1
|
Zhang H, Zhang W, Zheng X, Li Y. scRDEN: single-cell dynamic gene rank differential expression network and robust trajectory inference. Sci Rep 2025; 15:16963. [PMID: 40374885 DOI: 10.1038/s41598-025-01969-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 05/09/2025] [Indexed: 05/18/2025] Open
Abstract
The remarkable advancement of single-cell RNA sequencing (scRNA-seq) technology has empowered researchers to probe gene expression at the single-cell level with unprecedented precision. To gain a profound understanding of the heterogeneity inherent in cell fate determination, a central challenge lies in the comprehensive analysis of the dynamic regulatory alterations that underlie transcriptional differences and the accurate inference of the differentiation trajectory. Here, we propose the method scRDEN, a robust framework that infers important cell sub-populations and differential expression networks of multiple genes along the differentiation directions of each branch by converting the unstable gene expression values in cells into relatively stable gene-gene interactions (global features) and extracting the order of differential expression (network features), and further integrating the expression features of different dimension reduction methods. When applied to five published scRNA-seq datasets from human and mouse cell differentiation, scRDEN not only successfully captures the stable cell subpopulations with potential marker genes, measures the transcriptional differences of gene pairs to identify the rank differential expression network along the differentiation direction of each branch. In addition, in multiple gene rank differential expression networks, the rank expression directly related to transcription factors/marker genes shows a significant strengthening and weakening trend along with their expression changes, and the distribution of diversity and cluster coefficient show a non-monotonic change trend, including the cases of increasing first and then decreasing or decreasing first and then increasing. This may correspond to the mechanism of cells gradually differentiating into stable functions. It is particularly noteworthy that scRDEN method yielded exceptional results when applied to the large-scale, multi-branched, double-batch mouse dentate gyrus data. This outstanding performance provides novel and valuable insights into large-scale, multi-batch trajectory inference and the study of transcriptional mechanism regulation during the processes of differentiation and development.
Collapse
Affiliation(s)
- Han Zhang
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China
| | - Wei Zhang
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China
| | - Xiaoying Zheng
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China.
| | - Yuanyuan Li
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China.
| |
Collapse
|
2
|
Li Z, Yang M, Ma X, Zhou C, Meng F, Shi P, Hu P, Liang B, Jiang Q, Zhang L, Liu X, Shi T, Lai C, Zhang T, Song H. A Functionally Conserved yet Dynamically Evolving Toolkit Underpinning Molluscan Biomineralization: Insights From Shell and Radula. Integr Zool 2025. [PMID: 40248912 DOI: 10.1111/1749-4877.12978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/19/2025]
Abstract
The molluscan shell and radula constitute pivotal molluscan innovations, each characterized by distinct functions and diverse forms, regulated by the highly specific biomineralization regulatory networks. Despite their paramount importance, the conserved components and adaptive evolutionary processes governing these regulatory networks remain unresolved. To address this knowledge gap, we advocate for the integration of data from less-explored lineages, such as Scaphopoda, as an essential step. This study presents the inaugural comprehensive transcriptome analysis of Pictodentalium vernedei, a representative species of Scaphopoda distinguished by a unique and evolutionarily conserved shell morphology and radula structure. Furthermore, comparative transcriptome/genome analyses are employed to unravel the conservatism and evolutionary innovation of the involved biomineralization regulatory elements. Our findings underscore the central role of secretomes in governing biomineralization processes, and we identified a fundamental set of 26 domains within molluscan secretomes, forming an essential functional protein domain repertoire necessary for the transformation of inorganic ions into biomineralized structures. This core biomineralization toolkit has undergone independent expansion and lineage-specific recruitment, giving rise to novel, modular domain architectures. This may be essential for the functional specialization and morphological diversification of shell and radula structures. These evolutionary processes are driven by the independent co-option of ancient genes and the emergence of novel de novo genes. This comprehensive investigation not only contributes insights into the evolution of molluscan biomineralization structures but also establishes avenues for further scholarly exploration.
Collapse
Affiliation(s)
- Zhuoqing Li
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meijie Yang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinghao Ma
- Shouguang City Marine Fishery Development Center, Weifang, China
| | - Cong Zhou
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fanyu Meng
- Lianyungang City Ganyu District Zhewang Town Agriculture Rural and Social Undertakings Bureau, Lianyungang, China
| | - Pu Shi
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pengpeng Hu
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bin Liang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingtian Jiang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Lili Zhang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Xiaoyan Liu
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Qingdao Agricultural University, Qingdao, China
| | - Tingyu Shi
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Changping Lai
- Lianyungang Blue Carbon Marine Technology Co., Lianyungang, China
| | - Tao Zhang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Song
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Kyurkchiyan S, Petkova V, Stancheva G, Stancheva I, Dimitrov S, Dobriyanova V, Popova D, Kaneva R, M Popov T. Co-expression of miRNA players in advanced laryngeal carcinoma - Insights into the roles of miR-93-5p, miR-145-5p, and miR-210-3p. BIOMOLECULES & BIOMEDICINE 2025; 25:1052-1062. [PMID: 39412136 PMCID: PMC11984375 DOI: 10.17305/bb.2024.10947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 04/04/2025]
Abstract
Advanced laryngeal squamous cell carcinoma (LSCC) is the second most prevalent type of head and neck squamous cell carcinoma (HNSCC). Identifying microRNAs (miRNAs) related to key regulatory molecules or mechanisms could offer an alternative approach to developing new treatment strategies. The aim of our study is to evaluate significant correlations among deregulated miRNAs in advanced laryngeal carcinoma and to analyze, in silico, their strength of association, targets, and the most deregulated pathways. Several miRNAs demonstrated promising co-expression results, specifically miR-93-5p, miR-145-5p, and miR-210-3p. Their expressions were explored and further validated in a large set of in vivo advanced LSCC samples, which were subsequently used for bioinformatics and enrichment analyses. Our results highlight the significant roles of miR-93-5p, miR-145-5p, and miR-210-3p in regulating major pathways linked to the cell cycle via epithelial-to-mesenchymal transition (EMT), PI3K/Akt signaling, hypoxia, metabolism, apoptosis, angiogenesis, and metastasis. The associations between the expressions of these miRNAs and patients' clinical features could be central to the progression of advanced LSCC. Overall, our study provides important insights into the co-expression and regulatory networks of miR-93-5p, miR-145-5p, and miR-210-3p in advanced laryngeal carcinoma, underscoring their potential as therapeutic targets or biomarkers for this aggressive cancer. Further research is needed to elucidate the specific mechanisms through which these miRNAs contribute to the pathogenesis and progression of laryngeal carcinoma.
Collapse
Affiliation(s)
- Silva Kyurkchiyan
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Veronika Petkova
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Gergana Stancheva
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Iglika Stancheva
- Department of Ear, Nose and Throat Diseases, University Multiprofile Hospital for Active Treatment “Tsaritsa Yoanna - ISUL,” Medical University of Sofia, Sofia, Bulgaria
| | - Stoyan Dimitrov
- Department of Ear, Nose and Throat Diseases, University Multiprofile Hospital for Active Treatment “Tsaritsa Yoanna - ISUL,” Medical University of Sofia, Sofia, Bulgaria
| | - Venera Dobriyanova
- Department of Ear, Nose and Throat Diseases, University Multiprofile Hospital for Active Treatment “Tsaritsa Yoanna - ISUL,” Medical University of Sofia, Sofia, Bulgaria
| | - Diana Popova
- Department of Ear, Nose and Throat Diseases, University Multiprofile Hospital for Active Treatment “Tsaritsa Yoanna - ISUL,” Medical University of Sofia, Sofia, Bulgaria
| | - Radka Kaneva
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Todor M Popov
- Department of Ear, Nose and Throat Diseases, University Multiprofile Hospital for Active Treatment “Tsaritsa Yoanna - ISUL,” Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
4
|
Cheng S, Wei Y, Zhou Y, Xu Z, Wright DN, Liu J, Peng Y. Deciphering genomic codes using advanced natural language processing techniques: a scoping review. J Am Med Inform Assoc 2025; 32:761-772. [PMID: 39998912 PMCID: PMC12005631 DOI: 10.1093/jamia/ocaf029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVES The vast and complex nature of human genomic sequencing data presents challenges for effective analysis. This review aims to investigate the application of natural language processing (NLP) techniques, particularly large language models (LLMs) and transformer architectures, in deciphering genomic codes, focusing on tokenization, transformer models, and regulatory annotation prediction. The goal of this review is to assess data and model accessibility in the most recent literature, gaining a better understanding of the existing capabilities and constraints of these tools in processing genomic sequencing data. MATERIALS AND METHODS Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, our scoping review was conducted across PubMed, Medline, Scopus, Web of Science, Embase, and ACM Digital Library. Studies were included if they focused on NLP methodologies applied to genomic sequencing data analysis, without restrictions on publication date or article type. RESULTS A total of 26 studies published between 2021 and April 2024 were selected for review. The review highlights that tokenization and transformer models enhance the processing and understanding of genomic data, with applications in predicting regulatory annotations like transcription-factor binding sites and chromatin accessibility. DISCUSSION The application of NLP and LLMs to genomic sequencing data interpretation is a promising field that can help streamline the processing of large-scale genomic data while also providing a better understanding of its complex structures. It has the potential to drive advancements in personalized medicine by offering more efficient and scalable solutions for genomic analysis. Further research is also needed to discuss and overcome current limitations, enhancing model transparency and applicability. CONCLUSION This review highlights the growing role of NLP, particularly LLMs, in genomic sequencing data analysis. While these models improve data processing and regulatory annotation prediction, challenges remain in accessibility and interpretability. Further research is needed to refine their application in genomics.
Collapse
Affiliation(s)
- Shuyan Cheng
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, United States
| | - Yishu Wei
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, United States
| | - Yiliang Zhou
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, United States
| | - Zihan Xu
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, United States
| | - Drew N Wright
- Samuel J. Wood Library & C.V. Starr Biomedical Information Center, Weill Cornell Medicine, New York, NY 10065, United States
| | - Jinze Liu
- School of Public Health, Virginia Commonwealth University, Richmond, VA 23219, United States
| | - Yifan Peng
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, United States
| |
Collapse
|
5
|
Bekhbat M, Block AM, Dickinson SY, Tharp GK, Bosinger SE, Felger JC. Neurotransmitter and metabolic effects of interferon-alpha in association with decreased striatal dopamine in a non-human primate model of cytokine-Induced depression. Brain Behav Immun 2025; 125:308-318. [PMID: 39826580 PMCID: PMC11903159 DOI: 10.1016/j.bbi.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Inflammatory stimuli administered to humans and laboratory animals affect mesolimbic and nigrostriatal dopaminergic pathways in association with impaired motivation and motor activity. Alterations in dopaminergic corticostriatal reward and motor circuits have also been observed in depressed patients with increased peripheral inflammatory markers. The effects of peripheral inflammation on dopaminergic pathways and associated neurobiologic mechanisms and consequences have been difficult to measure in patients. Postmortem tissue (n = 11) from an established, translationally-relevant non-human primate model of cytokine-induced depressive behavior involving chronic interferon-alpha (IFN-a) administration was utilized herein to explore the molecular mechanisms of peripheral cytokine effects on striatal dopamine. Dopamine (but not serotonin or norepinephrine) was decreased in the nucleus accumbens (NAcc) and putamen of IFN-a-treated animals (p < 0.05). IFN-a had no effect on number of striatal neurons or dopamine terminal density, suggesting no overt neurodegenerative changes. RNA sequencing examined in the caudate, putamen, substantia nigra, and prefrontal cortical subregions revealed that while IFN-a nominally up-regulated limited numbers of genes enriching inflammatory signaling pathways in all regions, robust, whole genome-significant effects of IFN-a were observed specifically in putamen. Genes upregulated in the putamen primarily enriched synaptic signaling, glutamate receptor signaling, and inflammatory/metabolic pathways downstream of IFN-a, including MAPK and PI3K/AKT cascades. Conversely, gene transcripts reduced by IFN-a enriched oxidative phosphorylation (OXPHOS), protein translation, and pathways regulated by dopamine receptors. Unsupervised clustering identified a gene co-expression module in the putamen that was associated with both IFN-a treatment and low dopamine levels, which enriched similar inflammatory, metabolic, and synaptic signaling pathways. IFN-a-induced reductions in dopamine further correlated with genes related to excitotoxic glutamate, kynurenine, and altered dopamine receptor signaling (r = 0.78-97, p < 0.05). These findings provide insight into the immunologic mechanisms and neurobiological consequences of peripheral inflammation effects on dopamine, which may inform novel treatment strategies targeting inflammatory, metabolic or neurotransmitter systems in depressed patients with high inflammation.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Andrew M Block
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06030, USA
| | - Sarah Y Dickinson
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gregory K Tharp
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven E Bosinger
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
6
|
Yang H, Zhang C, Kim W, Shi M, Kiliclioglu M, Bayram C, Bolar I, Tozlu ÖÖ, Baba C, Yuksel N, Yildirim S, Iqbal S, Sebhaoui J, Hacımuftuoglu A, Uhlen M, Boren J, Turkez H, Mardinoglu A. Multi-tissue network analysis reveals the effect of JNK inhibition on dietary sucrose-induced metabolic dysfunction in rats. eLife 2025; 13:RP98427. [PMID: 39932177 PMCID: PMC11813226 DOI: 10.7554/elife.98427] [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: 02/13/2025] Open
Abstract
Excessive consumption of sucrose, in the form of sugar-sweetened beverages, has been implicated in the pathogenesis of metabolic dysfunction-associated fatty liver disease (MAFLD) and other related metabolic syndromes. The c-Jun N-terminal kinase (JNK) pathway plays a crucial role in response to dietary stressors, and it was demonstrated that the inhibition of the JNK pathway could potentially be used in the treatment of MAFLD. However, the intricate mechanisms underlying these interventions remain incompletely understood given their multifaceted effects across multiple tissues. In this study, we challenged rats with sucrose-sweetened water and investigated the potential effects of JNK inhibition by employing network analysis based on the transcriptome profiling obtained from hepatic and extrahepatic tissues, including visceral white adipose tissue, skeletal muscle, and brain. Our data demonstrate that JNK inhibition by JNK-IN-5A effectively reduces the circulating triglyceride accumulation and inflammation in rats subjected to sucrose consumption. Coexpression analysis and genome-scale metabolic modeling reveal that sucrose overconsumption primarily induces transcriptional dysfunction related to fatty acid and oxidative metabolism in the liver and adipose tissues, which are largely rectified after JNK inhibition at a clinically relevant dose. Skeletal muscle exhibited minimal transcriptional changes to sucrose overconsumption but underwent substantial metabolic adaptation following the JNK inhibition. Overall, our data provides novel insights into the molecular basis by which JNK inhibition exerts its metabolic effect in the metabolically active tissues. Furthermore, our findings underpin the critical role of extrahepatic metabolism in the development of diet-induced steatosis, offering valuable guidance for future studies focused on JNK-targeting for effective treatment of MAFLD.
Collapse
Affiliation(s)
- Hong Yang
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Woonghee Kim
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Mengnan Shi
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Metin Kiliclioglu
- Department of Pathology, Veterinary Faculty, Atatürk UniversityErzurumTurkiye
| | - Cemil Bayram
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk UniversityErzurumTurkiye
| | - Ismail Bolar
- Department of Pathology, Veterinary Faculty, Atatürk UniversityErzurumTurkiye
| | - Özlem Özdemir Tozlu
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical UniversityErzurumTurkiye
| | - Cem Baba
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical UniversityErzurumTurkiye
| | - Nursena Yuksel
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical UniversityErzurumTurkiye
| | - Serkan Yildirim
- Department of Pathology, Veterinary Faculty, Atatürk UniversityErzurumTurkiye
- Department of Pathology, Faculty of Veterinary Medicine, Kyrgyz-Türkish Manas ÜniversityBishkekKyrgyzstan
| | - Shazia Iqbal
- Trustlife Labs Drug Research and Development CenterIstanbulTurkiye
| | - Jihad Sebhaoui
- Trustlife Labs Drug Research and Development CenterIstanbulTurkiye
| | - Ahmet Hacımuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk UniversityErzurumTurkiye
| | - Matthias Uhlen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University HospitalGothenburgSweden
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk UniversityErzurumTurkiye
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College LondonLondonUnited Kingdom
| |
Collapse
|
7
|
Zhang X, Liu Q. A graph neural network approach for hierarchical mapping of breast cancer protein communities. BMC Bioinformatics 2025; 26:23. [PMID: 39838298 PMCID: PMC11749236 DOI: 10.1186/s12859-024-06015-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Comprehensively mapping the hierarchical structure of breast cancer protein communities and identifying potential biomarkers from them is a promising way for breast cancer research. Existing approaches are subjective and fail to take information from protein sequences into consideration. Deep learning can automatically learn features from protein sequences and protein-protein interactions for hierarchical clustering. RESULTS Using a large amount of publicly available proteomics data, we created a hierarchical tree for breast cancer protein communities using a novel hierarchical graph neural network, with the supervision of gene ontology terms and assistance of a pre-trained deep contextual language model. Then, a group-lasso algorithm was applied to identify protein communities that are under both mutation burden and survival burden, undergo significant alterations when targeted by specific drug molecules, and show cancer-dependent perturbations. The resulting hierarchical map of protein communities shows how gene-level mutations and survival information converge on protein communities at different scales. Internal validity of the model was established through the convergence on BRCA2 as a breast cancer hotspot. Further overlaps with breast cancer cell dependencies revealed SUPT6H and RAD21, along with their respective protein systems, HOST:37 and HOST:861, as potential biomarkers. Using gene-level perturbation data of the HOST:37 and HOST:861 gene sets, three FDA-approved drugs with high therapeutic value were selected as potential treatments to be further evaluated. These drugs include mercaptopurine, pioglitazone, and colchicine. CONCLUSION The proposed graph neural network approach to analyzing breast cancer protein communities in a hierarchical structure provides a novel perspective on breast cancer prognosis and treatment. By targeting entire gene sets, we were able to evaluate the prognostic and therapeutic value of genes (or gene sets) at different levels, from gene-level to system-level biology. Cancer-specific gene dependencies provide additional context for pinpointing cancer-related systems and drug-induced alterations can highlight potential therapeutic targets. These identified protein communities, in conjunction with other protein communities under strong mutation and survival burdens, can potentially be used as clinical biomarkers for breast cancer.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Applied Computer Science, University of Winnipeg, Winnipeg, MB, R3B 2E9, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E 0W2, Canada
| | - Qian Liu
- Department of Applied Computer Science, University of Winnipeg, Winnipeg, MB, R3B 2E9, Canada.
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E 0W2, Canada.
| |
Collapse
|
8
|
Kastendiek N, Coletti R, Gross T, Lopes MB. Exploring glioma heterogeneity through omics networks: from gene network discovery to causal insights and patient stratification. BioData Min 2024; 17:56. [PMID: 39696678 DOI: 10.1186/s13040-024-00411-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Gliomas are primary malignant brain tumors with a typically poor prognosis, exhibiting significant heterogeneity across different cancer types. Each glioma type possesses distinct molecular characteristics determining patient prognosis and therapeutic options. This study aims to explore the molecular complexity of gliomas at the transcriptome level, employing a comprehensive approach grounded in network discovery. The graphical lasso method was used to estimate a gene co-expression network for each glioma type from a transcriptomics dataset. Causality was subsequently inferred from correlation networks by estimating the Jacobian matrix. The networks were then analyzed for gene importance using centrality measures and modularity detection, leading to the selection of genes that might play an important role in the disease. To explore the pathways and biological functions these genes are involved in, KEGG and Gene Ontology (GO) enrichment analyses on the disclosed gene sets were performed, highlighting the significance of the genes selected across several relevent pathways and GO terms. Spectral clustering based on patient similarity networks was applied to stratify patients into groups with similar molecular characteristics and to assess whether the resulting clusters align with the diagnosed glioma type. The results presented highlight the ability of the proposed methodology to uncover relevant genes associated with glioma intertumoral heterogeneity. Further investigation might encompass biological validation of the putative biomarkers disclosed.
Collapse
Affiliation(s)
- Nina Kastendiek
- Institute for Chemistry and Biology of the Marine Environment, University of Oldenburg, Oldenburg, 26129, Germany
| | - Roberta Coletti
- Center for Mathematics and Applications (NOVA Math), NOVA School of Science and Technology (NOVA FCT), Caparica, 2829-516, Portugal
| | - Thilo Gross
- Institute for Chemistry and Biology of the Marine Environment, University of Oldenburg, Oldenburg, 26129, Germany
- Helmholtz Institute for Functional Marine Biodiversity (HIFMB), Oldenburg, 26129, Germany
- Alfred Wegener Institute, Helmholtz Center for Polar and Marine Research, Bremerhaven, 27570, Germany
| | - Marta B Lopes
- Center for Mathematics and Applications (NOVA Math), NOVA School of Science and Technology (NOVA FCT), Caparica, 2829-516, Portugal.
- UNIDEMI, Department of Mechanical and Industrial Engineering, NOVA School of Science and Technology (NOVA FCT), Caparica, 2829-516, Portugal.
| |
Collapse
|
9
|
Feng X, Wu W, Liu F. AH-6809 mediated regulation of lung adenocarcinoma metastasis through NLRP7 and prognostic analysis of key metastasis-related genes. Front Pharmacol 2024; 15:1486265. [PMID: 39697539 PMCID: PMC11652142 DOI: 10.3389/fphar.2024.1486265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 09/30/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction Lung adenocarcinoma (LUAD) has become one of the leading causes of cancer-related deaths globally, with metastasis representing the most lethal stage of the disease. Despite significant advances in diagnostic and therapeutic strategies for LUAD, the mechanisms enabling cancer cells to breach the blood-brain barrier remain poorly understood. While genomic profiling has shed light on the nature of primary tumors, the genetic drivers and clinical relevance of LUAD metastasis are still largely unexplored. Objectives This study aims to investigate the genomic differences between brain-metastatic and non-brain-metastatic LUAD, identify potential prognostic biomarkers, and evaluate the efficacy of AH-6809 in modulating key molecular pathways involved in LUAD metastasis, with a focus on post-translational modifications (PTMs). Methods Genomic analyses were performed using data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) between brain-metastatic and non-metastatic LUAD samples were identified. Key gene modules were determined using Weighted Gene Co-expression Network Analysis (WGCNA), and their prognostic significance was assessed through Kaplan-Meier analysis. Cellular experiments, including CCK8 and qRT-PCR assays, were conducted to evaluate the anti-cancer effects of AH-6809 in LUAD cells. Apoptosis and inflammatory marker expression were assessed using immunofluorescence. Results Genomic analysis differentiated brain-metastatic from non-brain-metastatic LUAD and identified NLRP7, FIBCD1, and ELF5 as prognostic markers. AH-6809 significantly suppressed LUAD cell proliferation, promoted apoptosis, and modulated epithelial-mesenchymal transition (EMT) markers. These effects were reversed upon NLRP7 knockdown, highlighting its role in metastasis. Literature analysis further supported AH-6809's tumor-suppressive activity, particularly in NLRP7 knockdown cells, where it inhibited cell growth and facilitated apoptosis. AH-6809 was also found to affect SUMO1-mediated PTMs and downregulate EMT markers, including VIM and CDH2. NLRP7 knockdown partially reversed these effects. Immunofluorescence revealed enhanced apoptosis and inflammation in lung cancer cells, especially in NLRP7 knockdown cells treated with AH-6809. The regulatory mechanisms involve SUMO1-mediated post-translational modifications and NQO1. Further studies are required to elucidate the molecular mechanisms and assess the clinical potential of these findings. Conclusion These findings demonstrate the critical role of NLRP7 and associated genes in LUAD metastasis and suggest that AH-6809 holds promise as a potential therapeutic agent for brain-metastatic LUAD.
Collapse
Affiliation(s)
- Xu Feng
- Department of Neurointerventional, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Wei Wu
- Department of Acupuncture, Jin Zhou Hospital of Traditional Chinese Medicine, Jinzhou, China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou MedicalUniversity, Jinzhou, China
| |
Collapse
|
10
|
Long S, Xia Y, Liang L, Yang Y, Xie H, Wang X. PyNetCor: a high-performance Python package for large-scale correlation analysis. NAR Genom Bioinform 2024; 6:lqae177. [PMID: 39703431 PMCID: PMC11655297 DOI: 10.1093/nargab/lqae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
The development of multi-omics technologies has generated an abundance of biological datasets, providing valuable resources for investigating potential relationships within complex biological systems. However, most correlation analysis tools face computational challenges when dealing with these high-dimensional datasets containing millions of features. Here, we introduce pyNetCor, a fast and scalable tool for constructing correlation networks on large-scale and high-dimensional data. PyNetCor features optimized algorithms for both full correlation coefficient matrix computation and top-k correlation search, outperforming other tools in the field in terms of runtime and memory consumption. It utilizes a linear interpolation strategy to rapidly estimate P-values and achieve false discovery rate control, demonstrating a speedup of over 110 times compared to existing methods. Overall, pyNetCor supports large-scale correlation analysis, a crucial foundational step for various bioinformatics workflows, and can be easily integrated into downstream applications to accelerate the process of extracting biological insights from data.
Collapse
Affiliation(s)
- Shibin Long
- Department of Data Science, 01Life Institute, Shenzhen 518000, China
| | - Yan Xia
- Department of Data Science, 01Life Institute, Shenzhen 518000, China
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Lifeng Liang
- Department of Data Science, 01Life Institute, Shenzhen 518000, China
| | - Ying Yang
- Department of Data Science, 01Life Institute, Shenzhen 518000, China
| | - Hailiang Xie
- Department of Data Science, 01Life Institute, Shenzhen 518000, China
| | - Xiaokai Wang
- Department of Data Science, 01Life Institute, Shenzhen 518000, China
| |
Collapse
|
11
|
Cheng S, Wei Y, Zhou Y, Xu Z, Wright DN, Liu J, Peng Y. Deciphering genomic codes using advanced NLP techniques: a scoping review. ARXIV 2024:arXiv:2411.16084v1. [PMID: 39650606 PMCID: PMC11623714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Objectives The vast and complex nature of human genomic sequencing data presents challenges for effective analysis. This review aims to investigate the application of Natural Language Processing (NLP) techniques, particularly Large Language Models (LLMs) and transformer architectures, in deciphering genomic codes, focusing on tokenization, transformer models, and regulatory annotation prediction. This review aims to assess data and model accessibility in the most recent literature, gaining a better understanding of the existing capabilities and constraints of these tools in processing genomic sequencing data. Methods Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, our scoping review was conducted across PubMed, Medline, Scopus, Web of Science, Embase, and ACM Digital Library. Studies were included if they focused on NLP methodologies applied to genomic sequencing data analysis, without restrictions on publication date or article type. Results A total of 26 studies published between 2021 and April 2024 were selected for review. The review highlights that tokenization and transformer models enhance the processing and understanding of genomic data, with applications in predicting regulatory annotations like transcription-factor binding sites and chromatin accessibility. Discussion The application of NLP and LLMs to genomic sequencing data interpretation is a promising field that can help streamline the processing of large-scale genomic data while providing a better understanding of its complex structures. It can potentially drive advancements in personalized medicine by offering more efficient and scalable solutions for genomic analysis. Further research is needed to discuss and overcome limitations, enhancing model transparency and applicability.
Collapse
Affiliation(s)
- Shuyan Cheng
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065
| | - Yishu Wei
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065
| | - Yiliang Zhou
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065
| | - Zihan Xu
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065
| | - Drew N Wright
- Samuel J. Wood Library & C.V. Starr Biomedical Information Center, Weill Cornell Medicine, New York, NY 10065
| | - Jinze Liu
- School of Public Health, Virginia Commonwealth University, Richmond, VA 23219
| | - Yifan Peng
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065
| |
Collapse
|
12
|
Defilippo A, Giorgi FM, Veltri P, Guzzi PH. Understanding complex systems through differential causal networks. Sci Rep 2024; 14:27431. [PMID: 39521851 PMCID: PMC11550418 DOI: 10.1038/s41598-024-78606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
In the evolving landscape of data science and computational biology, Causal Networks (CNs) have emerged as a robust framework for modelling causal relationships among elements of complex systems derived from experimental data. CNs can efficiently model causal relationships emerging in a single system while comparing multiple systems, allowing to understand rewiring in different cells, tissues, and physiological states with a deeper perspective. Despite the existence of network models, namely differential networks, that have been used to compare coexpression and correlation structures, causality needs to be introduced in differential analysis to robustly provide direction to the edges of such networks, in order to better understand the flows of information, and also to better intervene in their functioning, for example for agricultural or pharmacological purposes. Resolved to reach this ambitious goal, we introduce Differential Causal Networks (DCNs), a novel framework that represents differences between two existing CNs. A DCN is obtained from experimental data by comparing two CNs, and it is a power tool for highlighting differences in causal relations. After a careful definition and design of DCNs, we test our algorithm to model possible differential causal relationships between genes responsible for the onset of type 2 diabetes mellitus-related pathologies considering patients' sex at the tissue level. DCNs allowed us to shed light on causal differences between sexes across nine tissues. We also compare differences among three possible definitions of DCNs to highlight similarities and differences of biological importance. Code, Data and Supplementary Information are available at https://github.com/hguzzi/DifferentialCausalNetworks .
Collapse
Affiliation(s)
- Annamaria Defilippo
- Department of Surgical and Medical Sciences, Magna Graecia University of Catanzaro, Catanzaro, 88100, Italy
| | | | - Pierangelo Veltri
- Department of Computer Science, Modelling and Electronics (DIMES), University of Calabria, Rende, 87036, Italy
| | - Pietro Hiram Guzzi
- Department of Surgical and Medical Sciences, Magna Graecia University of Catanzaro, Catanzaro, 88100, Italy.
| |
Collapse
|
13
|
Ge Y, Li T, Feng X, Wu M, Liu H. Structured feature ranking for genomic marker identification accommodating multiple types of networks. Biometrics 2024; 80:ujae158. [PMID: 39745855 DOI: 10.1093/biomtc/ujae158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 10/04/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
Numerous statistical methods have been developed to search for genomic markers associated with the development, progression, and response to treatment of complex diseases. Among them, feature ranking plays a vital role due to its intuitive formulation and computational efficiency. However, most of the existing methods are based on the marginal importance of molecular predictors and share the limitation that the dependence (network) structures among predictors are not well accommodated, where a disease phenotype usually reflects various biological processes that interact in a complex network. In this paper, we propose a structured feature ranking method for identifying genomic markers, where such network structures are effectively accommodated using Laplacian regularization. The proposed method innovatively investigates multiple network scenarios, where the networks can be known a priori and data-dependently estimated. In addition, we rigorously explore the noise and uncertainty in the networks and control their impacts with proper selection of tuning parameters. These characteristics make the proposed method enjoy especially broad applicability. Theoretical result of our proposal is rigorously established. Compared to the original marginal measure, the proposed network structured measure can achieve sure screening properties with a faster convergence rate under mild conditions. Extensive simulations and analysis of The Cancer Genome Atlas melanoma data demonstrate the improvement of finite sample performance and practical usefulness of the proposed method.
Collapse
Affiliation(s)
- Yeheng Ge
- School of Statistics and Data Science, Shanghai University of Finance and Economics, 777 Guoding Road, Shanghai 200433, China
| | - Tao Li
- School of Statistics and Data Science, Shanghai University of Finance and Economics, 777 Guoding Road, Shanghai 200433, China
| | - Xingdong Feng
- School of Statistics and Data Science, Shanghai University of Finance and Economics, 777 Guoding Road, Shanghai 200433, China
| | - Mengyun Wu
- School of Statistics and Data Science, Shanghai University of Finance and Economics, 777 Guoding Road, Shanghai 200433, China
| | - Hailong Liu
- Department of Urology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| |
Collapse
|
14
|
Lv H, Wang A, Ling J, Li Y, He Y, Luo H, Ye H, Yao W, Su S, He W. Multi-organ transcriptomics analysis of a slowly growing fish rock carp (Procypris rabaudi) reveals insights into mechanism of growth rate regulation. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101337. [PMID: 39423654 DOI: 10.1016/j.cbd.2024.101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
To explore the patterns of differentially expressed genes (DEGs) associated with different growth rates in rock carp (Procypris rabaudi), transcriptome sequencing was performed on the muscle, liver, and brain tissues of rock carp. Subsequently, bioinformatics analysis was conducted, and 2129, 1380, and 415 DEGs were identified in the muscle, liver, and brain tissues, respectively. GO enrichment and KEGG pathway analysis revealed that genes related to appetite regulation, protein degradation and digestion, lipid transport and metabolisms, and glycolysis/gluconeogenesis were upregulated in individuals with slower growth rates. Differential expression analysis identified 21 genes associated with feeding and metabolism across three tissues, including mc4r, npy, and npry in brain tissue; fatp, fabp, pparα, and apo in liver tissue; and prss, ctrl, and cela in muscle tissue. All these genes were upregulated in the slow-growing fish. Furthermore, weighted gene co-expression network analyses, including three modules (yellow, turquoise, and brown), significantly associated with growth. A network map that included these three modules enabled the identification of a series of hub genes, including rp13a, ube2o, h6pd, etc. These genes may be key candidate genes regulating the growth of rock carp. This study contributes to a deeper understanding of the growth control mechanism in rock carp and offers a scientific basis for efficient breeding and species improvement.
Collapse
Affiliation(s)
- Hongsen Lv
- College of Fisheries, Southwest University, Chongqing 400715, China
| | - Anxiang Wang
- College of Fisheries, Southwest University, Chongqing 400715, China
| | - Jingning Ling
- College of Fisheries, Southwest University, Chongqing 400715, China
| | - Yixiao Li
- College of Fisheries, Southwest University, Chongqing 400715, China
| | - Yuanfa He
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China
| | - Hui Luo
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China
| | - Hua Ye
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China
| | - Weizhi Yao
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China
| | - Shengqi Su
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China
| | - Wenping He
- College of Fisheries, Southwest University, Chongqing 400715, China; Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China.
| |
Collapse
|
15
|
Li Y, Du Y, Wang M, Ai D. CSER: a gene regulatory network construction method based on causal strength and ensemble regression. Front Genet 2024; 15:1481787. [PMID: 39371416 PMCID: PMC11449711 DOI: 10.3389/fgene.2024.1481787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction Gene regulatory networks (GRNs) reveal the intricate interactions between and among genes, and understanding these interactions is essential for revealing the molecular mechanisms of cancer. However, existing algorithms for constructing GRNs may confuse regulatory relationships and complicate the determination of network directionality. Methods We propose a new method to construct GRNs based on causal strength and ensemble regression (CSER) to overcome these issues. CSER uses conditional mutual inclusive information to quantify the causal associations between genes, eliminating indirect regulation and marginal genes. It considers linear and nonlinear features and uses ensemble regression to infer the direction and interaction (activation or regression) from regulatory to target genes. Results Compared to traditional algorithms, CSER can construct directed networks and infer the type of regulation, thus demonstrating higher accuracy on simulated datasets. Here, using real gene expression data, we applied CSER to construct a colorectal cancer GRN and successfully identified several key regulatory genes closely related to colorectal cancer (CRC), including ADAMDEC1, CLDN8, and GNA11. Discussion Importantly, by integrating immune cell and microbial data, we revealed the complex interactions between the CRC gene regulatory network and the tumor microenvironment, providing additional new biomarkers and therapeutic targets for the early diagnosis and prognosis of CRC.
Collapse
Affiliation(s)
| | | | | | - Dongmei Ai
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| |
Collapse
|
16
|
Garcia Lopez A, Schäuble S, Sae-Ong T, Seelbinder B, Bauer M, Giamarellos-Bourboulis EJ, Singer M, Lukaszewski R, Panagiotou G. Risk assessment with gene expression markers in sepsis development. Cell Rep Med 2024; 5:101712. [PMID: 39232497 PMCID: PMC11528229 DOI: 10.1016/j.xcrm.2024.101712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/21/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
Infection is a commonplace, usually self-limiting, condition but can lead to sepsis, a severe life-threatening dysregulated host response. We investigate the individual phenotypic predisposition to developing uncomplicated infection or sepsis in a large cohort of non-infected patients undergoing major elective surgery. Whole-blood RNA sequencing analysis was performed on preoperative samples from 267 patients. These patients developed postoperative infection with (n = 77) or without (n = 49) sepsis, developed non-infectious systemic inflammatory response (n = 31), or had an uncomplicated postoperative course (n = 110). Machine learning classification models built on preoperative transcriptomic signatures predict postoperative outcomes including sepsis with an area under the curve of up to 0.910 (mean 0.855) and sensitivity/specificity up to 0.767/0.804 (mean 0.746/0.769). Our models, confirmed by quantitative reverse-transcription PCR (RT-qPCR), potentially offer a risk prediction tool for the development of postoperative sepsis with implications for patient management. They identify an individual predisposition to developing sepsis that warrants further exploration to better understand the underlying pathophysiology.
Collapse
Affiliation(s)
- Albert Garcia Lopez
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Tongta Sae-Ong
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Bastian Seelbinder
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | | | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Roman Lukaszewski
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany; Friedrich Schiller University, Institute of Microbiology, Faculty of Biological Sciences, 07743 Jena, Germany; Department of Medicine, University of Hong Kong, Hong Kong SAR, China; Jena University Hospital, Friedrich Schiller University Jena, 07743 Jena, Germany.
| |
Collapse
|
17
|
Xu C, Xia P, Li J, Lewis KB, Ciombor KK, Wang L, Smith JJ, Beauchamp RD, Chen XS. Discovery and validation of a 10-gene predictive signature for response to adjuvant chemotherapy in stage II and III colon cancer. Cell Rep Med 2024; 5:101661. [PMID: 39059386 PMCID: PMC11384724 DOI: 10.1016/j.xcrm.2024.101661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/30/2023] [Accepted: 07/02/2024] [Indexed: 07/28/2024]
Abstract
Identifying patients with stage II and III colon cancer who will benefit from 5-fluorouracil (5-FU)-based adjuvant chemotherapy is crucial for the advancement of personalized cancer therapy. We employ a semi-supervised machine learning approach to analyze a large dataset with 933 stage II and III colon cancer samples. Our analysis leverages gene regulatory networks to discover an 18-gene prognostic signature and to explore a 10-gene signature that potentially predicts chemotherapy benefits. The 10-gene signature demonstrates strong prognostic power and shows promising potential to predict chemotherapy benefits. We establish a robust clinical assay on the NanoString nCounter platform, validated in a retrospective formalin-fixed paraffin-embedded (FFPE) cohort, which represents an important step toward clinical application. Our study lays the groundwork for improving adjuvant chemotherapy and potentially expanding into immunotherapy decision-making in colon cancer. Future prospective studies are needed to validate and establish the clinical utility of the 10-gene signature in clinical settings.
Collapse
Affiliation(s)
- Chaohan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Peng Xia
- School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Jie Li
- Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, China
| | - Keeli B Lewis
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kristen K Ciombor
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lily Wang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - R Daniel Beauchamp
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - X Steven Chen
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
18
|
Wang Y, Hui X, Wang H, Chen H. Boosting Volatile fatty acids (VFAs) production in fermentation microorganisms through genes expression control: Unraveling the role of iron homeostasis transcription factors. WATER RESEARCH 2024; 259:121850. [PMID: 38851109 DOI: 10.1016/j.watres.2024.121850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/06/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Iron (Fe0, Fe (II), and Fe (III)) has been previously documented to upregulate the expression of key genes, enhancing the production of volatile fatty acids (VFAs) to achieve waste/wastewater resource recovery. However, the precise mechanism by why iron influences gene expression remains unclear. This study applied iron-assisted fermentation systems to explore the behind enhancing mechanism by constructing regulon networks among genes, microbes, and transcription factors. In iron-conditioned systems, a significant enhancement in VFAs production and upregulation of genes expression (1.19-3.92 folds) related to organic conversion and the electron transfer chain was observed. Besides, gene co-expression network and Procrustes analysis identified ten hub transcription factors (e.g., arsR, crp, iscR, perR) and their major contributors (genus) (e.g., Paludibacter, Acinetobacter, Tolumonas). Further analysis suggested that most of hub transcription factors were implicated in iron homeostasis regulation, which speculated that the induced iron homeostasis transcription factors probably effectively regulated the expression of genes encoding enzymes involving in VFAs production and electron transfer of functional microbes, in the case of Paludibacter, Acinetobacter, and Tolumonas while regulating the iron homeostasis, resulting in the efficient production of VFAs in iron-conditioned systems. This study might contribute to an enhanced understanding of the underlying genetic mechanisms by why iron influences gene expression regulation of microbes, which also provides a genetic theoretical basis for improving system VFAs production and resource recovery.
Collapse
Affiliation(s)
- Yanqiong Wang
- National Engineering Research Center for Urban Pollution Control, State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Xuesong Hui
- National Engineering Research Center for Urban Pollution Control, State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Hongwu Wang
- National Engineering Research Center for Urban Pollution Control, State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China; Key Laboratory of Urban Water Supply, Water Saving and Water Environment Governance in the Yangtze River Delta of Ministry of Water Resources, Tongji University, Shanghai 200092, China.
| | - Hongbin Chen
- National Engineering Research Center for Urban Pollution Control, State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| |
Collapse
|
19
|
Fazli M, Bertram R, Striegel DA. Multi-layer Bundling as a New Approach for Determining Multi-scale Correlations Within a High-Dimensional Dataset. Bull Math Biol 2024; 86:105. [PMID: 38995438 PMCID: PMC11245441 DOI: 10.1007/s11538-024-01335-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
The growing complexity of biological data has spurred the development of innovative computational techniques to extract meaningful information and uncover hidden patterns within vast datasets. Biological networks, such as gene regulatory networks and protein-protein interaction networks, hold critical insights into biological features' connections and functions. Integrating and analyzing high-dimensional data, particularly in gene expression studies, stands prominent among the challenges in deciphering these networks. Clustering methods play a crucial role in addressing these challenges, with spectral clustering emerging as a potent unsupervised technique considering intrinsic geometric structures. However, spectral clustering's user-defined cluster number can lead to inconsistent and sometimes orthogonal clustering regimes. We propose the Multi-layer Bundling (MLB) method to address this limitation, combining multiple prominent clustering regimes to offer a comprehensive data view. We call the outcome clusters "bundles". This approach refines clustering outcomes, unravels hierarchical organization, and identifies bridge elements mediating communication between network components. By layering clustering results, MLB provides a global-to-local view of biological feature clusters enabling insights into intricate biological systems. Furthermore, the method enhances bundle network predictions by integrating the bundle co-cluster matrix with the affinity matrix. The versatility of MLB extends beyond biological networks, making it applicable to various domains where understanding complex relationships and patterns is needed.
Collapse
Affiliation(s)
- Mehran Fazli
- Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Dr, Bethesda, MD, 20817, USA.
| | - Richard Bertram
- Department of Mathematics, Florida State University, 1017 Academic Way, Tallahassee, FL, 32306, USA
- Programs in Neuroscience and Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA
| | - Deborah A Striegel
- Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Dr, Bethesda, MD, 20817, USA
| |
Collapse
|
20
|
Rich A, Acar O, Carvunis AR. Massively integrated coexpression analysis reveals transcriptional regulation, evolution and cellular implications of the yeast noncanonical translatome. Genome Biol 2024; 25:183. [PMID: 38978079 PMCID: PMC11232214 DOI: 10.1186/s13059-024-03287-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 05/20/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Recent studies uncovered pervasive transcription and translation of thousands of noncanonical open reading frames (nORFs) outside of annotated genes. The contribution of nORFs to cellular phenotypes is difficult to infer using conventional approaches because nORFs tend to be short, of recent de novo origins, and lowly expressed. Here we develop a dedicated coexpression analysis framework that accounts for low expression to investigate the transcriptional regulation, evolution, and potential cellular roles of nORFs in Saccharomyces cerevisiae. RESULTS Our results reveal that nORFs tend to be preferentially coexpressed with genes involved in cellular transport or homeostasis but rarely with genes involved in RNA processing. Mechanistically, we discover that young de novo nORFs located downstream of conserved genes tend to leverage their neighbors' promoters through transcription readthrough, resulting in high coexpression and high expression levels. Transcriptional piggybacking also influences the coexpression profiles of young de novo nORFs located upstream of genes, but to a lesser extent and without detectable impact on expression levels. Transcriptional piggybacking influences, but does not determine, the transcription profiles of de novo nORFs emerging nearby genes. About 40% of nORFs are not strongly coexpressed with any gene but are transcriptionally regulated nonetheless and tend to form entirely new transcription modules. We offer a web browser interface ( https://carvunislab.csb.pitt.edu/shiny/coexpression/ ) to efficiently query, visualize, and download our coexpression inferences. CONCLUSIONS Our results suggest that nORF transcription is highly regulated. Our coexpression dataset serves as an unprecedented resource for unraveling how nORFs integrate into cellular networks, contribute to cellular phenotypes, and evolve.
Collapse
Affiliation(s)
- April Rich
- Joint Carnegie Mellon University-University of Pittsburgh, University of Pittsburgh Computational Biology PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Center for Evolutionary Biology and Medicine (CEBaM), University of Pittsburgh, Pittsburgh, PA, USA
| | - Omer Acar
- Joint Carnegie Mellon University-University of Pittsburgh, University of Pittsburgh Computational Biology PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Center for Evolutionary Biology and Medicine (CEBaM), University of Pittsburgh, Pittsburgh, PA, USA
| | - Anne-Ruxandra Carvunis
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Pittsburgh Center for Evolutionary Biology and Medicine (CEBaM), University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Li Y, Wang Q, Zheng X, Xu B, Hu W, Zhang J, Kong X, Zhou Y, Huang T, Zhou Y. ScHGSC-IGDC: Identifying genes with differential correlations of high-grade serous ovarian cancer based on single-cell RNA sequencing analysis. Heliyon 2024; 10:e32909. [PMID: 38975079 PMCID: PMC11226911 DOI: 10.1016/j.heliyon.2024.e32909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
Due to the high heterogeneity of ovarian cancer (OC), it occupies the main cause of cancer-related death among women. As the most aggressive and frequent subtype of OC, high-grade serous cancer (HGSC) represents around 70 % of all patients. With the booming progress of single-cell RNA sequencing (scRNA-seq), unique and subtle changes among different cell states have been identified including novel risk genes and pathways. Here, our present study aims to identify differentially correlated core genes between normal and tumor status through HGSC scRNA-seq data analysis. R package high-dimension Weighted Gene Co-expression Network Analysis (hdWGCNA) was implemented for building gene interaction networks based on HGSC scRNA-seq data. DiffCorr was integrated for identifying differentially correlated genes between tumor and their adjacent normal counterparts. Software Cytoscape was implemented for constructing and visualizing biological networks. Real-time qPCR (RT-qPCR) was utilized to confirm expression pattern of new genes. We introduced ScHGSC-IGDC (Identifying Genes with Differential Correlations of HGSC based on scRNA-seq analysis), an in silico framework for identifying core genes in the development of HGSC. We detected thirty-four modules in the network. Scores of new genes with opposite correlations with others such as NDUFS5, TMSB4X, SERPINE2 and ITPR2 were identified. Further survival and literature validation emphasized their great values in the HGSC management. Meanwhile, RT-qPCR verified expression pattern of NDUFS5, TMSB4X, SERPINE2 and ITPR2 in human OC cell lines and tissues. Our research offered novel perspectives on the gene modulatory mechanisms from single cell resolution, guiding network based algorithms in cancer etiology field.
Collapse
Affiliation(s)
- Yuanqi Li
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, 213003, China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003, China
| | - Qi Wang
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, 213003, China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003, China
| | - Xiao Zheng
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, 213003, China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003, China
| | - Bin Xu
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, 213003, China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003, China
| | - Wenwei Hu
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, 213003, China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003, China
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Xiangyin Kong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi Zhou
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, 213003, China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003, China
| | - Tao Huang
- Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - You Zhou
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, 213003, China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003, China
| |
Collapse
|
22
|
Velazquez-Caldelas TE, Zamora-Fuentes JM, Hernandez-Lemus E. Coordinated inflammation and immune response transcriptional regulation in breast cancer molecular subtypes. Front Immunol 2024; 15:1357726. [PMID: 38983850 PMCID: PMC11231215 DOI: 10.3389/fimmu.2024.1357726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/03/2024] [Indexed: 07/11/2024] Open
Abstract
Breast cancer, characterized by its complexity and diversity, presents significant challenges in understanding its underlying biology. In this study, we employed gene co-expression network analysis to investigate the gene composition and functional patterns in breast cancer subtypes and normal breast tissue. Our objective was to elucidate the detailed immunological features distinguishing these tumors at the transcriptional level and to explore their implications for diagnosis and treatment. The analysis identified nine distinct gene module clusters, each representing unique transcriptional signatures within breast cancer subtypes and normal tissue. Interestingly, while some clusters exhibited high similarity in gene composition between normal tissue and certain subtypes, others showed lower similarity and shared traits. These clusters provided insights into the immune responses within breast cancer subtypes, revealing diverse immunological functions, including innate and adaptive immune responses. Our findings contribute to a deeper understanding of the molecular mechanisms underlying breast cancer subtypes and highlight their unique characteristics. The immunological signatures identified in this study hold potential implications for diagnostic and therapeutic strategies. Additionally, the network-based approach introduced herein presents a valuable framework for understanding the complexities of other diseases and elucidating their underlying biology.
Collapse
Affiliation(s)
| | | | - Enrique Hernandez-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
23
|
Wang S, Stroup EK, Wang TY, Yang R, Ji Z. Comparative analyses of gene networks mediating cancer metastatic potentials across lineage types. Brief Bioinform 2024; 25:bbae357. [PMID: 39041189 PMCID: PMC11262869 DOI: 10.1093/bib/bbae357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/21/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024] Open
Abstract
Studies have identified genes and molecular pathways regulating cancer metastasis. However, it remains largely unknown whether metastatic potentials of cancer cells from different lineage types are driven by the same or different gene networks. Here, we aim to address this question through integrative analyses of 493 human cancer cells' transcriptomic profiles and their metastatic potentials in vivo. Using an unsupervised approach and considering both gene coexpression and protein-protein interaction networks, we identify different gene networks associated with various biological pathways (i.e. inflammation, cell cycle, and RNA translation), the expression of which are correlated with metastatic potentials across subsets of lineage types. By developing a regularized random forest regression model, we show that the combination of the gene module features expressed in the native cancer cells can predict their metastatic potentials with an overall Pearson correlation coefficient of 0.90. By analyzing transcriptomic profile data from cancer patients, we show that these networks are conserved in vivo and contribute to cancer aggressiveness. The intrinsic expression levels of these networks are correlated with drug sensitivity. Altogether, our study provides novel comparative insights into cancer cells' intrinsic gene networks mediating metastatic potentials across different lineage types, and our results can potentially be useful for designing personalized treatments for metastatic cancers.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60628, United States
| | - Emily K Stroup
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Chicago, IL 60611, United States
| | - Ting-You Wang
- Department of Urology, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Chicago, IL 60611, United States
| | - Rendong Yang
- Department of Urology, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Chicago, IL 60611, United States
| | - Zhe Ji
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60628, United States
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Chicago, IL 60611, United States
| |
Collapse
|
24
|
Preetam S, Duhita Mondal D, Mukerjee N, Naser SS, Tabish TA, Thorat N. Revolutionizing Cancer Treatment: The Promising Horizon of Zein Nanosystems. ACS Biomater Sci Eng 2024; 10:1946-1965. [PMID: 38427627 PMCID: PMC11005017 DOI: 10.1021/acsbiomaterials.3c01540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/03/2024]
Abstract
Various nanomaterials have recently become fascinating tools in cancer diagnostic applications because of their multifunctional and inherent molecular characteristics that support efficient diagnosis and image-guided therapy. Zein nanoparticles are a protein derived from maize. It belongs to the class of prolamins possessing a spherical structure with conformational properties similar to those of conventional globular proteins like ribonuclease and insulin. Zein nanoparticles have gained massive interest over the past couple of years owing to their natural hydrophilicity, ease of functionalization, biodegradability, and biocompatibility, thereby improving oral bioavailability, nanoparticle targeting, and prolonged drug administration. Thus, zein nanoparticles are becoming a promising candidate for precision cancer drug delivery. This review highlights the clinical significance of applying zein nanosystems for cancer theragnostic─moreover, the role of zein nanosystems for cancer drug delivery, anticancer agents, and gene therapy. Finally, the difficulties and potential uses of these NPs in cancer treatment and detection are discussed. This review will pave the way for researchers to develop theranostic strategies for precision medicine utilizing zein nanosystems.
Collapse
Affiliation(s)
- Subham Preetam
- Department
of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, South Korea
| | - Deb Duhita Mondal
- Department
of Biotechnology, Heritage Institute of
Technology, Kolkata, West Bengal 700107, India
| | - Nobendu Mukerjee
- Centre
for Global Health Research, Saveetha Medical
College and Hospital, Chennai 602105, India
- Department
of Science and Engineering, Novel Global
Community and Educational Foundation, Hebasham 2770, NSW, Australia
| | | | - Tanveer A. Tabish
- Division
of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7BN, United Kingdom
| | - Nanasaheb Thorat
- Nuffield
Department of Women’s & Reproductive Health, Medical Science
Division, John Radcliffe Hospital University
of Oxford, Oxford, OX3 9DU, United Kingdom
- Department
of Physics, Bernal Institute and Limerick
Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick V94T9PX, Ireland
| |
Collapse
|
25
|
Liao Y, Li S, Ji G. Graphene oxide stimulated low-temperature denitrification activity of microbial communities in lake sediments by enhancing anabolism and inhibiting cellular respiration. CHEMOSPHERE 2024; 350:141090. [PMID: 38169199 DOI: 10.1016/j.chemosphere.2023.141090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024]
Abstract
Nitrate pollution in fresh water is becoming increasingly serious. In this study, the effects of temperature and graphene oxide materials on the potential functions of denitrification communities in lake sediments were investigated by metagenome. The addition of graphene oxide significantly affected the abundance of denitrification genes such as Nap, Nos, and enhanced the contribution of Pseudomonas, making low temperature and material addition conducive to the denitrification process. Module network implied that low temperature increased the centrality of denitrification in community functions. At low temperatures, graphene oxide enhanced community anabolism by stimulation organic carbon consumption and regulating the gene abundance in the citric acid cycle and the semi-phosphorylation Entner-Doudoroff, thus possibly stimulating extracellular polymeric substances (EPS) synthesis and secretion. In addition, graphene oxide may also regulate the transfer of reducing electrons from NADH to denitrifying enzymes by affecting the gene abundances of complex I and complex IV.
Collapse
Affiliation(s)
- Yinhao Liao
- Key Laboratory of Water and Sediment Sciences, Ministry of Education, Department of Environmental Engineering, Peking University, Beijing, 100871, China; Institute of Whole Process Consulting, Chongqing CISDI Engineering Consulting Co. Ltd., Chongqing, 400013, China
| | - Shengjie Li
- Key Laboratory of Water and Sediment Sciences, Ministry of Education, Department of Environmental Engineering, Peking University, Beijing, 100871, China
| | - Guodong Ji
- Key Laboratory of Water and Sediment Sciences, Ministry of Education, Department of Environmental Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
26
|
Muthamil S, Muthuramalingam P, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Shin H, Park JH. Unlocking Prognostic Genes and Multi-Targeted Therapeutic Bioactives from Herbal Medicines to Combat Cancer-Associated Cachexia: A Transcriptomics and Network Pharmacology Approach. Int J Mol Sci 2023; 25:156. [PMID: 38203330 PMCID: PMC10778733 DOI: 10.3390/ijms25010156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Cachexia is a devastating fat tissue and muscle wasting syndrome associated with every major chronic illness, including cancer, chronic obstructive pulmonary disease, kidney disease, AIDS, and heart failure. Despite two decades of intense research, cachexia remains under-recognized by oncologists. While numerous drug candidates have been proposed for cachexia treatment, none have achieved clinical success. Only a few drugs are approved by the FDA for cachexia therapy, but a very low success rate is observed among patients. Currently, the identification of drugs from herbal medicines is a frontier research area for many diseases. In this milieu, network pharmacology, transcriptomics, cheminformatics, and molecular docking approaches were used to identify potential bioactive compounds from herbal medicines for the treatment of cancer-related cachexia. The network pharmacology approach is used to select the 32 unique genes from 238 genes involved in cachexia-related pathways, which are targeted by 34 phytocompounds identified from 12 different herbal medicines used for the treatment of muscle wasting in many countries. Gene expression profiling and functional enrichment analysis are applied to decipher the role of unique genes in cancer-associated cachexia pathways. In addition, the pharmacological properties and molecular interactions of the phytocompounds were analyzed to find the target compounds for cachexia therapy. Altogether, combined omics and network pharmacology approaches were used in the current study to untangle the complex prognostic genes involved in cachexia and phytocompounds with anti-cachectic efficacy. However, further functional and experimental validations are required to confirm the efficacy of these phytocompounds as commercial drug candidates for cancer-associated cachexia.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea; (S.M.); (H.-Y.K.); (H.-J.J.); (J.-H.L.); (U.C.S.)
| | - Pandiyan Muthuramalingam
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52725, Republic of Korea; (P.M.); (H.S.)
| | - Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea; (S.M.); (H.-Y.K.); (H.-J.J.); (J.-H.L.); (U.C.S.)
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea; (S.M.); (H.-Y.K.); (H.-J.J.); (J.-H.L.); (U.C.S.)
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea; (S.M.); (H.-Y.K.); (H.-J.J.); (J.-H.L.); (U.C.S.)
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea; (S.M.); (H.-Y.K.); (H.-J.J.); (J.-H.L.); (U.C.S.)
| | - Younghoon Go
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea;
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea;
| | - Hyunsuk Shin
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52725, Republic of Korea; (P.M.); (H.S.)
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea; (S.M.); (H.-Y.K.); (H.-J.J.); (J.-H.L.); (U.C.S.)
- Korean Convergence Medicine Major, University of Science & Technology (UST), KIOM Campus, Daejeon 34054, Republic of Korea
| |
Collapse
|
27
|
Trujillo-Ortíz R, Espinal-Enríquez J, Hernández-Lemus E. The Role of Transcription Factors in the Loss of Inter-Chromosomal Co-Expression for Breast Cancer Subtypes. Int J Mol Sci 2023; 24:17564. [PMID: 38139393 PMCID: PMC10743684 DOI: 10.3390/ijms242417564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Breast cancer encompasses a diverse array of subtypes, each exhibiting distinct clinical characteristics and treatment responses. Unraveling the underlying regulatory mechanisms that govern gene expression patterns in these subtypes is essential for advancing our understanding of breast cancer biology. Gene co-expression networks (GCNs) help us identify groups of genes that work in coordination. Previous research has revealed a marked reduction in the interaction of genes located on different chromosomes within GCNs for breast cancer, as well as for lung, kidney, and hematopoietic cancers. However, the reasons behind why genes on the same chromosome often co-express remain unclear. In this study, we investigate the role of transcription factors in shaping gene co-expression networks within the four main breast cancer subtypes: Luminal A, Luminal B, HER2+, and Basal, along with normal breast tissue. We identify communities within each GCN and calculate the transcription factors that may regulate these communities, comparing the results across different phenotypes. Our findings indicate that, in general, regulatory behavior is to a large extent similar among breast cancer molecular subtypes and even in healthy networks. This suggests that transcription factor motif usage does not fully determine long-range co-expression patterns. Specific transcription factor motifs, such as CCGGAAG, appear frequently across all phenotypes, even involving multiple highly connected transcription factors. Additionally, certain transcription factors exhibit unique actions in specific subtypes but with limited influence. Our research demonstrates that the loss of inter-chromosomal co-expression is not solely attributable to transcription factor regulation. Although the exact mechanism responsible for this phenomenon remains elusive, this work contributes to a better understanding of gene expression regulatory programs in breast cancer.
Collapse
Affiliation(s)
- Rodrigo Trujillo-Ortíz
- Computational Genomics Division, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
| | - Jesús Espinal-Enríquez
- Computational Genomics Division, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City 01010, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City 01010, Mexico
| |
Collapse
|
28
|
Martins FB, Aono AH, Moraes ADCL, Ferreira RCU, Vilela MDM, Pessoa-Filho M, Rodrigues-Motta M, Simeão RM, de Souza AP. Genome-wide family prediction unveils molecular mechanisms underlying the regulation of agronomic traits in Urochloa ruziziensis. FRONTIERS IN PLANT SCIENCE 2023; 14:1303417. [PMID: 38148869 PMCID: PMC10749977 DOI: 10.3389/fpls.2023.1303417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/15/2023] [Indexed: 12/28/2023]
Abstract
Tropical forage grasses, particularly those belonging to the Urochloa genus, play a crucial role in cattle production and serve as the main food source for animals in tropical and subtropical regions. The majority of these species are apomictic and tetraploid, highlighting the significance of U. ruziziensis, a sexual diploid species that can be tetraploidized for use in interspecific crosses with apomictic species. As a means to support breeding programs, our study investigates the feasibility of genome-wide family prediction in U. ruziziensis families to predict agronomic traits. Fifty half-sibling families were assessed for green matter yield, dry matter yield, regrowth capacity, leaf dry matter, and stem dry matter across different clippings established in contrasting seasons with varying available water capacity. Genotyping was performed using a genotyping-by-sequencing approach based on DNA samples from family pools. In addition to conventional genomic prediction methods, machine learning and feature selection algorithms were employed to reduce the necessary number of markers for prediction and enhance predictive accuracy across phenotypes. To explore the regulation of agronomic traits, our study evaluated the significance of selected markers for prediction using a tree-based approach, potentially linking these regions to quantitative trait loci (QTLs). In a multiomic approach, genes from the species transcriptome were mapped and correlated to those markers. A gene coexpression network was modeled with gene expression estimates from a diverse set of U. ruziziensis genotypes, enabling a comprehensive investigation of molecular mechanisms associated with these regions. The heritabilities of the evaluated traits ranged from 0.44 to 0.92. A total of 28,106 filtered SNPs were used to predict phenotypic measurements, achieving a mean predictive ability of 0.762. By employing feature selection techniques, we could reduce the dimensionality of SNP datasets, revealing potential genotype-phenotype associations. The functional annotation of genes near these markers revealed associations with auxin transport and biosynthesis of lignin, flavonol, and folic acid. Further exploration with the gene coexpression network uncovered associations with DNA metabolism, stress response, and circadian rhythm. These genes and regions represent important targets for expanding our understanding of the metabolic regulation of agronomic traits and offer valuable insights applicable to species breeding. Our work represents an innovative contribution to molecular breeding techniques for tropical forages, presenting a viable marker-assisted breeding approach and identifying target regions for future molecular studies on these agronomic traits.
Collapse
Affiliation(s)
- Felipe Bitencourt Martins
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Alexandre Hild Aono
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Aline da Costa Lima Moraes
- Department of Plant Biology, Biology Institute, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | | | - Marco Pessoa-Filho
- Embrapa Cerrados, Brazilian Agricultural Research Corporation, Brasília, Brazil
| | | | - Rosangela Maria Simeão
- Embrapa Gado de Corte, Brazilian Agricultural Research Corporation, Campo Grande, Mato Grosso, Brazil
| | - Anete Pereira de Souza
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Plant Biology, Biology Institute, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
29
|
Gong J, Li Q. Comparative Transcriptome and WGCNA Analysis Reveal Molecular Responses to Salinity Change in Larvae of the Iwagaki Oyster Crassostrea Nippona. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1031-1042. [PMID: 37872465 DOI: 10.1007/s10126-023-10257-w] [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: 08/19/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
The Iwagaki oyster Crassostrea nippona is an important aquaculture species with significant potential for large-scale oyster farming. It is susceptible to the fluctuated salinity in the coastal area. In this study, we compared the transcriptome of Crassostrea nippona larvae under variant conditions with low-salinity stress (28, 20, 15, 10, and 5 practical salinity units (psu)) for 24 h. KEGG enrichment analysis of differentially expressed genes (DEGs) from pairwise comparisons identified several free amino acid metabolism pathway (taurine and hypotaurine, arginine and proline, glycine, and beta-alanine) contributing to the salinity change adaptation and activated "lysosome" and "apoptosis" pathway in response to the low-salinity stress (10 and 5 psu). Trend analysis revealed sustained upregulation of transmembrane transport-related genes (such as SLC family) and downregulation of ribosomal protein synthesis genes faced with decreasing salinities. In addition, 9 biomarkers in response to low-salinity stress were identified through weighted gene co-expression network analysis (WGCNA) and validated by qRT-PCR. Our transcriptome analysis provides a comprehensive view of the molecular mechanisms and regulatory networks underlying the adaptive responses of oyster larvae to hypo-salinity conditions. These findings contribute to our understanding of the complex biological processes involved in oyster resilience and adaptation to changing environmental conditions.
Collapse
Affiliation(s)
- Jianwen Gong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
30
|
Bekhbat M, Drake J, Reed EC, Lauten TH, Natour T, Vladimirov VI, Case AJ. Repeated social defeat stress leads to immunometabolic shifts in innate immune cells of the spleen. Brain Behav Immun Health 2023; 34:100690. [PMID: 37791319 PMCID: PMC10543777 DOI: 10.1016/j.bbih.2023.100690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/05/2023] Open
Abstract
Psychosocial stress has been shown to prime peripheral innate immune cells, which take on hyper-inflammatory phenotypes and are implicated in depressive-like behavior in mouse models. However, the impact of stress on cellular metabolic states that are thought to fuel inflammatory phenotypes in immune cells are unknown. Using single cell RNA-sequencing, we investigated mRNA enrichment of immunometabolic pathways in innate immune cells of the spleen in mice subjected to repeated social defeat stress (RSDS) or no stress (NS). RSDS mice displayed a significant increase in the number of splenic macrophages and granulocytes (p < 0.05) compared to NS littermates. RSDS-upregulated genes in macrophages, monocytes, and granulocytes significantly enriched immunometabolic pathways thought to play a role in myeloid-driven inflammation (glycolysis, HIF-1 signaling, MTORC1 signaling) as well as pathways related to oxidative phosphorylation (OXPHOS) and oxidative stress (p < 0.05 and FDR<0.1). These results suggest that the metabolic enhancement reflected by upregulation of glycolytic and OXPHOS pathways may be important for cellular proliferation of splenic macrophages and granulocytes following repeated stress exposure. A better understanding of these intracellular metabolic mechanisms may ultimately help develop novel strategies to reverse the impact of stress and associated peripheral immune changes on the brain and behavior.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, 30322, USA
| | - John Drake
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
| | - Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| | - Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| | - Vladimir I. Vladimirov
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Psychiatry, University of Arizona, Phoenix, AZ, 85004, USA
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| |
Collapse
|
31
|
Li F, Dong Z, Zhao Q, Payne P, Province M, Cruchaga C, Zhang M, Zhao T, Chen Y. Highly accurate disease diagnosis and highly reproducible biomarker identification with PathFormer. RESEARCH SQUARE 2023:rs.3.rs-3576068. [PMID: 38014034 PMCID: PMC10680938 DOI: 10.21203/rs.3.rs-3576068/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Biomarker identification is critical for precise disease diagnosis and understanding disease pathogenesis in omics data analysis, like using fold change and regression analysis. Graph neural networks (GNNs) have been the dominant deep learning model for analyzing graph-structured data. However, we found two major limitations of existing GNNs in omics data analysis, i.e., limited-prediction/diagnosis accuracy and limited-reproducible biomarker identification capacity across multiple datasets. The root of the challenges is the unique graph structure of biological signaling pathways, which consists of a large number of targets and intensive and complex signaling interactions among these targets. To resolve these two challenges, in this study, we presented a novel GNN model architecture, named PathFormer , which systematically integrate signaling network, priori knowledge and omics data to rank biomarkers and predict disease diagnosis. In the comparison results, PathFormer outperformed existing GNN models significantly in terms of highly accurate prediction capability (~ 30% accuracy improvement in disease diagnosis compared with existing GNN models) and high reproducibility of biomarker ranking across different datasets. The improvement was confirmed using two independent Alzheimer's Disease (AD) and cancer transcriptomic datasets. The PathFormer model can be directly applied to other omics data analysis studies.
Collapse
|
32
|
Xu P, Zhang B. Multiscale network modeling reveals the gene regulatory landscape driving cancer prognosis in 32 cancer types. Genome Res 2023; 33:1806-1817. [PMID: 37907329 PMCID: PMC10691533 DOI: 10.1101/gr.278063.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/22/2023] [Indexed: 11/02/2023]
Abstract
Cancer is a complex disease with diverse molecular mechanisms that affect patient prognosis. Network-based approaches are effective in revealing a holistic picture of cancer prognosis and gene interactions. However, a comprehensive landscape of coexpression networks and prognostic gene modules across multiple cancer types remains elusive. In this study, we performed a systematic analysis of coexpression networks in 32 cancer types. Our analysis identified 4749 prognostic modules that play a vital role in regulating cancer progression. Integrative epigenomic analyses revealed that these modules were regulated by interactions between gene expression and methylation. Coregulated genes of network modules were enriched in chromosome cytobands and preferentially localized in open chromatin regions. The preserved network modules formed 330 module clusters that resided in chromosome hot spots. The cancer-type-specific prognostic modules participated in unique essential biological processes in different cancer types. Overall, our study provides rich resources of prevalent gene networks and underlying multiscale regulatory mechanisms driving cancer prognosis, which lay a foundation for biomarker discovery and therapeutic target development.
Collapse
Affiliation(s)
- Peng Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA;
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA;
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
33
|
Fang X, Weng Y, Zheng X. Involvement of CCL2 and CH25H Genes and TNF signaling pathways in mast cell activation and pathogenesis of chronic spontaneous urticaria. Front Immunol 2023; 14:1247432. [PMID: 37646031 PMCID: PMC10461452 DOI: 10.3389/fimmu.2023.1247432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Chronic spontaneous urticaria (CSU), a mast cell-driven disease, substantially affects the quality of life. While genetics affect CSU susceptibility and severity, the specific genetic factors associated with mast cell activation in CSU remain elusive. We aimed to identify key genetic factors and investigate their roles in CSU pathogenesis. Two gene expression datasets from the Gene Expression Omnibus were merged and validated using principal component analysis and boxplots. The merged dataset was subjected to limma and weighted gene co-expression network analyses. Genes whose expression correlated highly with CSU were identified and analyzed using Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. As GSEA, GO, and KEGG analyses highlighted the importance of chemokine (C-C motif) ligand 2 (CCL2) and cholesterol 25-hydroxylase (CH25H) gene and tumor necrosis factor (TNF) signaling pathways in CSU; the three corresponding genes were knocked down in human mast cell line-1 (HMC-1), followed by incubation with thrombin to mimic CSU pathogenesis. CCL2, CH25H, and TNF knockdown reduced excitability and cytokine production in HMC-1. Our findings suggest that genes involved in the CCL2, CH25H, and TNF pathways play crucial roles in CSU pathogenesis, providing insights into potential therapeutic targets for CSU treatment.
Collapse
Affiliation(s)
- Xiaobin Fang
- Department of Anesthesiology/Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Yueyi Weng
- Department of Anesthesiology/Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University & Fujian Emergency Medical Center, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Critical Medicine, Fuzhou, Fujian, China
- Fujian Provincial Co-constructed Laboratory of “Belt and Road”, Fuzhou, Fujian, China
| |
Collapse
|
34
|
Nakamura-García AK, Espinal-Enríquez J. Pseudogenes in Cancer: State of the Art. Cancers (Basel) 2023; 15:4024. [PMID: 37627052 PMCID: PMC10452131 DOI: 10.3390/cancers15164024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Pseudogenes are duplicates of protein-coding genes that have accumulated multiple detrimental alterations, rendering them unable to produce the protein they encode. Initially disregarded as "junk DNA" due to their perceived lack of functionality, research on their biological roles has been hindered by this assumption. Nevertheless, recent focus has shifted towards these molecules due to their abnormal expression in cancer phenotypes. In this review, our objective is to provide a thorough overview of the current understanding of pseudogene formation, the mechanisms governing their expression, and the roles they may play in promoting tumorigenesis.
Collapse
|
35
|
Ahmed F, Samantasinghar A, Manzoor Soomro A, Kim S, Hyun Choi K. A systematic review of computational approaches to understand cancer biology for informed drug repurposing. J Biomed Inform 2023; 142:104373. [PMID: 37120047 DOI: 10.1016/j.jbi.2023.104373] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/25/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
Cancer is the second leading cause of death globally, trailing only heart disease. In the United States alone, 1.9 million new cancer cases and 609,360 deaths were recorded for 2022. Unfortunately, the success rate for new cancer drug development remains less than 10%, making the disease particularly challenging. This low success rate is largely attributed to the complex and poorly understood nature of cancer etiology. Therefore, it is critical to find alternative approaches to understanding cancer biology and developing effective treatments. One such approach is drug repurposing, which offers a shorter drug development timeline and lower costs while increasing the likelihood of success. In this review, we provide a comprehensive analysis of computational approaches for understanding cancer biology, including systems biology, multi-omics, and pathway analysis. Additionally, we examine the use of these methods for drug repurposing in cancer, including the databases and tools that are used for cancer research. Finally, we present case studies of drug repurposing, discussing their limitations and offering recommendations for future research in this area.
Collapse
Affiliation(s)
- Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea
| | | | | | - Sejong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| |
Collapse
|
36
|
Zhang JJ, Shen Y, Chen XY, Jiang ML, Yuan FH, Xie SL, Zhang J, Xu F. Integrative network-based analysis on multiple Gene Expression Omnibus datasets identifies novel immune molecular markers implicated in non-alcoholic steatohepatitis. Front Endocrinol (Lausanne) 2023; 14:1115890. [PMID: 37008925 PMCID: PMC10061151 DOI: 10.3389/fendo.2023.1115890] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
Introduction Non-alcoholic steatohepatitis (NASH), an advanced subtype of non-alcoholic fatty liver disease (NAFLD), has becoming the most important aetiology for end-stage liver disease, such as cirrhosis and hepatocellular carcinoma. This study were designed to explore novel genes associated with NASH. Methods Here, five independent Gene Expression Omnibus (GEO) datasets were combined into a single cohort and analyzed using network biology approaches. Results 11 modules identified by weighted gene co-expression network analysis (WGCNA) showed significant association with the status of NASH. Further characterization of four gene modules of interest demonstrated that molecular pathology of NASH involves the upregulation of hub genes related to immune response, cholesterol and lipid metabolic process, extracellular matrix organization, and the downregulation of hub genes related to cellular amino acid catabolic, respectively. After DEGs enrichment analysis and module preservation analysis, the Turquoise module associated with immune response displayed a remarkably correlation with NASH status. Hub genes with high degree of connectivity in the module, including CD53, LCP1, LAPTM5, NCKAP1L, C3AR1, PLEK, FCER1G, HLA-DRA and SRGN were further verified in clinical samples and mouse model of NASH. Moreover, single-cell RNA-seq analysis showed that those key genes were expressed by distinct immune cells such as microphages, natural killer, dendritic, T and B cells. Finally, the potential transcription factors of Turquoise module were characterized, including NFKB1, STAT3, RFX5, ILF3, ELF1, SPI1, ETS1 and CEBPA, the expression of which increased with NASH progression. Discussion In conclusion, our integrative analysis will contribute to the understanding of NASH and may enable the development of potential biomarkers for NASH therapy.
Collapse
Affiliation(s)
- Jun-jie Zhang
- Center for Molecular Pathology, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yan Shen
- Department of Publication Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Xiao-yuan Chen
- Department of Publication Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Man-lei Jiang
- Department of Hepatology, The Affiliated Fifth People’s Hospital of Ganzhou, Gannan Medical University, Ganzhou, China
| | - Feng-hua Yuan
- Center for Molecular Pathology, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Shui-lian Xie
- Center for Molecular Pathology, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jie Zhang
- Department of Hepatology, The Affiliated Fifth People’s Hospital of Ganzhou, Gannan Medical University, Ganzhou, China
| | - Fei Xu
- Department of Hepatology, The Affiliated Fifth People’s Hospital of Ganzhou, Gannan Medical University, Ganzhou, China
| |
Collapse
|
37
|
Carmona-Mora P, Knepp B, Jickling GC, Zhan X, Hakoupian M, Hull H, Alomar N, Amini H, Sharp FR, Stamova B, Ander BP. Monocyte, neutrophil, and whole blood transcriptome dynamics following ischemic stroke. BMC Med 2023; 21:65. [PMID: 36803375 PMCID: PMC9942321 DOI: 10.1186/s12916-023-02766-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 12/21/2022] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND After ischemic stroke (IS), peripheral leukocytes infiltrate the damaged region and modulate the response to injury. Peripheral blood cells display distinctive gene expression signatures post-IS and these transcriptional programs reflect changes in immune responses to IS. Dissecting the temporal dynamics of gene expression after IS improves our understanding of immune and clotting responses at the molecular and cellular level that are involved in acute brain injury and may assist with time-targeted, cell-specific therapy. METHODS The transcriptomic profiles from peripheral monocytes, neutrophils, and whole blood from 38 ischemic stroke patients and 18 controls were analyzed with RNA-seq as a function of time and etiology after stroke. Differential expression analyses were performed at 0-24 h, 24-48 h, and >48 h following stroke. RESULTS Unique patterns of temporal gene expression and pathways were distinguished for monocytes, neutrophils, and whole blood with enrichment of interleukin signaling pathways for different time points and stroke etiologies. Compared to control subjects, gene expression was generally upregulated in neutrophils and generally downregulated in monocytes over all times for cardioembolic, large vessel, and small vessel strokes. Self-organizing maps identified gene clusters with similar trajectories of gene expression over time for different stroke causes and sample types. Weighted Gene Co-expression Network Analyses identified modules of co-expressed genes that significantly varied with time after stroke and included hub genes of immunoglobulin genes in whole blood. CONCLUSIONS Altogether, the identified genes and pathways are critical for understanding how the immune and clotting systems change over time after stroke. This study identifies potential time- and cell-specific biomarkers and treatment targets.
Collapse
Affiliation(s)
- Paulina Carmona-Mora
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA.
| | - Bodie Knepp
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Glen C Jickling
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, 87 Avenue & 114 Street, Edmonton, AB, T6G 2J7, Canada
| | - Xinhua Zhan
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Marisa Hakoupian
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Heather Hull
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Noor Alomar
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Hajar Amini
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Frank R Sharp
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Boryana Stamova
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Bradley P Ander
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| |
Collapse
|
38
|
Lanciano T, Savino A, Porcu F, Cittaro D, Bonchi F, Provero P. Contrast subgraphs allow comparing homogeneous and heterogeneous networks derived from omics data. Gigascience 2022; 12:giad010. [PMID: 36852877 PMCID: PMC9972522 DOI: 10.1093/gigascience/giad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/30/2022] [Accepted: 02/08/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Biological networks are often used to describe the relationships between relevant entities, particularly genes and proteins, and are a powerful tool for functional genomics. Many important biological problems can be investigated by comparing biological networks between different conditions or networks obtained with different techniques. FINDINGS We show that contrast subgraphs, a recently introduced technique to identify the most important structural differences between 2 networks, provide a versatile tool for comparing gene and protein networks of diverse origin. We demonstrate the use of contrast subgraphs in the comparison of coexpression networks derived from different subtypes of breast cancer, coexpression networks derived from transcriptomic and proteomic data, and protein-protein interaction networks assayed in different cell lines. CONCLUSIONS These examples demonstrate how contrast subgraphs can provide new insight in functional genomics by extracting the gene/protein modules whose connectivity is most altered between 2 conditions or experimental techniques.
Collapse
Affiliation(s)
| | - Aurora Savino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin 10126, Italy
| | | | - Davide Cittaro
- Center for Omics Sciences, San Raffaele Scientific Institute IRCSS, Milan 20132, Italy
| | | | - Paolo Provero
- Center for Omics Sciences, San Raffaele Scientific Institute IRCSS, Milan 20132, Italy
- Department of Neurosciences “Rita Levi Montalcini,” University of Turin, Turin 10126, Italy
| |
Collapse
|
39
|
Ma C, Li C, Ma H, Yu D, Zhang Y, Zhang D, Su T, Wu J, Wang X, Zhang L, Chen CL, Zhang YE. Pan-cancer surveys indicate cell cycle-related roles of primate-specific genes in tumors and embryonic cerebrum. Genome Biol 2022; 23:251. [PMID: 36474250 PMCID: PMC9724437 DOI: 10.1186/s13059-022-02821-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Despite having been extensively studied, it remains largely unclear why humans bear a particularly high risk of cancer. The antagonistic pleiotropy hypothesis predicts that primate-specific genes (PSGs) tend to promote tumorigenesis, while the molecular atavism hypothesis predicts that PSGs involved in tumors may represent recently derived duplicates of unicellular genes. However, these predictions have not been tested. RESULTS By taking advantage of pan-cancer genomic data, we find the upregulation of PSGs across 13 cancer types, which is facilitated by copy-number gain and promoter hypomethylation. Meta-analyses indicate that upregulated PSGs (uPSGs) tend to promote tumorigenesis and to play cell cycle-related roles. The cell cycle-related uPSGs predominantly represent derived duplicates of unicellular genes. We prioritize 15 uPSGs and perform an in-depth analysis of one unicellular gene-derived duplicate involved in the cell cycle, DDX11. Genome-wide screening data and knockdown experiments demonstrate that DDX11 is broadly essential across cancer cell lines. Importantly, non-neutral amino acid substitution patterns and increased expression indicate that DDX11 has been under positive selection. Finally, we find that cell cycle-related uPSGs are also preferentially upregulated in the highly proliferative embryonic cerebrum. CONCLUSIONS Consistent with the predictions of the atavism and antagonistic pleiotropy hypotheses, primate-specific genes, especially those PSGs derived from cell cycle-related genes that emerged in unicellular ancestors, contribute to the early proliferation of the human cerebrum at the cost of hitchhiking by similarly highly proliferative cancer cells.
Collapse
Affiliation(s)
- Chenyu Ma
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunyan Li
- School of Engineering Medicine, Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), and Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China
| | - Huijing Ma
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Daqi Yu
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yufei Zhang
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Dan Zhang
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Tianhan Su
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianmin Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaoyue Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Chun-Long Chen
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3244, Dynamics of Genetic Information, 75005, Paris, France
| | - Yong E Zhang
- Key Laboratory of Zoological Systematics and Evolution & State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
40
|
Knepp B, Ander BP, Jickling GC, Hull H, Yee AH, Ng K, Rodriguez F, Carmona-Mora P, Amini H, Zhan X, Hakoupian M, Alomar N, Sharp FR, Stamova B. Gene expression changes implicate specific peripheral immune responses to Deep and Lobar Intracerebral Hemorrhages in humans. BRAIN HEMORRHAGES 2022; 3:155-176. [PMID: 36936603 PMCID: PMC10019834 DOI: 10.1016/j.hest.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The peripheral immune system response to Intracerebral Hemorrhage (ICH) may differ with ICH in different brain locations. Thus, we investigated peripheral blood mRNA expression of Deep ICH, Lobar ICH, and vascular risk factor-matched control subjects (n = 59). Deep ICH subjects usually had hypertension. Some Lobar ICH subjects had cerebral amyloid angiopathy (CAA). Genes and gene networks in Deep ICH and Lobar ICH were compared to controls. We found 774 differentially expressed genes (DEGs) and 2 co-expressed gene modules associated with Deep ICH, and 441 DEGs and 5 modules associated with Lobar ICH. Pathway enrichment showed some common immune/inflammatory responses between locations including Autophagy, T Cell Receptor, Inflammasome, and Neuroinflammation Signaling. Th2, Interferon, GP6, and BEX2 Signaling were unique to Deep ICH. Necroptosis Signaling, Protein Ubiquitination, Amyloid Processing, and various RNA Processing terms were unique to Lobar ICH. Finding amyloid processing pathways in blood of Lobar ICH patients suggests peripheral immune cells may participate in processes leading to perivascular/vascular amyloid in CAA vessels and/or are involved in its removal. This study identifies distinct peripheral blood transcriptome architectures in Deep and Lobar ICH, emphasizes the need for considering location in ICH studies/clinical trials, and presents potential location-specific treatment targets.
Collapse
Affiliation(s)
- Bodie Knepp
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Bradley P. Ander
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Glen C. Jickling
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Heather Hull
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Alan H. Yee
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Kwan Ng
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Fernando Rodriguez
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Paulina Carmona-Mora
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Hajar Amini
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Xinhua Zhan
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Marisa Hakoupian
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Noor Alomar
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Frank R. Sharp
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Boryana Stamova
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
41
|
Saha Detroja T, Detroja R, Mukherjee S, Samson AO. Identifying Hub Genes Associated with Neoadjuvant Chemotherapy Resistance in Breast Cancer and Potential Drug Repurposing for the Development of Precision Medicine. Int J Mol Sci 2022; 23:ijms232012628. [PMID: 36293493 PMCID: PMC9603969 DOI: 10.3390/ijms232012628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is the second leading cause of morbidity and mortality in women worldwide. Despite advancements in the clinical application of neoadjuvant chemotherapy (NAC), drug resistance remains a major concern hindering treatment efficacy. Thus, identifying the key genes involved in driving NAC resistance and targeting them with known potential FDA-approved drugs could be applied to advance the precision medicine strategy. With this aim, we performed an integrative bioinformatics study to identify the key genes associated with NAC resistance in breast cancer and then performed the drug repurposing to identify the potential drugs which could use in combination with NAC to overcome drug resistance. In this study, we used publicly available RNA-seq datasets from the samples of breast cancer patients sensitive and resistant to chemotherapy and identified a total of 1446 differentially expressed genes in NAC-resistant breast cancer patients. Next, we performed gene co-expression network analysis to identify significantly co-expressed gene modules, followed by MCC (Multiple Correlation Clustering) clustering algorithms and identified 33 key hub genes associated with NAC resistance. mRNA–miRNA network analysis highlighted the potential impact of these hub genes in altering the regulatory network in NAC-resistance breast cancer cells. Further, several hub genes were found to be significantly involved in the poor overall survival of breast cancer patients. Finally, we identified FDA-approved drugs which could be useful for potential drug repurposing against those hub genes. Altogether, our findings provide new insight into the molecular mechanisms of NAC resistance and pave the way for drug repurposing techniques and personalized treatment to overcome NAC resistance in breast cancer.
Collapse
Affiliation(s)
| | - Rajesh Detroja
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Sumit Mukherjee
- Department of Computer Science, Ben-Gurion University, Beer-Sheva 8410501, Israel
- Correspondence: (S.M.); (A.O.S.)
| | - Abraham O. Samson
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
- Correspondence: (S.M.); (A.O.S.)
| |
Collapse
|
42
|
Zhang Y, Shu H, Mumtaz MA, Hao Y, Li L, He Y, Jin W, Li C, Zhou Y, Lu X, Fu H, Wang Z. Transcriptome and Metabolome Analysis of Color Changes during Fruit Development of Pepper ( Capsicum baccatum). Int J Mol Sci 2022; 23:12524. [PMID: 36293402 PMCID: PMC9604368 DOI: 10.3390/ijms232012524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/17/2022] Open
Abstract
Fruit color is one of the most critical characteristics of pepper. In this study, pepper (Capsicum baccatum L.) fruits with four trans-coloring periods were used as experimental materials to explore the color conversion mechanism of pepper fruit. By transcriptome and metabolome analysis, we identified a total of 307 flavonoid metabolites, 68 carotenoid metabolites, 29 DEGs associated with flavonoid biosynthesis, and 30 DEGs related to carotenoid biosynthesis. Through WGCNA (weighted gene co-expression network analysis) analysis, positively correlated modules with flavonoids and carotenoids were identified, and hub genes associated with flavonoid and carotenoid synthesis and transport were anticipated. We identified Pinobanksin, Naringenin Chalcone, and Naringenin as key metabolites in the flavonoid biosynthetic pathway catalyzed by the key genes chalcone synthase (CHS CQW23_29123, CQW23_29380, CQW23_12748), cinnamic acid 4-hydroxylase (C4H CQW23_16085, CQW23_16084), cytochrome P450 (CYP450 CQW23_19845, CQW23_24900). In addition, phytoene synthase (PSY CQW23_09483), phytoene dehydrogenase (PDS CQW23_11317), zeta-carotene desaturase (ZDS CQW23_19986), lycopene beta cyclase (LYC CQW23_09027), zeaxanthin epoxidase (ZEP CQW23_05387), 9-cis-epoxycarotenoid dioxygenase (NCED CQW23_17736), capsanthin/capsorubin synthase (CCS CQW23_30321) are key genes in the carotenoid biosynthetic pathway, catalyzing the synthesis of key metabolites such as Phytoene, Lycopene, β-carotene and ε-carotene. We also found that transcription factor families such as p450 and NBARC could play important roles in the biosynthesis of flavonoids and carotenoids in pepper fruits. These results provide new insights into the interaction mechanisms of genes and metabolites involved in the biosynthesis of flavonoids and carotenoids in pepper fruit leading to color changes in pepper fruit.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
- Hainan Yazhou Bay Seed Laboratory, Sanya 572025, China
| | - Huangying Shu
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Muhammad Ali Mumtaz
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Yuanyuan Hao
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Lin Li
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Yongjie He
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Weiheng Jin
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Caichao Li
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Yan Zhou
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Xu Lu
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Huizhen Fu
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
| | - Zhiwei Wang
- Key Laboratory for Quality Regulation of Tropical Horticultural Crops of Hainan Province, School of Horticulture, Hainan University, Haikou 570228, China
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
- Hainan Yazhou Bay Seed Laboratory, Sanya 572025, China
| |
Collapse
|
43
|
Wang SY, Huang YH, Liang YJ, Wu JC. Gene coexpression network analysis identifies hubs in hepatitis B virus-associated hepatocellular carcinoma. J Chin Med Assoc 2022; 85:972-980. [PMID: 35801949 DOI: 10.1097/jcma.0000000000000772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is among the leading causes of cancer-related death worldwide. The molecular pathogenesis of HCC involves multiple signaling pathways. This study utilizes systems and bioinformatic approaches to investigate the pathogenesis of HCC. METHODS Gene expression microarray data were obtained from 50 patients with chronic hepatitis B and HCC. There were 1649 differentially expressed genes inferred from tumorous and nontumorous datasets. Weighted gene coexpression network analysis (WGCNA) was performed to construct clustered coexpressed gene modules. Statistical analysis was used to study the correlation between gene coexpression networks and demographic features of patients. Functional annotation and pathway inference were explored for each coexpression network. Network analysis identified hub genes of the prognostic gene coexpression network. The hub genes were further validated with a public database. RESULT Five distinct gene coexpression networks were identified by WGCNA. A distinct coexpressed gene network was significantly correlated with HCC prognosis. Pathway analysis of this network revealed extensive integration with cell cycle regulation. Ten hub genes of this gene network were inferred from protein-protein interaction network analysis and further validated in an external validation dataset. Survival analysis showed that lower expression of the 10-gene signature had better overall survival and recurrence-free survival. CONCLUSION This study identified a crucial gene coexpression network associated with the prognosis of hepatitis B virus-related HCC. The identified hub genes may provide insights for HCC pathogenesis and may be potential prognostic markers or therapeutic targets.
Collapse
Affiliation(s)
- Shen-Yung Wang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Division of Gastroenterology and Hepatology, Department of Medicine, MacKay Memorial Hospital, Taipei, Taiwan, ROC
| | - Yen-Hua Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Center for Systems and Synthetic Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yuh-Jin Liang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Medical Research Department, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Jaw-Ching Wu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Medical Research Department, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
44
|
The Molecular Network behind Volatile Aroma Formation in Pear (Pyrus spp. Panguxiang) Revealed by Transcriptome Profiling via Fatty Acid Metabolic Pathways. Life (Basel) 2022; 12:life12101494. [PMID: 36294930 PMCID: PMC9605550 DOI: 10.3390/life12101494] [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: 08/28/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Pears are popular table fruits, grown and consumed worldwide for their excellent color, aroma, and taste. Volatile aroma is an important factor affecting fruit quality, and the fatty acid metabolism pathway is important in synthesizing volatile aromas. Most of the white pear varieties cultivated in China are not strongly scented, which significantly affects their overall quality. Panguxiang is a white pear cultivar, but its aroma has unique components and is strong. The study of the mechanisms by which aroma is formed in Panguxiang is, therefore, essential to improving the quality of the fruit. The study analyzed physiological and transcriptome factors to reveal the molecular network behind volatile aroma formation in Panguxiang. The samples of Panguxiang fruit were collected in two (fruit development at 60, 90, 120, and 147 days, and fruit storage at 0, 7, 14, 21, and 28 days) periods. A total of nine sample stages were used for RNA extraction and paired-end sequencing. In addition, RNA quantification and qualification, library preparation and sequencing, data analysis and gene annotation, gene co-expression network analysis, and validation of DEGs through quantitative real-time PCR (qRT-;PCR) were performed in this study. The WGCNA identified yellow functional modules and several biological and metabolic pathways related to fatty acid formation. Finally, we identified seven and eight hub genes in the fatty acid synthesis and fatty acid metabolism pathways, respectively. Further analysis of the co-expression network allowed us to identify several key transcription factors related to the volatile aroma, including AP2/ERF-ERF, C3H, MYB, NAC, C2H2, GRAS, and Trihelix, which may also be involved in the fatty acid synthesis. This study lays a theoretical foundation for studying volatile compounds in pear fruits and provides a theoretical basis for related research in other fruits.
Collapse
|
45
|
Game-theoretic link relevance indexing on genome-wide expression dataset identifies putative salient genes with potential etiological and diapeutics role in colorectal cancer. Sci Rep 2022; 12:13409. [PMID: 35927308 PMCID: PMC9352798 DOI: 10.1038/s41598-022-17266-0] [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: 10/01/2021] [Accepted: 07/22/2022] [Indexed: 11/08/2022] Open
Abstract
Diapeutics gene markers in colorectal cancer (CRC) can help manage mortality caused by the disease. We applied a game-theoretic link relevance Index (LRI) scoring on the high-throughput whole-genome transcriptome dataset to identify salient genes in CRC and obtained 126 salient genes with LRI score greater than zero. The biomarkers database lacks preliminary information on the salient genes as biomarkers for all the available cancer cell types. The salient genes revealed eleven, one and six overrepresentations for major Biological Processes, Molecular Function, and Cellular components. However, no enrichment with respect to chromosome location was found for the salient genes. Significantly high enrichments were observed for several KEGG, Reactome and PPI terms. The survival analysis of top protein-coding salient genes exhibited superior prognostic characteristics for CRC. MIR143HG, AMOTL1, ACTG2 and other salient genes lack sufficient information regarding their etiological role in CRC. Further investigation in LRI methodology and salient genes to augment the existing knowledge base may create new milestones in CRC diapeutics.
Collapse
|
46
|
Gene networks under circadian control exhibit diurnal organization in primate organs. Commun Biol 2022; 5:764. [PMID: 35906476 PMCID: PMC9334736 DOI: 10.1038/s42003-022-03722-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 07/14/2022] [Indexed: 12/11/2022] Open
Abstract
Mammalian organs are individually controlled by autonomous circadian clocks. At the molecular level, this process is defined by the cyclical co-expression of both core transcription factors and their downstream targets across time. While interactions between these molecular clocks are necessary for proper homeostasis, these features remain undefined. Here, we utilize integrative analysis of a baboon diurnal transcriptome atlas to characterize the properties of gene networks under circadian control. We found that 53.4% (8120) of baboon genes are oscillating body-wide. Additionally, two basic network modes were observed at the systems level: daytime and nighttime mode. Daytime networks were enriched for genes involved in metabolism, while nighttime networks were enriched for genes associated with growth and cellular signaling. A substantial number of diseases only form significant disease modules at either daytime or nighttime. In addition, a majority of SARS-CoV-2-related genes and modules are rhythmically expressed, which have significant network proximities with circadian regulators. Our data suggest that synchronization amongst circadian gene networks is necessary for proper homeostatic functions and circadian regulators have close interactions with SARS-CoV-2 infection. Integrative analysis of the high-resolution baboon diurnal transcriptome, provides insights into the effect of circadian rhythm on the whole-body primate gene network.
Collapse
|
47
|
Comparative Analysis of Gene Correlation Networks of Breast Cancer Patients Based on Mutations in TP53. Biomolecules 2022; 12:biom12070979. [PMID: 35883535 PMCID: PMC9313229 DOI: 10.3390/biom12070979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 12/10/2022] Open
Abstract
Breast cancer is one of the most prevalent cancers in females, with more than 450,000 deaths each year worldwide. Among the subtypes of breast cancer, basal-like breast cancer, also known as triple-negative breast cancer, shows the lowest survival rate and does not have effective treatments yet. Somatic mutations in the TP53 gene frequently occur across all breast cancer subtypes, but comparative analysis of gene correlations with respect to mutations in TP53 has not been done so far. The primary goal of this study is to identify gene correlations in two groups of breast cancer patients and to derive potential prognostic gene pairs for breast cancer. We partitioned breast cancer patients into two groups: one group with a mutated TP53 gene (mTP53) and the other with a wild-type TP53 gene (wtTP53). For every gene pair, we computed the hazard ratio using the Cox proportional hazard model and constructed gene correlation networks (GCNs) enriched with prognostic information. Our GCN is more informative than typical GCNs in the sense that it indicates the type of correlation between genes, the concordance index, and the prognostic type of a gene. Comparative analysis of correlation patterns and survival time of the two groups revealed several interesting findings. First, we found several new gene pairs with opposite correlations in the two GCNs and the difference in their correlation patterns was the most prominent in the basal-like subtype of breast cancer. Second, we obtained potential prognostic genes for breast cancer patients with a wild-type TP53 gene. From a comparative analysis of GCNs of mTP53 and wtTP53, we found several gene pairs that show significantly different correlation patterns in the basal-like breast cancer subtype and obtained prognostic genes for patients with a wild-type TP53 gene. The GCNs and prognostic genes identified in this study will be informative for the prognosis of survival and for selecting a drug target for breast cancer, in particular for basal-like breast cancer. To the best of our knowledge, this is the first attempt to construct GCNs for breast cancer patients with or without mutations in the TP53 gene and to find prognostic genes accordingly.
Collapse
|
48
|
Bekhbat M, Ulukaya GB, Bhasin MK, Felger JC, Miller AH. Cellular and immunometabolic mechanisms of inflammation in depression: Preliminary findings from single cell RNA sequencing and a tribute to Bruce McEwen. Neurobiol Stress 2022; 19:100462. [PMID: 35655933 PMCID: PMC9152104 DOI: 10.1016/j.ynstr.2022.100462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 11/04/2022] Open
Abstract
Inflammation is associated with symptoms of anhedonia, a core feature of major depression (MD). We have shown that MD patients with high inflammation as measured by plasma C-reactive protein (CRP) and anhedonia display gene signatures of metabolic reprograming (e.g., shift to glycolysis) necessary to sustain cellular immune activation. To gain preliminary insight into the immune cell subsets and transcriptomic signatures that underlie increased inflammation and its relationship with behavior in MD at the single-cell (sc) level, herein we conducted scRNA-Seq on peripheral blood mononuclear cells from a subset of medically-stable, unmedicated MD outpatients. Three MD patients with high CRP (>3 mg/L) before and two weeks after anti-inflammatory challenge with the tumor necrosis factor antagonist infliximab and three patients with low CRP (≤3 mg/L) were studied. Cell clusters were identified using a Single Cell Wizard pipeline, followed by pathway analysis. CD14+ and CD16+ monocytes were more abundant in MD patients with high CRP and were reduced by 29% and 55% respectively after infliximab treatment. Within CD14+ and CD16+ monocytes, genes upregulated in high CRP patients were enriched for inflammatory (phagocytosis, complement, leukocyte migration) and immunometabolic (hypoxia-inducible factor [HIF]-1, aerobic glycolysis) pathways. Shifts in CD4+ T cell subsets included ∼30% and ∼10% lower abundance of CD4+ central memory (TCM) and naïve cells and ∼50% increase in effector memory-like (TEM-like) cells in high versus low CRP patients. TCM cells of high CRP patients displayed downregulation of the oxidative phosphorylation (OXPHOS) pathway, a main energy source in this cell type. Following infliximab, changes in the number of CD14+ monocytes and CD4+ TEM-like cells predicted improvements in anhedonia scores (r = 1.0, p < 0.001). In sum, monocytes and CD4+ T cells from MD patients with increased inflammation exhibited immunometabolic reprograming in association with symptoms of anhedonia. These findings are the first step toward determining the cellular and molecular immune pathways associated with inflammatory phenotypes in MD, which may lead to novel immunomodulatory treatments of psychiatric illnesses with increased inflammation.
Collapse
|
49
|
Arshad Z, McDonald JF. A computational approach to generate highly conserved gene co-expression networks with RNA-seq data. STAR Protoc 2022; 3:101432. [PMID: 35677606 PMCID: PMC9168722 DOI: 10.1016/j.xpro.2022.101432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Zainab Arshad
- Integrated Cancer Research Center, School of Biological Sciences, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30619, USA
| | - John F. McDonald
- Integrated Cancer Research Center, School of Biological Sciences, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30619, USA
- Corresponding author
| |
Collapse
|
50
|
Jahan K, Yin Z, Zhang Y, Yan X, Nie H. Gene Co-Expression Network Analysis Reveals the Correlation Patterns Among Genes in Different Temperature Stress Adaptation of Manila Clam. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:542-554. [PMID: 35482153 DOI: 10.1007/s10126-022-10117-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The Manila clam (Ruditapes philippinarum) is one of the most important aquaculture species and widely distributed along the coasts of China, Japan, and Korea. Due to its wide distribution, it can tolerate a wide range of temperature. Studying the gene expression profiles of clam gills had found differentially expressed genes (DEGs) and pathway involved in temperature stress tolerance. A systematic study of cellular response to temperature stress may provide insights into the mechanism of acquired tolerance. Here, weighted gene co-expression network analysis (WGCNA) was carried out using RNA-seq data from gill transcriptome in response to high and low temperature stress. There are a total 32 gene modules, of which 18 gene modules were identified as temperature-related modules. Blue module was one significantly correlated with temperature which was associated with cellular metabolism, apoptosis pathway, ER stress, and others.
Collapse
Affiliation(s)
- Kifat Jahan
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Zhihui Yin
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Yanming Zhang
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Xiwu Yan
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Hongtao Nie
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|