51
|
Sameni M, Mirmotalebisohi SA, Dehghan Z, Abooshahab R, Khazaei-Poul Y, Mozafar M, Zali H. Deciphering molecular mechanisms of SARS-CoV-2 pathogenesis and drug repurposing through GRN motifs: a comprehensive systems biology study. 3 Biotech 2023; 13:117. [PMID: 37070032 PMCID: PMC10090260 DOI: 10.1007/s13205-023-03518-x] [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: 09/13/2022] [Accepted: 02/13/2023] [Indexed: 03/28/2023] Open
Abstract
The world has recently been plagued by a new coronavirus infection called SARS-CoV-2. This virus may lead to severe acute respiratory syndrome followed by multiple organ failure. SARS-CoV-2 has approximately 80-90% genetic similarity to SARS-CoV. Given the limited omics data available for host response to the viruses (more limited data for SARS-CoV-2), we attempted to unveil the crucial molecular mechanisms underlying the SARS-CoV-2 pathogenesis by comparing its regulatory network motifs with SARS-CoV. We also attempted to identify the non-shared crucial molecules and their functions to predict the specific mechanisms for each infection and the processes responsible for their different manifestations. Deciphering the crucial shared and non-shared mechanisms at the molecular level and signaling pathways underlying both diseases may help shed light on their pathogenesis and pave the way for other new drug repurposing against COVID-19. We constructed the GRNs for host response to SARS-CoV and SARS-CoV-2 pathogens (in vitro) and identified the significant 3-node regulatory motifs by analyzing them topologically and functionally. We attempted to identify the shared and non-shared regulatory elements and signaling pathways between their host responses. Interestingly, our findings indicated that NFKB1, JUN, STAT1, FOS, KLF4, and EGR1 were the critical shared TFs between motif-related subnetworks in both SARS and COVID-1, which are considered genes with specific functions in the immune response. Enrichment analysis revealed that the NOD-like receptor signaling, TNF signaling, and influenza A pathway were among the first significant pathways shared between SARS and COVID-19 up-regulated DEGs networks, and the term "metabolic pathways" (hsa01100) among the down-regulated DEGs networks. WEE1, PMAIP1, and TSC22D2 were identified as the top three hubs specific to SARS. However, MYPN, SPRY4, and APOL6 were the tops specific to COVID-19 in vitro. The term "Complement and coagulation cascades" pathway was identified as the first top non-shared pathway for COVID-19 and the MAPK signaling pathway for SARS. We used the identified crucial DEGs to construct a drug-gene interaction network to propose some drug candidates. Zinc chloride, Fostamatinib, Copper, Tirofiban, Tretinoin, and Levocarnitine were the six drugs with higher scores in our drug-gene network analysis. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03518-x.
Collapse
Affiliation(s)
- Marzieh Sameni
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Mirmotalebisohi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Yalda Khazaei-Poul
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mozafar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
52
|
Danzi F, Pacchiana R, Mafficini A, Scupoli MT, Scarpa A, Donadelli M, Fiore A. To metabolomics and beyond: a technological portfolio to investigate cancer metabolism. Signal Transduct Target Ther 2023; 8:137. [PMID: 36949046 PMCID: PMC10033890 DOI: 10.1038/s41392-023-01380-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 03/24/2023] Open
Abstract
Tumour cells have exquisite flexibility in reprogramming their metabolism in order to support tumour initiation, progression, metastasis and resistance to therapies. These reprogrammed activities include a complete rewiring of the bioenergetic, biosynthetic and redox status to sustain the increased energetic demand of the cells. Over the last decades, the cancer metabolism field has seen an explosion of new biochemical technologies giving more tools than ever before to navigate this complexity. Within a cell or a tissue, the metabolites constitute the direct signature of the molecular phenotype and thus their profiling has concrete clinical applications in oncology. Metabolomics and fluxomics, are key technological approaches that mainly revolutionized the field enabling researchers to have both a qualitative and mechanistic model of the biochemical activities in cancer. Furthermore, the upgrade from bulk to single-cell analysis technologies provided unprecedented opportunity to investigate cancer biology at cellular resolution allowing an in depth quantitative analysis of complex and heterogenous diseases. More recently, the advent of functional genomic screening allowed the identification of molecular pathways, cellular processes, biomarkers and novel therapeutic targets that in concert with other technologies allow patient stratification and identification of new treatment regimens. This review is intended to be a guide for researchers to cancer metabolism, highlighting current and emerging technologies, emphasizing advantages, disadvantages and applications with the potential of leading the development of innovative anti-cancer therapies.
Collapse
Affiliation(s)
- Federica Danzi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Maria T Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Biology and Genetics Section, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy.
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
53
|
Ouyang Y, Lu W, Wang Y, Wang B, Li F, Li X, Bai Y, Wang Y. Integrated analysis of mRNA and extrachromosomal circular DNA profiles to identify the potential mRNA biomarkers in breast cancer. Gene 2023; 857:147174. [PMID: 36627094 DOI: 10.1016/j.gene.2023.147174] [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: 09/29/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 01/08/2023]
Abstract
Extrachromosomal circular DNAs (eccDNAs) have been proved an inseparable relationship with cancer, based on the biological mechanisms of its biogenesis and impact on tumorigenesis, but still lacked of methods to analyze its function on the pathogenesis and progression of breast cancer (BC). The mRNA and eccDNA from BC cell samples (MDA-MB-453 and MCF-12A) were extracted with the removal of rRNA and linear DNA, respectively. High-throughput sequencing and bioinformatics analysis were performed to explore their expression level and molecular characterization of eccDNA. A total number of 161,062 eccDNA ranging from 33 bp to 54229 bp were detected with a median size of 1143 bp, distributed on all chromosomes and enriched on chromosome 20 the most. EccDNAs located in exons, upstream and downstream 2 kb regions were significantly increased compared with background. Analysis of eccDNA-related differentially expressed genes (eccDEGs) showed that FAT2 properly separated the two cells. CTNNB1, CACNA2D2 and CACNA1D were the hub genes with higher degrees in critical modules. All these four genes were significantly differentially expressed between breast invasive carcinoma (BRCA) tissues and normal ones. FAT2 and CTNNB1 correlated with significantly different overall survival (OS) when differentially expressed. The four genes showed a strong correlation with each other significantly and changed between tumor and normal samples. The results showed the potential of FAT2, CTNNB1, CACNA2D2 and CACNA1D as biomarkers with analysis of both DEGs and eccDEGs, which might assist in clinical medical treatment.
Collapse
Affiliation(s)
- Yunfei Ouyang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Wenxiang Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Ying Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Bangting Wang
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210096, PR China
| | - Fuyu Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Xiaohan Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Yunfei Bai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China.
| | - Yan Wang
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210096, PR China.
| |
Collapse
|
54
|
Zhu H, Li Y, Guo J, Feng S, Ge H, Gu C, Wang M, Nie R, Li N, Wang Y, Wang H, Zhong J, Qian X, He G. Integrated proteomic and phosphoproteomic analysis for characterization of colorectal cancer. J Proteomics 2023; 274:104808. [PMID: 36596410 DOI: 10.1016/j.jprot.2022.104808] [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: 08/03/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 01/02/2023]
Abstract
Proteins and translationally modified proteins like phosphoproteins have essential regulatory roles in tumorigenesis. This study attempts to elucidate the dysregulated proteins driving colorectal cancer (CRC). To explore the differential proteins, we performed iTRAQ labeling proteomics and TMT labeling phosphoproteomics analysis of CRC tissues and adjacent non-cancerous tissues. The functions of quantified proteins were analyzed using Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and Subcellular localization analysis. Depending on the results, we identified 330 differential proteins and 82 phosphoproteins in CRC. GO and KEGG analyses demonstrated that protein changes were primarily associated with regulating biological and metabolic processes through binding to other molecules. Co-expression relationships between proteomic and phosphoproteomic analysis revealed that TMC5, SMC4, SLBP, VSIG2, and NDRG2 were significantly dysregulated differential proteins. Additionally, based on the predicted co-expression proteins, we identified that the stem-loop binding protein (SLBP) was up-regulated in CRC cells and promoted the proliferation and migration of CRC. This study reports an integrated proteomic and phosphoproteomic analysis of CRC to discern the functional impact of protein alterations and provides a candidate diagnostic biomarker or therapeutic target for CRC. SIGNIFICANCE: Combining one or more high-throughput omics technologies with bioinformatics to analyze biological samples and explore the links between biomolecules and their functions can provide more comprehensive and multi-level insights for disease mechanism research. Proteomics, phosphoproteomics, metabolomics and their combined analysis play an important role in the auxiliary diagnosis, the discovery of biomarkers and novel therapeutic targets for colorectal cancer. In this integrated proteomic and phosphoproteomic analysis, we identified proteins and phosphoproteins in colorectal cancer tissue and analyzed potential mechanisms contributing to progression in colorectal cancer. The results of this study provide a foundation to focus future experiments on the contribution of altered protein and phosphorylation patterns to prevention and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Huifang Zhu
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Yongzhen Li
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Jingyu Guo
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Shuang Feng
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Hong Ge
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Chuansha Gu
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Mengyao Wang
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Ruicong Nie
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Na Li
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Yongxia Wang
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Haijun Wang
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Jiateng Zhong
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China
| | - Xinlai Qian
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China.
| | - Guoyang He
- Department of Pathology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang City, Henan Province, China.
| |
Collapse
|
55
|
Srinath A, Xie B, Li Y, Sone JY, Romanos S, Chen C, Sharma A, Polster S, Dorrestein PC, Weldon KC, DeBiasse D, Moore T, Lightle R, Koskimäki J, Zhang D, Stadnik A, Piedad K, Hagan M, Shkoukani A, Carrión-Penagos J, Bi D, Shen L, Shenkar R, Ji Y, Sidebottom A, Pamer E, Gilbert JA, Kahn ML, D'Souza M, Sulakhe D, Awad IA, Girard R. Plasma metabolites with mechanistic and clinical links to the neurovascular disease cavernous angioma. COMMUNICATIONS MEDICINE 2023; 3:35. [PMID: 36869161 PMCID: PMC9984539 DOI: 10.1038/s43856-023-00265-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Cavernous angiomas (CAs) affect 0.5% of the population, predisposing to serious neurologic sequelae from brain bleeding. A leaky gut epithelium associated with a permissive gut microbiome, was identified in patients who develop CAs, favoring lipid polysaccharide producing bacterial species. Micro-ribonucleic acids along with plasma levels of proteins reflecting angiogenesis and inflammation were also previously correlated with CA and CA with symptomatic hemorrhage. METHODS The plasma metabolome of CA patients and CA patients with symptomatic hemorrhage was assessed using liquid-chromatography mass spectrometry. Differential metabolites were identified using partial least squares-discriminant analysis (p < 0.05, FDR corrected). Interactions between these metabolites and the previously established CA transcriptome, microbiome, and differential proteins were queried for mechanistic relevance. Differential metabolites in CA patients with symptomatic hemorrhage were then validated in an independent, propensity matched cohort. A machine learning-implemented, Bayesian approach was used to integrate proteins, micro-RNAs and metabolites to develop a diagnostic model for CA patients with symptomatic hemorrhage. RESULTS Here we identify plasma metabolites, including cholic acid and hypoxanthine distinguishing CA patients, while arachidonic and linoleic acids distinguish those with symptomatic hemorrhage. Plasma metabolites are linked to the permissive microbiome genes, and to previously implicated disease mechanisms. The metabolites distinguishing CA with symptomatic hemorrhage are validated in an independent propensity-matched cohort, and their integration, along with levels of circulating miRNAs, enhance the performance of plasma protein biomarkers (up to 85% sensitivity and 80% specificity). CONCLUSIONS Plasma metabolites reflect CAs and their hemorrhagic activity. A model of their multiomic integration is applicable to other pathologies.
Collapse
Affiliation(s)
- Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Bingqing Xie
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Ying Li
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, 150001, Harbin, Heilongjiang, China
| | - Je Yeong Sone
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Chang Chen
- Bioinformatics Core, Center for Research Informatics, The University of Chicago, Chicago, IL, 60637, USA
| | - Anukriti Sharma
- Department of Surgery, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Sean Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Pieter C Dorrestein
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Department of Pharmacology, The University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Kelly C Weldon
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Dorothy DeBiasse
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Janne Koskimäki
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Kristina Piedad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Matthew Hagan
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Abdallah Shkoukani
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Julián Carrión-Penagos
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Dehua Bi
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Le Shen
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Yuan Ji
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Ashley Sidebottom
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Eric Pamer
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Jack A Gilbert
- Department of Surgery, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Mark D'Souza
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Dinanath Sulakhe
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA.
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| |
Collapse
|
56
|
Metabolomics in Corneal Diseases: A Narrative Review from Clinical Aspects. Metabolites 2023; 13:metabo13030380. [PMID: 36984820 PMCID: PMC10055016 DOI: 10.3390/metabo13030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Corneal pathologies may have subtle manifestations in the initial stages, delaying diagnosis and timely treatment. This can lead to irreversible visual loss. Metabolomics is a rapidly developing field that allows the study of metabolites in a system, providing a complementary tool in the early diagnosis and management of corneal diseases. Early identification of biomarkers is key to prevent disease progression. The advancement of nuclear magnetic resonance and mass spectrometry allows the identification of new biomarkers in the analysis of tear, cornea, and aqueous humor. Novel perspectives on disease mechanisms are identified, which provide vital information for potential targeted therapies in the future. Current treatments are analyzed at a molecular level to offer further information regarding their efficacy. In this article, we provide a comprehensive review of the metabolomic studies undertaken in the cornea and various pathologies such as dry eye disease, Sjogren’s syndrome, keratoconus, post-refractive surgery, contact lens wearers, and diabetic corneas. Lastly, we discuss the exciting future that metabolomics plays in cornea research.
Collapse
|
57
|
Tang J, Xu Q, Tang K, Ye X, Cao Z, Zou M, Zeng J, Guan X, Han J, Wang Y, Yang L, Lin Y, Jiang K, Chen X, Zhao Y, Tian D, Li C, Shen W, Du X. Susceptibility identification for seasonal influenza A/H3N2 based on baseline blood transcriptome. Front Immunol 2023; 13:1048774. [PMID: 36713410 PMCID: PMC9878565 DOI: 10.3389/fimmu.2022.1048774] [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: 09/20/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Influenza susceptibility difference is a widely existing trait that has great practical significance for the accurate prevention and control of influenza. Methods Here, we focused on the human susceptibility to the seasonal influenza A/H3N2 of healthy adults at baseline level. Whole blood expression data for influenza A/H3N2 susceptibility from GEO were collected firstly (30 symptomatic and 19 asymptomatic). Then to explore the differences at baseline, a suite of systems biology approaches - the differential expression analysis, co-expression network analysis, and immune cell frequencies analysis were utilized. Results We found the baseline condition, especially immune condition between symptomatic and asymptomatic, was different. Co-expression module that is positively related to asymptomatic is also related to immune cell type of naïve B cell. Function enrichment analysis showed significantly correlation with "B cell receptor signaling pathway", "immune response-activating cell surface receptor signaling pathway" and so on. Also, modules that are positively related to symptomatic are also correlated to immune cell type of neutrophils, with function enrichment analysis showing significantly correlations with "response to bacterium", "inflammatory response", "cAMP-dependent protein kinase complex" and so on. Responses of symptomatic and asymptomatic hosts after virus exposure show differences on resisting the virus, with more effective frontline defense for asymptomatic hosts. A prediction model was also built based on only baseline transcription information to differentiate symptomatic and asymptomatic population with accuracy of 0.79. Discussion The results not only improve our understanding of the immune system and influenza susceptibility, but also provide a new direction for precise and targeted prevention and therapy of influenza.
Collapse
Affiliation(s)
- Jing Tang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Qiumei Xu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Kang Tang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Ye
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zicheng Cao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,School of Public Health, Shantou University, Shantou, China
| | - Min Zou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Jinfeng Zeng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Xinyan Guan
- Department of Chronic Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, China
| | - Jinglin Han
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Yihan Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Lan Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yishan Lin
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Kaiao Jiang
- Palos Verdes Peninsula High School, Rancho Palos Verdes, CA, United States
| | - Xiaoliang Chen
- Department of Chronic Disease Control and Prevention, Shenzhen Guangming District Center for Disease Control and Prevention, Shenzhen, China
| | - Yang Zhao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Dechao Tian
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Chunwei Li
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Shen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,*Correspondence: Xiangjun Du, ; Wei Shen,
| | - Xiangjun Du
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China,*Correspondence: Xiangjun Du, ; Wei Shen,
| |
Collapse
|
58
|
Punetha A, Kotiya D. Advancements in Oncoproteomics Technologies: Treading toward Translation into Clinical Practice. Proteomes 2023; 11:2. [PMID: 36648960 PMCID: PMC9844371 DOI: 10.3390/proteomes11010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Proteomics continues to forge significant strides in the discovery of essential biological processes, uncovering valuable information on the identity, global protein abundance, protein modifications, proteoform levels, and signal transduction pathways. Cancer is a complicated and heterogeneous disease, and the onset and progression involve multiple dysregulated proteoforms and their downstream signaling pathways. These are modulated by various factors such as molecular, genetic, tissue, cellular, ethnic/racial, socioeconomic status, environmental, and demographic differences that vary with time. The knowledge of cancer has improved the treatment and clinical management; however, the survival rates have not increased significantly, and cancer remains a major cause of mortality. Oncoproteomics studies help to develop and validate proteomics technologies for routine application in clinical laboratories for (1) diagnostic and prognostic categorization of cancer, (2) real-time monitoring of treatment, (3) assessing drug efficacy and toxicity, (4) therapeutic modulations based on the changes with prognosis and drug resistance, and (5) personalized medication. Investigation of tumor-specific proteomic profiles in conjunction with healthy controls provides crucial information in mechanistic studies on tumorigenesis, metastasis, and drug resistance. This review provides an overview of proteomics technologies that assist the discovery of novel drug targets, biomarkers for early detection, surveillance, prognosis, drug monitoring, and tailoring therapy to the cancer patient. The information gained from such technologies has drastically improved cancer research. We further provide exemplars from recent oncoproteomics applications in the discovery of biomarkers in various cancers, drug discovery, and clinical treatment. Overall, the future of oncoproteomics holds enormous potential for translating technologies from the bench to the bedside.
Collapse
Affiliation(s)
- Ankita Punetha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers University, 225 Warren St., Newark, NJ 07103, USA
| | - Deepak Kotiya
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 South Limestone St., Lexington, KY 40536, USA
| |
Collapse
|
59
|
Liu C, Duan Y, Zhou Q, Wang Y, Gao Y, Kan H, Hu J. A classification method of gastric cancer subtype based on residual graph convolution network. Front Genet 2023; 13:1090394. [PMID: 36685956 PMCID: PMC9845413 DOI: 10.3389/fgene.2022.1090394] [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: 11/05/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Clinical diagnosis and treatment of tumors are greatly complicated by their heterogeneity, and the subtype classification of cancer frequently plays a significant role in the subsequent treatment of tumors. Presently, the majority of studies rely far too heavily on gene expression data, omitting the enormous power of multi-omics fusion data and the potential for patient similarities. Method: In this study, we created a gastric cancer subtype classification model called RRGCN based on residual graph convolutional network (GCN) using multi-omics fusion data and patient similarity network. Given the multi-omics data's high dimensionality, we built an artificial neural network Autoencoder (AE) to reduce the dimensionality of the data and extract hidden layer features. The model is then built using the feature data. In addition, we computed the correlation between patients using the Pearson correlation coefficient, and this relationship between patients forms the edge of the graph structure. Four graph convolutional network layers and two residual networks with skip connections make up RRGCN, which reduces the amount of information lost during transmission between layers and prevents model degradation. Results: The results show that RRGCN significantly outperforms other classification methods with an accuracy as high as 0.87 when compared to four other traditional machine learning methods and deep learning models. Conclusion: In terms of subtype classification, RRGCN excels in all areas and has the potential to offer fresh perspectives on disease mechanisms and disease progression. It has the potential to be used for a broader range of disorders and to aid in clinical diagnosis.
Collapse
Affiliation(s)
- Can Liu
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui Computer Application Research Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Hefei, Anhui, China
| | - Yuchen Duan
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Qingqing Zhou
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yongkang Wang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui Computer Application Research Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Hefei, Anhui, China
| | - Yong Gao
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui Computer Application Research Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Hefei, Anhui, China
| | - Hongxing Kan
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui Computer Application Research Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Hefei, Anhui, China
| | - Jili Hu
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Anhui Computer Application Research Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Hefei, Anhui, China
| |
Collapse
|
60
|
Chalikiopoulou C, Gómez-Tamayo JC, Katsila T. Untargeted Metabolomics for Disease-Specific Signatures. Methods Mol Biol 2023; 2571:71-81. [PMID: 36152151 DOI: 10.1007/978-1-0716-2699-3_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Human diseases account for complex traits that usually exhibit markedly diverse clinical manifestations coming from a series of pathogenic processes that shape heterogeneous phenotypes. Considering that correlation does not imply causation as well as population differences and/or inter-individual variability, disease-specific signatures are becoming critical for biomarker discovery. Untargeted metabolomics is deemed to be a powerful approach to delineate molecular pathways of prime interest. Metabotypes capture the interplay of genomics and environmental influences per se. Untargeted metabolomics share the charm of being not only hypothesis-driven but also hypothesis-generating. Notwithstanding, the applicability of untargeted metabolomics toward clinically relevant outcomes depend on wet- and dry-lab procedures in the context of elegant study designs with clear rationale. As ideal may be far from feasible, herein we provide recommendations to combat sample mishandling that adversely affect data outcomes and if so, deal with imbalanced datasets toward data integrity.
Collapse
Affiliation(s)
| | | | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece.
| |
Collapse
|
61
|
Hassan T, Firdous P, Nissar K, Ahmad MB, Imtiyaz Z. Role of proteomics in surgical oncology. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
62
|
Qu X, Ma J, Gao H, Zhang Y, Zhai J, Gong J, Song Y, Hu T. Integration of metabolomics and proteomics analysis to explore the mechanism of neurotoxicity induced by receipt of isoniazid and rifampicin in mice. Neurotoxicology 2023; 94:24-34. [PMID: 36347327 DOI: 10.1016/j.neuro.2022.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/06/2022]
Abstract
Isoniazid (INH) and rifampicin (RIF) are co-administered in tuberculosis treatment but can cause neurotoxicity, and the mechanism is not known. To explore this mechanism, we employed an integrated approach using metabolomics analysis (MA) and proteomics analysis (PA). Male mice were divided into three groups and administered vehicle (control group), or co-administered INH (120 mg/kg) and RIF (240 mg/kg), for 7 or 14 days. Mice brains were collected for mass spectrometry-based PA and MA plus lipidomics analysis. Measurement of brain levels of malondialdehyde and superoxide dismutase revealed time-dependent brain injury after exposure to INH+RIF for 7 and 14 days. Also, 422 proteins, 35 metabolites, and 21 lipids were dysregulated and identified. MA demonstrated "purine metabolism," "phenylalanine, tyrosine and tryptophan biosynthesis," "biosynthesis of unsaturated fatty acids," "phenylalanine metabolism," and "arginine biosynthesis" to be disturbed significantly. PA demonstrated pathways such as "lipids," "amino acids," and "energy metabolism" to be disrupted. Peroxisome proliferator-activated receptor (PPAR) pathways were changed in energy metabolism, which led to the neurotoxicity induced by INH+RIF. Immunohistochemical analyses of PPARs in mice brains verified that PPAR-α and -γ expression was downregulated. PPAR-α and -γ activation might be a key target for alleviating INH+RIF-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiaoyu Qu
- Department of Pharmacy, The First Hospital of Jilin University, 130021 Changchun, China
| | - Jie Ma
- Department of Pharmacy, The First Hospital of Jilin University, 130021 Changchun, China
| | - Huan Gao
- Department of Pharmacy, The First Hospital of Jilin University, 130021 Changchun, China
| | - Yueming Zhang
- Department of Pharmacy, The First Hospital of Jilin University, 130021 Changchun, China
| | - Jinghui Zhai
- Department of Pharmacy, The First Hospital of Jilin University, 130021 Changchun, China
| | - Jiawei Gong
- Department of Pharmacy, The First Hospital of Jilin University, 130021 Changchun, China
| | - Yanqing Song
- Department of Pharmacy, The First Hospital of Jilin University, 130021 Changchun, China.
| | - Tingting Hu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130021 Changchun, China.
| |
Collapse
|
63
|
He DN, Wang N, Wen XL, Li XH, Guo Y, Fu SH, Xiong FF, Wu ZY, Zhu X, Gao XL, Wang ZZ, Wang HJ. Multi-omics analysis reveals a molecular landscape of the early recurrence and early metastasis in pan-cancer. Front Genet 2023; 14:1061364. [PMID: 37152984 PMCID: PMC10157260 DOI: 10.3389/fgene.2023.1061364] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Cancer remains a formidable challenge in medicine due to its propensity for recurrence and metastasis, which can result in unfavorable treatment outcomes. This challenge is particularly acute for early-stage patients, who may experience recurrence and metastasis without timely detection. Here, we first analyzed the differences in clinical characteristics among the primary tumor, recurrent tumor, and metastatic tumor in different stages of cancer, which may be caused by the molecular level. Moreover, the importance of predicting early cancer recurrence and metastasis is emphasized by survival analyses. Next, we used a multi-omics approach to identify key molecular changes associated with early cancer recurrence and metastasis and discovered that early metastasis in cancer demonstrated a high degree of genomic and cellular heterogeneity. We performed statistical comparisons for each level of omics data including gene expression, mutation, copy number variation, immune cell infiltration, and cell status. Then, various analytical techniques, such as proportional hazard model and Fisher's exact test, were used to identify specific genes or immune characteristics associated with early cancer recurrence and metastasis. For example, we observed that the overexpression of BPIFB1 and high initial B-cell infiltration levels are linked to early cancer recurrence, while the overexpression or amplification of ANKRD22 and LIPM, mutation of IGHA1 and MUC16, high fibroblast infiltration level, M1 polarization of macrophages, cellular status of DNA repair are all linked to early cancer metastasis. These findings have led us to construct classifiers, and the average area under the curve (AUC) of these classifiers was greater than 0.75 in The Cancer Genome Atlas (TCGA) cancer patients, confirming that the features we identified could be biomarkers for predicting recurrence and metastasis of early cancer. Finally, we identified specific early sensitive targets for targeted therapy and immune checkpoint inhibitor therapy. Once the biomarkers we identified changed, treatment-sensitive targets can be treated accordingly. Our study has comprehensively characterized the multi-omics characteristics and identified a panel of biomarkers of early cancer recurrence and metastasis. Overall, it provides a valuable resource for cancer recurrence and metastasis research and improves our understanding of the underlying mechanisms driving early cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Dan-ni He
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Na Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Xiao-Ling Wen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Xu-Hua Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Yu Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Shu-heng Fu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Fei-fan Xiong
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Zhe-yu Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xu Zhu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Xiao-ling Gao
- The Medical Laboratory Center, Hainan General Hospital, Haikou, China
- *Correspondence: Hong-jiu Wang, ; Zhen-zhen Wang, ; Xiao-ling Gao,
| | - Zhen-zhen Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- *Correspondence: Hong-jiu Wang, ; Zhen-zhen Wang, ; Xiao-ling Gao,
| | - Hong-jiu Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- *Correspondence: Hong-jiu Wang, ; Zhen-zhen Wang, ; Xiao-ling Gao,
| |
Collapse
|
64
|
Virzì GM, Mattiotti M, de Cal M, Ronco C, Zanella M, De Rosa S. Endotoxin in Sepsis: Methods for LPS Detection and the Use of Omics Techniques. Diagnostics (Basel) 2022; 13:diagnostics13010079. [PMID: 36611371 PMCID: PMC9818564 DOI: 10.3390/diagnostics13010079] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Lipopolysaccharide (LPS) or endotoxin, the major cell wall component of Gram-negative bacteria, plays a pivotal role in the pathogenesis of sepsis. It is able to activate the host defense system through interaction with Toll-like receptor 4, thus triggering pro-inflammatory mechanisms. A large amount of LPS induces inappropriate activation of the immune system, triggering an exaggerated inflammatory response and consequent extensive organ injury, providing the basis of sepsis damage. In this review, we will briefly describe endotoxin's molecular structure and its main pathogenetic action during sepsis. In addition, we will summarize the main different available methods for endotoxin detection with a special focus on the wider spectrum offered by omics technologies (genomics, transcriptomics, proteomics, and metabolomics) and promising applications of these in the identification of specific biomarkers for sepsis.
Collapse
Affiliation(s)
- Grazia Maria Virzì
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, 36100 Vicenza, Italy
- IRRIV—International Renal Research Institute Vicenza, 36100 Vicenza, Italy
- Correspondence: ; Tel.: +39-0444753650; Fax: +39-0444753949
| | - Maria Mattiotti
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, 36100 Vicenza, Italy
- IRRIV—International Renal Research Institute Vicenza, 36100 Vicenza, Italy
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS—Azienda Ospedaliero-Universitaria di Bologna, Department of Experimental Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Massimo de Cal
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, 36100 Vicenza, Italy
- IRRIV—International Renal Research Institute Vicenza, 36100 Vicenza, Italy
| | - Claudio Ronco
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, 36100 Vicenza, Italy
- IRRIV—International Renal Research Institute Vicenza, 36100 Vicenza, Italy
| | - Monica Zanella
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, 36100 Vicenza, Italy
- IRRIV—International Renal Research Institute Vicenza, 36100 Vicenza, Italy
| | - Silvia De Rosa
- IRRIV—International Renal Research Institute Vicenza, 36100 Vicenza, Italy
- Centre for Medical Sciences—CISMed, University of Trento, Via S. Maria Maddalena 1, 38122 Trento, Italy
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS Trento, 38122 Trento, Italy
| |
Collapse
|
65
|
Bonnechère B. Integrating Rehabilomics into the Multi-Omics Approach in the Management of Multiple Sclerosis: The Way for Precision Medicine? Genes (Basel) 2022; 14:63. [PMID: 36672802 PMCID: PMC9858788 DOI: 10.3390/genes14010063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Over recent years, significant improvements have been made in the understanding of (epi)genetics and neuropathophysiological mechanisms driving the different forms of multiple sclerosis (MS). For example, the role and importance of the bidirectional communications between the brain and the gut-also referred to as the gut-brain axis-in the pathogenesis of MS is receiving increasing interest in recent years and is probably one of the most promising areas of research for the management of people with MS. However, despite these important advances, it must be noted that these data are not-yet-used in rehabilitation. Neurorehabilitation is a cornerstone of MS patient management, and there are many techniques available to clinicians and patients, including technology-supported rehabilitation. In this paper, we will discuss how new findings on the gut microbiome could help us to better understand how rehabilitation can improve motor and cognitive functions. We will also see how the data gathered during the rehabilitation can help to get a better diagnosis of the patients. Finally, we will discuss how these new techniques can better guide rehabilitation to lead to precision rehabilitation and ultimately increase the quality of patient care.
Collapse
Affiliation(s)
- Bruno Bonnechère
- REVAL Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, 3590 Diepenbeek, Belgium;
- Technology-Supported and Data-Driven Rehabilitation, Data Science Institute, Hasselt University, 3590 Diepenbeek, Belgium
| |
Collapse
|
66
|
Li Z, Zhang Y, Fang J, Xu Z, Zhang H, Mao M, Chen Y, Zhang L, Pian C. NcPath: a novel platform for visualization and enrichment analysis of human non-coding RNA and KEGG signaling pathways. Bioinformatics 2022; 39:6917072. [PMID: 36525367 PMCID: PMC9825761 DOI: 10.1093/bioinformatics/btac812] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/10/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
SUMMARY Non-coding RNAs play important roles in transcriptional processes and participate in the regulation of various biological functions, in particular miRNAs and lncRNAs. Despite their importance for several biological functions, the existing signaling pathway databases do not include information on miRNA and lncRNA. Here, we redesigned a novel pathway database named NcPath by integrating and visualizing a total of 178 308 human experimentally validated miRNA-target interactions (MTIs), 32 282 experimentally verified lncRNA-target interactions (LTIs) and 4837 experimentally validated human ceRNA networks across 222 KEGG pathways (including 27 sub-categories). To expand the application potential of the redesigned NcPath database, we identified 556 798 reliable lncRNA-protein-coding genes (PCG) interaction pairs by integrating co-expression relations, ceRNA relations, co-TF-binding interactions, co-histone-modification interactions, cis-regulation relations and lncPro Tool predictions between lncRNAs and PCG. In addition, to determine the pathways in which miRNA/lncRNA targets are involved, we performed a KEGG enrichment analysis using a hypergeometric test. The NcPath database also provides information on MTIs/LTIs/ceRNA networks, PubMed IDs, gene annotations and the experimental verification method used. In summary, the NcPath database will serve as an important and continually updated platform that provides annotation and visualization of the pathways on which non-coding RNAs (miRNA and lncRNA) are involved, and provide support to multimodal non-coding RNAs enrichment analysis. The NcPath database is freely accessible at http://ncpath.pianlab.cn/. AVAILABILITY AND IMPLEMENTATION NcPath database is freely available at http://ncpath.pianlab.cn/. The code and manual to use NcPath can be found at https://github.com/Marscolono/NcPath/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zutan Li
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuan Zhang
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingya Fang
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhihui Xu
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing 210023, China
| | - Hao Zhang
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Minfang Mao
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | - Cong Pian
- To whom correspondence should be addressed. or or
| |
Collapse
|
67
|
Lin R, Xie B, Xie L, Ge J, Li S. Integrated proteomics and metabolomics analysis of lumbar in a rat model of osteoporosis treated with Gushukang capsules. BMC Complement Med Ther 2022; 22:333. [PMID: 36522793 PMCID: PMC9756464 DOI: 10.1186/s12906-022-03807-7] [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: 04/27/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Gushukang (GSK) capsules are a Chinese patented medicine that is widely used in clinics for the treatment of osteoporosis (OP). Animal experiments have revealed that the bone mineral density of osteoporotic rats increase after treatment with GSK capsules. However, the specific mechanism and target of GSK in the treatment of osteoporosis are unclear. Further studies are needed. METHODS Metabolomics (GC/MS) and proteomics (TMT-LC-MC/MC) with bioinformatics (KEGG pathway enrichment), correlation analysis (Pearson correlation matrix), and joint pathway analysis (MetaboAnalyst) were employed to determine the underlying mechanisms of GSK. The differential expression proteins were verified by WB experiment. RESULTS The regulation of proteins, i.e., Cant1, Gstz1, Aldh3b1, Bid, and Slc1a3, in the common metabolic pathway of differential proteins and metabolites between GSK/OP and OP/SHAM was corrected in the GSK group. The regulation of 12 metabolites (tyramine, thymidine, deoxycytidine, cytosine, L-Aspartate, etc.) were differential in the common enrichment metabolic pathway between GSK /OP and OP/SHAM. Differential proteins and metabolites jointly regulate 11 metabolic pathways, such as purine metabolism, pyrimidine metabolism, histidine metabolism, beta-alanine metabolism, and so on. CONCLUSION GSK may protect bone metabolism in osteoporotic rats by affecting nucleotide metabolism, amino acid metabolism, and the immune system.
Collapse
Affiliation(s)
- Ruohui Lin
- Basic Research Institute, Fujian Academy of Chinese Medical Sciences, Fuzhou, 350003 Fujian China ,Fujian Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Osteoporosis, Fuzhou, 350003 Fujian China
| | - Bingying Xie
- Basic Research Institute, Fujian Academy of Chinese Medical Sciences, Fuzhou, 350003 Fujian China ,Fujian Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Osteoporosis, Fuzhou, 350003 Fujian China
| | - Lihua Xie
- Basic Research Institute, Fujian Academy of Chinese Medical Sciences, Fuzhou, 350003 Fujian China ,Fujian Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Osteoporosis, Fuzhou, 350003 Fujian China
| | - Jirong Ge
- Basic Research Institute, Fujian Academy of Chinese Medical Sciences, Fuzhou, 350003 Fujian China ,Fujian Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Osteoporosis, Fuzhou, 350003 Fujian China
| | - Shengqiang Li
- Basic Research Institute, Fujian Academy of Chinese Medical Sciences, Fuzhou, 350003 Fujian China ,Fujian Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Osteoporosis, Fuzhou, 350003 Fujian China
| |
Collapse
|
68
|
Chen S, Xu H, Guo C, Liu Z, Han X. Editorial: The role of multi-omics variants in tumor immunity and immunotherapy. Front Immunol 2022; 13:1098825. [PMID: 36524118 PMCID: PMC9745164 DOI: 10.3389/fimmu.2022.1098825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shuang Chen
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China,*Correspondence: Xinwei Han, ; Zaoqu Liu,
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China,*Correspondence: Xinwei Han, ; Zaoqu Liu,
| |
Collapse
|
69
|
Liu X, Yang C, Chen P, Zhang L, Cao Y. The uses of transcriptomics and lipidomics indicated that direct contact with graphene oxide altered lipid homeostasis through ER stress in 3D human brain organoids. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 849:157815. [PMID: 35931159 DOI: 10.1016/j.scitotenv.2022.157815] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/29/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
The potential uses of graphene-based nanomaterials (NMs) in various fields lead to the concern about their neurotoxicity, considering that graphene-based NMs are capable to cross blood brain barrier (BBB) and enter central nervous system (CNS). Although previous studies reported the possibility of graphene-based NM exposure to alter lipid homeostasis in animals or cultured neurons, recent studies suggested the need to use 3D human brain organoids for mechanism-based toxicological studies as this model might better recapitulate the complex human brains. Herein, we used multi-omics techniques to investigate the mechanisms of graphene oxide (GO) on lipid homeostasis in a novel 3D brain organoid model. We found that 50 μg/mL GO induced cytotoxicity but not superoxide. RNA-sequencing data showed that 50 μg/mL GO significantly up-regulated and down-regulated 80 and 121 genes, respectively. Furthermore, we found that GO exposure altered biological molecule metabolism pathways including lipid metabolism. Consistently, lipidomics data supported dose-dependent alteration of lipid profiles by GO in 3D brain organoids. Interestingly, co-exposure to GO and endoplasmic reticulum (ER) stress inhibitor 4-phenylbutyric acid (4-PBA) decreased most of the lipid classes compared with the exposure of GO only. We further verified that exposure to GO promoted ER stress marker GRP78 proteins, which in turn activated IRE1α/XBP-1 axis, and these changes were partially or completely inhibited by 4-PBA. These results proved that direct contact with GO disrupted lipid homeostasis through the activation of ER stress. As 3D brain organoids resemble human brains, these data might be better extrapolated to humans.
Collapse
Affiliation(s)
- Xudong Liu
- Department of Food science and Engineering, Moutai Institute, Renhuai 564507, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation and Application, Zhejiang Ocean University, Zhoushan, Zhejiang Province 316022, China
| | - P Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada; Advanced Materials Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250014, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada.
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang 421001, China.
| |
Collapse
|
70
|
Dylla L, Higgins HM, Piper C, Poisson SN, Herson PS, Monte AA. Sex as a biological variable in determining the metabolic changes influencing acute ischemic stroke outcomes-Where is the data: A systematic review. Front Neurol 2022; 13:1026431. [PMID: 36504643 PMCID: PMC9729945 DOI: 10.3389/fneur.2022.1026431] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Women continue to face a greater lifetime morbidity and mortality from stroke and have been shown to respond differently to stroke treatments compared to men. Since 2016, updated National Institutes of Health (NIH) policies require research studies to consider sex as a biological variable. However, the way in which this policy affects study design, analysis, and reporting is variable, with few studies performing and reporting a subgroup analysis based on biological sex. In acute ischemic stroke, the underlying biological explanation for sex-based differences in patient outcomes and response to treatments remains understudied. We performed a systematic review of preclinical and clinical research studies that explored sex differences in the metabolic response to acute ischemic stroke as it relates to neurological outcomes. Through a literature search in Ovid Medline, Embase, and Web of Science, 1,004 potential references were identified for screening. After abstract and full-text review, we identified only two studies which assessed metabolic response to acute ischemic stroke (within 72 h of last known well) and neurological outcome [Barthel Index, modified Rankin Scale (mRS) or an equivalent in preclinical models] and reported results based on biological sex. One article was a preclinical rat model and the other a clinical cohort study. In both studies, metabolites involved in amino acid metabolism, energy metabolism, fat metabolism, or oxidative stress were identified. We review these results and link to additional articles that use metabolomics to identify metabolites differentially expressed by sex or regulated based on stroke outcomes, but not both. The results of this systematic review should not only help identify targets in need of further investigation to improve the understanding of sex differences in the pathophysiology of acute ischemic stroke, but also highlight the critical need to expand the incorporation of sex as a biological variable in acute stroke research beyond simply including both sexes and reporting the proportion of males/females in each population studied.
Collapse
Affiliation(s)
- Layne Dylla
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Hannah M. Higgins
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Christi Piper
- Strauss Health Sciences Library, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sharon N. Poisson
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Paco S. Herson
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, United States
| | - Andrew A. Monte
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
71
|
Agamah FE, Bayjanov JR, Niehues A, Njoku KF, Skelton M, Mazandu GK, Ederveen THA, Mulder N, Chimusa ER, 't Hoen PAC. Computational approaches for network-based integrative multi-omics analysis. Front Mol Biosci 2022; 9:967205. [PMID: 36452456 PMCID: PMC9703081 DOI: 10.3389/fmolb.2022.967205] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/20/2022] [Indexed: 08/27/2023] Open
Abstract
Advances in omics technologies allow for holistic studies into biological systems. These studies rely on integrative data analysis techniques to obtain a comprehensive view of the dynamics of cellular processes, and molecular mechanisms. Network-based integrative approaches have revolutionized multi-omics analysis by providing the framework to represent interactions between multiple different omics-layers in a graph, which may faithfully reflect the molecular wiring in a cell. Here we review network-based multi-omics/multi-modal integrative analytical approaches. We classify these approaches according to the type of omics data supported, the methods and/or algorithms implemented, their node and/or edge weighting components, and their ability to identify key nodes and subnetworks. We show how these approaches can be used to identify biomarkers, disease subtypes, crosstalk, causality, and molecular drivers of physiological and pathological mechanisms. We provide insight into the most appropriate methods and tools for research questions as showcased around the aetiology and treatment of COVID-19 that can be informed by multi-omics data integration. We conclude with an overview of challenges associated with multi-omics network-based analysis, such as reproducibility, heterogeneity, (biological) interpretability of the results, and we highlight some future directions for network-based integration.
Collapse
Affiliation(s)
- Francis E. Agamah
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jumamurat R. Bayjanov
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anna Niehues
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kelechi F. Njoku
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gaston K. Mazandu
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- African Institute for Mathematical Sciences, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. 't Hoen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
72
|
Cheema Z, Kwinta P, Moreira A, Tovar M, Mustafa SB. Big Data for Tiny Patients: A Precision Medicine Approach to Bronchopulmonary Dysplasia. Pediatr Ann 2022; 51:e396-e404. [PMID: 36215088 DOI: 10.3928/19382359-20220803-06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease of extreme prematurity. Despite more than 50 years of research, current treatments are ineffective, and clinicians are largely unable to accurately predict which neonates the condition will develop in. A deeper understanding of the molecular mechanisms underlying the characteristic arrest in lung development are warranted. Integrating high-fidelity technology from precision medicine approaches may fill this gap and provide the tools necessary to identify biomarkers and targetable pathways. In this review, we describe insights garnered from current studies using omics for BPD prediction and stratification. We conclude by describing novel programs that will integrate multi-omics in efforts to better understand and treat the pathogenesis of BPD. [Pediatr Ann. 2022;51(10):e396-e404.].
Collapse
|
73
|
Wang X, Zhang HG, Xiong X, Hong C, Weber GM, Brat GA, Bonzel CL, Luo Y, Duan R, Palmer NP, Hutch MR, Gutiérrez-Sacristán A, Bellazzi R, Chiovato L, Cho K, Dagliati A, Estiri H, García-Barrio N, Griffier R, Hanauer DA, Ho YL, Holmes JH, Keller MS, Klann MEng JG, L'Yi S, Lozano-Zahonero S, Maidlow SE, Makoudjou A, Malovini A, Moal B, Moore JH, Morris M, Mowery DL, Murphy SN, Neuraz A, Yuan Ngiam K, Omenn GS, Patel LP, Pedrera-Jiménez M, Prunotto A, Jebathilagam Samayamuthu M, Sanz Vidorreta FJ, Schriver ER, Schubert P, Serrano-Balazote P, South AM, Tan ALM, Tan BWL, Tibollo V, Tippmann P, Visweswaran S, Xia Z, Yuan W, Zöller D, Kohane IS, Avillach P, Guo Z, Cai T. SurvMaximin: Robust federated approach to transporting survival risk prediction models. J Biomed Inform 2022; 134:104176. [PMID: 36007785 PMCID: PMC9707637 DOI: 10.1016/j.jbi.2022.104176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/18/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVE For multi-center heterogeneous Real-World Data (RWD) with time-to-event outcomes and high-dimensional features, we propose the SurvMaximin algorithm to estimate Cox model feature coefficients for a target population by borrowing summary information from a set of health care centers without sharing patient-level information. MATERIALS AND METHODS For each of the centers from which we want to borrow information to improve the prediction performance for the target population, a penalized Cox model is fitted to estimate feature coefficients for the center. Using estimated feature coefficients and the covariance matrix of the target population, we then obtain a SurvMaximin estimated set of feature coefficients for the target population. The target population can be an entire cohort comprised of all centers, corresponding to federated learning, or a single center, corresponding to transfer learning. RESULTS Simulation studies and a real-world international electronic health records application study, with 15 participating health care centers across three countries (France, Germany, and the U.S.), show that the proposed SurvMaximin algorithm achieves comparable or higher accuracy compared with the estimator using only the information of the target site and other existing methods. The SurvMaximin estimator is robust to variations in sample sizes and estimated feature coefficients between centers, which amounts to significantly improved estimates for target sites with fewer observations. CONCLUSIONS The SurvMaximin method is well suited for both federated and transfer learning in the high-dimensional survival analysis setting. SurvMaximin only requires a one-time summary information exchange from participating centers. Estimated regression vectors can be very heterogeneous. SurvMaximin provides robust Cox feature coefficient estimates without outcome information in the target population and is privacy-preserving.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Harrison G Zhang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Xin Xiong
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Chuan Hong
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Griffin M Weber
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Gabriel A Brat
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Clara-Lea Bonzel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yuan Luo
- Department of Preventive Medicine Northwestern University, Chicago, IL, USA
| | - Rui Duan
- Department of Biostatistics, Harvard University, Boston, MA, USA
| | - Nathan P Palmer
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Meghan R Hutch
- Department of Preventive Medicine Northwestern University, Chicago, IL, USA
| | | | - Riccardo Bellazzi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Luca Chiovato
- Unit of Internal Medicine and Endocrinology, Istituti Clinici Scientifici Maugeri SpA SB IRCCS, Pavia, Italy
| | - Kelly Cho
- Population Health and Data Science, VA Boston Healthcare System, Boston, MA, USA; Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Arianna Dagliati
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Hossein Estiri
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Romain Griffier
- IAM unit, Bordeaux University Hospital, Bordeaux, France; INSERM Bordeaux Population Health ERIAS TEAM, ERIAS - Inserm U1219 BPH, Bordeaux, France
| | - David A Hanauer
- Department of Learning Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - John H Holmes
- Department of Biostatistics, Epidemiology, and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark S Keller
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | | | - Sehi L'Yi
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Sara Lozano-Zahonero
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Sarah E Maidlow
- Michigan Institute for Clinical and Health Research (MICHR) Informatics, University of Michigan, Ann Arbor, MI, USA
| | - Adeline Makoudjou
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Alberto Malovini
- Laboratory of Informatics and Systems Engineering for Clinical Research, Istituti Clinici Scientifici Maugeri SpA SB IRCCS, Pavia, Italy
| | - Bertrand Moal
- IAM unit, Bordeaux University Hospital, Bordeaux, France
| | - Jason H Moore
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michele Morris
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Danielle L Mowery
- Department of Biostatistics, Epidemiology, and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shawn N Murphy
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Antoine Neuraz
- Department of biomedical informatics, Hôpital Necker-Enfants Malade, Assistance Publique Hôpitaux de Paris (APHP), University of Paris, Paris, France
| | - Kee Yuan Ngiam
- Department of Biomedical informatics, WiSDM, National University Health Systems, Singapore
| | - Gilbert S Omenn
- Depts of Computational Medicine & Bioinformatics, Internal Medicine, Human Genetics, Public Health University of Michigan, Ann Arbor, MI, USA
| | - Lav P Patel
- Department of Internal Medicine, Division of Medical Informatics, University Of Kansas Medical Center
| | | | - Andrea Prunotto
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | | | | | - Emily R Schriver
- Data Analytics Center, University of Pennsylvania Health System, Philadelphia, PA, USA
| | - Petra Schubert
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | | | - Andrew M South
- Department of Pediatrics-Section of Nephrology, Brenner Children's, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Amelia L M Tan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Byorn W L Tan
- Department of Medicine, National University Hospital, Singapore
| | - Valentina Tibollo
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Patric Tippmann
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Shyam Visweswaran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zongqi Xia
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - William Yuan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Daniela Zöller
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Isaac S Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Zijian Guo
- Department of Statistics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Tianxi Cai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
74
|
Levy J, Silva AM, De Carli E, Cacau LT, de Alvarenga JFR, Fiamoncini J, Benseñor IM, Lotufo PA, Marchioni DM. Biomarkers of Fruit Intake Using a Targeted Metabolomics Approach: an Observational Cross-Sectional Analysis of the ELSA-Brasil Study. J Nutr 2022; 152:2023-2030. [PMID: 35641174 DOI: 10.1093/jn/nxac115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/11/2022] [Accepted: 05/24/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Advances in technology have led to the identification of a greater number of metabolites related to diet. Although fruit intake biomarkers have been reported in some studies, these findings require further replication, considering the relevance of fruits for diet quality and health. OBJECTIVES The aim of this study was to explore the associations of a set of potential urinary biomarkers of diet, assessed using a targeted metabolomics approach, with self-reported fruit intake data in participants of a computer-assisted 24-h dietary recall (GloboDiet software) validation study. METHODS A total of 93 individuals aged 43-72 y, 54% female, participated in this study. The subjects were a subsample of the Longitudinal Study of Adult Health (ELSA-Brasil). A 24-h dietary recall was obtained with the aid of GloboDiet software matching a 24-h urine sample from each participant. Candidate biomarkers were selected in a literature search and identified in urine by LC coupled to high-resolution MS. Spearman correlation analyses were performed between fruit intake and each biomarker. RESULTS Spearman correlation analysis showed that total fruits intake was significantly correlated with citric acid (ρ = 0.213, P = 0.041), ferulic acid sulfate I (ρ = 0.240, P = 0.020), hesperetin glucuronide/homoeriodictyol glucuronide (ρ = 0.303, P = 0.003), hydroxyhippuric acid (ρ = 0.239, P = 0.021), homovanillic alcohol sulfate (ρ = 0.339, P = 0.001), methylgallic acid sulfate (ρ = 0.268, P = 0.009), naringenin glucuronide (NG; ρ = 0.278, P = 0.007), proline betaine (PB; ρ = 0.305, P = 0.003), syringic acid sulfate (ρ = 0.210, P = 0.044), and sinapic acid sulfate (ρ = 0.412, P < 0.001). Among them, 3 have been described in literature as promising biomarkers for intake of total fruit, oranges, and citrus fruit: NG, hesperetin glucuronide, and PB. CONCLUSIONS Associations of total fruits intake with urinary measurements indicate the potential usefulness of dietary biomarkers in the Brazilian population as a complement to self-reported dietary assessments.
Collapse
Affiliation(s)
- Jessica Levy
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Alexsandro Macedo Silva
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Eduardo De Carli
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Leandro Teixeira Cacau
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - José Fernando Rinaldi de Alvarenga
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jarlei Fiamoncini
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Isabela Martins Benseñor
- Clinical and Epidemiological Research Center, University Hospital, University of São Paulo, São Paulo, Brazil
| | - Paulo Andrade Lotufo
- Clinical and Epidemiological Research Center, University Hospital, University of São Paulo, São Paulo, Brazil
| | - Dirce Maria Marchioni
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
75
|
Titanji BK, Wang Z, Chen J, Hui Q, So-Armah K, Freiberg M, Justice AC, Ke X, Marconi VC, Sun YV. Soluble CD14-associated DNA methylation sites predict mortality among men with HIV infection. AIDS 2022; 36:1563-1571. [PMID: 35979830 PMCID: PMC9394925 DOI: 10.1097/qad.0000000000003279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Elevated plasma levels of sCD14 predict all-cause mortality in people with HIV (PWH). Epigenetic regulation plays a key role in infection and inflammation. To reveal the epigenetic relationships between sCD14, immune function and disease progression among PWH, we conducted an epigenome-wide association study (EWAS) of sCD14 and investigated the relationship with mortality. DESIGN AND METHODS DNA methylation (DNAm) levels of peripheral blood samples from PWH in the Veterans Aging Cohort Study (VACS) were measured using the Illumina Infinium Methylation 450K (n = 549) and EPIC (850K) BeadChip (n = 526). Adjusted for covariates and multiple testing, we conducted an epigenome-wide discovery, replication, and meta-analysis to identify significant associations with sCD14. We then examined and replicated the relationship between the principal epigenetic sites and survival using Cox regression models. FINDINGS We identified 118 DNAm sites significantly associated with sCD14 in the meta-analysis of 1075 PWH. The principal associated DNAm sites mapped to genes (e.g. STAT1, PARP9, IFITM1, MX1, and IFIT1) related to inflammation and antiviral response. Adjusting for multiple testing, 10 of 118 sCD14-associated DNAm sites significantly predicted survival time conditional on sCD14 levels. CONCLUSION The identification of DNAm sites independently predicting survival may improve our understanding of prognosis and potential therapeutic targets among PWH.
Collapse
Affiliation(s)
- Boghuma K Titanji
- Division of Infectious Diseases, Emory University School of Medicine
| | - Zeyuan Wang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Qin Hui
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | | | - Matthew Freiberg
- Cardiovascular Medicine Division and Tennessee Valley Healthcare System, Vanderbilt University Medical Center, Nashville, TN
| | - Amy C Justice
- Yale University School of Medicine, New Haven
- Connecticut Veteran Health System, West Haven, CT
| | - Xu Ke
- Yale University School of Medicine, New Haven
- Connecticut Veteran Health System, West Haven, CT
| | - Vincent C Marconi
- Division of Infectious Diseases, Emory University School of Medicine
- Atlanta Veterans Affairs Healthcare System, Decatur
- Hubert Department of Global Health, Rollins School of Public Health
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
- Atlanta Veterans Affairs Healthcare System, Decatur
| |
Collapse
|
76
|
Metabolomics and Biomarkers in Retinal and Choroidal Vascular Diseases. Metabolites 2022; 12:metabo12090814. [PMID: 36144219 PMCID: PMC9503269 DOI: 10.3390/metabo12090814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022] Open
Abstract
The retina is one of the most important structures in the eye, and the vascular health of the retina and choroid is critical to visual function. Metabolomics provides an analytical approach to endogenous small molecule metabolites in organisms, summarizes the results of “gene-environment interactions”, and is an ideal analytical tool to obtain “biomarkers” related to disease information. This study discusses the metabolic changes in neovascular diseases involving the retina and discusses the progress of the study from the perspective of metabolomics design and analysis. This study advocates a comparative strategy based on existing studies, which encompasses optimization of the performance of newly identified biomarkers and the consideration of the basis of existing studies, which facilitates quality control of newly discovered biomarkers and is recommended as an additional reference strategy for new biomarker discovery. Finally, by describing the metabolic mechanisms of retinal and choroidal neovascularization, based on the results of existing studies, this study provides potential opportunities to find new therapeutic approaches.
Collapse
|
77
|
Network Pharmacology of Adaptogens in the Assessment of Their Pleiotropic Therapeutic Activity. Pharmaceuticals (Basel) 2022; 15:ph15091051. [PMID: 36145272 PMCID: PMC9504187 DOI: 10.3390/ph15091051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 02/07/2023] Open
Abstract
The reductionist concept, based on the ligand–receptor interaction, is not a suitable model for adaptogens, and herbal preparations affect multiple physiological functions, revealing polyvalent pharmacological activities, and are traditionally used in many conditions. This review, for the first time, provides a rationale for the pleiotropic therapeutic efficacy of adaptogens based on evidence from recent gene expression studies in target cells and where the network pharmacology and systems biology approaches were applied. The specific molecular targets and adaptive stress response signaling mechanisms involved in nonspecific modes of action of adaptogens are identified.
Collapse
|
78
|
Pan J, Ma B, Hou X, Li C, Xiong T, Gong Y, Song F. The construction of transcriptional risk scores for breast cancer based on lightGBM and multiple omics data. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:12353-12370. [PMID: 36654001 DOI: 10.3934/mbe.2022576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
BACKGROUND Polygenic risk score (PRS) can evaluate the individual-level genetic risk of breast cancer. However, standalone single nucleotide polymorphisms (SNP) data used for PRS may not provide satisfactory prediction accuracy. Additionally, current PRS models based on linear regression have insufficient power to leverage non-linear effects from thousands of associated SNPs. Here, we proposed a transcriptional risk score (TRS) based on multiple omics data to estimate the risk of breast cancer. METHODS The multiple omics data and clinical data of breast invasive carcinoma (BRCA) were collected from the cancer genome atlas (TCGA) and the gene expression omnibus (GEO). First, we developed a novel TRS model for BRCA utilizing single omic data and LightGBM algorithm. Subsequently, we built a combination model of TRS derived from each omic data to further improve the prediction accuracy. Finally, we performed association analysis and prognosis prediction to evaluate the utility of the TRS generated by our method. RESULTS The proposed TRS model achieved better predictive performance than the linear models and other ML methods in single omic dataset. An independent validation dataset also verified the effectiveness of our model. Moreover, the combination of the TRS can efficiently strengthen prediction accuracy. The analysis of prevalence and the associations of the TRS with phenotypes including case-control and cancer stage indicated that the risk of breast cancer increases with the increases of TRS. The survival analysis also suggested that TRS for the cancer stage is an effective prognostic metric of breast cancer patients. CONCLUSIONS Our proposed TRS model expanded the current definition of PRS from standalone SNP data to multiple omics data and outperformed the linear models, which may provide a powerful tool for diagnostic and prognostic prediction of breast cancer.
Collapse
Affiliation(s)
- Jianqiao Pan
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Tianjin, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Baoshan Ma
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Xiaoyu Hou
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Chongyang Li
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Tong Xiong
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Yi Gong
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Tianjin, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| |
Collapse
|
79
|
Metabolomics: A New Approach in the Evaluation of Effects in Human Beings and Wildlife Associated with Environmental Exposition to POPs. TOXICS 2022; 10:toxics10070380. [PMID: 35878286 PMCID: PMC9320281 DOI: 10.3390/toxics10070380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/28/2022] [Accepted: 07/06/2022] [Indexed: 12/10/2022]
Abstract
Human beings and wild organisms are exposed daily to a broad range of environmental stressors. Among them are the persistent organic pollutants that can trigger adverse effects on these organisms due to their toxicity properties. There is evidence that metabolomics can be used to identify biomarkers of effect by altering the profiles of endogenous metabolites in biological fluids or tissues. This approach is relatively new and has been used in vitro studies mainly. Therefore, this review addresses those that have used metabolomics as a key tool to identify metabolites associated with environmental exposure to POPs in wildlife and human populations and that can be used as biomarkers of effect. The published results suggest that the metabolic pathways that produce energy, fatty acids, and amino acids are commonly affected by POPs. Furthermore, these pathways can be promoters of additional effects. In the future, metabolomics combined with other omics will improve understanding of the origin, development, and progression of the effects caused by environmental exposure.
Collapse
|
80
|
Effects of Storage Temperature on Indica-Japonica Hybrid Rice Metabolites, Analyzed Using Liquid Chromatography and Mass Spectrometry. Int J Mol Sci 2022; 23:ijms23137421. [PMID: 35806428 PMCID: PMC9266784 DOI: 10.3390/ijms23137421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 12/04/2022] Open
Abstract
The Yongyou series of indica-japonica hybrid rice has excellent production potential and storage performance. However, little is known about the underlying mechanism of its storage resistance. In this study, Yongyou 1540 rice (Oryza sativa cv. yongyou 1540) was stored at different temperatures, and the storability was validated though measuring nutritional components and apparent change. In addition, a broad-targeted metabolomic approach coupled with liquid chromatography-mass spectrometry was applied to analyze the metabolite changes. The study found that under high temperature storage conditions (35 °C), Yongyou 1540 was not significantly worse in terms of fatty acid value, whiteness value, and changes in electron microscope profile. A total of 19 key differential metabolites were screened, and lipid metabolites related to palmitoleic acid were found to affect the aging of rice. At the same time, two substances, guanosine 3′,5′-cyclophosphate and pipecolic acid, were beneficial to enhance the resistance of rice under harsh storage conditions, thereby delaying the deterioration of its quality and maintaining its quality. Significant regulation of galactose metabolism, alanine, aspartate and glutamate metabolism, butyrate metabolism, and arginine and proline metabolism pathways were probably responsible for the good storage capacity of Yongyou 1540.
Collapse
|
81
|
Cong Y, Endo T. Multi-Omics and Artificial Intelligence-Guided Drug Repositioning: Prospects, Challenges, and Lessons Learned from COVID-19. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:361-371. [PMID: 35759424 DOI: 10.1089/omi.2022.0068] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drug repurposing is of interest for therapeutics innovation in many human diseases including coronavirus disease 2019 (COVID-19). Methodological innovations in drug repurposing are currently being empowered by convergence of omics systems science and digital transformation of life sciences. This expert review article offers a systematic summary of the application of artificial intelligence (AI), particularly machine learning (ML), to drug repurposing and classifies and introduces the common clustering, dimensionality reduction, and other methods. We highlight, as a present-day high-profile example, the involvement of AI/ML-based drug discovery in the COVID-19 pandemic and discuss the collection and sharing of diverse data types, and the possible futures awaiting drug repurposing in an era of AI/ML and digital technologies. The article provides new insights on convergence of multi-omics and AI-based drug repurposing. We conclude with reflections on the various pathways to expedite innovation in drug development through drug repurposing for prompt responses to the current COVID-19 pandemic and future ecological crises in the 21st century.
Collapse
Affiliation(s)
- Yi Cong
- Laboratory of Information Biology, Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Toshinori Endo
- Laboratory of Information Biology, Information Science and Technology, Hokkaido University, Sapporo, Japan
| |
Collapse
|
82
|
Fan H, Zhang J, Zou B, He Z. The Role of CEP55 Expression in Tumor Immune Response and Prognosis of Patients with Non-small Cell lung Cancer. ARCHIVES OF IRANIAN MEDICINE 2022; 25:432-442. [DOI: 10.34172/aim.2022.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 11/20/2021] [Indexed: 11/06/2022]
Abstract
Background: With the continuous advancement of diagnostic methods, more and more early-stage Non-small cell lung cancer (NSCLC) patients are diagnosed. Although many scholars have devoted substantial efforts to investigate the pathogenesis and prognosis of NSCLC, its molecular mechanism is still not well explained. Methods: We retrieved three gene datasets GSE10072, GSE19188 and GSE40791 from the Gene Expression Omnibus (GEO) database and screened and identified differentially expressed genes (DEGs). Then, we performed KEGG and GO functional enrichment analysis, survival analysis, risk analysis and prognosis analysis on the selected hub genes. We constructed a protein-protein interaction (PPI) network, and used the STRING database and Cytoscape software. Results: The biological process analysis showed that these genes were mainly enriched in cell division and nuclear division. Survival analysis showed that the genes of CEP55 (centrosomal protein 55), NMU (neuromedin U), CAV1 (Caveolin 1), TBX3 (T-box transcription factor 3), FBLN1 (fibulin 1) and SYNM (synemin) may be involved in the development, invasion or metastasis of NSCLC (P<0.05, logFC>1). Prognostic analysis and independent prognostic analysis showed that the expression of these hub gene-related mRNAs was related to the prognostic risk of NSCLC. Risk analysis showed that the selected hub genes were closely related to the overall survival time of patients with NSCLC. Conclusion: The DEGs and hub genes screened and identified in this study will help us to understand the molecular mechanisms of NSCLC, and CEP55 expression affects the survival and prognosis of patients with NSCLC, and participates in tumor immune response.
Collapse
Affiliation(s)
- Haiyin Fan
- Thoracic Department, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Jin Zhang
- Ultrasound Department, Jiangxi Chest Hospital, Nanchang, Jiangxi, China
| | - Bin Zou
- Thoracic Department, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Zhisheng He
- Thoracic Department, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
83
|
Vitório JG, Duarte-Andrade FF, Pereira TDSF, Melo Braga MND, Canuto GAB, Macedo AND, Lebron YAR, Moreira VR, Felicori LF, Lange LC, Souza Santos LVD, Larsen MR, Gomes CC, Gomez RS. Integrated proteomics, phosphoproteomics and metabolomics analyses reveal similarities amongst giant cell granulomas of the jaws with different genetic mutations. J Oral Pathol Med 2022; 51:666-673. [PMID: 35706152 DOI: 10.1111/jop.13327] [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: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Giant cell granuloma of the jaws are benign osteolytic lesions of the jaws. These lesions are genetically characterized by mutually exclusive somatic mutations at TRPV4, KRAS, and FGFR1, and a fourth molecular subgroup which is wild-type for the three mutations. Irrespective of the molecular background, giant cell granulomas show MAPK/ERK activation. However, it remains unclear if these mutations lead to differences in their molecular signaling in giant cell granulomas. METHODS Metabolomics, proteomics and phosphoproteomics analyses were carried out in formalin-fixed paraffin-embedded samples of giant cell granuloma of the jaws. The study cohort consisted of five lesions harboring mutations in FGFR1, six in KRAS, five in TRPV4 and five that were wild-type for these mutations. RESULTS Lesions harboring KRAS or FGFR1 mutations showed overall similar proteomics and metabolomics profiles. In all four groups, metabolic pathways showed similarity in apoptosis, cell signaling, gene expression, cell differentiation and erythrocyte activity. Lesions harboring TRPV4 mutations showed a greater number of enriched pathways related to tissue architecture. On the other hand, the wild-type group presented increased number of enriched pathways related to protein metabolism compared to the other groups. CONCLUSION Despite some minor differences, our results revealed an overall similar molecular profile among the groups with different mutational profile at the metabolic, proteic and phosphopeptidic levels. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jéssica Gardone Vitório
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Filipe Fideles Duarte-Andrade
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Thais Dos Santos Fontes Pereira
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Marcella Nunes de Melo Braga
- Department of Biochemistry and Immunology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gisele André Baptista Canuto
- Department of Analytical Chemistry, Institute of Chemistry, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Adriana Nori de Macedo
- Department of Analytical Chemistry, Institute of Chemistry, Universidade Federal da Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Yuri Abner Rocha Lebron
- Department of Department of Sanitary and Environmental Engineering, Engineer School, Universidade Federal de Minas Gerais (UFMG),, Belo Horizonte, Brazil
| | - Victor Rezende Moreira
- Department of Department of Sanitary and Environmental Engineering, Engineer School, Universidade Federal de Minas Gerais (UFMG),, Belo Horizonte, Brazil
| | - Liza Figueiredo Felicori
- Department of Biochemistry and Immunology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Liséte Celina Lange
- Department of Department of Sanitary and Environmental Engineering, Engineer School, Universidade Federal de Minas Gerais (UFMG),, Belo Horizonte, Brazil
| | - Lucilaine Valéria de Souza Santos
- Department of Department of Sanitary and Environmental Engineering, Engineer School, Universidade Federal de Minas Gerais (UFMG),, Belo Horizonte, Brazil
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark
| | - Carolina Cavalieri Gomes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
84
|
Cheema AK, Li Y, Moulton J, Girgis M, Wise SY, Carpenter A, Fatanmi OO, Singh VK. Identification of novel biomarkers for acute radiation injury using multi-omics approach and nonhuman primate model. Int J Radiat Oncol Biol Phys 2022; 114:310-320. [PMID: 35675853 DOI: 10.1016/j.ijrobp.2022.05.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/02/2022] [Accepted: 05/28/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE The availability of validated biomarkers to assess radiation exposure and to assist in developing medical countermeasures remains an unmet need. METHODS AND MATERIALS We used a cobalt-60 gamma-irradiated nonhuman primate (NHP) model to delineate a multi-omics-based serum probability index of radiation exposure. Both male and female NHPs were irradiated with different doses ranging from 6.0 to 8.5 Gy, with 0.5 Gy increments between doses. We leveraged high-resolution mass spectrometry for analysis of metabolites, lipids, and proteins at 1, 2, and 6 days post-irradiation in NHP serum. RESULTS A logistic regression model was implemented to develop a 4-analyte panel to stratify irradiated NHPs from unirradiated with high accuracy that was agnostic for all doses of gamma-rays tested in the study, up to six days after exposure. This panel was comprised of Serpin Family A9, acetylcarnitine, PC (16:0/22:6), and suberylglycine, which showed 2 - 4-fold elevation in serum abundance upon irradiation in NHPs and can potentially be translated as a molecular diagnostic for human use following larger validation studies. CONCLUSIONS Taken together, this study, for the first time, demonstrates the utility of a combinatorial molecular characterization approach using an NHP model for developing minimally invasive assays from small volumes of blood that can be effectively used for radiation exposure assessments.
Collapse
Affiliation(s)
- Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Department of Biochemistry; Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA.
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Department of Biochemistry
| | - Joanna Moulton
- Department of Oncology, Lombardi Comprehensive Cancer Center, Department of Biochemistry
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Department of Biochemistry
| | - Stephen Y Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alana Carpenter
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Oluseyi O Fatanmi
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
85
|
Li X, Zhang L, Li T, Li S, Wu W, Zhao L, Xie P, Yang J, Li P, Zhang Y, Xiao H, Yu Y, Zhao Z. Abplatin (IV) inhibited tumor growth on a patient derived cancer model of hepatocellular carcinoma and its comparative multi-omics study with cisplatin. J Nanobiotechnology 2022; 20:258. [PMID: 35659243 PMCID: PMC9164404 DOI: 10.1186/s12951-022-01465-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/18/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cisplatin, the alkylating agent of platinum(II) (Pt(II)), is the most common antitumor drug in clinic; however, it has many side effects, therefore it is higly desired to develop low toxicity platinum(IV) (Pt(IV)) drugs. Multi-omics analysis, as a powerful tool, has been frequently employed for the mechanism study of a certain therapy at the molecular level, which might be helpful for elucidating the mechanism of platinum drugs and facilitating their clinical application. METHODS Strating form cisplatin, a hydrophobic Pt(IV) prodrug (CisPt(IV)) with two hydrophobic aliphatic chains was synthesized, and further encapsulated with a drug carrier, human serum albumin (HSA), to form nanoparticles, namely AbPlatin(IV). The anticancer effect of AbPlatin(IV) was investigated in vitro and in vivo. Moreover, transcriptomics, metabolomics and lipidomics were performed to explore the mechanism of AbPlatin(IV). RESULTS Compared with cisplatin, Abplatin(IV) exhibited better tumor-targeting effect and greater tumor inhibition rate. Lipidomics study showed that Abplatin(IV) might induce the changes of BEL-7404 cell membrane, and cause the disorder of glycerophospholipids and sphingolipids. In addition, transcriptomics and metabolomics study showed that Abplatin(IV) significantly disturbed the purine metabolism pathway. CONCLUSIONS This research highlighted the development of Abplatin(IV) and the use of multi-omics for the mechanism elucidation of prodrug, which is the key to the clinical translation of prodrug.
Collapse
Affiliation(s)
- Xing Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
- Graduate School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
- College of Life Science and Technology; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Tuo Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Shumu Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
| | - Wenjing Wu
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
- Graduate School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingyu Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
- Graduate School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peng Xie
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jinqi Yang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
- Graduate School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peipei Li
- Graduate School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yangyang Zhang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China
| | - Haihua Xiao
- Graduate School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingjie Yu
- College of Life Science and Technology; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Zhenwen Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing, 100190, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
86
|
Abstract
Metabolomics emerged as an important tool to gain insights on how the body responds to therapeutic interventions. Bariatric surgery is the most effective treatment for severe obesity and obesity-related co-morbidities. Our aim was to conduct a systematic review of the available data on metabolomics profiles that characterize patients submitted to different bariatric surgery procedures, which could be useful to predict clinical outcomes including weight loss and type 2 diabetes remission. For that, the Preferred Reporting Items for Systematic Reviews and Meta-Analyses - PRISMA guidelines were followed. Data from forty-seven original study reports addressing metabolomics profiles induced by bariatric surgery that met eligibility criteria were compiled and summarized. Amino acids, lipids, energy-related and gut microbiota-related were the metabolite classes most influenced by bariatric surgery. Among these, higher pre-operative levels of specific lipids including phospholipids, long-chain fatty acids and bile acids were associated with post-operative T2D remission. As conclusion, metabolite profiling could become a useful tool to predict long term response to different bariatric surgery procedures, allowing more personalized interventions and improved healthcare resources allocation.
Collapse
Affiliation(s)
- Matilde Vaz
- Endocrine & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Sofia S Pereira
- Endocrine & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Mariana P Monteiro
- Endocrine & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.
- Department of Anatomy, School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal.
| |
Collapse
|
87
|
Huang Y, Chen R, Yang S, Chen Y, Lü X. The Mechanism of Interaction Between Gold Nanoparticles and Human Dermal Fibroblasts Based on Integrative Analysis of Transcriptomics and Metabolomics Data. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The aim of this paper was to combine transcriptomics and metabolomics to analyze the mechanism of gold nanoparticles (GNPs) on human dermal fibroblasts (HDFs). First, 20-nm GNPs were prepared, and the differentially expressed genes in HDFs were subsequently screened by transcriptome
sequencing technology after 4, 8, and 24 h of treatment with GNPs. By comparing the metabolic pathways in which the metabolites obtained in a previous study were involved, the pathways involving both genes and metabolites were filtered, and the differentially expressed genes and metabolites
with upstream and downstream relationships were screened out. The gene–metabolite–metabolic pathway network was further constructed, and the functions of metabolic pathways, genes and metabolites in the important network were analyzed and experimentally verified. The results of
transcriptome sequencing experiments showed that 1904, 1216 and 489 genes were differentially expressed in HDFs after 4, 8 and 24 h of treatment with GNPs, and these genes were involved in 270, 235 and 163 biological pathways, respectively. Through the comparison and analysis of the metabolic
pathways affected by the metabolites, 7, 3 and 2 metabolic pathways with genes and metabolites exhibiting upstream and downstream relationships were identified. Through analysis of the gene–metabolite–metabolic pathway network, 4 important metabolic pathways, 9 genes and 7 metabolites
were identified. Combined with the results of verification experiments on oxidative stress, apoptosis, the cell cycle, the cytoskeleton and cell adhesion, it was found that GNPs regulated the synthesis of downstream metabolites through upstream genes in important metabolic pathways. GNPs inhibited
oxidative stress and thus did not induce significant apoptosis, but they exerted effects on several cellular functions, including arresting the cell cycle and affecting the cytoskeleton and cell adhesion.
Collapse
Affiliation(s)
- Yan Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, Jiangsu, PR China
| | - Rong Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, Jiangsu, PR China
| | - Shuci Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, Jiangsu, PR China
| | - Ye Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, Jiangsu, PR China
| | - Xiaoying Lü
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, Jiangsu, PR China
| |
Collapse
|
88
|
Ren Y, Yu G, Shi C, Liu L, Guo Q, Han C, Zhang D, Zhang L, Liu B, Gao H, Zeng J, Zhou Y, Qiu Y, Wei J, Luo Y, Zhu F, Li X, Wu Q, Li B, Fu W, Tong Y, Meng J, Fang Y, Dong J, Feng Y, Xie S, Yang Q, Yang H, Wang Y, Zhang J, Gu H, Xuan H, Zou G, Luo C, Huang L, Yang B, Dong Y, Zhao J, Han J, Zhang X, Huang H. Majorbio Cloud: A one-stop, comprehensive bioinformatic platform for multiomics analyses. IMETA 2022; 1:e12. [PMID: 38868573 PMCID: PMC10989754 DOI: 10.1002/imt2.12] [Citation(s) in RCA: 242] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 06/14/2024]
Abstract
The platform consists of three modules, which are pre-configured bioinformatic pipelines, cloud toolsets, and online omics' courses. The pre-configured bioinformatic pipelines not only combine analytic tools for metagenomics, genomes, transcriptome, proteomics and metabolomics, but also provide users with powerful and convenient interactive analysis reports, which allow them to analyze and mine data independently. As a useful supplement to the bioinformatics pipelines, a wide range of cloud toolsets can further meet the needs of users for daily biological data processing, statistics, and visualization. The rich online courses of multi-omics also provide a state-of-art platform to researchers in interactive communication and knowledge sharing.
Collapse
Affiliation(s)
- Yi Ren
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Guo Yu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Caiping Shi
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Linmeng Liu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Quan Guo
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Chang Han
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Dan Zhang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Lei Zhang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Binxu Liu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Hao Gao
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Jing Zeng
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yong Zhou
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yuhan Qiu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Jian Wei
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yanchun Luo
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Fengjuan Zhu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Xiaojie Li
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Qin Wu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Bing Li
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Wenyao Fu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yanli Tong
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Jie Meng
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yahong Fang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Jie Dong
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yitong Feng
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Shichang Xie
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Qianqian Yang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Hui Yang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yan Wang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Junbiao Zhang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Haidong Gu
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Hongdong Xuan
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Guanqing Zou
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Chun Luo
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Long Huang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Bing Yang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Yachen Dong
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Jianhua Zhao
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Jichen Han
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Xianglin Zhang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| | - Huasheng Huang
- Shanghai Majorbio Bio‐pharm Technology Co., Ltd.ShanghaiChina
| |
Collapse
|
89
|
Donati VL, Madsen L, Middelboe M, Strube ML, Dalsgaard I. The Gut Microbiota of Healthy and Flavobacterium psychrophilum-Infected Rainbow Trout Fry Is Shaped by Antibiotics and Phage Therapies. Front Microbiol 2022; 13:771296. [PMID: 35620089 PMCID: PMC9128845 DOI: 10.3389/fmicb.2022.771296] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/07/2022] [Indexed: 01/15/2023] Open
Abstract
In the aquaculture sector, there is an increased interest in developing environmentally friendly alternatives to antibiotics in the treatment and prevention of bacterial infections. This requires an understanding of the effects of different treatments on the fish microbiota as a measure for improving the fish health status. In this study, we focused on the freshwater pathogen Flavobacterium psychrophilum and investigated the effects of antibiotics (florfenicol) and phage therapies on the gut microbiota of healthy and infected rainbow trout fry (1–2 g). Florfenicol-coated feed was administered for 10 days, starting two days after the infection procedure. A two-component mix of phage targeting F. psychrophilum (FpV4 and FPSV-D22) was continuously delivered by feed with a prophylactic period of 12 days. Samples of the distal intestine were collected over time (day -1 and 1, 8, and 33 days post-infection) and analyzed by community analysis targeting the 16S rRNA gene (V3–V4 region). Results showed the dysbiosis effect caused both by the infection and by florfenicol administration. Shifts in the overall composition were detected by β-diversity analysis, and changes in specific populations were observed during taxonomic mapping. Measures of α-diversity were only affected in infected fish (large variation observed 1 and 8 dpi). These community alterations disappeared again when fish recovered from the infection and the antibiotic treatment was terminated (33 dpi). Interestingly, phage addition altered the microbiota of the fish independently of the presence of their target bacterium. The overall gut bacterial community in fish fed phage-treated feed was different from the controls at each time point as revealed by β-diversity analysis. However, it was not possible to identify specific bacterial populations responsible for these changes except for an increase of lactic acid bacteria 33 dpi. Overall, the results indicate that the administered phages might affect the complex network of phage-bacteria interactions in the fish gut. Nevertheless, we did not observe negative effects on fish health or growth, and further studies should be directed in understanding if these changes are beneficial or not for the fish health with an additional focus on the host immune response.
Collapse
Affiliation(s)
- Valentina Laura Donati
- Unit for Fish and Shellfish Diseases, National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lone Madsen
- Unit for Fish and Shellfish Diseases, National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mathias Middelboe
- Marine Biological Section, University of Copenhagen, Helsingør, Denmark
| | - Mikael Lenz Strube
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Inger Dalsgaard
- Unit for Fish and Shellfish Diseases, National Institute of Aquatic Resources, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
90
|
Li J, Wang L, Ding J, Cheng Y, Diao L, Li L, Zhang Y, Yin T. Multiomics Studies Investigating Recurrent Pregnancy Loss: An Effective Tool for Mechanism Exploration. Front Immunol 2022; 13:826198. [PMID: 35572542 PMCID: PMC9094436 DOI: 10.3389/fimmu.2022.826198] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/31/2022] [Indexed: 12/18/2022] Open
Abstract
Patients with recurrent pregnancy loss (RPL) account for approximately 1%-5% of women aiming to achieve childbirth. Although studies have shown that RPL is associated with failure of endometrial decidualization, placental dysfunction, and immune microenvironment disorder at the maternal-fetal interface, the exact pathogenesis remains unknown. With the development of high-throughput technology, more studies have focused on the genomics, transcriptomics, proteomics and metabolomics of RPL, and new gene mutations and new biomarkers of RPL have been discovered, providing an opportunity to explore the pathogenesis of RPL from different biological processes. Bioinformatics analyses of these differentially expressed genes, proteins and metabolites also reflect the biological pathways involved in RPL, laying a foundation for further research. In this review, we summarize the findings of omics studies investigating decidual tissue, villous tissue and blood from patients with RPL and identify some possible limitations of current studies.
Collapse
Affiliation(s)
- Jianan Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Linlin Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Jinli Ding
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanxiang Cheng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Longfei Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Yan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tailang Yin
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
91
|
Manoharan A, Sambandam R, Ballambattu VB. Genetics of atrial fibrillation-an update of recent findings. Mol Biol Rep 2022; 49:8121-8129. [PMID: 35587846 DOI: 10.1007/s11033-022-07420-2] [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: 10/22/2021] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia and a major risk factor for stroke, heart failure, and premature death. AF has a strong genetic predisposition. This review highlights the recent findings on the genetics of AF from genome-wide association studies (GWAS) and high-throughput sequencing studies. The consensus from GWAS implies that AF is both polygenic and pleiotropic in nature. With the advent of whole-genome sequencing and whole-exome sequencing, rare variants associated with AF pathogenesis have been identified. The recent studies have contributed towards better understanding of AF pathogenesis.
Collapse
Affiliation(s)
- Aarthi Manoharan
- Multi-Disciplinary Center for Biomedical Research, Vinayaka Mission's Research Foundation, Aarupadai Veedu Medical College and Hospital, Puducherry, 607402, India
| | - Ravikumar Sambandam
- Multi-Disciplinary Center for Biomedical Research, Vinayaka Mission's Research Foundation, Aarupadai Veedu Medical College and Hospital, Puducherry, 607402, India.
| | - Vishnu Bhat Ballambattu
- Multi-Disciplinary Center for Biomedical Research, Vinayaka Mission's Research Foundation, Aarupadai Veedu Medical College and Hospital, Puducherry, 607402, India
| |
Collapse
|
92
|
Liang Y, Xie J, Zhang Q, Wang X, Gou S, Lin L, Chen T, Ge W, Zhuang Z, Lian M, Chen F, Li N, Ouyang Z, Lai C, Liu X, Li L, Ye Y, Wu H, Wang K, Lai L. AGBE: a dual deaminase-mediated base editor by fusing CGBE with ABE for creating a saturated mutant population with multiple editing patterns. Nucleic Acids Res 2022; 50:5384-5399. [PMID: 35544322 PMCID: PMC9122597 DOI: 10.1093/nar/gkac353] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 12/26/2022] Open
Abstract
Establishing saturated mutagenesis in a specific gene through gene editing is an efficient approach for identifying the relationships between mutations and the corresponding phenotypes. CRISPR/Cas9-based sgRNA library screening often creates indel mutations with multiple nucleotides. Single base editors and dual deaminase-mediated base editors can achieve only one and two types of base substitutions, respectively. A new glycosylase base editor (CGBE) system, in which the uracil glycosylase inhibitor (UGI) is replaced with uracil-DNA glycosylase (UNG), was recently reported to efficiently induce multiple base conversions, including C-to-G, C-to-T and C-to-A. In this study, we fused a CGBE with ABE to develop a new type of dual deaminase-mediated base editing system, the AGBE system, that can simultaneously introduce 4 types of base conversions (C-to-G, C-to-T, C-to-A and A-to-G) as well as indels with a single sgRNA in mammalian cells. AGBEs can be used to establish saturated mutant populations for verification of the functions and consequences of multiple gene mutation patterns, including single-nucleotide variants (SNVs) and indels, through high-throughput screening.
Collapse
Affiliation(s)
- Yanhui Liang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingke Xie
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Quanjun Zhang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Xiaomin Wang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Shixue Gou
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Lihui Lin
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Tao Chen
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Weikai Ge
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Zhenpeng Zhuang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Lian
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Fangbing Chen
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Nan Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Zhen Ouyang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Chengdan Lai
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Xiaoyi Liu
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinghua Ye
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Han Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Kepin Wang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Liangxue Lai
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China.,Sanya institute of Swine resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China.,Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
93
|
Luan G, Wang M, Yuan J, Bu X, Song J, Wang C, Zhang L. Regulatory network identified by pulmonary transcriptome and proteome profiling reveals extensive change of tumor-related genes in microRNA-21 knockout mice. J Cancer Res Clin Oncol 2022; 148:1919-1929. [PMID: 35511299 DOI: 10.1007/s00432-022-03967-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/21/2022] [Indexed: 01/08/2023]
Abstract
PURPOSE MicroRNA-21 (miR-21) is a well-known oncomiR and plays key roles in regulating various biological processes related to pulmonary diseases, especially lung carcinoma. The regulatory roles and downstream targets of miR-21 remain far from well understood. We aimed to identify miR-21-gene regulatory network in lung tissue. METHODS Transcriptome and proteome analyses were performed on lung tissues from miR-21 knockout (KO) mice and their wildtype (WT) littermates. Differentially expressed genes (DEGs) and proteins (DEPs) between miR-21KO and WT were analyzed, and correlation analysis was performed between transcriptional and translational level. DEPs were used for prediction of miR-21 target genes and construction of co-expression network. RESULTS Comparing with WT mice, 820 DEGs and 623 DEPs were identified in lung tissues of miR-21KO mice. Upregulated DEGs and DEPs were both significantly enriched in pathways of metabolism of xenobiotics by cytochrome P450, drug metabolism, and chemical carcinogenesis. Of the 31 molecules commonly identified in DEGs and DEPs, 9 upregulated genes were tumor suppressor genes while 8 downregulated genes were oncogenes, and 12 genes showed closely positive correlation between mRNA and protein expression. Real-time PCR validation results were consistent with the omics data. Among the upregulated DEPs in miR-21KO mice, 21 genes were predicted as miR-21 targets. The miR-21 regulatory network was constructed by target genes and their highly co-expressed proteins, which identified the miR-21 target Itih4 as a hub gene. CONCLUSION MiR-21-gene regulatory network was constructed in mouse lung tissue. MiR-21KO resulted in extensive upregulation of tumor suppressor genes and downregulation of oncogenes.
Collapse
Affiliation(s)
- Ge Luan
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, 100005, China
| | - Ming Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, 100005, China
| | - Jing Yuan
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, 100005, China
| | - Xiangting Bu
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, 100005, China
| | - Jing Song
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, 100005, China
| | - Chengshuo Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China.
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, 100005, China.
| | - Luo Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730, China.
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, 100005, China.
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, No. 1, DongJiaoMinXiang, DongCheng District, Beijing, 100730, China.
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
94
|
Noor F, Tahir ul Qamar M, Ashfaq UA, Albutti A, Alwashmi ASS, Aljasir MA. Network Pharmacology Approach for Medicinal Plants: Review and Assessment. Pharmaceuticals (Basel) 2022; 15:572. [PMID: 35631398 PMCID: PMC9143318 DOI: 10.3390/ph15050572] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/13/2022] Open
Abstract
Natural products have played a critical role in medicine due to their ability to bind and modulate cellular targets involved in disease. Medicinal plants hold a variety of bioactive scaffolds for the treatment of multiple disorders. The less adverse effects, affordability, and easy accessibility highlight their potential in traditional remedies. Identifying pharmacological targets from active ingredients of medicinal plants has become a hot topic for biomedical research to generate innovative therapies. By developing an unprecedented opportunity for the systematic investigation of traditional medicines, network pharmacology is evolving as a systematic paradigm and becoming a frontier research field of drug discovery and development. The advancement of network pharmacology has opened up new avenues for understanding the complex bioactive components found in various medicinal plants. This study is attributed to a comprehensive summary of network pharmacology based on current research, highlighting various active ingredients, related techniques/tools/databases, and drug discovery and development applications. Moreover, this study would serve as a protocol for discovering novel compounds to explore the full range of biological potential of traditionally used plants. We have attempted to cover this vast topic in the review form. We hope it will serve as a significant pioneer for researchers working with medicinal plants by employing network pharmacology approaches.
Collapse
Affiliation(s)
- Fatima Noor
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000, Pakistan; (F.N.); (M.T.u.Q.)
| | - Muhammad Tahir ul Qamar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000, Pakistan; (F.N.); (M.T.u.Q.)
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000, Pakistan; (F.N.); (M.T.u.Q.)
| | - Aqel Albutti
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Ameen S. S. Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (A.S.S.A.); (M.A.A.)
| | - Mohammad Abdullah Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (A.S.S.A.); (M.A.A.)
| |
Collapse
|
95
|
Oliveira NC, Phelan L, Labate CA, Cônsoli FL. Non-targeted metabolomics reveals differences in the gut metabolic profile of the fall armyworm strains when feeding different food sources. JOURNAL OF INSECT PHYSIOLOGY 2022; 139:104400. [PMID: 35598778 DOI: 10.1016/j.jinsphys.2022.104400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Spodoptera frugiperda (fall armyworm - FAW) is an important polyphagous agricultural pest feeding on nearly 350 host plants. FAW is undergoing incipient speciation with two well-characterized host-adapted strains, the "corn" (CS) and "rice" (RS) strains, which are morphologically identical but carry several genes under positive selection for host adaptation. We used non-targeted metabolomics based on gas chromatography/mass spectrometry to identify differences in metabolite profiles of the larval gut of CS and RS feeding on different host plants. Larvae were fed on artificial diet, maize, rice, or cotton leaves from eclosion to the sixth instar, when they had their midgut dissected for analysis. This study revealed that the midgut metabolome of FAW varied due to larval diet and differed between the FAW host-adapted strains. Additionally, we identified several candidate metabolites that may be involved in the adaptation of CS and RS to their host plants. Our findings provide clues toward the gut metabolic activities of the FAW strains.
Collapse
Affiliation(s)
- Nathalia C Oliveira
- Insect Interactions Laboratory, Department of Entomology and Acarology, Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Larry Phelan
- Department of Entomology, OARDC, The Ohio State University, Wooster, OH, United States
| | - Carlos A Labate
- Multi-User Proteomics, Metabolomics and Lipidomics Laboratory, Department of Genetics, Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Fernando L Cônsoli
- Insect Interactions Laboratory, Department of Entomology and Acarology, Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil.
| |
Collapse
|
96
|
Zhang X, Zhou Z, Xu H, Liu CT. Integrative clustering methods for multi-omics data. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL STATISTICS 2022; 14. [PMID: 35573155 PMCID: PMC9097984 DOI: 10.1002/wics.1553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Integrative analysis of multi-omics data has drawn much attention from the scientific community due to the technological advancements which have generated various omics data. Leveraging these multi-omics data potentially provides a more comprehensive view of the disease mechanism or biological processes. Integrative multi-omics clustering is an unsupervised integrative method specifically used to find coherent groups of samples or features by utilizing information across multi-omics data. It aims to better stratify diseases and to suggest biological mechanisms and potential targeted therapies for the diseases. However, applying integrative multi-omics clustering is both statistically and computationally challenging due to various reasons such as high dimensionality and heterogeneity. In this review, we summarized integrative multi-omics clustering methods into three general categories: concatenated clustering, clustering of clusters, and interactive clustering based on when and how the multi-omics data are processed for clustering. We further classified the methods into different approaches under each category based on the main statistical strategy used during clustering. In addition, we have provided recommended practices tailored to four real-life scenarios to help researchers to strategize their selection in integrative multi-omics clustering methods for their future studies.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Zhenwei Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
97
|
Yu CT, Chao BN, Barajas R, Haznadar M, Maruvada P, Nicastro HL, Ross SA, Verma M, Rogers S, Zanetti KA. An evaluation of the National Institutes of Health grants portfolio: identifying opportunities and challenges for multi-omics research that leverage metabolomics data. Metabolomics 2022; 18:29. [PMID: 35488937 PMCID: PMC9056487 DOI: 10.1007/s11306-022-01878-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Through the systematic large-scale profiling of metabolites, metabolomics provides a tool for biomarker discovery and improving disease monitoring, diagnosis, prognosis, and treatment response, as well as for delineating disease mechanisms and etiology. As a downstream product of the genome and epigenome, transcriptome, and proteome activity, the metabolome can be considered as being the most proximal correlate to the phenotype. Integration of metabolomics data with other -omics data in multi-omics analyses has the potential to advance understanding of human disease development and treatment. AIM OF REVIEW To understand the current funding and potential research opportunities for when metabolomics is used in human multi-omics studies, we cross-sectionally evaluated National Institutes of Health (NIH)-funded grants to examine the use of metabolomics data when collected with at least one other -omics data type. First, we aimed to determine what types of multi-omics studies included metabolomics data collection. Then, we looked at those multi-omics studies to examine how often grants employed an integrative analysis approach using metabolomics data. KEY SCIENTIFIC CONCEPTS OF REVIEW We observed that the majority of NIH-funded multi-omics studies that include metabolomics data performed integration, but to a limited extent, with integration primarily incorporating only one other -omics data type. Some opportunities to improve data integration may include increasing confidence in metabolite identification, as well as addressing variability between -omics approach requirements and -omics data incompatibility.
Collapse
Affiliation(s)
- Catherine T Yu
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Brittany N Chao
- Office of Workforce Planning and Development, National Cancer Institute, Rockville, MD, USA
| | - Rolando Barajas
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Majda Haznadar
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Rockville, MD, USA
| | - Padma Maruvada
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Holly L Nicastro
- Office of Nutrition Research, National Institutes of Health, Bethesda, MD, USA
| | - Sharon A Ross
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | - Mukesh Verma
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Scott Rogers
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Krista A Zanetti
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
98
|
Liu Z, Xu J, Que S, Geng L, Zhou L, Mardinoglu A, Zheng S. Recent Progress and Future Direction for the Application of Multiomics Data in Clinical Liver Transplantation. J Clin Transl Hepatol 2022; 10:363-373. [PMID: 35528975 PMCID: PMC9039708 DOI: 10.14218/jcth.2021.00219] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/14/2021] [Accepted: 10/07/2021] [Indexed: 12/04/2022] Open
Abstract
Omics data address key issues in liver transplantation (LT) as the most effective therapeutic means for end-stage liver disease. The purpose of this study was to review the current application and future direction for omics in LT. We reviewed the use of multiomics to elucidate the pathogenesis leading to LT and prognostication. Future directions with respect to the use of omics in LT are also described based on perspectives of surgeons with experience in omics. Significant molecules were identified and summarized based on omics, with a focus on post-transplant liver fibrosis, early allograft dysfunction, tumor recurrence, and graft failure. We emphasized the importance omics for clinicians who perform LTs and prioritized the directions that should be established. We also outlined the ideal workflow for omics in LT. In step with advances in technology, the quality of omics data can be guaranteed using an improved algorithm at a lower price. Concerns should be addressed on the translational value of omics for better therapeutic effects in patients undergoing LT.
Collapse
Affiliation(s)
- Zhengtao Liu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuping Que
- DingXiang Clinics, Hangzhou, Zhejiang, China
| | - Lei Geng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lin Zhou
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
- Correspondence to: Adil Mardinoglu, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden. ORCID: https://orcid.org/0000-0002-4254-6090. Tel: +46-31-772-3140, Fax: +46-31-772-3801, E-mail: ; Shusen Zheng, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China. ORCID: https://orcid.org/0000-0003-1459-8261. Tel/Fax: +86-571-87236570, E-mail:
| | - Shusen Zheng
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Correspondence to: Adil Mardinoglu, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden. ORCID: https://orcid.org/0000-0002-4254-6090. Tel: +46-31-772-3140, Fax: +46-31-772-3801, E-mail: ; Shusen Zheng, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China. ORCID: https://orcid.org/0000-0003-1459-8261. Tel/Fax: +86-571-87236570, E-mail:
| |
Collapse
|
99
|
Vahabi N, Michailidis G. Unsupervised Multi-Omics Data Integration Methods: A Comprehensive Review. Front Genet 2022; 13:854752. [PMID: 35391796 PMCID: PMC8981526 DOI: 10.3389/fgene.2022.854752] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/28/2022] [Indexed: 12/26/2022] Open
Abstract
Through the developments of Omics technologies and dissemination of large-scale datasets, such as those from The Cancer Genome Atlas, Alzheimer’s Disease Neuroimaging Initiative, and Genotype-Tissue Expression, it is becoming increasingly possible to study complex biological processes and disease mechanisms more holistically. However, to obtain a comprehensive view of these complex systems, it is crucial to integrate data across various Omics modalities, and also leverage external knowledge available in biological databases. This review aims to provide an overview of multi-Omics data integration methods with different statistical approaches, focusing on unsupervised learning tasks, including disease onset prediction, biomarker discovery, disease subtyping, module discovery, and network/pathway analysis. We also briefly review feature selection methods, multi-Omics data sets, and resources/tools that constitute critical components for carrying out the integration.
Collapse
Affiliation(s)
- Nasim Vahabi
- Informatics Institute, University of Florida, Gainesville, FL, United States
| | - George Michailidis
- Informatics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
100
|
Graupera I, Isus L, Coll M, Pose E, Díaz A, Vallverdú J, Rubio-Tomás T, Martínez-Sánchez C, Huelin P, Llopis M, Solé C, Fondevila C, Lozano JJ, Sancho-Bru P, Ginès P, Aloy P. Molecular characterization of chronic liver disease dynamics: from liver fibrosis to acute-on-chronic liver failure. JHEP Rep 2022; 4:100482. [PMID: 35540106 PMCID: PMC9079303 DOI: 10.1016/j.jhepr.2022.100482] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/22/2022] [Accepted: 03/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background & Aims The molecular mechanisms driving the progression from early-chronic liver disease (CLD) to cirrhosis and, finally, acute-on-chronic liver failure (ACLF) are largely unknown. Our aim was to develop a protein network-based approach to investigate molecular pathways driving progression from early-CLD to ACLF. Methods Transcriptome analysis was performed on liver biopsies from patients at different liver disease stages, including fibrosis, compensated cirrhosis, decompensated cirrhosis and ACLF, and control healthy livers. We created 9 liver-specific disease-related protein-protein interaction networks capturing key pathophysiological processes potentially related to CLD. We used these networks as a framework and performed gene set-enrichment analysis (GSEA) to identify dynamic gene profiles of disease progression. Results Principal component analyses revealed that samples clustered according to the disease stage. GSEA of the defined processes showed an upregulation of inflammation, fibrosis and apoptosis networks throughout disease progression. Interestingly, we did not find significant gene expression differences between compensated and decompensated cirrhosis, while ACLF showed acute expression changes in all the defined liver disease-related networks. The analyses of disease progression patterns identified ascending and descending expression profiles associated with ACLF onset. Functional analyses showed that ascending profiles were associated with inflammation, fibrosis, apoptosis, senescence and carcinogenesis networks, while descending profiles were mainly related to oxidative stress and genetic factors. We confirmed by qPCR the upregulation of genes of the ascending profile and validated our findings in an independent patient cohort. Conclusion ACLF is characterized by a specific hepatic gene expression pattern related to inflammation, fibrosis, apoptosis, senescence and carcinogenesis. Moreover, the observed profile is significantly different from that of compensated and decompensated cirrhosis, supporting the hypothesis that ACLF should be considered a distinct entity. Lay summary By using transjugular biopsies obtained from patients at different stages of chronic liver disease, we unveil the molecular pathogenic mechanisms implicated in the progression of chronic liver disease to cirrhosis and acute-on-chronic liver failure. The most relevant finding in this study is that patients with acute-on-chronic liver failure present a specific hepatic gene expression pattern distinct from that of patients at earlier disease stages. This gene expression pattern is mostly related to inflammation, fibrosis, angiogenesis, and senescence and apoptosis pathways in the liver. We unveiled the molecular pathogenic mechanisms implicated in the progression of chronic liver disease to cirrhosis and ACLF. ACLF presents a specific hepatic gene expression pattern distinct from that of patients at earlier disease stages. Gene expression pattern of ACLF is mostly related to inflammation, fibrosis, angiogenesis, senescence and apoptosis pathways in the liver.
Collapse
|