1
|
Duran I, Tsurumi A. Evaluating transcriptional alterations associated with ageing and developing age prediction models based on the human blood transcriptome. Biogerontology 2025; 26:86. [PMID: 40186010 DOI: 10.1007/s10522-025-10216-z] [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: 02/09/2024] [Accepted: 03/04/2025] [Indexed: 04/07/2025]
Abstract
Ageing-related DNA methylome and proteome changes and machine-learned ageing clock models have been described previously; however, there is a dearth of ageing clock prediction models based on human blood transcript information. Applying various machine learning algorithms is expected to aid in the development of age prediction models. Using blood transcriptome data from healthy subjects ranging in age from 21 to 90 in the 10 K Immunomes repository, we evaluated differentially regulated transcripts, assessed enriched gene ontology, pathway and disease ontology analysis to characterize biological functions associated with the genes associated with age. Furthermore, we constructed and compared age prediction models developed by applying the Least Absolute Shrinkage and Selection Operator (LASSO), Elastic Net (EN), eXtreme Gradient Boosting (XGBoost) and Light Gradient-Boosting Machine (LightGBM) algorithms. Compared to LASSO (7 genes) and EN (9 genes) regularized regression, XGBoost (142 genes) and LightGBM (149 genes) Gradient Boosted Decision Tree methods performed better in this dataset (training set r = 0.836 (LASSO), 0.837 (EN), 1.000 (XGBoost) and 0.995 (LightGBM); test set: r = 0.883 (LASSO), 0.876 (EN), 0.931 (XGBoost) and 0.915 (LightGBM); external validation set: r = 0.535 (LASSO), 0.534 (EN), 0.591 (XGBoost) and 0.645 (LightGBM)). Blood transcriptome-based age prediction models may provide a simple method to monitor biological ageing, and provide additional molecular insight. Future studies to externally validate these models in various diverse large populations and molecular studies to elucidate the underlying mechanisms by which the gene expression levels may be related to ageing phenotypes would be advantageous.
Collapse
Affiliation(s)
- Ivan Duran
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Boston, MA, 02114, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Boston, MA, 02114, USA.
- Shriners Hospitals for Children-Boston, 51 Blossom St., Boston, MA, 02114, USA.
| |
Collapse
|
2
|
Briller S, Ben David G, Amir Y, Atzmon G, Somekh J. A computational framework for detecting inter-tissue gene-expression coordination changes with aging. Sci Rep 2025; 15:11014. [PMID: 40164681 PMCID: PMC11958765 DOI: 10.1038/s41598-025-94043-9] [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: 04/24/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Aging is a complex and systematic biological process that involves multiple genes and biological pathways across different tissues. While existing studies focus on tissue-specific aging factors, the inter-tissue interplay between molecular pathways during aging remains insufficiently explored. To bridge this gap, we propose a novel computational framework to identify the effect of aging on the coordinated patterns of gene-expression across multiple tissues. Our framework includes (1) an adjusted multi-tissue weighted gene co-expression network analysis, (2) differential network connectivity analysis between age groups and (3) machine learning models, XGBoost and Random Forest (RF) fed by gene expression levels and lower-dimensional pathway score space, to identify unique key inter-tissue genes and biological pathways for classifying aging. We applied our approach to three representative tissues: Adipose-Subcutaneous, Muscle-Skeletal and Brain-Cortex. The RF model demonstrated the best performance in predicting age group (AUC < 88%) highlighting key genes involved in inter-tissue coordination processes in aging. We also identified the inter-tissue involvement of lipid metabolism, immune system, and cell communication pathways during aging and detected distinct aging pathways manifested between tissues. The proposed framework highlights the importance of inter-tissue coordination processes underlying aging and provides valuable insights into aging mechanisms which can further assist in the development of therapeutic strategies promoting healthy aging.
Collapse
Affiliation(s)
- Shaked Briller
- Department of Information Systems, University of Haifa, Haifa, Israel
| | - Gil Ben David
- Department of Human Biology, University of Haifa, Haifa, Israel
| | - Yam Amir
- Department of Human Biology, University of Haifa, Haifa, Israel
- Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Gil Atzmon
- Department of Human Biology, University of Haifa, Haifa, Israel
| | - Judith Somekh
- Department of Information Systems, University of Haifa, Haifa, Israel.
| |
Collapse
|
3
|
Sobin L, Barcus M, Branton PA, Engel KB, Keen J, Tabor D, Ardlie KG, Greytak SR, Roche N, Luke B, Vaught J, Guan P, Moore HM. Histologic and Quality Assessment of Genotype-Tissue Expression (GTEx) Research Samples: A Large Postmortem Tissue Collection. Arch Pathol Lab Med 2025; 149:217-232. [PMID: 38797720 DOI: 10.5858/arpa.2023-0467-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2024] [Indexed: 05/29/2024]
Abstract
CONTEXT.— The National Institutes of Health Genotype-Tissue Expression (GTEx) project was developed to elucidate how genetic variation influences gene expression in multiple normal tissues procured from postmortem donors. OBJECTIVE.— To provide critical insight into a biospecimen's suitability for subsequent analysis, each biospecimen underwent quality assessment measures that included evaluation for underlying disease and potential effects introduced by preanalytic factors. DESIGN.— Electronic images of each tissue collected from nearly 1000 postmortem donors were evaluated by board-certified pathologists for the extent of autolysis, tissue purity, and the type and abundance of any extraneous tissue. Tissue-specific differences in the severity of autolysis and RNA integrity were evaluated, as were potential relationships between these markers and the duration of postmortem interval and rapidity of death. RESULTS.— Tissue-specific challenges in the procurement and preservation of the nearly 30 000 tissue specimens collected during the GTEx project are summarized. Differences in the degree of autolysis and RNA integrity number were observed among the 40 tissue types evaluated, and tissue-specific susceptibilities to the duration of postmortem interval and rapidity of death were observed. CONCLUSIONS.— Ninety-five percent of tissues were of sufficient quality to support RNA sequencing analysis. Biospecimens, annotated whole slide images, de-identified clinical data, and genomic data generated for GTEx represent a high-quality and comprehensive resource for the scientific community that has contributed to its use in approximately 1695 articles. Biospecimens and data collected under the GTEx project are available via the GTEx portal and authorized access to the Database of Genotypes and Phenotypes; procedures and whole slide images are available from the National Cancer Institute.
Collapse
Affiliation(s)
- Leslie Sobin
- From Leidos Biomedical Research, Inc, Rockville, Maryland (Sobin, Barcus, Tabor, Roche, Luke)
| | - Mary Barcus
- From Leidos Biomedical Research, Inc, Rockville, Maryland (Sobin, Barcus, Tabor, Roche, Luke)
| | - Philip A Branton
- Biorepositories and Biospecimen Research Branch, Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland (Branton, Keen, Vaught, Guan, Moore)
| | - Kelly B Engel
- Preferred Scientific Group, North Bethesda, Maryland (Engel)
| | - Judy Keen
- Biorepositories and Biospecimen Research Branch, Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland (Branton, Keen, Vaught, Guan, Moore)
| | - David Tabor
- From Leidos Biomedical Research, Inc, Rockville, Maryland (Sobin, Barcus, Tabor, Roche, Luke)
| | - Kristin G Ardlie
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts (Ardlie)
| | | | - Nancy Roche
- From Leidos Biomedical Research, Inc, Rockville, Maryland (Sobin, Barcus, Tabor, Roche, Luke)
| | - Brian Luke
- From Leidos Biomedical Research, Inc, Rockville, Maryland (Sobin, Barcus, Tabor, Roche, Luke)
| | - Jim Vaught
- Biorepositories and Biospecimen Research Branch, Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland (Branton, Keen, Vaught, Guan, Moore)
| | - Ping Guan
- Biorepositories and Biospecimen Research Branch, Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland (Branton, Keen, Vaught, Guan, Moore)
| | - Helen M Moore
- Biorepositories and Biospecimen Research Branch, Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland (Branton, Keen, Vaught, Guan, Moore)
| |
Collapse
|
4
|
Munshi S, Alarbi AM, Zheng H, Kuplicki R, Burrows K, Figueroa-Hall LK, Victor TA, Aupperle RL, Khalsa SS, Paulus MP, Teague TK, Savitz J. Increased expression of ER stress, inflammasome activation, and mitochondrial biogenesis-related genes in peripheral blood mononuclear cells in major depressive disorder. Mol Psychiatry 2025; 30:574-586. [PMID: 39174649 PMCID: PMC12054637 DOI: 10.1038/s41380-024-02695-2] [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: 11/05/2023] [Accepted: 08/09/2024] [Indexed: 08/24/2024]
Abstract
A subset of major depressive disorder (MDD) is characterized by immune system dysfunction, but the intracellular origin of these immune changes remains unclear. Here we tested the hypothesis that abnormalities in endoplasmic reticulum (ER) stress, inflammasome activity and mitochondrial biogenesis contribute to the development of systemic inflammation in MDD. RT-qPCR was used to measure mRNA expression of key organellar genes from peripheral blood mononuclear cells (PBMCs) isolated from 186 MDD and 67 healthy control (HC) subjects. The comparative CT (2-ΔΔCT) method was applied to quantify mRNA expression using GAPDH as the reference gene. After controlling for age, sex, BMI, and medication status using linear regression models, expression of the inflammasome (NLRC4 and NLRP3) and the ER stress (XBP1u, XBP1s, and ATF4) genes was found to be significantly increased in the MDD versus the HC group. Sensitivity analyses excluding covariates yielded similar results. After excluding outliers, expression of the inflammasome genes was no longer statistically significant but expression of the ER stress genes (XBP1u, XBP1s, and ATF4) remained significant and the mitochondrial biogenesis gene, MFN2, was significantly increased in the MDD group. NLRC4 and MFN2 were positively correlated with serum C-reactive protein concentrations, while ASC trended significant. The altered expression of inflammasome activation, ER stress, and mitochondrial biogenesis pathway components suggest that dysfunction of these organelles may play a role in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| | - Ahlam M Alarbi
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
| | - Haixia Zheng
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Rayus Kuplicki
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Kaiping Burrows
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Leandra K Figueroa-Hall
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Teresa A Victor
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Robin L Aupperle
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California at Los Angeles, 300 UCLA Medical Plaza, Los Angeles, CA, 90095, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - T Kent Teague
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
- Department of Biochemistry and Microbiology, Center for Health Sciences, Oklahoma State University, 1111 W. 17th St., Tulsa, OK, 74107, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| |
Collapse
|
5
|
Kramer NE, Byun S, Coryell P, D’Costa S, Thulson E, Kim H, Parkus SM, Bond ML, Klein ER, Shine J, Chubinskaya S, Love MI, Mohlke KL, Diekman BO, Loeser RF, Phanstiel DH. Response eQTLs, chromatin accessibility, and 3D chromatin structure in chondrocytes provide mechanistic insight into osteoarthritis risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592567. [PMID: 38952796 PMCID: PMC11216363 DOI: 10.1101/2024.05.05.592567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Osteoarthritis (OA) poses a significant healthcare burden with limited treatment options. While genome-wide association studies (GWAS) have identified over 100 OA-associated loci, translating these findings into therapeutic targets remains challenging. Integrating expression quantitative trait loci (eQTL), 3D chromatin structure, and other genomic approaches with OA GWAS data offers a promising approach to elucidate disease mechanisms; however, comprehensive eQTL maps in OA-relevant tissues and conditions remain scarce. We mapped gene expression, chromatin accessibility, and 3D chromatin structure in primary human articular chondrocytes in both resting and OA-mimicking conditions. We identified thousands of differentially expressed genes, including those associated with differences in sex and age. RNA-seq in chondrocytes from 101 donors across two conditions uncovered 3782 unique eGenes, including 420 that exhibited strong and significant condition-specific effects. Colocalization with OA GWAS signals revealed 13 putative OA risk genes, 10 of which have not been previously identified. Chromatin accessibility and 3D chromatin structure provided insights into the mechanisms and conditional specificity of these variants. Our findings shed light on OA pathogenesis and highlight potential targets for therapeutic development.
Collapse
Affiliation(s)
- Nicole E Kramer
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Seyoun Byun
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Philip Coryell
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan D’Costa
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eliza Thulson
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - HyunAh Kim
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sylvie M Parkus
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Marielle L Bond
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Emma R Klein
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susanna Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michael I Love
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27695, USA
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Douglas H Phanstiel
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Takasugi M, Nonaka Y, Takemura K, Yoshida Y, Stein F, Schwarz JJ, Adachi J, Satoh J, Ito S, Tombline G, Biashad SA, Seluanov A, Gorbunova V, Ohtani N. An atlas of the aging mouse proteome reveals the features of age-related post-transcriptional dysregulation. Nat Commun 2024; 15:8520. [PMID: 39353907 PMCID: PMC11445428 DOI: 10.1038/s41467-024-52845-x] [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: 07/03/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
To what extent and how post-transcriptional dysregulation affects aging proteome remains unclear. Here, we provide proteomic data of whole-tissue lysates (WTL) and low-solubility protein-enriched fractions (LSF) of major tissues collected from mice of 6, 15, 24, and 30 months of age. Low-solubility proteins are preferentially affected by age and the analysis of LSF doubles the number of proteins identified to be differentially expressed with age. Simultaneous analysis of proteome and transcriptome using the same tissue homogenates reveals the features of age-related post-transcriptional dysregulation. Post-transcriptional dysregulation becomes evident especially after 24 months of age and age-related post-transcriptional dysregulation leads to accumulation of core matrisome proteins and reduction of mitochondrial membrane proteins in multiple tissues. Based on our in-depth proteomic data and sample-matched transcriptome data of adult, middle-aged, old, and geriatric mice, we construct the Mouse aging proteomic atlas ( https://aging-proteomics.info/ ), which provides a thorough and integrative view of age-related gene expression changes.
Collapse
Affiliation(s)
- Masaki Takasugi
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan.
| | - Yoshiki Nonaka
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Kazuaki Takemura
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Yuya Yoshida
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Frank Stein
- Proteomic Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | | | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Junko Satoh
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Gregory Tombline
- Department of Biology, University of Rochester, Rochester, NY, USA
| | | | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Naoko Ohtani
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
7
|
Hatefi-Shogae S, Emadi-Baygi M, Ghaedi-Heydari R. Analysis of Human Papillomavirus-Associated Cervical Cancer Differentially Expressed Genes and Identification of Prognostic Factors using Integrated Bioinformatics Approaches. Adv Biomed Res 2024; 13:78. [PMID: 39512411 PMCID: PMC11542694 DOI: 10.4103/abr.abr_338_23] [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: 09/05/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 11/15/2024] Open
Abstract
Background Human papillomavirus (HPV)-induced cervical cancer progresses through a series of steps. Despite our limited understanding of the mechanisms driving this progression, identifying the key genes involved could significantly improve early detection and treatment. Materials and Methods Two gene expression profiles of GSE9750 and GSE6791, which included cervical cancer HPV-positive and -negative samples, were evaluated using the R limma package with established cut-off criteria of P value < 0.05 and | fold change| ≥ 1. KEGG pathway enrichment was performed to identify potential pathways. Weighted gene co-expression network analysis (WGCNA) was used to discover co-expressed gene modules and trait-module connections. Results Considering the defined criteria, 115 differentially expressed genes (DEGs) were identified. The DEG's KEGG pathway enrichment analysis revealed enrichment in highly relevant pathways to the HPV infection, including cell cycle, viral carcinogenesis, autophagy-animal, Epstein-Barr virus infection, human T-cell leukemia virus 1 infection, and microRNAs in cancer. WGCNA results in 13 co-expression modules, and the magenta module is identified with significant relations to HPV, cervical cancer stage, and metastasis traits. The survival analysis identified BEX1 and CDC45 as potential prognostic factors in HPV-associated cervical cancer. Conclusion The innovation of our work lies in identifying essential genes associated with the multi-step process of cervical carcinogenesis. In fact, the current study has the potential to give a distinct viewpoint on the molecular pathways linked to cervical cancer. Considering the potential importance of the hub genes, we recommend conducting in-depth wet lab research to determine their impact on the biological mechanisms of cervical cancer.
Collapse
Affiliation(s)
- Saba Hatefi-Shogae
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Modjtaba Emadi-Baygi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Rasoul Ghaedi-Heydari
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
8
|
Gómez-Pascual A, Rocamora-Pérez G, Ibanez L, Botía JA. Targeted co-expression networks for the study of traits. Sci Rep 2024; 14:16675. [PMID: 39030261 PMCID: PMC11271532 DOI: 10.1038/s41598-024-67329-7] [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: 12/28/2023] [Accepted: 07/10/2024] [Indexed: 07/21/2024] Open
Abstract
Weighted Gene Co-expression Network Analysis (WGCNA) is a widely used approach for the generation of gene co-expression networks. However, networks generated with this tool usually create large modules with a large set of functional annotations hard to decipher. We have developed TGCN, a new method to create Targeted Gene Co-expression Networks. This method identifies the transcripts that best predict the trait of interest based on gene expression using a refinement of the LASSO regression. Then, it builds the co-expression modules around those transcripts. Algorithm properties were characterized using the expression of 13 brain regions from the Genotype-Tissue Expression project. When comparing our method with WGCNA, TGCN networks lead to more precise modules that have more specific and yet rich biological meaning. Then, we illustrate its applicability by creating an APP-TGCN on The Religious Orders Study and Memory and Aging Project dataset, aiming to identify the molecular pathways specifically associated with APP role in Alzheimer's disease. Main biological findings were further validated in two independent cohorts. In conclusion, we provide a new framework that serves to create targeted networks that are smaller, biologically relevant and useful in high throughput hypothesis driven research. The TGCN R package is available on Github: https://github.com/aliciagp/TGCN .
Collapse
Affiliation(s)
- A Gómez-Pascual
- Communications Engineering and Information Department, University of Murcia, 30100, Murcia, Spain
| | - G Rocamora-Pérez
- Department of Genetics and Genomic Medicine Research and Teaching, UCL GOS Institute of Child Health, London, WC1N 1EH, UK
| | - L Ibanez
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - J A Botía
- Communications Engineering and Information Department, University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
9
|
Lorenzetti WR, Ibelli AMG, Peixoto JDO, Savoldi IR, Mores MAZ, de Souza Romano G, do Carmo KB, Ledur MC. The downregulation of genes encoding muscle proteins have a potential role in the development of scrotal hernia in pigs. Mol Biol Rep 2024; 51:822. [PMID: 39023774 DOI: 10.1007/s11033-024-09766-1] [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: 02/15/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Testicular descent is a physiological process regulated by many factors. Eventually, disturbances in the embryological/fetal development path facilitate the occurrence of scrotal hernia, a congenital malformation characterized by the presence of intestinal portions within the scrotal sac due to the abnormal expansion of the inguinal ring. In pigs, some genes have been related to this anomaly, but the genetic mechanisms involved remain unclear. This study aimed to investigate the expression profile of a set of genes potentially involved with the manifestation of scrotal hernia in the inguinal ring tissue. METHODS AND RESULTS Tissue samples from the inguinal ring/canal of normal and scrotal hernia-affected male pigs with approximately 30 days of age were used. Relative expression analysis was performed using qPCR to confirm the expression profile of 17 candidate genes previously identified in an RNA-Seq study. Among them, the Myosin heavy chain 1 (MYH1), Desmin (DES), and Troponin 1 (TNNI1) genes were differentially expressed between groups and had reduced levels of expression in the affected animals. These genes encode proteins involved in the formation of muscle tissue, which seems to be important for increasing the resistance of the inguinal ring to the abdominal pressure, which is essential to avoid the occurrence of scrotal hernia. CONCLUSIONS The downregulation of muscular candidate genes in the inguinal tissue clarifies the genetic mechanisms involved with this anomaly in its primary site, providing useful information for developing strategies to control this malformation in pigs and other mammals.
Collapse
Affiliation(s)
- William Raphael Lorenzetti
- Programa de Pós-graduação em Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, Rua Beloni Trombeta Zanin 680E, Chapecó, Santa Catarina, 89815-630, Brazil
| | - Adriana Mércia Guaratini Ibelli
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838, Guarapuava, Paraná, 85040-167, Brazil
- Embrapa Pecuária Sudeste, Rodovia Washington Luiz, Km 234, São Carlos, São Paulo, 13560-970, Brazil
| | - Jane de Oliveira Peixoto
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838, Guarapuava, Paraná, 85040-167, Brazil
| | - Igor Ricardo Savoldi
- Programa de Pós-graduação em Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, Rua Beloni Trombeta Zanin 680E, Chapecó, Santa Catarina, 89815-630, Brazil
- Laudo laboratório Avícola, Rodovia BR-365, Morumbi, Uberlândia, Minas Gerais, 38407180, Brazil
| | - Marcos Antônio Zanella Mores
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil
| | | | - Kamilla Bleil do Carmo
- Universidade do Contestado, Concórdia, Santa Catarina, Brazil
- Instituto Federal Catarinense, Rodovia SC 283, km 17, Concórdia, Santa Catarina, 89703-720, Brazil
| | - Mônica Corrêa Ledur
- Programa de Pós-graduação em Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, Rua Beloni Trombeta Zanin 680E, Chapecó, Santa Catarina, 89815-630, Brazil.
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil.
| |
Collapse
|
10
|
Rafikova E, Nemirovich-Danchenko N, Ogmen A, Parfenenkova A, Velikanova A, Tikhonov S, Peshkin L, Rafikov K, Spiridonova O, Belova Y, Glinin T, Egorova A, Batin M. Open Genes-a new comprehensive database of human genes associated with aging and longevity. Nucleic Acids Res 2024; 52:D950-D962. [PMID: 37665017 PMCID: PMC10768108 DOI: 10.1093/nar/gkad712] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/31/2023] [Accepted: 08/19/2023] [Indexed: 09/05/2023] Open
Abstract
The Open Genes database was created to enhance and simplify the search for potential aging therapy targets. We collected data on 2402 genes associated with aging and developed convenient tools for searching and comparing gene features. A comprehensive description of genes has been provided, including lifespan-extending interventions, age-related changes, longevity associations, gene evolution, associations with diseases and hallmarks of aging, and functions of gene products. For each experiment, we presented the necessary structured data for evaluating the experiment's quality and interpreting the study's findings. Our goal was to stay objective and precise while connecting a particular gene to human aging. We distinguished six types of studies and 12 criteria for adding genes to our database. Genes were classified according to the confidence level of the link between the gene and aging. All the data collected in a database are provided both by an API and a user interface. The database is publicly available on a website at https://open-genes.org/.
Collapse
Affiliation(s)
- Ekaterina Rafikova
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
| | - Nikolay Nemirovich-Danchenko
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
- Faculty of Cytology and Genetics, National Research Tomsk State University, Tomsk 634050, Russia
| | - Anna Ogmen
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul 34342, Turkey
| | - Anna Parfenenkova
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
| | | | - Stanislav Tikhonov
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow 119991, Russia
| | - Leonid Peshkin
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Konstantin Rafikov
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
| | - Olga Spiridonova
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
| | - Yulia Belova
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
| | - Timofey Glinin
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
- Endocrine Neoplasia Laboratory, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Genetics & Biotechnology, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Anastasia Egorova
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
| | - Mikhail Batin
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
| |
Collapse
|
11
|
Scarpa J. Improving liver transplant outcomes with transplant-omics and network biology. Curr Opin Organ Transplant 2023; 28:412-418. [PMID: 37706301 DOI: 10.1097/mot.0000000000001100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
PURPOSE OF REVIEW Molecular omics data is increasingly ubiquitous throughout medicine. In organ transplantation, recent large-scale research efforts are generating the 'transplant-ome' - the entire set of molecular omics data, including the genome, transcriptome, proteome, and metabolome. Importantly, early studies in anesthesiology have demonstrated how perioperative interventions alter molecular profiles in various patient populations. The next step for anesthesiologists and intensivists will be to tailor perioperative care to the transplant-ome of individual liver transplant patients. RECENT FINDINGS In liver transplant patients, elements of the transplant-ome predict complications and point to novel interventions. Importantly, molecular profiles of both the donor organ and recipient contribute to this risk, and interventions like normothermic machine perfusion influence these profiles. As we can now measure various omics molecules simultaneously, we can begin to understand how these molecules interact to form molecular networks and emerging technologies offer noninvasive and continuous ways to measure these networks throughout the perioperative period. Molecules that regulate these networks are likely mediators of complications and actionable clinical targets throughout the perioperative period. SUMMARY The transplant-ome can be used to tailor perioperative care to the individual liver transplant patient. Monitoring molecular networks continuously and noninvasively would provide new opportunities to optimize perioperative management.
Collapse
Affiliation(s)
- Joseph Scarpa
- Department of Anesthesiology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
12
|
Newman LE, Testard C, DeCasien AR, Chiou KL, Watowich MM, Janiak MC, Pavez-Fox MA, Sanchez Rosado MR, Cooper EB, Costa CE, Petersen RM, Montague MJ, Platt ML, Brent LJN, Snyder-Mackler N, Higham JP. The biology of aging in a social world: Insights from free-ranging rhesus macaques. Neurosci Biobehav Rev 2023; 154:105424. [PMID: 37827475 PMCID: PMC10872885 DOI: 10.1016/j.neubiorev.2023.105424] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Social adversity can increase the age-associated risk of disease and death, yet the biological mechanisms that link social adversities to aging remain poorly understood. Long-term naturalistic studies of nonhuman animals are crucial for integrating observations of social behavior throughout an individual's life with detailed anatomical, physiological, and molecular measurements. Here, we synthesize the body of research from one such naturalistic study system, Cayo Santiago, which is home to the world's longest continuously monitored free-ranging population of rhesus macaques (Macaca mulatta). We review recent studies of age-related variation in morphology, gene regulation, microbiome composition, and immune function. We also discuss ecological and social modifiers of age-markers in this population. In particular, we summarize how a major natural disaster, Hurricane Maria, affected rhesus macaque physiology and social structure and highlight the context-dependent and domain-specific nature of aging modifiers. Finally, we conclude by providing directions for future study, on Cayo Santiago and elsewhere, that will further our understanding of aging across different domains and how social adversity modifies aging processes.
Collapse
Affiliation(s)
- Laura E Newman
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA.
| | - Camille Testard
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA.
| | - Alex R DeCasien
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Kenneth L Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Marina M Watowich
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA; Department of Biology, University of Washington, Seattle, WA, USA
| | - Mareike C Janiak
- Department of Anthropology, New York University, New York, NY, USA
| | - Melissa A Pavez-Fox
- Centre for Research in Animal Behaviour, University of Exeter, United Kingdom
| | | | - Eve B Cooper
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA
| | - Christina E Costa
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA
| | - Rachel M Petersen
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA; Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren J N Brent
- Centre for Research in Animal Behaviour, University of Exeter, United Kingdom
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - James P Higham
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA
| |
Collapse
|
13
|
Kovalskaia VA, Cherevatova TB, Polyakov AV, Ryzhkova OP. Molecular basis and genetics of hypohidrotic ectodermal dysplasias. Vavilovskii Zhurnal Genet Selektsii 2023; 27:676-683. [PMID: 38023809 PMCID: PMC10643535 DOI: 10.18699/vjgb-23-78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/06/2023] [Accepted: 03/24/2023] [Indexed: 12/01/2023] Open
Abstract
Ectodermal dysplasia (ED) is a heterogeneous group of hereditary diseases of the skin and its appendages, which are characterized by impaired development and/or homeostasis of two or more ectoderm derivatives, including: hair, teeth, nails, sweat glands and their modifications (mammary glands, for instance). The overall prevalence of ectodermal dysplasia remains precisely unknown not only in Russia, but also in the world, nor is known the contribution of individual genes to its structure. This complicates the DNA diagnosis establishment of this disease due to the lack of an accurate diagnostic algorithm and a universal cost-effective method of analysis. To date, the most highly-researched genes involved in the development of anhydrous or hypohidrotic forms of ED are EDA, EDAR, EDARADD and WNT10A. The ectodysplasin A (EDA) gene is the cause of the most common X-linked form of ED, a gene from the Wnt family (WNT10A) is responsible for the autosomal recessive form of the disease, and two other genes (EDAR and EDARADD) can cause both autosomal recessive and autosomal dominant forms. This review provides the characteristics of the genes involved in ED, their mutation spectra, the level of their expression in human tissues, as well as the interrelation of the aforementioned genes. The domain structures of the corresponding proteins are considered, as well as the molecular genetic pathways in which they are involved. Animal models for studying this disorder are also taken into consideration. Due to the cross-species genes conservation, their mutations cause the disruption of the development of ectoderm derivatives not only in humans, but also in mice, cows, dogs, and even fish. It can be exploited for a better understanding of the etiopathogenesis of ectodermal dysplasias. Moreover, this article brings up the possibility of recurrent mutations in the EDA and WNT10A genes. The review also presents data on promising approaches for intrauterine ED treatment.
Collapse
Affiliation(s)
| | | | - A V Polyakov
- Research Centre for Medical Genetics, Moscow, Russia
| | - O P Ryzhkova
- Research Centre for Medical Genetics, Moscow, Russia
| |
Collapse
|
14
|
Salignon J, Faridani OR, Miliotis T, Janssens GE, Chen P, Zarrouki B, Sandberg R, Davidsson P, Riedel CG. Age prediction from human blood plasma using proteomic and small RNA data: a comparative analysis. Aging (Albany NY) 2023; 15:5240-5265. [PMID: 37341993 PMCID: PMC10333066 DOI: 10.18632/aging.204787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 05/26/2023] [Indexed: 06/22/2023]
Abstract
Aging clocks, built from comprehensive molecular data, have emerged as promising tools in medicine, forensics, and ecological research. However, few studies have compared the suitability of different molecular data types to predict age in the same cohort and whether combining them would improve predictions. Here, we explored this at the level of proteins and small RNAs in 103 human blood plasma samples. First, we used a two-step mass spectrometry approach measuring 612 proteins to select and quantify 21 proteins that changed in abundance with age. Notably, proteins increasing with age were enriched for components of the complement system. Next, we used small RNA sequencing to select and quantify a set of 315 small RNAs that changed in abundance with age. Most of these were microRNAs (miRNAs), downregulated with age, and predicted to target genes related to growth, cancer, and senescence. Finally, we used the collected data to build age-predictive models. Among the different types of molecules, proteins yielded the most accurate model (R² = 0.59 ± 0.02), followed by miRNAs as the best-performing class of small RNAs (R² = 0.54 ± 0.02). Interestingly, the use of protein and miRNA data together improved predictions (R2 = 0.70 ± 0.01). Future work using larger sample sizes and a validation dataset will be necessary to confirm these results. Nevertheless, our study suggests that combining proteomic and miRNA data yields superior age predictions, possibly by capturing a broader range of age-related physiological changes. It will be interesting to determine if combining different molecular data types works as a general strategy to improve future aging clocks.
Collapse
Affiliation(s)
- Jérôme Salignon
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Huddinge 14157, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 14157, Sweden
| | - Omid R. Faridani
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Huddinge 14157, Sweden
- Lowy Cancer Research Centre, School of Medical Sciences, University of New South Wales, Sydney, Australia
- Garvan Institute of Medical Research, Sydney, Australia
| | - Tasso Miliotis
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Georges E. Janssens
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Huddinge 14157, Sweden
| | - Ping Chen
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Huddinge 14157, Sweden
| | - Bader Zarrouki
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rickard Sandberg
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Huddinge 14157, Sweden
- Department of Cellular and Molecular Biology, Ludwig Institute for Cancer Research, Karolinska Institutet, Solna 17165, Sweden
| | - Pia Davidsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christian G. Riedel
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Huddinge 14157, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 14157, Sweden
| |
Collapse
|
15
|
Nehlin JO. Senolytic and senomorphic interventions to defy senescence-associated mitochondrial dysfunction. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:217-247. [PMID: 37437979 DOI: 10.1016/bs.apcsb.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The accumulation of senescent cells in the aging individual is associated with an increase in the occurrence of age-associated pathologies that contribute to poor health, frailty, and mortality. The number and type of senescent cells is viewed as a contributor to the body's senescence burden. Cellular models of senescence are based on induction of senescence in cultured cells in the laboratory. One type of senescence is triggered by mitochondrial dysfunction. There are several indications that mitochondria defects contribute to body aging. Senotherapeutics, targeting senescent cells, have been shown to induce their lysis by means of senolytics, or repress expression of their secretome, by means of senomorphics, senostatics or gerosuppressors. An outline of the mechanism of action of various senotherapeutics targeting mitochondria and senescence-associated mitochondria dysfunction will be here addressed. The combination of geroprotective interventions together with senotherapeutics will help to strengthen mitochondrial energy metabolism, biogenesis and turnover, and lengthen the mitochondria healthspan, minimizing one of several molecular pathways contributing to the aging phenotype.
Collapse
Affiliation(s)
- Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital, Amager and Hvidovre, Hvidovre, Denmark.
| |
Collapse
|
16
|
De Man R, McDonough JE, Adams TS, Manning EP, Myers G, Vos R, Ceulemans L, Dupont L, Vanaudenaerde BM, Wuyts WA, Rosas IO, Hagood JS, Ambalavanan N, Niklason L, Hansen KC, Yan X, Kaminski N. A Multi-omic Analysis of the Human Lung Reveals Distinct Cell Specific Aging and Senescence Molecular Programs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.536722. [PMID: 37131739 PMCID: PMC10153177 DOI: 10.1101/2023.04.19.536722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Age is a major risk factor for lung disease. To understand the mechanisms underlying this association, we characterized the changing cellular, genomic, transcriptional, and epigenetic landscape of lung aging using bulk and single-cell RNAseq (scRNAseq) data. Our analysis revealed age-associated gene networks that reflected hallmarks of aging, including mitochondrial dysfunction, inflammation, and cellular senescence. Cell type deconvolution revealed age-associated changes in the cellular composition of the lung: decreased alveolar epithelial cells and increased fibroblasts and endothelial cells. In the alveolar microenvironment, aging is characterized by decreased AT2B cells and reduced surfactant production, a finding that was validated by scRNAseq and IHC. We showed that a previously reported senescence signature, SenMayo, captures cells expressing canonical senescence markers. SenMayo signature also identified cell-type specific senescence-associated co-expression modules that have distinct molecular functions, including ECM regulation, cell signaling, and damage response pathways. Analysis of somatic mutations showed that burden was highest in lymphocytes and endothelial cells and was associated with high expression of senescence signature. Finally, aging and senescence gene expression modules were associated with differentially methylated regions, with inflammatory markers such as IL1B, IL6R, and TNF being significantly regulated with age. Our findings provide new insights into the mechanisms underlying lung aging and may have implications for the development of interventions to prevent or treat age-related lung diseases.
Collapse
Affiliation(s)
- Ruben De Man
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - John E McDonough
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Taylor S Adams
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Edward P Manning
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Greg Myers
- Department of Pediatrics (Division of Pulmonology) and Marsico Lung Institute, University of North Carolina at Chapel Hill
| | - Robin Vos
- Department of Respiratory Medicine, KU Leuven, Leuven, Belgium
| | | | - Lieven Dupont
- Department of Respiratory Medicine, KU Leuven, Leuven, Belgium
| | | | - Wim A Wuyts
- Department of Respiratory Medicine, KU Leuven, Leuven, Belgium
| | - Ivan O Rosas
- Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - James S. Hagood
- Department of Pediatrics (Division of Pulmonology) and Marsico Lung Institute, University of North Carolina at Chapel Hill
| | | | - Laura Niklason
- Department of Anesthesiology, Yale School of Medicine; and Humacyte Global Inc
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Xiting Yan
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
17
|
Newman LE, Testard C, DeCasien AR, Chiou KL, Watowich MM, Janiak MC, Pavez-Fox MA, Rosado MRS, Cooper EB, Costa CE, Petersen RM, Montague MJ, Platt ML, Brent LJ, Snyder-Mackler N, Higham JP. The biology of aging in a social world:insights from free-ranging rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.28.525893. [PMID: 36747827 PMCID: PMC9900930 DOI: 10.1101/2023.01.28.525893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Social adversity can increase the age-associated risk of disease and death, yet the biological mechanisms that link social adversities to aging remain poorly understood. Long-term naturalistic studies of nonhuman animals are crucial for integrating observations of social behavior throughout an individual's life with detailed anatomical, physiological, and molecular measurements. Here, we synthesize the body of research from one such naturalistic study system, Cayo Santiago Island, which is home to the world's longest continuously monitored free-ranging population of rhesus macaques. We review recent studies of age-related variation in morphology, gene regulation, microbiome composition, and immune function. We also discuss ecological and social modifiers of age-markers in this population. In particular, we summarize how a major natural disaster, Hurricane Maria, affected rhesus macaque physiology and social structure and highlight the context-dependent and domain-specific nature of aging modifiers. Finally, we conclude by providing directions for future study, on Cayo Santiago and elsewhere, that will further our understanding of aging across different domains and how social adversity modifies aging processes.
Collapse
Affiliation(s)
- Laura E. Newman
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| | - Camille Testard
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex R. DeCasien
- Section on Developmental Neurogenomics, National Institutes of Mental Health, Bethesda, Maryland, USA
| | - Kenneth L. Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Marina M. Watowich
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Mareike C. Janiak
- Department of Anthropology, New York University, New York, New York, USA
| | | | | | - Eve B. Cooper
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| | - Christina E. Costa
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| | - Rachel M. Petersen
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Michael J. Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren J.N. Brent
- Centre for Research in Animal Behaviour, University of Exeter, United Kingdom
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - James P. Higham
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| |
Collapse
|
18
|
Stevanovic M, Lazic A, Schwirtlich M, Stanisavljevic Ninkovic D. The Role of SOX Transcription Factors in Ageing and Age-Related Diseases. Int J Mol Sci 2023; 24:851. [PMID: 36614288 PMCID: PMC9821406 DOI: 10.3390/ijms24010851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
The quest for eternal youth and immortality is as old as humankind. Ageing is an inevitable physiological process accompanied by many functional declines that are driving factors for age-related diseases. Stem cell exhaustion is one of the major hallmarks of ageing. The SOX transcription factors play well-known roles in self-renewal and differentiation of both embryonic and adult stem cells. As a consequence of ageing, the repertoire of adult stem cells present in various organs steadily declines, and their dysfunction/death could lead to reduced regenerative potential and development of age-related diseases. Thus, restoring the function of aged stem cells, inducing their regenerative potential, and slowing down the ageing process are critical for improving the health span and, consequently, the lifespan of humans. Reprograming factors, including SOX family members, emerge as crucial players in rejuvenation. This review focuses on the roles of SOX transcription factors in stem cell exhaustion and age-related diseases, including neurodegenerative diseases, visual deterioration, chronic obstructive pulmonary disease, osteoporosis, and age-related cancers. A better understanding of the molecular mechanisms of ageing and the roles of SOX transcription factors in this process could open new avenues for developing novel strategies that will delay ageing and prevent age-related diseases.
Collapse
Affiliation(s)
- Milena Stevanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11158 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11000 Belgrade, Serbia
| | - Andrijana Lazic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | - Marija Schwirtlich
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | | |
Collapse
|
19
|
Sturm G, Monzel AS, Karan KR, Michelson J, Ware SA, Cardenas A, Lin J, Bris C, Santhanam B, Murphy MP, Levine ME, Horvath S, Belsky DW, Wang S, Procaccio V, Kaufman BA, Hirano M, Picard M. A multi-omics longitudinal aging dataset in primary human fibroblasts with mitochondrial perturbations. Sci Data 2022; 9:751. [PMID: 36463290 PMCID: PMC9719499 DOI: 10.1038/s41597-022-01852-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Aging is a process of progressive change. To develop biological models of aging, longitudinal datasets with high temporal resolution are needed. Here we report a multi-omics longitudinal dataset for cultured primary human fibroblasts measured across their replicative lifespans. Fibroblasts were sourced from both healthy donors (n = 6) and individuals with lifespan-shortening mitochondrial disease (n = 3). The dataset includes cytological, bioenergetic, DNA methylation, gene expression, secreted proteins, mitochondrial DNA copy number and mutations, cell-free DNA, telomere length, and whole-genome sequencing data. This dataset enables the bridging of mechanistic processes of aging as outlined by the "hallmarks of aging", with the descriptive characterization of aging such as epigenetic age clocks. Here we focus on bridging the gap for the hallmark mitochondrial metabolism. Our dataset includes measurement of healthy cells, and cells subjected to over a dozen experimental manipulations targeting oxidative phosphorylation (OxPhos), glycolysis, and glucocorticoid signaling, among others. These experiments provide opportunities to test how cellular energetics affect the biology of cellular aging. All data are publicly available at our webtool: https://columbia-picard.shinyapps.io/shinyapp-Lifespan_Study/.
Collapse
Affiliation(s)
- Gabriel Sturm
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Anna S Monzel
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Kalpita R Karan
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeremy Michelson
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah A Ware
- University of Pittsburgh, School of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine and Vascular Medicine Institute, Pittsburgh, PA, USA
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, 94305, USA
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Céline Bris
- UMR CNRS 6015, INSERM U1083, MITOVASC, SFR ICAT, Université d'Angers, Angers, F-49000, France
- Department of Genetics, CHU Angers, Angers, F-49000, France
| | - Balaji Santhanam
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Michael P Murphy
- MRC-Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Morgan E Levine
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
- Altos Labs, San Diego, USA
| | - Steve Horvath
- Altos Labs, San Diego, USA
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Daniel W Belsky
- Department of Epidemiology & Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Shuang Wang
- Department of Biostatistics, Columbia University Irving Medical Center, New York, NY, USA
| | - Vincent Procaccio
- UMR CNRS 6015, INSERM U1083, MITOVASC, SFR ICAT, Université d'Angers, Angers, F-49000, France
- Department of Genetics, CHU Angers, Angers, F-49000, France
| | - Brett A Kaufman
- University of Pittsburgh, School of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine and Vascular Medicine Institute, Pittsburgh, PA, USA
| | - Michio Hirano
- Merritt Center and Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.
- Merritt Center and Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
20
|
Brinkley TE, Justice JN, Basu S, Bauer SR, Loh KP, Mukli P, Ng TKS, Turney IC, Ferrucci L, Cummings SR, Kritchevsky SB. Research priorities for measuring biologic age: summary and future directions from the Research Centers Collaborative Network Workshop. GeroScience 2022; 44:2573-2583. [PMID: 36242692 PMCID: PMC9768050 DOI: 10.1007/s11357-022-00661-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023] Open
Abstract
Biologic aging reflects the genetic, molecular, and cellular changes underlying the development of morbidity and mortality with advancing chronological age. As several potential mechanisms have been identified, there is a growing interest in developing robust measures of biologic age that can better reflect the underlying biology of aging and predict age-related outcomes. To support this endeavor, the Research Centers Collaborative Network (RCCN) conducted a workshop in January 2022 to discuss emerging concepts in the field and identify opportunities to move the science forward. This paper presents workshop proceedings and summarizes the identified research needs, priorities, and recommendations for measuring biologic age. The highest priorities identified were the need for more robust measures, longitudinal studies, multidisciplinary collaborations, and translational approaches.
Collapse
Affiliation(s)
- Tina E Brinkley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Jamie N Justice
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shubhashrita Basu
- Center for Demography of Health and Aging, University of Wisconsin, Madison, WI, USA
| | - Scott R Bauer
- Departments of Medicine and Urology, University of California and San Francisco VA Medical Center, San Francisco, CA, USA
| | - Kah Poh Loh
- Division of Hematology/Oncology, Department of Medicine, University of Rochester Medical Center, James P. Wilmot Cancer Institute, Rochester, NY, USA
| | - Peter Mukli
- Department of Biochemistry/Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ted Kheng Siang Ng
- Edson College of Nursing and Health Innovation, Arizona State University, Tempe, AZ, USA
| | - Indira C Turney
- Department of Neurology, Columbia University Medical Center, New York City, NY, USA
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute On Aging, NIH, Baltimore, MD, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute and the Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
21
|
He B, Wang K, Xiang J, Bing P, Tang M, Tian G, Guo C, Xu M, Yang J. DGHNE: network enhancement-based method in identifying disease-causing genes through a heterogeneous biomedical network. Brief Bioinform 2022; 23:6712302. [PMID: 36151744 DOI: 10.1093/bib/bbac405] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/01/2022] [Accepted: 08/21/2022] [Indexed: 12/14/2022] Open
Abstract
The identification of disease-causing genes is critical for mechanistic understanding of disease etiology and clinical manipulation in disease prevention and treatment. Yet the existing approaches in tackling this question are inadequate in accuracy and efficiency, demanding computational methods with higher identification power. Here, we proposed a new method called DGHNE to identify disease-causing genes through a heterogeneous biomedical network empowered by network enhancement. First, a disease-disease association network was constructed by the cosine similarity scores between phenotype annotation vectors of diseases, and a new heterogeneous biomedical network was constructed by using disease-gene associations to connect the disease-disease network and gene-gene network. Then, the heterogeneous biomedical network was further enhanced by using network embedding based on the Gaussian random projection. Finally, network propagation was used to identify candidate genes in the enhanced network. We applied DGHNE together with five other methods into the most updated disease-gene association database termed DisGeNet. Compared with all other methods, DGHNE displayed the highest area under the receiver operating characteristic curve and the precision-recall curve, as well as the highest precision and recall, in both the global 5-fold cross-validation and predicting new disease-gene associations. We further performed DGHNE in identifying the candidate causal genes of Parkinson's disease and diabetes mellitus, and the genes connecting hyperglycemia and diabetes mellitus. In all cases, the predicted causing genes were enriched in disease-associated gene ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways, and the gene-disease associations were highly evidenced by independent experimental studies.
Collapse
Affiliation(s)
- Binsheng He
- Academician Workstation, Changsha Medical University, Changsha 410219, China.,Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, P. R. China.,School of pharmacy, Changsha Medical University, Changsha 410219, P. R. China
| | - Kun Wang
- School of Mathematical Sciences, Ocean University of China, Qingdao 266100, China
| | - Ju Xiang
- Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Pingping Bing
- Academician Workstation, Changsha Medical University, Changsha 410219, China.,Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, P. R. China.,School of pharmacy, Changsha Medical University, Changsha 410219, P. R. China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang 212001, Jiangsu, China
| | - Geng Tian
- Geneis (Beijing) Co., Ltd., Beijing 100102, China
| | - Cheng Guo
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Miao Xu
- Broad institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Jialiang Yang
- Academician Workstation, Changsha Medical University, Changsha 410219, China.,Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, P. R. China.,School of pharmacy, Changsha Medical University, Changsha 410219, P. R. China.,Geneis (Beijing) Co., Ltd., Beijing 100102, China
| |
Collapse
|
22
|
Tong X, Li WX, Liang J, Zheng Y, Dai SX. Two different aging paths in human blood revealed by integrated analysis of gene Expression, mutation and alternative splicing. Gene 2022; 829:146501. [PMID: 35452709 DOI: 10.1016/j.gene.2022.146501] [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: 01/16/2022] [Revised: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 11/04/2022]
Abstract
Aging is a complex life process that human organs and tissues steadily and continuously decline. Aging has huge heterogeneity, which shows different aging rates among different individuals and in different tissues of the same individual. Many studies of aging are often contradictory and show little common signature. The integrated analysis of these transcriptome datasets will provide an unbiased global view of the aging process. Here, we integrated 8 transcriptome datasets including 757 samples from healthy human blood to study aging from three aspects of gene expression, mutations, and alternative splicing. Surprisingly, we found that transcriptome changes in blood are relatively independent of the chronological age. Further pseudotime analysis revealed two different aging paths (AgingPath1 and AgingPath2) in human blood. The differentially expressed genes (DEGs) along the two paths showed a limited overlap and are enriched in different biological processes. The mutations of DEGs in AgingPath1 are significantly increased in the aging process, while the opposite trend was observed in AgingPath2. Expression quantitative trait loci (eQTL) and splicing quantitative trait loci (sQTL) analysis identified 304 important mutations that can affect both gene expression and alternative splicing during aging. Finally, by comparison between aging and Alzheimer's disease, we identified 37 common DEGs in AgingPath1, AgingPath2 and Alzheimer's disease. These genes may contribute to the shift from aging state to Alzheimer's disease. In summary, this study revealed the two aging paths and the related genes and mutations, which provides a new insight into aging and aging-related disease.
Collapse
Affiliation(s)
- Xin Tong
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Wen-Xing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jihao Liang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yang Zheng
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Shao-Xing Dai
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China.
| |
Collapse
|
23
|
Informative SNP Selection Based on a Fuzzy Clustering and Improved Binary Particle Swarm Optimization Algorithm. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3837579. [PMID: 35756402 PMCID: PMC9225903 DOI: 10.1155/2022/3837579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/14/2022] [Accepted: 04/30/2022] [Indexed: 12/04/2022]
Abstract
Single-nucleotide polymorphism (SNP) involves the replacement of a single nucleotide in a deoxyribonucleic acid (DNA) sequence and is often linked to the development of specific diseases. Although current genotyping methods can tag SNP loci within biological samples to provide accurate genetic information for a disease associated, they have limited prediction accuracy. Furthermore, they are complex to perform and may result in the prediction of an excessive number of tag SNP loci, which may not always be associated with the disease. Therefore in this manuscript, we aimed to evaluate the impact of a newly optimized fuzzy clustering and binary particle swarm optimization algorithm (FCBPSO) on the accuracy and running time of informative SNP selection. Fuzzy clustering and FCBPSO were first applied to identify the equivalence relation and the candidate tag SNP set to reduce the redundancy between loci. The FCBPSO algorithm was then optimized and used to obtain the final tag SNP set. The prediction performance and running time of the newly developed model were compared with other traditional methods, including NMC, SPSO, and MCMR. The prediction accuracy of the FCBPSO algorithm was always higher than that of the other algorithms especially as the number of tag SNPs increased. However, when the number of tag SNPs was low, the prediction accuracy of FCBPSO was slightly lower than that of MCMR (add prediction accuracy values for each algorithm). However, the running time of the FCBPSO algorithm was always lower than that of MCMR. FCBPSO not only reduced the size and dimension of the optimization problem but also simplified the training of the prediction model. This improved the prediction accuracy of the model and reduced the running time when compared with other traditional methods.
Collapse
|
24
|
Liu CL, Chien MN, Hsu YC, Cheng SP. Transcriptomic Characteristics Associated With Aging in the Thyroid Gland. Front Nutr 2022; 9:859702. [PMID: 35694164 PMCID: PMC9174607 DOI: 10.3389/fnut.2022.859702] [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: 01/21/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022] Open
Abstract
The aging thyroid is associated with a plethora of morphological and functional changes. Limited studies have addressed the gene expression signature in the aging thyroid, except for sporadic reports using data from postmortem samples in the Genotype-Tissue Expression (GTEx) project. In this investigation, we analyzed the RNA sequencing data of 58 samples of normal-appearing counterpart thyroid tissues from The Cancer Genome Atlas. Aging-correlated genes were identified by determining the Spearman rank-order correlation between patient age and gene expression level. Additionally, we performed gene set enrichment analysis and conducted a weighted correlation network analysis. The results were compared with those analyzed using the GTEx data. The over-represented protein class of aging-correlated genes is mainly metabolite interconversion enzymes. Our analyses identified alterations in immune and inflammatory responses, mitochondrial functions, cytoskeletal proteins, as well as amino acid and cytochrome P450 metabolism. There was no significant association between thyroid differentiation and age. Our findings may shed molecular light on thyroid disorders in the geriatric population.
Collapse
Affiliation(s)
- Chien-Liang Liu
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Ming-Nan Chien
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- *Correspondence: Shih-Ping Cheng
| |
Collapse
|
25
|
Liu J, Lan Y, Tian G, Yang J. A Systematic Framework for Identifying Prognostic Genes in the Tumor Microenvironment of Colon Cancer. Front Oncol 2022; 12:899156. [PMID: 35664768 PMCID: PMC9161737 DOI: 10.3389/fonc.2022.899156] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 12/23/2022] Open
Abstract
As one of the most common cancers of the digestive system, colon cancer is a predominant cause of cancer-related deaths worldwide. To investigate prognostic genes in the tumor microenvironment of colon cancer, we collected 461 colon adenocarcinoma (COAD) and 172 rectal adenocarcinoma (READ) samples from The Cancer Genome Atlas (TCGA) database, and calculated the stromal and immune scores of each sample. We demonstrated that stromal and immune scores were significantly associated with colon cancer stages. By analyzing differentially expressed genes (DEGs) between two stromal and immune score groups, we identified 952 common DEGs. The significantly enriched Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) terms for these DEGs were associated with T-cell activation, immune receptor activity, and cytokine–cytokine receptor interaction. Through univariate Cox regression analysis, we identified 22 prognostic genes. Furthermore, nine key prognostic genes, namely, HOXC8, SRPX, CCL22, CD72, IGLON5, SERPING1, PCOLCE2, FABP4, and ARL4C, were identified using the LASSO Cox regression analysis. The risk score of each sample was calculated using the gene expression of the nine genes. Patients with high-risk scores had a poorer prognosis than those with low-risk scores. The prognostic model established with the nine-gene signature was able to effectively predict the outcome of colon cancer patients. Our findings may help in the clinical decisions and improve the prognosis for colon cancer.
Collapse
Affiliation(s)
- Jinyang Liu
- Department of Sciences, Geneis Beijing Co., Ltd., Beijing, China
- Department of Data Mining,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Yu Lan
- Department of Sciences, Geneis Beijing Co., Ltd., Beijing, China
- Department of Data Mining,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Geng Tian
- Department of Sciences, Geneis Beijing Co., Ltd., Beijing, China
- Department of Data Mining,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Jialiang Yang
- Department of Sciences, Geneis Beijing Co., Ltd., Beijing, China
- Department of Data Mining,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
- PhD Workstation, Chifeng Municipal Hospital, Chifeng, China
- *Correspondence: Jialiang Yang,
| |
Collapse
|
26
|
Exploring the Association between Glutathione Metabolism and Ferroptosis in Osteoblasts with Disuse Osteoporosis and the Key Genes Connecting them. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4914727. [PMID: 35602340 PMCID: PMC9119747 DOI: 10.1155/2022/4914727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022]
Abstract
Disused osteoporosis is a kind of osteoporosis, a common age-related disease. Neurological disorders are major risk factors for osteoporosis. Though there are many studies on disuse osteoporosis, the genetic mechanisms for the association between glutathione metabolism and ferroptosis in osteoblasts with disuse osteoporosis are still unclear. The purpose of this study is to explore the key genes and other related mechanism of ferroptosis and glutathione metabolism in osteoblast differentiation and disuse osteoporosis. By weighted gene coexpression network analysis (WGCNA), the process of osteoblast differentiation-related genes was studied in GSE30393. And the related functional pathways were found through the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. By combining GSE1367 and GSE100933 together, key genes which were separately bound up with glutathione metabolism and ferroptosis were located. The correlation of these key genes was analyzed by the Pearson correlation coefficient. GSTM1 targeted agonist glutathione (GSH) selected by connectivity map (CMap) analysis was used to interfere with the molding disused osteoporosis process in MC3T3-E1 cells. RT-PCR and intracellular reactive oxygen species (ROS) were performed. Two important pathways, glutathione metabolism and ferroptosis pathways, were found. GSTM1 and TFRC were thought as key genes in disuse osteoporosis osteoblasts with the two mechanisms. The two genes have a strong negative correlation. Our experiment results showed that the expression of TFRC was consistent with the negative correlation with the activation process of GSTM1. The strong relationship between the two genes was proved. Glutathione metabolism and ferroptosis are important in the normal differentiation of osteoblasts and the process of disuse osteoporosis. GSTM1 and TFRC were the key genes. The two genes interact with each other, which can be seen as a bridge between the two pathways. The two genes participate in the process of reducing ROS in disuse osteoporosis osteoblasts.
Collapse
|
27
|
Goida J, Pilmane M. The Evaluation of FGFR1, FGFR2 and FOXO1 in Orofacial Cleft Tissue. CHILDREN 2022; 9:children9040516. [PMID: 35455561 PMCID: PMC9032315 DOI: 10.3390/children9040516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/21/2022]
Abstract
Although cleft lip with or without cleft palate (CL/P) is one of the most common congenital anomalies worldwide, the morphopathogenesis of non-syndromic orofacial clefts is still unclear. Many candidate genes have been proposed to play a causal role; however, only a few have been confirmed, leaving many still to be assessed. Taking into account the significance of FGFR1, FGFR2 and FOXO1 in embryogenesis, the aim of this work was to detect and compare the three candidate genes in cleft-affected lip and palatine tissue. Ten soft tissue samples were taken during cheiloplasty and veloplasty. The signals of the candidate genes were visualized using chromogenic in situ hybridization and analyzed using a semi-quantitative method. No statistically important difference in the distribution of FGFR1, FGFR2 and FOXO1 between neither the patients’ lip and vomer mucosa nor the control group was observed. Statistically significant very strong and strong correlations were found between genes in the lip and palatine tissue. The expression of FGFR1, FGFR2 and FOXO1 in cleft-affected lip and palatine tissue seems to be highly individual. Numerous intercorrelations between the genes do not exclude their role in the possible complex morphopathogenesis of orofacial clefts.
Collapse
|
28
|
McElroy GS, Chakrabarty RP, D'Alessandro KB, Hu YS, Vasan K, Tan J, Stoolman JS, Weinberg SE, Steinert EM, Reyfman PA, Singer BD, Ladiges WC, Gao L, Lopéz-Barneo J, Ridge K, Budinger GRS, Chandel NS. Reduced expression of mitochondrial complex I subunit Ndufs2 does not impact healthspan in mice. Sci Rep 2022; 12:5196. [PMID: 35338200 PMCID: PMC8956724 DOI: 10.1038/s41598-022-09074-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/16/2022] [Indexed: 01/01/2023] Open
Abstract
Aging in mammals leads to reduction in genes encoding the 45-subunit mitochondrial electron transport chain complex I. It has been hypothesized that normal aging and age-related diseases such as Parkinson’s disease are in part due to modest decrease in expression of mitochondrial complex I subunits. By contrast, diminishing expression of mitochondrial complex I genes in lower organisms increases lifespan. Furthermore, metformin, a putative complex I inhibitor, increases healthspan in mice and humans. In the present study, we investigated whether loss of one allele of Ndufs2, the catalytic subunit of mitochondrial complex I, impacts healthspan and lifespan in mice. Our results indicate that Ndufs2 hemizygous mice (Ndufs2+/−) show no overt impairment in aging-related motor function, learning, tissue histology, organismal metabolism, or sensitivity to metformin in a C57BL6/J background. Despite a significant reduction of Ndufs2 mRNA, the mice do not demonstrate a significant decrease in complex I function. However, there are detectable transcriptomic changes in individual cell types and tissues due to loss of one allele of Ndufs2. Our data indicate that a 50% decline in mRNA of the core mitochondrial complex I subunit Ndufs2 is neither beneficial nor detrimental to healthspan.
Collapse
Affiliation(s)
- Gregory S McElroy
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ram P Chakrabarty
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karis B D'Alessandro
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yuan-Shih Hu
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karthik Vasan
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jerica Tan
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Joshua S Stoolman
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Samuel E Weinberg
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth M Steinert
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul A Reyfman
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin D Singer
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Warren C Ladiges
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José Lopéz-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Karen Ridge
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Scott Budinger
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Navdeep S Chandel
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
29
|
Izgi H, Han D, Isildak U, Huang S, Kocabiyik E, Khaitovich P, Somel M, Dönertaş HM. Inter-tissue convergence of gene expression during ageing suggests age-related loss of tissue and cellular identity. eLife 2022; 11:68048. [PMID: 35098922 PMCID: PMC8880995 DOI: 10.7554/elife.68048] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Developmental trajectories of gene expression may reverse in their direction during ageing, a phenomenon previously linked to cellular identity loss. Our analysis of cerebral cortex, lung, liver and muscle transcriptomes of 16 mice, covering development and ageing intervals, revealed widespread but tissue-specific ageing-associated expression reversals. Cumulatively, these reversals create a unique phenomenon: mammalian tissue transcriptomes diverge from each other during postnatal development, but during ageing, they tend to converge towards similar expression levels, a process we term Divergence followed by Convergence, or DiCo. We found that DiCo was most prevalent among tissue-specific genes and associated with loss of tissue identity, which is confirmed using data from independent mouse and human datasets. Further, using publicly available single-cell transcriptome data, we showed that DiCo could be driven both by alterations in tissue cell type composition and also by cell-autonomous expression changes within particular cell types.
Collapse
Affiliation(s)
- Hamit Izgi
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - DingDing Han
- CAS Key Laboratory of Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Ulas Isildak
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Shuyun Huang
- CAS Key Laboratory of Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Ece Kocabiyik
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Philipp Khaitovich
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow, Russian Federation
| | - Mehmet Somel
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | | |
Collapse
|
30
|
Cai Z, Deng X, Jia J, Wang D, Yuan G. Ectodysplasin A/Ectodysplasin A Receptor System and Their Roles in Multiple Diseases. Front Physiol 2021; 12:788411. [PMID: 34938205 PMCID: PMC8685516 DOI: 10.3389/fphys.2021.788411] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022] Open
Abstract
Ectodysplasin A (EDA) is a member of the tumor necrosis factor (TNF) family of ligands that was initially reported to induce the formation of various ectodermal derivatives during normal prenatal development. EDA exerts its biological activity as two splice variants, namely, EDA-A1 and EDA-A2. The former binds to the EDA receptor (EDAR), resulting in the recruitment of the intracellular EDAR-associated death domain (EDARADD) adapter protein and the activation of the NF-κB signaling pathway, while the latter binds to a different receptor, EDA2R, also known as X-linked ectodermal dysplasia receptor (XEDAR). Inactivation mutation of the EDA gene or the genes coding for its receptors can result in hypohidrosis ectodermal dysplasia (HED), a condition that is characterized by oligotrichosis, edentulosis or oligodontia, and oligohidrosis or anhidrosis. Recently, as a new liver factor, EDA is gradually known and endowed with some new functions. EDA levels were observed to be upregulated in several metabolic diseases, such as non-alcoholic fatty liver disease (NAFLD), obesity, and insulin resistance. In addition, EDA and its receptors have been implicated in tumor pathogenesis through the regulation of tumor cell proliferation, apoptosis, differentiation, and migration. Here, we first review the role of EDA and its two-receptor system in various signaling pathways and then discuss the physiological and pathological roles of EDA and its receptors.
Collapse
Affiliation(s)
- Zhensheng Cai
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xia Deng
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jue Jia
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Dong Wang
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guoyue Yuan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
31
|
Guo Y, Ju Y, Chen D, Wang L. Research on the Computational Prediction of Essential Genes. Front Cell Dev Biol 2021; 9:803608. [PMID: 34938741 PMCID: PMC8685449 DOI: 10.3389/fcell.2021.803608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/22/2021] [Indexed: 11/19/2022] Open
Abstract
Genes, the nucleotide sequences that encode a polypeptide chain or functional RNA, are the basic genetic unit controlling biological traits. They are the guarantee of the basic structures and functions in organisms, and they store information related to biological factors and processes such as blood type, gestation, growth, and apoptosis. The environment and genetics jointly affect important physiological processes such as reproduction, cell division, and protein synthesis. Genes are related to a wide range of phenomena including growth, decline, illness, aging, and death. During the evolution of organisms, there is a class of genes that exist in a conserved form in multiple species. These genes are often located on the dominant strand of DNA and tend to have higher expression levels. The protein encoded by it usually either performs very important functions or is responsible for maintaining and repairing these essential functions. Such genes are called persistent genes. Among them, the irreplaceable part of the body’s life activities is the essential gene. For example, when starch is the only source of energy, the genes related to starch digestion are essential genes. Without them, the organism will die because it cannot obtain enough energy to maintain basic functions. The function of the proteins encoded by these genes is thought to be fundamental to life. Nowadays, DNA can be extracted from blood, saliva, or tissue cells for genetic testing, and detailed genetic information can be obtained using the most advanced scientific instruments and technologies. The information gained from genetic testing is useful to assess the potential risks of disease, and to help determine the prognosis and development of diseases. Such information is also useful for developing personalized medication and providing targeted health guidance to improve the quality of life. Therefore, it is of great theoretical and practical significance to identify important and essential genes. In this paper, the research status of essential genes and the essential genome database of bacteria are reviewed, the computational prediction method of essential genes based on communication coding theory is expounded, and the significance and practical application value of essential genes are discussed.
Collapse
Affiliation(s)
- Yuxin Guo
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China.,Key Laboratory of Computational Science and Application of Hainan Province, Haikou, China.,Key Laboratory of Data Science and Intelligence Education, Hainan Normal University, Ministry of Education, Haikou, China.,School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Ying Ju
- School of Informatics, Xiamen University, Xiamen, China
| | - Dong Chen
- College of Electrical and Information Engineering, Quzhou University, Quzhou, China
| | - Lihong Wang
- Beidahuang Industry Group General Hospital, Harbin, China
| |
Collapse
|
32
|
Peng S, Zeng L, Haure-Mirande JV, Wang M, Huffman DM, Haroutunian V, Ehrlich ME, Zhang B, Tu Z. Transcriptomic Changes Highly Similar to Alzheimer's Disease Are Observed in a Subpopulation of Individuals During Normal Brain Aging. Front Aging Neurosci 2021; 13:711524. [PMID: 34924992 PMCID: PMC8675870 DOI: 10.3389/fnagi.2021.711524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is a major risk factor for late-onset Alzheimer’s disease (LOAD). How aging contributes to the development of LOAD remains elusive. In this study, we examined multiple large-scale transcriptomic datasets from both normal aging and LOAD brains to understand the molecular interconnection between aging and LOAD. We found that shared gene expression changes between aging and LOAD are mostly seen in the hippocampal and several cortical regions. In the hippocampus, the expression of phosphoprotein, alternative splicing and cytoskeleton genes are commonly changed in both aging and AD, while synapse, ion transport, and synaptic vesicle genes are commonly down-regulated. Aging-specific changes are associated with acetylation and methylation, while LOAD-specific changes are more related to glycoprotein (both up- and down-regulations), inflammatory response (up-regulation), myelin sheath and lipoprotein (down-regulation). We also found that normal aging brain transcriptomes from relatively young donors (45–70 years old) clustered into several subgroups and some subgroups showed gene expression changes highly similar to those seen in LOAD brains. Using brain transcriptomic datasets from another cohort of older individuals (>70 years), we found that samples from cognitively normal older individuals clustered with the “healthy aging” subgroup while AD samples mainly clustered with the “AD similar” subgroups. This may imply that individuals in the healthy aging subgroup will likely remain cognitively normal when they become older and vice versa. In summary, our results suggest that on the transcriptome level, aging and LOAD have strong interconnections in some brain regions in a subpopulation of cognitively normal aging individuals. This supports the theory that the initiation of LOAD occurs decades earlier than the manifestation of clinical phenotype and it may be essential to closely study the “normal brain aging” to identify the very early molecular events that may lead to LOAD development.
Collapse
Affiliation(s)
- Shouneng Peng
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Lu Zeng
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | | | - Minghui Wang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Derek M Huffman
- Department of Medicine, Albert Einstein College of Medicine, New York City, NY, United States.,Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, United States.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York City, NY, United States
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, United States
| | - Michelle E Ehrlich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Bin Zhang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
33
|
Guo Y, Wu C, Yuan Z, Wang Y, Liang Z, Wang Y, Zhang Y, Xu L. Gene-Based Testing of Interactions Using XGBoost in Genome-Wide Association Studies. Front Cell Dev Biol 2021; 9:801113. [PMID: 34977040 PMCID: PMC8716787 DOI: 10.3389/fcell.2021.801113] [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: 10/24/2021] [Accepted: 11/23/2021] [Indexed: 11/30/2022] Open
Abstract
Among the myriad of statistical methods that identify gene–gene interactions in the realm of qualitative genome-wide association studies, gene-based interactions are not only powerful statistically, but also they are interpretable biologically. However, they have limited statistical detection by making assumptions on the association between traits and single nucleotide polymorphisms. Thus, a gene-based method (GGInt-XGBoost) originated from XGBoost is proposed in this article. Assuming that log odds ratio of disease traits satisfies the additive relationship if the pair of genes had no interactions, the difference in error between the XGBoost model with and without additive constraint could indicate gene–gene interaction; we then used a permutation-based statistical test to assess this difference and to provide a statistical p-value to represent the significance of the interaction. Experimental results on both simulation and real data showed that our approach had superior performance than previous experiments to detect gene–gene interactions.
Collapse
Affiliation(s)
- Yingjie Guo
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Chenxi Wu
- Department of Mathematics, University of Wisconsin-Madison, Madison, WI, United States
| | - Zhian Yuan
- Research Institute of Big Data Science and Industry, Shanxi University, Taiyuan, China
| | - Yansu Wang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Zhen Liang
- School of Life Science, Shanxi University, Taiyuan, China
| | - Yang Wang
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Yi Zhang
- Beidahuang Industry Group General Hospital, Harbin, China
- *Correspondence: Yi Zhang, ; Lei Xu,
| | - Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
- *Correspondence: Yi Zhang, ; Lei Xu,
| |
Collapse
|
34
|
Genetically regulated expression in late-onset Alzheimer's disease implicates risk genes within known and novel loci. Transl Psychiatry 2021; 11:618. [PMID: 34873149 PMCID: PMC8648734 DOI: 10.1038/s41398-021-01677-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 12/22/2022] Open
Abstract
Late-onset Alzheimer disease (LOAD) is highly polygenic, with a heritability estimated between 40 and 80%, yet risk variants identified in genome-wide studies explain only ~8% of phenotypic variance. Due to its increased power and interpretability, genetically regulated expression (GReX) analysis is an emerging approach to investigate the genetic mechanisms of complex diseases. Here, we conducted GReX analysis within and across 51 tissues on 39 LOAD GWAS data sets comprising 58,713 cases and controls from the Alzheimer's Disease Genetics Consortium (ADGC) and the International Genomics of Alzheimer's Project (IGAP). Meta-analysis across studies identified 216 unique significant genes, including 72 with no previously reported LOAD GWAS associations. Cross-brain-tissue and cross-GTEx models revealed eight additional genes significantly associated with LOAD. Conditional analysis of previously reported loci using established LOAD-risk variants identified eight genes reaching genome-wide significance independent of known signals. Moreover, the proportion of SNP-based heritability is highly enriched in genes identified by GReX analysis. In summary, GReX-based meta-analysis in LOAD identifies 216 genes (including 72 novel genes), illuminating the role of gene regulatory models in LOAD.
Collapse
|
35
|
Lee KH, Kim DY, Kim W. Regulation of Gene Expression by Telomere Position Effect. Int J Mol Sci 2021; 22:ijms222312807. [PMID: 34884608 PMCID: PMC8657463 DOI: 10.3390/ijms222312807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022] Open
Abstract
Many diseases that involve malignant tumors in the elderly affect the quality of human life; therefore, the relationship between aging and pathogenesis in geriatric diseases must be under-stood to develop appropriate treatments for these diseases. Recent reports have shown that epigenetic regulation caused by changes in the local chromatin structure plays an essential role in aging. This review provides an overview of the roles of telomere shortening on genomic structural changes during an age-dependent shift in gene expression. Telomere shortening is one of the most prominent events that is involved in cellular aging and it affects global gene expression through genome rearrangement. This review provides novel insights into the roles of telomere shortening in disease-affected cells during pathogenesis and suggests novel therapeutic approaches.
Collapse
Affiliation(s)
- Kyung-Ha Lee
- Division of Cosmetic Science and Technology, Daegu Haany University, Gyeongsan 38610, Korea;
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
- Correspondence: (D.-Y.K.); (W.K.)
| | - Wanil Kim
- Department of Biochemistry, Department of Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 52727, Korea
- Correspondence: (D.-Y.K.); (W.K.)
| |
Collapse
|
36
|
Lang J, Zhu R, Sun X, Zhu S, Li T, Shi X, Sun Y, Yang Z, Wang W, Bing P, He B, Tian G. Evaluation of the MGISEQ-2000 Sequencing Platform for Illumina Target Capture Sequencing Libraries. Front Genet 2021; 12:730519. [PMID: 34777467 PMCID: PMC8578046 DOI: 10.3389/fgene.2021.730519] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/24/2021] [Indexed: 01/19/2023] Open
Abstract
Illumina is the leading sequencing platform in the next-generation sequencing (NGS) market globally. In recent years, MGI Tech has presented a series of new sequencers, including DNBSEQ-T7, MGISEQ-2000 and MGISEQ-200. As a complex application of NGS, cancer-detecting panels pose increasing demands for the high accuracy and sensitivity of sequencing and data analysis. In this study, we used the same capture DNA libraries constructed based on the Illumina protocol to evaluate the performance of the Illumina Nextseq500 and MGISEQ-2000 sequencing platforms. We found that the two platforms had high consistency in the results of hotspot mutation analysis; more importantly, we found that there was a significant loss of fragments in the 101-133 bp size range on the MGISEQ-2000 sequencing platform for Illumina libraries, but not for the capture DNA libraries prepared based on the MGISEQ protocol. This phenomenon may indicate fragment selection or low fragment ligation efficiency during the DNA circularization step, which is a unique step of the MGISEQ-2000 sequence platform. In conclusion, these different sequencing libraries and corresponding sequencing platforms are compatible with each other, but protocol and platform selection need to be carefully evaluated in combination with research purpose.
Collapse
Affiliation(s)
- Jidong Lang
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China.,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China.,Academician Workstation, Changsha Medical University, Changsha, China
| | - Rongrong Zhu
- Vascular Surgery Department, Tsinghua University Affiliated Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Xue Sun
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China
| | - Siyu Zhu
- Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA, United States
| | - Tianbao Li
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China.,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Xiaoli Shi
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China
| | - Yanqi Sun
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China
| | - Zhou Yang
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China
| | - Weiwei Wang
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China.,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Pingping Bing
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Binsheng He
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Geng Tian
- Bioinformatics and R and D Department, Geneis (Beijing) Co. Ltd., Beijing, China.,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| |
Collapse
|
37
|
Xiang C, Ni H, Wang Z, Ji B, Wang B, Shi X, Wu W, Liu N, Gu Y, Ma D, Liu H. Agent Repurposing for the Treatment of Advanced Stage Diffuse Large B-Cell Lymphoma Based on Gene Expression and Network Perturbation Analysis. Front Genet 2021; 12:756784. [PMID: 34721544 PMCID: PMC8551569 DOI: 10.3389/fgene.2021.756784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022] Open
Abstract
Over 50% of diffuse large B-cell lymphoma (DLBCL) patients are diagnosed at an advanced stage. Although there are a few therapeutic strategies for DLBCL, most of them are more effective in limited-stage cancer patients. The prognosis of patients with advanced-stage DLBCL is usually poor with frequent recurrence and metastasis. In this study, we aimed to identify gene expression and network differences between limited- and advanced-stage DLBCL patients, with the goal of identifying potential agents that could be used to relieve the severity of DLBCL. Specifically, RNA sequencing data of DLBCL patients at different clinical stages were collected from the cancer genome atlas (TCGA). Differentially expressed genes were identified using DESeq2, and then, weighted gene correlation network analysis (WGCNA) and differential module analysis were performed to find variations between different stages. In addition, important genes were extracted by key driver analysis, and potential agents for DLBCL were identified according to gene-expression perturbations and the Crowd Extracted Expression of Differential Signatures (CREEDS) drug signature database. As a result, 20 up-regulated and 73 down-regulated genes were identified and 79 gene co-expression modules were found using WGCNA, among which, the thistle1 module was highly related to the clinical stage of DLBCL. KEGG pathway and GO enrichment analyses of genes in the thistle1 module indicated that DLBCL progression was mainly related to the NOD-like receptor signaling pathway, neutrophil activation, secretory granule membrane, and carboxylic acid binding. A total of 47 key drivers were identified through key driver analysis with 11 up-regulated key driver genes and 36 down-regulated key diver genes in advanced-stage DLBCL patients. Five genes (MMP1, RAB6C, ACCSL, RGS21 and MOCOS) appeared as hub genes, being closely related to the occurrence and development of DLBCL. Finally, both differentially expressed genes and key driver genes were subjected to CREEDS analysis, and 10 potential agents were predicted to have the potential for application in advanced-stage DLBCL patients. In conclusion, we propose a novel pipeline to utilize perturbed gene-expression signatures during DLBCL progression for identifying agents, and we successfully utilized this approach to generate a list of promising compounds.
Collapse
Affiliation(s)
- Chenxi Xiang
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Huimin Ni
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Zhina Wang
- Department of Oncology, Emergency General Hospital, Beijing, China
| | - Binbin Ji
- Genies Beijing Co., Ltd., Beijing, China.,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Bo Wang
- Genies Beijing Co., Ltd., Beijing, China.,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Xiaoli Shi
- Genies Beijing Co., Ltd., Beijing, China.,Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Wanna Wu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Nian Liu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Ying Gu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Dongshen Ma
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Liu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
38
|
Perrot N, Pelletier W, Bourgault J, Couture C, Li Z, Mitchell PL, Ghodsian N, Bossé Y, Thériault S, Mathieu P, Arsenault BJ. A trans-omic Mendelian randomization study of parental lifespan uncovers novel aging biology and therapeutic candidates for chronic diseases. Aging Cell 2021; 20:e13497. [PMID: 34704651 PMCID: PMC8590095 DOI: 10.1111/acel.13497] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 05/20/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022] Open
Abstract
The study of parental lifespan has emerged as an innovative tool to advance aging biology and our understanding of the genetic architecture of human longevity and aging-associated diseases. Here, we leveraged summary statistics of a genome-wide association study including over one million parental lifespans to identify genetically regulated genes from the Genotype-Tissue Expression project. Through a combination of multi-tissue transcriptome-wide association analyses and genetic colocalization, we identified novel genes that may be associated with parental lifespan. Mendelian randomization (MR) analyses also identified circulating proteins and metabolites causally associated with parental lifespan and chronic diseases offering new drug repositioning opportunities such as those targeting apolipoprotein-B-containing lipoproteins. Liver expression of HP, the gene encoding haptoglobin, and plasma haptoglobin levels were causally linked with parental lifespan. Phenome-wide MR analyses were used to map genetically regulated genes, proteins and metabolites with other human traits as well as the disease-related phenome in the FinnGen cohorts (n = 135,638). Altogether, this study identified new candidate genes, circulating proteins and metabolites that may influence human aging as well as potential therapeutic targets for chronic diseases that warrant further investigation.
Collapse
Affiliation(s)
- Nicolas Perrot
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
- Department of MedicineFaculty of MedicineUniversité LavalQuébecQCCanada
| | - William Pelletier
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
- Department of MedicineFaculty of MedicineUniversité LavalQuébecQCCanada
| | - Jérôme Bourgault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
| | - Christian Couture
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
| | - Zhonglin Li
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
| | - Patricia L. Mitchell
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
| | - Nooshin Ghodsian
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
| | - Yohan Bossé
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
- Department of Molecular MedicineFaculty of MedicineUniversité LavalQuébecQCCanada
| | - Sébastien Thériault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
- Department of Molecular Biology, Medical Biochemistry and PathologyFaculty of MedicineUniversité LavalQuébecQCCanada
| | - Patrick Mathieu
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
- Department of SurgeryFaculty of MedicineUniversité LavalQuébecQCCanada
| | - Benoit J. Arsenault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de QuébecQuébecQCCanada
- Department of MedicineFaculty of MedicineUniversité LavalQuébecQCCanada
| |
Collapse
|
39
|
Potrony M, Haddad TS, Tell-Martí G, Gimenez-Xavier P, Leon C, Pevida M, Mateu J, Badenas C, Carrera C, Malvehy J, Aguilera P, Llames S, Escámez MJ, Puig-Butillé JA, Del Río M, Puig S. DNA Repair and Immune Response Pathways Are Deregulated in Melanocyte-Keratinocyte Co-cultures Derived From the Healthy Skin of Familial Melanoma Patients. Front Med (Lausanne) 2021; 8:692341. [PMID: 34660619 PMCID: PMC8517393 DOI: 10.3389/fmed.2021.692341] [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: 04/08/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Familial melanoma accounts for 10% of cases, being CDKN2A the main high-risk gene. However, the mechanisms underlying melanomagenesis in these cases remain poorly understood. Our aim was to analyze the transcriptome of melanocyte-keratinocyte co-cultures derived from healthy skin from familial melanoma patients vs. controls, to unveil pathways involved in melanoma development in at-risk individuals. Accordingly, primary melanocyte-keratinocyte co-cultures were established from the healthy skin biopsies of 16 unrelated familial melanoma patients (8 CDKN2A mutant, 8 CDKN2A wild-type) and 7 healthy controls. Whole transcriptome was captured using the SurePrint G3 Human Microarray. Transcriptome analyses included: differential gene expression, functional enrichment, and protein-protein interaction (PPI) networks. We identified a gene profile associated with familial melanoma independently of CDKN2A germline status. Functional enrichment analysis of this profile showed a downregulation of pathways related to DNA repair and immune response in familial melanoma (P < 0.05). In addition, the PPI network analysis revealed a network that consisted of double-stranded DNA repair genes (including BRCA1, BRCA2, BRIP1, and FANCA), immune response genes, and regulation of chromosome segregation. The hub gene was BRCA1. In conclusion, the constitutive deregulation of BRCA1 pathway genes and the immune response in healthy skin could be a mechanism related to melanoma risk.
Collapse
Affiliation(s)
- Miriam Potrony
- Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Tariq Sami Haddad
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Tell-Martí
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Pol Gimenez-Xavier
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Leon
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioingeniería, Universidad Carlos III de Madrid, Leganés, Spain.,Cátedra de Medicina Regenerativa y Bioingeniería de Tejidos, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Marta Pevida
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Tissue Engineering Unit, Centro Comunitario de Sangre y Tejidos de Asturias, Oviedo, Spain.,Instituto Universitario Fdez-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Oviedo, Spain
| | - Judit Mateu
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Celia Badenas
- Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Carrera
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Josep Malvehy
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Paula Aguilera
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Sara Llames
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Cátedra de Medicina Regenerativa y Bioingeniería de Tejidos, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.,Tissue Engineering Unit, Centro Comunitario de Sangre y Tejidos de Asturias, Oviedo, Spain.,Instituto Universitario Fdez-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Oviedo, Spain
| | - Maria José Escámez
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioingeniería, Universidad Carlos III de Madrid, Leganés, Spain.,Cátedra de Medicina Regenerativa y Bioingeniería de Tejidos, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.,Centro de Investigaciones Energéticas Mediambientales y Tecnonlógicas, Madrid, Spain
| | - Joan A Puig-Butillé
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Molecular Biology Core, Biomedical Diagnostic Center, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Marcela Del Río
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioingeniería, Universidad Carlos III de Madrid, Leganés, Spain.,Cátedra de Medicina Regenerativa y Bioingeniería de Tejidos, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.,Centro de Investigaciones Energéticas Mediambientales y Tecnonlógicas, Madrid, Spain
| | - Susana Puig
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Ma H, Liu Z, Li H, Guo X, Guo S, Qu P, Wang Y. Bioinformatics Analysis Reveals MCM3 as an Important Prognostic Marker in Cervical Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:8494260. [PMID: 34671420 PMCID: PMC8523256 DOI: 10.1155/2021/8494260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 12/09/2022]
Abstract
The minichromosome maintenance complex 3 (MCM3) is essential for the regulation of DNA replication and cell cycle progression. However, the expression and prognostic values of MCM3 in cervical cancer (CC) have not been well-studied. Herein, we investigated the expression patterns and survival data of MCM3 in cervical cancer patients from the ONCOMINE, GEPIA, Human Protein Atlas, UALCAN, Kaplan-Meier Plotter, and LinkedOmics databases. The expression level of MCM3 is negatively correlated with advanced tumor stage and metastatic status. Specifically, MCM3 is significantly differentially expressed between patients in stage 1 and stage 3 cervical cancer with p value 0.0138. Similarly, the p values between stage 1 and stage 4 cervical cancer, between stage 2 and stage 3, and between stage 2 and stage 4 are 0.00089, 0.0244, and 0.00197, respectively. Not only that, cervical cancer patients with high mRNA expression of MCM3 may indicate longer overall survival but indicate shorter relapse-free survival. PRIM2 and MCM6 are positively correlated genes of MCM3. Bioinformatics analysis revealed that MCM3 might be considered a biological indicator for prognostic evaluation of cervical cancer. However, it is currently limited to bioinformatics analysis, and more clinical tissue specimens and cell experiments are needed to further explore the role of MCM3 in the occurrence and progression of cervical cancer.
Collapse
Affiliation(s)
- Hui Ma
- Department of Gynecology, The Secondary Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin 300211, China
| | - Zhen Liu
- Department of Gynecology, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng 024099, China
| | - Honglin Li
- Department of Gynecology, The Secondary Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin 300211, China
| | - Xuewang Guo
- Department of Gynecology, The Secondary Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin 300211, China
| | - Sujie Guo
- Department of Gynecology, The Secondary Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin 300211, China
| | - Pengpeng Qu
- Department of Gynecology Oncology, Tianjin Central Hospital of Gynecology & Obstetrics, 156 Sanmalu, Nankai, Tianjin 300100, China
| | - Yuquan Wang
- Department of Gynecology, The Secondary Hospital of Tianjin Medical University, No. 23 Pingjiang Road, Hexi District, Tianjin 300211, China
| |
Collapse
|
41
|
Zhao L, Li Y, Wang Y, Gao Q, Ge Z, Sun X, Li Y. Development and Validation of a Nomogram for the Prediction of Hospital Mortality of Patients With Encephalopathy Caused by Microbial Infection: A Retrospective Cohort Study. Front Microbiol 2021; 12:737066. [PMID: 34489922 PMCID: PMC8417384 DOI: 10.3389/fmicb.2021.737066] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Background Hospital mortality is high for patients with encephalopathy caused by microbial infection. Microbial infections often induce sepsis. The damage to the central nervous system (CNS) is defined as sepsis-associated encephalopathy (SAE). However, the relationship between pathogenic microorganisms and the prognosis of SAE patients is still unclear, especially gut microbiota, and there is no clinical tool to predict hospital mortality for SAE patients. The study aimed to explore the relationship between pathogenic microorganisms and the hospital mortality of SAE patients and develop a nomogram for the prediction of hospital mortality in SAE patients. Methods The study is a retrospective cohort study. The lasso regression model was used for data dimension reduction and feature selection. Model of hospital mortality of SAE patients was developed by multivariable Cox regression analysis. Calibration and discrimination were used to assess the performance of the nomogram. Decision curve analysis (DCA) to evaluate the clinical utility of the model. Results Unfortunately, the results of our study did not find intestinal infection and microorganisms of the gastrointestinal (such as: Escherichia coli) that are related to the prognosis of SAE. Lasso regression and multivariate Cox regression indicated that factors including respiratory failure, lactate, international normalized ratio (INR), albumin, SpO2, temperature, and renal replacement therapy were significantly correlated with hospital mortality. The AUC of 0.812 under the nomogram was more than that of the Simplified Acute Physiology Score (0.745), indicating excellent discrimination. DCA demonstrated that using the nomogram or including the prognostic signature score status was better than without the nomogram or using the SAPS II at predicting hospital mortality. Conclusion The prognosis of SAE patients has nothing to do with intestinal and microbial infections. We developed a nomogram that predicts hospital mortality in patients with SAE according to clinical data. The nomogram exhibited excellent discrimination and calibration capacity, favoring its clinical utility.
Collapse
Affiliation(s)
- Lina Zhao
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Yun Li
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Yunying Wang
- Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Qian Gao
- Department of Neurology, Yidu Central Hospital Affiliated to Weifang Medical University, Weifang, China
| | - Zengzheng Ge
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xibo Sun
- Department of Neurology, Yidu Central Hospital Affiliated to Weifang Medical University, Weifang, China
| | - Yi Li
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Beckmann ND, Comella PH, Cheng E, Lepow L, Beckmann AG, Tyler SR, Mouskas K, Simons NW, Hoffman GE, Francoeur NJ, Del Valle DM, Kang G, Do A, Moya E, Wilkins L, Le Berichel J, Chang C, Marvin R, Calorossi S, Lansky A, Walker L, Yi N, Yu A, Chung J, Hartnett M, Eaton M, Hatem S, Jamal H, Akyatan A, Tabachnikova A, Liharska LE, Cotter L, Fennessy B, Vaid A, Barturen G, Shah H, Wang YC, Sridhar SH, Soto J, Bose S, Madrid K, Ellis E, Merzier E, Vlachos K, Fishman N, Tin M, Smith M, Xie H, Patel M, Nie K, Argueta K, Harris J, Karekar N, Batchelor C, Lacunza J, Yishak M, Tuballes K, Scott I, Kumar A, Jaladanki S, Agashe C, Thompson R, Clark E, Losic B, Peters L, Roussos P, Zhu J, Wang W, Kasarskis A, Glicksberg BS, Nadkarni G, Bogunovic D, Elaiho C, Gangadharan S, Ofori-Amanfo G, Alesso-Carra K, Onel K, Wilson KM, Argmann C, Bunyavanich S, Alarcón-Riquelme ME, Marron TU, Rahman A, Kim-Schulze S, Gnjatic S, Gelb BD, Merad M, Sebra R, Schadt EE, Charney AW. Downregulation of exhausted cytotoxic T cells in gene expression networks of multisystem inflammatory syndrome in children. Nat Commun 2021; 12:4854. [PMID: 34381049 PMCID: PMC8357784 DOI: 10.1038/s41467-021-24981-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) presents with fever, inflammation and pathology of multiple organs in individuals under 21 years of age in the weeks following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although an autoimmune pathogenesis has been proposed, the genes, pathways and cell types causal to this new disease remain unknown. Here we perform RNA sequencing of blood from patients with MIS-C and controls to find disease-associated genes clustered in a co-expression module annotated to CD56dimCD57+ natural killer (NK) cells and exhausted CD8+ T cells. A similar transcriptome signature is replicated in an independent cohort of Kawasaki disease (KD), the related condition after which MIS-C was initially named. Probing a probabilistic causal network previously constructed from over 1,000 blood transcriptomes both validates the structure of this module and reveals nine key regulators, including TBX21, a central coordinator of exhausted CD8+ T cell differentiation. Together, this unbiased, transcriptome-wide survey implicates downregulation of NK cells and cytotoxic T cell exhaustion in the pathogenesis of MIS-C.
Collapse
Affiliation(s)
- Noam D Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA.
| | - Phillip H Comella
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Lepow
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aviva G Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott R Tyler
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Konstantinos Mouskas
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicole W Simons
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nancy J Francoeur
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | | | - Gurpawan Kang
- Department of Medicine, Division of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anh Do
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Emily Moya
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lillian Wilkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Le Berichel
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christie Chang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Marvin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sharlene Calorossi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alona Lansky
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Walker
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nancy Yi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alex Yu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Chung
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Melody Eaton
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandra Hatem
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hajra Jamal
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alara Akyatan
- Department of of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra Tabachnikova
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lora E Liharska
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liam Cotter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Fennessy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Akhil Vaid
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guillermo Barturen
- Department of Medical Genomics, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government (GENYO), Granada, Spain
| | - Hardik Shah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ying-Chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shwetha Hara Sridhar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan Soto
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Swaroop Bose
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Kent Madrid
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Ethan Ellis
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Elyze Merzier
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Konstantinos Vlachos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Nataly Fishman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Manying Tin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Melissa Smith
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Hui Xie
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kimberly Argueta
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jocelyn Harris
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neha Karekar
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Craig Batchelor
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jose Lacunza
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mahlet Yishak
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kevin Tuballes
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ieisha Scott
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arvind Kumar
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Suraj Jaladanki
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charuta Agashe
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ryan Thompson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Evan Clark
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bojan Losic
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Peters
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panagiotis Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jun Zhu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenhui Wang
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Benjamin S Glicksberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish Nadkarni
- Mount Sinai COVID Informatics Center, New York, NY, USA
- Department of Medicine, Mount Sinai, New York, NY, USA
- Hasso Plattner Institute for Digital Health at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute for Personalized Medicine, New York, NY, USA
| | - Dusan Bogunovic
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cordelia Elaiho
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandeep Gangadharan
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Ofori-Amanfo
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kasey Alesso-Carra
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenan Onel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karen M Wilson
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta E Alarcón-Riquelme
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas U Marron
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adeeb Rahman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce D Gelb
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Black Family Stem Cell Institute, New York, NY, USA
- Sema4, a Mount Sinai Venture, Stamford, CT, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA.
- Sema4, a Mount Sinai Venture, Stamford, CT, USA.
| | - Alexander W Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mount Sinai COVID Informatics Center, New York, NY, USA.
| |
Collapse
|
43
|
Walkiewicz D, Wicik Z, Puzianowska-Kuznicka M. Gonadotropin-releasing hormone receptor pathway affects the function of human EBV-transformed B lymphocytes in an age-independent way. Exp Gerontol 2021; 152:111471. [PMID: 34256116 DOI: 10.1016/j.exger.2021.111471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/27/2022]
Abstract
Immune system function changes during aging, but the molecular mechanisms of this phenomenon are not fully understood. The present study identified pathways that are associated with age-associated changes in human B lymphocytes. Initial in silico analysis of 1355 genes involved in aging revealed the strongest association (p = 4.36E-21) with the gonadotropin-releasing hormone receptor (GnRHR) pathway. Extended analysis of 2736 aging-related genes using updated databases confirmed such association (p = 2.41E-16). Genes involved in both aging and the GnRHR pathway were significantly involved in lymphocyte B and T activation and aging-related phenotypes, including hyperinsulinemia and diabetes, arthritis, cerebrovascular disease, and cancers. We, therefore, examined non-tumorigenic Epstein-Barr virus (EBV)-transformed B-lymphocyte cell lines that originated from 12 young subjects (20-31 years old) and 10 centenarians (100-102 years old). Gonadotropin-releasing hormone I (GnRH-I) and GnRHR levels did not depend on the age of the cell donors. Inhibition of the GnRHR pathway age-independently decreased cell proliferation (p < 0.001) and increased apoptosis (p < 0.001). However, the decrease in immunoglobulin G synthesis (p < 0.01) was twice as high in centenarian cells than in young cells. In conclusion, the GnRHR pathway regulated essential properties of B lymphocytes. However, upon EBV transformation, memory class-switched B cells became the dominant cell subpopulation. Therefore, the observed effects of GnRHR inhibition were attributable to this subpopulation.
Collapse
Affiliation(s)
- Dorota Walkiewicz
- Department of Human Epigenetics, Mossakowski Medical Research Institute, PAS, 02-106 Warsaw, Poland; Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
| | - Zofia Wicik
- Department of Human Epigenetics, Mossakowski Medical Research Institute, PAS, 02-106 Warsaw, Poland.
| | - Monika Puzianowska-Kuznicka
- Department of Human Epigenetics, Mossakowski Medical Research Institute, PAS, 02-106 Warsaw, Poland; Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland.
| |
Collapse
|
44
|
Emechebe U, Nelson JW, Alkayed NJ, Kaul S, Adey AC, Barnes AP. Age-dependent transcriptional alterations in cardiac endothelial cells. Physiol Genomics 2021; 53:295-308. [PMID: 34097533 PMCID: PMC8321782 DOI: 10.1152/physiolgenomics.00037.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023] Open
Abstract
Aging is a significant risk factor for cardiovascular disease. Despite the fact that endothelial cells play critical roles in cardiovascular function and disease, the molecular impact of aging on this cell population in many organ systems remains unknown. In this study, we sought to determine age-associated transcriptional alterations in cardiac endothelial cells. Highly enriched populations of endothelial cells (ECs) isolated from the heart, brain, and kidney of young (3 mo) and aged (24 mo) C57/BL6 mice were profiled for RNA expression via bulk RNA sequencing. Approximately 700 cardiac endothelial transcripts significantly differ by age. Gene set enrichment analysis indicated similar patterns for cellular pathway perturbations. Receptor-ligand comparisons indicated parallel alterations in age-affected circulating factors and cardiac endothelial-expressed receptors. Gene and pathway enrichment analyses show that age-related transcriptional response of cardiac endothelial cells is distinct from that of endothelial cells derived from the brain or kidney vascular bed. Furthermore, single-cell analysis identified nine distinct EC subtypes and shows that the Apelin Receptor-enriched subtype is reduced with age in mouse heart. Finally, we identify age-dysregulated genes in specific aged cardiac endothelial subtypes.
Collapse
Affiliation(s)
- Uchenna Emechebe
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Jonathan W Nelson
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Nabil J Alkayed
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
| | - Sanjiv Kaul
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Andrew C Adey
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Cancer Early Detection Advanced Research Institute, Oregon Health and Science University, Portland, Oregon
| | - Anthony P Barnes
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
45
|
Ramos‐Marquès E, García‐Mendívil L, Pérez‐Zabalza M, Santander‐Badules H, Srinivasan S, Oliveros JC, Torres‐Pérez R, Cebollada A, Vallejo‐Gil JM, Fresneda‐Roldán PC, Fañanás‐Mastral J, Vázquez‐Sancho M, Matamala‐Adell M, Sorribas‐Berjón JF, Bellido‑Morales JA, Mancebón‑Sierra FJ, Vaca‑Núñez AS, Ballester‐Cuenca C, Jiménez‐Navarro M, Villaescusa JM, Garrido‐Huéscar E, Segovia‐Roldán M, Oliván‐Viguera A, Gómez‐González C, Muñiz G, Diez E, Ordovás L, Pueyo E. Chronological and biological aging of the human left ventricular myocardium: Analysis of microRNAs contribution. Aging Cell 2021; 20:e13383. [PMID: 34092006 PMCID: PMC8282276 DOI: 10.1111/acel.13383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 11/29/2022] Open
Abstract
Aging is the main risk factor for cardiovascular diseases. In humans, cardiac aging remains poorly characterized. Most studies are based on chronological age (CA) and disregard biological age (BA), the actual physiological age (result of the aging rate on the organ structure and function), thus yielding potentially imperfect outcomes. Deciphering the molecular basis of ventricular aging, especially by BA, could lead to major progresses in cardiac research. We aim to describe the transcriptome dynamics of the aging left ventricle (LV) in humans according to both CA and BA and characterize the contribution of microRNAs, key transcriptional regulators. BA is measured using two CA-associated transcriptional markers: CDKN2A expression, a cell senescence marker, and apparent age (AppAge), a highly complex transcriptional index. Bioinformatics analysis of 132 LV samples shows that CDKN2A expression and AppAge represent transcriptomic changes better than CA. Both BA markers are biologically validated in relation to an aging phenotype associated with heart dysfunction, the amount of cardiac fibrosis. BA-based analyses uncover depleted cardiac-specific processes, among other relevant functions, that are undetected by CA. Twenty BA-related microRNAs are identified, and two of them highly heart-enriched that are present in plasma. We describe a microRNA-gene regulatory network related to cardiac processes that are partially validated in vitro and in LV samples from living donors. We prove the higher sensitivity of BA over CA to explain transcriptomic changes in the aging myocardium and report novel molecular insights into human LV biological aging. Our results can find application in future therapeutic and biomarker research.
Collapse
Affiliation(s)
- Estel Ramos‐Marquès
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Laura García‐Mendívil
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - María Pérez‐Zabalza
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Hazel Santander‐Badules
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
| | - Sabarathinam Srinivasan
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Juan Carlos Oliveros
- Bioinformatics for Genomics and Proteomics National Center of Biotechnology‐ Spanish National Research Council Madrid Spain
| | - Rafael Torres‐Pérez
- Bioinformatics for Genomics and Proteomics National Center of Biotechnology‐ Spanish National Research Council Madrid Spain
| | | | | | | | | | - Manuel Vázquez‐Sancho
- Department of Cardiovascular Surgery University Hospital Miguel Servet Zaragoza Spain
| | - Marta Matamala‐Adell
- Department of Cardiovascular Surgery University Hospital Miguel Servet Zaragoza Spain
| | | | | | | | | | | | - Manuel Jiménez‐Navarro
- Heart Area Hospital Clínico Universitario Virgen de la Victoria, CIBERCV IBIMA, Universidad de Málaga, UMA Málaga Spain
| | - José Manuel Villaescusa
- UGC Heart Area Cardiovascular Surgery Department Hospital Universitario Virgen de la Victoria de Málaga Fundación Pública Andaluza para la Investigación de Málaga en Biomedicina y Salud (FIMABIS) CIBERCV Enfermedades Cardiovasculares Instituto de Salud Carlos III University of Málaga Madrid Spain
| | - Elisa Garrido‐Huéscar
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Margarita Segovia‐Roldán
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | - Aida Oliván‐Viguera
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
| | | | - Gorka Muñiz
- Department of Pathology San Jorge Hospital Huesca Spain
| | - Emiliano Diez
- Institute of Experimental Medicine and Biology of Cuyo (IMBECU) CONICET Mendoza Argentina
| | - Laura Ordovás
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
- ARAID Foundation Zaragoza Spain
| | - Esther Pueyo
- Biomedical Signal Interpretation and Computational Simulation group (BSICoS) Aragón Institute of Engineering Research University of Zaragoza Zaragoza Spain
- BSICoSIIS Aragón Zaragoza Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER‐BBN) Zaragoza Spain
| |
Collapse
|
46
|
Oh E, Kim JH, Um J, Jung DW, Williams DR, Lee H. Genome-Wide Transcriptomic Analysis of Non-Tumorigenic Tissues Reveals Aging-Related Prognostic Markers and Drug Targets in Renal Cell Carcinoma. Cancers (Basel) 2021; 13:3045. [PMID: 34207247 PMCID: PMC8234889 DOI: 10.3390/cancers13123045] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 01/03/2023] Open
Abstract
The relationship between expression of aging-related genes in normal tissues and cancer patient survival has not been assessed. We developed a genome-wide transcriptomic analysis approach for normal tissues adjacent to the tumor to identify aging-related transcripts associated with survival outcome, and applied it to 12 cancer types. As a result, five aging-related genes (DUSP22, MAPK14, MAPKAPK3, STAT1, and VCP) in normal tissues were found to be significantly associated with a worse survival outcome in patients with renal cell carcinoma (RCC). This computational approach was investigated using nontumorigenic immune cells purified from young and aged mice. Aged immune cells showed upregulated expression of all five aging-related genes and promoted RCC invasion compared to young immune cells. Further studies revealed DUSP22 as a regulator and druggable target of metastasis. DUSP22 gene knockdown reduced RCC invasion and the small molecule inhibitor BML-260 prevented RCC dissemination in a tumor/immune cell xenograft model. Overall, these results demonstrate that deciphering the relationship between aging-related gene expression in normal tissues and cancer patient survival can provide new prognostic markers, regulators of tumorigenesis and novel targets for drug development.
Collapse
Affiliation(s)
- Euiyoung Oh
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea;
| | - Jun-Hyeong Kim
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - JungIn Um
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - Da-Woon Jung
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - Darren R. Williams
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - Hyunju Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea;
| |
Collapse
|
47
|
Beehler K, Nikpay M, Lau P, Dang AT, Lagace TA, Soubeyrand S, McPherson R. A Common Polymorphism in the FADS1 Locus Links miR1908 to Low-Density Lipoprotein Cholesterol Through BMP1. Arterioscler Thromb Vasc Biol 2021; 41:2252-2262. [PMID: 34134519 DOI: 10.1161/atvbaha.121.316473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kaitlyn Beehler
- Atherogenomics Laboratory (K.B., M.N., P.L., A.-T.D., S.S., R.M.), University of Ottawa Heart Institute, Canada
| | - Majid Nikpay
- Atherogenomics Laboratory (K.B., M.N., P.L., A.-T.D., S.S., R.M.), University of Ottawa Heart Institute, Canada
| | - Paulina Lau
- Atherogenomics Laboratory (K.B., M.N., P.L., A.-T.D., S.S., R.M.), University of Ottawa Heart Institute, Canada
| | - Anh-Thu Dang
- Atherogenomics Laboratory (K.B., M.N., P.L., A.-T.D., S.S., R.M.), University of Ottawa Heart Institute, Canada
| | - Thomas A Lagace
- Lipoprotein Receptor Biology Laboratory (T.A.L.), University of Ottawa Heart Institute, Canada
| | - Sébastien Soubeyrand
- Atherogenomics Laboratory (K.B., M.N., P.L., A.-T.D., S.S., R.M.), University of Ottawa Heart Institute, Canada
| | - Ruth McPherson
- Atherogenomics Laboratory (K.B., M.N., P.L., A.-T.D., S.S., R.M.), University of Ottawa Heart Institute, Canada
| |
Collapse
|
48
|
Richard D, Capellini TD. Shifting epigenetic contexts influence regulatory variation and disease risk. Aging (Albany NY) 2021; 13:15699-15749. [PMID: 34138751 PMCID: PMC8266365 DOI: 10.18632/aging.203194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/01/2021] [Indexed: 11/25/2022]
Abstract
Epigenetic shifts are a hallmark of aging that impact transcriptional networks at regulatory level. These shifts may modify the effects of genetic regulatory variants during aging and contribute to disease pathomechanism. However, these shifts occur on the backdrop of epigenetic changes experienced throughout an individual's development into adulthood; thus, the phenotypic, and ultimately fitness, effects of regulatory variants subject to developmental- versus aging-related epigenetic shifts may differ considerably. Natural selection therefore may act differently on variants depending on their changing epigenetic context, which we propose as a novel lens through which to consider regulatory sequence evolution and phenotypic effects. Here, we define genomic regions subjected to altered chromatin accessibility as tissues transition from their fetal to adult forms, and subsequently from early to late adulthood. Based on these epigenomic datasets, we examine patterns of evolutionary constraint and potential functional impacts of sequence variation (e.g., genetic disease risk associations). We find that while the signals observed with developmental epigenetic changes are consistent with stronger fitness consequences (i.e., negative selection pressures), they tend to have weaker effects on genetic risk associations for aging-related diseases. Conversely, we see stronger effects of variants with increased local accessibility in adult tissues, strongest in young adult when compared to old. We propose a model for how epigenetic status of a region may influence the effects of evolutionary relevant sequence variation, and suggest that such a perspective on gene regulatory networks may elucidate our understanding of aging biology.
Collapse
Affiliation(s)
- Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
49
|
Ubaida-Mohien C, Moaddel R, Moore AZ, Kuo PL, Faghri F, Tharakan R, Tanaka T, Nalls MA, Ferrucci L. Proteomics and Epidemiological Models of Human Aging. Front Physiol 2021; 12:674013. [PMID: 34135771 PMCID: PMC8202502 DOI: 10.3389/fphys.2021.674013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/03/2021] [Indexed: 12/21/2022] Open
Abstract
Human aging is associated with a decline of physical and cognitive function and high susceptibility to chronic diseases, which is influenced by genetics, epigenetics, environmental, and socio-economic status. In order to identify the factors that modulate the aging process, established measures of aging mechanisms are required, that are both robust and feasible in humans. It is also necessary to connect these measures to the phenotypes of aging and their functional consequences. In this review, we focus on how this has been addressed from an epidemiologic perspective using proteomics. The key aspects of epidemiological models of aging can be incorporated into proteomics and other omics which can provide critical detailed information on the molecular and biological processes that change with age, thus unveiling underlying mechanisms that drive multiple chronic conditions and frailty, and ideally facilitating the identification of new effective approaches for prevention and treatment.
Collapse
Affiliation(s)
- Ceereena Ubaida-Mohien
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Ann Zenobia Moore
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Pei-Lun Kuo
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Faraz Faghri
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, United States
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
- Data Tecnica International, Glen Echo, MD, United States
| | - Ravi Tharakan
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Toshiko Tanaka
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Mike A. Nalls
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, United States
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
- Data Tecnica International, Glen Echo, MD, United States
| | - Luigi Ferrucci
- Biomedical Research Center, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| |
Collapse
|
50
|
Molecular evolution and the decline of purifying selection with age. Nat Commun 2021; 12:2657. [PMID: 33976227 PMCID: PMC8113359 DOI: 10.1038/s41467-021-22981-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/06/2021] [Indexed: 12/18/2022] Open
Abstract
Life history theory predicts that the intensity of selection declines with age, and this trend should impact how genes expressed at different ages evolve. Here we find consistent relationships between a gene’s age of expression and patterns of molecular evolution in two mammals (the human Homo sapiens and the mouse Mus musculus) and two insects (the malaria mosquito Anopheles gambiae and the fruit fly Drosophila melanogaster). When expressed later in life, genes fix nonsynonymous mutations more frequently, are more polymorphic for nonsynonymous mutations, and have shorter evolutionary lifespans, relative to those expressed early. The latter pattern is explained by a simple evolutionary model. Further, early-expressed genes tend to be enriched in similar gene ontology terms across species, while late-expressed genes show no such consistency. In humans, late-expressed genes are more likely to be linked to cancer and to segregate for dominant disease-causing mutations. Last, the effective strength of selection (Nes) decreases and the fraction of beneficial mutations increases with a gene’s age of expression. These results are consistent with the diminishing efficacy of purifying selection with age, as proposed by Medawar’s classic hypothesis for the evolution of senescence, and provide links between life history theory and molecular evolution. A fundamental principle of evolutionary theory is that the force of natural selection is weaker on traits expressed late in life relative to traits expressed early. Here, the authors find strong and consistent patterns of molecular evolution reflecting this principle in four species of animals, including humans.
Collapse
|