1
|
Rodrigues R, Sousa C, Barros A, Vale N. Chlamydia trachomatis: From Urogenital Infections to the Pathway of Infertility. Genes (Basel) 2025; 16:205. [PMID: 40004534 PMCID: PMC11855039 DOI: 10.3390/genes16020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Chlamydia trachomatis (CT) is a major cause of sexually transmitted infections (STIs) worldwide, with significant implications for reproductive health. The bacterium's genome contains highly polymorphic regions, influencing both the type and severity of infections. These genetic variations, particularly those occurring in the major outer membrane protein (MOMP) gene, are critical for classifying the bacterium into distinct serovars and enable CT to adapt to diverse host environments, contributing to its immune evasion, persistence, and pathogenicity. Persistent or untreated urogenital infections can lead to chronic inflammation, tissue damage, and pelvic inflammatory disease, ultimately increasing the risk of ectopic pregnancy, spontaneous abortion, and infertility. This review consolidates current knowledge on the genetic diversity of CT, its potential role in modulating infection outcomes, and its immune evasion mechanisms. By integrating scientific evidence linking chlamydial infections to infertility, we underscore the urgent need for targeted research to address this critical public health challenge.
Collapse
Affiliation(s)
- Rafaela Rodrigues
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, Leça do Balio, 4465-671 Matosinhos, Portugal
| | - Carlos Sousa
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, Leça do Balio, 4465-671 Matosinhos, Portugal
| | - Alberto Barros
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Centre for Reproductive Genetics Alberto Barros, 4100-012 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
2
|
Meewes C, Gupta K, Geisler WM. Role of microRNAs in immune regulation and pathogenesis of Chlamydia trachomatis and Chlamydia muridarum infections: a rapid review. Microbes Infect 2024; 26:105397. [PMID: 39025257 PMCID: PMC11609027 DOI: 10.1016/j.micinf.2024.105397] [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: 02/21/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
MicroRNAs in Chlamydia trachomatis (CT) and Chlamydia muridarum (CM) infections are an emerging topic of research that provide knowledge that could advance vaccine development and strategies for managing infection. This rapid review summarizes human and murine studies on miRNA expression in CT and CM infections in vivo and ex vivo.
Collapse
Affiliation(s)
- Chloe Meewes
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kanupriya Gupta
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - William M Geisler
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
3
|
Ji DN, Jin SD, Jiang Y, Xu FY, Fan SW, Zhao YL, Liu XQ, Sun H, Cheng WZ, Zhang XY, Guan XX, Zhang BW, Du ZM, Wang Y, Wang N, Zhang R, Zhang MY, Xu CQ. CircNSD1 promotes cardiac fibrosis through targeting the miR-429-3p/SULF1/Wnt/β-catenin signaling pathway. Acta Pharmacol Sin 2024; 45:2092-2106. [PMID: 38760544 PMCID: PMC11420342 DOI: 10.1038/s41401-024-01296-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/17/2024] [Indexed: 05/19/2024]
Abstract
Cardiac fibrosis is a detrimental pathological process, which constitutes the key factor for adverse cardiac structural remodeling leading to heart failure and other critical conditions. Circular RNAs (circRNAs) have emerged as important regulators of various cardiovascular diseases. It is known that several circRNAs regulate gene expression and pathological processes by binding miRNAs. In this study we investigated whether a novel circRNA, named circNSD1, and miR-429-3p formed an axis that controls cardiac fibrosis. We established a mouse model of myocardial infarction (MI) for in vivo studies and a cellular model of cardiac fibrogenesis in primary cultured mouse cardiac fibroblasts treated with TGF-β1. We showed that miR-429-3p was markedly downregulated in the cardiac fibrosis models. Through gain- and loss-of-function studies we confirmed miR-429-3p as a negative regulator of cardiac fibrosis. In searching for the upstream regulator of miR-429-3p, we identified circNSD1 that we subsequently demonstrated as an endogenous sponge of miR-429-3p. In MI mice, knockdown of circNSD1 alleviated cardiac fibrosis. Moreover, silence of human circNSD1 suppressed the proliferation and collagen production in human cardiac fibroblasts in vitro. We revealed that circNSD1 directly bound miR-429-3p, thereby upregulating SULF1 expression and activating the Wnt/β-catenin pathway. Collectively, circNSD1 may be a novel target for the treatment of cardiac fibrosis and associated cardiac disease.
Collapse
Affiliation(s)
- Dong-Ni Ji
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Sai-di Jin
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Yuan Jiang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Fei-Yong Xu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Shu-Wei Fan
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yi-Lin Zhao
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Xin-Qi Liu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Hao Sun
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Wen-Zheng Cheng
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Xin-Yue Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Xiao-Xiang Guan
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Bo-Wen Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Zhi-Min Du
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Ying Wang
- Center of Chronic Diseases and Drug Research of Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Ning Wang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
| | - Rong Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China.
| | - Ming-Yu Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China.
| | - Chao-Qian Xu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
4
|
Galdiero M, Trotta C, Schettino MT, Cirillo L, Sasso FP, Petrillo F, Petrillo A. Normospermic Patients Infected With Ureaplasma parvum: Role of Dysregulated miR-122-5p, miR-34c-5, and miR-141-3p. Pathog Immun 2024; 8:16-36. [PMID: 38223489 PMCID: PMC10783813 DOI: 10.20411/pai.v8i2.603] [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: 06/14/2023] [Accepted: 10/20/2023] [Indexed: 01/16/2024] Open
Abstract
Background Ureaplasma parvum (UP) is a causative agent of non-gonococcal urethritis, involved in the pathogenesis of prostatitis and epididymitis, and it could impair human fertility. Although UP infection is a frequent cause of male infertility the study evidence assessing their prevalence and the association in patients with infertility is still scarce. The molecular processes leading to defects in spermatozoa quality are not completely investigated. MicroRNAs (miRNAs) have been extensively reported as gene regulatory molecules on post-transcriptional levels involved in various biological processes such as gametogenesis, embryogenesis, and the quality of sperm, oocyte, and embryos. Methods Therefore, the study design was to demonstrate that miRNAs in body fluids like sperm could be utilized as non-invasive diagnostic biomarkers for pathological and physiological conditions such as infertility. A post-hoc bioinformatics analysis was carried out to predict the pathways modulated by the miRNAs dysregulated in the differently motile spermatozoa. Results Here it is shown that normospermic patients infected by UP had spermatozoa with increased quantity of superoxide anions, reduced expression of miR-122-5p, miR-34c-5, and increased miR-141-3p compared with non-infected normospermic patients. This corresponded to a reduction of sperm motility in normospermic infected patients compared with normospermic non-infected ones. A target gene prediction presumed that an essential role of these miRNAs resided in the regulation of lipid kinase activity, accounting for the changes in the constitution of spermatozoa membrane lipids caused by UP. Conclusions Altogether, the data underline the influence of UP on epigenetic mechanisms regulating spermatozoa motility.
Collapse
Affiliation(s)
- Marilena Galdiero
- Department of Experimental Medicine, Section of Microbiology and Virology, University of Campania Luigi Vanvitelli, 80138, Naples, Italy. Department of Experimental Medicine, Section of Pharmacology, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Carolo Trotta
- Department of Gynecology and Obstetrics University of Campania Luigi Vanvitelli Naples Italy
| | - Maria Teresa Schettino
- Department of Gynecology and Obstetrics University of Campania Luigi Vanvitelli Naples Italy
| | - Luigi Cirillo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples Federico II, Naples Italy
| | | | - Francesco Petrillo
- Department of Experimental Medicine, Section of Microbiology and Virology, University of Campania Luigi Vanvitelli, 80138, Naples, Italy. Department of Experimental Medicine, Section of Pharmacology, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | | |
Collapse
|
5
|
Zhang Y, Bai H, Zhang W, Gao J, Gao C, Deng T, Liu X, Sun X, Liu Y, Wang N, Wu Y. miR-212/132 attenuates OVA-induced airway inflammation by inhibiting mast cells activation through MRGPRX2 and ASAP1. Exp Cell Res 2023; 433:113828. [PMID: 37875175 DOI: 10.1016/j.yexcr.2023.113828] [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: 01/22/2023] [Revised: 09/25/2023] [Accepted: 10/19/2023] [Indexed: 10/26/2023]
Abstract
Allergic asthma is a chronic inflammatory disease of airways involving complex mechanisms, including MAS-related GPR family member X2 (MRGPRX2) and its orthologue MRGPRB2 on mast cells (MCs). Although miRNAs have been previously shown to related to allergic asthma, the role of miR-212/132 in this process has not been studied. In this study, the predicted pairing of miRNAs and MRGPRX2 (MRGPRB2) mRNAs was carried out by online databases and the function was verify using in vivo and in vitro experiments. Database prediction showed that miR-212/132 interact with MRGPRX2 and MRGPRB2. miR-212/132 mimics alleviated MRGPRB2 mRNA expression as well as pathology changes in lungs and AHR of mice with airway inflammation in vivo. The expression level of MRGPRB2 in the mice lungs after inhaled OVA was also decreased by miR-212/132 mimics. Meanwhile, miR-212/132 inhibited MCs degranulation and cytokines release triggered by C48/80 in vitro. Further, ASAP1 (ARF GTPase-Activating Protein 1) was selected from the junction related pathways using RNAseq and KEGG enrichment. ASAP1 mRNA level was upregulated in airway inflammation and MCs activation and decreased by miR-212/132 mimics. miR-212/132 attenuated OVA-induced airway inflammation by inhibiting MCs activation through MRGPRX2 and ASAP1.
Collapse
Affiliation(s)
- Yongjing Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Haoyun Bai
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Wen Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Jie Gao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Chang Gao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Tingting Deng
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Xinyu Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, 157 West 5th Road, Xi'an, Shaanxi, China
| | - Xiuzhen Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, 157 West 5th Road, Xi'an, Shaanxi, China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, 157 West 5th Road, Xi'an, Shaanxi, China
| | - Nan Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China.
| | - Yuanyuan Wu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, 157 West 5th Road, Xi'an, Shaanxi, China.
| |
Collapse
|
6
|
Ray A, Bhati T, Arora R, Rastogi S. Progesterone-mediated immunoregulation of cytokine signaling by miRNA-133a and 101-3p in Chlamydia trachomatis-associated recurrent spontaneous abortion. Mol Immunol 2023; 164:47-57. [PMID: 37952361 DOI: 10.1016/j.molimm.2023.10.012] [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: 06/20/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023]
Abstract
miRNAs regulate the expression of various genes involved in cellular and metabolic pathways in pregnancy related complications including recurrent spontaneous abortion (RSA). Modulation of progesterone and associated pro-inflammatory cytokines by miRNAs in Chlamydia trachomatis-associated RSA is still under investigation. Present study aimed to evaluate the expression/correlation of serum-circulating miRNAs-133a, 101-3p, 320b, 146b-5p, 24, 559, progesterone and few cytokines in C. trachomatis-positive spontaneous aborters. Non-heparinized blood and urine was collected from 120 patients with history of RSA (Group I) and 120 patients with ≥ 2 successful deliveries (Group II) attending Department of Obstetrics and Gynecology, Safdarjung hospital, New Delhi, India. C. trachomatis detection was performed by PCR and chlamydial load by real time PCR. Progesterone concentration was estimated by ELISA. miRNAs and cytokine expression was studied by quantitative real-time PCR and correlated with progesterone expression. Twenty six patients were found to be positive for C. trachomatis. miRNAs- 133a, 101-3p showed maximum upregulation in infected versus control patients. miRNA expression showed positive correlation with chlamydial load. Progesterone concentration showed significant decrease while cytokines (IL-6, IFN-γ, TNF-α) were significantly upregulated in C. trachomatis-positive patients. Positive correlation was observed between expression of miRNAs-133a and 101-3p and cytokines while negative correlation was observed with progesterone in infected RSA patients. Correlation between progesterone and cytokines was found to be significantly negative in infected RSA patients. Although further validation is required, the study concludes that miR-133a and 101-3p are of clinical importance and have a role in immunoregulation of progesterone and cytokines in infection associated RSA.
Collapse
Affiliation(s)
- Ankita Ray
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital campus, Post Box no. 4909, New Delhi 110029, India
| | - Tanu Bhati
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital campus, Post Box no. 4909, New Delhi 110029, India
| | - Renu Arora
- Department of Obstetrics and Gynecology, Vardhman Mahavir Medical College (VMMC) and Safdarjung Hospital, New Delhi 110029, India
| | - Sangita Rastogi
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital campus, Post Box no. 4909, New Delhi 110029, India.
| |
Collapse
|
7
|
Stein RA, Thompson LM. Epigenetic changes induced by pathogenic Chlamydia spp. Pathog Dis 2023; 81:ftad034. [PMID: 38031337 DOI: 10.1093/femspd/ftad034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/16/2023] [Accepted: 11/28/2023] [Indexed: 12/01/2023] Open
Abstract
Chlamydia trachomatis, C. pneumoniae, and C. psittaci, the three Chlamydia species known to cause human disease, have been collectively linked to several pathologies, including conjunctivitis, trachoma, respiratory disease, acute and chronic urogenital infections and their complications, and psittacosis. In vitro, animal, and human studies also established additional correlations, such as between C. pneumoniae and atherosclerosis and between C. trachomatis and ovarian cancer. As part of their survival and pathogenesis strategies as obligate intracellular bacteria, Chlamydia spp. modulate all three major types of epigenetic changes, which include deoxyribonucleic acid (DNA) methylation, histone post-translational modifications, and microRNA-mediated gene silencing. Some of these epigenetic changes may be implicated in key aspects of pathogenesis, such as the ability of the Chlamydia spp. to induce epithelial-to-mesenchymal transition, interfere with DNA damage repair, suppress cholesterol efflux from infected macrophages, act as a co-factor in human papillomavirus (HPV)-mediated cervical cancer, prevent apoptosis, and preserve the integrity of mitochondrial networks in infected host cells. A better understanding of the individual and collective contribution of epigenetic changes to pathogenesis will enhance our knowledge about the biology of Chlamydia spp. and facilitate the development of novel therapies and biomarkers. Pathogenic Chlamydia spp. contribute to epigenetically-mediated gene expression changes in host cells by multiple mechanisms.
Collapse
Affiliation(s)
- Richard A Stein
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, United States
| | - Lily M Thompson
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, United States
| |
Collapse
|
8
|
Liu Y, Xiang J, Hu X, Wang H, Sun Y. Expression profile screening and bioinformatics analysis of CircRNA, LncRNA, and mRNA in HeLa cells infected with Chlamydia muridarum. Arch Microbiol 2022; 204:352. [PMID: 35622163 DOI: 10.1007/s00203-022-02941-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 11/02/2022]
Abstract
We have previously shown that circRNAs in host cells are involved in the process of Chlamydia trachomatis infection. In this study we aimed to identify significantly altered circRNAs/lncRNAs/mRNAs in Chlamydia muridarum infected cells and investigate their biological functions in the interaction between Chlamydia muridarum and host cells. For this purpose, circRNA, lncRNA and mRNA expression profiles were screened and identified in HeLa cells with or without Chlamydia muridarum infection by microarray. Bioinformatics analyses including Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) analysis were then carried out and the circRNA-miRNA ceRNA network was constructed. The differentially expressed circRNAs and lncRNAs were selected for validation by RT-qPCR. The results shown that a total of 834 circRNAs, 2149 lncRNAs and 1283 mRNAs were found to be differentially expressed. Enrichment analysis of GO and KEGG showed that the dysregulated genes involved nuclear-transcribed mRNA catabolic process, protein binding, RNA catabolic process and translation, the MAPK signaling pathway, apoptosis, Toll-like receptor signaling pathway, cAMP signaling pathway and Notch signaling pathway may play important roles in Chlamydia infection. Our study provides a systematic outlook on the potential function of non-coding RNAs in the molecular basis of Chlamydia infection.
Collapse
Affiliation(s)
- Yuanjun Liu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Junqiu Xiang
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xinyue Hu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Huiping Wang
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yina Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
9
|
Huang X, Liufu Q, Xu R, Chen X, Liu M, Han J, Guan H, Ma C. Integrating lncRNAs and mRNAs Expression Profiles in Penicillin-Induced Persistent Chlamydial Infection in HeLa Cells. Front Mol Biosci 2022; 9:744901. [PMID: 35252346 PMCID: PMC8890745 DOI: 10.3389/fmolb.2022.744901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Chlamydia trachomatis (C. trachomatis) is a major etiological agent of sexually transmitted infection. Some stressing conditions can result in persistent chlamydial infection, which is thought to be associated with severe complications including ectopic pregnancy and tubal factor infertility. Long noncoding RNAs (lncRNAs) have been identified as key modulators in many biological processes. Nevertheless, the role of lncRNAs in persistent chlamydial infection is still unclear. In this study, we used lncRNA and mRNA microarray to identify the global lncRNAs and mRNAs expression in penicillin-induced persistent chlamydial infection in HeLa cells as well as the control group (HeLa cells without C. trachomatis infection). Among 1005 differentially expressed lncRNAs, 585 lncRNAs were upregulated and 420 downregulated in persistent chlamydial infection, while 410 mRNAs were identified to express differentially, of which 113 mRNAs were upregulated and 297 downregulated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis with differentially expressed genes were performed. We then constructed the lncRNA-miRNA-mRNA competing endogenous RNAs (ceRNAs) network. Four mRNAs were validated to be changed by quantitative real-time PCR which were correlated with the microarray result. Integration of protein-protein interaction network was constructed and hub genes were identified. These findings provide a new perspective on the molecular mechanisms of penicillin-induced persistent chlamydial infection.
Collapse
Affiliation(s)
- Xiaobao Huang
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Liufu
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Xu
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Chen
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mingna Liu
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiande Han
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongyu Guan
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Chunguang Ma, ; Hongyu Guan,
| | - Chunguang Ma
- Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Chunguang Ma, ; Hongyu Guan,
| |
Collapse
|
10
|
Tang H, Lin Y, Huang L, Hu J. MiR-223-3p Regulates Autophagy and Inflammation by Targeting ATG16L1 in Fusarium solani-Induced Keratitis. Invest Ophthalmol Vis Sci 2022; 63:41. [PMID: 35089329 PMCID: PMC8802027 DOI: 10.1167/iovs.63.1.41] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/05/2022] [Indexed: 12/17/2022] Open
Abstract
Purpose Increasing evidence suggested that microRNAs (miRs) are implicated in the regulation of the inflammatory response and autophagy in multiple diseases. The present study aimed to explore the effect of miR-223-3p on inflammation and autophagy in fungal keratitis (FK). Methods An FK mouse model was established, and primary corneal stromal cells were isolated by inoculation with Fusarium solani. The expression of miR-223-3p was determined by quantitative RT-PCR. Subsequently, the target gene of miR-223-3p was identified by a dual-luciferase reporter assay. The levels of miR-223-3p were altered by transfecting miR agomir/antagomir to evaluate its effects. Slit-lamp biomicroscopy and hematoxylin and eosin staining were employed to detect corneal damage. The levels of autophagy were assessed by immunofluorescence, Western blotting, mRFP-GFP-LC3 fluorescence microscopy, and electron microscopy. In addition, inflammation was demonstrated by determining the proinflammatory mediators IL-1β and TNF-ɑ. Results Our data suggested that miR-223-3p was increased and that autophagic flux was impaired in mouse FK. Then, we confirmed that autophagy-related gene 16L1 (ATG16L1) was a potential target of miR-223-3p and that this miR negatively regulated the expression of ATG16L1. The inhibition of miR-223-3p attenuated inflammation in FK, reduced P62 expression, and increased the ratio of LC3-II/LC3-I, whereas the overexpression of miR-223-3p displayed the opposite results. Conclusions Taken together, miR-223-3p might regulate autophagy via targeting ATG16L1 in experimental F. solani keratitis and is associated with the inflammatory response. MiR-223-3p might be a potential therapeutic target for FK.
Collapse
Affiliation(s)
- Hanfeng Tang
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fu Zhou, China
| | - Yi Lin
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fu Zhou, China
| | - Liwei Huang
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fu Zhou, China
| | - Jianzhang Hu
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fu Zhou, China
| |
Collapse
|
11
|
Ling H, Luo L, Dai X, Chen H. Fallopian tubal infertility: the result of Chlamydia trachomatis-induced fallopian tubal fibrosis. Mol Cell Biochem 2021; 477:205-212. [PMID: 34652537 DOI: 10.1007/s11010-021-04270-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/29/2021] [Indexed: 11/26/2022]
Abstract
Chlamydia trachomatis is one of the most common pathogens of sexually transmitted diseases, and its incidence in genital tract infections is now 4.7% in south China. Infertility is the end result of C. trachomatis-induced fallopian tubal fibrosis and is receiving intense attention from scientists worldwide. To reduce the incidence of infertility, it is important to understand the pathology-related changes of the genital tract where C. trachomatis infection is significant, especially the mechanism of fibrosis formation. During fibrosis development, the fallopian tube becomes sticky and occluded, which will eventually lead to tubal infertility. At present, the mechanism of fallopian tubal fibrosis induced by C. trachomatis infection is unclear. Our study attempted to summarize the possible mechanisms of fibrosis caused by C. trachomatis infection in the fallopian tube by reviewing published studies and further providing potential therapeutic targets to reduce the occurrence of infertility. This study also provides ideas for future research. Factors leading to fallopian tube fibrosis include inflammatory factors, miRNA, ECT, cHSP, and host factors. We hypothesized that C. trachomatis mediates the transcription and translation of EMT and ECM via upregulating TGF signaling pathway, which leads to the formation of fallopian tube fibrosis and ultimately to tubal infertility.
Collapse
Affiliation(s)
- Hua Ling
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China
| | - Lipei Luo
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China
| | - Xingui Dai
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China.
- The First People's Hospital of Chenzhou, Chenzhou, 423000, People's Republic of China.
- The First Affiliated Hospital of Xiangnan University, Chenzhou, 423000, People's Republic of China.
| | - Hongliang Chen
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China.
- The First People's Hospital of Chenzhou, Chenzhou, 423000, People's Republic of China.
- The First Affiliated Hospital of Xiangnan University, Chenzhou, 423000, People's Republic of China.
| |
Collapse
|
12
|
Ray A, Bhati T, Arora R, Pradhan D, Parvez S, Rastogi S. Differential expression of urine-circulating micro-RNAs in Chlamydia trachomatis-induced recurrent spontaneous aborters. Microb Pathog 2021; 160:105156. [PMID: 34418493 DOI: 10.1016/j.micpath.2021.105156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/01/2021] [Accepted: 08/17/2021] [Indexed: 12/29/2022]
Abstract
Studies behind mechanisms of Chlamydia trachomatis-induced recurrent spontaneous abortion is still in its infancy. Possible strategy for preventing recurrent spontaneous abortion at molecular level is needed. Despite its multifactorial aetiology, Chlamydia trachomatis is important cause of RSA. However, mechanism leading to RSA in C. trachomatis-positive patients is not understood and novel strategies are needed. It is hypothesized that microRNAs play important role in RSA regulation during infection. Study aimed to elucidate expression/role of urine-circulating miRs-320b, 221-3p, 146b-5p,-16,-24,-559 in recurrent spontaneous aborters with C. trachomatis infection and to find their target genes by bioinformatic analysis. First-void urine was collected from 30 non-pregnant women with RSA (Group I) and 30 non-pregnant women with ≥2 successful deliveries (Group II; Controls) attending Department of Obstetrics and Gynaecology, Vardhman Mahavir Medical College, Safdarjung hospital, New Delhi (India). PCR was performed to detect C. trachomatis. Expression of miRNAs was studied by quantitative real-time PCR while target genes/functional annotations were predicted by GO/KEGG databases. Data was statistically evaluated. 05 RSA patients were C. trachomatis-positive. Group I was subdivided into Group Ia (C. trachomatis-positive RSA; n = 5) and Group Ib (C. trachomatis-negative RSA; internal controls). miR-320b, -221-3p, -146b-5p, -16, -24 were significantly upregulated (miR-16 showed maximum 4.3 fold-change) while miR-559 was downregulated (0.5 fold-change) in Group Ia versus controls ('p'<0.001). Bioinformatic analysis revealed that target genes of miRNAs in RSA are involved in apoptosis and AMPK signalling pathways. Results showed differential expression of miRNAs implyingmiR-16 and miR-559 as potential biomarkers of RSA in infected women. Furthermore, network of genes of differentially expressed miRNAs regulates RSA by targeting gene function in apoptosis, cell adhesion and angiogenesis.
Collapse
Affiliation(s)
- Ankita Ray
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box No. 4909, New Delhi, 110029, India
| | - Tanu Bhati
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box No. 4909, New Delhi, 110029, India
| | - Renu Arora
- Department of Obstetrics and Gynaecology, Vardhman Mahavir Medical College (VMMC) and Safdarjung Hospital, New Delhi, 110029, India
| | - Dibyabhaba Pradhan
- ICMR Computational Genomics Centre, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi, 110062, India
| | - Sangita Rastogi
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box No. 4909, New Delhi, 110029, India.
| |
Collapse
|
13
|
Inhibiting miR-129-5p alleviates inflammation and modulates autophagy by targeting ATG14 in fungal keratitis. Exp Eye Res 2021; 211:108731. [PMID: 34411602 DOI: 10.1016/j.exer.2021.108731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 07/16/2021] [Accepted: 08/13/2021] [Indexed: 01/04/2023]
Abstract
To investigate the role of miR-129-5p in inflammation and autophagy in fungal keratitis, we established a keratitis mouse model infected with Fusarium solani (F. solani) and conducted experiments on corneal stromal cells infected with F. solani. The expression of miR-129-5p was detected via quantitative real-time polymerase chain reaction (PCR). The miR-129-5p antagomir was used to transfect cells and mice to study the regulatory role of miR-129-5p in autophagy and inflammation after fungal infection. The expression of Beclin1 and LC3B and colocalization of LC3B with lysosomes were detected via Western blotting and immunofluorescence. CCK-8 was used to determine the viability of corneal stromal cells. The expression of IL-1β were detected by ELISA. Bioinformatics software was used to predict the potential targets of miR-129-5p, which were verified by a luciferase reporter gene assay. RT-PCR showed that miR-129-5p expression in mouse corneas was significantly increased after infection with F. solani. Subconjunctival injection of the miR-129-5p antagomir significantly enhanced the proteins Beclin-1 and LC3B. At the same time, inhibiting miR-129-5p expression could reduce the inflammatory response in FK and significantly increase the viability of corneal stromal cells infected with F. solan. Moreover, the dual luciferase reporter assay indicated that Atg14 was a direct target of miR-129-5p. Our study shows that miR-129-5p is a novel small molecule that regulates autophagy by targeting Atg14, indicating that it may be a proinflammatory and therapeutic target for fungal keratitis.
Collapse
|
14
|
Lundy SR, Abney K, Ellerson D, Igietseme JU, Carroll D, Eko FO, Omosun YO. MiR-378b Modulates Chlamydia-Induced Upper Genital Tract Pathology. Pathogens 2021; 10:566. [PMID: 34067003 PMCID: PMC8151610 DOI: 10.3390/pathogens10050566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 04/17/2023] Open
Abstract
Genital Chlamydia trachomatis infection causes severe reproductive pathologies such as salpingitis and pelvic inflammatory disease that can lead to tubal factor infertility. MicroRNAs (miRNAs) are evolutionarily conserved regulators of mammalian gene expression in development, immunity and pathophysiologic processes during inflammation and infection, including Chlamydia infection. Among the miRNAs involved in regulating host responses and pathologic outcome of Chlamydia infection, we have shown that miR-378b was significantly differentially expressed during primary infection and reinfection. In this study, we tested the hypothesis that miR-378b is involved in the pathological outcome of Chlamydia infection. We developed miR-378b knockout mice (miR-378b-/-) using Crispr/Cas and infected them along with their wild-type (WT) control with Chlamydia to compare the infectivity and reproductive pathologies. The results showed that miR-378b-/- mice were unable to clear the infection compared to WT mice; also, miR-378b-/- mice exhibited a relatively higher Chlamydia burden throughout the duration of infection. However, gross pathology results showed that miR-378b-/- mice had significantly reduced uterine dilatations and pathologic lesions after two infections compared to WT mice. In addition, the pregnancy and fertility rates for infected miR-378b-/- mice showed protection from Chlamydia-induced infertility with fertility rate that was comparable to uninfected WT mice. These results are intriguing as they suggest that miR-378b is important in regulating host immune responses that control Chlamydial replication and drive the inflammation that causes complications such as infertility. The finding has important implications for biomarkers of Chlamydial complications and targets for prevention of disease.
Collapse
Affiliation(s)
- Stephanie R. Lundy
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (S.R.L.); (K.A.); (J.U.I.); (F.O.E.)
| | - Kobe Abney
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (S.R.L.); (K.A.); (J.U.I.); (F.O.E.)
- Department of Chemistry and Biochemistry, Spelman College, Atlanta, GA 30310, USA
| | - Debra Ellerson
- Centers for Disease Control & Prevention (CDC), Atlanta, GA 30333, USA; (D.E.); (D.C.)
| | - Joseph U. Igietseme
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (S.R.L.); (K.A.); (J.U.I.); (F.O.E.)
- Centers for Disease Control & Prevention (CDC), Atlanta, GA 30333, USA; (D.E.); (D.C.)
| | - Darin Carroll
- Centers for Disease Control & Prevention (CDC), Atlanta, GA 30333, USA; (D.E.); (D.C.)
| | - Francis O. Eko
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (S.R.L.); (K.A.); (J.U.I.); (F.O.E.)
| | - Yusuf O. Omosun
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (S.R.L.); (K.A.); (J.U.I.); (F.O.E.)
- Centers for Disease Control & Prevention (CDC), Atlanta, GA 30333, USA; (D.E.); (D.C.)
| |
Collapse
|
15
|
de la Maza LM, Darville TL, Pal S. Chlamydia trachomatis vaccines for genital infections: where are we and how far is there to go? Expert Rev Vaccines 2021; 20:421-435. [PMID: 33682583 DOI: 10.1080/14760584.2021.1899817] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen in the world. Antibiotic treatment does not prevent against reinfection and a vaccine is not yet available. AREAS COVERED We focus the review on the progress made of our understanding of the immunological responses required for a vaccine to elicit protection, and on the antigens, adjuvants, routes of immunization and delivery systems that have been tested in animal models. PubMed and Google Scholar were used to search publication on these topics for the last 5 years and recent Reviews were examined. EXPERT OPINION The first Phase 1 clinical trial of a C. trachomatis vaccine to protect against genital infections was successfully completed. We expect that, in the next five years, additional vaccine clinical trials will be implemented.
Collapse
Affiliation(s)
- Luis M de la Maza
- Department of Pathology and Laboratory Medicine Medical Sciences, I, Room D440 University of California, Irvine, California, USA
| | - Toni L Darville
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine Medical Sciences, I, Room D440 University of California, Irvine, California, USA
| |
Collapse
|
16
|
Keck J, Chambers JP, Yu JJ, Cheng X, Christenson LK, Guentzel MN, Gupta R, Arulanandam BP. Modulation of Immune Response to Chlamydia muridarum by Host miR-135a. Front Cell Infect Microbiol 2021; 11:638058. [PMID: 33928045 PMCID: PMC8076868 DOI: 10.3389/fcimb.2021.638058] [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: 12/04/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Previously, our laboratory established the role of small, noncoding RNA species, i.e., microRNA (miRNA) including miR-135a in anti-chlamydial immunity in infected hosts. We report here chlamydial infection results in decreased miR-135a expression in mouse genital tissue and a fibroblast cell line. Several chemokine and chemokine receptor genes (including CXCL10, CCR5) associated with chlamydial pathogenesis were identified in silico to contain putative miR-135a binding sequence(s) in the 3' untranslated region. The role of miR-135a in the host immune response was investigated using exogenous miR-135a mimic to restore the immune phenotype associated with decreased miR-135a following Chlamydia muridarum (Cm) infection. We observed miR-135a regulation of Cm-primed bone marrow derived dendritic cells (BMDC) via activation of Cm-immune CD4+ T cells for clonal expansion and CCR5 expression. Using a transwell cell migration assay, we explore the role of miR-135a in regulation of genital tract CXCL10 expression and recruitment of CXCR3+ CD4+ T cells via the CXCL10/CXCR3 axis. Collectively, data reported here support miR-135a affecting multiple cellular processes in response to chlamydial infection.
Collapse
Affiliation(s)
- Jonathon Keck
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - James P Chambers
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Xingguo Cheng
- Department of Materials & Bioengineering, Southwest Research Institute, San Antonio, TX, United States
| | - Lane K Christenson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - M N Guentzel
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Bernard P Arulanandam
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
17
|
Howard S, Richardson S, Benyeogor I, Omosun Y, Dye K, Medhavi F, Lundy S, Adebayo O, Igietseme JU, Eko FO. Differential miRNA Profiles Correlate With Disparate Immunity Outcomes Associated With Vaccine Immunization and Chlamydial Infection. Front Immunol 2021; 12:625318. [PMID: 33692799 PMCID: PMC7937703 DOI: 10.3389/fimmu.2021.625318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Vaccine-induced immune responses following immunization with promising Chlamydia vaccines protected experimental animals from Chlamydia-induced upper genital tract pathologies and infertility. In contrast, primary genital infection with live Chlamydia does not protect against these pathologies. We hypothesized that differential miRNA profiles induced in the upper genital tracts (UGT) of mice correlate with the disparate immunity vs. pathologic outcomes associated with vaccine immunization and chlamydial infection. Thus, miRNA expression profiles in the UGT of mice after Chlamydia infection (Live EB) and immunization with dendritic cell (DC)-based vaccine (DC vaccine) or VCG-based vaccine (VCG vaccine) were compared using the NanoString nCounter Mouse miRNA assay. Of the 602 miRNAs differentially expressed (DE) in the UGT of immunized and infected mice, we selected 58 with counts >100 and p-values < 0.05 for further analysis. Interestingly, vaccine immunization and Chlamydia infection induced the expression of distinct miRNA profiles with a higher proportion in vaccine-immunized compared to Chlamydia infected mice; DC vaccine (41), VCG vaccine (23), and Live EB (15). Hierarchical clustering analysis showed notable differences in the uniquely DE miRNAs for each experimental group, with DC vaccine showing the highest number (21 up-regulated, five down-regulated), VCG vaccine (two up-regulated, five down-regulated), and live EB (two up-regulated, four down-regulated). The DC vaccine-immunized group showed the highest number (21 up-regulated and five down-regulated compared to two up-regulated and four down-regulated in the live Chlamydia infected group). Pathway analysis showed that the DE miRNAs target genes that regulate several biological processes and functions associated with immune response and inflammation. These results suggest that the induction of differential miRNA expression plays a significant role in the disparate immunity outcomes associated with Chlamydia infection and vaccination.
Collapse
Affiliation(s)
- Simone Howard
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Shakyra Richardson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Ifeyinwa Benyeogor
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kamran Dye
- Department of Chemistry, Morehouse College, Atlanta, GA, United States
| | - Fnu Medhavi
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Stephanie Lundy
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Olayinka Adebayo
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Joseph U. Igietseme
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
18
|
Endocervical miRNA Expression Profiles in Women Positive for Chlamydia trachomatis with Clinical Signs and/or Symptoms Are Distinct from Those in Women Positive for Chlamydia trachomatis without Signs and Symptoms. Infect Immun 2020; 88:IAI.00057-20. [PMID: 32690634 DOI: 10.1128/iai.00057-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/10/2020] [Indexed: 12/26/2022] Open
Abstract
Chlamydia trachomatis is the leading cause of sexually transmitted infections that may progress to pelvic inflammatory disease and infertility. No effective vaccine exists for Chlamydia, nor are there biomarkers available that readily predict disease progression. In this cross-sectional pilot study, we recruited symptomatic and asymptomatic women with C. trachomatis (CT) infection and asymptomatic, uninfected control women from an urban sexually transmitted disease clinic to determine if there were differences in microRNA (miRNA) expression. Infected women with signs and/or symptoms (CTSS) have distinct miRNA profiles compared to asymptomatic infected women (CTNS). In the CTSS group, miR-142 and -147 showed 2.2- to 6.9-fold increases in expression. In the CTNS group, miR-449c, -6779, -519d, -449a, and -2467 showed 3.9- to 9.0-fold increases in expression. In the CTNS group, cyclins and cell cycle regulation and IL-17 pathways were likely downregulated, while the same signaling pathways were upregulated in the CTSS group. In addition, in the CTSS group, additional inflammatory pathways associated with TNFR1 and IL-8 appear to be upregulated. The miRNA expression patterns differ between CT-infected symptomatic and asymptomatic women, and these differences may warrant further study.
Collapse
|
19
|
Ruiz-Roso MB, Gil-Zamorano J, López de Las Hazas MC, Tomé-Carneiro J, Crespo MC, Latasa MJ, Briand O, Sánchez-López D, Ortiz AI, Visioli F, Martínez JA, Dávalos A. Intestinal Lipid Metabolism Genes Regulated by miRNAs. Front Genet 2020; 11:707. [PMID: 32742270 PMCID: PMC7366872 DOI: 10.3389/fgene.2020.00707] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) crucial roles in translation repression and post-transcriptional adjustments contribute to regulate intestinal lipid metabolism. Even though their actions in different metabolic tissues have been elucidated, their intestinal activity is yet unclear. We aimed to investigate intestinal miRNA-regulated lipid metabolism-related genes, by creating an intestinal-specific Dicer1 knockout (Int-Dicer1 KO) mouse model, with a depletion of microRNAs in enterocytes. The levels of 83 cholesterol and lipoprotein metabolism-related genes were assessed in the intestinal mucosa of Int-Dicer1 KO and Wild Type C57BL/6 (WT) littermates mice at baseline and 2 h after an oral lipid challenge. Among the 18 genes selected for further validation, Hmgcs2, Acat1 and Olr1 were found to be strong candidates to be modulated by miRNAs in enterocytes and intestinal organoids. Moreover, we report that intestinal miRNAs contribute to the regulation of intestinal epithelial differentiation. Twenty-nine common miRNAs found in the intestines were analyzed for their potential to target any of the three candidate genes found and validated by miRNA-transfection assays in Caco-2 cells. MiR-31-5p, miR-99b-5p, miR-200a-5p, miR-200b-5p and miR-425-5p are major regulators of these lipid metabolism-related genes. Our data provide new evidence on the potential of intestinal miRNAs as therapeutic targets in lipid metabolism-associated pathologies.
Collapse
Affiliation(s)
- María Belén Ruiz-Roso
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Judit Gil-Zamorano
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - María Carmen López de Las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Joao Tomé-Carneiro
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - María Carmen Crespo
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - María Jesús Latasa
- Research Program, Innovation, Communication and Education Program, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Olivier Briand
- University of Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Daniel Sánchez-López
- University of Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Ana I Ortiz
- Servicio de Cirugía Experimental, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Francesco Visioli
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain.,Department of Molecular Medicine, University of Padua, Padua, Italy
| | - J Alfredo Martínez
- Department of Nutrition and Physiology, Center for Nutrition Research, University of Navarra, IDISNA Navarra, Pamplona, Spain.,Centre of Biomedical Research in Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.,Cardiometabolic Nutrition Group, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| |
Collapse
|
20
|
Wang J, Wang Y, Zhang H, Gao W, Lu M, Liu W, Li Y, Yin Z. Forkhead box C1 promotes the pathology of osteoarthritis by upregulating β-catenin in synovial fibroblasts. FEBS J 2019; 287:3065-3087. [PMID: 31837247 DOI: 10.1111/febs.15178] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/17/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a joint disease characterized by articular cartilage degeneration, and no effective treatment is available. The OA classification has shifted from a cartilage-only disease to a whole-joint disease, and the synovial membrane plays an important role. Therefore, studies are needed to identify additional genes that regulate the pathological changes in the synovial membrane to develop a promising therapeutic strategy for OA. Here, we validated that the expression of forkhead box protein C1 (FoxC1) and β-catenin was upregulated in OA synovial membranes and synovial fibroblasts (SFs). Gain- and loss-of-function studies revealed that FoxC1 overexpression promoted, whilst silencing inhibited OA synovial fibroblast (OASF) proliferation and pro-inflammatory cytokine [interleukin 6 (IL-6), interleukin 8 (IL-8) and tumour necrosis factor-α (TNF-α)] production. FoxC1 overexpression increased β-catenin mRNA, total and nuclear protein expression in OASFs and upregulated a disintegrin and metalloproteinase with thrombospondin motif 5 (ADAMTS-5), fibronectin, matrix metalloproteinase 3 (MMP3) and matrix metalloproteinase 13 (MMP13) mRNA and total protein expression in OASFs. Conversely, FoxC1 knockdown reduced β-catenin mRNA, total and nuclear protein expression in OASFs and reduced ADAMTS-5, fibronectin, MMP3 and MMP13 mRNA and total protein expression in OASFs. β-catenin mediates FoxC1-induced pathological changes (proliferation, catabolic regulation and inflammation) in OASFs. MicroRNA-200a-3p (miR-200a-3p) binds to the 3'-UTR of FoxC1 and mediates FoxC1 expression. Intra-articular FoxC1-specific siRNA transfection hindered OA development in mice. Therefore, our results demonstrate the key role FoxC1 plays in vivo and in vitro in OA synovial pathology, possibly identifying a potential novel therapeutic target for OA.
Collapse
Affiliation(s)
- Jun Wang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, China
| | - Yin Wang
- Department of Plastic Surgery, The Fourth Affiliated Hospital of Anhui Medical University, China
| | - Hui Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, China
| | - Weilu Gao
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, China
| | - Ming Lu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, China
| | - Wendong Liu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, China
| | - Yetian Li
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, China
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, China
| |
Collapse
|
21
|
Wu H, Bao Y, Wang L, Li X, Sun J. Mycobacterium marinum down-regulates miR-148a in macrophages in an EsxA-dependent manner. Int Immunopharmacol 2019; 73:41-48. [PMID: 31078924 DOI: 10.1016/j.intimp.2019.04.056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/08/2019] [Accepted: 04/25/2019] [Indexed: 12/13/2022]
Abstract
As a key virulence factor of Mycobacterium tuberculosis, EsxA is not only involved in phagosome rupture, but also functions in stimulation of immune responses in macrophages. Here, we report thatmiR-148a is down-regulated in the macrophages infected with Mycobacterium marinum (Mm). Using the knockout strain Mm∆EsxA/B, recombinant EsxA, EsxB and EsxA/B heterodimer proteins, we provide evidence that down-regulation of miR-148ais dependent on EsxA, and up-regulation of miR-148a reduces Mm intracellular survival. Moreover, up-regulation of miR-148a down-regulates the pro-inflammatory cytokines (e.g. TNF-α and IL-1β) and the TLR4-mediated NF-κB activation. Together, miR-148a may function as an anti-inflammation modulator in responses to mycobacterial infection. Regulation of miR-148a may provide a novel venue in development of therapies in tuberculosis.
Collapse
Affiliation(s)
- Haichong Wu
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA; Current address: Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Yanqing Bao
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | - Lin Wang
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | - Xiujun Li
- Department of Chemistry, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | - Jianjun Sun
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA.
| |
Collapse
|
22
|
Aguilar C, Mano M, Eulalio A. Multifaceted Roles of microRNAs in Host-Bacterial Pathogen Interaction. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0002-2019. [PMID: 31152522 PMCID: PMC11026079 DOI: 10.1128/microbiolspec.bai-0002-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a well-characterized class of small noncoding RNAs that act as major posttranscriptional regulators of gene expression. Accordingly, miRNAs have been associated with a wide range of fundamental biological processes and implicated in human diseases. During the past decade, miRNAs have also been recognized for their role in the complex interplay between the host and bacterial pathogens, either as part of the host response to counteract infection or as a molecular strategy employed by bacteria to subvert host pathways for their own benefit. Importantly, the characterization of downstream miRNA targets and their underlying mechanisms of action has uncovered novel molecular factors and pathways relevant to infection. In this article, we review the current knowledge of the miRNA response to bacterial infection, focusing on different bacterial pathogens, including Salmonella enterica, Listeria monocytogenes, Mycobacterium spp., and Helicobacter pylori, among others.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-Based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
- RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
23
|
Benyeogor I, Simoneaux T, Wu Y, Lundy S, George Z, Ryans K, McKeithen D, Pais R, Ellerson D, Lorenz WW, Omosun T, Thompson W, Eko FO, Black CM, Blas-Machado U, Igietseme JU, He Q, Omosun Y. A unique insight into the MiRNA profile during genital chlamydial infection. BMC Genomics 2019; 20:143. [PMID: 30777008 PMCID: PMC6379932 DOI: 10.1186/s12864-019-5495-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
Background Genital C. trachomatis infection may cause pelvic inflammatory disease (PID) that can lead to tubal factor infertility (TFI). Understanding the pathogenesis of chlamydial complications including the pathophysiological processes within the female host genital tract is important in preventing adverse pathology. MicroRNAs regulate several pathophysiological processes of infectious and non-infectious etiologies. In this study, we tested the hypothesis that the miRNA profile of single and repeat genital chlamydial infections will be different and that these differences will be time dependent. Thus, we analyzed and compared differentially expressed mice genital tract miRNAs after single and repeat chlamydia infections using a C. muridarum mouse model. Mice were sacrificed and their genital tract tissues were collected at 1, 2, 4, and 8 weeks after a single and repeat chlamydia infections. Histopathology, and miRNA sequencing were performed. Results Histopathology presentation showed that the oviduct and uterus of reinfected mice were more inflamed, distended and dilated compared to mice infected once. The miRNAs expression profile was different in the reproductive tissues after a reinfection, with a greater number of miRNAs expressed after reinfection. Also, the number of miRNAs expressed each week after chlamydia infection and reinfection varied, with weeks eight and one having the highest number of differentially expressed miRNAs for chlamydia infection and reinfection respectively. Ten miRNAs; mmu-miR-378b, mmu-miR-204-5p, mmu-miR-151-5p, mmu-miR-142-3p, mmu-miR-128-3p, mmu-miR-335-3p, mmu-miR-195a-3p, mmu-miR-142-5p, mmu-miR-106a-5p and mmu-miR-92a-3p were common in both primary chlamydia infection and reinfection. Pathway analysis showed that, amongst other functions, the differentially regulated miRNAs control pathways involved in cellular and tissue development, disease conditions and toxicity. Conclusions This study provides insights into the changes in miRNA expression over time after chlamydia infection and reinfection, as well as the pathways they regulate to determine pathological outcomes. The miRNAs networks generated in our study shows that there are differences in the focus molecules involved in significant biological functions in chlamydia infection and reinfection, implying that chlamydial pathogenesis occurs differently for each type of infection and that this could be important when determining treatments regime and disease outcome. The study underscores the crucial role of host factors in chlamydia pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12864-019-5495-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ifeyinwa Benyeogor
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Tankya Simoneaux
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Yuehao Wu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Stephanie Lundy
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Zenas George
- Centers for Disease Control & Prevention (CDC), Atlanta, GA, 30333, USA
| | - Khamia Ryans
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Danielle McKeithen
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Roshan Pais
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Debra Ellerson
- Centers for Disease Control & Prevention (CDC), Atlanta, GA, 30333, USA
| | - W Walter Lorenz
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Tolulope Omosun
- Department of Physical Sciences, Georgia State University, Covington, GA, 30014, USA
| | - Winston Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Francis O Eko
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA
| | - Carolyn M Black
- Centers for Disease Control & Prevention (CDC), Atlanta, GA, 30333, USA
| | - Uriel Blas-Machado
- Department of Pathology, University of Georgia, College of Veterinary Medicine, Athens, GA, 30602, USA
| | - Joseph U Igietseme
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA.,Centers for Disease Control & Prevention (CDC), Atlanta, GA, 30333, USA
| | - Qing He
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA.,Centers for Disease Control & Prevention (CDC), Atlanta, GA, 30333, USA
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, 720 Westview Drive, S.W, Atlanta, GA, 30310, USA. .,Centers for Disease Control & Prevention (CDC), Atlanta, GA, 30333, USA.
| |
Collapse
|
24
|
Poston TB, O'Connell CM, Girardi J, Sullivan JE, Nagarajan UM, Marinov A, Scurlock AM, Darville T. T Cell-Independent Gamma Interferon and B Cells Cooperate To Prevent Mortality Associated with Disseminated Chlamydia muridarum Genital Tract Infection. Infect Immun 2018; 86:e00143-18. [PMID: 29661927 PMCID: PMC6013674 DOI: 10.1128/iai.00143-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
CD4 T cells and antibody are required for optimal acquired immunity to Chlamydia muridarum genital tract infection, and T cell-mediated gamma interferon (IFN-γ) production is necessary to clear infection in the absence of humoral immunity. However, the role of T cell-independent immune responses during primary infection remains unclear. We investigated this question by inoculating wild-type and immune-deficient mice with C. muridarum CM001, a clonal isolate capable of enhanced extragenital replication. Genital inoculation of wild-type mice resulted in transient dissemination to the lungs and spleen that then was rapidly cleared from these organs. However, CM001 genital infection proved lethal for STAT1-/- and IFNG-/- mice, in which IFN-γ signaling was absent, and for Rag1-/- mice, which lacked T and B cells and in which innate IFN-γ signaling was retained. In contrast, B cell-deficient muMT mice, which can generate a Th1 response, and T cell-deficient mice with intact B cell and innate IFN-γ signaling survived. These data collectively indicate that IFN-γ prevents lethal CM001 dissemination in the absence of T cells and suggests a B cell corequirement. Adoptive transfer of convalescent-phase immune serum but not naive IgM to Rag1-/- mice infected with CM001 significantly increased the survival time, while transfer of naive B cells completely rescued Rag1-/- mice from CM001 lethality. Protection was associated with a significant reduction in the lung chlamydial burden of genitally infected mice. These data reveal an important cooperation between T cell-independent B cell responses and innate IFN-γ in chlamydial host defense and suggest that interactions between T cell-independent antibody and IFN-γ are essential for limiting extragenital dissemination.
Collapse
Affiliation(s)
- Taylor B Poston
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Catherine M O'Connell
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jenna Girardi
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jeanne E Sullivan
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Uma M Nagarajan
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Anthony Marinov
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy M Scurlock
- Department of Pediatrics, Arkansas Children's Hospital, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Toni Darville
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
25
|
Eledge MR, Yeruva L. Host and pathogen interface: microRNAs are modulators of disease outcome. Microbes Infect 2017; 20:410-415. [PMID: 28889971 DOI: 10.1016/j.micinf.2017.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 12/17/2022]
Abstract
Chlamydiae are a group of intracellular bacterium that infect a range of hosts and are responsible for the most common sexual transmitted infections, which could be the result of a plethora of factors leading to varied pathological outcomes. This review aims to show that Chlamydia possibly manipulates host defenses through microRNAs interaction.
Collapse
Affiliation(s)
- Michael R Eledge
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Arkansas Children's Research Institute, Little Rock, AR, USA; Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Laxmi Yeruva
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Arkansas Children's Research Institute, Little Rock, AR, USA; Arkansas Children's Nutrition Center, Little Rock, AR, USA.
| |
Collapse
|
26
|
Keck J, Gupta R, Christenson LK, Arulanandam BP. MicroRNA mediated regulation of immunity against gram-negative bacteria. Int Rev Immunol 2017; 36:287-299. [PMID: 28800263 PMCID: PMC6904929 DOI: 10.1080/08830185.2017.1347649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Evidence over the last couple decades has comprehensively established that short, highly conserved, non-coding RNA species called microRNA (miRNA) exhibit the ability to regulate expression and function of host genes at the messenger RNA (mRNA) level. MicroRNAs play key regulatory roles in immune cell development, differentiation, and protective function. Intrinsic host immune response to invading pathogens rely on intricate orchestrated events in the development of innate and adaptive arms of immunity. We discuss the involvement of miRNAs in regulating these processes against gram negative pathogens in this review.
Collapse
Affiliation(s)
- Jonathon Keck
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| | - Lane K. Christenson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| |
Collapse
|