1
|
Valero P, Silva K, Valenzuela-Hinrichsen A, Vásquez A, Espinoza F, Lira F, Cornejo M, Fuentes G, González D, Moore-Carrasco R, van der Beek EM, Hillebrands JL, van Goor H, Grismaldo A, Sobrevia L. Shortcomings, limitations and gaps in physiological roles of extracellular vesicles in obesity. J Physiol 2024. [PMID: 39470472 DOI: 10.1113/jp286955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in mediating communication between cells across species and kingdoms. The intercellular communication facilitated by EVs through autocrine and paracrine signalling mechanisms is essential for cell survival, maintaining normal metabolic functions and ensuring overall bodily homeostasis and health. Extracellular vesicles are present in various bodily fluids, such as pleural effusions, plasma, breast milk, amniotic fluid, semen and saliva. Additionally, the generation and release of EVs contribute to the removal of cellular waste. Patients with obesity exhibit a higher release and amount of circulating EVs than individuals with normal weight. This increased EV release in obesity might contribute to the inflammatory state characteristic of this metabolic condition, because higher levels of pro-inflammatory molecules are found within their cargo. However, interpreting results related to EV abundance, cargo and biological actions can be complicated by several factors; these include variations in cell sources, a wide age range (from children to the elderly), a mix of females and males, medication use and health status, a range of body weights (from normal weight to morbid obesity) and differences between in vitro assays using cell lines versus primary cultures. This article addresses the shortcomings, limitations and gaps in knowledge, providing a framework for enhancing our understanding of the physiological effects of EVs on obesity.
Collapse
Affiliation(s)
- Paola Valero
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Katherin Silva
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute of Chemistry, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Andrés Valenzuela-Hinrichsen
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonia Vásquez
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernanda Espinoza
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernanda Lira
- Faculty of Medicine, Universidad de Antofagasta, Antofagasta, Chile
| | - Marcelo Cornejo
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta, Chile
| | - Gonzalo Fuentes
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Daniel González
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | | | - Eline M van der Beek
- Department of Pediatrics, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
- Nestlé Institute for Health Sciences, Nestlé Research, Societé des Produits de Nestlé, Lausanne, Switzerland
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Adriana Grismaldo
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- **Faculty of Excellence program, School of Medicine and Health Sciences, The Institute for Obesity Research (IOR), Eutra, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
| | - Luis Sobrevia
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- **Faculty of Excellence program, School of Medicine and Health Sciences, The Institute for Obesity Research (IOR), Eutra, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
2
|
Mukhtar F, Guarnieri A, Brancazio N, Falcone M, Di Naro M, Azeem M, Zubair M, Nicolosi D, Di Marco R, Petronio Petronio G. The role of Mycobacterium tuberculosis exosomal miRNAs in host pathogen cross-talk as diagnostic and therapeutic biomarkers. Front Microbiol 2024; 15:1441781. [PMID: 39176271 PMCID: PMC11340542 DOI: 10.3389/fmicb.2024.1441781] [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: 05/31/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Tuberculosis (TB) is a global threat, affecting one-quarter of the world's population. The World Health Organization (WHO) reports that 6 million people die annually due to chronic illnesses, a statistic that includes TB-related deaths. This high mortality is attributed to factors such as the emergence of drug-resistant strains and the exceptional survival mechanisms of Mycobacterium tuberculosis (MTB). Recently, microRNAs (miRNAs) have garnered attention for their crucial role in TB pathogenesis, surpassing typical small RNAs (sRNA) in their ability to alter the host's immune response. For instance, miR-155, miR-125b, and miR-29a have been identified as key players in the immune response to MTB, particularly in modulating macrophages, T cells, and cytokine production. While sRNAs are restricted to within cells, exo-miRNAs are secreted from MTB-infected macrophages. These exo-miRNAs modify the function of surrounding cells to favor the bacterium, perpetuating the infection cycle. Another significant aspect is that the expression of these miRNAs affects specific genes and pathways involved in immune functions, suggesting their potential use in diagnosing TB and as therapeutic targets. This review compiles existing information on the immunomodulatory function of exosomal miRNAs from MTB, particularly focusing on disease progression and the scientific potential of this approach compared to existing diagnostic techniques. Thus, the aim of the study is to understand the role of exosomal miRNAs in TB and to explore their potential for developing novel diagnostic and therapeutic methods.
Collapse
Affiliation(s)
- Farwa Mukhtar
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| | - Antonio Guarnieri
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| | - Natasha Brancazio
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| | - Marilina Falcone
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| | - Maria Di Naro
- Department of Drug and Health Sciences, Università degli Studi di Catania, Catania, Italy
| | - Muhammad Azeem
- Department of Precision Medicine in the Medical, Surgical and Critical Care Area (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Muhammad Zubair
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Daria Nicolosi
- Department of Drug and Health Sciences, Università degli Studi di Catania, Catania, Italy
| | - Roberto Di Marco
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| | - Giulio Petronio Petronio
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| |
Collapse
|
3
|
Sun X, Li W, Zhao L, Fan K, Qin F, Shi L, Gao F, Zheng C. Current landscape of exosomes in tuberculosis development, diagnosis, and treatment applications. Front Immunol 2024; 15:1401867. [PMID: 38846947 PMCID: PMC11153741 DOI: 10.3389/fimmu.2024.1401867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/09/2024] [Indexed: 06/09/2024] Open
Abstract
Tuberculosis (TB), caused by the bacterial pathogen Mycobacterium tuberculosis (MTB), remains one of the most prevalent and deadly infectious diseases worldwide. Currently, there are complex interactions between host cells and pathogens in TB. The onset, progression, and regression of TB are correlated not only with the virulence of MTB but also with the immunity of TB patients. Exosomes are cell-secreted membrane-bound nanovesicles with lipid bilayers that contain a variety of biomolecules, such as metabolites, lipids, proteins, and nucleic acids. Exosome-mediated cell-cell communication and interactions with the microenvironment represent crucial mechanisms through which exosomes exert their functional effects. Exosomes harbor a wide range of regulatory roles in physiological and pathological conditions, including MTB infection. Exosomes can regulate the immune response, metabolism, and cellular death to remodel the progression of MTB infection. During MTB infection, exosomes display distinctive profiles and quantities that may act as diagnostic biomarkers, suggesting that exosomes provide a revealing glimpse into the evolving landscape of MTB infections. Furthermore, exosomes derived from MTB and mesenchymal stem cells can be harnessed as vaccine platforms and drug delivery vehicles for the precise targeting and treatment of TB. In this review, we highlight the functions and mechanisms through which exosomes influence the progression of TB. Additionally, we unravel the critical significance of exosomal constituents in the diagnosis and therapeutic applications of TB, aiming to offer novel perspectives and strategies for combating TB.
Collapse
Affiliation(s)
- Xuezhi Sun
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Wei Li
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Li Zhao
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Ke Fan
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Fenfen Qin
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Liwen Shi
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunlan Zheng
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| |
Collapse
|
4
|
Zhang L, Chi J, Wu H, Xia X, Xu C, Hao H, Liu Z. Extracellular vesicles and endothelial dysfunction in infectious diseases. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e148. [PMID: 38938849 PMCID: PMC11080793 DOI: 10.1002/jex2.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 06/29/2024]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of mortality and morbidity globally. Studies have shown that infections especially bacteraemia and sepsis are associated with increased risks for endothelial dysfunction and related CVDs including atherosclerosis. Extracellular vesicles (EVs) are small, sealed membrane-derived structures that are released into body fluids and blood from cells and/or microbes and are critically involved in a variety of important cell functions and disease development, including intercellular communications, immune responses and inflammation. It is known that EVs-mediated mechanism(s) is important in the development of endothelial dysfunction in infections with a diverse spectrum of microorganisms including Escherichia coli, Candida albicans, SARS-CoV-2 (the virus for COVID-19) and Helicobacter pylori. H. pylori infection is one of the most common infections globally. During H. pylori infection, EVs can carry H. pylori components, such as lipopolysaccharide, cytotoxin-associated gene A, or vacuolating cytotoxin A, and transfer these substances into endothelial cells, triggering inflammatory responses and endothelial dysfunction. This review is to illustrate the important role of EVs in the pathogenesis of infectious diseases, and the development of endothelial dysfunction in infectious diseases especially H. pylori infection, and to discuss the potential mechanisms and clinical implications.
Collapse
Affiliation(s)
- Linfang Zhang
- Department of GastroenterologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
- Center for Precision Medicine and Division of Cardiovascular MedicineDepartment of MedicineUniversity of Missouri School of MedicineColumbiaMissouriUSA
| | - Jingshu Chi
- Center for Precision Medicine and Division of Cardiovascular MedicineDepartment of MedicineUniversity of Missouri School of MedicineColumbiaMissouriUSA
- Department of Gastroenterologythe Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Hao Wu
- Center for Precision Medicine and Division of Cardiovascular MedicineDepartment of MedicineUniversity of Missouri School of MedicineColumbiaMissouriUSA
| | - Xiujuan Xia
- Center for Precision Medicine and Division of Cardiovascular MedicineDepartment of MedicineUniversity of Missouri School of MedicineColumbiaMissouriUSA
| | - Canxia Xu
- Department of Gastroenterologythe Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular MedicineDepartment of MedicineUniversity of Missouri School of MedicineColumbiaMissouriUSA
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular MedicineDepartment of MedicineUniversity of Missouri School of MedicineColumbiaMissouriUSA
| |
Collapse
|
5
|
Abstract
Extracellular vesicles (EVs) are membrane-bound structures released by cells and have become significant players in immune system functioning, primarily by facilitating cell-to-cell communication. Immune cells like neutrophils and dendritic cells release EVs containing bioactive molecules that modulate chemotaxis, activate immune cells, and induce inflammation. EVs also contribute to antigen presentation, lymphocyte activation, and immune tolerance. Moreover, EVs play pivotal roles in antimicrobial host defense. They deliver microbial antigens to antigen-presenting cells (APCs), triggering immune responses, or act as decoys to neutralize virulence factors and toxins. This review discusses host and microbial EVs' multifaceted roles in innate and adaptive immunity, highlighting their involvement in immune cell development, antigen presentation, and antimicrobial responses.
Collapse
Affiliation(s)
- Puja Kumari
- Department of Immunology, University of Connecticut Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Skylar S. Wright
- Department of Immunology, University of Connecticut Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Vijay A. Rathinam
- Department of Immunology, University of Connecticut Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| |
Collapse
|
6
|
Wang N, Yao Y, Qian Y, Qiu D, Cao H, Xiang H, Wang J. Cargoes of exosomes function as potential biomarkers for Mycobacterium tuberculosis infection. Front Immunol 2023; 14:1254347. [PMID: 37928531 PMCID: PMC10622749 DOI: 10.3389/fimmu.2023.1254347] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
Exosomes as double-membrane vesicles contain various contents of lipids, proteins, mRNAs and non-coding RNAs, and involve in multiple physiological processes, for instance intercellular communication and immunomodulation. Currently, numerous studies found that the components of exosomal proteins, nucleic acids or lipids released from host cells are altered following infection with Mycobacterium tuberculosis. Exosomal contents provide excellent biomarkers for the auxiliary diagnosis, efficacy evaluation, and prognosis of tuberculosis. This study aimed to review the current literatures detailing the functions of exosomes in the procedure of M. tuberculosis infection, and determine the potential values of exosomes as biomarkers to assist in the diagnosis and monitoring of tuberculosis.
Collapse
Affiliation(s)
- Nan Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, China
| | - Yongliang Yao
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, China
| | - Yingfen Qian
- Department of Clinical Laboratory, Kunshan Fourth People’s Hospital, Suzhou, Jiangsu, China
| | - Dewen Qiu
- Department of Clinical Laboratory, Jiangxi Maternal and Child Health Hospital Maternal and Child Heath Hospital of Nanchang College, Nanchang, China
| | - Hui Cao
- Department of Food and Nutrition Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Huayuan Xiang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, China
| | - Jianjun Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Liu J, Li Y, Liu T, Shi Y, Wang Y, Wu J, Qi Y. Novel Biomarker Panel of Let-7d-5p and MiR-140-5p Can Distinguish Latent Tuberculosis Infection from Active Tuberculosis Patients. Infect Drug Resist 2023; 16:3847-3859. [PMID: 37346367 PMCID: PMC10281287 DOI: 10.2147/idr.s412116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
Background Mycobacterium tuberculosis (Mtb) survives inside a human host for a long time in the form of latent tuberculosis infection (LTBI). Latent infection of tuberculosis has the opportunity of developing into active tuberculosis (ATB), which has greatly endangered human health. The existing diagnostic methods cannot effectively distinguish LTBI from ATB. Therefore, more effective diagnostic biomarkers and methods are urgently needed. Methods Here, we screened the GEO data set, conducted joint differential analysis and target gene enrichment analysis, after filtering the disease-related database, we screened the differential miRNA related to TB. The qPCR was used to verify the miRNAs in 84 serum samples. Different combinations of biomarkers were evaluated by logistic regression to obtain a biomarker panel with good performance for diagnosing LTBI. Results A panel with two miRNAs (hsa-let-7d-5p, hsa-miR-140-5p) was established to differentiate LTBI from ATB. Receiver operating characteristic (ROC) curve showed that the area under the curve (AUC) are 0.930 (sensitivity = 100%, specificity = 88.5%) and 0.923 (sensitivity = 100%, specificity = 92.3%) with the biomarker panel for the training set and test set respectively. Conclusion The findings indicated that the logistic regression model built by let-7d-5p and miR-140-5p has the ability to distinguish LTBI from active TB patients.
Collapse
Affiliation(s)
- Jiaxing Liu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, People’s Republic of China
| | - Ye Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230000, People’s Republic of China
| | - Ting Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230000, People’s Republic of China
| | - Yuru Shi
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230000, People’s Republic of China
| | - Yun Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230000, People’s Republic of China
| | - Jing Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230000, People’s Republic of China
| | - Yingjie Qi
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230000, People’s Republic of China
| |
Collapse
|
8
|
Wang L, Xiong Y, Fu B, Guo D, Zaky MY, Lin X, Wu H. MicroRNAs as immune regulators and biomarkers in tuberculosis. Front Immunol 2022; 13:1027472. [PMID: 36389769 PMCID: PMC9647078 DOI: 10.3389/fimmu.2022.1027472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 07/26/2023] Open
Abstract
Tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), is one of the most lethal infectious disease worldwide, and it greatly affects human health. Some diagnostic and therapeutic methods are available to effectively prevent and treat TB; however, only a few systematic studies have described the roles of microRNAs (miRNAs) in TB. Combining multiple clinical datasets and previous studies on Mtb and miRNAs, we state that pathogens can exploit interactions between miRNAs and other biomolecules to avoid host mechanisms of immune-mediated clearance and survive in host cells for a long time. During the interaction between Mtb and host cells, miRNA expression levels are altered, resulting in the changes in the miRNA-mediated regulation of host cell metabolism, inflammatory responses, apoptosis, and autophagy. In addition, differential miRNA expression can be used to distinguish healthy individuals, patients with TB, and patients with latent TB. This review summarizes the roles of miRNAs in immune regulation and their application as biomarkers in TB. These findings could provide new opportunities for the diagnosis and treatment of TB.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Yan Xiong
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Beibei Fu
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Dong Guo
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Mohamed Y. Zaky
- Department of Zoology, Molecular Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Xiaoyuan Lin
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Haibo Wu
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
9
|
Simper JD, Perez E, Schlesinger LS, Azad AK. Resistance and Susceptibility Immune Factors at Play during Mycobacterium tuberculosis Infection of Macrophages. Pathogens 2022; 11:1153. [PMID: 36297211 PMCID: PMC9611686 DOI: 10.3390/pathogens11101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB), caused by infection with Mycobacterium tuberculosis (M.tb), is responsible for >1.5 million deaths worldwide annually. Innate immune cells, especially macrophages, are the first to encounter M.tb, and their response dictates the course of infection. During infection, macrophages exert a variety of immune factors involved in either controlling or promoting the growth of M.tb. Research on this topic has been performed in both in vitro and in vivo animal models with discrepant results in some cases based on the model of study. Herein, we review macrophage resistance and susceptibility immune factors, focusing primarily on recent advances in the field. We include macrophage cellular pathways, bioeffector proteins and molecules, cytokines and chemokines, associated microbiological factors and bacterial strains, and host genetic factors in innate immune genes. Recent advances in mechanisms underlying macrophage resistance and susceptibility factors will aid in the successful development of host-directed therapeutics, a topic emphasized throughout this review.
Collapse
Affiliation(s)
- Jan D. Simper
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Department of Microbiology, Immunology and Molecular Genetics, UT Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Esteban Perez
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Translational Sciences Program, UT Health San Antonio Graduate School of Biomedical Sciences, San Antonio, TX 78229, USA
| | - Larry S. Schlesinger
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
| | - Abul K. Azad
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
| |
Collapse
|
10
|
Zhou L, Luo L, Luo L, Ding Y, Lu Z, Feng D, Xiao Y. Macrophage-Secreted Exosomal HCG11 Promotes Autophagy in Antigen 85B-Infected Macrophages and Inhibits Fibroblast Fibrosis to Affect Tracheobronchial Tuberculosis Progression via the miR-601/Sirtuin 1 Axis. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background: Tracheobronchial tuberculosis (TBTB) is a serious threat to human health. We aimed to explore the potential regulatory mechanism by which macrophages secrete exosomes that regulate TBTB progression. Methods: Bioinformatics analysis predicted lncRNAs with low
expression in TBTB. Macrophage-derived exosomes were isolated and identified. HCG11 was knocked down and overexpressed, and miR-601 was overexpressed. ELISA was utilized to measure TGF-β, IL-8, IL-6 and IFN-γ levels. Based on bioinformatics prediction and dual-luciferase
assay analysis, lncRNA HCG11 bound to miR-601, and miR-601 bound to SIRT1. The mRNA or protein expressions of lncRNA HCG11, miR- 601, SIRT1, PI3K/Akt/mTOR pathway-related factors, ATG5 and LC3B, as well as COL-1, MMP2, Timp-1 and Timp-3, were evaluated. Results: HCG11 was expressed
at low levels in TBTB patients. Macrophage-secreted exosomes inhibited Ag85B-induced macrophage proinflammatory response and promoted autophagy. Moreover, normal macrophage (MØ)-exo-derived HCG11 could inhibit Ag85B-induced macrophage proinflammatory response and promote autophagy.
HCG11 bound to miR-601, and miR-601 bound to SIRT1. HCG11 inhibited miR-601 to upregulate SIRT1. In addition, MØ-exo-derived HCG11 reduced Ag85B-induced fibroblast hyperproliferation and extracellular matrix deposition through the miR-601/SIRT1 axis. Conclusion: Macrophage-secreted
exosomal HCG11 promotes autophagy in Ag85B-infected macrophages and inhibits fibroblast fibrosis to affect TBTB progression via the miR-601/SIRT1 axis.
Collapse
Affiliation(s)
- Lei Zhou
- Endoscopy Center, Hunan Chest Hospital, Changsha, 410016, China
| | - Li Luo
- Endoscopy Center, Hunan Chest Hospital, Changsha, 410016, China
| | - Linzi Luo
- Endoscopy Center, Hunan Chest Hospital, Changsha, 410016, China
| | - Yan Ding
- Endoscopy Center, Hunan Chest Hospital, Changsha, 410016, China
| | - Zhibin Lu
- Endoscopy Center, Hunan Chest Hospital, Changsha, 410016, China
| | - Dan Feng
- Endoscopy Center, Hunan Chest Hospital, Changsha, 410016, China
| | - Yangbao Xiao
- Endoscopy Center, Hunan Chest Hospital, Changsha, 410016, China
| |
Collapse
|
11
|
Luo D, Wu J, Liu Y, Li P, Liang X, Xiao S, Qi Z, Liu T, Pan J. Overexpression of VPS11 antagonizes the promoting effect of miR-542-3p on Mycobacterium tuberculosis survival in macrophages by regulating autophagy. Microb Pathog 2022; 169:105609. [PMID: 35662671 DOI: 10.1016/j.micpath.2022.105609] [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: 02/08/2022] [Revised: 05/26/2022] [Accepted: 05/28/2022] [Indexed: 10/18/2022]
Abstract
Impaired autophagy is an important cause of Mycobacterium tuberculosis survival in macrophages. VPS11 is an important regulator of autophagy; decreased VPS11 expression has been observed in macrophages after tuberculosis (TB) infection. Gene ontology data revealed that various miRNAs (for example, miR-542-3p) were upregulated in macrophages upon TB infection; thus, these miRNAs were likely to reduce VPS11 expression. In this study, both TB patients and healthy subjects were enrolled, and the levels of VPS11 and some miRNAs in their blood macrophages were measured. Moreover, various macrophages were cultured and infected with M. tuberculosis. Luciferase reporter, RNA pulldown, and RNA immunoprecipitation assays were performed to determine the regulatory effect of miR-542-3p on VPS11 expression. Results showed that VPS11 expression was downregulated, whereas miR-542-3p expression was upregulated in blood macrophages after TB infection. TB infection reduced VPS11 levels in two human macrophages in vitro, but not in mouse macrophages. This might be because the seed sequence exists in the VPS11 3' untranslated region in humans, but is absent in mice and rats. miR-542-3p promoted M. tuberculosis survival in human macrophages, but VPS11 overexpression antagonized the promoting effect of miR-542-3p. Further, VPS11 was confirmed as a target of miR-542-3p. Overexpression of VPS11 or depletion of miR-542-3p promoted autophagy, which was suppressed upon TB infection. In summary, VPS11 overexpression antagonized the promoting effect of miR-542-3p on M. tuberculosis survival in macrophages by regulating autophagy.
Collapse
Affiliation(s)
- Dan Luo
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Jialing Wu
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Yinyin Liu
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Peng Li
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Xianzhi Liang
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Sifang Xiao
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Zhiqiang Qi
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Ting Liu
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China
| | - Jianhua Pan
- Department of laboratory, Changsha Central Hospital Affiliated to Nanhua University, 410004, China.
| |
Collapse
|
12
|
Zheng L, Wei F, Li G. The crosstalk between bacteria and host autophagy: host defense or bacteria offense. J Microbiol 2022; 60:451-460. [DOI: 10.1007/s12275-022-2009-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/18/2022] [Accepted: 03/29/2022] [Indexed: 12/26/2022]
|
13
|
Mourenza Á, Lorente-Torres B, Durante E, Llano-Verdeja J, Aparicio JF, Fernández-López A, Gil JA, Mateos LM, Letek M. Understanding microRNAs in the Context of Infection to Find New Treatments against Human Bacterial Pathogens. Antibiotics (Basel) 2022; 11:356. [PMID: 35326819 PMCID: PMC8944844 DOI: 10.3390/antibiotics11030356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
The development of RNA-based anti-infectives has gained interest with the successful application of mRNA-based vaccines. Small RNAs are molecules of RNA of <200 nucleotides in length that may control the expression of specific genes. Small RNAs include small interference RNAs (siRNAs), Piwi-interacting RNAs (piRNAs), or microRNAs (miRNAs). Notably, the role of miRNAs on the post-transcriptional regulation of gene expression has been studied in detail in the context of cancer and many other genetic diseases. However, it is also becoming apparent that some human miRNAs possess important antimicrobial roles by silencing host genes essential for the progress of bacterial or viral infections. Therefore, their potential use as novel antimicrobial therapies has gained interest during the last decade. The challenges of the transport and delivery of miRNAs to target cells are important, but recent research with exosomes is overcoming the limitations in RNA-cellular uptake, avoiding their degradation. Therefore, in this review, we have summarised the latest developments in the exosomal delivery of miRNA-based therapies, which may soon be another complementary treatment to pathogen-targeted antibiotics that could help solve the problem caused by multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Álvaro Mourenza
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Blanca Lorente-Torres
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Elena Durante
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- L’Università di Urbino Carlo Bo, Via Aurelio Saffi, 2, 61029 Urbino, Italy
| | - Jesús Llano-Verdeja
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Jesús F. Aparicio
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
| | - Arsenio Fernández-López
- Departamento de Biología Molecular, Área de Biología Celular, Universidad de León, 24071 León, Spain;
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Neural Therapies SL, Campus de Vegazana s/n, 24071 León, Spain
| | - José A. Gil
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Luis M. Mateos
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Michal Letek
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (B.L.-T.); (E.D.); (J.L.-V.); (J.F.A.); (J.A.G.)
- Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
| |
Collapse
|
14
|
Li Y, Sun L, Liu J, Xu G, Hu Y, Qin A. Down-regulation of GAS5 has diagnostic value for tuberculosis and regulates the inflammatory response in mycobacterium tuberculosis infected THP-1 cells. Tuberculosis (Edinb) 2022; 132:102141. [PMID: 34808575 DOI: 10.1016/j.tube.2021.102141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aimed to investigate the expression of long non-coding RNA (lncRNA) growth arrest-special transcript 5 (GAS5) in the serum of tuberculosis (TB) patients and discuss the mechanism of GAS5 in TB by establishing an in-vitro TB cell model. METHODS Serum expressions of GAS5 and miR-18a-5p were determined by quantitative real-time PCR (qRT-PCR). The effects of GAS5 on macrophage cell viability and the inflammatory response after MTB infection were assessed by CCK-8 and ELISA. Luciferase reporter gene assay was applied to delve into the potential target gene of GAS5. RESULTS The expression of GAS5 in TB patients was down-regulated, while miR-18a-5p was up-regulated, and the serum inflammatory factors were negatively correlated with the expression level of GAS5. MTB infection induced significant upregulation on the cell viability and inflammatory response but the acceleration effect could be rescued by GAS5-overexpression. Meanwhile, miR-18a-5p was recognized as the target gene of GAS5. CONCLUSION This study indicated that the expression level of GAS5 in the serum of TB patients was decreased, while in the cells infected with MTB, the down-regulated GAS5 might develop a role in facilitating the cell vitality and the inflammatory response by adsorbing miR-18a-5p in the form of molecular sponge.
Collapse
Affiliation(s)
- Yusong Li
- Department of Laboratory Medicine, The Fourth People's Hospital of Huai'an, Jiangsu, 223001, China
| | - Lihua Sun
- Department of Laboratory Medicine, The Fourth People's Hospital of Huai'an, Jiangsu, 223001, China
| | - Juan Liu
- Department of Laboratory Medicine, The Fourth People's Hospital of Huai'an, Jiangsu, 223001, China
| | - Guoying Xu
- School of Medical Technology, Jiangsu College of Nursing, Jiangsu, 223007, China
| | - Yan Hu
- Clinical Laboratory and Pathology Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, China.
| | - Andong Qin
- Department of Laboratory Medicine, The Fourth People's Hospital of Huai'an, Jiangsu, 223001, China.
| |
Collapse
|
15
|
Ning QY, Liu N, Wu JZ, Hu DF, Wei Q, Zhou JA, Zou J, Zang N, Li GJ. Serum Exosomal microRNA Profiling in AIDS Complicated with Talaromyces marneffei Infection. Infect Drug Resist 2021; 14:4931-4948. [PMID: 34858034 PMCID: PMC8630369 DOI: 10.2147/idr.s338321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/04/2021] [Indexed: 11/24/2022] Open
Abstract
Introduction In order to find the early diagnostic markers of AIDS combined with TM infection, we detected and analyzed the serum exosomal miRNAs of AIDS patients with or without TM infection. Materials and Methods We profiled the expression levels of miRNAs via RNA sequencing in serum exosomes from the pooled samples of 17 AIDS patients combined without TM infection and 15 AIDS combined with TM infection patients. For external validation, we validated these results using real-time quantification polymerase chain reaction (qRT-PCR) in an independent cohort of 35 AIDS patients combined without TM infection and 33 AIDS combined with TM infection patients. Finally, bioinformatics was used to predict the target genes and pathways of meaningful miRNAs. Results A total of 131 serum exosomal miRNAs including 73 up-regulated and 59 down-regulated miRNAs were found to be differentially expressed (log2FC≥1 and FDR <0.01) in the two groups. A validation analysis revealed that three miRNAs (miR-192-5p, miR-194-5p and miR-1246) were upregulated in exosomes from AIDS combined with TM infection patients. ROC analyses showed that the AUC in combined diagnosis of the three miRNAs was 0.742, and the diagnostic sensitivity and specificity were 0.568 and 0.861, respectively. In the biological process analysis, all the 3 miRNAs were involved in transcription, DNA-templated and positive regulation of transcription from RNA polymerase II promoter. At the same time, the related pathways were involved in TGF-β signaling pathway, AMPK signaling pathway, Wnt signaling pathway, MAPK signaling pathway, cGMP-PKG and cAMP signaling pathway, etc. Conclusion miR-192-5p, miR-194-5p and miR-1246 in serum exosomes might be a potential biomarker for AIDS combined with TM infection patients, which may be involved in TGF-β signaling pathway, AMPK signaling pathway, Wnt signaling pathway, MAPK signaling pathway, cGMP-PKG and cAMP signaling pathway, etc. Further research is needed on the biological functions and mechanisms of these miRNAs.
Collapse
Affiliation(s)
- Qiu-Yue Ning
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Na Liu
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Ji-Zhou Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Die-Fei Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Qi Wei
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jin-Ai Zhou
- Youjiang Medical University for Nationalities, Baise, Guangxi, People's Republic of China
| | - Jun Zou
- The Fourth People's Hospital of Nanning, Nanning, Guangxi, People's Republic of China
| | - Ning Zang
- School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, People's Republic of China.,Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Guo-Jian Li
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
16
|
Yan Z, Wang H, Mu L, Hu ZD, Zheng WQ. Regulatory roles of extracellular vesicles in immune responses against Mycobacterium tuberculosis infection. World J Clin Cases 2021; 9:7311-7318. [PMID: 34616797 PMCID: PMC8464473 DOI: 10.12998/wjcc.v9.i25.7311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/19/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are cystic vesicles naturally released by most mammalian cells and bacteria. EV contents include proteins, lipids, and nucleic acids. EVs can act as messengers to transmit a variety of molecules to recipient cells and thus play important regulatory roles in intercellular signal transduction. EVs, released by either a host cell or a pathogen, can carry pathogen-associated antigens and thus act as modulators of immune responses. EVs derived from Mycobacterium tuberculosis (Mtb)-infected cells can regulate the innate immune response through various pathways, such as regulating the release of inflammatory cytokines. In addition, EVs can mediate antigen presentation and regulate the adaptive immune response by transmitting immunoregulatory molecules to T helper cells. In this review, we summarize the regulatory roles of EVs in the immune response against Mtb.
Collapse
Affiliation(s)
- Zhi Yan
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Hua Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Lan Mu
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Zhi-De Hu
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Wen-Qi Zheng
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| |
Collapse
|
17
|
Hu ZQ, Li Q, Hu ZH, Liu HC, Rao CL, Zhang MJ, Xia YP, Deng L, Mao XH, Fang Y. MicroRNA-146a inhibits autophagy to maintain the intracellular survival of Burkholderia pseudomallei by targeting LIPA. Microb Pathog 2021; 158:104969. [PMID: 34044047 DOI: 10.1016/j.micpath.2021.104969] [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: 02/01/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 11/26/2022]
Abstract
Burkholderia pseudomallei is the etiological agent of melioidosis, which is an emerging infectious disease endemic to many tropical regions. Autophagy is an intrinsic cellular process that degrades cytoplasmic components and plays an important role in protecting the host against pathogens. Like many intracellular pathogens, B. pseudomallei can evade the autophagy-dependent cellular clearance. However, the underlying mechanism remains unclear. In this study, we applied a combination of multiple assays to monitor autophagy processes and found that B. pseudomallei induced an incomplete autophagic flux and eliminate autophagy clearance in macrophages by blocking autophagosome-lysosome fusion. Based on a high-throughput microarray screening, we found that LIPA (lysosomal acid LIPAse A) was downregulated during B. pseudomallei infection. MiR-146a was then identified to be specifically upregulated upon infection with B. pseudomallei and further regulated LIPA expression by interacting with 3'UTR of LIPA. Furthermore, overexpression of miR-146a contributed to the defect of autophagic flux caused by B. pseudomallei and was beneficial for the survival of B. pseudomallei in macrophages. Therefore, our findings suggest that miR-146a inhibits autophagy via posttranscriptional suppression of LIPA expression to maintain B. pseudomallei survival in macrophages.
Collapse
Affiliation(s)
- Zhi-Qiang Hu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China; Shigatse Branch, Xinqiao Hospital, Army 953 Hospital, Army Medical University, Shigatse, 857000, PR China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Zhen-Hong Hu
- Department of Respiratory, General Hospital of Center Theater of PLA, PLA's Health Service Scientific Research Plan, Wuhan, PR China
| | - Hai-Chao Liu
- Department of Respiratory, General Hospital of Center Theater of PLA, PLA's Health Service Scientific Research Plan, Wuhan, PR China
| | - Cheng-Long Rao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Mei-Juan Zhang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yu-Pei Xia
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Ling Deng
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xu-Hu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China.
| | - Yao Fang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Third Military Medical University (Army Medical University), Chongqing, PR China; Department of Respiratory, General Hospital of Center Theater of PLA, PLA's Health Service Scientific Research Plan, Wuhan, PR China.
| |
Collapse
|
18
|
Sun YF, Pi J, Xu JF. Emerging Role of Exosomes in Tuberculosis: From Immunity Regulations to Vaccine and Immunotherapy. Front Immunol 2021; 12:628973. [PMID: 33868247 PMCID: PMC8047325 DOI: 10.3389/fimmu.2021.628973] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/05/2021] [Indexed: 01/08/2023] Open
Abstract
Exosomes are cell-derived nanovesicles carrying protein, lipid, and nucleic acid for secreting cells, and act as significant signal transport vectors for cell-cell communication and immune modulation. Immune-cell-derived exosomes have been found to contain molecules involved in immunological pathways, such as MHCII, cytokines, and pathogenic antigens. Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), remains one of the most fatal infectious diseases. The pathogen for tuberculosis escapes the immune defense and continues to replicate despite rigorous and complicate host cell mechanisms. The infected-cell-derived exosomes under this circumstance are found to trigger different immune responses, such as inflammation, antigen presentation, and activate subsequent pathways, highlighting the critical role of exosomes in anti-MTB immune response. Additionally, as a novel kind of delivery system, exosomes show potential in developing new vaccination and treatment of tuberculosis. We here summarize recent research progress regarding exosomes in the immune environment during MTB infection, and further discuss the potential of exosomes as delivery system for novel anti-MTB vaccines and therapies.
Collapse
Affiliation(s)
- Yin-Fu Sun
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Jun-Fa Xu
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| |
Collapse
|
19
|
Xing H, Tan J, Miao Y, Lv Y, Zhang Q. Crosstalk between exosomes and autophagy: A review of molecular mechanisms and therapies. J Cell Mol Med 2021; 25:2297-2308. [PMID: 33506641 PMCID: PMC7933923 DOI: 10.1111/jcmm.16276] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 12/12/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022] Open
Abstract
Exosomes are extracellular vesicles that primarily exist in bodily fluids such as blood. Autophagy is an intracellular degradation process, which, along with exosomes, can significantly influence human health and has therefore attracted considerable attention in recent years. Exosomes have been shown to regulate the intracellular autophagic process, which, in turn, affects the circulating exosomes. However, crosstalk between exosomal and autophagic pathways is highly complex, depends primarily on the environment, and varies greatly in different diseases. In addition, studies have demonstrated that exosomes, from specific cell, can mitigate several diseases by regulating autophagy, which can also affect the excessive release of some harmful exosomes. This phenomenon lays a theoretical foundation for the improvement of many diseases. Herein, we review the mechanisms and clinical significance of the association and regulation of exosomes and autophagy, in order to provide a new perspective for the prevention and treatment of associated diseases.
Collapse
Affiliation(s)
- Huifang Xing
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | | | - Yingmei Lv
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
20
|
Biadglegne F, König B, Rodloff AC, Dorhoi A, Sack U. Composition and Clinical Significance of Exosomes in Tuberculosis: A Systematic Literature Review. J Clin Med 2021; 10:E145. [PMID: 33406750 PMCID: PMC7795701 DOI: 10.3390/jcm10010145] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis (TB) remains a major health issue worldwide. In order to contain TB infections, improved vaccines as well as accurate and reliable diagnostic tools are desirable. Exosomes are employed for the diagnosis of various diseases. At present, research on exosomes in TB is still at the preliminary stage. Recent studies have described isolation and characterization of Mycobacterium tuberculosis (Mtb) derived exosomes in vivo and in vitro. Mtb-derived exosomes (Mtbexo) may be critical for TB pathogenesis by delivering mycobacterial-derived components to the recipient cells. Proteomic and transcriptomic analysis of Mtbexo have revealed a variety of proteins and miRNA, which are utilized by the TB bacteria for pathogenesis. Exosomes has been isolated in body fluids, are amenable for fast detection, and could contribute as diagnostic or prognostic biomarker to disease control. Extraction of exosomes from biological fluids is essential for the exosome research and requires careful standardization for TB. In this review, we summarized the different studies on Mtbexo molecules, including protein and miRNA and the method used to detect exosomes in biological fluids and cell culture supernatants. Thus, the detection of Mtbexo molecules in biological fluids may have a potential to expedite the diagnosis of TB infection. Moreover, the analysis of Mtbexo may generate new aspects in vaccine development.
Collapse
Affiliation(s)
- Fantahun Biadglegne
- College of Medicine and Health Sciences, Bahir Dar University, 79 Bahir Dar, Ethiopia
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (B.K.); (A.C.R.)
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany;
| | - Brigitte König
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (B.K.); (A.C.R.)
| | - Arne C. Rodloff
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (B.K.); (A.C.R.)
| | - Anca Dorhoi
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany;
| | - Ulrich Sack
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany;
| |
Collapse
|
21
|
Silwal P, Paik S, Kim JK, Yoshimori T, Jo EK. Regulatory Mechanisms of Autophagy-Targeted Antimicrobial Therapeutics Against Mycobacterial Infection. Front Cell Infect Microbiol 2021; 11:633360. [PMID: 33828998 PMCID: PMC8019938 DOI: 10.3389/fcimb.2021.633360] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/08/2021] [Indexed: 01/25/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an intracellular pathogen causing human tuberculosis, an infectious disease that still remains as a global health problem. Autophagy, a lysosomal degradative process, has emerged as a critical pathway to restrict intracellular Mtb growth through enhancement of phagosomal maturation. Indeed, several autophagy-modulating agents show promise as host-directed therapeutics for Mtb infection. In this Review, we discuss recent progress in our understanding the molecular mechanisms underlying the action of autophagy-modulating agents to overcome the immune escape strategies mediated by Mtb. The factors and pathways that govern such mechanisms include adenosine 5'-monophosphate-activated protein kinase, Akt/mammalian TOR kinase, Wnt signaling, transcription factor EB, cathelicidins, inflammation, endoplasmic reticulum stress, and autophagy-related genes. A further understanding of these mechanisms will facilitate the development of host-directed therapies against tuberculosis as well as infections with other intracellular bacteria targeted by autophagic degradation.
Collapse
Affiliation(s)
- Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
- *Correspondence: Eun-Kyeong Jo,
| |
Collapse
|
22
|
Extracellular RNAs in Bacterial Infections: From Emerging Key Players on Host-Pathogen Interactions to Exploitable Biomarkers and Therapeutic Targets. Int J Mol Sci 2020; 21:ijms21249634. [PMID: 33348812 PMCID: PMC7766527 DOI: 10.3390/ijms21249634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/04/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are key regulators of post-transcriptional gene expression in prokaryotic and eukaryotic organisms. These molecules can interact with mRNAs or proteins, affecting a variety of cellular functions. Emerging evidence shows that intra/inter-species and trans-kingdom regulation can also be achieved with exogenous RNAs, which are exported to the extracellular medium, mainly through vesicles. In bacteria, membrane vesicles (MVs) seem to be the more common way of extracellular communication. In several bacterial pathogens, MVs have been described as a delivery system of ncRNAs that upon entry into the host cell, regulate their immune response. The aim of the present work is to review this recently described mode of host-pathogen communication and to foster further research on this topic envisaging their exploitation in the design of novel therapeutic and diagnostic strategies to fight bacterial infections.
Collapse
|
23
|
Mourenza Á, Gil JA, Mateos LM, Letek M. Novel Treatments against Mycobacterium tuberculosis Based on Drug Repurposing. Antibiotics (Basel) 2020; 9:E550. [PMID: 32872158 PMCID: PMC7557778 DOI: 10.3390/antibiotics9090550] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/30/2022] Open
Abstract
Tuberculosis is the leading cause of death, worldwide, due to a bacterial pathogen. This respiratory disease is caused by the intracellular pathogen Mycobacterium tuberculosis and produces 1.5 million deaths every year. The incidence of tuberculosis has decreased during the last decade, but the emergence of MultiDrug-Resistant (MDR-TB) and Extensively Drug-Resistant (XDR-TB) strains of M. tuberculosis is generating a new health alarm. Therefore, the development of novel therapies based on repurposed drugs against MDR-TB and XDR-TB have recently gathered significant interest. Recent evidence, focused on the role of host molecular factors on M. tuberculosis intracellular survival, allowed the identification of new host-directed therapies. Interestingly, the mechanism of action of many of these therapies is linked to the activation of autophagy (e.g., nitazoxanide or imatinib) and other well-known molecular pathways such as apoptosis (e.g., cisplatin and calycopterin). Here, we review the latest developments on the identification of novel antimicrobials against tuberculosis (including avermectins, eltrombopag, or fluvastatin), new host-targeting therapies (e.g., corticoids, fosfamatinib or carfilzomib) and the host molecular factors required for a mycobacterial infection that could be promising targets for future drug development.
Collapse
Affiliation(s)
- Álvaro Mourenza
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
| | - José A. Gil
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Luis M. Mateos
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Michal Letek
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
| |
Collapse
|
24
|
Ouyang Y, Tang Y, Fu L, Peng S, Wu W, Tan D, Fu X. Exosomes secreted by chronic hepatitis B patients with PNALT and liver inflammation grade ≥ A2 promoted the progression of liver cancer by transferring miR-25-3p to inhibit the co-expression of TCF21 and HHIP. Cell Prolif 2020; 53:e12833. [PMID: 32525231 PMCID: PMC7377934 DOI: 10.1111/cpr.12833] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES The current study aimed to investigate the mechanism by which exosomes secreted by CHB patients with PNALT and liver inflammation grade (≥A2) affected the development of liver cancer. MATERIALS AND METHODS Gene expression was assessed by RT-PCR, Western blotting and immunohistochemistry. CCK-8, colony formation, transwell, scratch-wound and flow cytometry assays were used to detect cell viability, proliferation, apoptosis and metastasis. The interaction of TCF21 and HHIP was assessed by co-immunoprecipitation assay. Luciferase reporter was used to detect the combination of TCF21/HHIP and miR-25-3p. Xenograft studies in nude mice manifested tumour growth ability of miR-25-3p. Bioinformatics analyses were conducted using TargetScan, EVmiRNA, TCGA, GEO, DAVID, COEXPEDIA, UALCAN, UCSC and the Human Protein Atlas databases. RESULTS CHB-PNALT-Exo (≥A2) promoted the proliferation and metastasis of HepG2.2.15 cells. miR-25-3p was upregulated in CHB-PNALT-Exo (≥A2). miR-25-3p overexpression promoted cell proliferation and metastasis and was related to poor survival in patients with CHB-PNALT (≥A2). The cell proliferation- and metastasis-promoting functions of CHB-PNALT-Exo (≥A2) were abolished by miR-25-3p inhibitors. TCF21 directly interacted with HHIP. Inhibition of TCF21 or HHIP promoted cell proliferation and metastasis. Knockdown of TCF21 or HHIP counteracted the effects of CHB-PNALT-Exo (≥A2) containing miR-25-3p inhibitor on cell proliferation, metastasis and the expression of Ki67, E-cadherin and caspase-3/-9. CONCLUSIONS Transfer of miR-25-3p by CHB-PNALT-Exo promoted the development of liver cancer by inhibiting the co-expression of TCF21 and HHIP.
Collapse
Affiliation(s)
- Yi Ouyang
- Key Laboratory of Viral HepatitisDepartment of Infectious DiseasesXiangya HospitalCentral South UniversityChangshaChina
| | - Yujing Tang
- Department of Second Area of Liver DiseaseXia'men Hospital of Chinese MedicineXia'menChina
| | - Lei Fu
- Key Laboratory of Viral HepatitisDepartment of Infectious DiseasesXiangya HospitalCentral South UniversityChangshaChina
| | - Shifang Peng
- Key Laboratory of Viral HepatitisDepartment of Infectious DiseasesXiangya HospitalCentral South UniversityChangshaChina
| | - Wanfeng Wu
- School of the Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaChina
| | - Deming Tan
- Key Laboratory of Viral HepatitisDepartment of Infectious DiseasesXiangya HospitalCentral South UniversityChangshaChina
| | - Xiaoyu Fu
- Key Laboratory of Viral HepatitisDepartment of Infectious DiseasesXiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
25
|
Shi Q, Wang J, Yang Z, Liu Y. CircAGFG1modulates autophagy and apoptosis of macrophages infected by Mycobacterium tuberculosis via the Notch signaling pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:645. [PMID: 32566582 PMCID: PMC7290638 DOI: 10.21037/atm.2020-20-3048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Recent studies have revealed the involvement of circular RNAs (circRNAs) in the control and elimination of invading Mycobacterium tuberculosis (Mtb) by macrophages. However, the regulatory mechanism of circAGFG1 in macrophages infected by Mtb has not been fully explored. In this study, we sought to investigate the role of circAGFG1 on autophagy and apoptosis of Mtb-infected macrophages and reveal its the molecular mechanism. Methods The expression of circAGFG1 in macrophages from patients with active tuberculosis and in Mtb-treated macrophages in vitro was explored. Then, the effect of circAGFG1 on autophagy and apoptosis of Mtb-infected macrophages was evaluated by flow cytometry, electron microscope, immunofluorescence, and Western blotting. Bioinformatics analysis was used to identify and validate the downstream regulatory pathway of circAGFG1, miRNA-1257/Notch. For further analysis, the role of miRNA-1257 on autophagy and apoptosis was assessed. Results In vitro, ectopic expression of circAGFG1 upregulated autophagy and reduced apoptosis significantly in Mtb-infected cells. Notch levels were discovered to be increased by the silencing effect of circAGFG1 on miRNA-1257 expression. miRNA-1257 was found to noticeably reduce autophagy and promote macrophage apoptosis. Increased circAGFG1 expression, decreased monocyte apoptosis, and enhanced autophagy were found in macrophages from patients with active tuberculosis. Conclusions In active tuberculosis, circAGFG1 enhances autophagy and reduces apoptosis via the miRNA-1257/Notch axis; this provides new therapeutic targets for tuberculosis patients.
Collapse
Affiliation(s)
- Qinghong Shi
- Department of Clinical Laboratory, The Third Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jingying Wang
- Department of Clinical Laboratory, The Third Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Zhe Yang
- Department of Radiological, The Second hospital of Jilin University, Changchun 130021, China
| | - Yang Liu
- Department of Radiological, The Second hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
26
|
Larabi A, Barnich N, Nguyen HTT. Emerging Role of Exosomes in Diagnosis and Treatment of Infectious and Inflammatory Bowel Diseases. Cells 2020; 9:cells9051111. [PMID: 32365813 PMCID: PMC7290936 DOI: 10.3390/cells9051111] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
To communicate with each other, cells release exosomes that transfer their composition, including lipids, proteins and nucleic acids, to neighboring cells, thus playing a role in various pathophysiological processes. During an infection with pathogenic bacteria, such as adherent-invasive E. coli (AIEC) associated with Crohn disease, exosomes secreted by infected cells can have an impact on the innate immune responses of surrounding cells to infection. Furthermore, inflammation can be amplified via the exosomal shuttle during infection with pathogenic bacteria, which could contribute to the development of the associated disease. Since these vesicles can be released in various biological fluids, changes in exosomal content may provide a means for the identification of non-invasive biomarkers for infectious and inflammatory bowel diseases. Moreover, evidence suggests that exosomes could be used as vaccines to prime the immune system to recognize and kill invading pathogens, and as therapeutic components relieving intestinal inflammation. Here, we summarize the current knowledge on the role of exosomes in bacterial infections and highlight their potential use as biomarkers, vaccines and conveyers of therapeutic molecules in inflammatory bowel diseases.
Collapse
|
27
|
The Messenger Apps of the cell: Extracellular Vesicles as Regulatory Messengers of Microglial Function in the CNS. J Neuroimmune Pharmacol 2020; 15:473-486. [PMID: 32337651 DOI: 10.1007/s11481-020-09916-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 03/20/2020] [Indexed: 02/08/2023]
Abstract
The intense effort of investigators, in particular during the past decade, has highlighted the importance of extracellular vesicles (EVs) such as exosomes in regulating both innate and adaptive immunity in the course of a variety of infections, with clear implications for development of novel vaccines, therapeutics, and diagnostics. Current and future efforts now need to focus strongly on teasing apart the intricate and complex molecular mechanisms that operate during EV regulation of immunity. In this review, we discuss recent advances that bear on our current understanding of how EVs, including exosomes, can contribute to the innate immune functions of microglia within the central nervous system (CNS), and we also highlight future important mechanistic questions that need to be addressed. In particular, recent findings that highlight the crosstalk between autophagy and exosome pathways and their implications for innate immune functions of microglia will be presented. Microglial activation has been shown to play a key role in neuroAIDS, a neuro-infectious disease for which the importance of exosome functions, including exosome-autophagy interplay, has been reported. The importance of exosomes and exosome-autophagy crosstalk involving microglia has also been shown for the Parkinson's disease (PD), a neurodegenerative disease that is thought to be linked with immune dysfunction and involve infectious agents as trigger. Considering the accumulation of recent findings and the vibrancy of the EV field, we anticipate that future studies will continue to have a deep impact on our understanding of the CNS pathologies that are influenced by the functions of microglia and of the infectious disease mechanisms in general. Graphical Abstract.
Collapse
|
28
|
Yuan Q, Chen H, Yang Y, Fu Y, Yi Z. miR-18a promotes Mycobacterial survival in macrophages via inhibiting autophagy by down-regulation of ATM. J Cell Mol Med 2019; 24:2004-2012. [PMID: 31845528 PMCID: PMC6991191 DOI: 10.1111/jcmm.14899] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/24/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is one of leading causes of global deaths. This study aimed to explore the role of miR-18a in RAW264.7 cells response to Mtb infection. Exosomes derived from Mtb-infected cells were isolated and further validated by size, transmission electron microscopy and Western blot. RT-PCR was utilized to measure miR-18a expression. Cell viability and ultrastructure were examined by CFU counting, CCK-8 and electron microscope, respectively. Potential target genes of miR-18a were predicted with bioinformatics and further confirmed using RT-PCR, Western blot and laser confocal microscope analysis, respectively. LC3, AMPK and mTOR were measured using Western blot. We found that miR-18a was induced both in Mtb-infected RAW264.7 cells and its derived exosomes compared with the controls. In addition, up-regulation of miR-18a promoted intracellular Mtb survival, attenuated cell viability and reduced LC3-II level, while its down-regulation had the opposite effect. miR-18a overexpression suppressed level of ATM, one possible target of miR-18a, while its underexpression enhanced ATM. We also found that inhibition of ATM induced LC3-II decrease in Mtb-infected cells and could reverse the increase of LC3-II caused by inhibition of miR-18a. Moreover, down-regulation of miR-18a increased p-AMPK level while reduction of ATM could reverse the change. Taken together, our results suggest that miR-18a is up-regulated in macrophages response to Mtb infection, and it promotes intracellular Mtb survival through repressing autophagic process by down-regulation of ATM pathway. This provides new thought for TB pathogenesis, diagnosis and treatment.
Collapse
Affiliation(s)
- Qiulu Yuan
- Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Department of Laboratory Medicine, Weifang Medical University, Weifang, China
| | - Haotian Chen
- Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Department of Laboratory Medicine, Weifang Medical University, Weifang, China
| | - Yuxin Yang
- Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Department of Laboratory Medicine, Weifang Medical University, Weifang, China
| | - Yurong Fu
- Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Department of Medical Microbiology, Clinical Medicine College, Weifang Medical University, Weifang, China
| | - Zhengjun Yi
- Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Department of Laboratory Medicine, Weifang Medical University, Weifang, China
| |
Collapse
|