1
|
Pandkar MR, Shukla S. Epigenetics and alternative splicing in cancer: old enemies, new perspectives. Biochem J 2024; 481:1497-1518. [PMID: 39422322 DOI: 10.1042/bcj20240221] [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: 05/08/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
In recent years, significant strides in both conceptual understanding and technological capabilities have bolstered our comprehension of the factors underpinning cancer initiation and progression. While substantial insights have unraveled the molecular mechanisms driving carcinogenesis, there has been an overshadowing of the critical contribution made by epigenetic pathways, which works in concert with genetics. Mounting evidence demonstrates cancer as a complex interplay between genetics and epigenetics. Notably, epigenetic elements play a pivotal role in governing alternative pre-mRNA splicing, a primary contributor to protein diversity. In this review, we have provided detailed insights into the bidirectional communication between epigenetic modifiers and alternative splicing, providing examples of specific genes and isoforms affected. Notably, succinct discussion on targeting epigenetic regulators and the potential of the emerging field of epigenome editing to modulate splicing patterns is also presented. In summary, this review offers valuable insights into the intricate interplay between epigenetics and alternative splicing in cancer, paving the way for novel approaches to understanding and targeting this critical process.
Collapse
Affiliation(s)
- Madhura R Pandkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India
| |
Collapse
|
2
|
Madrid A, Papale LA, Bergmann PE, Breen C, Clark LR, Asthana S, Johnson SC, Keleş S, Hogan KJ, Alisch RS. Whole genome methylation sequencing in blood from persons with mild cognitive impairment and dementia due to Alzheimer's disease identifies cognitive status. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615196. [PMID: 39386499 PMCID: PMC11463426 DOI: 10.1101/2024.09.26.615196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Whole genome methylation sequencing (WGMS) in blood identifies differential DNA methylation in persons with late-onset dementia due to Alzheimer's disease (AD) but has not been tested in persons with mild cognitive impairment (MCI). METHODS We used WGMS to compare DNA methylation levels at 25,244,219 CpG loci in 382 blood samples from 99 persons with MCI, 109 with AD, and 174 who are cognitively unimpaired (CU). RESULTS WGMS identified 9,756 differentially methylated positions (DMPs) in persons with MCI, including 1,743 differentially methylated genes encoding proteins in biological pathways related to synapse organization, dendrite development, and ion transport. 447 DMPs exhibit progressively increasing or decreasing DNA methylation levels between CU, MCI, and AD that correspond to cognitive status. DISCUSSION WGMS identifies DMPs in known and newly detected genes in blood from persons with MCI and AD that support blood DNA methylation levels as candidate biomarkers of cognitive status.
Collapse
Affiliation(s)
- Andy Madrid
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Ligia A. Papale
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Phillip E. Bergmann
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Coleman Breen
- Department of Statistics, University of Wisconsin, Medical Sciences Center, 1300 University Ave Room 1220, Madison, WI 53706 USA
| | - Lindsay R. Clark
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 600 Highland Ave, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 600 Highland Ave, Madison, WI 53792, USA
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 600 Highland Ave, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Sündüz Keleş
- Department of Statistics, University of Wisconsin, Medical Sciences Center, 1300 University Ave Room 1220, Madison, WI 53706 USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Kirk J. Hogan
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| | - Reid S. Alisch
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792, USA
| |
Collapse
|
3
|
Minervini G. Feature Paper in Oral Physiology and Pathology. Life (Basel) 2024; 14:895. [PMID: 39063647 PMCID: PMC11278310 DOI: 10.3390/life14070895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
In the realm of life sciences, the journal 'Life' has consistently served as a beacon for groundbreaking research and scientific discovery [...].
Collapse
Affiliation(s)
- Giuseppe Minervini
- Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, Tamil Nadu, India;
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania “Luigi Vanvitelli”, 80121 Naples, Italy
| |
Collapse
|
4
|
Yin J, Fu J, Xu J, Chen C, Zhu H, Wang B, Yu C, Yang X, Cai R, Li M, Ji K, Wu W, Zhao Y, Zheng Z, Pu Y, Zheng L. Integrated analysis of m6A regulator-mediated RNA methylation modification patterns and immune characteristics in Sjögren's syndrome. Heliyon 2024; 10:e28645. [PMID: 38596085 PMCID: PMC11002070 DOI: 10.1016/j.heliyon.2024.e28645] [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: 01/23/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/11/2024] Open
Abstract
The epigenetic modifier N6-methyladenosine (m6A), recognized as the most prevalent internal modification in messenger RNA (mRNA), has recently emerged as a pivotal player in immune regulation. Its dysregulation has been implicated in the pathogenesis of various autoimmune conditions. However, the implications of m6A modification within the immune microenvironment of Sjögren's syndrome (SS), a chronic autoimmune disorder characterized by exocrine gland dysfunction, remain unexplored. Herein, we leverage an integrative analysis combining public database resources and novel sequencing data to investigate the expression profiles of m6A regulatory genes in SS. Our cohort comprised 220 patients diagnosed with SS and 62 healthy individuals, enabling a comprehensive evaluation of peripheral blood at the transcriptomic level. We report a significant association between SS and altered expression of key m6A regulators, with these changes closely tied to the activation of CD4+ T cells. Employing a random forest (RF) algorithm, we identified crucial genes contributing to the disease phenotype, which facilitated the development of a robust diagnostic model via multivariate logistic regression analysis. Further, unsupervised clustering revealed two distinct m6A modification patterns, which were significantly associated with variations in immunocyte infiltration, immune response activity, and biological function enrichment in SS. Subsequently, we proceeded with a screening process aimed at identifying genes that were differentially expressed (DEGs) between the two groups distinguished by m6A modification. Leveraging these DEGs, we employed weight gene co-expression network analysis (WGCNA) to uncover sets of genes that exhibited strong co-variance and hub genes that were closely linked to m6A modification. Through rigorous analysis, we identified three critical m6A regulators - METTL3, ALKBH5, and YTHDF1 - alongside two m6A-related hub genes, COMMD8 and SRP9. These elements collectively underscore a complex but discernible pattern of m6A modification that appears to be integrally linked with SS's pathogenesis. Our findings not only illuminate the significant correlation between m6A modification and the immune microenvironment in SS but also lay the groundwork for a deeper understanding of m6A regulatory mechanisms. More importantly, the identification of these key regulators and hub genes opens new avenues for the diagnosis and treatment of SS, presenting potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Junhao Yin
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Jiayao Fu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Jiabao Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Changyu Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Hanyi Zhu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Baoli Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Chuangqi Yu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Xiujuan Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Ruiyu Cai
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Mengyang Li
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Kaihan Ji
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Wanning Wu
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yijie Zhao
- Department of Oral and Maxillofacial Surgery, Shanghai Stomatological Hospital, Fudan University, 1258 Fuxin Zhong Road, Shanghai 200031, China
| | - Zhanglong Zheng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yiping Pu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| | - Lingyan Zheng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Institute of Stomatology, Shanghai, China
| |
Collapse
|
5
|
Caldo D, Massarini E, Rucci M, Deaglio S, Ferracini R. Epigenetics in Knee Osteoarthritis: A 2020-2023 Update Systematic Review. Life (Basel) 2024; 14:269. [PMID: 38398778 PMCID: PMC10890710 DOI: 10.3390/life14020269] [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: 12/29/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Osteoarthritis is a leading cause of disability in the world. The scientific literature highlights the critical importance of epigenetic regulatory effects, intertwined with biomechanical and biochemical peculiar conditions within each musculoskeletal district. While the contribution of genetic and epigenetic factors to knee OA is well-recognized, their precise role in disease management remains an area of active research. Such a field is particularly heterogeneous, calling for regular analysis and summarizing of the data that constantly emerge in the scientific literature, often sparse and scant of integration. The aim of this study was to systematically identify and synthesize all new evidence that emerged in human and animal model studies published between 2020 and 2023. This was necessary because, to the best of our knowledge, articles published before 2019 (and partly 2020) had already been included in systematic reviews that allowed to identify the ones concerning the knee joint. The review was carried out in accordance with Preferential Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Only peer-reviewed articles were considered for inclusion. A total of 40 studies were identified, showing promising results in terms either of biomarker identification, new insight in mechanism of action or potential therapeutic targets for knee OA. DNA methylation, histone modification and ncRNA were all mechanisms involved in epigenetic regulation of the knee. Most recent evidence suggests that epigenetics is a most promising field with the long-term goal of improving understanding and management of knee OA, but a variety of research approaches need greater consolidation.
Collapse
Affiliation(s)
- Davide Caldo
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Eugenia Massarini
- Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università di Genova, 16126 Genua, Italy
| | - Massimiliano Rucci
- Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università di Genova, 16126 Genua, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Riccardo Ferracini
- Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università di Genova, 16126 Genua, Italy
- Ospedale Koelliker, Corso Galileo Ferraris 247/255, 10134 Turin, Italy
| |
Collapse
|
6
|
Breen C, Papale LA, Clark LR, Bergmann PE, Madrid A, Asthana S, Johnson SC, Keleş S, Alisch RS, Hogan KJ. Whole genome methylation sequencing in blood identifies extensive differential DNA methylation in late-onset dementia due to Alzheimer's disease. Alzheimers Dement 2024; 20:1050-1062. [PMID: 37856321 PMCID: PMC10916976 DOI: 10.1002/alz.13514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/17/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION DNA microarray-based studies report differentially methylated positions (DMPs) in blood between late-onset dementia due to Alzheimer's disease (AD) and cognitively unimpaired individuals, but interrogate < 4% of the genome. METHODS We used whole genome methylation sequencing (WGMS) to quantify DNA methylation levels at 25,409,826 CpG loci in 281 blood samples from 108 AD and 173 cognitively unimpaired individuals. RESULTS WGMS identified 28,038 DMPs throughout the human methylome, including 2707 differentially methylated genes (e.g., SORCS3, GABA, and PICALM) encoding proteins in biological pathways relevant to AD such as synaptic membrane, cation channel complex, and glutamatergic synapse. One hundred seventy-three differentially methylated blood-specific enhancers interact with the promoters of 95 genes that are differentially expressed in blood from persons with and without AD. DISCUSSION WGMS identifies differentially methylated CpGs in known and newly detected genes and enhancers in blood from persons with and without AD. HIGHLIGHTS Whole genome DNA methylation levels were quantified in blood from persons with and without Alzheimer's disease (AD). Twenty-eight thousand thirty-eight differentially methylated positions (DMPs) were identified. Two thousand seven hundred seven genes comprise DMPs. Forty-eight of 75 independent genetic risk loci for AD have DMPs. One thousand five hundred sixty-eight blood-specific enhancers comprise DMPs, 173 of which interact with the promoters of 95 genes that are differentially expressed in blood from persons with and without AD.
Collapse
Affiliation(s)
- Coleman Breen
- Department of StatisticsUniversity of Wisconsin, Medical Sciences CenterMadisonWisconsinUSA
| | - Ligia A. Papale
- Department of Neurological SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Lindsay R. Clark
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton Memorial Veterans HospitalMadisonWisconsinUSA
| | - Phillip E. Bergmann
- Department of Neurological SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Andy Madrid
- Department of Neurological SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sanjay Asthana
- Geriatric Research Education and Clinical CenterWilliam S. Middleton Memorial Veterans HospitalMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton Memorial Veterans HospitalMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sündüz Keleş
- Department of StatisticsUniversity of Wisconsin, Medical Sciences CenterMadisonWisconsinUSA
- Department of Biostatistics and Medical InformaticsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Reid S. Alisch
- Department of Neurological SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Kirk J. Hogan
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of AnesthesiologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| |
Collapse
|
7
|
Ji Q, Jiang X, Wang M, Xin Z, Zhang W, Qu J, Liu GH. Multimodal Omics Approaches to Aging and Age-Related Diseases. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:56-71. [PMID: 38605908 PMCID: PMC11003952 DOI: 10.1007/s43657-023-00125-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 04/13/2024]
Abstract
Aging is associated with a progressive decline in physiological capacities and an increased risk of aging-associated disorders. An increasing body of experimental evidence shows that aging is a complex biological process coordinately regulated by multiple factors at different molecular layers. Thus, it is difficult to delineate the overall systematic aging changes based on single-layer data. Instead, multimodal omics approaches, in which data are acquired and analyzed using complementary omics technologies, such as genomics, transcriptomics, and epigenomics, are needed for gaining insights into the precise molecular regulatory mechanisms that trigger aging. In recent years, multimodal omics sequencing technologies that can reveal complex regulatory networks and specific phenotypic changes have been developed and widely applied to decode aging and age-related diseases. This review summarizes the classification and progress of multimodal omics approaches, as well as the rapidly growing number of articles reporting on their application in the field of aging research, and outlines new developments in the clinical treatment of age-related diseases based on omics technologies.
Collapse
Affiliation(s)
- Qianzhao Ji
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
| | - Xiaoyu Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
| | - Minxian Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zijuan Xin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100190 China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100190 China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053 China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| |
Collapse
|
8
|
Wu Q, Deng Z, Zhang W, Pan X, Choi KS, Zuo Y, Shen HB, Yu DJ. MLNGCF: circRNA-disease associations prediction with multilayer attention neural graph-based collaborative filtering. Bioinformatics 2023; 39:btad499. [PMID: 37561093 PMCID: PMC10457666 DOI: 10.1093/bioinformatics/btad499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/17/2023] [Accepted: 08/09/2023] [Indexed: 08/11/2023] Open
Abstract
MOTIVATION CircRNAs play a critical regulatory role in physiological processes, and the abnormal expression of circRNAs can mediate the processes of diseases. Therefore, exploring circRNAs-disease associations is gradually becoming an important area of research. Due to the high cost of validating circRNA-disease associations using traditional wet-lab experiments, novel computational methods based on machine learning are gaining more and more attention in this field. However, current computational methods suffer to insufficient consideration of latent features in circRNA-disease interactions. RESULTS In this study, a multilayer attention neural graph-based collaborative filtering (MLNGCF) is proposed. MLNGCF first enhances multiple biological information with autoencoder as the initial features of circRNAs and diseases. Then, by constructing a central network of different diseases and circRNAs, a multilayer cooperative attention-based message propagation is performed on the central network to obtain the high-order features of circRNAs and diseases. A neural network-based collaborative filtering is constructed to predict the unknown circRNA-disease associations and update the model parameters. Experiments on the benchmark datasets demonstrate that MLNGCF outperforms state-of-the-art methods, and the prediction results are supported by the literature in the case studies. AVAILABILITY AND IMPLEMENTATION The source codes and benchmark datasets of MLNGCF are available at https://github.com/ABard0/MLNGCF.
Collapse
Affiliation(s)
- Qunzhuo Wu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Zhaohong Deng
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Wei Zhang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Xiaoyong Pan
- Institute of Image Processing and Pattern Recognition, Shanghai Jiaotong University, Shanghai, China
| | - Kup-Sze Choi
- The Centre for Smart Health, The Hong Kong Polytechnic University, Hong Kong
| | - Yun Zuo
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Hong-Bin Shen
- Institute of Image Processing and Pattern Recognition, Shanghai Jiaotong University, Shanghai, China
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing, China
| |
Collapse
|
9
|
Kiełbowski K, Herian M, Bakinowska E, Banach B, Sroczyński T, Pawlik A. The Role of Genetics and Epigenetic Regulation in the Pathogenesis of Osteoarthritis. Int J Mol Sci 2023; 24:11655. [PMID: 37511413 PMCID: PMC10381003 DOI: 10.3390/ijms241411655] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Osteoarthritis (OA) is progressive disease characterised by cartilage degradation, subchondral bone remodelling and inflammation of the synovium. The disease is associated with obesity, mechanical load and age. However, multiple pro-inflammatory immune mediators regulate the expression of metalloproteinases, which take part in cartilage degradation. Furthermore, genetic factors also contribute to OA susceptibility. Recent studies have highlighted that epigenetic mechanisms may regulate the expression of OA-associated genes. This review aims to present the mechanisms of OA pathogenesis and summarise current evidence regarding the role of genetics and epigenetics in this process.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (M.H.); (E.B.); (B.B.); (T.S.)
| |
Collapse
|
10
|
Huang H, Dong X, Mao K, Pan W, Nie B, Jiang L. Identification of key candidate genes and pathways in rheumatoid arthritis and osteoarthritis by integrated bioinformatical analysis. Front Genet 2023; 14:1083615. [PMID: 36861127 PMCID: PMC9968929 DOI: 10.3389/fgene.2023.1083615] [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: 10/29/2022] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are the most common joint disorders. Although they have shown analogous clinical manifestations, the pathogenesis of RA and OA are different. In this study, we used the online Gene Expression Omnibus (GEO) microarray expression profiling dataset GSE153015 to identify gene signatures between RA and OA joints. The relevant data on 8 subjects obtained from large joints of RA patients (RA-LJ), 8 subjects obtained from small joints of RA patients (RA-SJ), and 4 subjects with OA were investigated. Differentially expressed genes (DEGs) were screened. Functional enrichment analysis of DEGs including the Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified, which were mainly associated with T cell activation or chemokine activity. Besides, protein-protein interaction (PPI) network analysis was performed, and key modules were identified. Hub genes of RA-LJ and OA groups were screened, they were CD8A, GZMB, CCL5, CD2, and CXCL9, whereas CD8A, CD2, IL7R, CD27, and GZMB were hub genes of RA-SJ and OA group. The novel DEGs and functional pathways between RA and OA identified in this study may provide new insight into the underlying molecular mechanisms and therapeutic strategies of RA and OA.
Collapse
Affiliation(s)
- Huijing Huang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyi Dong
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kaimin Mao
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wanwan Pan
- Yankuang New Journey General Hospital, Jingning, Shandong, China
| | - Bin’en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lindi Jiang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China,*Correspondence: Lindi Jiang,
| |
Collapse
|
11
|
Paterson C, Bozic I, Smith MJ, Hoad X, Evans DGR. A mechanistic mathematical model of initiation and malignant transformation in sporadic vestibular schwannoma. Br J Cancer 2022; 127:1843-1857. [PMID: 36097176 PMCID: PMC9643471 DOI: 10.1038/s41416-022-01955-8] [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: 10/28/2021] [Revised: 07/13/2022] [Accepted: 08/08/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND A vestibular schwannoma (VS) is a relatively rare, benign tumour of the eighth cranial nerve, often involving alterations to the gene NF2. Previous mathematical models of schwannoma incidence have not attempted to account for alterations in specific genes, and could not distinguish between nonsense mutations and loss of heterozygosity (LOH). METHODS Here, we present a mechanistic approach to modelling initiation and malignant transformation in schwannoma. Each parameter is associated with a specific gene or mechanism operative in Schwann cells, and can be determined by combining incidence data with empirical frequencies of pathogenic variants and LOH. RESULTS This results in new estimates for the base-pair mutation rate u = 4.48 × 10-10 and the rate of LOH = 2.03 × 10-6/yr in Schwann cells. In addition to new parameter estimates, we extend the approach to estimate the risk of both spontaneous and radiation-induced malignant transformation. DISCUSSION We conclude that radiotherapy is likely to have a negligible excess risk of malignancy for sporadic VS, with a possible exception of rapidly growing tumours.
Collapse
Affiliation(s)
- Chay Paterson
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK.
| | - Ivana Bozic
- Department of Applied Mathematics, University of Washington, Seattle, WA, USA
| | - Miriam J Smith
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Xanthe Hoad
- Radiation Protection Group, Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - D Gareth R Evans
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
12
|
Lu J, Annunziata F, Sirvinskas D, Omrani O, Li H, Rasa SMM, Krepelova A, Adam L, Neri F. Establishment and evaluation of module-based immune-associated gene signature to predict overall survival in patients of colon adenocarcinoma. J Biomed Sci 2022; 29:81. [PMID: 36229806 PMCID: PMC9563160 DOI: 10.1186/s12929-022-00867-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/04/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Patients with colon adenocarcinoma (COAD) exhibit significant heterogeneity in overall survival. The current tumor-node-metastasis staging system is insufficient to provide a precise prediction for prognosis. Identification and evaluation of new risk models by using big cancer data may provide a good way to identify prognosis-related signature. METHODS We integrated different datasets and applied bioinformatic and statistical methods to construct a robust immune-associated risk model for COAD prognosis. Furthermore, a nomogram was constructed based on the gene signature and clinicopathological features to improve risk stratification and quantify risk assessment for individual patients. RESULTS The immune-associated risk model discriminated high-risk patients in our investigated and validated cohorts. Survival analyses demonstrated that our gene signature served as an independent risk factor for overall survival and the nomogram exhibited high accuracy. Functional analysis interpreted the correlation between our risk model and its role in prognosis by classifying groups with different immune activities. Remarkably, patients in the low-risk group showed higher immune activity, while those in the high-risk group displayed a lower immune activity. CONCLUSIONS Our study provides a novel tool that may contribute to the optimization of risk stratification for survival and personalized management of COAD.
Collapse
Affiliation(s)
- Jing Lu
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Francesco Annunziata
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Dovydas Sirvinskas
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Omid Omrani
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Huahui Li
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Seyed Mohammad Mahdi Rasa
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Anna Krepelova
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Lisa Adam
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Francesco Neri
- grid.418245.e0000 0000 9999 5706Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany ,grid.7605.40000 0001 2336 6580Present Address: Life Sciences and Systems Biology Department, University of Torino, MBC, via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
13
|
Tong L, Yu H, Huang X, Shen J, Xiao G, Chen L, Wang H, Xing L, Chen D. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res 2022; 10:60. [PMID: 36127328 PMCID: PMC9489702 DOI: 10.1038/s41413-022-00226-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/27/2022] [Accepted: 06/19/2022] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease that causes painful swelling and permanent damage to the joints in the body. The molecular mechanisms of OA are currently unknown. OA is a heterogeneous disease that affects the entire joint, and multiple tissues are altered during OA development. To better understand the pathological mechanisms of OA, new approaches, methods, and techniques need to be used to understand OA pathogenesis. In this review, we first focus on the epigenetic regulation of OA, with a particular focus on DNA methylation, histone modification, and microRNA regulation, followed by a summary of several key mediators in OA-associated pain. We then introduce several innovative techniques that have been and will continue to be used in the fields of OA and OA-associated pain, such as CRISPR, scRNA sequencing, and lineage tracing. Next, we discuss the timely updates concerning cell death regulation in OA pathology, including pyroptosis, ferroptosis, and autophagy, as well as their individual roles in OA and potential molecular targets in treating OA. Finally, our review highlights new directions on the role of the synovial lymphatic system in OA. An improved understanding of OA pathogenesis will aid in the development of more specific and effective therapeutic interventions for OA.
Collapse
Affiliation(s)
- Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China
| | - Huan Yu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xingyun Huang
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jie Shen
- Department of Orthopedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huaiyu Wang
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lianping Xing
- Department of Pathology and Laboratory of Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China.
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
14
|
Kim M, Jiang X, Lauter K, Ismayilzada E, Shams S. Secure human action recognition by encrypted neural network inference. Nat Commun 2022; 13:4799. [PMID: 35970834 PMCID: PMC9378731 DOI: 10.1038/s41467-022-32168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
Advanced computer vision technology can provide near real-time home monitoring to support "aging in place" by detecting falls and symptoms related to seizures and stroke. Affordable webcams, together with cloud computing services (to run machine learning algorithms), can potentially bring significant social benefits. However, it has not been deployed in practice because of privacy concerns. In this paper, we propose a strategy that uses homomorphic encryption to resolve this dilemma, which guarantees information confidentiality while retaining action detection. Our protocol for secure inference can distinguish falls from activities of daily living with 86.21% sensitivity and 99.14% specificity, with an average inference latency of 1.2 seconds and 2.4 seconds on real-world test datasets using small and large neural nets, respectively. We show that our method enables a 613x speedup over the latency-optimized LoLa and achieves an average of 3.1x throughput increase in secure inference compared to the throughput-optimized nGraph-HE2.
Collapse
Affiliation(s)
- Miran Kim
- Department of Mathematics, Hanyang University, Seoul, Republic of Korea.
- Department of Computer Science, Hanyang University, Seoul, Republic of Korea.
| | - Xiaoqian Jiang
- Center for Secure Artificial intelligence For hEalthcare (SAFE), School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, USA
| | | | - Elkhan Ismayilzada
- Department of Computer Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Shayan Shams
- Department of Applied Data Science, San Jose State University, San Jose, CA, USA.
| |
Collapse
|
15
|
Brumwell A, Aubourg G, Hussain J, Parker E, Deehan DJ, Rice SJ, Loughlin J. Identification of TMEM129, encoding a ubiquitin-protein ligase, as an effector gene of osteoarthritis genetic risk. Arthritis Res Ther 2022; 24:189. [PMID: 35941660 PMCID: PMC9358880 DOI: 10.1186/s13075-022-02882-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoarthritis is highly heritable and genome-wide studies have identified single nucleotide polymorphisms (SNPs) associated with the disease. One such locus is marked by SNP rs11732213 (T > C). Genotype at rs11732213 correlates with the methylation levels of nearby CpG dinucleotides (CpGs), forming a methylation quantitative trait locus (mQTL). This study investigated the regulatory activity of the CpGs to identify a target gene of the locus. METHODS Nucleic acids were extracted from the articular cartilage of osteoarthritis patients. Samples were genotyped, and DNA methylation was quantified by pyrosequencing at 14 CpGs within a 259-bp interval. CpGs were tested for enhancer effects in immortalised chondrocytes using a reporter gene assay. DNA methylation at the locus was altered using targeted epigenome editing, with the impact on gene expression determined using quantitative polymerase chain reaction. RESULTS rs11732213 genotype correlated with DNA methylation at nine CpGs, which formed a differentially methylated region (DMR), with the osteoarthritis risk allele T corresponding to reduced levels of methylation. The DMR acted as an enhancer and demethylation of the CpGs altered expression of TMEM129. Allelic imbalance in TMEM129 expression was identified in cartilage, with under-expression of the risk allele. CONCLUSIONS TMEM129 is a target of osteoarthritis genetic risk at this locus. Genotype at rs11732213 impacts DNA methylation at the enhancer, which, in turn, modulates TMEM129 expression. TMEM129 encodes an enzyme involved in protein degradation within the endoplasmic reticulum, a process previously implicated in osteoarthritis. TMEM129 is a compelling osteoarthritis susceptibility target.
Collapse
Affiliation(s)
- Abby Brumwell
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, UK
| | - Guillaume Aubourg
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, UK
| | - Juhel Hussain
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, UK
| | - Eleanor Parker
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, UK
| | - David J Deehan
- Freeman Hospital, Newcastle University Teaching Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Sarah J Rice
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, UK
| | - John Loughlin
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, UK.
| |
Collapse
|
16
|
Albalwy F, McDermott JH, Newman WG, Brass A, Davies A. A blockchain-based framework to support pharmacogenetic data sharing. THE PHARMACOGENOMICS JOURNAL 2022; 22:264-275. [PMID: 35869255 DOI: 10.1038/s41397-022-00285-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 12/11/2022]
Abstract
The successful implementation of pharmacogenetics (PGx) into clinical practice requires patient genomic data to be shared between stakeholders in multiple settings. This creates a number of barriers to widespread adoption of PGx, including privacy concerns related to the storage and movement of identifiable genomic data. Informatic solutions that support secure and equitable data access for genomic data are therefore important to PGx. Here we propose a methodology that uses smart contracts implemented on a blockchain-based framework, PGxChain, to address this issue. The design requirements for PGxChain were identified through a systematic literature review, identifying technical challenges and barriers impeding the clinical implementation of pharmacogenomics. These requirements included security and privacy, accessibility, interoperability, traceability and legal compliance. A proof-of-concept implementation based on Ethereum was then developed that met the design requirements. PGxChain's performance was examined using Hyperledger Caliper for latency, throughput, and transaction success rate. The findings clearly indicate that blockchain technology offers considerable potential to advance pharmacogenetic data sharing, particularly with regard to PGx data security and privacy, large-scale accessibility of PGx data, PGx data interoperability between multiple health care providers and compliance with data-sharing laws and regulations.
Collapse
Affiliation(s)
- F Albalwy
- Department of Computer Science, Kilburn Building, University of Manchester, Oxford Road, Manchester, M13 9PL, UK. .,Department of Computer Science, College of Computer Science and Engineering, Taibah University, Madinah, Saudi Arabia. .,Division of Informatics, Imaging and Data Sciences, Stopford Building, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| | - J H McDermott
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK.,Division of Evolution Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - W G Newman
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK.,Division of Evolution Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - A Brass
- Department of Computer Science, Kilburn Building, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.,Division of Informatics, Imaging and Data Sciences, Stopford Building, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - A Davies
- Division of Informatics, Imaging and Data Sciences, Stopford Building, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
17
|
Samy A, Maher MA, Abdelsalam NA, Badr E. SARS-CoV-2 potential drugs, drug targets, and biomarkers: a viral-host interaction network-based analysis. Sci Rep 2022; 12:11934. [PMID: 35831333 PMCID: PMC9279364 DOI: 10.1038/s41598-022-15898-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/30/2022] [Indexed: 12/13/2022] Open
Abstract
COVID-19 is a global pandemic impacting the daily living of millions. As variants of the virus evolve, a complete comprehension of the disease and drug targets becomes a decisive duty. The Omicron variant, for example, has a notably high transmission rate verified in 155 countries. We performed integrative transcriptomic and network analyses to identify drug targets and diagnostic biomarkers and repurpose FDA-approved drugs for SARS-CoV-2. Upon the enrichment of 464 differentially expressed genes, pathways regulating the host cell cycle were significant. Regulatory and interaction networks featured hsa-mir-93-5p and hsa-mir-17-5p as blood biomarkers while hsa-mir-15b-5p as an antiviral agent. MYB, RRM2, ERG, CENPF, CIT, and TOP2A are potential drug targets for treatment. HMOX1 is suggested as a prognostic biomarker. Enhancing HMOX1 expression by neem plant extract might be a therapeutic alternative. We constructed a drug-gene network for FDA-approved drugs to be repurposed against the infection. The key drugs retrieved were members of anthracyclines, mitotic inhibitors, anti-tumor antibiotics, and CDK1 inhibitors. Additionally, hydroxyquinone and digitoxin are potent TOP2A inhibitors. Hydroxyurea, cytarabine, gemcitabine, sotalol, and amiodarone can also be redirected against COVID-19. The analysis enforced the repositioning of fluorouracil and doxorubicin, especially that they have multiple drug targets, hence less probability of resistance.
Collapse
Affiliation(s)
- Asmaa Samy
- University of Science and Technology, Zewail City, Giza, 12578, Egypt
| | - Mohamed A Maher
- University of Science and Technology, Zewail City, Giza, 12578, Egypt
| | - Nehal Adel Abdelsalam
- University of Science and Technology, Zewail City, Giza, 12578, Egypt.,Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Eman Badr
- University of Science and Technology, Zewail City, Giza, 12578, Egypt. .,Faculty of Computers and Artificial Intelligence, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
18
|
Gikaro JM, Xiong H, Lin F. Activity limitation and participation restriction in Osteoarthritis and Rheumatoid arthritis: findings based on the National Health and Nutritional Examination Survey. BMC Musculoskelet Disord 2022; 23:647. [PMID: 35794600 PMCID: PMC9258218 DOI: 10.1186/s12891-022-05607-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/27/2022] [Indexed: 11/12/2022] Open
Abstract
Background Osteoarthritis (OA) and Rheumatoid arthritis (RA) are the most common joint diseases leading to chronic pain and disability. Given the chronicity and disabling nature of OA and RA, they are likely to influence full participation of individuals in the society. An activity limitation occurs when a person has difficulty executing an activity; a participation restriction is experienced when a person has difficulty participating in a real-life situation. The aim of this study was to examine the associations between OA and RA and the domains of activity limitation and participation restriction. Methods A cross-sectional study design comprised 3604 adults from the 2009 to 2018 National Health and Nutrition Examination Survey (NHANES). All participants aged ≥ 20 years with complete data were included. Activity limitation and participation restriction were assessed by reported difficulty in performing 14 tasks selected from Physical Functioning Questionnaire. Data on OA and RA were obtained from Medical Conditions Questionnaire. Weighted logistic regression model was used to examine the associations between OA and RA and the selected tasks. Results Over 36% of participants had limitations. Both OA (OR = 2.11) and RA (OR = 2.36) were positively associated with activity limitation and participation restriction (p < 0.001). Poor or fair health was associated with difficulty in physical functioning, with highest odds observed in leisure activities (OR = 2.05), followed by difficulty in attending social events (OR = 1.99), walking for a quarter mile (OR = 1.97), preparing meals (OR = 1.93) and walking up ten steps (OR = 1.92). Conclusion Adults with OA and RA had nearly similar odds of having activity limitations and participation restrictions. Difficulty in executing most activities of daily living (ADLs) has significant association with poor or fair health. Holistic interdisciplinary care to individuals with OA or RA focusing on ADLs and environmental factors may improve health status.
Collapse
|
19
|
Oncogenic role and potential regulatory mechanism of topoisomerase IIα in a pan-cancer analysis. Sci Rep 2022; 12:11161. [PMID: 35778520 PMCID: PMC9249858 DOI: 10.1038/s41598-022-15205-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/20/2022] [Indexed: 11/08/2022] Open
Abstract
Topoisomerase IIα (TOP2A) plays an oncogenic role in multiple tumor types. However, no pan-cancer analysis about the function and the upstream molecular mechanism of TOP2A is available. For the first time, we analyzed potential oncogenic roles of TOP2A in 33 cancer types via The Cancer Genome Atlas (TCGA) database. Overexpression of TOP2A was existed in almost all cancer types, and related to poor prognosis and advanced pathological stages in most cases. Besides, the high frequency of TOP2A genetic alterations was observed in several cancer types, and related to prognosis in some cases. Moreover, we conduct upstream miRNAs and lncRNAs of TOP2A to establish ceRNA networks in kidney renal clear cell carcinoma (SNHG3-miR-139-5p), kidney renal papillary cell carcinoma (TMEM147-AS1/N4BP2L2-IT2/THUMPD3-AS1/ERICD/TTN-AS1/SH3BP5-AS1/THRB-IT1/SNHG3/NEAT1-miR-139-5p), liver hepatocellular carcinoma (SNHG3/THUMPD3-AS1/NUTM2B-AS1/NUTM2A-AS1-miR-139-5p and SNHG6/GSEC/SNHG1/SNHG14/LINC00265/MIR3142HG-miR-101-3p) and lung adenocarcinoma (TYMSOS/HELLPAR/SNHG1/GSEC/SNHG6-miR-101-3p). TOP2A expression was generally positively correlated with cancer associated fibroblasts, M0 and M1 macrophages in most cancer types. Furthermore, TOP2A was positively associated with expression of immune checkpoints (CD274, CTLA4, HAVCR2, LAG3, PDCD1 and TIGIT) in most cancer types. Our first TOP2A pan-cancer study contributes to understanding the prognostic roles, immunological roles and potential upstream molecular mechanism of TOP2A in different cancers.
Collapse
|
20
|
Teng Q, Liu Z, Song Y, Han K, Lu Y. A survey on the interpretability of deep learning in medical diagnosis. MULTIMEDIA SYSTEMS 2022; 28:2335-2355. [PMID: 35789785 PMCID: PMC9243744 DOI: 10.1007/s00530-022-00960-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
Deep learning has demonstrated remarkable performance in the medical domain, with accuracy that rivals or even exceeds that of human experts. However, it has a significant problem that these models are "black-box" structures, which means they are opaque, non-intuitive, and difficult for people to understand. This creates a barrier to the application of deep learning models in clinical practice due to lack of interpretability, trust, and transparency. To overcome this problem, several studies on interpretability have been proposed. Therefore, in this paper, we comprehensively review the interpretability of deep learning in medical diagnosis based on the current literature, including some common interpretability methods used in the medical domain, various applications with interpretability for disease diagnosis, prevalent evaluation metrics, and several disease datasets. In addition, the challenges of interpretability and future research directions are also discussed here. To the best of our knowledge, this is the first time that various applications of interpretability methods for disease diagnosis have been summarized.
Collapse
Affiliation(s)
- Qiaoying Teng
- School of Computer Science, Jilin Normal University, Siping, 136000 China
| | - Zhe Liu
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, 212013 China
| | - Yuqing Song
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, 212013 China
| | - Kai Han
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, 212013 China
| | - Yang Lu
- School of Computer Science, Jilin Normal University, Siping, 136000 China
| |
Collapse
|
21
|
Izda V, Martin J, Sturdy C, Jeffries MA. DNA methylation and noncoding RNA in OA: Recent findings and methodological advances. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 3. [PMID: 35360044 PMCID: PMC8966627 DOI: 10.1016/j.ocarto.2021.100208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Introduction: Osteoarthritis (OA) is a chronic musculoskeletal disease characterized by progressive loss of joint function. Historically, it has been characterized as a disease caused by mechanical trauma, so-called ‘wear and tear’. Over the past two decades, it has come to be understood as a complex systemic disorder involving gene-environmental interactions. Epigenetic changes have been increasingly implicated. Recent improvements in microarray and next-generation sequencing (NGS) technologies have allowed for ever more complex evaluations of epigenetic aberrations associated with the development and progression of OA. Methods: A systematic review was conducted in the Pubmed database. We curated studies that presented the results of DNA methylation and noncoding RNA research in human OA and OA animal models since 1985. Results: Herein, we discuss recent findings and methodological advancements in OA epigenetics, including a discussion of DNA methylation, including microarray and NGS studies, and noncoding RNAs. Beyond cartilage, we also highlight studies in subchondral bone and peripheral blood mononuclear cells, which highlight widespread and potentially clinically important alterations in epigenetic patterns seen in OA patients. Finally, we discuss epigenetic editing approaches in the context of OA. Conclusions: Although a substantial body of literature has already been published in OA, much is still unknown. Future OA epigenetics studies will no doubt continue to broaden our understanding of underlying pathophysiology and perhaps offer novel diagnostics and/or treatments for human OA.
Collapse
Affiliation(s)
- Vladislav Izda
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK, USA
| | - Jake Martin
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK, USA
| | - Cassandra Sturdy
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK, USA
| | - Matlock A. Jeffries
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK, USA
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, And Allergy, Oklahoma City, OK, USA
- Corresponding author. Oklahoma Medical Research Foundation, 825 NE 13th Street, Laboratory MC400, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
22
|
Salie MT, Yang J, Ramírez Medina CR, Zühlke LJ, Chishala C, Ntsekhe M, Gitura B, Ogendo S, Okello E, Lwabi P, Musuku J, Mtaja A, Hugo-Hamman C, El-Sayed A, Damasceno A, Mocumbi A, Bode-Thomas F, Yilgwan C, Amusa GA, Nkereuwem E, Shaboodien G, Da Silva R, Lee DCH, Frain S, Geifman N, Whetton AD, Keavney B, Engel ME. Data-independent acquisition mass spectrometry in severe rheumatic heart disease (RHD) identifies a proteomic signature showing ongoing inflammation and effectively classifying RHD cases. Clin Proteomics 2022; 19:7. [PMID: 35317720 PMCID: PMC8939134 DOI: 10.1186/s12014-022-09345-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rheumatic heart disease (RHD) remains a major source of morbidity and mortality in developing countries. A deeper insight into the pathogenetic mechanisms underlying RHD could provide opportunities for drug repurposing, guide recommendations for secondary penicillin prophylaxis, and/or inform development of near-patient diagnostics. METHODS We performed quantitative proteomics using Sequential Windowed Acquisition of All Theoretical Fragment Ion Mass Spectrometry (SWATH-MS) to screen protein expression in 215 African patients with severe RHD, and 230 controls. We applied a machine learning (ML) approach to feature selection among the 366 proteins quantifiable in at least 40% of samples, using the Boruta wrapper algorithm. The case-control differences and contribution to Area Under the Receiver Operating Curve (AUC) for each of the 56 proteins identified by the Boruta algorithm were calculated by Logistic Regression adjusted for age, sex and BMI. Biological pathways and functions enriched for proteins were identified using ClueGo pathway analyses. RESULTS Adiponectin, complement component C7 and fibulin-1, a component of heart valve matrix, were significantly higher in cases when compared with controls. Ficolin-3, a protein with calcium-independent lectin activity that activates the complement pathway, was lower in cases than controls. The top six biomarkers from the Boruta analyses conferred an AUC of 0.90 indicating excellent discriminatory capacity between RHD cases and controls. CONCLUSIONS These results support the presence of an ongoing inflammatory response in RHD, at a time when severe valve disease has developed, and distant from previous episodes of acute rheumatic fever. This biomarker signature could have potential utility in recognizing different degrees of ongoing inflammation in RHD patients, which may, in turn, be related to prognostic severity.
Collapse
Affiliation(s)
- M Taariq Salie
- AFROStrep Research Group, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Jing Yang
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Carlos R Ramírez Medina
- Division of Informatics, Imaging, and Data Sciences, University of Manchester, Manchester , UK
| | - Liesl J Zühlke
- Division of Paediatric Cardiology, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and University of Cape Town, Cape Town, South Africa
| | - Chishala Chishala
- Division of Cardiology, University of Cape Town & Groote Schuur Hospital, Cape Town, South Africa
| | - Mpiko Ntsekhe
- Division of Cardiology, University of Cape Town & Groote Schuur Hospital, Cape Town, South Africa
| | - Bernard Gitura
- Cardiology Department of Medicine, Kenyatta National Hospital, University of Nairobi, Nairobi, Kenya
| | - Stephen Ogendo
- Department of Surgery, University of Nairobi, Nairobi, Kenya
| | - Emmy Okello
- Departments of Adult and Pediatric Cardiology, Uganda Heart Institute, Kampala, Uganda
| | - Peter Lwabi
- Departments of Adult and Pediatric Cardiology, Uganda Heart Institute, Kampala, Uganda
| | - John Musuku
- University Teaching Hospital-Children's Hospital, University of Zambia, Lusaka, Zambia
| | - Agnes Mtaja
- University Teaching Hospital-Children's Hospital, University of Zambia, Lusaka, Zambia
| | - Christopher Hugo-Hamman
- Division of Paediatric Cardiology, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and University of Cape Town, Cape Town, South Africa
- Rheumatic Heart Disease Clinic, Windhoek Central Hospital, Windhoek, Namibia
| | - Ahmed El-Sayed
- Department of Cardiothoracic Surgery, Alshaab Teaching Hospital, Alazhari Health Research Center, Alzaiem Alazhari University, Khartoum, Sudan
| | - Albertino Damasceno
- Faculty of Medicine, Eduardo Mondlane University/Nucleo de Investigaçao, Departamento de Medicina, Hospital Central de Maputo, Maputo, Mozambique
| | - Ana Mocumbi
- Faculdade de Medicina, Universidade Eduardo Mondlane, Maputo, Mozambique
- Division of Non Communicable Diseases, Instituto Nacional de Saude, Vila de Marracuene, Mozambique
| | - Fidelia Bode-Thomas
- Departments of Paediatrics, Jos University Teaching Hospital, Jos, Plateau State, Nigeria
| | - Christopher Yilgwan
- Departments of Paediatrics, Jos University Teaching Hospital, Jos, Plateau State, Nigeria
| | - Ganiyu A Amusa
- Department of Medicine, University of Jos and Jos University Teaching Hospital, Jos, Nigeria
| | - Esin Nkereuwem
- Departments of Paediatrics, Jos University Teaching Hospital, Jos, Plateau State, Nigeria
| | - Gasnat Shaboodien
- Department of Medicine and Cape Heart Institute (CHI), University of Cape Town, Cape Town, South Africa
| | - Rachael Da Silva
- Stoller Biomarker Discovery Institute, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Dave Chi Hoo Lee
- Stoller Biomarker Discovery Institute, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Simon Frain
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Nophar Geifman
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Anthony D Whetton
- Faculty of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Manchester Heart Institute, Manchester University NHS Foundation Trust, Manchester, UK
| | - Mark E Engel
- AFROStrep Research Group, Department of Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
23
|
Computational identification of host genomic biomarkers highlighting their functions, pathways and regulators that influence SARS-CoV-2 infections and drug repurposing. Sci Rep 2022; 12:4279. [PMID: 35277538 PMCID: PMC8915158 DOI: 10.1038/s41598-022-08073-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 02/15/2022] [Indexed: 12/13/2022] Open
Abstract
The pandemic threat of COVID-19 has severely destroyed human life as well as the economy around the world. Although, the vaccination has reduced the outspread, but people are still suffering due to the unstable RNA sequence patterns of SARS-CoV-2 which demands supplementary drugs. To explore novel drug target proteins, in this study, a transcriptomics RNA-Seq data generated from SARS-CoV-2 infection and control samples were analyzed. We identified 109 differentially expressed genes (DEGs) that were utilized to identify 10 hub-genes/proteins (TLR2, USP53, GUCY1A2, SNRPD2, NEDD9, IGF2, CXCL2, KLF6, PAG1 and ZFP36) by the protein–protein interaction (PPI) network analysis. The GO functional and KEGG pathway enrichment analyses of hub-DEGs revealed some important functions and signaling pathways that are significantly associated with SARS-CoV-2 infections. The interaction network analysis identified 5 TFs proteins and 6 miRNAs as the key regulators of hub-DEGs. Considering 10 hub-proteins and 5 key TFs-proteins as drug target receptors, we performed their docking analysis with the SARS-CoV-2 3CL protease-guided top listed 90 FDA approved drugs. We found Torin-2, Rapamycin, Radotinib, Ivermectin, Thiostrepton, Tacrolimus and Daclatasvir as the top ranked seven candidate drugs. We investigated their resistance performance against the already published COVID-19 causing top-ranked 11 independent and 8 protonated receptor proteins by molecular docking analysis and found their strong binding affinities, which indicates that the proposed drugs are effective against the state-of-the-arts alternatives independent receptor proteins also. Finally, we investigated the stability of top three drugs (Torin-2, Rapamycin and Radotinib) by using 100 ns MD-based MM-PBSA simulations with the two top-ranked proposed receptors (TLR2, USP53) and independent receptors (IRF7, STAT1), and observed their stable performance. Therefore, the proposed drugs might play a vital role for the treatment against different variants of SARS-CoV-2 infections.
Collapse
|
24
|
Leung AWS, Leung HCM, Wong CL, Zheng ZX, Lui WW, Luk HM, Lo IFM, Luo R, Lam TW. ECNano: A cost-effective workflow for target enrichment sequencing and accurate variant calling on 4800 clinically significant genes using a single MinION flowcell. BMC Med Genomics 2022; 15:43. [PMID: 35246132 PMCID: PMC8895767 DOI: 10.1186/s12920-022-01190-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 02/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The application of long-read sequencing using the Oxford Nanopore Technologies (ONT) MinION sequencer is getting more diverse in the medical field. Having a high sequencing error of ONT and limited throughput from a single MinION flowcell, however, limits its applicability for accurate variant detection. Medical exome sequencing (MES) targets clinically significant exon regions, allowing rapid and comprehensive screening of pathogenic variants. By applying MES with MinION sequencing, the technology can achieve a more uniform capture of the target regions, shorter turnaround time, and lower sequencing cost per sample. METHOD We introduced a cost-effective optimized workflow, ECNano, comprising a wet-lab protocol and bioinformatics analysis, for accurate variant detection at 4800 clinically important genes and regions using a single MinION flowcell. The ECNano wet-lab protocol was optimized to perform long-read target enrichment and ONT library preparation to stably generate high-quality MES data with adequate coverage. The subsequent variant-calling workflow, Clair-ensemble, adopted a fast RNN-based variant caller, Clair, and was optimized for target enrichment data. To evaluate its performance and practicality, ECNano was tested on both reference DNA samples and patient samples. RESULTS ECNano achieved deep on-target depth of coverage (DoC) at average > 100× and > 98% uniformity using one MinION flowcell. For accurate ONT variant calling, the generated reads sufficiently covered 98.9% of pathogenic positions listed in ClinVar, with 98.96% having at least 30× DoC. ECNano obtained an average read length of 1000 bp. The long reads of ECNano also covered the adjacent splice sites well, with 98.5% of positions having ≥ 30× DoC. Clair-ensemble achieved > 99% recall and accuracy for SNV calling. The whole workflow from wet-lab protocol to variant detection was completed within three days. CONCLUSION We presented ECNano, an out-of-the-box workflow comprising (1) a wet-lab protocol for ONT target enrichment sequencing and (2) a downstream variant detection workflow, Clair-ensemble. The workflow is cost-effective, with a short turnaround time for high accuracy variant calling in 4800 clinically significant genes and regions using a single MinION flowcell. The long-read exon captured data has potential for further development, promoting the application of long-read sequencing in personalized disease treatment and risk prediction.
Collapse
Affiliation(s)
- Amy Wing-Sze Leung
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | | | - Chak-Lim Wong
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Zhen-Xian Zheng
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Wui-Wang Lui
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Ho-Ming Luk
- Department of Health, Clinical Genetic Service, Hong Kong, SAR, China
| | - Ivan Fai-Man Lo
- Department of Health, Clinical Genetic Service, Hong Kong, SAR, China
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, China.
| | - Tak-Wah Lam
- Department of Computer Science, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
25
|
Rego-Pérez I, Durán-Sotuela A, Ramos-Louro P, Blanco FJ. Genetic biomarkers in osteoarthritis: a quick overview. Fac Rev 2022; 10:78. [PMID: 35028644 PMCID: PMC8725648 DOI: 10.12703/r/10-78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Osteoarthritis (OA) is a chronic musculoskeletal disease with a polygenic and heterogeneous nature. In addition, when clinical manifestations appear, the evolution of the disease is usually already irreversible. Therefore, the efforts on OA research are focused mainly on the discovery of therapeutic targets and reliable biomarkers that permit the early identification of different OA-related parameters such as diagnosis, prognosis, or phenotype identification. To date, potential candidate protein biomarkers have been associated with different aspects of the disease; however, there is currently no gold standard. In this sense, genomic data could act as complementary biomarkers of diagnosis and prognosis or even help to identify therapeutic targets of the disease. In this review, we will describe the most recent advances in genetic biomarkers in OA over the past three years.
Collapse
Affiliation(s)
- Ignacio Rego-Pérez
- Unidad de Genómica. Grupo de Investigación en Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidade da Coruña (UDC). C/ As Xubias de Arriba 84, 15006, A Coruña, España
| | - Alejandro Durán-Sotuela
- Unidad de Genómica. Grupo de Investigación en Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidade da Coruña (UDC). C/ As Xubias de Arriba 84, 15006, A Coruña, España
| | - Paula Ramos-Louro
- Unidad de Genómica. Grupo de Investigación en Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidade da Coruña (UDC). C/ As Xubias de Arriba 84, 15006, A Coruña, España
| | - Francisco J Blanco
- Unidad de Genómica. Grupo de Investigación en Reumatología (GIR). Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidade da Coruña (UDC). C/ As Xubias de Arriba 84, 15006, A Coruña, España
- Universidade da Coruña (UDC), Grupo de Investigación en Reumatología y Salud. Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, 15008, A Coruña, España
| |
Collapse
|
26
|
Hosseini K, Ranjbar M, Pirpour Tazehkand A, Asgharian P, Montazersaheb S, Tarhriz V, Ghasemnejad T. Evaluation of exosomal non-coding RNAs in cancer using high-throughput sequencing. J Transl Med 2022; 20:30. [PMID: 35033106 PMCID: PMC8760667 DOI: 10.1186/s12967-022-03231-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022] Open
Abstract
Clinical oncologists need more reliable and non-invasive diagnostic and prognostic biomarkers to follow-up cancer patients. However, the existing biomarkers are often invasive and costly, emphasizing the need for the development of biomarkers to provide convenient and precise detection. Extracellular vesicles especially exosomes have recently been the focus of translational research to develop non-invasive and reliable biomarkers for several diseases such as cancers, suggesting as a valuable source of tumor markers. Exosomes are nano-sized extracellular vesicles secreted by various living cells that can be found in all body fluids including serum, urine, saliva, cerebrospinal fluid, and ascites. Different molecular and genetic contents of their origin such as nucleic acids, proteins, lipids, and glycans in a stable form make exosomes a promising approach for various cancers' diagnoses, prediction, and follow-up in a minimally invasive manner. Since exosomes are used by cancer cells for intercellular communication, they play a critical role in the disease process, highlighting the importance of their use as clinically relevant biomarkers. However, regardless of the advantages that exosome-based diagnostics have, they suffer from problems regarding their isolation, detection, and characterization of their contents. This study reviews the history and biogenesis of exosomes and discusses non-coding RNAs (ncRNAs) and their potential as tumor markers in different types of cancer, with a focus on next generation sequencing (NGS) as a detection method. Moreover, the advantages and challenges associated with exosome-based diagnostics are also presented.
Collapse
Affiliation(s)
- Kamran Hosseini
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Ranjbar
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Pirpour Tazehkand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parina Asgharian
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Tohid Ghasemnejad
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Gadepalli VS, Kim H, Liu Y, Han T, Cheng L. XDeathDB: a visualization platform for cell death molecular interactions. Cell Death Dis 2021; 12:1156. [PMID: 34907160 PMCID: PMC8669630 DOI: 10.1038/s41419-021-04397-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/07/2021] [Accepted: 11/09/2021] [Indexed: 12/23/2022]
Abstract
Lots of cell death initiator and effector molecules, signalling pathways and subcellular sites have been identified as key mediators in both cell death processes in cancer. The XDeathDB visualization platform provides a comprehensive cell death and their crosstalk resource for deciphering the signaling network organization of interactions among different cell death modes associated with 1461 cancer types and COVID-19, with an aim to understand the molecular mechanisms of physiological cell death in disease and facilitate systems-oriented novel drug discovery in inducing cell deaths properly. Apoptosis, autosis, efferocytosis, ferroptosis, immunogenic cell death, intrinsic apoptosis, lysosomal cell death, mitotic cell death, mitochondrial permeability transition, necroptosis, parthanatos, and pyroptosis related to 12 cell deaths and their crosstalk can be observed systematically by the platform. Big data for cell death gene-disease associations, gene-cell death pathway associations, pathway-cell death mode associations, and cell death-cell death associations is collected by literature review articles and public database from iRefIndex, STRING, BioGRID, Reactom, Pathway's commons, DisGeNET, DrugBank, and Therapeutic Target Database (TTD). An interactive webtool, XDeathDB, is built by web applications with R-Shiny, JavaScript (JS) and Shiny Server Iso. With this platform, users can search specific interactions from vast interdependent networks that occur in the realm of cell death. A multilayer spectral graph clustering method that performs convex layer aggregation to identify crosstalk function among cell death modes for a specific cancer. 147 hallmark genes of cell death could be observed in detail in these networks. These potential druggable targets are displayed systematically and tailoring networks to visualize specified relations is available to fulfil user-specific needs. Users can access XDeathDB for free at https://pcm2019.shinyapps.io/XDeathDB/ .
Collapse
Affiliation(s)
- Venkat Sundar Gadepalli
- Research Information Technology, College of Medicine, Ohio State University, 1585 Neil Ave, Columbus, OH, 43210, USA
| | - Hangil Kim
- 1Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Yueze Liu
- The Grainger College of Engineering, The University of Illinois-Urbana-Champaign, Urbana and Champaign, Champaign, IL, 61801, USA
| | - Tao Han
- 1Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Lijun Cheng
- 1Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
28
|
Chen J, Guo JT. Structural and functional analysis of somatic coding and UTR indels in breast and lung cancer genomes. Sci Rep 2021; 11:21178. [PMID: 34707120 PMCID: PMC8551294 DOI: 10.1038/s41598-021-00583-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/14/2021] [Indexed: 11/24/2022] Open
Abstract
Insertions and deletions (Indels) represent one of the major variation types in the human genome and have been implicated in diseases including cancer. To study the features of somatic indels in different cancer genomes, we investigated the indels from two large samples of cancer types: invasive breast carcinoma (BRCA) and lung adenocarcinoma (LUAD). Besides mapping somatic indels in both coding and untranslated regions (UTRs) from the cancer whole exome sequences, we investigated the overlap between these indels and transcription factor binding sites (TFBSs), the key elements for regulation of gene expression that have been found in both coding and non-coding sequences. Compared to the germline indels in healthy genomes, somatic indels contain more coding indels with higher than expected frame-shift (FS) indels in cancer genomes. LUAD has a higher ratio of deletions and higher coding and FS indel rates than BRCA. More importantly, these somatic indels in cancer genomes tend to locate in sequences with important functions, which can affect the core secondary structures of proteins and have a bigger overlap with predicted TFBSs in coding regions than the germline indels. The somatic CDS indels are also enriched in highly conserved nucleotides when compared with germline CDS indels.
Collapse
Affiliation(s)
- Jing Chen
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Jun-Tao Guo
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA.
| |
Collapse
|
29
|
Zhao X, Ji J, Wang S, Wang R, Yu Q, Li D. The regulatory pattern of target gene expression by aberrant enhancer methylation in glioblastoma. BMC Bioinformatics 2021; 22:420. [PMID: 34482818 PMCID: PMC8420065 DOI: 10.1186/s12859-021-04345-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/23/2021] [Indexed: 12/21/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most common and aggressive primary malignant brain tumor with grim prognosis. Aberrant DNA methylation is an epigenetic mechanism that promotes GBM carcinogenesis, while the function of DNA methylation at enhancer regions in GBM remains poorly described. Results We integrated multi-omics data to identify differential methylation enhancer region (DMER)-genes and revealed global enhancer hypomethylation in GBM. In addition, a DMER-mediated target genes regulatory network and functional enrichment analysis of target genes that might be regulated by hypomethylation enhancer regions showed that aberrant enhancer regions could contribute to tumorigenesis and progression in GBM. Further, we identified 22 modules in which lncRNAs and mRNAs synergistically competed with each other. Finally, through the construction of drug-target association networks, our study identified potential small-molecule drugs for GBM treatment. Conclusions Our study provides novel insights for understanding the regulation of aberrant enhancer region methylation and developing methylation-based biomarkers for the diagnosis and treatment of GBM. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04345-8.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Jianghuai Ji
- Department of Radiation Physics, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China.,Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310022, People's Republic of China
| | - Shijia Wang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Rendong Wang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Qiuhong Yu
- Department of Hyperbaric Oxygen, Beijing Tiantan Hospital, Capital Medical University, 119 Nansihuan Xi Lu, Fengtai District, Beijing, 100070, People's Republic of China.
| | - Dongguo Li
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing, 100069, People's Republic of China. .,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
30
|
Visconti VV, Cariati I, Fittipaldi S, Iundusi R, Gasbarra E, Tarantino U, Botta A. DNA Methylation Signatures of Bone Metabolism in Osteoporosis and Osteoarthritis Aging-Related Diseases: An Updated Review. Int J Mol Sci 2021; 22:ijms22084244. [PMID: 33921902 PMCID: PMC8072687 DOI: 10.3390/ijms22084244] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 01/03/2023] Open
Abstract
DNA methylation is one of the most studied epigenetic mechanisms that play a pivotal role in regulating gene expression. The epigenetic component is strongly involved in aging-bone diseases, such as osteoporosis and osteoarthritis. Both are complex multi-factorial late-onset disorders that represent a globally widespread health problem, highlighting a crucial point of investigations in many scientific studies. In recent years, new findings on the role of DNA methylation in the pathogenesis of aging-bone diseases have emerged. The aim of this systematic review is to update knowledge in the field of DNA methylation associated with osteoporosis and osteoarthritis, focusing on the specific tissues involved in both pathological conditions.
Collapse
Affiliation(s)
- Virginia Veronica Visconti
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.V.V.); (I.C.); (S.F.); (A.B.)
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (R.I.); (E.G.)
| | - Ida Cariati
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.V.V.); (I.C.); (S.F.); (A.B.)
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (R.I.); (E.G.)
| | - Simona Fittipaldi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.V.V.); (I.C.); (S.F.); (A.B.)
| | - Riccardo Iundusi
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (R.I.); (E.G.)
| | - Elena Gasbarra
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (R.I.); (E.G.)
| | - Umberto Tarantino
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (R.I.); (E.G.)
- Department of Clinical Science and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
- Correspondence:
| | - Annalisa Botta
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.V.V.); (I.C.); (S.F.); (A.B.)
| |
Collapse
|
31
|
Wang HY, Zhao JP, Zheng CH. SUSCC: Secondary Construction of Feature Space based on UMAP for Rapid and Accurate Clustering Large-scale Single Cell RNA-seq Data. Interdiscip Sci 2021; 13:83-90. [PMID: 33475958 DOI: 10.1007/s12539-020-00411-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/08/2020] [Accepted: 12/19/2020] [Indexed: 10/22/2022]
Abstract
Clustering is a common method to identify cell types in single cell analysis, but the increasing size of scRNA-seq datasets brings challenges to single cell clustering. Therefore, it is an urgent need to design a faster and more accurate clustering method for large-scale scRNA-seq data. In this paper, we proposed a new method for single cell clustering. First, a count matrix is constructed through normalization and gene filtration. Second, the raw data of gene expression matrix are projected to feature space constructed by secondary construction of feature space based on UMAP (Uniform Manifold Approximation and Projection). Third, the low-dimensional matrix on the feature space is randomly divided into two sub-matrices according to a certain proportion for clustering and classifying, respectively. Finally, one subset is clustered by k-means algorithm and then the other subset is classified by k-nearest neighbor algorithm based on clustering results. Experimental results show that our method can cluster the scRNA-seq datasets effectively.
Collapse
Affiliation(s)
- Hai-Yun Wang
- College of Mathematics and System Sciences, Xinjiang University, Urumqi, China
| | - Jian-Ping Zhao
- College of Mathematics and System Sciences, Xinjiang University, Urumqi, China. .,Institute of Mathematics and Physics, Xinjiang University, Urumqi, China.
| | - Chun-Hou Zheng
- College of Mathematics and System Sciences, Xinjiang University, Urumqi, China. .,College of Computer Science and Technology, Anhui University, Hefei, China.
| |
Collapse
|
32
|
Malemud CJ. Role of epigenetics in the pathogenesis and progression of osteoarthritis AK Sorial et al. Multi-tissue epigenetic analysis of the osteoarthritis susceptibility locus mapping to the plectin gene PLEC. Osteoarthritis Cartilage 2020; 28:1401-1402. [PMID: 32891768 DOI: 10.1016/j.joca.2020.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/24/2020] [Indexed: 02/02/2023]
Affiliation(s)
- C J Malemud
- Department of Medicine, Division of Rheumatic Diseases, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, Room 207, 2061 Cornell Road, Cleveland, Ohio, 44106-5076, United States.
| |
Collapse
|
33
|
Gardini ES, Chen GG, Fiacco S, Mernone L, Willi J, Turecki G, Ehlert U. Differential ESR1 Promoter Methylation in the Peripheral Blood-Findings from the Women 40+ Healthy Aging Study. Int J Mol Sci 2020; 21:E3654. [PMID: 32455834 PMCID: PMC7279168 DOI: 10.3390/ijms21103654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Background Estrogen receptor α (ERα) contributes to maintaining biological processes preserving health during aging. DNA methylation changes of ERα gene (ESR1) were established as playing a direct role in the regulation of ERα levels. In this study, we hypothesized decreased DNA methylation of ESR1 associated with postmenopause, lower estradiol (E2) levels, and increased age among healthy middle-aged and older women. Methods We assessed DNA methylation of ESR1 promoter region from dried blood spots (DBSs) and E2 from saliva samples in 130 healthy women aged 40-73 years. Results We found that postmenopause and lower E2 levels were associated with lower DNA methylation of a distal regulatory region, but not with DNA methylation of proximal promoters. Conclusion Our results indicate that decreased methylation of ESR1 cytosine-phosphate-guanine island (CpGI) shore may be associated with conditions of lower E2 in older healthy women.
Collapse
Affiliation(s)
- Elena S. Gardini
- Clinical Psychology and Psychotherapy, Department of Psychology, University of Zurich, 8050 Zurich, Switzerland; (E.S.G.); (S.F.); (L.M.); (J.W.)
- University Research Priority Program (URPP) Dynamics of Healthy Aging, University of Zurich, 8050 Zurich, Switzerland
| | - Gary G. Chen
- Douglas Hospital Research Center, McGill University, Montreal, QC H4H 1R3, Canada; (G.G.C.); (G.T.)
| | - Serena Fiacco
- Clinical Psychology and Psychotherapy, Department of Psychology, University of Zurich, 8050 Zurich, Switzerland; (E.S.G.); (S.F.); (L.M.); (J.W.)
- University Research Priority Program (URPP) Dynamics of Healthy Aging, University of Zurich, 8050 Zurich, Switzerland
| | - Laura Mernone
- Clinical Psychology and Psychotherapy, Department of Psychology, University of Zurich, 8050 Zurich, Switzerland; (E.S.G.); (S.F.); (L.M.); (J.W.)
- University Research Priority Program (URPP) Dynamics of Healthy Aging, University of Zurich, 8050 Zurich, Switzerland
| | - Jasmine Willi
- Clinical Psychology and Psychotherapy, Department of Psychology, University of Zurich, 8050 Zurich, Switzerland; (E.S.G.); (S.F.); (L.M.); (J.W.)
- University Research Priority Program (URPP) Dynamics of Healthy Aging, University of Zurich, 8050 Zurich, Switzerland
| | - Gustavo Turecki
- Douglas Hospital Research Center, McGill University, Montreal, QC H4H 1R3, Canada; (G.G.C.); (G.T.)
| | - Ulrike Ehlert
- Clinical Psychology and Psychotherapy, Department of Psychology, University of Zurich, 8050 Zurich, Switzerland; (E.S.G.); (S.F.); (L.M.); (J.W.)
- University Research Priority Program (URPP) Dynamics of Healthy Aging, University of Zurich, 8050 Zurich, Switzerland
| |
Collapse
|