1
|
Zhao F, Li Y, Chen L, Yao B. Identification of brain region-specific landscape and functions of clustered circRNAs in Alzheimer's disease using circMeta2. Commun Biol 2024; 7:1353. [PMID: 39427093 PMCID: PMC11490488 DOI: 10.1038/s42003-024-07060-1] [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: 03/21/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder with regulatory RNAs playing significant roles in its etiology. Circular RNAs (CircRNA) are enriched in human brains and contribute to AD progression. Many circRNA isoforms derived from same gene loci share common back splicing sites, thus often form clusters and work as a group to additively regulate their downstream targets. Unfortunately, the coordinated role of clustered circRNAs is often overlooked in individual circRNA differential expression (DE) analysis. To address these challenges, we develop circMeta2, a computational tool designed to perform DE analysis focused on circRNA clusters, equipped with modules tailored for both a small sample of biological replicates and a large-scale population study. Using circMeta2, we identify brain region-specific circRNA clusters from six distinct brain regions in the ENCODE datasets, as well as brain region-specific alteration of circRNA clusters signatures associated with AD from Mount Sinai brain bank (MSBB) AD study. We also illustrate how AD-associated circRNA clusters within and across different brain regions work coordinately to contribute to AD etiology by impacting miRNA-mediated gene expression and identified key circRNA clusters that associated with AD progression and severity. Our study demonstrates circMeta2 as a highly accuracy and robust tool for analyzing circRNA clusters, offering valuable molecular insights into AD pathology.
Collapse
Affiliation(s)
- Fengdi Zhao
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, FL, USA.
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
2
|
Willis C, White JD, Minto MS, Quach BC, Han S, Tao R, Shin JH, Deep-Soboslay A, Hyde TM, Mayfield RD, Webb BT, Johnson EO, Kleinman JE, Bierut LJ, Hancock DB. Gene expression differences associated with alcohol use disorder in human brain. Mol Psychiatry 2024:10.1038/s41380-024-02777-1. [PMID: 39394458 DOI: 10.1038/s41380-024-02777-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Abstract
Excessive alcohol consumption is a leading cause of preventable death worldwide. To improve understanding of neurobiological mechanisms associated with alcohol use disorder (AUD) in humans, we compared gene expression data from deceased individuals with and without AUD across two addiction-relevant brain regions: the nucleus accumbens (NAc) and dorsolateral prefrontal cortex (DLPFC). Bulk RNA-seq data from NAc and DLPFC (N ≥50 with AUD, ≥46 non-AUD) were analyzed for differential gene expression using modified negative binomial regression adjusting for technical and biological covariates. The region-level results were meta-analyzed with those from an independent dataset (NNAc = 28 AUD, 29 non-AUD; NPFC = 66 AUD, 77 non-AUD). We further tested for heritability enrichment of AUD-related phenotypes, gene co-expression networks, gene ontology enrichment, and drug repurposing. We identified 176 differentially expressed genes (DEGs; 12 in both regions, 78 in NAc only, 86 in DLPFC only) for AUD in our new dataset. After meta-analyzing with published data, we identified 476 AUD DEGs (25 in both regions, 29 in NAc only, 422 in PFC only). Of these DEGs, 17 were significant when looked up in GWAS of problematic alcohol use or drinks per week. Gene co-expression analysis showed both concordant and unique gene networks across brain regions. We also identified 29 and 436 drug compounds that target DEGs from our meta-analysis in NAc and PFC, respectively. This study identified robust AUD-associated DEGs, contributing novel neurobiological insights into AUD and highlighting genes targeted by known drug compounds, generating opportunity for drug repurposing to treat AUD.
Collapse
Affiliation(s)
- Caryn Willis
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA.
| | - Julie D White
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Melyssa S Minto
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Bryan C Quach
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Shizhong Han
- Lieber Institute for Brain Development (LIBD), Baltimore, MD, USA
| | - Ran Tao
- Lieber Institute for Brain Development (LIBD), Baltimore, MD, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development (LIBD), Baltimore, MD, USA
| | | | - Thomas M Hyde
- Lieber Institute for Brain Development (LIBD), Baltimore, MD, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA
| | - Bradley T Webb
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Eric O Johnson
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
- Fellow Program, RTI International, Research Triangle Park, NC, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development (LIBD), Baltimore, MD, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Dana B Hancock
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
3
|
Kamińska K, Świderska B, Malinowska A, Grzesiak M. Tandem mass tag-based proteomic analysis of granulosa and theca interna cells of the porcine ovarian follicle following in vitro treatment with vitamin D 3 and insulin alone or in combination. J Proteomics 2024; 310:105318. [PMID: 39284438 DOI: 10.1016/j.jprot.2024.105318] [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: 06/19/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
This study was performed to investigate the proteomic basis underlying the interaction between vitamin D3 (VD) and insulin (I) within ovarian follicle using the pig as a model. Porcine antral follicles were incubated in vitro for 12 h with VD alone and I alone or in combination (VD + I) or with no treatment as the control (C). In total, 7690 and 7467 proteins were identified in the granulosa and theca interna compartments, respectively. Comparative proteomic analysis revealed 97 differentially abundant proteins (DAPs) within the granulosa layer and 11 DAPs within the theca interna layer. In the granulosa compartment, VD affected proteome leading to the promotion of cell proliferation, whereas I influenced mainly proteins related to cellular adhesion. The VD + I treatment induced granulosa cell proliferation probably via the DAPs involved in DNA synthesis and the cell cycle regulation. In the theca interna layer, VD alone or in co-treatment with I affected DAPs associated with cholesterol transport and lipid and steroid metabolic processes that was further confirmed by diminished lipid droplet accumulation. SIGNIFICANCE: The application of quantitative proteomics demonstrated for the first time the complexity of VD and I interactions in porcine ovarian follicle, providing a framework for understanding the molecular mechanisms underlying their cross-talk. Although identified DAPs were related to crucial ovarian processes, including the granulosa cell proliferation and cholesterol transport in the theca interna layer, novel molecular pathways underlying these processes have been proposed. The identified unique proteins may serve as indicators of VD and I interactions in both follicle layers, and could be useful biomarkers of ovarian pathologies characterized by impaired VD and I levels, such as polycystic ovary syndrome.
Collapse
Affiliation(s)
- Kinga Kamińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Bianka Świderska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Agata Malinowska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Małgorzata Grzesiak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland.
| |
Collapse
|
4
|
Puerta R, de Rojas I, García-González P, Olivé C, Sotolongo-Grau O, García-Sánchez A, García-Gutiérrez F, Montrreal L, Pablo Tartari J, Sanabria Á, Pytel V, Lage C, Quintela I, Aguilera N, Rodriguez-Rodriguez E, Alarcón-Martín E, Orellana A, Pastor P, Pérez-Tur J, Piñol-Ripoll G, de Munian AL, García-Alberca JM, Royo JL, Bullido MJ, Álvarez V, Real LM, Anchuelo AC, Gómez-Garre D, Larrad MTM, Franco-Macías E, Mir P, Medina M, Sánchez-Valle R, Dols-Icardo O, Sáez ME, Carracedo Á, Tárraga L, Alegret M, Valero S, Marquié M, Boada M, Juan PS, Cavazos JE, Cabrera A, Cano A. Connecting genomic and proteomic signatures of amyloid burden in the brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313124. [PMID: 39281766 PMCID: PMC11398581 DOI: 10.1101/2024.09.06.24313124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Alzheimer's disease (AD) has a high heritable component characteristic of complex diseases, yet many of the genetic risk factors remain unknown. We combined genome-wide association studies (GWAS) on amyloid endophenotypes measured in cerebrospinal fluid (CSF) and positron emission tomography (PET) as surrogates of amyloid pathology, which may be helpful to understand the underlying biology of the disease. Methods We performed a meta-analysis of GWAS of CSF Aβ42 and PET measures combining six independent cohorts (n=2,076). Due to the opposite effect direction of Aβ phenotypes in CSF and PET measures, only genetic signals in the opposite direction were considered for analysis (n=376,599). Polygenic risk scores (PRS) were calculated and evaluated for AD status and amyloid endophenotypes. We then searched the CSF proteome signature of brain amyloidosis using SOMAscan proteomic data (Ace cohort, n=1,008) and connected it with GWAS results of loci modulating amyloidosis. Finally, we compared our results with a large meta-analysis using publicly available datasets in CSF (n=13,409) and PET (n=13,116). This combined approach enabled the identification of overlapping genes and proteins associated with amyloid burden and the assessment of their biological significance using enrichment analyses. Results After filtering the meta-GWAS, we observed genome-wide significance in the rs429358-APOE locus and nine suggestive hits were annotated. We replicated the APOE loci using the large CSF-PET meta-GWAS and identified multiple AD-associated genes as well as the novel GADL1 locus. Additionally, we found a significant association between the AD PRS and amyloid levels, whereas no significant association was found between any Aβ PRS with AD risk. CSF SOMAscan analysis identified 1,387 FDR-significant proteins associated with CSF Aβ42 levels. The overlap among GWAS loci and proteins associated with amyloid burden was very poor (n=35). The enrichment analysis of overlapping hits strongly suggested several signalling pathways connecting amyloidosis with the anchored component of the plasma membrane, synapse physiology and mental disorders that were replicated in the large CSF-PET meta-analysis. Conclusions The strategy of combining CSF and PET amyloid endophenotypes GWAS with CSF proteome analyses might be effective for identifying signals associated with the AD pathological process and elucidate causative molecular mechanisms behind the amyloid mobilization in AD.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- Universitat de Barcelona (UB)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | | | | | | | - Laura Montrreal
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nuria Aguilera
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | | | - Adelina Orellana
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
- The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Jordi Pérez-Tur
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unitat de Genètica Molecular, Institut de Biomedicina de València-CSIC, Valencia, Spain
- Unidad Mixta de Neurologia Genètica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Hospital Universitari Santa Maria de Lleida, Lleida, Spain
- Institut de Recerca Biomedica de Lleida (IRBLLeida), Lleida, Spain
| | - Adolfo López de Munian
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Department of Neurology. Hospital Universitario Donostia. San Sebastian, Spain
- Department of Neurosciences. Faculty of Medicine and Nursery. University of the Basque Country, San Sebastián, Spain
- Neurosciences Area. Instituto Biodonostia. San Sebastian, Spain
| | - Jose María García-Alberca
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer Research Center & Memory Clinic, Andalusian Institute for Neuroscience, Málaga, Spain
| | - Jose Luís Royo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
| | - María Jesús Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC)
- Instituto de Investigacion Sanitaria ‘Hospital la Paz’ (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid
| | - Victoria Álvarez
- Laboratorio de Genética. Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Luis Miguel Real
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
- Unidad Clínica de Enfermedades Infecciosas y Microbiología.Hospital Universitario de Valme, Sevilla, Spain
| | - Arturo Corbatón Anchuelo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
| | - Dulcenombre Gómez-Garre
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos; Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid (UCM)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - María Teresa Martínez Larrad
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Emilio Franco-Macías
- Dementia Unit, Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Pablo Mir
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Miguel Medina
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center
| | - Raquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders unit. Service of Neurology. Hospital Clínic of Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Department of Neurology, Sant Pau Memory Unit, Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica – CIBERER-IDIS, Santiago de Compostela, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montse Alegret
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pascual Sánchez Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| | - Alfredo Cabrera
- Neuroscience Therapeutic Area, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Amanda Cano
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Alzheimer’s Disease Neuroimaging Initiative.
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| |
Collapse
|
5
|
Cogan JA, Benova N, Kuklinkova R, Boyne JR, Anene CA. Meta-analysis of RNA interaction profiles of RNA-binding protein using the RBPInper tool. BIOINFORMATICS ADVANCES 2024; 4:vbae127. [PMID: 39233897 PMCID: PMC11374027 DOI: 10.1093/bioadv/vbae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/29/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
Motivation Recent RNA-centric experimental methods have significantly expanded our knowledge of proteins with known RNA-binding functions. However, the complete regulatory network and pathways for many of these RNA-binding proteins (RBPs) in different cellular contexts remain unknown. Although critical to understanding the role of RBPs in health and disease, experimentally mapping the RBP-RNA interactomes in every single context is an impossible task due the cost and manpower required. Additionally, identifying relevant RNAs bound by RBPs is challenging due to their diverse binding modes and function. Results To address these challenges, we developed RBP interaction mapper RBPInper an integrative framework that discovers global RBP interactome using statistical data fusion. Experiments on splicing factor proline and glutamine rich (SFPQ) datasets revealed cogent global SFPQ interactome. Several biological processes associated with this interactome were previously linked with SFPQ function. Furthermore, we conducted tests using independent dataset to assess the transferability of the SFPQ interactome to another context. The results demonstrated robust utility in generating interactomes that transfers to unseen cellular context. Overall, RBPInper is a fast and user-friendly method that enables a systems-level understanding of RBP functions by integrating multiple molecular datasets. The tool is designed with a focus on simplicity, minimal dependencies, and straightforward input requirements. This intentional design aims to empower everyday biologists, making it easy for them to incorporate the tool into their research. Availability and implementation The source code, documentation, and installation instructions as well as results for use case are freely available at https://github.com/AneneLab/RBPInper. A user can easily compile similar datasets for a target RBP.
Collapse
Affiliation(s)
- Joseph A Cogan
- School of Biological Sciences, University of Huddersfield, Huddersfield, HD1 3DH, United Kingdom
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Natalia Benova
- Centre for Biomedical Science Research, School of Health, Leeds Beckett University, Leeds, LS1 3HE, United Kingdom
| | - Rene Kuklinkova
- Centre for Biomedical Science Research, School of Health, Leeds Beckett University, Leeds, LS1 3HE, United Kingdom
| | - James R Boyne
- Centre for Biomedical Science Research, School of Health, Leeds Beckett University, Leeds, LS1 3HE, United Kingdom
| | - Chinedu A Anene
- Centre for Biomedical Science Research, School of Health, Leeds Beckett University, Leeds, LS1 3HE, United Kingdom
- Centre for Cancer Genomics and Computation Biology, Barts Cancer Institute, Queen Mary University of London, London, E1 4NS, United Kingdom
| |
Collapse
|
6
|
Zhu B, Liu S, David NL, Dion W, Doshi NK, Siegel LB, Amorim T, Andrews RE, Kumar GVN, Li H, Irfan S, Pesaresi T, Sharma AX, Sun M, Fazeli PK, Steinhauser ML. Evidence for ~12-h ultradian gene programs in humans. NPJ BIOLOGICAL TIMING AND SLEEP 2024; 1:4. [PMID: 39148626 PMCID: PMC11325440 DOI: 10.1038/s44323-024-00005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024]
Abstract
Mice and many marine organisms exhibit ~12-h ultradian rhythms, however, direct evidence of ~12-h ultradian rhythms in humans is lacking. Here, we performed prospective, temporal transcriptome profiling of peripheral white blood cells from three healthy humans. All three participants independently exhibited robust ~12-h transcriptional rhythms in molecular programs involved in RNA and protein metabolism, with strong homology to circatidal gene programs previously identified in Cnidarian marine species.
Collapse
Affiliation(s)
- Bokai Zhu
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Silvia Liu
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Natalie L David
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - William Dion
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Nandini K Doshi
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Lauren B Siegel
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Tânia Amorim
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Rosemary E Andrews
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - G V Naveen Kumar
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Hanwen Li
- Department of Statistics, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA USA
| | - Saad Irfan
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Tristan Pesaresi
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Ankit X Sharma
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Michelle Sun
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Pouneh K Fazeli
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Matthew L Steinhauser
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| |
Collapse
|
7
|
Alonso-García M, Gutiérrez-Gil B, Pelayo R, Fonseca PAS, Marina H, Arranz JJ, Suárez-Vega A. A meta-analysis approach for annotation and identification of lncRNAs controlling perirenal fat deposition in suckling lambs. Anim Biotechnol 2024; 35:2374328. [PMID: 39003576 DOI: 10.1080/10495398.2024.2374328] [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] [Indexed: 07/15/2024]
Abstract
Long non-coding RNAs (lncRNAs) are being studied in farm animals due to their association with traits of economic interest, such as fat deposition. Based on the analysis of perirenal fat transcriptomes, this research explored the relevance of these regulatory elements to fat deposition in suckling lambs. To that end, meta-analysis techniques have been implemented to efficiently characterize and detect differentially expressed transcripts from two different RNA-seq datasets, one including samples of two sheep breeds that differ in fat deposition features, Churra and Assaf (n = 14), and one generated from Assaf suckling lambs with different fat deposition levels (n = 8). The joint analysis of the 22 perirenal fat RNA-seq samples with the FEELnc software allowed the detection of 3953 novel lncRNAs. After the meta-analysis, 251 differentially expressed genes were identified, 21 of which were novel lncRNAs. Additionally, a co-expression analysis revealed that, in suckling lambs, lncRNAs may play a role in controlling angiogenesis and thermogenesis, processes highlighted in relation to high and low fat deposition levels, respectively. Overall, while providing information that could be applied for the improvement of suckling lamb carcass traits, this study offers insights into the biology of perirenal fat deposition regulation in mammals.
Collapse
Affiliation(s)
- María Alonso-García
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Beatriz Gutiérrez-Gil
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Rocío Pelayo
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Pablo A S Fonseca
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Héctor Marina
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Juan José Arranz
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Aroa Suárez-Vega
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| |
Collapse
|
8
|
Tian J, Jia K, Wang T, Guo L, Xuan Z, Michaelis EK, Swerdlow RH, Du H. Hippocampal transcriptome-wide association study and pathway analysis of mitochondrial solute carriers in Alzheimer's disease. Transl Psychiatry 2024; 14:250. [PMID: 38858380 PMCID: PMC11164935 DOI: 10.1038/s41398-024-02958-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/12/2024] Open
Abstract
The etiopathogenesis of late-onset Alzheimer's disease (AD) is increasingly recognized as the result of the combination of the aging process, toxic proteins, brain dysmetabolism, and genetic risks. Although the role of mitochondrial dysfunction in the pathogenesis of AD has been well-appreciated, the interaction between mitochondrial function and genetic variability in promoting dementia is still poorly understood. In this study, by tissue-specific transcriptome-wide association study (TWAS) and further meta-analysis, we examined the genetic association between mitochondrial solute carrier family (SLC25) genes and AD in three independent cohorts and identified three AD-susceptibility genes, including SLC25A10, SLC25A17, and SLC25A22. Integrative analysis using neuroimaging data and hippocampal TWAS-predicted gene expression of the three susceptibility genes showed an inverse correlation of SLC25A22 with hippocampal atrophy rate in AD patients, which outweighed the impacts of sex, age, and apolipoprotein E4 (ApoE4). Furthermore, SLC25A22 downregulation demonstrated an association with AD onset, as compared with the other two transcriptome-wide significant genes. Pathway and network analysis related hippocampal SLC25A22 downregulation to defects in neuronal function and development, echoing the enrichment of SLC25A22 expression in human glutamatergic neurons. The most parsimonious interpretation of the results is that we have identified AD-susceptibility genes in the SLC25 family through the prediction of hippocampal gene expression. Moreover, our findings mechanistically yield insight into the mitochondrial cascade hypothesis of AD and pave the way for the future development of diagnostic tools for the early prevention of AD from a perspective of precision medicine by targeting the mitochondria-related genes.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Kun Jia
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Tienju Wang
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Lan Guo
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Zhenyu Xuan
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Elias K Michaelis
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Russell H Swerdlow
- Alzheimer's Disease Research Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Heng Du
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA.
- Alzheimer's Disease Research Center, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
9
|
Antonatos C, Georgakilas GK, Evangelou E, Vasilopoulos Y. Transcriptomic meta-analysis characterizes molecular commonalities between psoriasis and obesity. Genes Immun 2024; 25:179-187. [PMID: 38580831 DOI: 10.1038/s41435-024-00271-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
Despite the abundance of epidemiological evidence for the high comorbid rate between psoriasis and obesity, systematic approaches to common inflammatory mechanisms have not been adequately explored. We performed a meta-analysis of publicly available RNA-sequencing datasets to unveil putative mechanisms that are postulated to exacerbate both diseases, utilizing both late-stage, disease-specific meta-analyses and consensus gene co-expression network (cWGCNA). Single-gene meta-analyses reported several common inflammatory mechanisms fostered by the perturbed expression profile of inflammatory cells. Assessment of gene overlaps between both diseases revealed significant overlaps between up- (n = 170, P value = 6.07 × 10-65) and down-regulated (n = 49, P value = 7.1 × 10-7) genes, associated with increased T cell response and activated transcription factors. Our cWGCNA approach disentangled 48 consensus modules, associated with either the differentiation of leukocytes or metabolic pathways with similar correlation signals in both diseases. Notably, all our analyses confirmed the association of the perturbed T helper (Th)17 differentiation pathway in both diseases. Our novel findings through whole transcriptomic analyses characterize the inflammatory commonalities between psoriasis and obesity implying the assessment of several expression profiles that could serve as putative comorbid disease progression biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece
| | - Georgios K Georgakilas
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece
- Information Management Systems Institute (IMSI), ATHENA Research Center, 15125, Athens, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110, Ioannina, Greece
- Department of Epidemiology & Biostatistics, MRC Centre for Environment and Health, Imperial College London, London, W2 1PG, UK
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece.
| |
Collapse
|
10
|
Carter JK, Quach BC, Willis C, Minto MS, Hancock DB, Montalvo-Ortiz J, Corradin O, Logan RW, Walss-Bass C, Maher BS, Johnson EO. Identifying novel gene dysregulation associated with opioid overdose death: A meta-analysis of differential gene expression in human prefrontal cortex. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.12.24301153. [PMID: 38260365 PMCID: PMC10802752 DOI: 10.1101/2024.01.12.24301153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Only recently have human postmortem brain studies of differential gene expression (DGE) associated with opioid overdose death (OOD) been published; sample sizes from these studies have been modest (N = 40-153). To increase statistical power to identify OOD-associated genes, we leveraged human prefrontal cortex RNAseq data from four independent OOD studies and conducted a transcriptome-wide DGE meta-analysis (N = 285). Using a unified gene expression data processing and analysis framework across studies, we meta-analyzed 20 098 genes and found 335 significant differentially expressed genes (DEGs) by OOD status (false discovery rate < 0.05). Of these, 66 DEGs were among the list of 303 genes reported as OOD-associated in prior prefrontal cortex molecular studies, including genes/gene families (e.g., OPRK1, NPAS4, DUSP, EGR). The remaining 269 DEGs were not previously reported (e.g., NR4A2, SYT1, HCRTR2, BDNF). There was little evidence of genetic drivers for the observed differences in gene expression between opioid addiction cases and controls. Enrichment analyses for the DEGs across molecular pathway and biological process databases highlight an interconnected set of genes and pathways from orexin and tyrosine kinase receptors through MEK/ERK/MAPK signaling to affect neuronal plasticity.
Collapse
Affiliation(s)
- Javan K. Carter
- Omics, Epidemiology, and Analytics Program, RTI International, Research Triangle Park, North Carolina, USA
| | - Bryan C. Quach
- Omics, Epidemiology, and Analytics Program, RTI International, Research Triangle Park, North Carolina, USA
| | - Caryn Willis
- Omics, Epidemiology, and Analytics Program, RTI International, Research Triangle Park, North Carolina, USA
| | - Melyssa S. Minto
- Omics, Epidemiology, and Analytics Program, RTI International, Research Triangle Park, North Carolina, USA
| | | | - Dana B. Hancock
- Omics, Epidemiology, and Analytics Program, RTI International, Research Triangle Park, North Carolina, USA
| | - Janitza Montalvo-Ortiz
- Department of Psychiatry, Division of Human Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Clinical Neurosciences Division, National Center of PTSD, VA CT Healthcare System, West Haven, Connecticut, USA
| | - Olivia Corradin
- Whitehead Institute Biomedical Research, Cambridge, Massachusetts, USA
| | - Ryan W. Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Consuelo Walss-Bass
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
- MD Anderson Cancer Center University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Brion S. Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Eric Otto Johnson
- Omics, Epidemiology, and Analytics Program, RTI International, Research Triangle Park, North Carolina, USA
- Fellow Program, RTI International, Research Triangle Park, North Carolina, USA
| |
Collapse
|
11
|
Jayanetti WT, Sikdar S. Empirically adjusted fixed-effects meta-analysis methods in genomic studies. Stat Appl Genet Mol Biol 2024; 23:sagmb-2023-0041. [PMID: 39340124 DOI: 10.1515/sagmb-2023-0041] [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: 12/14/2023] [Accepted: 09/10/2024] [Indexed: 09/30/2024]
Abstract
In recent years, meta-analyzing summary results from multiple studies has become a common practice in genomic research, leading to a significant improvement in the power of statistical detection compared to an individual genomic study. Meta analysis methods that combine statistical estimates across studies are known to be statistically more powerful than those combining statistical significance measures. An approach combining effect size estimates based on a fixed-effects model, called METAL, has gained extreme popularity to perform the former type of meta-analysis. In this article, we discuss the limitations of METAL due to its dependence on the theoretical null distribution, leading to incorrect significance testing results. Through various simulation studies and real genomic data application, we show how modifying the z-scores in METAL, using an empirical null distribution, can significantly improve the results, especially in presence of hidden confounders. For the estimation of the null distribution, we consider two different approaches, and we highlight the scenarios when one null estimation approach outperforms the other. This article will allow researchers to gain an insight into the importance of using an empirical null distribution in the fixed-effects meta-analysis as well as in choosing the appropriate empirical null distribution estimation approach.
Collapse
Affiliation(s)
- Wimarsha T Jayanetti
- Department of Statistical Sciences, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Sinjini Sikdar
- Department of Mathematics and Statistics, 6042 Old Dominion University , Norfolk, VA 23529, USA
| |
Collapse
|
12
|
He C, Thai PK, Bertrand L, Jayarathne A, van Mourik L, Phuc DH, Banks A, Mueller JF, Wang XF. Calibration and Application of PUF Disk Passive Air Samplers To Assess Chlorinated Paraffins in Ambient Air in Australia, China, and Vietnam. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:21061-21070. [PMID: 37939218 DOI: 10.1021/acs.est.3c06703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Ambient air samples were collected in Brisbane (Australia), Dalian (China), and Hanoi (Vietnam) during Mar 2013-Feb 2018 using polyurethane foam based passive air samplers. A sampling rate calibration experiment was conducted for chlorinated paraffins (CPs, i.e., short-chain, medium-chain, and long-chain CPs), where the sampling rates were 4.5 ± 0.7, 4.8 ± 0.3, and 4.8 ± 2.1 m3 day-1 for SCCPs, MCCPs, and LCCPs, respectively. The atmospheric concentration of CPs was then calculated and the medians of ∑CPs were 0.079, 1.0, and 0.89 ng m-3 in Brisbane, Dalian, and Hanoi, respectively. The concentration of CPs in Brisbane's air remained at low levels, with no significant differences observed between the city background site and the city center site, indicating limited usage and production of CPs in this city. The highest concentration of MCCPs was detected in Dalian, while the highest concentration of SCCPs was detected in Hanoi. A decrease of SCCP concentration and an increase of MCCPs' were found in Brisbane's air from 2016 to 2018, while increasing trends for both SCCPs and MCCPs were observed in Dalian. These results indicated impacts from different sources of CPs in the investigated cities.
Collapse
Affiliation(s)
- Chang He
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, P. R. China
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Key Laboratory for City Cluster Environmental Safety and Green Development of the Ministry of Education, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, P. R. China
- QAEHS, Queensland Alliance for Environmental Health Sciences, The University of Queensland, 4102 Brisbane, Australia
| | - Phong K Thai
- QAEHS, Queensland Alliance for Environmental Health Sciences, The University of Queensland, 4102 Brisbane, Australia
| | - Lidwina Bertrand
- CIBICI- CONICET and Universidad Nacional de Córdoba, Facultad Ciencias Químicas, Dpto. Bioquímica Clínica, 5000 Córdoba, Argentina
| | - Ayomi Jayarathne
- QAEHS, Queensland Alliance for Environmental Health Sciences, The University of Queensland, 4102 Brisbane, Australia
| | - Louise van Mourik
- Department of Environment and Health, Vrije Universiteit, De Boelelaan 1087, 1081 HV Amsterdam, The Netherlands
| | - Dam Hoang Phuc
- Hanoi University of Science and Technology, Hanoi 10999, Viet Nam
| | - Andrew Banks
- QAEHS, Queensland Alliance for Environmental Health Sciences, The University of Queensland, 4102 Brisbane, Australia
- Racing Science Centre, Queensland Racing Integrity Commission, 4010 Brisbane, Australia
| | - Jochen F Mueller
- QAEHS, Queensland Alliance for Environmental Health Sciences, The University of Queensland, 4102 Brisbane, Australia
| | - Xianyu Fisher Wang
- QAEHS, Queensland Alliance for Environmental Health Sciences, The University of Queensland, 4102 Brisbane, Australia
| |
Collapse
|
13
|
Roointan A, Ghaeidamini M, Shafieizadegan S, Hudkins KL, Gholaminejad A. Metabolome panels as potential noninvasive biomarkers for primary glomerulonephritis sub-types: meta-analysis of profiling metabolomics studies. Sci Rep 2023; 13:20325. [PMID: 37990116 PMCID: PMC10663527 DOI: 10.1038/s41598-023-47800-7] [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: 03/13/2023] [Accepted: 11/18/2023] [Indexed: 11/23/2023] Open
Abstract
Primary glomerulonephritis diseases (PGDs) are known as the top causes of chronic kidney disease worldwide. Renal biopsy, an invasive method, is the main approach to diagnose PGDs. Studying the metabolome profiles of kidney diseases is an inclusive approach to identify the disease's underlying pathways and discover novel non-invasive biomarkers. So far, different experiments have explored the metabolome profiles in different PGDs, but the inconsistencies might hinder their clinical translations. The main goal of this meta-analysis study was to achieve consensus panels of dysregulated metabolites in PGD sub-types. The PGDs-related metabolome profiles from urine samples in humans were selected in a comprehensive search. Amanida package in R software was utilized for performing the meta-analysis. Through sub-type analyses, the consensus list of metabolites in each category was obtained. To identify the most affected pathways, functional enrichment analysis was performed. Also, a gene-metabolite network was constructed to identify the key metabolites and their connected proteins. After a vigorous search, among the 11 selected studies (15 metabolite profiles), 270 dysregulated metabolites were recognized in urine of 1154 PGDs and control samples. Through sub-type analyses by Amanida package, the consensus list of metabolites in each category was obtained. Top dysregulated metabolites (vote score of ≥ 4 or ≤ - 4) in PGDs urines were selected as main panel of meta-metabolites including glucose, leucine, choline, betaine, dimethylamine, fumaric acid, citric acid, 3-hydroxyisovaleric acid, pyruvic acid, isobutyric acid, and hippuric acid. The enrichment analyses results revealed the involvement of different biological pathways such as the TCA cycle and amino acid metabolisms in the pathogenesis of PGDs. The constructed metabolite-gene interaction network revealed the high centralities of several metabolites, including pyruvic acid, leucine, and choline. The identified metabolite panels could shed a light on the underlying pathological pathways and be considered as non-invasive biomarkers for the diagnosis of PGD sub-types.
Collapse
Affiliation(s)
- Amir Roointan
- Regenerative Medicine Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Hezar Jarib St., Isfahan, 81746-73461, Iran
| | - Maryam Ghaeidamini
- Regenerative Medicine Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Hezar Jarib St., Isfahan, 81746-73461, Iran
| | - Saba Shafieizadegan
- Regenerative Medicine Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Hezar Jarib St., Isfahan, 81746-73461, Iran
| | - Kelly L Hudkins
- Department of Laboratory Medicine and Pathology, University of Washington, School of Medicine, Seattle, USA
| | - Alieh Gholaminejad
- Regenerative Medicine Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Hezar Jarib St., Isfahan, 81746-73461, Iran.
| |
Collapse
|
14
|
G K AV, Gogoi G, Kachappilly MC, Rangarajan A, Pandya HJ. Label-free multimodal electro-thermo-mechanical (ETM) phenotyping as a novel biomarker to differentiate between normal, benign, and cancerous breast biopsy tissues. J Biol Eng 2023; 17:68. [PMID: 37957665 PMCID: PMC10644568 DOI: 10.1186/s13036-023-00388-y] [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: 05/06/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Technologies for quick and label-free diagnosis of malignancies from breast tissues have the potential to be a significant adjunct to routine diagnostics. The biophysical phenotypes of breast tissues, such as its electrical, thermal, and mechanical properties (ETM), have the potential to serve as novel markers to differentiate between normal, benign, and malignant tissue. RESULTS We report a system-of-biochips (SoB) integrated into a semi-automated mechatronic system that can characterize breast biopsy tissues using electro-thermo-mechanical sensing. The SoB, fabricated on silicon using microfabrication techniques, can measure the electrical impedance (Z), thermal conductivity (K), mechanical stiffness (k), and viscoelastic stress relaxation (%R) of the samples. The key sensing elements of the biochips include interdigitated electrodes, resistance temperature detectors, microheaters, and a micromachined diaphragm with piezoresistive bridges. Multi-modal ETM measurements performed on formalin-fixed tumour and adjacent normal breast biopsy samples from N = 14 subjects were able to differentiate between invasive ductal carcinoma (malignant), fibroadenoma (benign), and adjacent normal (healthy) tissues with a root mean square error of 0.2419 using a Gaussian process classifier. Carcinoma tissues were observed to have the highest mean impedance (110018.8 ± 20293.8 Ω) and stiffness (0.076 ± 0.009 kNm-1) and the lowest thermal conductivity (0.189 ± 0.019 Wm-1 K-1) amongst the three groups, while the fibroadenoma samples had the highest percentage relaxation in normalized load (47.8 ± 5.12%). CONCLUSIONS The work presents a novel strategy to characterize the multi-modal biophysical phenotype of breast biopsy tissues to aid in cancer diagnosis from small-sized tumour samples. The methodology envisions to supplement the existing technology gap in the analysis of breast tissue samples in the pathology laboratories to aid the diagnostic workflow.
Collapse
Affiliation(s)
- Anil Vishnu G K
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Gayatri Gogoi
- Department of Pathology, Assam Medical College, Dibrugarh, Assam, 786002, India
| | - Midhun C Kachappilly
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Annapoorni Rangarajan
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Hardik J Pandya
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, Karnataka, 560012, India.
- Centre for Product Design and Manufacturing, Indian Institute of Science, Bangalore, Karnataka, 560012, India.
| |
Collapse
|
15
|
Teh WT, Chung J, Holdsworth-Carson SJ, Donoghue JF, Healey M, Rees HC, Bittinger S, Obers V, Sloggett C, Kendarsari R, Fung JN, Mortlock S, Montgomery GW, Girling JE, Rogers PAW. A molecular staging model for accurately dating the endometrial biopsy. Nat Commun 2023; 14:6222. [PMID: 37798294 PMCID: PMC10556104 DOI: 10.1038/s41467-023-41979-z] [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: 07/15/2022] [Accepted: 09/26/2023] [Indexed: 10/07/2023] Open
Abstract
Natural variability in menstrual cycle length, coupled with rapid changes in endometrial gene expression, makes it difficult to accurately define and compare different stages of the endometrial cycle. Here we develop and validate a method for precisely determining endometrial cycle stage based on global gene expression. Our 'molecular staging model' reveals significant and remarkably synchronised daily changes in expression for over 3400 endometrial genes throughout the cycle, with the most dramatic changes occurring during the secretory phase. Our study significantly extends existing data on the endometrial transcriptome, and for the first time enables identification of differentially expressed endometrial genes with increasing age and different ethnicities. It also allows reinterpretation of all endometrial RNA-seq and array data that has been published to date. Our molecular staging model will significantly advance understanding of endometrial-related disorders that affect nearly all women at some stage of their lives, such as heavy menstrual bleeding, endometriosis, adenomyosis, and recurrent implantation failure.
Collapse
Affiliation(s)
- W T Teh
- University of Melbourne Department of Obstetrics and Gynaecology, Melbourne, Victoria, Australia
- Royal Women's Hospital, Melbourne, Victoria, Australia
- Melbourne IVF, Melbourne, Victoria, Australia
| | - J Chung
- University of Melbourne Department of Obstetrics and Gynaecology, Melbourne, Victoria, Australia
- Melbourne Bioinformatics, University of Melbourne, Melbourne, Victoria, Australia
| | - S J Holdsworth-Carson
- University of Melbourne Department of Obstetrics and Gynaecology, Melbourne, Victoria, Australia
- Royal Women's Hospital, Melbourne, Victoria, Australia
- Julia Argyrou Endometriosis Centre, Epworth HealthCare, Richmond, Victoria, Australia
| | - J F Donoghue
- University of Melbourne Department of Obstetrics and Gynaecology, Melbourne, Victoria, Australia
- Royal Women's Hospital, Melbourne, Victoria, Australia
| | - M Healey
- University of Melbourne Department of Obstetrics and Gynaecology, Melbourne, Victoria, Australia
- Royal Women's Hospital, Melbourne, Victoria, Australia
| | - H C Rees
- Royal Women's Hospital, Melbourne, Victoria, Australia
- Royal Children's Hospital, Melbourne, Victoria, Australia
| | - S Bittinger
- Royal Women's Hospital, Melbourne, Victoria, Australia
- Royal Children's Hospital, Melbourne, Victoria, Australia
| | - V Obers
- Melbourne Pathology, Collingwood, Victoria, Australia
| | - C Sloggett
- Melbourne Bioinformatics, University of Melbourne, Melbourne, Victoria, Australia
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute, Melbourne, Victoria, Australia
| | - R Kendarsari
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
- Illumina Inc. 11 Biopolis Way, Singapore, 138667, Singapore
| | - J N Fung
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - S Mortlock
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - G W Montgomery
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - J E Girling
- University of Melbourne Department of Obstetrics and Gynaecology, Melbourne, Victoria, Australia
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, Aotearoa, New Zealand
| | - P A W Rogers
- University of Melbourne Department of Obstetrics and Gynaecology, Melbourne, Victoria, Australia.
- Royal Women's Hospital, Melbourne, Victoria, Australia.
| |
Collapse
|
16
|
Fotiadis P, Cieslak M, He X, Caciagli L, Ouellet M, Satterthwaite TD, Shinohara RT, Bassett DS. Myelination and excitation-inhibition balance synergistically shape structure-function coupling across the human cortex. Nat Commun 2023; 14:6115. [PMID: 37777569 PMCID: PMC10542365 DOI: 10.1038/s41467-023-41686-9] [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: 11/16/2022] [Accepted: 09/08/2023] [Indexed: 10/02/2023] Open
Abstract
Recent work has demonstrated that the relationship between structural and functional connectivity varies regionally across the human brain, with reduced coupling emerging along the sensory-association cortical hierarchy. The biological underpinnings driving this expression, however, remain largely unknown. Here, we postulate that intracortical myelination and excitation-inhibition (EI) balance mediate the heterogeneous expression of structure-function coupling (SFC) and its temporal variance across the cortical hierarchy. We employ atlas- and voxel-based connectivity approaches to analyze neuroimaging data acquired from two groups of healthy participants. Our findings are consistent across six complementary processing pipelines: 1) SFC and its temporal variance respectively decrease and increase across the unimodal-transmodal and granular-agranular gradients; 2) increased myelination and lower EI-ratio are associated with more rigid SFC and restricted moment-to-moment SFC fluctuations; 3) a gradual shift from EI-ratio to myelination as the principal predictor of SFC occurs when traversing from granular to agranular cortical regions. Collectively, our work delivers a framework to conceptualize structure-function relationships in the human brain, paving the way for an improved understanding of how demyelination and/or EI-imbalances induce reorganization in brain disorders.
Collapse
Affiliation(s)
- Panagiotis Fotiadis
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Matthew Cieslak
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xiaosong He
- Department of Psychology, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Lorenzo Caciagli
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mathieu Ouellet
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Theodore D Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Russell T Shinohara
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Biomedical Image Computing & Analytics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dani S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Santa Fe Institute, Santa Fe, NM, 87501, USA.
| |
Collapse
|
17
|
Colicchio JM, Amstutz CL, Garcia N, Prabhu KN, Cairns TM, Akman M, Gottilla T, Gollery T, Stricklin SL, Bayer TS. A tool for rapid, automated characterization of population epigenomics in plants. Sci Rep 2023; 13:12915. [PMID: 37591855 PMCID: PMC10435466 DOI: 10.1038/s41598-023-38356-7] [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: 02/16/2023] [Accepted: 07/06/2023] [Indexed: 08/19/2023] Open
Abstract
Epigenetic variation in plant populations is an important factor in determining phenotype and adaptation to the environment. However, while advances have been made in the molecular and computational methods to analyze the methylation status of a given sample of DNA, tools to profile and compare the methylomes of multiple individual plants or groups of plants at high resolution and low cost are lacking. Here, we describe a computational approach and R package (sounDMR) that leverages the benefits of long read nanopore sequencing to enable robust identification of differential methylation from complex experimental designs, as well as assess the variability within treatment groups and identify individual plants of interest. We demonstrate the utility of this approach by profiling a population of Arabidopsis thaliana exposed to a demethylating agent and identify genomic regions of high epigenetic variability between individuals. Given the low cost of nanopore sequencing devices and the ease of sample preparation, these results show that high resolution epigenetic profiling of plant populations can be made more broadly accessible in plant breeding and biotechnology.
Collapse
Affiliation(s)
| | | | | | | | | | - Melis Akman
- Sound Agriculture Company, Emeryville, CA, USA
| | | | | | | | | |
Collapse
|
18
|
Rivas E. RNA covariation at helix-level resolution for the identification of evolutionarily conserved RNA structure. PLoS Comput Biol 2023; 19:e1011262. [PMID: 37450549 PMCID: PMC10370758 DOI: 10.1371/journal.pcbi.1011262] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Many biologically important RNAs fold into specific 3D structures conserved through evolution. Knowing when an RNA sequence includes a conserved RNA structure that could lead to new biology is not trivial and depends on clues left behind by conservation in the form of covariation and variation. For that purpose, the R-scape statistical test was created to identify from alignments of RNA sequences, the base pairs that significantly covary above phylogenetic expectation. R-scape treats base pairs as independent units. However, RNA base pairs do not occur in isolation. The Watson-Crick (WC) base pairs stack together forming helices that constitute the scaffold that facilitates the formation of the non-WC base pairs, and ultimately the complete 3D structure. The helix-forming WC base pairs carry most of the covariation signal in an RNA structure. Here, I introduce a new measure of statistically significant covariation at helix-level by aggregation of the covariation significance and covariation power calculated at base-pair-level resolution. Performance benchmarks show that helix-level aggregated covariation increases sensitivity in the detection of evolutionarily conserved RNA structure without sacrificing specificity. This additional helix-level sensitivity reveals an artifact that results from using covariation to build an alignment for a hypothetical structure and then testing the alignment for whether its covariation significantly supports the structure. Helix-level reanalysis of the evolutionary evidence for a selection of long non-coding RNAs (lncRNAs) reinforces the evidence against these lncRNAs having a conserved secondary structure.
Collapse
Affiliation(s)
- Elena Rivas
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
| |
Collapse
|
19
|
Tyshkovskiy A, Ma S, Shindyapina AV, Tikhonov S, Lee SG, Bozaykut P, Castro JP, Seluanov A, Schork NJ, Gorbunova V, Dmitriev SE, Miller RA, Gladyshev VN. Distinct longevity mechanisms across and within species and their association with aging. Cell 2023; 186:2929-2949.e20. [PMID: 37269831 PMCID: PMC11192172 DOI: 10.1016/j.cell.2023.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2022] [Accepted: 05/02/2023] [Indexed: 06/05/2023]
Abstract
Lifespan varies within and across species, but the general principles of its control remain unclear. Here, we conducted multi-tissue RNA-seq analyses across 41 mammalian species, identifying longevity signatures and examining their relationship with transcriptomic biomarkers of aging and established lifespan-extending interventions. An integrative analysis uncovered shared longevity mechanisms within and across species, including downregulated Igf1 and upregulated mitochondrial translation genes, and unique features, such as distinct regulation of the innate immune response and cellular respiration. Signatures of long-lived species were positively correlated with age-related changes and enriched for evolutionarily ancient essential genes, involved in proteolysis and PI3K-Akt signaling. Conversely, lifespan-extending interventions counteracted aging patterns and affected younger, mutable genes enriched for energy metabolism. The identified biomarkers revealed longevity interventions, including KU0063794, which extended mouse lifespan and healthspan. Overall, this study uncovers universal and distinct strategies of lifespan regulation within and across species and provides tools for discovering longevity interventions.
Collapse
Affiliation(s)
- Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Siming Ma
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stanislav Tikhonov
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Sang-Goo Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Perinur Bozaykut
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - José P Castro
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Aging and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Nicholas J Schork
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Sergey E Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
20
|
Wuest SE, Schulz L, Rana S, Frommelt J, Ehmig M, Pires ND, Grossniklaus U, Hardtke CS, Hammes UZ, Schmid B, Niklaus PA. Single-gene resolution of diversity-driven overyielding in plant genotype mixtures. Nat Commun 2023; 14:3379. [PMID: 37291153 PMCID: PMC10250416 DOI: 10.1038/s41467-023-39130-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
In plant communities, diversity often increases productivity and functioning, but the specific underlying drivers are difficult to identify. Most ecological theories attribute positive diversity effects to complementary niches occupied by different species or genotypes. However, the specific nature of niche complementarity often remains unclear, including how it is expressed in terms of trait differences between plants. Here, we use a gene-centred approach to study positive diversity effects in mixtures of natural Arabidopsis thaliana genotypes. Using two orthogonal genetic mapping approaches, we find that between-plant allelic differences at the AtSUC8 locus are strongly associated with mixture overyielding. AtSUC8 encodes a proton-sucrose symporter and is expressed in root tissues. Genetic variation in AtSUC8 affects the biochemical activities of protein variants and natural variation at this locus is associated with different sensitivities of root growth to changes in substrate pH. We thus speculate that - in the particular case studied here - evolutionary divergence along an edaphic gradient resulted in the niche complementarity between genotypes that now drives overyielding in mixtures. Identifying genes important for ecosystem functioning may ultimately allow linking ecological processes to evolutionary drivers, help identify traits underlying positive diversity effects, and facilitate the development of high-performance crop variety mixtures.
Collapse
Affiliation(s)
- Samuel E Wuest
- Department of Evolutionary Biology and Environmental Studies and Zurich-Basel Plant Science Center, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
- Department of Plant and Microbial Biology and Zurich-Basel Plant Science Center, University of Zurich, Zollikerstrasse 107, 8008, Zurich, Switzerland.
- Department of Geography, Remote Sensing Laboratories, University of Zurich, 8057, Zurich, Switzerland.
- Agroscope, Group Breeding Research, Mueller-Thurgau-Strasse 29, 8820, Waedenswil, Switzerland.
| | - Lukas Schulz
- Plant Systems Biology, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Surbhi Rana
- Department of Plant Molecular Biology, University of Lausanne, Biophore Building, Lausanne, 1015, Switzerland
- Department of Crop Genetics, John Innes Centre, Norwich Research Park, Colney Ln, Norwich, NR4 7UH, United Kingdom
| | - Julia Frommelt
- Department of Evolutionary Biology and Environmental Studies and Zurich-Basel Plant Science Center, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Merten Ehmig
- Department of Systematic and Evolutionary Botany, University of Zurich, Zollikerstrasse 107, 8008, Zürich, Switzerland
| | - Nuno D Pires
- Department of Plant and Microbial Biology and Zurich-Basel Plant Science Center, University of Zurich, Zollikerstrasse 107, 8008, Zurich, Switzerland
| | - Ueli Grossniklaus
- Department of Plant and Microbial Biology and Zurich-Basel Plant Science Center, University of Zurich, Zollikerstrasse 107, 8008, Zurich, Switzerland
| | - Christian S Hardtke
- Department of Plant Molecular Biology, University of Lausanne, Biophore Building, Lausanne, 1015, Switzerland
| | - Ulrich Z Hammes
- Plant Systems Biology, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Bernhard Schmid
- Department of Evolutionary Biology and Environmental Studies and Zurich-Basel Plant Science Center, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Geography, Remote Sensing Laboratories, University of Zurich, 8057, Zurich, Switzerland
| | - Pascal A Niklaus
- Department of Evolutionary Biology and Environmental Studies and Zurich-Basel Plant Science Center, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| |
Collapse
|
21
|
Miyashita M, Bell JSK, Wenric S, Karaesmen E, Rhead B, Kase M, Kaneva K, De La Vega FM, Zheng Y, Yoshimatsu TF, Khramtsova G, Liu F, Zhao F, Howard FM, Nanda R, Beaubier N, White KP, Huo D, Olopade OI. Molecular profiling of a real-world breast cancer cohort with genetically inferred ancestries reveals actionable tumor biology differences between European ancestry and African ancestry patient populations. Breast Cancer Res 2023; 25:58. [PMID: 37231433 DOI: 10.1186/s13058-023-01627-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/27/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Endocrine-resistant HR+/HER2- breast cancer (BC) and triple-negative BC (TNBC) are of interest for molecularly informed treatment due to their aggressive natures and limited treatment profiles. Patients of African Ancestry (AA) experience higher rates of TNBC and mortality than European Ancestry (EA) patients, despite lower overall BC incidence. Here, we compare the molecular landscapes of AA and EA patients with HR+/HER2- BC and TNBC in a real-world cohort to promote equity in precision oncology by illuminating the heterogeneity of potentially druggable genomic and transcriptomic pathways. METHODS De-identified records from patients with TNBC or HR+/HER2- BC in the Tempus Database were randomly selected (N = 5000), with most having stage IV disease. Mutations, gene expression, and transcriptional signatures were evaluated from next-generation sequencing data. Genetic ancestry was estimated from DNA-seq. Differences in mutational prevalence, gene expression, and transcriptional signatures between AA and EA were compared. EA patients were used as the reference population for log fold-changes (logFC) in expression. RESULTS After applying inclusion criteria, 3433 samples were evaluated (n = 623 AA and n = 2810 EA). Observed patterns of dysregulated pathways demonstrated significant heterogeneity among the two groups. Notably, PIK3CA mutations were significantly lower in AA HR+/HER2- tumors (AA = 34% vs. EA = 42%, P < 0.05) and the overall cohort (AA = 28% vs. EA = 37%, P = 2.08e-05). Conversely, KMT2C mutation was significantly more frequent in AA than EA TNBC (23% vs. 12%, P < 0.05) and HR+/HER2- (24% vs. 15%, P = 3e-03) tumors. Across all subtypes and stages, over 8000 genes were differentially expressed between the two ancestral groups including RPL10 (logFC = 2.26, P = 1.70e-162), HSPA1A (logFC = - 2.73, P = 2.43e-49), ATRX (logFC = - 1.93, P = 5.89e-83), and NUTM2F (logFC = 2.28, P = 3.22e-196). Ten differentially expressed gene sets were identified among stage IV HR+/HER2- tumors, of which four were considered relevant to BC treatment and were significantly enriched in EA: ERBB2_UP.V1_UP (P = 3.95e-06), LTE2_UP.V1_UP (P = 2.90e-05), HALLMARK_FATTY_ACID_METABOLISM (P = 0.0073), and HALLMARK_ANDROGEN_RESPONSE (P = 0.0074). CONCLUSIONS We observed significant differences in mutational spectra, gene expression, and relevant transcriptional signatures between patients with genetically determined African and European ancestries, particularly within the HR+/HER2- BC and TNBC subtypes. These findings could guide future development of treatment strategies by providing opportunities for biomarker-informed research and, ultimately, clinical decisions for precision oncology care in diverse populations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Fang Liu
- The University of Chicago, Chicago, IL, USA
| | | | | | - Rita Nanda
- The University of Chicago, Chicago, IL, USA
| | | | - Kevin P White
- Tempus Inc, Chicago, IL, USA
- National University Singapore, Queenstown, Singapore
| | | | | |
Collapse
|
22
|
Rivas E. RNA covariation at helix-level resolution for the identification of evolutionarily conserved RNA structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.14.536965. [PMID: 37131783 PMCID: PMC10153129 DOI: 10.1101/2023.04.14.536965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Many biologically important RNAs fold into specific 3D structures conserved through evolution. Knowing when an RNA sequence includes a conserved RNA structure that could lead to new biology is not trivial and depends on clues left behind by conservation in the form of covariation and variation. For that purpose, the R-scape statistical test was created to identify from alignments of RNA sequences, the base pairs that significantly covary above phylogenetic expectation. R-scape treats base pairs as independent units. However, RNA base pairs do not occur in isolation. The Watson-Crick (WC) base pairs stack together forming helices that constitute the scaffold that facilitates the formation of the non-WC base pairs, and ultimately the complete 3D structure. The helix-forming WC base pairs carry most of the covariation signal in an RNA structure. Here, I introduce a new measure of statistically significant covariation at helix-level by aggregation of the covariation significance and covariation power calculated at base-pair-level resolution. Performance benchmarks show that helix-level aggregated covariation increases sensitivity in the detection of evolutionarily conserved RNA structure without sacrificing specificity. This additional helix-level sensitivity reveals an artifact that results from using covariation to build an alignment for a hypothetical structure and then testing the alignment for whether its covariation significantly supports the structure. Helix-level reanalysis of the evolutionary evidence for a selection of long non-coding RNAs (lncRNAs) reinforces the evidence against these lncRNAs having a conserved secondary structure. Availability Helix aggregated E-values are integrated in the R-scape software package (version 2.0.0.p and higher). The R-scape web server eddylab.org/R-scape includes a link to download the source code. Contact elenarivas@fas.harvard.edu. Supplementary information Supplementary data and code are provided with this manuscript at rivaslab.org .
Collapse
|
23
|
Nguyen HCT, Baik B, Yoon S, Park T, Nam D. Benchmarking integration of single-cell differential expression. Nat Commun 2023; 14:1570. [PMID: 36944632 PMCID: PMC10030080 DOI: 10.1038/s41467-023-37126-3] [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: 06/03/2022] [Accepted: 03/03/2023] [Indexed: 03/23/2023] Open
Abstract
Integration of single-cell RNA sequencing data between different samples has been a major challenge for analyzing cell populations. However, strategies to integrate differential expression analysis of single-cell data remain underinvestigated. Here, we benchmark 46 workflows for differential expression analysis of single-cell data with multiple batches. We show that batch effects, sequencing depth and data sparsity substantially impact their performances. Notably, we find that the use of batch-corrected data rarely improves the analysis for sparse data, whereas batch covariate modeling improves the analysis for substantial batch effects. We show that for low depth data, single-cell techniques based on zero-inflation model deteriorate the performance, whereas the analysis of uncorrected data using limmatrend, Wilcoxon test and fixed effects model performs well. We suggest several high-performance methods under different conditions based on various simulation and real data analyses. Additionally, we demonstrate that differential expression analysis for a specific cell type outperforms that of large-scale bulk sample data in prioritizing disease-related genes.
Collapse
Affiliation(s)
- Hai C T Nguyen
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Bukyung Baik
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Sora Yoon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dougu Nam
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
- Department of Mathematical Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
| |
Collapse
|
24
|
He B, Xiao Y, Liang H, Huang Q, Du Y, Li Y, Garmire D, Sun D, Garmire LX. ASGARD is A Single-cell Guided Pipeline to Aid Repurposing of Drugs. Nat Commun 2023; 14:993. [PMID: 36813801 PMCID: PMC9945835 DOI: 10.1038/s41467-023-36637-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Single-cell RNA sequencing technology has enabled in-depth analysis of intercellular heterogeneity in various diseases. However, its full potential for precision medicine has yet to be reached. Towards this, we propose A Single-cell Guided Pipeline to Aid Repurposing of Drugs (ASGARD) that defines a drug score to recommend drugs by considering all cell clusters to address the intercellular heterogeneity within each patient. ASGARD shows significantly better average accuracy on single-drug therapy compared to two bulk-cell-based drug repurposing methods. We also demonstrated that it performs considerably better than other cell cluster-level predicting methods. In addition, we validate ASGARD using the drug response prediction method TRANSACT with Triple-Negative-Breast-Cancer patient samples. We find that many top-ranked drugs are either approved by the Food and Drug Administration or in clinical trials treating corresponding diseases. In conclusion, ASGARD is a promising drug repurposing recommendation tool guided by single-cell RNA-seq for personalized medicine. ASGARD is free for educational use at https://github.com/lanagarmire/ASGARD .
Collapse
Affiliation(s)
- Bing He
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yao Xiao
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Haodong Liang
- Department of Statistics, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Qianhui Huang
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yuheng Du
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yijun Li
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - David Garmire
- Department of Electrical Engineering and Computer Science, College of Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Lana X Garmire
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Ham H, Park T. Combining p-values from various statistical methods for microbiome data. Front Microbiol 2022; 13:990870. [PMID: 36439799 PMCID: PMC9686280 DOI: 10.3389/fmicb.2022.990870] [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: 07/10/2022] [Accepted: 10/11/2022] [Indexed: 08/30/2023] Open
Abstract
MOTIVATION In the field of microbiome analysis, there exist various statistical methods that have been developed for identifying differentially expressed features, that account for the overdispersion and the high sparsity of microbiome data. However, due to the differences in statistical models or test formulations, it is quite often to have inconsistent significance results across statistical methods, that makes it difficult to determine the importance of microbiome taxa. Thus, it is practically important to have the integration of the result from all statistical methods to determine the importance of microbiome taxa. A standard meta-analysis is a powerful tool for integrative analysis and it provides a summary measure by combining p-values from various statistical methods. While there are many meta-analyses available, it is not easy to choose the best meta-analysis that is the most suitable for microbiome data. RESULTS In this study, we investigated which meta-analysis method most adequately represents the importance of microbiome taxa. We considered Fisher's method, minimum value of p method, Simes method, Stouffer's method, Kost method, and Cauchy combination test. Through simulation studies, we showed that Cauchy combination test provides the best combined value of p in the sense that it performed the best among the examined methods while controlling the type 1 error rates. Furthermore, it produced high rank similarity with the true ranks. Through the real data application of colorectal cancer microbiome data, we demonstrated that the most highly ranked microbiome taxa by Cauchy combination test have been reported to be associated with colorectal cancer.
Collapse
Affiliation(s)
- Hyeonjung Ham
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, South Korea
| | - Taesung Park
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, South Korea
- Departement of Statistics, Seoul National University, Seoul, South Korea
| |
Collapse
|
26
|
Chakravarthy A, Reddin I, Henderson S, Dong C, Kirkwood N, Jeyakumar M, Rodriguez DR, Martinez NG, McDermott J, Su X, Egawa N, Fjeldbo CS, Skingen VE, Lyng H, Halle MK, Krakstad C, Soleiman A, Sprung S, Lechner M, Ellis PJI, Wass M, Michaelis M, Fiegl H, Salvesen H, Thomas GJ, Doorbar J, Chester K, Feber A, Fenton TR. Integrated analysis of cervical squamous cell carcinoma cohorts from three continents reveals conserved subtypes of prognostic significance. Nat Commun 2022; 13:5818. [PMID: 36207323 PMCID: PMC9547055 DOI: 10.1038/s41467-022-33544-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 09/15/2022] [Indexed: 11/10/2022] Open
Abstract
Human papillomavirus (HPV)-associated cervical cancer is a leading cause of cancer deaths in women. Here we present an integrated multi-omic analysis of 643 cervical squamous cell carcinomas (CSCC, the most common histological variant of cervical cancer), representing patient populations from the USA, Europe and Sub-Saharan Africa and identify two CSCC subtypes (C1 and C2) with differing prognosis. C1 and C2 tumours can be driven by either of the two most common HPV types in cervical cancer (16 and 18) and while HPV16 and HPV18 are overrepresented among C1 and C2 tumours respectively, the prognostic difference between groups is not due to HPV type. C2 tumours, which comprise approximately 20% of CSCCs across these cohorts, display distinct genomic alterations, including loss or mutation of the STK11 tumour suppressor gene, increased expression of several immune checkpoint genes and differences in the tumour immune microenvironment that may explain the shorter survival associated with this group. In conclusion, we identify two therapy-relevant CSCC subtypes that share the same defining characteristics across three geographically diverse cohorts. Human papillomavirus (HPV) is a known cause of cervical cancer. Here, the authors perform a multi-omic analysis using published cervical squamous cell carcinoma cohorts from the USA, Europe, and SubSaharan Africa and identify two cervical squamous cell carcinoma subtypes that display prognostic differences.
Collapse
Affiliation(s)
- Ankur Chakravarthy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ian Reddin
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Stephen Henderson
- UCL Cancer Institute, Bill Lyons Informatics Centre, University College London, London, UK
| | - Cindy Dong
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, UK
| | - Nerissa Kirkwood
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, UK
| | - Maxmilan Jeyakumar
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, UK
| | | | | | | | | | - Nagayasau Egawa
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | | | - Heidi Lyng
- Department of Radiation Biology, Oslo University Hospital, Oslo, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| | - Mari Kyllesø Halle
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway; Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Camilla Krakstad
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway; Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Afschin Soleiman
- INNPATH, Institute of Pathology, Tirol Kliniken Innsbruck, Innsbruck, Austria
| | - Susanne Sprung
- Institute of Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matt Lechner
- UCL Cancer Institute, University College London, London, UK
| | - Peter J I Ellis
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, UK
| | - Mark Wass
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, UK
| | - Martin Michaelis
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, UK
| | - Heidi Fiegl
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helga Salvesen
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway; Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gareth J Thomas
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Kerry Chester
- UCL Cancer Institute, University College London, London, UK.
| | - Andrew Feber
- Centre for Molecular Pathology, Royal Marsden Hospital Trust, London, UK. .,Division of Surgery and Interventional Science, University College London, London, UK.
| | - Tim R Fenton
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK. .,School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, UK. .,Institute for Life Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
27
|
Arencibia A, Salazar LA. Microarray meta-analysis reveals IL6 and p38β/MAPK11 as potential targets of hsa-miR-124 in endothelial progenitor cells: Implications for stent re-endothelization in diabetic patients. Front Cardiovasc Med 2022; 9:964721. [PMID: 36176980 PMCID: PMC9513120 DOI: 10.3389/fcvm.2022.964721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Circulating endothelial progenitor cells (EPCs) play an important role in the repair processes of damaged vessels, favoring re-endothelization of stented vessels to minimize restenosis. EPCs number and function is diminished in patients with type 2 diabetes, a known risk factor for restenosis. Considering the impact of EPCs in vascular injury repair, we conducted a meta-analysis of microarray to assess the transcriptomic profile and determine target genes during the differentiation process of EPCs into mature ECs. Five microarray datasets, including 13 EPC and 12 EC samples were analyzed, using the online tool ExpressAnalyst. Differentially expressed genes (DEGs) analysis was done by Limma method, with an | log2FC| > 1 and FDR < 0.05. Combined p-value by Fisher exact method was computed for the intersection of datasets. There were 3,267 DEGs, 1,539 up-regulated and 1,728 down-regulated in EPCs, with 407 common DEGs in at least four datasets. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed enrichment for terms related to “AGE-RAGE signaling pathway in diabetic complications.” Intersection of common DEGs, KEGG pathways genes and genes in protein-protein interaction network (PPI) identified four key genes, two up-regulated (IL1B and STAT5A) and two down-regulated (IL6 and MAPK11). MicroRNA enrichment analysis of common DEGs depicted five hub microRNA targeting 175 DEGs, including STAT5A, IL6 and MAPK11, with hsa-miR-124 as common regulator. This group of genes and microRNAs could serve as biomarkers of EPCs differentiation during coronary stenting as well as potential therapeutic targets to improve stent re-endothelization, especially in diabetic patients.
Collapse
|
28
|
Barnewall RJ, Marsh IB, Quinn JC. Meta-Analysis of qPCR for Bovine Respiratory Disease Based on MIQE Guidelines. Front Mol Biosci 2022; 9:902401. [PMID: 35923462 PMCID: PMC9340069 DOI: 10.3389/fmolb.2022.902401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Qualitative and quantitative PCR-based tests are widely used in both diagnostics and research to assess the prevalence of disease-causing pathogens in veterinary medicine. The efficacy of these tests, usually measured in terms of sensitivity and specificity, is critical in confirming or excluding a clinical diagnosis. We undertook a meta-analysis to assess the inherent value of published PCR diagnostic approaches used to confirm and quantify bacteria and viruses associated with bovine respiratory disease (BRD) in cattle. This review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A thorough search of nine electronic databases (Web of Science, EBSCOhost, Cambridge journals online, ProQuest, PubMed, Sage journals online, ScienceDirect, Wiley online library and MEDLINE) was undertaken to find studies that had reported on the use of PCR and/or qPCR for the detection and/or quantification of BRD associated organisms. All studies meeting the inclusion criteria for reporting quantitative PCR for identification of BRD associated microorganisms were included in the analysis. Studies were then assessed on the applications of the Minimum Information for Publication of Quantitative Real-Time PCR Experiment (MIQE) and PCR primer/probe sequences were extracted and tested for in silico specificity using a high level of stringency. Fourteen full-text articles were included in this study. Of these, 79% of the analysed articles did not report the application of the MIQE guidelines in their study. High stringency in silico testing of 144 previously published PCR primer/probe sequences found many to have questionable specificity. This review identified a high occurrence of primer/probe sequences with a variable in silico specificity such that this may have implications for the accuracy of reporting. Although this analysis was only applied to one specific disease state, identification of animals suspected to be suffering from bovine respiratory disease, there appears to be more broadly a need for veterinary diagnostic studies to adopt international best practice for reporting of quantitative PCR diagnostic data to be both accurate and comparable between studies and methodologies.
Collapse
Affiliation(s)
- Rebecca J. Barnewall
- School of Agricultural, Environmental and Veterinary Science, Charles Sturt University, Wagga Wagga, NSW, Australia
- Gulbali Institute, Wagga Wagga, NSW, Australia
| | - Ian B. Marsh
- NSW DPI, Elizabeth Macarthur Agricultural Institute, Menangle, NSW, Australia
| | - Jane C. Quinn
- School of Agricultural, Environmental and Veterinary Science, Charles Sturt University, Wagga Wagga, NSW, Australia
- Gulbali Institute, Wagga Wagga, NSW, Australia
- *Correspondence: Jane C. Quinn,
| |
Collapse
|
29
|
Wang X, Gardner K, Tegegn MB, Dalgard CL, Alba C, Menzel S, Patel H, Pirooznia M, Fu YP, Seifuddin FT, Thein SL. Genetic variants of PKLR are associated with acute pain in sickle cell disease. Blood Adv 2022; 6:3535-3540. [PMID: 35271708 PMCID: PMC9198922 DOI: 10.1182/bloodadvances.2021006668] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/01/2022] [Indexed: 11/20/2022] Open
Abstract
Acute pain, the most prominent complication of sickle cell disease (SCD), results from vaso-occlusion triggered by sickling of deoxygenated red blood cells (RBCs). Concentration of 2,3-diphosphoglycerate (2,3-DPG) in RBCs promotes deoxygenation by preferentially binding to the low-affinity T conformation of HbS. 2,3-DPG is an intermediate substrate in the glycolytic pathway in which pyruvate kinase (gene PKLR, protein PKR) is a rate-limiting enzyme; variants in PKLR may affect PKR activity, 2,3-DPG levels in RBCs, RBC sickling, and acute pain episodes (APEs). We performed a candidate gene association study using 2 cohorts: 242 adult SCD-HbSS patients and 977 children with SCD-HbSS or SCD-HbSβ0 thalassemia. Seven of 47 PKLR variants evaluated in the adult cohort were associated with hospitalization: intron 4, rs2071053; intron 2, rs8177970, rs116244351, rs114455416, rs12741350, rs3020781, and rs8177964. All 7 variants showed consistent effect directions in both cohorts and remained significant in weighted Fisher's meta-analyses of the adult and pediatric cohorts using P < .0071 as threshold to correct for multiple testing. Allele-specific expression analyses in an independent cohort of 52 SCD adults showed that the intronic variants are likely to influence APE by affecting expression of PKLR, although the causal variant and mechanism are not defined.
Collapse
Affiliation(s)
- Xunde Wang
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD
| | - Kate Gardner
- School of Cancer & Pharmaceutical Sciences, King’s College London, London, United Kingdom
- Department of Haematology, Guy and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Mickias B. Tegegn
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology & Genetics, and
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Camille Alba
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD
| | - Stephan Menzel
- School of Cancer & Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Hamel Patel
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | | | - Yi-Ping Fu
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | | | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD
| |
Collapse
|
30
|
Sun X, Xu H, Liu G, Chen J, Xu J, Li M, Liu L. A Robust Immuno-Prognostic Model of Non-Muscle-Invasive Bladder Cancer Indicates Dynamic Interaction in Tumor Immune Microenvironment Contributes to Cancer Progression. Front Genet 2022; 13:833989. [PMID: 35719408 PMCID: PMC9205430 DOI: 10.3389/fgene.2022.833989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/28/2022] [Indexed: 12/24/2022] Open
Abstract
Non-muscle-invasive bladder cancer (NMIBC) accounts for more than 70% of urothelial cancer. More than half of NMIBC patients experience recurrence, progression, or metastasis, which essentially reduces life quality and survival time. Identifying the high-risk patients prone to progression remains the primary concern of risk management of NMIBC. In this study, we included 1370 NMIBC transcripts data from nine public datasets, identified nine tumor-infiltrating marker cells highly related to the survival of NMIBC, quantified the cells’ proportion by self-defined differentially expressed signature genes, and established a robust immuno-prognostic model dividing NMIBC patients into low-risk versus high-risk progression groups. Our model implies that the loss of crosstalk between tumor cells and adjacent normal epithelium, along with enriched cell proliferation signals, may facilitate tumor progression. Thus, evaluating tumor progression should consider various components in the tumor immune microenvironment instead of the single marker in a single dimension. Moreover, we also appeal to the necessity of using appropriate meta-analysis methods to integrate the evidence from multiple sources in the feature selection step from large-scale heterogeneous omics data such as our study.
Collapse
Affiliation(s)
- Xiaomeng Sun
- Institutes of Biomedical Sciences and School of Basic Medical Sciences, Fudan University, Shanghai, China
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai, China
| | - Huilin Xu
- Institutes of Biomedical Sciences and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Gang Liu
- Institutes of Biomedical Sciences and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiani Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jinrong Xu
- Department of Electronic Engineering, Taiyuan Institute of Technology, Taiyuan, China
- *Correspondence: Jinrong Xu, ; Mingming Li, ; Lei Liu,
| | - Mingming Li
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
- *Correspondence: Jinrong Xu, ; Mingming Li, ; Lei Liu,
| | - Lei Liu
- Institutes of Biomedical Sciences and School of Basic Medical Sciences, Fudan University, Shanghai, China
- *Correspondence: Jinrong Xu, ; Mingming Li, ; Lei Liu,
| |
Collapse
|
31
|
Hou CD, Yang TS. Distribution of weighted Lancaster’s statistic for combining independent or dependent P-values, with applications to human genetic studies. COMMUN STAT-THEOR M 2022. [DOI: 10.1080/03610926.2022.2046088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Chia-Ding Hou
- Department of Statistics and Information Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ti-Sung Yang
- Department of Statistics and Information Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
32
|
De Silva K, Demmer RT, Jönsson D, Mousa A, Forbes A, Enticott J. A data-driven biocomputing pipeline with meta-analysis on high throughput transcriptomics to identify genome-wide miRNA markers associated with type 2 diabetes. Heliyon 2022; 8:e08886. [PMID: 35169647 PMCID: PMC8829580 DOI: 10.1016/j.heliyon.2022.e08886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/23/2021] [Accepted: 01/29/2022] [Indexed: 12/12/2022] Open
|
33
|
Moeckli B, Delaune V, Prados J, Tihy M, Peloso A, Oldani G, Delmi T, Slits F, Gex Q, Rubbia-Brandt L, Goossens N, Lacotte S, Toso C. Impact of Maternal Obesity on Liver Disease in the Offspring: A Comprehensive Transcriptomic Analysis and Confirmation of Results in a Murine Model. Biomedicines 2022; 10:biomedicines10020294. [PMID: 35203502 PMCID: PMC8869223 DOI: 10.3390/biomedicines10020294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/16/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The global obesity epidemic particularly affects women of reproductive age. Offspring of obese mothers suffer from an increased risk of liver disease but the molecular mechanisms involved remain unknown. We performed an integrative genomic analysis of datasets that investigated the impact of maternal obesity on the hepatic gene expression profile of the offspring in mice. Furthermore, we developed a murine model of maternal obesity and studied the development of liver disease and the gene expression profile of the top dysregulated genes by quantitative real-time polymerase chain reaction (qPCR). Our data are available for interactive exploration on our companion webpage. We identified five publicly available datasets relevant to our research question. Pathways involved in metabolism, the innate immune system, the clotting cascade, and the cell cycle were consistently dysregulated in the offspring of obese mothers. Concerning genes involved in the development of liver disease, Egfr, Vegfb, Wnt2,Pparg and six other genes were dysregulated in multiple independent datasets. In our own model, we observed a higher tendency towards the development of non-alcoholic liver disease (60 vs. 20%) and higher levels of alanine aminotransferase (41.0 vs. 12.5 IU/l, p = 0.008) in female offspring of obese mothers. Male offspring presented higher levels of liver fibrosis (2.4 vs. 0.6% relative surface area, p = 0.045). In a qPCR gene expression analysis of our own samples, we found Fgf21, Pparg, Ppard, and Casp6 to be dysregulated by maternal obesity. Maternal obesity represents a looming threat to the liver health of future generations. Our comprehensive transcriptomic analysis will help to better understand the mechanisms of the development of liver disease in the offspring of obese mothers and can give rise to further explorations.
Collapse
Affiliation(s)
- Beat Moeckli
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Vaihere Delaune
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Julien Prados
- Bioinformatics Support Platform, Services Communs de la Faculté, University of Geneva, 1206 Geneva, Switzerland;
| | - Matthieu Tihy
- Division of Clinical Pathology, Geneva University Hospitals, 1205 Geneva, Switzerland; (M.T.); (L.R.-B.)
| | - Andrea Peloso
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Graziano Oldani
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Thomas Delmi
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
| | - Florence Slits
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
| | - Quentin Gex
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
| | - Laura Rubbia-Brandt
- Division of Clinical Pathology, Geneva University Hospitals, 1205 Geneva, Switzerland; (M.T.); (L.R.-B.)
| | - Nicolas Goossens
- Division of Gastroenterology, Geneva University Hospitals, 1205 Geneva, Switzerland;
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Correspondence:
| | - Christian Toso
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| |
Collapse
|
34
|
Llambrich M, Correig E, Gumà J, Brezmes J, Cumeras R. Amanida: an R package for meta-analysis of metabolomics non-integral data. Bioinformatics 2022; 38:583-585. [PMID: 34406360 PMCID: PMC8722753 DOI: 10.1093/bioinformatics/btab591] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 02/03/2023] Open
Abstract
SUMMARY The combination, analysis and evaluation of different studies which try to answer or solve the same scientific question, also known as a meta-analysis, plays a crucial role in answering relevant clinical relevant questions. Unfortunately, metabolomics studies rarely disclose all the statistical information needed to perform a meta-analysis. Here, we present a meta-analysis approach using only the most reported statistical parameters in this field: P-value and fold-change. The P-values are combined via Fisher's method and fold-changes by averaging, both weighted by the study size (n). The amanida package includes several visualization options: a volcano plot for quantitative results, a vote plot for total regulation behaviours (up/down regulations) for each compound, and a explore plot of the vote-counting results with the number of times a compound is found upregulated or downregulated. In this way, it is very easy to detect discrepancies between studies at a first glance. AVAILABILITY AND IMPLEMENTATION Amanida code and documentation are at CRAN and https://github.com/mariallr/amanida. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Maria Llambrich
- Department of Electrical Electronic Engineering and Automation, Universitat Rovira i Virgili, IISPV, 43007 Tarragona, Spain
- Metabolomics Interdisciplinary Group, Department of Nutrition and Metabolism, Institut d’Investigació Sanitària Pere Virgili, 43201 Reus, Catalonia, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Eudald Correig
- Department of Biostatistics, Universitat Rovira i Virgili, 43201 Reus, Catalonia, Spain
| | - Josep Gumà
- Oncology Department, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Jesús Brezmes
- Department of Electrical Electronic Engineering and Automation, Universitat Rovira i Virgili, IISPV, 43007 Tarragona, Spain
- Metabolomics Interdisciplinary Group, Department of Nutrition and Metabolism, Institut d’Investigació Sanitària Pere Virgili, 43201 Reus, Catalonia, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Raquel Cumeras
- Department of Electrical Electronic Engineering and Automation, Universitat Rovira i Virgili, IISPV, 43007 Tarragona, Spain
- Metabolomics Interdisciplinary Group, Department of Nutrition and Metabolism, Institut d’Investigació Sanitària Pere Virgili, 43201 Reus, Catalonia, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), ISCIII, 28029 Madrid, Spain
- West Coast Metabolomics Center, University of California Davis, CA 95616, USA
| |
Collapse
|
35
|
He B, Xiao Y, Liang H, Huang Q, Du Y, Li Y, Garmire D, Sun D, Garmire LX. ASGARD: A Single-cell Guided pipeline to Aid Repurposing of Drugs. ARXIV 2021:arXiv:2109.06377v4. [PMID: 34545335 PMCID: PMC8452105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Revised: 12/22/2022] [Indexed: 01/04/2023]
Abstract
Intercellular heterogeneity is a major obstacle to successful precision medicine. Single-cell RNA sequencing (scRNA-seq) technology has enabled in-depth analysis of intercellular heterogeneity in various diseases. However, its full potential for precision medicine has yet to be reached. Towards this, we propose a new drug recommendation system called: A Single-cell Guided Pipeline to Aid Repurposing of Drugs (ASGARD). ASGARD defines a novel drug score predicting drugs by considering all cell clusters to address the intercellular heterogeneity within each patient. We tested ASGARD on multiple diseases, including breast cancer, acute lymphoblastic leukemia, and coronavirus disease 2019 (COVID-19). On single-drug therapy, ASGARD shows significantly better average accuracy (AUC of 0.92) compared to two other bulk-cell-based drug repurposing methods (AUC of 0.80 and 0.76). It is also considerably better (AUC of 0.82) than other cell cluster level predicting methods (AUC of 0.67 and 0.55). In addition, ASGARD is also validated by the drug response prediction method TRANSACT with Triple-Negative-Breast-Cancer patient samples. Many top-ranked drugs are either approved by FDA or in clinical trials treating corresponding diseases. In silico cell-type specific drop-out experiments using triple-negative breast cancers show the importance of T cells in the tumor microenvironment in affecting drug predictions. In conclusion, ASGARD is a promising drug repurposing recommendation tool guided by single-cell RNA-seq for personalized medicine. ASGARD is free for educational use at https://github.com/lanagarmire/ASGARD.
Collapse
Affiliation(s)
- Bing He
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yao Xiao
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Haodong Liang
- Department of Statistics, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Qianhui Huang
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yuheng Du
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yijun Li
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - David Garmire
- Department of Electrical Engineering and Computer Science, College of Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Lana X. Garmire
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Mutations in Epigenetic Regulation Genes in Gastric Cancer. Cancers (Basel) 2021; 13:cancers13184586. [PMID: 34572812 PMCID: PMC8467700 DOI: 10.3390/cancers13184586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Epigenetic mechanisms, such as DNA methylation/demethylation, covalent modifications of histone proteins, and chromatin remodeling, create specific patterns of gene expression. Epigenetic deregulations are associated with oncogenesis, relapse of the disease and metastases, and can serve as a useful clinical marker. We assessed the clinical relevance of integrity of the genes coding for epigenetic regulator proteins by mutational profiling of 25 genes in 135 gastric cancer (GC) samples. Overall, mutations in the epigenetic regulation genes were found to be significantly associated with reduced overall survival of patients in the group with metastases and in the group with tumors with signet ring cells. We have also discovered mutual exclusivity of somatic mutations in the KMT2D, KMT2C, ARID1A, and CHD7 genes in our cohort. Our results suggest that mutations in epigenetic regulation genes may be valuable clinical markers and deserve further exploration in independent cohorts. Abstract We have performed mutational profiling of 25 genes involved in epigenetic processes on 135 gastric cancer (GC) samples. In total, we identified 79 somatic mutations in 49/135 (36%) samples. The minority (n = 8) of mutations was identified in DNA methylation/demethylation genes, while the majority (n = 41), in histone modifier genes, among which mutations were most commonly found in KMT2D and KMT2C. Somatic mutations in KMT2D, KMT2C, ARID1A and CHD7 were mutually exclusive (p = 0.038). Mutations in ARID1A were associated with distant metastases (p = 0.03). The overall survival of patients in the group with metastases and in the group with tumors with signet ring cells was significantly reduced in the presence of mutations in epigenetic regulation genes (p = 0.036 and p = 0.041, respectively). Separately, somatic mutations in chromatin remodeling genes correlate with low survival rate of patients without distant metastasis (p = 0.045) and in the presence of signet ring cells (p = 0.0014). Our results suggest that mutations in epigenetic regulation genes may be valuable clinical markers and deserve further exploration in independent cohorts.
Collapse
|