1
|
Li Q, Wang J, Zhao C. From Genomics to Metabolomics: Molecular Insights into Osteoporosis for Enhanced Diagnostic and Therapeutic Approaches. Biomedicines 2024; 12:2389. [PMID: 39457701 PMCID: PMC11505085 DOI: 10.3390/biomedicines12102389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Osteoporosis (OP) is a prevalent skeletal disorder characterized by decreased bone mineral density (BMD) and increased fracture risk. The advancements in omics technologies-genomics, transcriptomics, proteomics, and metabolomics-have provided significant insights into the molecular mechanisms driving OP. These technologies offer critical perspectives on genetic predispositions, gene expression regulation, protein signatures, and metabolic alterations, enabling the identification of novel biomarkers for diagnosis and therapeutic targets. This review underscores the potential of these multi-omics approaches to bridge the gap between basic research and clinical applications, paving the way for precision medicine in OP management. By integrating these technologies, researchers can contribute to improved diagnostics, preventative strategies, and treatments for patients suffering from OP and related conditions.
Collapse
Affiliation(s)
- Qingmei Li
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Jihan Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
| | - Congzhe Zhao
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| |
Collapse
|
2
|
Zhang X, Wang Y, Zheng M, Wei Q, Zhang R, Zhu K, Zhai Q, Xu Y. IMPC-based screening revealed that ROBO1 can regulate osteoporosis by inhibiting osteogenic differentiation. Front Cell Dev Biol 2024; 12:1450215. [PMID: 39439909 PMCID: PMC11494888 DOI: 10.3389/fcell.2024.1450215] [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: 07/10/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction The utilization of denosumab in treating osteoporosis highlights promising prospects for osteoporosis intervention guided by gene targets. While omics-based research into osteoporosis pathogenesis yields a plethora of potential gene targets for clinical transformation, identifying effective gene targets has posed challenges. Methods We first queried the omics data of osteoporosis clinical samples on PubMed, used International Mouse Phenotyping Consortium (IMPC) to screen differentially expressed genes, and conducted preliminary functional verification of candidate genes in human Saos2 cells through osteogenic differentiation and mineralization experiments. We then selected the candidate genes with the most significant effects on osteogenic differentiation and further verified the osteogenic differentiation and mineralization functions in mouse 3T3-E1 and bone marrow mesenchymal stem cells (BMSC). Finally, we used RNA-seq to explore the regulation of osteogenesis by the target gene. Results We identified PPP2R2A, RRBP1, HSPB6, SLC22A15, ADAMTS4, ATP8B1, CTNNB1, ROBO1, and EFR3B, which may contribute to osteoporosis. ROBO1 was the most significant regulator of osteogenesis in both human and mouse osteoblast. The inhibitory effect of Robo1 knockdown on osteogenic differentiation may be related to the activation of inflammatory signaling pathways. Conclusion Our study provides several novel molecular mechanisms involved in the pathogenesis of osteoporosis. ROBO1 is a potential target for osteoporosis intervention.
Collapse
Affiliation(s)
- Xiangzheng Zhang
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yike Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Zheng
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Wei
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ruizhi Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Keyu Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiaocheng Zhai
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Youjia Xu
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
3
|
James R, Subramanyam KN, Payva F, E AP, Tv VK, Sivaramakrishnan V, Ks S. In-silico analysis predicts disruption of normal angiogenesis as a causative factor in osteoporosis pathogenesis. BMC Genom Data 2024; 25:85. [PMID: 39379846 PMCID: PMC11460074 DOI: 10.1186/s12863-024-01269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Angiogenesis-osteogenesis coupling is critical for proper functioning and maintaining the health of bones. Any disruption in this coupling, associated with aging and disease, might lead to loss of bone mass. Osteoporosis (OP) is a debilitating bone metabolic disorder that affects the microarchitecture of bones, gradually leading to fracture. Computational analysis revealed that normal angiogenesis is disrupted during the progression of OP, especially postmenopausal osteoporosis (PMOP). The genes associated with OP and PMOP were retrieved from the DisGeNET database. Hub gene analysis and molecular pathway enrichment were performed via the Cytoscape plugins STRING, MCODE, CytoHubba, ClueGO and the web-based tool Enrichr. Twenty-eight (28) hub genes were identified, eight of which were transcription factors (HIF1A, JUN, TP53, ESR1, MYC, PPARG, RUNX2 and SOX9). Analysis of SNPs associated with hub genes via the gnomAD, I-Mutant2.0, MUpro, ConSurf and COACH servers revealed the substitution F201L in IL6 as the most deleterious. The IL6 protein was modeled in the SWISS-MODEL server and the substitution was analyzed via the YASARA FoldX plugin. A positive ΔΔG (1.936) of the F201L mutant indicates that the mutated structure is less stable than the wild-type structure is. Thirteen hub genes, including IL6 and the enriched molecular pathways were found to be profoundly involved in angiogenesis/endothelial function and immune signaling. Mechanical loading of bones through weight-bearing exercises can activate osteoblasts via mechanotransduction leading to increased bone formation. The present study suggests proper mechanical loading of bone as a preventive strategy for PMOP, by which angiogenesis and the immune status of the bone can be maintained. This in silico analysis could be used to understand the molecular etiology of OP and to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Remya James
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India.
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India.
| | - Koushik Narayan Subramanyam
- Department of Orthopaedics, Sri Sathya Sai Institute of Higher Medical Sciences, Prasanthigram, Puttaparthi, Andhra Pradesh, 515134, India
| | - Febby Payva
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India
| | - Amrisa Pavithra E
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India
| | - Vineeth Kumar Tv
- Department of Zoology, The Cochin College, Kochi, Kerala, 682002, India.
| | - Venketesh Sivaramakrishnan
- School of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthinilayam, Puttaparthi, Andhra Pradesh, 515134, India
| | - Santhy Ks
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India.
| |
Collapse
|
4
|
Mardinoglu A, Palsson BØ. Genome-scale models in human metabologenomics. Nat Rev Genet 2024:10.1038/s41576-024-00768-0. [PMID: 39300314 DOI: 10.1038/s41576-024-00768-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/22/2024]
Abstract
Metabologenomics integrates metabolomics with other omics data types to comprehensively study the genetic and environmental factors that influence metabolism. These multi-omics data can be incorporated into genome-scale metabolic models (GEMs), which are highly curated knowledge bases that explicitly account for genes, transcripts, proteins and metabolites. By including all known biochemical reactions catalysed by enzymes and transporters encoded in the human genome, GEMs analyse and predict the behaviour of complex metabolic networks. Continued advancements to the scale and scope of GEMs - from cells and tissues to microbiomes and the whole body - have helped to design effective treatments and develop better diagnostic tools for metabolic diseases. Furthermore, increasing amounts of multi-omics data are incorporated into GEMs to better identify the underlying mechanisms, biomarkers and potential drug targets of metabolic diseases.
Collapse
Affiliation(s)
- Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.
| | - Bernhard Ø Palsson
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA.
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
5
|
Zhao C, Su KJ, Wu C, Cao X, Sha Q, Li W, Luo Z, Tian Q, Qiu C, Zhao LJ, Liu A, Jiang L, Zhang X, Shen H, Zhou W, Deng HW. Multi-scale variational autoencoder for imputation of missing values in untargeted metabolomics using whole-genome sequencing data. Comput Biol Med 2024; 179:108813. [PMID: 38955127 PMCID: PMC11324385 DOI: 10.1016/j.compbiomed.2024.108813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Missing data is a common challenge in mass spectrometry-based metabolomics, which can lead to biased and incomplete analyses. The integration of whole-genome sequencing (WGS) data with metabolomics data has emerged as a promising approach to enhance the accuracy of data imputation in metabolomics studies. METHOD In this study, we propose a novel method that leverages the information from WGS data and reference metabolites to impute unknown metabolites. Our approach utilizes a multi-scale variational autoencoder to jointly model the burden score, polygenetic risk score (PGS), and linkage disequilibrium (LD) pruned single nucleotide polymorphisms (SNPs) for feature extraction and missing metabolomics data imputation. By learning the latent representations of both omics data, our method can effectively impute missing metabolomics values based on genomic information. RESULTS We evaluate the performance of our method on empirical metabolomics datasets with missing values and demonstrate its superiority compared to conventional imputation techniques. Using 35 template metabolites derived burden scores, PGS and LD-pruned SNPs, the proposed methods achieved R2-scores > 0.01 for 71.55 % of metabolites. CONCLUSION The integration of WGS data in metabolomics imputation not only improves data completeness but also enhances downstream analyses, paving the way for more comprehensive and accurate investigations of metabolic pathways and disease associations. Our findings offer valuable insights into the potential benefits of utilizing WGS data for metabolomics data imputation and underscore the importance of leveraging multi-modal data integration in precision medicine research.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Computer Science, Kennesaw State University, 680 Arntson Dr, Marietta, GA 30060
| | - Kuan-Jui Su
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Chong Wu
- Department of Biostatistics, University of Texas MD Anderson, Pickens Academic Tower, 1400 Pressler St., Houston, TX 77030
| | - Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI, 49931
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, 1400 Townsend Dr, Houghton, MI, 49931
| | - Wu Li
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Zhe Luo
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Qing Tian
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Chuan Qiu
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Lan Juan Zhao
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Anqi Liu
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Lindong Jiang
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Xiao Zhang
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Hui Shen
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| | - Weihua Zhou
- Department of Applied Computing, Michigan Technological University, 1400 Townsend Dr, Houghton, MI, 49931
- Center for Biocomputing and Digital Health, Institute of Computing and Cybersystems, and Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Hong-Wen Deng
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112
| |
Collapse
|
6
|
Gutierrez Reyes CD, Alejo-Jacuinde G, Perez Sanchez B, Chavez Reyes J, Onigbinde S, Mogut D, Hernández-Jasso I, Calderón-Vallejo D, Quintanar JL, Mechref Y. Multi Omics Applications in Biological Systems. Curr Issues Mol Biol 2024; 46:5777-5793. [PMID: 38921016 PMCID: PMC11202207 DOI: 10.3390/cimb46060345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Traditional methodologies often fall short in addressing the complexity of biological systems. In this regard, system biology omics have brought invaluable tools for conducting comprehensive analysis. Current sequencing capabilities have revolutionized genetics and genomics studies, as well as the characterization of transcriptional profiling and dynamics of several species and sample types. Biological systems experience complex biochemical processes involving thousands of molecules. These processes occur at different levels that can be studied using mass spectrometry-based (MS-based) analysis, enabling high-throughput proteomics, glycoproteomics, glycomics, metabolomics, and lipidomics analysis. Here, we present the most up-to-date techniques utilized in the completion of omics analysis. Additionally, we include some interesting examples of the applicability of multi omics to a variety of biological systems.
Collapse
Affiliation(s)
| | - Gerardo Alejo-Jacuinde
- Department of Plant and Soil Science, Institute of Genomics for Crop Abiotic Stress Tolerance (IGCAST), Texas Tech University, Lubbock, TX 79409, USA; (G.A.-J.); (B.P.S.)
| | - Benjamin Perez Sanchez
- Department of Plant and Soil Science, Institute of Genomics for Crop Abiotic Stress Tolerance (IGCAST), Texas Tech University, Lubbock, TX 79409, USA; (G.A.-J.); (B.P.S.)
| | - Jesus Chavez Reyes
- Center of Basic Sciences, Department of Physiology and Pharmacology, Autonomous University of Aguascalientes, Aguascalientes 20392, Mexico; (J.C.R.); (I.H.-J.); (D.C.-V.); (J.L.Q.)
| | - Sherifdeen Onigbinde
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA;
| | - Damir Mogut
- Department of Food Biochemistry, Faculty of Food Science, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Irma Hernández-Jasso
- Center of Basic Sciences, Department of Physiology and Pharmacology, Autonomous University of Aguascalientes, Aguascalientes 20392, Mexico; (J.C.R.); (I.H.-J.); (D.C.-V.); (J.L.Q.)
| | - Denisse Calderón-Vallejo
- Center of Basic Sciences, Department of Physiology and Pharmacology, Autonomous University of Aguascalientes, Aguascalientes 20392, Mexico; (J.C.R.); (I.H.-J.); (D.C.-V.); (J.L.Q.)
| | - J. Luis Quintanar
- Center of Basic Sciences, Department of Physiology and Pharmacology, Autonomous University of Aguascalientes, Aguascalientes 20392, Mexico; (J.C.R.); (I.H.-J.); (D.C.-V.); (J.L.Q.)
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA;
| |
Collapse
|
7
|
Saeed NM, Ramadan LA, El-Sabbagh WA, Said MA, Abdel-Rahman HM, Mekky RH. Exploring the anti-osteoporosis potential of Petroselinum crispum (Mill.) Fuss extract employing experimentally ovariectomized rat model and network pharmacology approach. Fitoterapia 2024; 175:105971. [PMID: 38663562 DOI: 10.1016/j.fitote.2024.105971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/11/2024] [Accepted: 04/21/2024] [Indexed: 04/30/2024]
Abstract
One of the most prevalent secondary osteoporosis is ovariectomy-induced osteoporosis. Parsley (Petroselinum crispum) has potent estrogenic and antioxidant properties and was used traditionally in the treatment of amenorrhea and dysmenorrhea. The present study aimed to characterize parsley leaf extract (PLE) employing RP-HPLC-MS-MS/MS-based method and possible protective effect in ovariectomized (OVX)-induced osteoporosis in rats was assessed. Rats were randomly assigned into SHAM group, OVX group, PLE + OVX group (150 mg/kg/day, p.o), and estradiol benzoate (E2) + OVX group (30 μg/kg/day, s.c). After eight weeks following ovariectomy, biomarkers of bone strength, bone resorption, oxidative stress and histopathology were carried out. A network pharmacology approach investigated the key targets and potential mechanisms by of PLE metabolites against osteoporosis using databases: PubChem, BindingDB server, DisGeNET, ShinyGO, and KEGG Pathway. Moreover, FunRich 3.1.3, Cytoscape 3.10.0, and MOE 2019.0102 softwares were used for network pharmacology analysis and molecular docking studies. Flavones and hydroxycinnamic acid derivatives were predominant among 38 metabolites in PLE. It significantly restored bone strength and bone resorption biomarkers, osteocalcin (OST), oxidative stress biomarkers and histopathological alterations. The employed network pharmacology approach revealed that 14 primary target genes were associated with decreasing the severity of osteoporosis. Molecular docking revealed that cGMP-PKG signaling pathway has the highest fold enrichment and its downstream PDE5A. Luteolin, diosmetin, and isorhamnetin derivatives affected mostly osteoporosis targets. PLE exhibited protective action against ovariectomy-induced osteoporosis in rats and may be a promising therapy for premenopausal bone loss. cGMP-PKG signaling pathway could be a promising target for PLE in treating osteoporosis.
Collapse
Affiliation(s)
- Noha M Saeed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, 11829 Cairo, Egypt.
| | - Laila A Ramadan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, 11829 Cairo, Egypt
| | - Walaa A El-Sabbagh
- Drug Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), 11787 Cairo, Egypt
| | - Mohamed A Said
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo 11829, Egypt
| | - Hanaa M Abdel-Rahman
- Department of Pharmacy Practice, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt; Department of Forensic Medicine and Toxicology, Faculty of Medicine, Ain Shams University, Cairo 11562, Egypt
| | - Reham Hassan Mekky
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, 11829, Cairo, Egypt..
| |
Collapse
|
8
|
Li C, Pan H, Liu W, Jin G, Liu W, Liang C, Jiang X. Discovery of novel serum biomarkers for diagnosing and predicting postmenopausal osteoporosis patients by 4D-label free protein omics. J Orthop Res 2023; 41:2713-2720. [PMID: 37203779 DOI: 10.1002/jor.25628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/24/2023] [Accepted: 05/16/2023] [Indexed: 05/20/2023]
Abstract
We aimed to identify protein biomarkers that could rapidly and accurately diagnose osteoporosis patients (OPs) using a highly sensitive proteomic immunoassay. Four-dimensional (4D) label-free proteomics analysis was performed to determine the differentially expressed proteins in serum collected from 10 postmenopausal osteoporosis patients and 6 non-osteoporosis patients. The ELISA method was used to select the predicted proteins for verification. Serum was taken from 36 postmenopausal osteoporosis patients and 36 healthy individuals from normal postmenopausal women. Receiver operating characteristic (ROC) curves were used to determine the diagnostic potential of this method. We validated the expression of these six proteins using ELISA. The CDH1, IGFBP2, and VWF of osteoporosis patients were significantly higher than those of the normal group. PNP was significantly lower than that in the normal group. And using ROC curve calculation, serum CDH1 had a cut-off of 3.78 ng/mL with a sensitivity of 84.4%, and PNP had a cut-off of 944.32 ng/mL with 88.9% sensitivity. These outcomes suggest that serum-level CHD1 and PNP have the potential power as effective indicators for the diagnosis of PMOP. Our results suggest that CHD1 and PNP might be associated with the pathogenesis of OP and would be helpful in diagnosing OP. Therefore, CHD1 and PNP may act as potential key markers in OP.
Collapse
Affiliation(s)
- Chunyan Li
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, China
- Department of Laboratory Medicine, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, Xicheng District, Beijing, China
| | - Haizhou Pan
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University, Hangzhou, China
- College of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Liu
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, China
| | - Guohong Jin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, China
| | - Wuzheng Liu
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, China
| | - Cuiying Liang
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, China
- Department of Laboratory Medicine, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, Xicheng District, Beijing, China
| | - Xieyuan Jiang
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, The 4th Medical College of Peking University, Beijing, China
| |
Collapse
|
9
|
Zhao C, Keyak JH, Cao X, Sha Q, Wu L, Luo Z, Zhao LJ, Tian Q, Serou M, Qiu C, Su KJ, Shen H, Deng HW, Zhou W. Multi-view information fusion using multi-view variational autoencoder to predict proximal femoral fracture load. Front Endocrinol (Lausanne) 2023; 14:1261088. [PMID: 38075049 PMCID: PMC10710145 DOI: 10.3389/fendo.2023.1261088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Background Hip fracture occurs when an applied force exceeds the force that the proximal femur can support (the fracture load or "strength") and can have devastating consequences with poor functional outcomes. Proximal femoral strengths for specific loading conditions can be computed by subject-specific finite element analysis (FEA) using quantitative computerized tomography (QCT) images. However, the radiation and availability of QCT limit its clinical usability. Alternative low-dose and widely available measurements, such as dual energy X-ray absorptiometry (DXA) and genetic factors, would be preferable for bone strength assessment. The aim of this paper is to design a deep learning-based model to predict proximal femoral strength using multi-view information fusion. Results We developed new models using multi-view variational autoencoder (MVAE) for feature representation learning and a product of expert (PoE) model for multi-view information fusion. We applied the proposed models to an in-house Louisiana Osteoporosis Study (LOS) cohort with 931 male subjects, including 345 African Americans and 586 Caucasians. We performed genome-wide association studies (GWAS) to select 256 genetic variants with the lowest p-values for each proximal femoral strength and integrated whole genome sequence (WGS) features and DXA-derived imaging features to predict proximal femoral strength. The best prediction model for fall fracture load was acquired by integrating WGS features and DXA-derived imaging features. The designed models achieved the mean absolute percentage error of 18.04%, 6.84% and 7.95% for predicting proximal femoral fracture loads using linear models of fall loading, nonlinear models of fall loading, and nonlinear models of stance loading, respectively. Conclusion The proposed models are capable of predicting proximal femoral strength using WGS features and DXA-derived imaging features. Though this tool is not a substitute for predicting FEA using QCT images, it would make improved assessment of hip fracture risk more widely available while avoiding the increased radiation exposure from QCT.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Applied Computing, Michigan Technological University, Houghton, MI, United States
| | - Joyce H. Keyak
- Department of Radiological Sciences, Department of Biomedical Engineering, Department of Mechanical and Aerospace Engineering, and Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, United States
| | - Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Li Wu
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Zhe Luo
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Lan-Juan Zhao
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Qing Tian
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Michael Serou
- Department of Radiology, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Chuan Qiu
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Kuan-Jui Su
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Hui Shen
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Hong-Wen Deng
- Division of Biomedical Informatics and Genomics, Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Weihua Zhou
- Department of Applied Computing, Michigan Technological University, Houghton, MI, United States
- Center for Biocomputing and Digital Health, Institute of Computing and Cybersystems, and Health Research Institute, Michigan Technological University, Houghton, MI, United States
| |
Collapse
|
10
|
Guo H, Lv X, Li Y, Li M. Attention-based GCN integrates multi-omics data for breast cancer subtype classification and patient-specific gene marker identification. Brief Funct Genomics 2023; 22:463-474. [PMID: 37114942 DOI: 10.1093/bfgp/elad013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/16/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer is a heterogeneous disease and can be divided into several subtypes with unique prognostic and molecular characteristics. The classification of breast cancer subtypes plays an important role in the precision treatment and prognosis of breast cancer. Benefitting from the relation-aware ability of a graph convolution network (GCN), we present a multi-omics integrative method, the attention-based GCN (AGCN), for breast cancer molecular subtype classification using messenger RNA expression, copy number variation and deoxyribonucleic acid methylation multi-omics data. In the extensive comparative studies, our AGCN models outperform state-of-the-art methods under different experimental conditions and both attention mechanisms and the graph convolution subnetwork play an important role in accurate cancer subtype classification. The layer-wise relevance propagation (LRP) algorithm is used for the interpretation of model decision, which can identify patient-specific important biomarkers that are reported to be related to the occurrence and development of breast cancer. Our results highlighted the effectiveness of the GCN and attention mechanisms in multi-omics integrative analysis and the implement of the LRP algorithm can provide biologically reasonable insights into model decision.
Collapse
Affiliation(s)
- Hui Guo
- College of Chemistry at Sichuan University
| | - Xiang Lv
- College of Chemistry at Sichuan University
| | - Yizhou Li
- College of Cyber Science and Engineering at Sichuan University
| | | |
Collapse
|
11
|
Marino C, Pagano I, Castaldo G, Grimaldi M, D’Elia M, Santoro A, Conte A, Molettieri P, Parisella C, Buonocore M, D’Ursi AM, Rastrelli L. Supplementing Low-Sodium Bicarbonate-Calcic (Lete) ® Water: Effects in Women on Bone and Systemic Metabolism. Metabolites 2023; 13:1109. [PMID: 37999205 PMCID: PMC10673306 DOI: 10.3390/metabo13111109] [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: 09/16/2023] [Revised: 10/03/2023] [Accepted: 10/19/2023] [Indexed: 11/25/2023] Open
Abstract
Calcium (Ca) represents about 40% of the total mineral mass, mainly in the bone, providing mechanical strength to the skeleton and teeth. An adequate Ca intake is necessary for bone growth and development in children and adolescents and for maintaining bone mineral loss in elderly age. Ca deficiency predisposes to osteopenia and osteoporosis. Healthy nutrition, including an adequate intake of Ca-rich food, is paramount to prevent and cure osteoporosis. Recently, several clinical studies have demonstrated that, in conditions of Ca dysmetabolism, Ca-rich mineral water is beneficial as a valuable source of Ca to be used as an alternative to caloric Ca-rich dairy products. Although promising, these data have been collected from small groups of participants. Moreover, they mainly regard the effect of Ca-rich mineral water on bone metabolism. In contrast, an investigation of the effect of Ca supplementation on systemic metabolism is needed to address the spreading of systemic metabolic dysfunction often associated with Ca dysmetabolism. In the present study, we analyzed urine and blood sera of 120 women in perimenopausal condition who were subjected for six months to 2l daily consumption of bicarbonate-calcium mineral water marketed under ®Lete. Remarkably, this water, in addition to being rich in calcium and bicarbonate, is also low in sodium. A complete set of laboratory tests was carried out to investigate whether the specific water composition was such to confirm the known therapeutic effects on bone metabolism. Second, but not least, urine and blood sera were analyzed using NMR-based metabolomic procedures to investigate, other than the action on Ca metabolism, potential system-wide metabolic effects. Our data show that Lete water is a valid supplement for compensating for Ca dysmetabolism and preserving bone health and integrity.
Collapse
Affiliation(s)
- Carmen Marino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (C.M.); (I.P.); (M.G.); (M.D.); (A.S.)
- Department of Pharmacy and Ph.D. Program in Drug Discovery and Development, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Imma Pagano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (C.M.); (I.P.); (M.G.); (M.D.); (A.S.)
- NutriKeto_LAB Unisa—“San Giuseppe Moscati” National Hospital (AORN), Contrada Amoretta, 83100 Avellino, Italy; (G.C.); (A.C.); (P.M.); (C.P.)
| | - Giuseppe Castaldo
- NutriKeto_LAB Unisa—“San Giuseppe Moscati” National Hospital (AORN), Contrada Amoretta, 83100 Avellino, Italy; (G.C.); (A.C.); (P.M.); (C.P.)
| | - Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (C.M.); (I.P.); (M.G.); (M.D.); (A.S.)
| | - Maria D’Elia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (C.M.); (I.P.); (M.G.); (M.D.); (A.S.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Department of Pharmacy, Scuola di Specializzazione in Farmacia Ospedaliera, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Angelo Santoro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (C.M.); (I.P.); (M.G.); (M.D.); (A.S.)
- Department of Pharmacy, Scuola di Specializzazione in Farmacia Ospedaliera, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Aurelio Conte
- NutriKeto_LAB Unisa—“San Giuseppe Moscati” National Hospital (AORN), Contrada Amoretta, 83100 Avellino, Italy; (G.C.); (A.C.); (P.M.); (C.P.)
| | - Paola Molettieri
- NutriKeto_LAB Unisa—“San Giuseppe Moscati” National Hospital (AORN), Contrada Amoretta, 83100 Avellino, Italy; (G.C.); (A.C.); (P.M.); (C.P.)
| | - Chiara Parisella
- NutriKeto_LAB Unisa—“San Giuseppe Moscati” National Hospital (AORN), Contrada Amoretta, 83100 Avellino, Italy; (G.C.); (A.C.); (P.M.); (C.P.)
| | - Michela Buonocore
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant’Angelo, Via Cinthia 21, 80126 Naples, Italy;
| | - Anna Maria D’Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (C.M.); (I.P.); (M.G.); (M.D.); (A.S.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Luca Rastrelli
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (C.M.); (I.P.); (M.G.); (M.D.); (A.S.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
12
|
Mullin BH, Ribet ABP, Pavlos NJ. Bone Trans-omics: Integrating Omics to Unveil Mechanistic Molecular Networks Regulating Bone Biology and Disease. Curr Osteoporos Rep 2023; 21:493-502. [PMID: 37410317 PMCID: PMC10543827 DOI: 10.1007/s11914-023-00812-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE OF REVIEW Recent advancements in "omics" technologies and bioinformatics have afforded researchers new tools to study bone biology in an unbiased and holistic way. The purpose of this review is to highlight recent studies integrating multi-omics data gathered from multiple molecular layers (i.e.; trans-omics) to reveal new molecular mechanisms that regulate bone biology and underpin skeletal diseases. RECENT FINDINGS Bone biologists have traditionally relied on single-omics technologies (genomics, transcriptomics, proteomics, and metabolomics) to profile measureable differences (both qualitative and quantitative) of individual molecular layers for biological discovery and to investigate mechanisms of disease. Recently, literature has grown on the implementation of integrative multi-omics to study bone biology, which combines computational and informatics support to connect multiple layers of data derived from individual "omic" platforms. This emerging discipline termed "trans-omics" has enabled bone biologists to identify and construct detailed molecular networks, unveiling new pathways and unexpected interactions that have advanced our mechanistic understanding of bone biology and disease. While the era of trans-omics is poised to revolutionize our capacity to answer more complex and diverse questions pertinent to bone pathobiology, it also brings new challenges that are inherent when trying to connect "Big Data" sets. A concerted effort between bone biologists and interdisciplinary scientists will undoubtedly be needed to extract physiologically and clinically meaningful data from bone trans-omics in order to advance its implementation in the field.
Collapse
Affiliation(s)
- Benjamin H Mullin
- Bone Biology & Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, 2nd Floor "M" Block QEII Medical Centre, Nedlands, WA, 6009, Australia
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
| | - Amy B P Ribet
- Bone Biology & Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, 2nd Floor "M" Block QEII Medical Centre, Nedlands, WA, 6009, Australia
| | - Nathan J Pavlos
- Bone Biology & Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, 2nd Floor "M" Block QEII Medical Centre, Nedlands, WA, 6009, Australia.
| |
Collapse
|
13
|
Lee RH, Bain J, Muehlbauer M, Ilkayeva O, Pieper C, Wixted D, Colón-Emeric C. Metabolic factors associated with incident fracture among older adults with type 2 diabetes mellitus: a nested case-control study. Osteoporos Int 2023; 34:1263-1268. [PMID: 37100949 PMCID: PMC10443052 DOI: 10.1007/s00198-023-06763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/18/2023] [Indexed: 04/28/2023]
Abstract
Older adults with type 2 diabetes mellitus have an increased risk of fracture despite a paradoxically higher average bone mineral density. This study identified additional markers of fracture risk in this at-risk population. Non-esterified fatty acids and the amino acids glutamine/glutamate and asparagine/aspartate were associated with incident fractures. PURPOSE Type 2 diabetes mellitus (T2D) is associated with an increased risk of fracture despite a paradoxically higher bone mineral density. Additional markers of fracture risk are needed to identify at-risk individuals. METHOD The MURDOCK study is an ongoing study, initiated in 2007, of residents in central North Carolina. At enrollment, participants completed health questionnaires and provided biospecimen samples. In this nested case-control analysis, incident fractures among adults with T2D, age ≥ 50 years, were identified by self-report and electronic medical record query. Fracture cases were matched 1:2 by age, gender, race/ethnicity, and BMI to those without incident fracture. Stored sera were analyzed for conventional metabolites and targeted metabolomics (amino acids and acylcarnitines). The association between incident fracture and metabolic profile was assessed using conditional logistic regression, controlled for multiple confounders including tobacco and alcohol use, medical comorbidities, and medications. RESULTS 107 incident fractures were identified with 210 matched controls. Targeted metabolomics analysis included 2 amino acid factors, consisting of: 1) the branched chain amino acids, phenylalanine and tyrosine; and 2) glutamine/glutamate, asparagine/aspartate, arginine, and serine [E/QD/NRS]. After controlling for multiple risk factors, E/QD/NRS was significantly associated with incident fracture (OR 2.50, 95% CI: 1.36-4.63). Non-esterified fatty acids were associated with lower odds of fracture (OR 0.17, 95% CI: 0.03-0.87). There were no associations with fracture among other conventional metabolites, acylcarnitine factors, nor the other amino acid factors. CONCLUSION Our results indicate novel biomarkers, and suggest potential mechanisms, of fracture risk among older adults with T2D.
Collapse
Affiliation(s)
- Richard H Lee
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Center for the Study of Aging and Human Development, Duke University, Durham, NC, USA.
| | - James Bain
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Center for the Study of Aging and Human Development, Duke University, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Michael Muehlbauer
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Carl Pieper
- Center for the Study of Aging and Human Development, Duke University, Durham, NC, USA
| | - Doug Wixted
- Duke Clinical and Translational Science Institute, Durham, NC, USA
| | - Cathleen Colón-Emeric
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Center for the Study of Aging and Human Development, Duke University, Durham, NC, USA
| |
Collapse
|
14
|
Kwoji ID, Aiyegoro OA, Okpeku M, Adeleke MA. 'Multi-omics' data integration: applications in probiotics studies. NPJ Sci Food 2023; 7:25. [PMID: 37277356 DOI: 10.1038/s41538-023-00199-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
The concept of probiotics is witnessing increasing attention due to its benefits in influencing the host microbiome and the modulation of host immunity through the strengthening of the gut barrier and stimulation of antibodies. These benefits, combined with the need for improved nutraceuticals, have resulted in the extensive characterization of probiotics leading to an outburst of data generated using several 'omics' technologies. The recent development in system biology approaches to microbial science is paving the way for integrating data generated from different omics techniques for understanding the flow of molecular information from one 'omics' level to the other with clear information on regulatory features and phenotypes. The limitations and tendencies of a 'single omics' application to ignore the influence of other molecular processes justify the need for 'multi-omics' application in probiotics selections and understanding its action on the host. Different omics techniques, including genomics, transcriptomics, proteomics, metabolomics and lipidomics, used for studying probiotics and their influence on the host and the microbiome are discussed in this review. Furthermore, the rationale for 'multi-omics' and multi-omics data integration platforms supporting probiotics and microbiome analyses was also elucidated. This review showed that multi-omics application is useful in selecting probiotics and understanding their functions on the host microbiome. Hence, recommend a multi-omics approach for holistically understanding probiotics and the microbiome.
Collapse
Affiliation(s)
- Iliya Dauda Kwoji
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090, Durban, South Africa
| | - Olayinka Ayobami Aiyegoro
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, Northwest, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090, Durban, South Africa
| | - Matthew Adekunle Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090, Durban, South Africa.
| |
Collapse
|
15
|
Yang J, Wu J. Discovery of potential biomarkers for osteoporosis diagnosis by individual omics and multi-omics technologies. Expert Rev Mol Diagn 2023:1-16. [PMID: 37140363 DOI: 10.1080/14737159.2023.2208750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
INTRODUCTION Global aging has made osteoporosis an increasingly serious public health problem. Osteoporotic fractures seriously affect the quality of life of patients and increase disability and mortality rates. Early diagnosis is important for timely intervention. The continuous development of individual- and multi-omics methods is helpful for the exploration and discovery of biomarkers for the diagnosis of osteoporosis. AREAS COVERED In this review, we first introduce the epidemiological status of osteoporosis and then describe the pathogenesis of osteoporosis. Furthermore, the latest progress in individual- and multi-omics technologies for exploring biomarkers for osteoporosis diagnosis is summarized. Moreover, we clarify the advantages and disadvantages of the application of osteoporosis biomarkers obtained using the omics method. Finally, we put forward valuable views on the future research direction of diagnostic biomarkers of osteoporosis. EXPERT OPINION Omics methods undoubtedly provide greatly contribute to the exploration of diagnostic biomarkers of osteoporosis; however, in the future, the clinical validity and clinical utility of the obtained potential biomarkers should be thoroughly examined. In addition, the improvement and optimization of the detection methods for different types of biomarkers and standardization of the detection process guarantee the reliability and accuracy of the detection results.
Collapse
Affiliation(s)
- Jing Yang
- Department of Clinical Laboratory Medicine, Beijing Jishuitan Hospital, Peking University, Beijing, China
| | - Jun Wu
- Department of Clinical Laboratory Medicine, Beijing Jishuitan Hospital, Peking University, Beijing, China
| |
Collapse
|
16
|
Tan Q, Møller AMJ, Qiu C, Madsen JS, Shen H, Bechmann T, Delaisse JM, Kristensen BW, Deng HW, Karasik D, Søe K. A variability in response of osteoclasts to zoledronic acid is mediated by smoking-associated modification in the DNA methylome. Clin Epigenetics 2023; 15:42. [PMID: 36915112 PMCID: PMC10012449 DOI: 10.1186/s13148-023-01449-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/15/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Clinical trials have shown zoledronic acid as a potent bisphosphonate in preventing bone loss, but with varying potency between patients. Human osteoclasts ex vivo reportedly displayed a variable sensitivity to zoledronic acid > 200-fold, determined by the half-maximal inhibitory concentration (IC50), with cigarette smoking as one of the reported contributors to this variation. To reveal the molecular basis of the smoking-mediated variation on treatment sensitivity, we performed a DNA methylome profiling on whole blood cells from 34 healthy female blood donors. Multiple regression models were fitted to associate DNA methylation with ex vivo determined IC50 values, smoking, and their interaction adjusting for age and cell compositions. RESULTS We identified 59 CpGs displaying genome-wide significance (p < 1e-08) with a false discovery rate (FDR) < 0.05 for the smoking-dependent association with IC50. Among them, 3 CpGs have p < 1e-08 and FDR < 2e-03. By comparing with genome-wide association studies, 15 significant CpGs were locally enriched (within < 50,000 bp) by SNPs associated with bone and body size measures. Furthermore, through a replication analysis using data from a published multi-omics association study on bone mineral density (BMD), we could validate that 29 out of the 59 CpGs were in close vicinity of genomic sites significantly associated with BMD. Gene Ontology (GO) analysis on genes linked to the 59 CpGs displaying smoking-dependent association with IC50, detected 18 significant GO terms including cation:cation antiporter activity, extracellular matrix conferring tensile strength, ligand-gated ion channel activity, etc. CONCLUSIONS: Our results suggest that smoking mediates individual sensitivity to zoledronic acid treatment through epigenetic regulation. Our novel findings could have important clinical implications since DNA methylation analysis may enable personalized zoledronic acid treatment.
Collapse
Affiliation(s)
- Qihua Tan
- grid.10825.3e0000 0001 0728 0170Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, 5000 Odense C, Denmark
| | - Anaïs Marie Julie Møller
- grid.10825.3e0000 0001 0728 0170Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, J. B. Winsløvs Vej 25, 1st Floor, 5000 Odense C, Denmark
- grid.10825.3e0000 0001 0728 0170Clinical Cell Biology, Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Chuan Qiu
- grid.265219.b0000 0001 2217 8588Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane Center of Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112 USA
| | - Jonna Skov Madsen
- grid.7143.10000 0004 0512 5013Department of Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- grid.10825.3e0000 0001 0728 0170Department of Regional Health Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Hui Shen
- grid.265219.b0000 0001 2217 8588Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane Center of Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112 USA
| | - Troels Bechmann
- grid.7143.10000 0004 0512 5013Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- grid.452681.c0000 0004 0639 1735Department of Oncology, Regional Hospital West Jutland, 7400 Herning, Denmark
| | - Jean-Marie Delaisse
- grid.10825.3e0000 0001 0728 0170Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, J. B. Winsløvs Vej 25, 1st Floor, 5000 Odense C, Denmark
- grid.7143.10000 0004 0512 5013Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
| | - Bjarne Winther Kristensen
- grid.7143.10000 0004 0512 5013Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- grid.10825.3e0000 0001 0728 0170Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Hong-Wen Deng
- grid.265219.b0000 0001 2217 8588Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane Center of Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112 USA
| | - David Karasik
- grid.22098.310000 0004 1937 0503Azrieli Faculty of Medicine, Bar-Ilan University, 130010 Safed, Israel
| | - Kent Søe
- grid.10825.3e0000 0001 0728 0170Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, J. B. Winsløvs Vej 25, 1st Floor, 5000 Odense C, Denmark
- grid.7143.10000 0004 0512 5013Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- grid.10825.3e0000 0001 0728 0170Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| |
Collapse
|
17
|
Mallik S, Sarkar A, Nath S, Maulik U, Das S, Pati SK, Ghosh S, Zhao Z. 3PNMF-MKL: A non-negative matrix factorization-based multiple kernel learning method for multi-modal data integration and its application to gene signature detection. Front Genet 2023; 14:1095330. [PMID: 36865387 PMCID: PMC9971618 DOI: 10.3389/fgene.2023.1095330] [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: 11/11/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
In this current era, biomedical big data handling is a challenging task. Interestingly, the integration of multi-modal data, followed by significant feature mining (gene signature detection), becomes a daunting task. Remembering this, here, we proposed a novel framework, namely, three-factor penalized, non-negative matrix factorization-based multiple kernel learning with soft margin hinge loss (3PNMF-MKL) for multi-modal data integration, followed by gene signature detection. In brief, limma, employing the empirical Bayes statistics, was initially applied to each individual molecular profile, and the statistically significant features were extracted, which was followed by the three-factor penalized non-negative matrix factorization method used for data/matrix fusion using the reduced feature sets. Multiple kernel learning models with soft margin hinge loss had been deployed to estimate average accuracy scores and the area under the curve (AUC). Gene modules had been identified by the consecutive analysis of average linkage clustering and dynamic tree cut. The best module containing the highest correlation was considered the potential gene signature. We utilized an acute myeloid leukemia cancer dataset from The Cancer Genome Atlas (TCGA) repository containing five molecular profiles. Our algorithm generated a 50-gene signature that achieved a high classification AUC score (viz., 0.827). We explored the functions of signature genes using pathway and Gene Ontology (GO) databases. Our method outperformed the state-of-the-art methods in terms of computing AUC. Furthermore, we included some comparative studies with other related methods to enhance the acceptability of our method. Finally, it can be notified that our algorithm can be applied to any multi-modal dataset for data integration, followed by gene module discovery.
Collapse
Affiliation(s)
- Saurav Mallik
- Department of Environmental Health, Harvard T H Chan School of public Health, Boston, MA, United States,*Correspondence: Saurav Mallik, , ; Zhongming Zhao,
| | - Anasua Sarkar
- Department of Computer Science & Engineering, Jadavpur University, Kolkata, India
| | - Sagnik Nath
- Department of Computer Science & Engineering, Jadavpur University, Kolkata, India
| | - Ujjwal Maulik
- Department of Computer Science & Engineering, Jadavpur University, Kolkata, India
| | - Supantha Das
- Department of Information Technology, Academy of Technology, Hooghly, West Bengal, India
| | - Soumen Kumar Pati
- Department of Bioinformatics, Maulana Abul Kalam Azad University, Kolkata, West Bengal, India
| | - Soumadip Ghosh
- Department of Computer Science & Engineering, Sister Nivedita University, New Town, West Bengal, India
| | - Zhongming Zhao
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States,Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States,*Correspondence: Saurav Mallik, , ; Zhongming Zhao,
| |
Collapse
|
18
|
Wang S, Greenbaum J, Qiu C, Gong Y, Wang Z, Lin X, Liu Y, He P, Meng X, Zhang Q, Shen H, Vemulapalli KC, Sanchez FL, Schiller MR, Xiao H, Deng H. Single-cell RNA sequencing reveals in vivo osteoimmunology interactions between the immune and skeletal systems. Front Endocrinol (Lausanne) 2023; 14:1107511. [PMID: 37051201 PMCID: PMC10083244 DOI: 10.3389/fendo.2023.1107511] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/10/2023] [Indexed: 03/28/2023] Open
Abstract
BACKGROUND While osteoimmunology interactions between the immune and skeletal systems are known to play an important role in osteoblast development, differentiation and bone metabolism related disease like osteoporosis, such interactions in either bone microenvironment or peripheral circulation in vivo at the single-cell resolution have not yet been characterized. METHODS We explored the osteoimmunology communications between immune cells and osteoblastic lineage cells (OBCs) by performing CellphoneDB and CellChat analyses with single-cell RNA sequencing (scRNA-seq) data from human femoral head. We also explored the osteoimmunology effects of immune cells in peripheral circulation on skeletal phenotypes. We used a scRNA-seq dataset of peripheral blood monocytes (PBMs) to perform deconvolution analysis. Then weighted gene co-expression network analysis (WGCNA) was used to identify monocyte subtype-specific subnetworks. We next used cell-specific network (CSN) and the least absolute shrinkage and selection operator (LASSO) to analyze the correlation of a gene subnetwork identified by WGCNA with bone mineral density (BMD). RESULTS We constructed immune cell and OBC communication networks and further identified L-R genes, such as JAG1 and NOTCH1/2, with ossification related functions. We also found a Mono4 related subnetwork that may relate to BMD variation in both older males and postmenopausal female subjects. CONCLUSIONS This is the first study to identify numerous ligand-receptor pairs that likely mediate signals between immune cells and osteoblastic lineage cells. This establishes a foundation to reveal advanced and in-depth osteoimmunology interactions to better understand the relationship between local bone microenvironment and immune cells in peripheral blood and the impact on bone phenotypes.
Collapse
Affiliation(s)
- Shengran Wang
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Jonathan Greenbaum
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Chuan Qiu
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Yun Gong
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Zun Wang
- Xiangya School of Nursing, Central South University, Changsha, China
| | - Xu Lin
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yong Liu
- Center for System Biology, Data Sciences and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
| | - Pei He
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xianghe Meng
- Center for System Biology, Data Sciences and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
| | - Qiang Zhang
- College of Public Health, Zhengzhou University, High-Tech Development Zone of States, Zhengzhou, China
| | - Hui Shen
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Krishna Chandra Vemulapalli
- Department of Orthopaedic Surgery, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Fernando L. Sanchez
- Department of Orthopaedic Surgery, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Martin R. Schiller
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
| | - Hongmei Xiao
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
- *Correspondence: Hongwen Deng, ; Hongmei Xiao,
| | - Hongwen Deng
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
- *Correspondence: Hongwen Deng, ; Hongmei Xiao,
| |
Collapse
|
19
|
Ri K, Lee-Okada HC, Yokomizo T. Omega-6 highly unsaturated fatty acids in Leydig cells facilitate male sex hormone production. Commun Biol 2022; 5:1001. [PMID: 36131086 PMCID: PMC9492697 DOI: 10.1038/s42003-022-03972-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Highly unsaturated fatty acids (HUFAs) are fatty acids with more than three double bonds in the molecule. Mammalian testes contain very high levels of omega-6 HUFAs compared with other tissues. However, the metabolic and biological significance of these HUFAs in the mammalian testis is poorly understood. Here we show that Leydig cells vigorously synthesize omega-6 HUFAs to facilitate male sex hormone production. In the testis, FADS2 (Fatty acid desaturase 2), the rate-limiting enzyme for HUFA biosynthesis, is highly expressed in Leydig cells. In this study, pharmacological and genetic inhibition of FADS2 drastically reduces the production of omega-6 HUFAs and male steroid hormones in Leydig cells; this reduction is significantly rescued by supplementation with omega-6 HUFAs. Mechanistically, hormone-sensitive lipase (HSL; also called LIPE), a lipase that supplies free cholesterol for steroid hormone production, preferentially hydrolyzes HUFA-containing cholesteryl esters as substrates. Taken together, our results demonstrate that Leydig cells highly express FADS2 to facilitate male steroid hormone production by accumulating omega-6 HUFA-containing cholesteryl esters, which serve as preferred substrates for HSL. These findings unveil a previously unrecognized importance of omega-6 HUFAs in the mammalian male reproductive system. Leydig cells highly express FADS2 to facilitate male steroid hormone production by accumulating omega-6 HUFA-containing cholesteryl esters, which serve as preferred substrates for hormone-sensitive lipase
Collapse
Affiliation(s)
- Keiken Ri
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hyeon-Cheol Lee-Okada
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Panahi N, Fahimfar N, Roshani S, Arjmand B, Gharibzadeh S, Shafiee G, Migliavacca E, Breuille D, Feige JN, Grzywinski Y, Corthesy J, Razi F, Heshmat R, Nabipour I, Farzadfar F, Soltani A, Larijani B, Ostovar A. Association of amino acid metabolites with osteoporosis, a metabolomic approach: Bushehr elderly health program. Metabolomics 2022; 18:63. [PMID: 35915271 DOI: 10.1007/s11306-022-01919-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION AND OBJECTIVES Amino acids are the most frequently reported metabolites associated with low bone mineral density (BMD) in metabolomics studies. We aimed to evaluate the association between amino acid metabolic profile and bone indices in the elderly population. METHODS 400 individuals were randomly selected from 2384 elderly men and women over 60 years participating in the second stage of the Bushehr elderly health (BEH) program, a population-based prospective cohort study that is being conducted in Bushehr, a southern province of Iran. Frozen plasma samples were used to measure 29 amino acid and derivatives metabolites using the UPLC-MS/MS-based targeted metabolomics platform. We conducted Elastic net regression analysis to detect the metabolites associated with BMD of different sites and lumbar spine trabecular bone score, and also to examine the ability of the measured metabolites to differentiate osteoporosis. RESULTS We adjusted the analysis for possible confounders (age, BMI, diabetes, smoking, physical activity, vitamin D level, and sex). Valine, leucine, isoleucine, and alanine in women and tryptophan in men were the most important amino acids inversely associated with osteoporosis (OR range from 0.77 to 0.89). Sarcosine, followed by tyrosine, asparagine, alpha aminobutyric acid, and ADMA in women and glutamine in men and when both women and men were considered together were the most discriminating amino acids detected in individuals with osteoporosis (OR range from 1.15 to 1.31). CONCLUSION We found several amino acid metabolites associated with possible bone status in elderly individuals. Further studies are required to evaluate the utility of these metabolites as clinical biomarkers for osteoporosis prediction and their effect on bone health as dietary supplements.
Collapse
Affiliation(s)
- Nekoo Panahi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Fahimfar
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahin Roshani
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Babak Arjmand
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Gita Shafiee
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Eugenia Migliavacca
- Nestlé Institute of Health Sciences, Nestlé Research, CH-1015, Lausanne, Switzerland
| | - Denis Breuille
- Nestlé Institute of Health Sciences, Nestlé Research, CH-1015, Lausanne, Switzerland
| | - Jerome N Feige
- Nestlé Institute of Health Sciences, Nestlé Research, CH-1015, Lausanne, Switzerland
| | - Yohan Grzywinski
- Institute of Food Safety and Analytical Science, Nestlé Research, CH-1015, Lausanne, Switzerland
| | - John Corthesy
- Institute of Food Safety and Analytical Science, Nestlé Research, CH-1015, Lausanne, Switzerland
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Farshad Farzadfar
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akbar Soltani
- Evidence-Based Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Ostovar
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Integration of Omics and Phenotypic Data for Precision Medicine. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2486:19-35. [PMID: 35437716 DOI: 10.1007/978-1-0716-2265-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Over the past two decades, biomedical research is moving toward a big-data-driven approach. The underlying causes of this transition include the ability to gather genetic or molecular profiles of humans faster, the increasing adoption of electronic health record (EHR) system, and the growing interest in linking omics and phenotypic data for analysis. The integration of individual's biology data (e.g., genomics, proteomics, metabolomics), and health-care data has created unprecedented opportunities for precision medicine, that is, a medical model that uses a patient's unique information, mainly genetic, to prevent, diagnose, or treat disease. This chapter reviewed the research opportunities and applications of integrating omics and phenotypic data for precision medicine, such as understanding the relationship between genotype and phenotype, disease subtyping, and diagnosis or prediction of adverse outcomes. We reviewed the recent advanced methods, particularly the machine learning and deep learning-based approaches used for harnessing and harmonizing the multiomics and phenotypic data to address these applications. We finally discussed the challenges and future directions.
Collapse
|
22
|
Zeng Z, Guo R, Wang Z, Yan H, Lv X, Zhao Q, Jiang X, Zhang C, Zhang D, Yang C, Li W, Zhang Z, Wang Q, Huang R, Li B, Hu X, Gao L. Circulating Monocytes Act as a Common Trigger for the Calcification Paradox of Osteoporosis and Carotid Atherosclerosis via TGFB1-SP1 and TNFSF10-NFKB1 Axis. Front Endocrinol (Lausanne) 2022; 13:944751. [PMID: 35937796 PMCID: PMC9354531 DOI: 10.3389/fendo.2022.944751] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Osteoporosis often occurs with carotid atherosclerosis and causes contradictory calcification across tissue in the same patient, which is called the "calcification paradox". Circulating monocytes may be responsible for this unbalanced ectopic calcification. Here, we aimed to show how CD14+ monocytes contribute to the pathophysiology of coexisting postmenopausal osteoporosis and carotid atherosclerosis. METHODS We comprehensively analyzed osteoporosis data from the mRNA array dataset GSE56814 and the scRNA-seq dataset GSM4423510. Carotid atherosclerosis data were obtained from the GSE23746 mRNA dataset and GSM4705591 scRNA-seq dataset. First, osteoblast and vascular SMC lineages were annotated based on their functional expression using gene set enrichment analysis and AUCell scoring. Next, pseudotime analysis was applied to draw their differentiated trajectory and identify the key gene expression changes in crossroads. Then, ligand-receptor interactions between CD14+ monocytes and osteoblast and vascular smooth muscle cell (SMC) lineages were annotated with iTALK. Finally, we selected calcification paradox-related expression in circulating monocytes with LASSO analysis. RESULTS First, we found a large proportion of delayed premature osteoblasts in osteoporosis and osteogenic SMCs in atherosclerosis. Second, CD14+ monocytes interacted with the intermediate cells of the premature osteoblast and osteogenic SMC lineage by delivering TGFB1 and TNFSF10. This interaction served as a trigger activating the transcription factors (TF) SP1 and NFKB1 to upregulate the inflammatory response and cell senescence and led to a retarded premature state in the osteoblast lineage and osteogenic transition in the SMC lineage. Then, 76.49% of common monocyte markers were upregulated in the circulating monocytes between the two diseases, which were related to chemotaxis and inflammatory responses. Finally, we identified 7 calcification paradox-related genes on circulating monocytes, which were upregulated in aging cells and downregulated in DNA repair cells, indicating that the aging monocytes contributed to the development of the two diseases. CONCLUSIONS Our work provides a perspective for understanding the triggering roles of CD14+ monocytes in the development of the calcification paradox in osteoporosis- and atherosclerosis-related cells based on combined scRNA and mRNA data. This study provided us with an elucidation of the mechanisms underlying the calcification paradox and could help in developing preventive and therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Bo Li
- *Correspondence: Liangbin Gao, ; Xumin Hu, ; Bo Li,
| | - Xumin Hu
- *Correspondence: Liangbin Gao, ; Xumin Hu, ; Bo Li,
| | - Liangbin Gao
- *Correspondence: Liangbin Gao, ; Xumin Hu, ; Bo Li,
| |
Collapse
|
23
|
Zhao Z, Cai Z, Chen A, Cai M, Yang K. Application of metabolomics in osteoporosis research. Front Endocrinol (Lausanne) 2022; 13:993253. [PMID: 36452325 PMCID: PMC9702081 DOI: 10.3389/fendo.2022.993253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022] Open
Abstract
Osteoporosis (OP) is a systemic disease characterized by bone metabolism imbalance and bone microstructure destruction, which causes serious social and economic burden. At present, the diagnosis and treatment of OP mainly rely on imaging combined with drugs. However, the existing pathogenic mechanisms, diagnosis and treatment strategies for OP are not clear and effective enough, and the disease progression that cannot reflect OP further restricts its effective treatment. The application of metabolomics has facilitated the study of OP, further exploring the mechanism and behavior of bone cells, prevention, and treatment of the disease from various metabolic perspectives, finally realizing the possibility of a holistic approach. In this review, we focus on the application of metabolomics in OP research, especially the newer systematic application of metabolomics and treatment with herbal medicine and their extracts. In addition, the prospects of clinical transformation in related fields are also discussed. The aim of this study is to highlight the use of metabolomics in OP research, especially in exploring the pathogenesis of OP and the therapeutic mechanisms of natural herbal medicine, for the benefit of interdisciplinary researchers including clinicians, biologists, and materials engineers.
Collapse
Affiliation(s)
- Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengwei Cai
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aopan Chen
- Department of Orthopaedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Ming Cai, ; Kai Yang,
| | - Kai Yang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Ming Cai, ; Kai Yang,
| |
Collapse
|
24
|
Panahi N, Arjmand B, Ostovar A, Kouhestani E, Heshmat R, Soltani A, Larijani B. Metabolomic biomarkers of low BMD: a systematic review. Osteoporos Int 2021; 32:2407-2431. [PMID: 34309694 DOI: 10.1007/s00198-021-06037-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022]
Abstract
Due to the metabolic nature of osteoporosis, this study was conducted to identify metabolomic studies investigating the metabolic profile of low bone mineral density (BMD) and osteoporosis. A comprehensive systematic literature search was conducted through PubMed, Web of Science, Scopus, and Embase databases up to April 08, 2020, to identify observational studies with cross-sectional or case-control designs investigating the metabolic profile of low BMD in adults using biofluid specimen via metabolomic platform. The quality assessment panel specified for the "omics"-based diagnostic research (QUADOMICS) tool was used to estimate the methodologic quality of the included studies. Ten untargeted and one targeted approach metabolomic studies investigating biomarkers in different biofluids through mass spectrometry or nuclear magnetic resonance platforms were included in the systematic review. Some metabolite panels, rather than individual metabolites, showed promising results in differentiating low BMD from normal. Candidate metabolites were of different categories including amino acids, followed by lipids and carbohydrates. Besides, certain pathways were suggested by some of the studies to be involved. This systematic review suggested that metabolic profiling could improve the diagnosis of low BMD. Despite valuable findings attained from each of these studies, there was great heterogeneity regarding the ethnicity and age of participants, samples, and the metabolomic platform. Further longitudinal studies are needed to validate the results and confirm the predictive role of metabolic profile on low BMD and fracture. It is also mandatory to address and minimize the heterogeneity in future studies by using reliable quantitative methods. Summary: Due to the metabolic nature of osteoporosis, researchers have considered metabolomic studies recently. This systematic review showed that metabolic profiling including different categories of metabolites could improve the diagnosis of low BMD. However, great heterogeneity was observed and it is mandatory to address and minimize the heterogeneity in future studies.
Collapse
Affiliation(s)
- N Panahi
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - B Arjmand
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - A Ostovar
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - E Kouhestani
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - R Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - A Soltani
- Evidence Based Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - B Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Abood A, Farber CR. Using "-omics" Data to Inform Genome-wide Association Studies (GWASs) in the Osteoporosis Field. Curr Osteoporos Rep 2021; 19:369-380. [PMID: 34125409 PMCID: PMC8767463 DOI: 10.1007/s11914-021-00684-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 01/12/2023]
Abstract
PURPOSE OF REVIEW Osteoporosis constitutes a major societal health problem. Genome-wide association studies (GWASs) have identified over 1100 loci influencing bone mineral density (BMD); however, few of the causal genes have been identified. Here, we review approaches that use "-omics" data and genetic- and systems genetics-based analytical strategies to facilitate causal gene discovery. RECENT FINDINGS The bone field is beginning to adopt approaches that are commonplace in other disease disciplines. The slower progress has been due in part to the lack of large-scale "omics" data on bone and bone cells. This is however changing, and approaches such as eQTL colocalization, transcriptome-wide association studies (TWASs), network, and integrative approaches are beginning to provide significant insight into the genes responsible for BMD GWAS associations. The use of "-omics" data to inform BMD GWASs has increased in recent years, leading to the identification of novel regulators of BMD in humans. The ultimate goal will be to use this information to develop more effective therapies to treat and ultimately prevent osteoporosis.
Collapse
Affiliation(s)
- Abdullah Abood
- Center for Public Health Genomics, University of Virginia, 800717, Charlottesville, VA, 22908, USA
- Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Charles R Farber
- Center for Public Health Genomics, University of Virginia, 800717, Charlottesville, VA, 22908, USA.
- Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA.
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
26
|
Alam MA, Qiu C, Shen H, Wang YP, Deng HW. A generalized kernel machine approach to identify higher-order composite effects in multi-view datasets, with application to adolescent brain development and osteoporosis. J Biomed Inform 2021; 120:103854. [PMID: 34237438 DOI: 10.1016/j.jbi.2021.103854] [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: 09/25/2020] [Revised: 05/28/2021] [Accepted: 06/28/2021] [Indexed: 10/20/2022]
Abstract
In recent years, a comprehensive study of complex disease with multi-view datasets (e.g., multi-omics and imaging scans) has been a focus and forefront in biomedical research. State-of-the-art biomedical technologies are enabling us to collect multi-view biomedical datasets for the study of complex diseases. While all the views of data tend to explore complementary information of disease, analysis of multi-view data with complex interactions is challenging for a deeper and holistic understanding of biological systems. In this paper, we propose a novel generalized kernel machine approach to identify higher-order composite effects in multi-view biomedical datasets (GKMAHCE). This generalized semi-parametric (a mixed-effect linear model) approach includes the marginal and joint Hadamard product of features from different views of data. The proposed kernel machine approach considers multi-view data as predictor variables to allow a more thorough and comprehensive modeling of a complex trait. We applied GKMAHCE approach to both synthesized datasets and real multi-view datasets from adolescent brain development and osteoporosis study. Our experiments demonstrate that the proposed method can effectively identify higher-order composite effects and suggest that corresponding features (genes, region of interests, and chemical taxonomies) function in a concerted effort. We show that the proposed method is more generalizable than existing ones. To promote reproducible research, the source code of the proposed method is available at.
Collapse
Affiliation(s)
- Md Ashad Alam
- Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, USA; Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112, USA.
| | - Chuan Qiu
- Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, USA; Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, USA; Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Yu-Ping Wang
- Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, USA; Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, USA; Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
27
|
Zhang L, Yang Y, Geng D, Wu Y. Identification of Potential Therapeutic Targets and Molecular Regulatory Mechanisms for Osteoporosis by Bioinformatics Methods. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8851421. [PMID: 33778083 PMCID: PMC7969088 DOI: 10.1155/2021/8851421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/06/2021] [Accepted: 02/08/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Osteoporosis is characterized by low bone mass, deterioration of bone tissue structure, and susceptibility to fracture. New and more suitable therapeutic targets need to be discovered. METHODS We collected osteoporosis-related datasets (GSE56815, GSE99624, and GSE63446). The methylation markers were obtained by differential analysis. Degree, DMNC, MCC, and MNC plug-ins were used to screen the important methylation markers in PPI network, then enrichment analysis was performed. ROC curve was used to evaluate the diagnostic effect of osteoporosis. In addition, we evaluated the difference in immune cell infiltration between osteoporotic patients and control by ssGSEA. Finally, differential miRNAs in osteoporosis were used to predict the regulators of key methylation markers. RESULTS A total of 2351 differentially expressed genes and 5246 differentially methylated positions were obtained between osteoporotic patients and controls. We identified 19 methylation markers by PPI network. They were mainly involved in biological functions and signaling pathways such as apoptosis and immune inflammation. HIST1H3G, MAP3K5, NOP2, OXA1L, and ZFPM2 with higher AUC values were considered key methylation markers. There were significant differences in immune cell infiltration between osteoporotic patients and controls, especially dendritic cells and natural killer cells. The correlation between MAP3K5 and immune cells was high, and its differential expression was also validated by other two datasets. In addition, NOP2 was predicted to be regulated by differentially expressed hsa-miR-3130-5p. CONCLUSION Our efforts aim to provide new methylation markers as therapeutic targets for osteoporosis to better treat osteoporosis in the future.
Collapse
Affiliation(s)
- Li Zhang
- Department of Geriatrics, The Municipal Hospital of Suzhou, Jiangsu, China
| | - Yunlong Yang
- Department of Geriatrics, The Municipal Hospital of Suzhou, Jiangsu, China
| | - Dechun Geng
- Department of orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Yonghua Wu
- Department of Geriatrics, The Municipal Hospital of Suzhou, Jiangsu, China
| |
Collapse
|
28
|
Wörheide MA, Krumsiek J, Kastenmüller G, Arnold M. Multi-omics integration in biomedical research - A metabolomics-centric review. Anal Chim Acta 2021; 1141:144-162. [PMID: 33248648 PMCID: PMC7701361 DOI: 10.1016/j.aca.2020.10.038] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Recent advances in high-throughput technologies have enabled the profiling of multiple layers of a biological system, including DNA sequence data (genomics), RNA expression levels (transcriptomics), and metabolite levels (metabolomics). This has led to the generation of vast amounts of biological data that can be integrated in so-called multi-omics studies to examine the complex molecular underpinnings of health and disease. Integrative analysis of such datasets is not straightforward and is particularly complicated by the high dimensionality and heterogeneity of the data and by the lack of universal analysis protocols. Previous reviews have discussed various strategies to address the challenges of data integration, elaborating on specific aspects, such as network inference or feature selection techniques. Thereby, the main focus has been on the integration of two omics layers in their relation to a phenotype of interest. In this review we provide an overview over a typical multi-omics workflow, focusing on integration methods that have the potential to combine metabolomics data with two or more omics. We discuss multiple integration concepts including data-driven, knowledge-based, simultaneous and step-wise approaches. We highlight the application of these methods in recent multi-omics studies, including large-scale integration efforts aiming at a global depiction of the complex relationships within and between different biological layers without focusing on a particular phenotype.
Collapse
Affiliation(s)
- Maria A Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jan Krumsiek
- Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
| |
Collapse
|
29
|
Wu ZL, Chen SY, Hu S, Jia X, Wang J, Lai SJ. Metabolomic and Proteomic Profiles Associated With Ketosis in Dairy Cows. Front Genet 2020; 11:551587. [PMID: 33391334 PMCID: PMC7772412 DOI: 10.3389/fgene.2020.551587] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 11/11/2020] [Indexed: 12/26/2022] Open
Abstract
Ketosis is a common metabolic disease in dairy cows during early lactation. However, information about the metabolomic and proteomic profiles associated with the incidence and progression of ketosis is still limited. In this study, an integrated metabolomics and proteomics approach was performed on blood serum sampled from cows diagnosed with clinical ketosis (case, ≥ 2.60 mmol/L plasma β-hydroxybutyrate; BHBA) and healthy controls (control, < 1.0 mmol/L BHBA). Samples were taken 2 weeks before parturition and 2 weeks after parturition from 19 animals (nine cases, 10 controls). All serum samples (n = 38) were subjected to Liquid Chromatography-Mass Spectrometry (LC-MS) based metabolomic analysis, and 20 samples underwent Data-Independent Acquisition (DIA) LC-MS based proteomic analysis. A total of 97 metabolites and 540 proteins were successfully identified, and multivariate analysis revealed significant differences in both metabolomic and proteomic profiles between cases and controls. We investigated clinical ketosis-associated metabolomic and proteomic changes using statistical analyses. Correlation analysis of statistically significant metabolites and proteins showed 78 strong correlations (correlation coefficient, R ≥ 0.7) between 38 metabolites and 25 proteins, which were then mapped to pathways using IMPaLA. Results showed that ketosis altered a wide range of metabolic pathways, such as metabolism, metabolism of proteins, gene expression and post-translational protein modification, vitamin metabolism, signaling, and disease related pathways. Findings presented here are relevant for identifying molecular targets for ketosis and biomarkers for ketosis detection during the transition period.
Collapse
Affiliation(s)
| | | | | | | | | | - Song-Jia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
30
|
Yu F, Xu C, Deng HW, Shen H. A novel computational strategy for DNA methylation imputation using mixture regression model (MRM). BMC Bioinformatics 2020; 21:552. [PMID: 33261550 PMCID: PMC7708217 DOI: 10.1186/s12859-020-03865-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND DNA methylation is an important heritable epigenetic mark that plays a crucial role in transcriptional regulation and the pathogenesis of various human disorders. The commonly used DNA methylation measurement approaches, e.g., Illumina Infinium HumanMethylation-27 and -450 BeadChip arrays (27 K and 450 K arrays) and reduced representation bisulfite sequencing (RRBS), only cover a small proportion of the total CpG sites in the human genome, which considerably limited the scope of the DNA methylation analysis in those studies. RESULTS We proposed a new computational strategy to impute the methylation value at the unmeasured CpG sites using the mixture of regression model (MRM) of radial basis functions, integrating information of neighboring CpGs and the similarities in local methylation patterns across subjects and across multiple genomic regions. Our method achieved a better imputation accuracy over a set of competing methods on both simulated and empirical data, particularly when the missing rate is high. By applying MRM to an RRBS dataset from subjects with low versus high bone mineral density (BMD), we recovered methylation values of ~ 300 K CpGs in the promoter regions of chromosome 17 and identified some novel differentially methylated CpGs that are significantly associated with BMD. CONCLUSIONS Our method is well applicable to the numerous methylation studies. By expanding the coverage of the methylation dataset to unmeasured sites, it can significantly enhance the discovery of novel differential methylation signals and thus reveal the mechanisms underlying various human disorders/traits.
Collapse
Affiliation(s)
- Fangtang Yu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Chao Xu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Hong-Wen Deng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Hui Shen
- Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
31
|
Randhawa V, Pathania S. Advancing from protein interactomes and gene co-expression networks towards multi-omics-based composite networks: approaches for predicting and extracting biological knowledge. Brief Funct Genomics 2020; 19:364-376. [PMID: 32678894 DOI: 10.1093/bfgp/elaa015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/31/2020] [Accepted: 06/15/2020] [Indexed: 01/17/2023] Open
Abstract
Prediction of biological interaction networks from single-omics data has been extensively implemented to understand various aspects of biological systems. However, more recently, there is a growing interest in integrating multi-omics datasets for the prediction of interactomes that provide a global view of biological systems with higher descriptive capability, as compared to single omics. In this review, we have discussed various computational approaches implemented to infer and analyze two of the most important and well studied interactomes: protein-protein interaction networks and gene co-expression networks. We have explicitly focused on recent methods and pipelines implemented to infer and extract biologically important information from these interactomes, starting from utilizing single-omics data and then progressing towards multi-omics data. Accordingly, recent examples and case studies are also briefly discussed. Overall, this review will provide a proper understanding of the latest developments in protein and gene network modelling and will also help in extracting practical knowledge from them.
Collapse
Affiliation(s)
- Vinay Randhawa
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Shivalika Pathania
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
32
|
Oh S, Jo Y, Jung S, Yoon S, Yoo KH. From genome sequencing to the discovery of potential biomarkers in liver disease. BMB Rep 2020. [PMID: 32475383 PMCID: PMC7330805 DOI: 10.5483/bmbrep.2020.53.6.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chronic liver disease progresses through several stages, fatty liver, steatohepatitis, cirrhosis, and eventually, it leads to hepatocellular carcinoma (HCC) over a long period of time. Since a large proportion of patients with HCC are accompanied by cirrhosis, it is considered to be an important factor in the diagnosis of liver cancer. This is because cirrhosis leads to an irreversible harmful effect, but the early stages of chronic liver disease could be reversed to a healthy state. Therefore, the discovery of biomarkers that could identify the early stages of chronic liver disease is important to prevent serious liver damage. Biomarker discovery at liver cancer and cirrhosis has enhanced the development of sequencing technology. Next generation sequencing (NGS) is one of the representative technical innovations in the biological field in the recent decades and it is the most important thing to design for research on what type of sequencing methods are suitable and how to handle the analysis steps for data integration. In this review, we comprehensively summarized NGS techniques for identifying genome, transcriptome, DNA methylome and 3D/4D chromatin structure, and introduced framework of processing data set and integrating multi-omics data for uncovering biomarkers.
Collapse
Affiliation(s)
- Sumin Oh
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Yeeun Jo
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Sungju Jung
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Sumin Yoon
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Kyung Hyun Yoo
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| |
Collapse
|
33
|
Chierici M, Bussola N, Marcolini A, Francescatto M, Zandonà A, Trastulla L, Agostinelli C, Jurman G, Furlanello C. Integrative Network Fusion: A Multi-Omics Approach in Molecular Profiling. Front Oncol 2020; 10:1065. [PMID: 32714870 PMCID: PMC7340129 DOI: 10.3389/fonc.2020.01065] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Recent technological advances and international efforts, such as The Cancer Genome Atlas (TCGA), have made available several pan-cancer datasets encompassing multiple omics layers with detailed clinical information in large collection of samples. The need has thus arisen for the development of computational methods aimed at improving cancer subtyping and biomarker identification from multi-modal data. Here we apply the Integrative Network Fusion (INF) pipeline, which combines multiple omics layers exploiting Similarity Network Fusion (SNF) within a machine learning predictive framework. INF includes a feature ranking scheme (rSNF) on SNF-integrated features, used by a classifier over juxtaposed multi-omics features (juXT). In particular, we show instances of INF implementing Random Forest (RF) and linear Support Vector Machine (LSVM) as the classifier, and two baseline RF and LSVM models are also trained on juXT. A compact RF model, called rSNFi, trained on the intersection of top-ranked biomarkers from the two approaches juXT and rSNF is finally derived. All the classifiers are run in a 10x5-fold cross-validation schema to warrant reproducibility, following the guidelines for an unbiased Data Analysis Plan by the US FDA-led initiatives MAQC/SEQC. INF is demonstrated on four classification tasks on three multi-modal TCGA oncogenomics datasets. Gene expression, protein expression and copy number variants are used to predict estrogen receptor status (BRCA-ER, N = 381) and breast invasive carcinoma subtypes (BRCA-subtypes, N = 305), while gene expression, miRNA expression and methylation data is used as predictor layers for acute myeloid leukemia and renal clear cell carcinoma survival (AML-OS, N = 157; KIRC-OS, N = 181). In test, INF achieved similar Matthews Correlation Coefficient (MCC) values and 97% to 83% smaller feature sizes (FS), compared with juXT for BRCA-ER (MCC: 0.83 vs. 0.80; FS: 56 vs. 1801) and BRCA-subtypes (0.84 vs. 0.80; 302 vs. 1801), improving KIRC-OS performance (0.38 vs. 0.31; 111 vs. 2319). INF predictions are generally more accurate in test than one-dimensional omics models, with smaller signatures too, where transcriptomics consistently play the leading role. Overall, the INF framework effectively integrates multiple data levels in oncogenomics classification tasks, improving over the performance of single layers alone and naive juxtaposition, and provides compact signature sizes.
Collapse
Affiliation(s)
| | - Nicole Bussola
- Fondazione Bruno Kessler, Trento, Italy
- University of Trento, Trento, Italy
| | | | - Margherita Francescatto
- Fondazione Bruno Kessler, Trento, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | | | | | | | | | | |
Collapse
|