Cargando…

RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis

PURPOSE: This study aimed to provide a comprehensive understanding of the genome-wide expression patterns in the synovial tissue samples of patients with rheumatoid arthritis (RA) to investigate the potential mechanisms regulating RA occurrence and development. METHODS: Transcription profiles of the...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Runrun, Jin, Yehua, Chang, Cen, Xu, Lingxia, Bian, Yanqin, Shen, Yu, Sun, Yang, Sun, Songtao, Schrodi, Steven J., Guo, Shicheng, He, Dongyi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9116426/
https://www.ncbi.nlm.nih.gov/pubmed/35602512
http://dx.doi.org/10.3389/fmed.2022.799440
_version_ 1784710110733402112
author Zhang, Runrun
Jin, Yehua
Chang, Cen
Xu, Lingxia
Bian, Yanqin
Shen, Yu
Sun, Yang
Sun, Songtao
Schrodi, Steven J.
Guo, Shicheng
He, Dongyi
author_facet Zhang, Runrun
Jin, Yehua
Chang, Cen
Xu, Lingxia
Bian, Yanqin
Shen, Yu
Sun, Yang
Sun, Songtao
Schrodi, Steven J.
Guo, Shicheng
He, Dongyi
author_sort Zhang, Runrun
collection PubMed
description PURPOSE: This study aimed to provide a comprehensive understanding of the genome-wide expression patterns in the synovial tissue samples of patients with rheumatoid arthritis (RA) to investigate the potential mechanisms regulating RA occurrence and development. METHODS: Transcription profiles of the synovial tissue samples from nine patients with RA and 15 patients with osteoarthritis (OA) (control) from the East Asian population were generated using RNA sequencing (RNA-seq). Gene set enrichment analysis (GSEA) was used to analyze all the detected genes and the differentially expressed genes (DEGs) were identified using DESeq. To further analyze the DEGs, the Gene Ontology (GO) functional enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. The protein–protein interaction (PPI) network of the DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and the hub genes were identified by topology clustering with the Molecular Complex Detection (MCODE)-Cytoscape. The most important hub genes were validated using quantitative real-time PCR (qRT-PCR). RESULTS: Of the 17,736 genes detected, 851 genes were identified as the DEGs (474 upregulated and 377 downregulated genes) using the false discovery rate (FDR) approach. GSEA revealed that the significantly enriched gene sets that positively correlated with RA were CD40 signaling overactivation, Th1 cytotoxic module, overactivation of the immune response, adaptive immune response, effective vs. memory CD8+ T cells (upregulated), and naïve vs. effective CD8+ T cells (downregulated). Biological process enrichment analysis showed that the DEGs were significantly enriched for signal transduction (P = 3.01 × 10(−6)), immune response (P = 1.65 × 10(−24)), and inflammatory response (P = 5.76 × 10(−10)). Molecule function enrichment analysis revealed that the DEGs were enriched in calcium ion binding (P = 1.26 × 10(−5)), receptor binding (P = 1.26 × 10(−5)), and cytokine activity (P = 2.01 × 10(−3)). Cellular component enrichment analysis revealed that the DEGs were significantly enriched in the plasma membrane (P = 1.91 × 10(−31)), an integral component of the membrane (P = 7.39 × 10(−13)), and extracellular region (P = 7.63 × 10(−11)). The KEGG pathway analysis showed that the DEGs were enriched in the cytokine–cytokine receptor interaction (P = 3.05 × 10(−17)), chemokine signaling (P = 3.50 × 10(−7)), T-cell receptor signaling (P = 5.17 × 10(−4)), and RA (P = 5.17 × 10(−4)) pathways. We confirmed that RA was correlated with the upregulation of the PPI network hub genes, such as CXCL13, CXCL6, CCR5, CXCR5, CCR2, CXCL3, and CXCL10, and the downregulation of the PPI network hub gene such as SSTR1. CONCLUSION: This study identified and validated the DEGs in the synovial tissue samples of patients with RA, which highlighted the activity of a subset of chemokine genes, thereby providing novel insights into the molecular mechanisms of RA pathogenesis and identifying potential diagnostic and therapeutic targets for RA.
format Online
Article
Text
id pubmed-9116426
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-91164262022-05-19 RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis Zhang, Runrun Jin, Yehua Chang, Cen Xu, Lingxia Bian, Yanqin Shen, Yu Sun, Yang Sun, Songtao Schrodi, Steven J. Guo, Shicheng He, Dongyi Front Med (Lausanne) Medicine PURPOSE: This study aimed to provide a comprehensive understanding of the genome-wide expression patterns in the synovial tissue samples of patients with rheumatoid arthritis (RA) to investigate the potential mechanisms regulating RA occurrence and development. METHODS: Transcription profiles of the synovial tissue samples from nine patients with RA and 15 patients with osteoarthritis (OA) (control) from the East Asian population were generated using RNA sequencing (RNA-seq). Gene set enrichment analysis (GSEA) was used to analyze all the detected genes and the differentially expressed genes (DEGs) were identified using DESeq. To further analyze the DEGs, the Gene Ontology (GO) functional enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. The protein–protein interaction (PPI) network of the DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and the hub genes were identified by topology clustering with the Molecular Complex Detection (MCODE)-Cytoscape. The most important hub genes were validated using quantitative real-time PCR (qRT-PCR). RESULTS: Of the 17,736 genes detected, 851 genes were identified as the DEGs (474 upregulated and 377 downregulated genes) using the false discovery rate (FDR) approach. GSEA revealed that the significantly enriched gene sets that positively correlated with RA were CD40 signaling overactivation, Th1 cytotoxic module, overactivation of the immune response, adaptive immune response, effective vs. memory CD8+ T cells (upregulated), and naïve vs. effective CD8+ T cells (downregulated). Biological process enrichment analysis showed that the DEGs were significantly enriched for signal transduction (P = 3.01 × 10(−6)), immune response (P = 1.65 × 10(−24)), and inflammatory response (P = 5.76 × 10(−10)). Molecule function enrichment analysis revealed that the DEGs were enriched in calcium ion binding (P = 1.26 × 10(−5)), receptor binding (P = 1.26 × 10(−5)), and cytokine activity (P = 2.01 × 10(−3)). Cellular component enrichment analysis revealed that the DEGs were significantly enriched in the plasma membrane (P = 1.91 × 10(−31)), an integral component of the membrane (P = 7.39 × 10(−13)), and extracellular region (P = 7.63 × 10(−11)). The KEGG pathway analysis showed that the DEGs were enriched in the cytokine–cytokine receptor interaction (P = 3.05 × 10(−17)), chemokine signaling (P = 3.50 × 10(−7)), T-cell receptor signaling (P = 5.17 × 10(−4)), and RA (P = 5.17 × 10(−4)) pathways. We confirmed that RA was correlated with the upregulation of the PPI network hub genes, such as CXCL13, CXCL6, CCR5, CXCR5, CCR2, CXCL3, and CXCL10, and the downregulation of the PPI network hub gene such as SSTR1. CONCLUSION: This study identified and validated the DEGs in the synovial tissue samples of patients with RA, which highlighted the activity of a subset of chemokine genes, thereby providing novel insights into the molecular mechanisms of RA pathogenesis and identifying potential diagnostic and therapeutic targets for RA. Frontiers Media S.A. 2022-05-04 /pmc/articles/PMC9116426/ /pubmed/35602512 http://dx.doi.org/10.3389/fmed.2022.799440 Text en Copyright © 2022 Zhang, Jin, Chang, Xu, Bian, Shen, Sun, Sun, Schrodi, Guo and He. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Medicine
Zhang, Runrun
Jin, Yehua
Chang, Cen
Xu, Lingxia
Bian, Yanqin
Shen, Yu
Sun, Yang
Sun, Songtao
Schrodi, Steven J.
Guo, Shicheng
He, Dongyi
RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis
title RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis
title_full RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis
title_fullStr RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis
title_full_unstemmed RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis
title_short RNA-seq and Network Analysis Reveal Unique Chemokine Activity Signatures in the Synovial Tissue of Patients With Rheumatoid Arthritis
title_sort rna-seq and network analysis reveal unique chemokine activity signatures in the synovial tissue of patients with rheumatoid arthritis
topic Medicine
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9116426/
https://www.ncbi.nlm.nih.gov/pubmed/35602512
http://dx.doi.org/10.3389/fmed.2022.799440
work_keys_str_mv AT zhangrunrun rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT jinyehua rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT changcen rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT xulingxia rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT bianyanqin rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT shenyu rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT sunyang rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT sunsongtao rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT schrodistevenj rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT guoshicheng rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis
AT hedongyi rnaseqandnetworkanalysisrevealuniquechemokineactivitysignaturesinthesynovialtissueofpatientswithrheumatoidarthritis