic UPD events minimizing the risk of severe genetic disorders. CONCLUSION Integrating STR-based UPD screening within PGT-SR workflows is a reliable and cost-effective strategy that enhances embryo selection and mitigates the risk of imprinting disorders. This approach improves reproductive outcomes for families with chromosomal rearrangements, offering a practical advancement in assisted reproduction.
Collapse
Affiliation(s)
- Marco Fabiani
- Department of Human Genetic, Altamedica, Rome, Italy.
| | | | | | - Chiara Alì
- Department of Human Genetic, Altamedica, Rome, Italy
| | | | | | | | | | - Giovanna Buonomo
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Danilo Cristofano
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Assunta Lippa
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Chiara Pasquale
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Mario Montanino Oliva
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | | | - Claudio Giorlandino
- Department of Human Genetic, Altamedica, Rome, Italy
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| |
Collapse
|
8
|
Sobral AF, Dinis-Oliveira RJ, Barbosa DJ. CRISPR-Cas technology in forensic investigations: Principles, applications, and ethical considerations. Forensic Sci Int Genet 2025; 74:103163. [PMID: 39437497 DOI: 10.1016/j.fsigen.2024.103163] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
CRISPR-Cas (Clustered Regularly Interspaced Short Palindromic Repeats and CRISPR-associated proteins) systems are adaptive immune systems originally present in bacteria, where they are essential to protect against external genetic elements, including viruses and plasmids. Taking advantage of this system, CRISPR-Cas-based technologies have emerged as incredible tools for precise genome editing, thus significantly advancing several research fields. Forensic sciences represent a multidisciplinary field that explores scientific methods to investigate and resolve legal issues, particularly criminal investigations and subject identification. Consequently, it plays a critical role in the justice system, providing scientific evidence to support judicial investigations. Although less explored, CRISPR-Cas-based methodologies demonstrate strong potential in the field of forensic sciences due to their high accuracy and sensitivity, including DNA profiling and identification, interpretation of crime scene investigations, detection of food contamination or fraud, and other aspects related to environmental forensics. However, using CRISPR-Cas-based methodologies in human samples raises several ethical issues and concerns regarding the potential misuse of individual genetic information. In this manuscript, we provide an overview of potential applications of CRISPR-Cas-based methodologies in several areas of forensic sciences and discuss the legal implications that challenge their routine implementation in this research field.
Collapse
Affiliation(s)
- Ana Filipa Sobral
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra 4585-116, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra 4585-116, Portugal.
| | - Ricardo Jorge Dinis-Oliveira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra 4585-116, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra 4585-116, Portugal; Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Porto 4200-319, Portugal; FOREN - Forensic Science Experts, Dr. Mário Moutinho Avenue, No. 33-A, Lisbon 1400-136, Portugal.
| | - Daniel José Barbosa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra 4585-116, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra 4585-116, Portugal.
| |
Collapse
|
9
|
Hulsegge I, Bouwman AC, Derks MFL. genomeprofile: Unveiling the genomic profile for livestock breeding through comprehensive SNP array-based genotyping. Anim Genet 2024; 55:793-797. [PMID: 39021305 DOI: 10.1111/age.13466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/21/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024]
Abstract
In livestock breeding, single nucleotide polymorphism arrays have become a cornerstone of modern livestock breeding. SNP arrays facilitate the identification of genetic markers linked to economically important traits and provide a powerful tool for predicting breeding values. However, conventional breeding programs often overlook additional genomic features contained in the SNP array data that can provide valuable insights into the genetic diversity, copy number variation, inbreeding levels and potential challenges in breeding lines. Here we present genomeprofile, a tool using SNP array-based genomic data, offering a comprehensive profile of breeding animals including the identification of copy number variants and runs of homozygosity, and screening for aneuploidy. By integrating these features into the breeding landscape, genomeprofile enables a more comprehensive picture of genomic variation, ultimately enhancing precision breeding strategies. To illustrate the practicality and efficacy of genomeprofile, we applied the tool to a dataset of four pig breeding lines. The genomeprofile tool is a user-friendly tool that processes genotype data in finalreport or plink ped format efficiently into useful output. The output contains copy number variations, runs of homozygosity, selection signatures, aneuploidy and inbreeding per individual and across populations. This allows breeding companies and researchers to identify unique individuals or regions in the genome of interest based on routinely collected data.
Collapse
Affiliation(s)
- Ina Hulsegge
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
- Centre for Genetic Resources, The Netherlands, Wageningen University and Research, Wageningen, The Netherlands
| | - Aniek C Bouwman
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Martijn F L Derks
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
10
|
Herrera-Luis E, Martin-Almeida M, Pino-Yanes M. Asthma-Genomic Advances Toward Risk Prediction. Clin Chest Med 2024; 45:599-610. [PMID: 39069324 PMCID: PMC11284279 DOI: 10.1016/j.ccm.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Asthma is a common complex airway disease whose prediction of disease risk and most severe outcomes is crucial in clinical practice for adequate clinical management. This review discusses the latest findings in asthma genomics and current obstacles faced in moving forward to translational medicine. While genome-wide association studies have provided valuable insights into the genetic basis of asthma, there are challenges that must be addressed to improve disease prediction, such as the need for diverse representation, the functional characterization of genetic variants identified, variant selection for genetic testing, and refining prediction models using polygenic risk scores.
Collapse
Affiliation(s)
- Esther Herrera-Luis
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, 615 N Wolfe Street, Baltimore, MD 21205, USA.
| | - Mario Martin-Almeida
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez, s/n. Facultad de Ciencias, San Cristóbal de La Laguna, S/C de Tenerife La Laguna 38200, Tenerife, Spain
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez, s/n. Facultad de Ciencias, San Cristóbal de La Laguna, S/C de Tenerife La Laguna 38200, Tenerife, Spain; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), San Cristóbal de La Laguna 38200, Tenerife, Spain
| |
Collapse
|
11
|
Burridge AJ, Winfield M, Przewieslik‐Allen A, Edwards KJ, Siddique I, Barral‐Arca R, Griffiths S, Cheng S, Huang Z, Feng C, Dreisigacker S, Bentley AR, Brown‐Guedira G, Barker GL. Development of a next generation SNP genotyping array for wheat. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:2235-2247. [PMID: 38520342 PMCID: PMC11258986 DOI: 10.1111/pbi.14341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/25/2024]
Abstract
High-throughput genotyping arrays have provided a cost-effective, reliable and interoperable system for genotyping hexaploid wheat and its relatives. Existing, highly cited arrays including our 35K Wheat Breeder's array and the Illumina 90K array were designed based on a limited amount of varietal sequence diversity and with imperfect knowledge of SNP positions. Recent progress in wheat sequencing has given us access to a vast pool of SNP diversity, whilst technological improvements have allowed us to fit significantly more probes onto a 384-well format Axiom array than previously possible. Here we describe a novel Axiom genotyping array, the 'Triticum aestivum Next Generation' array (TaNG), largely derived from whole genome skim sequencing of 204 elite wheat lines and 111 wheat landraces taken from the Watkins 'Core Collection'. We used a novel haplotype optimization approach to select SNPs with the highest combined varietal discrimination and a design iteration step to test and replace SNPs which failed to convert to reliable markers. The final design with 43 372 SNPs contains a combination of haplotype-optimized novel SNPs and legacy cross-platform markers. We show that this design has an improved distribution of SNPs compared to previous arrays and can be used to generate genetic maps with a significantly higher number of distinct bins than our previous array. We also demonstrate the improved performance of TaNGv1.1 for Genome-wide association studies (GWAS) and its utility for Copy Number Variation (CNV) analysis. The array is commercially available with supporting marker annotations and initial genotyping results freely available.
Collapse
Affiliation(s)
| | - Mark Winfield
- School of Biological SciencesUniversity of BristolBristolUK
| | | | | | - Imteaz Siddique
- Thermo Fisher Scientific3450 Central ExpresswaySanta ClaraCAUSA
| | | | | | - Shifeng Cheng
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Zejian Huang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Cong Feng
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | | | | | - Gina Brown‐Guedira
- Plant Science Research UnitUSDA Agricultural Research ServiceRaleighNCUSA
| | - Gary L. Barker
- School of Biological SciencesUniversity of BristolBristolUK
| |
Collapse
|
12
|
Incze E, Mangó K, Fekete F, Kiss ÁF, Póti Á, Harkó T, Moldvay J, Szüts D, Monostory K. Potential Association of Cytochrome P450 Copy Number Alteration in Tumour with Chemotherapy Resistance in Lung Adenocarcinoma Patients. Int J Mol Sci 2023; 24:13380. [PMID: 37686184 PMCID: PMC10487787 DOI: 10.3390/ijms241713380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Resistance to anticancer agents is a major obstacle to efficacious tumour therapy and responsible for high cancer-related mortality rates. Some resistance mechanisms are associated with pharmacokinetic variability in anticancer drug exposure due to genetic polymorphisms of drug-metabolizing cytochrome P450 (CYP) enzymes, whereas variations in tumoural metabolism as a consequence of CYP copy number alterations are assumed to contribute to the selection of resistant cells. A high-throughput quantitative polymerase chain reaction (qPCR)-based method was developed for detection of CYP copy number alterations in tumours, and a scoring system improved the identification of inappropriate reference genes that underwent deletion/multiplication in tumours. The copy numbers of both the target (CYP2C8, CYP3A4) and the reference genes (ALB, B2M, BCKDHA, F5, CD36, MPO, TBP, RPPH1) established in primary lung adenocarcinoma by the qPCR-based method were congruent with those determined by next-generation sequencing (for 10 genes, slope = 0.9498, r2 = 0.72). In treatment naïve adenocarcinoma samples, the copy number multiplication of paclitaxel-metabolizing CYP2C8 and/or CYP3A4 was more prevalent in non-responder patients with progressive disease/exit than in responders with complete remission. The high-throughput qPCR-based method can become an alternative approach to next-generation sequencing in routine clinical practice, and identification of altered CYP copy numbers may provide a promising biomarker for therapy-resistant tumours.
Collapse
Affiliation(s)
- Evelyn Incze
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok 2, H-1117 Budapest, Hungary; (E.I.); (K.M.); (F.F.); (Á.F.K.); (Á.P.); (D.S.)
- Doctoral School of Pharmaceutical Sciences, Semmelweis University, Üllői 26, H-1085 Budapest, Hungary
| | - Katalin Mangó
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok 2, H-1117 Budapest, Hungary; (E.I.); (K.M.); (F.F.); (Á.F.K.); (Á.P.); (D.S.)
- Doctoral School of Pharmaceutical Sciences, Semmelweis University, Üllői 26, H-1085 Budapest, Hungary
| | - Ferenc Fekete
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok 2, H-1117 Budapest, Hungary; (E.I.); (K.M.); (F.F.); (Á.F.K.); (Á.P.); (D.S.)
| | - Ádám Ferenc Kiss
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok 2, H-1117 Budapest, Hungary; (E.I.); (K.M.); (F.F.); (Á.F.K.); (Á.P.); (D.S.)
| | - Ádám Póti
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok 2, H-1117 Budapest, Hungary; (E.I.); (K.M.); (F.F.); (Á.F.K.); (Á.P.); (D.S.)
| | - Tünde Harkó
- Department of Pathology, National Korányi Institute of Pulmonology, Pihenő 1, H-1121 Budapest, Hungary;
| | - Judit Moldvay
- 1st Department of Pulmonology, National Korányi Institute of Pulmonology, Pihenő 1, H-1121 Budapest, Hungary;
| | - Dávid Szüts
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok 2, H-1117 Budapest, Hungary; (E.I.); (K.M.); (F.F.); (Á.F.K.); (Á.P.); (D.S.)
| | - Katalin Monostory
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok 2, H-1117 Budapest, Hungary; (E.I.); (K.M.); (F.F.); (Á.F.K.); (Á.P.); (D.S.)
| |
Collapse
|
13
|
Ortiz M, Jauset-Rubio M, Trummer O, Foessl I, Kodr D, Acero JL, Botero ML, Biggs P, Lenartowicz D, Trajanoska K, Rivadeneira F, Hocek M, Obermayer-Pietsch B, O’Sullivan CK. Generic Platform for the Multiplexed Targeted Electrochemical Detection of Osteoporosis-Associated Single Nucleotide Polymorphisms Using Recombinase Polymerase Solid-Phase Primer Elongation and Ferrocene-Modified Nucleoside Triphosphates. ACS CENTRAL SCIENCE 2023; 9:1591-1602. [PMID: 37637735 PMCID: PMC10450878 DOI: 10.1021/acscentsci.3c00243] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Indexed: 08/29/2023]
Abstract
Osteoporosis is a multifactorial disease influenced by genetic and environmental factors, which contributes to an increased risk of bone fracture, but early diagnosis of this disease cannot be achieved using current techniques. We describe a generic platform for the targeted electrochemical genotyping of SNPs identified by genome-wide association studies to be associated with a genetic predisposition to osteoporosis. The platform exploits isothermal solid-phase primer elongation with ferrocene-labeled nucleoside triphosphates. Thiolated reverse primers designed for each SNP were immobilized on individual gold electrodes of an array. These primers are designed to hybridize to the SNP site at their 3'OH terminal, and primer elongation occurs only where there is 100% complementarity, facilitating the identification and heterozygosity of each SNP under interrogation. The platform was applied to real blood samples, which were thermally lysed and directly used without the need for DNA extraction or purification. The results were validated using Taqman SNP genotyping assays and Sanger sequencing. The assay is complete in just 15 min with a total cost of 0.3€ per electrode. The platform is completely generic and has immense potential for deployment at the point of need in an automated device for targeted SNP genotyping with the only required end-user intervention being sample addition.
Collapse
Affiliation(s)
- Mayreli Ortiz
- INTERFIBIO
Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Miriam Jauset-Rubio
- INTERFIBIO
Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Olivia Trummer
- Division
of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Ines Foessl
- Division
of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - David Kodr
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo namesti 2, CZ 16610 Prague 6, Czech Republic
| | - Josep Lluís Acero
- INTERFIBIO
Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Mary Luz Botero
- INTERFIBIO
Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Phil Biggs
- Labman
Automation
Ltd., Seamer Hill, Stokesley, North Yorkshire, TS9 5NQ U.K.
| | - Daniel Lenartowicz
- Labman
Automation
Ltd., Seamer Hill, Stokesley, North Yorkshire, TS9 5NQ U.K.
| | - Katerina Trajanoska
- Department
of Internal Medicine, Erasmus MC, 40 3015 Rotterdam, The Netherlands
| | | | - Michal Hocek
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo namesti 2, CZ 16610 Prague 6, Czech Republic
- Department
of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, CZ-12843 Prague 2, Czech Republic
| | - Barbara Obermayer-Pietsch
- Division
of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Ciara K. O’Sullivan
- INTERFIBIO
Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Institució
Catalana de Recerca i Estudis Avancats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
14
|
Abondio P, Cilli E, Luiselli D. Human Pangenomics: Promises and Challenges of a Distributed Genomic Reference. Life (Basel) 2023; 13:1360. [PMID: 37374141 DOI: 10.3390/life13061360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
A pangenome is a collection of the common and unique genomes that are present in a given species. It combines the genetic information of all the genomes sampled, resulting in a large and diverse range of genetic material. Pangenomic analysis offers several advantages compared to traditional genomic research. For example, a pangenome is not bound by the physical constraints of a single genome, so it can capture more genetic variability. Thanks to the introduction of the concept of pangenome, it is possible to use exceedingly detailed sequence data to study the evolutionary history of two different species, or how populations within a species differ genetically. In the wake of the Human Pangenome Project, this review aims at discussing the advantages of the pangenome around human genetic variation, which are then framed around how pangenomic data can inform population genetics, phylogenetics, and public health policy by providing insights into the genetic basis of diseases or determining personalized treatments, targeting the specific genetic profile of an individual. Moreover, technical limitations, ethical concerns, and legal considerations are discussed.
Collapse
Affiliation(s)
- Paolo Abondio
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy
| | - Elisabetta Cilli
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy
| | - Donata Luiselli
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy
| |
Collapse
|
15
|
Iourov IY, Gerasimov AP, Zelenova MA, Ivanova NE, Kurinnaia OS, Zabrodskaya YM, Demidova IA, Barantsevich ER, Vasin KS, Kolotii AD, Ushanov VV, Sitovskaya DA, Lobzhanidze TBA, Iuditskaia ME, Iakushev NS, Zhumatov MM, Vorsanova SG, Samochernyh KA. Cytogenomic epileptology. Mol Cytogenet 2023; 16:1. [PMID: 36600272 PMCID: PMC9814426 DOI: 10.1186/s13039-022-00634-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
Molecular cytogenetic and cytogenomic studies have made a contribution to genetics of epilepsy. However, current genomic research of this devastative condition is generally focused on the molecular genetic aspects (i.e. gene hunting, detecting mutations in known epilepsy-associated genes, searching monogenic causes of epilepsy). Nonetheless, chromosomal abnormalities and copy number variants (CNVs) represent an important part of genetic defects causing epilepsy. Moreover, somatic chromosomal mosaicism and genome/chromosome instability seem to be a possible mechanism for a wide spectrum of epileptic conditions. This idea becomes even more attracting taking into account the potential of molecular neurocytogenetic (neurocytogenomic) studies of the epileptic brain. Unfortunately, analyses of chromosome numbers and structure in the affected brain or epileptogenic brain foci are rarely performed. Therefore, one may conclude that cytogenomic area of genomic epileptology is poorly researched. Accordingly, molecular cytogenetic and cytogenomic studies of the clinical cohorts and molecular neurocytogenetic analyses of the epileptic brain appear to be required. Here, we have performed a theoretical analysis to define the targets of the aforementioned studies and to highlight future directions for molecular cytogenetic and cytogenomic research of epileptic disorders in the widest sense. To succeed, we have formed a consortium, which is planned to perform at least a part of suggested research. Taking into account the nature of the communication, "cytogenomic epileptology" has been introduced to cover the research efforts in this field of medical genomics and epileptology. Additionally, initial results of studying cytogenomic variations in the Russian neurodevelopmental cohort are reviewed with special attention to epilepsy. In total, we have concluded that (i) epilepsy-associated cytogenomic variations require more profound research; (ii) ontological analyses of epilepsy genes affected by chromosomal rearrangements and/or CNVs with unraveling pathways implicating epilepsy-associated genes are beneficial for epileptology; (iii) molecular neurocytogenetic (neurocytogenomic) analysis of postoperative samples are warranted in patients suffering from epileptic disorders.
Collapse
Affiliation(s)
- Ivan Y Iourov
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia.
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia.
- Department of Medical Biological Disciplines, Belgorod State University, Belgorod, Russia.
| | - Alexandr P Gerasimov
- Research Laboratory of Pediatric Neurosurgery, Polenov Neurosurgical Institute, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Maria A Zelenova
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Natalya E Ivanova
- Scientific Department of Polenov Neurosurgical Institute, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Oksana S Kurinnaia
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Yulia M Zabrodskaya
- Research Laboratory of Pathomorphology of the Nervous System, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Irina A Demidova
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Evgeny R Barantsevich
- Postgraduate Neurology and Manual Medicine Department, Pavlov First Saint-Petersburg State Medical University, Saint Petersburg, Russia
| | - Kirill S Vasin
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Alexey D Kolotii
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Vseslav V Ushanov
- Department of Neurosurgery, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Darya A Sitovskaya
- Research Laboratory of Pathomorphology of the Nervous System, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | | | - Maria E Iuditskaia
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Nikita S Iakushev
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Muslim M Zhumatov
- Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russia
| | - Svetlana G Vorsanova
- Yurov's Laboratory of Molecular Genetics and Cytogenomics of the Brain, Mental Health Research Center, Moscow, Russia
- Vorsanova's Laboratory of Molecular Cytogenetics of Neuropsychiatric Diseases, Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Konstantin A Samochernyh
- Polenov Neurosurgical Institute, Almazov National Medical Research Centre, Saint Petersburg, Russia
| |
Collapse
|
16
|
Maihofer AX, Engchuan W, Huguet G, Klein M, MacDonald JR, Shanta O, Thiruvahindrapuram B, Jean-Louis M, Saci Z, Jacquemont S, Scherer SW, Ketema E, Aiello AE, Amstadter AB, Avdibegović E, Babic D, Baker DG, Bisson JI, Boks MP, Bolger EA, Bryant RA, Bustamante AC, Caldas-de-Almeida JM, Cardoso G, Deckert J, Delahanty DL, Domschke K, Dunlop BW, Dzubur-Kulenovic A, Evans A, Feeny NC, Franz CE, Gautam A, Geuze E, Goci A, Hammamieh R, Jakovljevic M, Jett M, Jones I, Kaufman ML, Kessler RC, King AP, Kremen WS, Lawford BR, Lebois LAM, Lewis C, Liberzon I, Linnstaedt SD, Lugonja B, Luykx JJ, Lyons MJ, Mavissakalian MR, McLaughlin KA, McLean SA, Mehta D, Mellor R, Morris CP, Muhie S, Orcutt HK, Peverill M, Ratanatharathorn A, Risbrough VB, Rizzo A, Roberts AL, Rothbaum AO, Rothbaum BO, Roy-Byrne P, Ruggiero KJ, Rutten BPF, Schijven D, Seng JS, Sheerin CM, Sorenson MA, Teicher MH, Uddin M, Ursano RJ, Vinkers CH, Voisey J, Weber H, Winternitz S, Xavier M, Yang R, McD Young R, Zoellner LA, Salem RM, Shaffer RA, Wu T, Ressler KJ, Stein MB, Koenen KC, Sebat J, Nievergelt CM. Rare copy number variation in posttraumatic stress disorder. Mol Psychiatry 2022; 27:5062-5069. [PMID: 36131047 PMCID: PMC9763110 DOI: 10.1038/s41380-022-01776-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 01/27/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a heritable (h2 = 24-71%) psychiatric illness. Copy number variation (CNV) is a form of rare genetic variation that has been implicated in the etiology of psychiatric disorders, but no large-scale investigation of CNV in PTSD has been performed. We present an association study of CNV burden and PTSD symptoms in a sample of 114,383 participants (13,036 cases and 101,347 controls) of European ancestry. CNVs were called using two calling algorithms and intersected to a consensus set. Quality control was performed to remove strong outlier samples. CNVs were examined for association with PTSD within each cohort using linear or logistic regression analysis adjusted for population structure and CNV quality metrics, then inverse variance weighted meta-analyzed across cohorts. We examined the genome-wide total span of CNVs, enrichment of CNVs within specified gene-sets, and CNVs overlapping individual genes and implicated neurodevelopmental regions. The total distance covered by deletions crossing over known neurodevelopmental CNV regions was significant (beta = 0.029, SE = 0.005, P = 6.3 × 10-8). The genome-wide neurodevelopmental CNV burden identified explains 0.034% of the variation in PTSD symptoms. The 15q11.2 BP1-BP2 microdeletion region was significantly associated with PTSD (beta = 0.0206, SE = 0.0056, P = 0.0002). No individual significant genes interrupted by CNV were identified. 22 gene pathways related to the function of the nervous system and brain were significant in pathway analysis (FDR q < 0.05), but these associations were not significant once NDD regions were removed. A larger sample size, better detection methods, and annotated resources of CNV are needed to explore this relationship further.
Collapse
Affiliation(s)
- Adam X Maihofer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, USA.
- Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA.
| | - Worrawat Engchuan
- The Hospital for Sick Children, Genetics and Genome Biology, Toronto, Ontario, Canada
- The Hospital for Sick Children, The Centre for Applied Genomics, Toronto, Ontario, Canada
| | - Guillaume Huguet
- Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montreal, Quebec, Canada
| | - Marieke Klein
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jeffrey R MacDonald
- The Hospital for Sick Children, Genetics and Genome Biology, Toronto, Ontario, Canada
| | - Omar Shanta
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, USA
| | | | - Martineau Jean-Louis
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montreal, Quebec, Canada
| | - Zohra Saci
- Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montreal, Quebec, Canada
| | - Sebastien Jacquemont
- Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montreal, Quebec, Canada
- Department of Genetics, Centre Hospitalier Universitaire Vaudois, Lausanne, Vaud, Switzerland
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Stephen W Scherer
- The Hospital for Sick Children, Genetics and Genome Biology, Toronto, Ontario, Canada
- University of Toronto, McLaughlin Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth Ketema
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Allison E Aiello
- Department of Epidemiology, Robert N Butler Columbia Aging Center, Columbia University, New York, NY, USA
| | - Ananda B Amstadter
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Richmond, VA, USA
| | - Esmina Avdibegović
- Department of Psychiatry, University Clinical Center of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Dragan Babic
- Department of Psychiatry, University Clinical Center of Mostar, Mostar, Bosnia and Herzegovina
| | - Dewleen G Baker
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Jonathan I Bisson
- MRC Centre for Psychiatric Genetics and Genomics, Cardiff University, National Centre for Mental Health, Cardiff, South Glamorgan, UK
| | - Marco P Boks
- Department of Psychiatry, UMC Utrecht Brain Center, Utrecht, the Netherlands
| | - Elizabeth A Bolger
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Richard A Bryant
- Department of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - Angela C Bustamante
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Graça Cardoso
- Lisbon Institute of Global Mental Health and Comprehensive Health Research Centre, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Jurgen Deckert
- University Hospital of Wuerzburg, Center of Mental Health, Psychiatry, Psychosomatics and Psychotherapy, Wuerzburg, Germany
| | - Douglas L Delahanty
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
- Research and Sponsored Programs, Kent State University, Kent, OH, USA
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Faculty of Medicine, Centre for Basics in Neuromodulation, University of Freiburg, Freiburg, Germany
| | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Alma Dzubur-Kulenovic
- Department of Psychiatry, University Clinical Center of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alexandra Evans
- MRC Centre for Psychiatric Genetics and Genomics, Cardiff University, National Centre for Mental Health, Cardiff, South Glamorgan, UK
| | - Norah C Feeny
- Department of Psychological Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Aarti Gautam
- Walter Reed Army Institute of Research, Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Silver Spring, MD, USA
| | - Elbert Geuze
- Netherlands Ministry of Defence, Brain Research and Innovation Centre, Utrecht, the Netherlands
- Department of Psychiatry, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, the Netherlands
| | - Aferdita Goci
- Department of Psychiatry, University Clinical Centre of Kosovo, Prishtina, Kosovo
| | - Rasha Hammamieh
- Walter Reed Army Institute of Research, Medical Readiness Systems Biology, Center for Military Psychiatry and Neuroscience, Silver Spring, MD, USA
| | - Miro Jakovljevic
- Department of Psychiatry, University Hospital Center of Zagreb, Zagreb, Croatia
| | - Marti Jett
- US Medical Research & Development Comm, Fort Detrick, MD, USA
- Walter Reed Army Institute of Research, Headquarter, Silver Spring, MD, USA
| | - Ian Jones
- MRC Centre for Psychiatric Genetics and Genomics, Cardiff University, National Centre for Mental Health, Cardiff, South Glamorgan, UK
| | - Milissa L Kaufman
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | - Anthony P King
- Ohio State University, College of Medicine, Institute for Behavioral Medicine Research, Columbus, OH, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Bruce R Lawford
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Lauren A M Lebois
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Catrin Lewis
- MRC Centre for Psychiatric Genetics and Genomics, Cardiff University, National Centre for Mental Health, Cardiff, South Glamorgan, UK
| | - Israel Liberzon
- Department of Psychiatry and Behavioral Sciences, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Sarah D Linnstaedt
- Institute for Trauma Recovery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bozo Lugonja
- MRC Centre for Psychiatric Genetics and Genomics, Cardiff University, National Centre for Mental Health, Cardiff, South Glamorgan, UK
| | - Jurjen J Luykx
- Department of Psychiatry, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, the Netherlands
- Department of Translational Neuroscience, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, the Netherlands
| | - Michael J Lyons
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | | | | | - Samuel A McLean
- Institute for Trauma Recovery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Emergency Medicine, UNC Institute for Trauma Recovery, Chapel Hill, NC, USA
| | - Divya Mehta
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD, Australia
- Queensland University of Technology, Centre for Genomics and Personalised Health, Kelvin Grove, QLD, Australia
| | - Rebecca Mellor
- Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Greenslopes, QLD, Australia
| | - Charles Phillip Morris
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Seid Muhie
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Holly K Orcutt
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Matthew Peverill
- Department of Psychology, University of Washington, Seattle, WA, USA
| | - Andrew Ratanatharathorn
- Department of Epidemiology, Columbia University Mailmain School of Public Health, New York, NY, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Albert Rizzo
- University of Southern California, Institute for Creative Technologies, Los Angeles, CA, USA
| | - Andrea L Roberts
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alex O Rothbaum
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Barbara O Rothbaum
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Peter Roy-Byrne
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Kenneth J Ruggiero
- Department of Nursing and Department of Psychiatry, Medical University of South Carolina, Charleston, SC, USA
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, Maastricht Universitair Medisch Centrum, School for Mental Health and Neuroscience, Maastricht, Limburg, the Netherlands
| | - Dick Schijven
- Department of Psychiatry, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, the Netherlands
- Department of Translational Neuroscience, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, the Netherlands
| | - Julia S Seng
- University of Michigan, School of Nursing, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Women's and Gender Studies, University of Michigan, Ann Arbor, MI, USA
- University of Michigan, Institute for Research on Women and Gender, Ann Arbor, MI, USA
| | - Christina M Sheerin
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Richmond, VA, USA
| | - Michael A Sorenson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Martin H Teicher
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Monica Uddin
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Robert J Ursano
- Department of Psychiatry, Uniformed Services University, Bethesda, MD, USA
| | - Christiaan H Vinkers
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, the Netherlands
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Joanne Voisey
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD, Australia
- Queensland University of Technology, Centre for Genomics and Personalised Health, Kelvin Grove, QLD, Australia
| | - Heike Weber
- University Hospital of Wuerzburg, Center of Mental Health, Psychiatry, Psychosomatics and Psychotherapy, Wuerzburg, Germany
| | - Sherry Winternitz
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Miguel Xavier
- Universidade Nova de Lisboa, Nova Medical School, Lisboa, Portugal
| | - Ruoting Yang
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Ross McD Young
- Queensland University of Technology, School of Clinical Sciences, Kelvin Grove, QLD, Australia
- University of the Sunshine Coast, The Chancellory, Sippy Downs, QLD, Australia
| | - Lori A Zoellner
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Rany M Salem
- University of California San Diego, Herbert Wertheim School of Public Health and Human Longevity Science, La Jolla, CA, USA
| | - Richard A Shaffer
- Department of Epidemiology and Health Sciences, Naval Health Research Center, San Diego, CA, USA
| | - Tianying Wu
- Division of Epidemiology and Biostatistics, San Diego State University, School of Public Health, San Diego, CA, USA
- University of California, San Diego, Moores Cancer Center, San Diego, CA, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Murray B Stein
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- University of California San Diego, School of Public Health, La Jolla, CA, USA
| | - Karestan C Koenen
- Broad Institute of MIT and Harvard, Stanley Center for Psychiatric Research, Cambridge, MA, USA
- Department of Epidemiology, Harvard T. H. School of Public Health, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan Sebat
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, Center of Excellence for Stress and Mental Health, San Diego, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
17
|
Integrated Analysis of Tumor Mutation Burden and Immune Infiltrates in Hepatocellular Carcinoma. Diagnostics (Basel) 2022; 12:diagnostics12081918. [PMID: 36010268 PMCID: PMC9406847 DOI: 10.3390/diagnostics12081918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Tumor mutation burdens (TMBs) act as an indicator of immunotherapeutic responsiveness in various tumors. However, the relationship between TMBs and immune cell infiltrates in hepatocellular carcinoma (HCC) is still obscure. The present study aimed to explore the potential diagnostic markers of TMBs for HCC and analyze the role of immune cell infiltration in this pathology. We used OA datasets from The Cancer Genome Atlas database. First, the “maftools” package was used to screen the highest mutation frequency in all samples. R software was used to identify differentially expressed genes (DEGs) according to mutation frequency and perform functional correlation analysis. Then, the gene ontology (GO) enrichment analysis was performed with “clusterProfiler”, “enrichplot”, and “ggplot2” packages. Finally, the correlations between diagnostic markers and infiltrating immune cells were analyzed, and CIBERSORT was used to evaluate the infiltration of immune cells in HCC tissues. As a result, we identified a total of 359 DEGs in this study. These DEGs may affect HCC prognosis by regulating fatty acid metabolism, hypoxia, and the P53 pathway. The top 15 genes were selected as the hub genes through PPI network analysis. SRSF1, SNRPA1, and SRSF3 showed strong similarities in biological effects, NCBP2 was demonstrated as a diagnostic marker of HCC, and high NCBP2 expression was significantly correlated with poor over survival (OS) in HCC. In addition, NCBP2 expression was correlated with the infiltration of B cells (r = 0.364, p = 3.30 × 10−12), CD8+ T cells (r = 0.295, p = 2.71 × 10−8), CD4+ T cells, (r = 0.484, p = 1.37 × 10−21), macrophages (r = 0.551, p = 1.97 × 10−28), neutrophils (r = 0.457, p = 3.26 × 10−19), and dendritic cells (r = 0.453, p = 1.97 × 10−18). Immune cell infiltration analysis revealed that the degree of central memory T-cell (Tcm) infiltration may be correlated with the HCC process. In conclusion, NCBP2 can be used as diagnostic markers of HCC, and immune cell infiltration plays an important role in the occurrence and progression of HCC.
Collapse
|