1
|
Kim HW, Lee JW, Yoon HS, Park HW, Lee YI, Lee SK, Whang J, Kim JS. Restriction of mitochondrial oxidation of glutamine or fatty acids enhances intracellular growth of Mycobacterium abscessus in macrophages. Virulence 2025; 16:2454323. [PMID: 39828906 PMCID: PMC11749347 DOI: 10.1080/21505594.2025.2454323] [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: 08/18/2024] [Revised: 11/28/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025] Open
Abstract
Mycobacterium abscessus (Mab), a nontuberculous mycobacterium, is increasing in prevalence worldwide and causes treatment-refractory pulmonary diseases. However, how Mab rewires macrophage energy metabolism to facilitate its survival is poorly understood. We compared the metabolic profiles of murine bone marrow-derived macrophages (BMDMs) infected with smooth (S)- and rough (R)-type Mab using extracellular flux technology. Mab infection shifted BMDMs towards a more energetic phenotype, marked by increased oxidative phosphorylation (OXPHOS) and glycolysis, with a significantly greater enhancement in OXPHOS. This metabolic adaptation was characterized by enhanced ATP production rates, particularly in cells infected with S-type Mab, highlighting OXPHOS as a key energy source. Notably, Mab infection also modulated mitochondrial substrate preferences, increasing fatty acid oxidation capabilities while revealing significant changes in glutamine dependency and flexibility. R-type Mab infections exhibited a marked decrease in glutamine reliance but enhanced metabolic flexibility and capacity. Furthermore, targeting metabolic pathways related to glutamine and fatty acid oxidation exacerbated Mab growth within macrophages, suggesting these pathways play a protective role against infection. These insights advance our understanding of Mab's impact on host cell metabolism and propose a novel avenue for therapeutic intervention. By manipulating host mitochondrial metabolism, we identify a potential host-directed therapeutic strategy against Mab, offering a promising alternative to conventional treatments beleaguered by drug resistance. This study underscores the importance of exploring metabolic interventions to combat Mab infection, paving the way for innovative approaches in the fight against this formidable pathogen.
Collapse
Affiliation(s)
- Ho Won Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Ji Won Lee
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Hoe Sun Yoon
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University Hospital and College of Medicine, Daejeon, South Korea
| | | | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, South Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC), Department of Research and Development, The Korean Institute of Tuberculosis, Osong, South Korea
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
- Department of Cell Biology, Konyang University Hospital and College of Medicine, Daejeon, South Korea
| |
Collapse
|
2
|
Ayeldeen G, Shaker OG, Gomaa M, Magdy MM, Elsamaloty N, Kamel AS, Senousy MA. Association of Epistatic Effects of lncRNA GAS5, miR-146a, IRAK-1, and miR-155 Genetic Variants with Multiple Sclerosis Risk and Severity. Mol Neurobiol 2025:10.1007/s12035-025-04876-8. [PMID: 40234289 DOI: 10.1007/s12035-025-04876-8] [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: 10/21/2024] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
The complex genetic architecture of heritability in multiple sclerosis (MS) remains undisclosed mainly. Epistasis (gene-gene interaction) substantially impacts MS; however, it is largely unexplored, especially among the non-coding RNA genes and their targets. The long non-coding RNA GAS5 exacerbates demyelination and sponges miR-146a and miR-155, impeccable contributors to MS pathogenesis. miR-146a negatively regulates the immune responses by targeting IRAK-1. We investigated the association of epistatic effects and haplotypes of five single nucleotide polymorphisms (SNPs), GAS5 rs2067079, miR-146a rs2910164 and rs57095329, IRAK-1 rs3027898, and miR-155 rs767649, with the risk of MS and its phenotypes. The expression quantitative trait locus (eQTL) associated with these variants was explored through bioinformatics analysis. The study enrolled 116 MS patients and 120 healthy controls. No strong linkage disequilibrium (D' ≥ 0.8) was detected among the studied SNPs. SNP-SNP interactions overlaid an overall magnified risk of MS and its phenotypes compared to the single-locus effects. After adjustment for multiple comparisons, the most significant interactions associated with the risk of overall MS and secondary-progressive MS were rs2067079-rs2910164, rs2910164-rs57095329, and rs3027898-rs767649. The last two former SNP-SNP interactions were highly associated with relapsing-remitting MS risk. The same pattern of interactions, as observed in association with MS risk, was female-specific. The CCAAA haplotype (alleles in the order of rs2067079, rs2910164, rs57095329, rs3027898, and rs767649) was protective against MS risk (CCAAA vs. CGAAT, adjusted OR = 0.14, 95% CI = 0.03-0.69, P = 0.009). Among MS patients, harboring the CGACT and CGAAT haplotypes was more prevalent in females and males, respectively. MS patients having EDSS ≥ 6 had a significantly higher frequency of the CCGCA haplotype than those with EDSS < 6. Functional analysis revealed rs2067079, rs57095329, and rs767649 as strong cis-eQTL regulating multiple genes, particularly in the brain and immune system. We propose that a magnified combined effect of GAS5, miR-146a, IRAK-1, and miR-155 genetic variants via epistatic interactions might impact the risk of MS and its phenotypes and could help in the risk stratification of MS patients.
Collapse
Affiliation(s)
- Ghada Ayeldeen
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Olfat G Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohammed Gomaa
- Department of Neurology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Mostafa M Magdy
- Department of Neurology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Nourhan Elsamaloty
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| | - Ahmed S Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Gesr El Suez St, Cairo, PO 11786, Egypt
| | - Mahmoud A Senousy
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt.
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
3
|
Nunes S, Bastos R, Marinho AI, Vieira R, Benício I, de Noronha MA, Lírio S, Brodskyn C, Tavares NM. Recent advances in the development and clinical application of miRNAs in infectious diseases. Noncoding RNA Res 2025; 10:41-54. [PMID: 39296638 PMCID: PMC11406675 DOI: 10.1016/j.ncrna.2024.09.005] [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: 03/25/2024] [Revised: 08/06/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
In the search for new biomarkers and therapeutic targets for infectious diseases, several molecules have been investigated. Small RNAs, known as microRNAs (miRs), are important regulators of gene expression, and have emerged as promising candidates for these purposes. MiRs are a class of small, endogenous non-coding RNAs that play critical roles in several human diseases, including host-pathogen interaction mechanisms. Recently, miRs signatures have been reported in different infectious diseases, opening new perspectives for molecular diagnosis and therapy. MiR profiles can discriminate between healthy individuals and patients, as well as distinguish different disease stages. Furthermore, the possibility of assessing miRs in biological fluids, such as serum and whole blood, renders these molecules feasible for the development of new non-invasive diagnostic and prognostic tools. In this manuscript, we will comprehensively describe miRs as biomarkers and therapeutic targets in infectious diseases and explore how they can contribute to the advance of existing and new tools. Additionally, we will discuss different miR analysis platforms to understand the obstacles and advances of this molecular approach and propose their potential clinical applications and contributions to public health.
Collapse
Affiliation(s)
- Sara Nunes
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
| | - Rana Bastos
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
| | - Ananda Isis Marinho
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
| | - Raissa Vieira
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
| | - Ingra Benício
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
| | | | - Sofia Lírio
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Bahiana School of Medicine and Public Health, Salvador, Brazil
| | - Cláudia Brodskyn
- Federal University of Bahia (UFBA), Salvador, Brazil
- Laboratory of Parasite-Host Interaction and Epidemiology (LaIPHE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Instituto Nacional de Ciência e Tecnologia (INCT) Iii - Instituto de Investigação Em Imunologia, São Paulo, Brazil
| | - Natalia Machado Tavares
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia (INCT) Iii - Instituto de Investigação Em Imunologia, São Paulo, Brazil
| |
Collapse
|
4
|
Cui X, Zhang F, Meng H, Yuan T, Li M, Yuan D, Fan X, Jia X, Wang Q, Xing L, Wu C. Transport of miR-766-3p to A549 cells by plasma-derived exosomes and its effect on intracellular survival of Mycobacterium tuberculosis by regulating NRAMP1 expression in A549 cells. Microbiol Res 2025; 290:127943. [PMID: 39504604 DOI: 10.1016/j.micres.2024.127943] [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: 05/15/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024]
Abstract
Exosomal microRNAs (miRNAs) in circulation were recognized as potential biomarkers for the diagnosis of multiple diseases. However, its potential as a diagnostic hallmark for tuberculosis (TB) has yet to be explored. Here, we comprehensively analyze miRNA profiles in exosomes derived from the plasma of active TB patients and healthy persons to evaluate its efficacy in TB diagnosis. Small-RNA transcriptomic profiling analysis identified a total of 14 differentially expressed miRNAs (DEmiRNAs), among which the diagnostic potential of exosomal miR-766-3p, miR-376c-3p, miR-1283, and miR-125a-5p was evident from their respective areas under the ROC curve, which were 0.8963, 0.8313, 0.8097, and 0.8050, respectively. The bioinformatics analysis and Luciferase reporter assays confirmed that the 3'-untranslated region of natural resistance-associated macrophage protein 1 (NRAMP1) mRNA was targeted by miR-766-3p. The exosomes could be internalized by the A549 cells in co-culturing experiments. Furthermore, both increased miR-766-3p and decreased NRAMP1 expression were observed in Mtb-infected A549 cells. MiR-766-3p overexpression reduced the NRAMP1 levels, but increased intracellular Mtb, suggesting that miR-766-3p may facilitate Mtb survival by targeting NRAMP1. Moreover, miR-766-3p-transfected cells exhibited increased apoptosis and reduced proliferation following Mtb infection. Taken together, circulating exosomal miR-766-3p, miR-1283, miR-125a-5p, and miR-376c-3p may serve as candidate hallmarks for TB diagnosis where the presence of miR-766-3p seems associated with the vulnerability to Mtb infection in humans and could be a new molecular target for therapeutic intervention of TB.
Collapse
Affiliation(s)
- Xiaogang Cui
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Fengfeng Zhang
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Hangting Meng
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Tianqi Yuan
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Miao Li
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Dan Yuan
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Xiaoxia Fan
- Taiyuan Fourth People's Hospital, Taiyuan 030053, China
| | - Xiaohui Jia
- Taiyuan Fourth People's Hospital, Taiyuan 030053, China
| | - Quanhong Wang
- Taiyuan Fourth People's Hospital, Taiyuan 030053, China.
| | - Li Xing
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China.
| | - Changxin Wu
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
5
|
Kushwaha S, Goel A, Singh AV. Serum microRNA Biomarker Expression in HIV and TB: A Concise Overview. Infect Disord Drug Targets 2025; 25:e18715265305638. [PMID: 39506419 DOI: 10.2174/0118715265305638240930054842] [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: 03/30/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 11/08/2024]
Abstract
Non-coding RNAs (ncRNAs), specifically MicroRNAs or miRNAs, are now understood to be essential regulators in the complex field of gene expression. By selectively binding to certain mRNA targets, these tiny RNA molecules control the expression of genes, leading to mRNA degradation or translational repression. The discovery of miRNAs has significantly advanced biomedical research, particularly in elucidating the molecular mechanisms underlying various diseases and exploring innovative therapeutic approaches. Recent progress in miRNA research has provided insights into their biogenesis, functional roles, and potential clinical applications. Despite the absence of established methodologies for clinical implementation, miRNAs show great promise as diagnostic and therapeutic agents for a wide array of diseases. Their distinctive attributes, such as high specificity, sensitivity, and accessibility, position them as ideal candidates for biomarker development and targeted therapy. Achieving a comprehensive understanding of miRNA biology and functionality is crucial to fully harnessing their potential in medicine. Ongoing research efforts aim to unravel the intricate mechanisms of miRNA-mediated gene regulation and to develop novel approaches for utilizing miRNAs in disease diagnosis, prognosis, and treatment. This review provides a comprehensive analysis of current knowledge on miRNAs, focusing on their biogenesis, regulatory mechanisms, and potential clinical applications. By synthesizing existing evidence and highlighting key research findings, this review aims to inspire further exploration into the diverse roles of miRNAs in health and disease. Ultimately, this endeavour could result in the development of innovative miRNA-based diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Shweta Kushwaha
- Department of Microbiology and Molecular Biology, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282004, Uttar Pradesh, India
- Department of Biotechnology, GLA University, Mathura, 281406, Uttar Pradesh, India
| | - Anjana Goel
- Department of Biotechnology, GLA University, Mathura, 281406, Uttar Pradesh, India
| | - Ajay Vir Singh
- Department of Microbiology and Molecular Biology, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282004, Uttar Pradesh, India
| |
Collapse
|
6
|
Doghish AS, Abulsoud AI, Nassar YA, Nasr SM, Mohammed OA, Abdel-Reheim MA, Rizk NI, Lutfy RH, Abdel Mageed SS, Ismail MA, Abd-Elhalim HM, Awad FA, Fayez SZ, Elimam H, Mansour RM. Harnessing miRNAs: A Novel Approach to Diagnosis and Treatment of Tuberculosis. J Biochem Mol Toxicol 2025; 39:e70119. [PMID: 39799557 DOI: 10.1002/jbt.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 01/15/2025]
Abstract
Mycobacterium tuberculosis (Mtb) complex, responsible for tuberculosis (TB) infection, continues to be a predominant global cause of mortality due to intricate host-pathogen interactions that affect disease progression. MicroRNAs (miRNAs), essential posttranscriptional regulators, have become pivotal modulators of these relationships. Recent findings indicate that miRNAs actively regulate immunological responses to Mtb complex by modulating autophagy, apoptosis, and immune cell activities. This has resulted in increased interest in miRNAs as prospective diagnostic indicators for TB, especially in differentiating active infection from latent or inactive stages. Variations in miRNA expression during Mtb infection indicate disease progression and offer insights into the immune response. Furthermore, miRNAs present potential as therapeutic targets in host-directed therapy (HDT) techniques for TB infection. This work examines the function of miRNAs in TB pathogenesis, with the objective of identifying particular miRNAs that regulate the immune response to the Mtb complex, evaluating their diagnostic value and exploring their therapeutic implications in host-directed therapy for TB infection. The objective is to enhance comprehension of how miRNAs can facilitate improved diagnosis and treatment of TB.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Yara A Nassar
- Department of Botany, Biotechnology and Its Application Program, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Sami Mohamed Nasr
- Biochemistry and Molecular Biology, Theodor Bilharz Research Institute, Giza, Egypt
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | | | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Radwa H Lutfy
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
| | - Menattallah A Ismail
- Applied Biotechnology Program, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Haytham M Abd-Elhalim
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
- Agricultural Research Center, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Farah A Awad
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Salma Zaki Fayez
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Molecular Biology and Biotechnology Department, School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
| |
Collapse
|
7
|
Arya R, Kumar S, Vinetz JM, Kim JJ, Chaurasia R. Unlocking the potential of miRNAs in detecting pulmonary tuberculosis: prospects and pitfalls. Expert Rev Mol Med 2024; 26:e32. [PMID: 39639643 PMCID: PMC11629464 DOI: 10.1017/erm.2024.29] [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: 12/04/2023] [Revised: 04/03/2024] [Accepted: 07/12/2024] [Indexed: 12/07/2024]
Abstract
Tuberculosis (TB) is one of the deadliest infectious diseases globally, ranking as 13th leading cause of mortality and morbidity. According to the Global Tuberculosis Report 2022, TB claimed the lives of 1.6 million people worldwide in 2021. Among the casualties, 1 870 000 individuals with HIV co-infections contributed to 6.7% of the total fatalities, accounting TB as the second most lethal infectious disease following COVID-19. In the quest to identify biomarkers for disease progression and anti-TB therapy, microRNAs (miRNAs) have gained attention due to their precise regulatory role in gene expression in disease stages and their ability to distinguish latent and active TB, enabling the development of early TB prognostic signatures. miRNAs are stable in biological fluids and therefore will be useful for non-invasive and broad sample collection. However, their inherent lack of specificity and experimental variations may lead to false-positive outcomes. These limitations can be overcome by integrating standard protocols with machine learning, presenting a novel tool for TB diagnostics and therapeutics. This review summarizes, discusses and highlights the potential of miRNAs as a biomarker, particularly their differential expression at disease stages. The review assesses the advantages and obstacles associated with miRNA-based diagnostic biomarkers in pulmonary TB and facilitates rapid, point-of-care testing.
Collapse
Affiliation(s)
- Rakesh Arya
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Surendra Kumar
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph M. Vinetz
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Jong Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Reetika Chaurasia
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
8
|
Li Z, Hu Y, Wang W, Zou F, Yang J, Gao W, Feng S, Chen G, Shi C, Cai Y, Deng G, Chen X. Integrating pathogen- and host-derived blood biomarkers for enhanced tuberculosis diagnosis: a comprehensive review. Front Immunol 2024; 15:1438989. [PMID: 39185416 PMCID: PMC11341448 DOI: 10.3389/fimmu.2024.1438989] [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: 05/27/2024] [Accepted: 07/24/2024] [Indexed: 08/27/2024] Open
Abstract
This review explores the evolving landscape of blood biomarkers in the diagnosis of tuberculosis (TB), focusing on biomarkers derived both from the pathogen and the host. These biomarkers provide critical insights that can improve diagnostic accuracy and timeliness, essential for effective TB management. The document highlights recent advancements in molecular techniques that have enhanced the detection and characterization of specific biomarkers. It also discusses the integration of these biomarkers into clinical practice, emphasizing their potential to revolutionize TB diagnostics by enabling more precise detection and monitoring of the disease progression. Challenges such as variability in biomarker expression and the need for standardized validation processes are addressed to ensure reliability across different populations and settings. The review calls for further research to refine these biomarkers and fully harness their potential in the fight against TB, suggesting a multidisciplinary approach to overcome existing barriers and optimize diagnostic strategies. This comprehensive analysis underscores the significance of blood biomarkers as invaluable tools in the global effort to control and eliminate TB.
Collapse
Affiliation(s)
- Zhaodong Li
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Yunlong Hu
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wenfei Wang
- National Clinical Research Center for Infectious Disease, The Third People's Hospital of Shenzhen, Southern University of Science and Technology, Shenzhen, China
| | - Fa Zou
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jing Yang
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei Gao
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - SiWan Feng
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Guanghuan Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Chenyan Shi
- Department of Preventive Medicine, School of Public Health, Shenzhen University, Shenzhen, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Guofang Deng
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
9
|
Qassim HA, Mohammed ST, Muhamed HJ. The impact of miRNA-155 in acute and chronic toxoplasmosis in Iraqi women. Acta Trop 2024; 255:107211. [PMID: 38678844 DOI: 10.1016/j.actatropica.2024.107211] [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: 12/20/2023] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 05/01/2024]
Abstract
Toxoplasmosis is a prevalent parasitic infection caused by Toxoplasma gondii known to induce complex immune responses, to control the infection. MicroRNAs (miRNAs) are a cluster of small noncoding RNAs that are reported to have regulatory functions in the immune response. The objective of this study is to assess the expression of miR-155 and its targets, Src homology-2 domain-containing inositol 5- phosphatase 1 (SHIP-1) and suppressor of cytokine signaling-1 (SOCS1), in non-pregnant Iraqi women seropositive for toxoplasmosis. The study included 55 non-pregnant women positive for toxoplasmosis (20 in the acute phase and 35 in the chronic phase) and 35 non-pregnant women negative for toxoplasmosis (control group). Serum samples were collected from all participants to investigate the expression of miR-155 by RT‒PCR, in addition to the levels of SOCS1 and SHIP-1 measured by ELISA. The results showed a significant increase in the expression of miR-155 in both groups of acute and chronic toxoplasmosis compared to the control group. Lower levels of SOCS1 and SHIP-1 were found in acutely infected women compared to those with chronic infection and non-infected women. These findings showed the possible critical impact of miR-155 on host immune response during T.gondii infection, proposing that miR-155 can be explored as a prospective target to support host immune response against infectious diseases, with special help in early detection and management of toxoplasmosis in high-risk immunocompromised patients. Further studies are needed to evaluate the molecular pathways by which miRNAs improve immunity against toxoplasmosis.
Collapse
Affiliation(s)
- Hiba A Qassim
- Department of Biology, College of Science, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Sabaa T Mohammed
- Department of Biology, College of Science, Al-Mustansiriyah University, Baghdad, Iraq
| | - Haider J Muhamed
- Department of Biology, College of Science, Al-Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
10
|
Li C, Wang J, Xu JF, Pi J, Zheng B. Roles of HIF-1α signaling in Mycobacterium tuberculosis infection: New targets for anti-TB therapeutics? Biochem Biophys Res Commun 2024; 711:149920. [PMID: 38615574 DOI: 10.1016/j.bbrc.2024.149920] [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: 02/15/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Tuberculosis (TB), a deadly infectious disease induced by Mycobacterium tuberculosis (Mtb), continues to be a global public health issue that kill millions of patents every year. Despite significant efforts have been paid to identify effective TB treatments, the emergence of drug-resistant strains of the disease and the presence of comorbidities in TB patients urges us to explore the detailed mechanisms involved in TB immunity and develop more effective innovative anti-TB strategies. HIF-1α, a protein involved in regulating cellular immune responses during TB infection, has been highlighted as a promising target for the development of novel strategies for TB treatment due to its critical roles in anti-TB host immunity. This review provides a summary of current research progress on the roles of HIF-1α in TB infection, highlighting its importance in regulating the host immune response upon Mtb infection and summarizing the influences and mechanisms of HIF-1α on anti-TB immunological responses of host cells. This review also discusses the various challenges associated with developing HIF-1α as a target for anti-TB therapies, including ensuring specificity and avoiding off-target effects on normal cell function, determining the regulation and expression of HIF-1α in TB patients, and developing drugs that can inhibit HIF-1α. More deep understanding of the molecular mechanisms involved in HIF-1α signaling, its impact on TB host status, and systematic animal testing and clinical trials may benefit the optimization of HIF-1α as a novel therapeutic target for TB.
Collapse
Affiliation(s)
- Chaowei Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jiajun Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Biying Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| |
Collapse
|
11
|
Müller SM, Jücker M. The Functional Roles of the Src Homology 2 Domain-Containing Inositol 5-Phosphatases SHIP1 and SHIP2 in the Pathogenesis of Human Diseases. Int J Mol Sci 2024; 25:5254. [PMID: 38791291 PMCID: PMC11121230 DOI: 10.3390/ijms25105254] [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: 04/10/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The src homology 2 domain-containing inositol 5-phosphatases SHIP1 and SHIP2 are two proteins involved in intracellular signaling pathways and have been linked to the pathogenesis of several diseases. Both protein paralogs are well known for their involvement in the formation of various kinds of cancer. SHIP1, which is expressed predominantly in hematopoietic cells, has been implicated as a tumor suppressor in leukemogenesis especially in myeloid leukemia, whereas SHIP2, which is expressed ubiquitously, has been implicated as an oncogene in a wider variety of cancer types and is suggested to be involved in the process of metastasis of carcinoma cells. However, there are numerous other diseases, such as inflammatory diseases as well as allergic responses, Alzheimer's disease, and stroke, in which SHIP1 can play a role. Moreover, SHIP2 overexpression was shown to correlate with opsismodysplasia and Alzheimer's disease, as well as metabolic diseases. The SHIP1-inhibitor 3-α-aminocholestane (3AC), and SHIP1-activators, such as AQX-435 and AQX-1125, and SHIP2-inhibitors, such as K161 and AS1949490, have been developed and partly tested in clinical trials, which indicates the importance of the SHIP-paralogs as possible targets in the therapy of those diseases. The aim of this article is to provide an overview of the current knowledge about the involvement of SHIP proteins in the pathogenesis of cancer and other human diseases and to create awareness that SHIP1 and SHIP2 are more than just tumor suppressors and oncogenes.
Collapse
Affiliation(s)
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| |
Collapse
|
12
|
Yoshida M, Kwon AT, Qin XY, Nishimura H, Maeda S, Miyamoto Y, Yoshida Y, Hoshino Y, Suzuki H. Transcriptome analysis of long non-coding RNAs in Mycobacterium avium complex-infected macrophages. Front Immunol 2024; 15:1374437. [PMID: 38711507 PMCID: PMC11070510 DOI: 10.3389/fimmu.2024.1374437] [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: 01/22/2024] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Mycobacterium avium complex (MAC) is a non-tuberculous mycobacterium widely distributed in the environment. Even though MAC infection is increasing in older women and immunocompromised patients, to our knowledge there has been no comprehensive analysis of the MAC-infected host-cell transcriptome-and particularly of long non-coding RNAs (lncRNAs). By using in vitro-cultured primary mouse bone-marrow-derived macrophages (BMDMs) and Cap analysis of gene expression, we analyzed the transcriptional and kinetic landscape of macrophage genes, with a focus on lncRNAs, during MAC infection. MAC infection of macrophages induced the expression of immune/inflammatory response genes and other genes similar to those involved in M1 macrophage activation, consistent with previous reports, although Nos2 (M1 activation) and Arg1 (M2 activation) had distinct expression profiles. We identified 31 upregulated and 30 downregulated lncRNA promoters corresponding respectively to 18 and 26 lncRNAs. Upregulated lncRNAs were clustered into two groups-early and late upregulated-predicted to be associated with immune activation and the immune response to infection, respectively. Furthermore, an Ingenuity Pathway Analysis revealed canonical pathways and upstream transcription regulators associated with differentially expressed lncRNAs. Several differentially expressed lncRNAs reported elsewhere underwent expressional changes upon M1 or M2 preactivation and subsequent MAC infection. Finally, we showed that expressional change of lncRNAs in MAC-infected BMDMs was mediated by toll-like receptor 2, although there may be other mechanisms that sense MAC infection. We identified differentially expressed lncRNAs in MAC-infected BMDMs, revealing diverse features that imply the distinct roles of these lncRNAs in MAC infection and macrophage polarization.
Collapse
Affiliation(s)
- Mitsunori Yoshida
- Department of Mycobacteriology, National Institute of Infectious Diseases, Higashi-Murayama, Tokyo, Japan
| | - Andrew Taejun Kwon
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Xian-Yang Qin
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Hajime Nishimura
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Shiori Maeda
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yuji Miyamoto
- Department of Mycobacteriology, National Institute of Infectious Diseases, Higashi-Murayama, Tokyo, Japan
| | - Yasuhiro Yoshida
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kita-Kyushu, Japan
| | - Yoshihiko Hoshino
- Department of Mycobacteriology, National Institute of Infectious Diseases, Higashi-Murayama, Tokyo, Japan
| | - Harukazu Suzuki
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| |
Collapse
|
13
|
Wazahat R, Zaidi R, Kumar P. Epigenetic regulations in Mycobacterium tuberculosis infection. Indian J Tuberc 2024; 71:204-212. [PMID: 38589125 DOI: 10.1016/j.ijtb.2023.06.011] [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: 04/06/2023] [Revised: 06/02/2023] [Accepted: 06/22/2023] [Indexed: 04/10/2024]
Abstract
Mycobacterium tuberculosis (Mtb) employs several sophisticated strategies to evade host immunity and facilitate its intracellular survival. One of them is the epigenetic manipulation of host chromatin by three strategies i.e., DNA methylation, histone modifications and miRNA involvement. A host-directed therapeutic can be an attractive approach that targets these host epigenetics or gene regulations and circumvent manipulation of host cell machinery by Mtb. Given the complexity of the nature of intracellular infection by Mtb, there are challenges in identifying the important host proteins, non-coding RNA or the secretory proteins of Mtb itself that directly or indirectly bring upon the epigenetic modifications in the host chromatin. Equally challenging is developing the methods of targeting these epigenetic factors through chemical or non-chemical approaches as host-directed therapeutics. The current review article briefly summarizes several of the epigenetic factors that serve to bring upon potential changes in the host transcriptional machinery and targets the immune system for immunosuppression and disease progression in Mtb infection.
Collapse
Affiliation(s)
- Rushna Wazahat
- Department of Biochemistry, Jamia Hamdard, New Delhi 110062, India.
| | - Rana Zaidi
- Department of Biochemistry, Jamia Hamdard, New Delhi 110062, India
| | - Pankaj Kumar
- Department of Biochemistry, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
14
|
Yao S, Liu B, Hu X, Tan Y, Liu K, He M, Wu B, Ahmad N, Su X, Zhang Y, Yi M. Diagnostic value of microRNAs in active tuberculosis based on quantitative and enrichment analyses. Diagn Microbiol Infect Dis 2024; 108:116172. [PMID: 38340483 DOI: 10.1016/j.diagmicrobio.2024.116172] [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: 09/10/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Tuberculosis (TB) infection remains a crucial global health challenge, with active tuberculosis (ATB) representing main infection source. MicroRNA (miRNA) has emerged as a potential diagnostic tool in this context. This study aims to identify candidate miRNAs for ATB diagnosis and explore their possible mechanisms. METHODS Differentially expressed miRNAs in ATB were summarized in qualitative analysis. The diagnostic values of miRNAs for ATB subtypes were assessed by overall sensitivity, specificity, and area under the curve. Additionally, we conducted enrichment analysis on miRNAs and target genes. RESULTS Over 100 differentially expressed miRNAs were identified, with miR-29 family being the most extensively studied. The miR-29 family demonstrated sensitivity, specificity, and area under the curve of 80 %, 80 % and 0.86 respectively for active pulmonary TB (PTB). The differentially expressed miR-29-target genes in PTB were enriched in immune-related pathways. CONCLUSIONS The miR-29 family exhibits good diagnostic value for active PTB and shows association with immune process.
Collapse
Affiliation(s)
- Shuoyi Yao
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Xiangya School of Medicine, Central South University, Changsha, China
| | - Bin Liu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinyue Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yun Tan
- School of Medicine, Changsha Social Work College, Changsha, China
| | - Kun Liu
- School of Life Sciences, Central South University, Changsha, China
| | - Meng He
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Bohan Wu
- School of Life Sciences, Central South University, Changsha, China
| | - Namra Ahmad
- School of Life Sciences, Central South University, Changsha, China
| | - Xiaoli Su
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhang
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Minhan Yi
- School of Life Sciences, Central South University, Changsha, China.
| |
Collapse
|
15
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
16
|
Santana-da-Silva MN, Sena-dos-Santos C, Cáceres-Durán MÁ, de Souza FG, Gobbo AR, Pinto P, Salgado CG, dos Santos SEB. ncRNAs: an unexplored cellular defense mechanism in leprosy. Front Genet 2023; 14:1295586. [PMID: 38116294 PMCID: PMC10729009 DOI: 10.3389/fgene.2023.1295586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023] Open
Abstract
Leprosy is an infectious disease primarily caused by the obligate intracellular parasite Mycobacterium leprae. Although it has been considered eradicated in many countries, leprosy continues to be a health issue in developing nations. Besides the social stigma associated with it, individuals affected by leprosy may experience nerve damage leading to physical disabilities if the disease is not properly treated or early diagnosed. Leprosy is recognized as a complex disease wherein socioenvironmental factors, immune response, and host genetics interact to contribute to its development. Recently, a new field of study called epigenetics has emerged, revealing that the immune response and other mechanisms related to infectious diseases can be influenced by noncoding RNAs. This review aims to summarize the significant advancements concerning non-coding RNAs in leprosy, discussing the key perspectives on this novel approach to comprehending the pathophysiology of the disease and identifying molecular markers. In our view, investigations on non-coding RNAs in leprosy hold promise and warrant increased attention from researches in this field.
Collapse
Affiliation(s)
- Mayara Natália Santana-da-Silva
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
- Laboratório de Imunologia, Seção de Virologia (SAVIR), Instituto Evandro Chagas, Ananindeua, Brazil
| | - Camille Sena-dos-Santos
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Miguel Ángel Cáceres-Durán
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Felipe Gouvea de Souza
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Angelica Rita Gobbo
- Laboratório de Dermato-Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Pablo Pinto
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
- Laboratório de Dermato-Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Claudio Guedes Salgado
- Laboratório de Dermato-Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Sidney Emanuel Batista dos Santos
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
- Laboratório de Dermato-Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| |
Collapse
|
17
|
Yeoh WJ, Krebs P. SHIP1 and its role for innate immune regulation-Novel targets for immunotherapy. Eur J Immunol 2023; 53:e2350446. [PMID: 37742135 DOI: 10.1002/eji.202350446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/03/2023] [Accepted: 09/21/2023] [Indexed: 09/25/2023]
Abstract
Phosphoinositide-3-kinase/AKT (PI3K/AKT) signaling plays key roles in the regulation of cellular activity in both health and disease. In immune cells, this PI3K/AKT pathway is critically regulated by the phosphoinositide phosphatase SHIP1, which has been reported to modulate the function of most immune subsets. In this review, we summarize our current knowledge of SHIP1 with a focus on innate immune cells, where we reflect on the most pertinent aspects described in the current literature. We also present several small-molecule agonists and antagonists of SHIP1 developed over the last two decades, which have led to improved outcomes in several preclinical models of disease. We outline these promising findings and put them in relation to human diseases with unmet medical needs, where we discuss the most attractive targets for immune therapies based on SHIP1 modulation.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Philippe Krebs
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Abdalla AE, Alanazi A, Abosalif KOA, Alameen AAM, Junaid K, Manni E, Talha AA, Ejaz H. MicroRNA-155, a double-blade sword regulator of innate tuberculosis immunity. Microb Pathog 2023; 185:106438. [PMID: 37925110 DOI: 10.1016/j.micpath.2023.106438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023]
Abstract
Tuberculosis (TB) is a chronic, life-threatening disease caused by unusual facultative intracellular bacteria, Mycobacterium tuberculosis. This bacterium has unique resistance to many antimicrobial agents and has become a major global health concern due to emerging multidrug-resistant strains. Additionally, it has developed multiple schemes to exploit host immune signaling and establish long-term survival within host tissues. Thus, understanding the pathways that govern the crosstalk between the bacterium and the immune system could provide a new avenue for therapeutic interventions. MicroRNAs (miRs) are short, noncoding, and regulator RNA molecules that control the expression of cellular genes by targeting their mRNAs post-transcriptionally. MiR-155 is one of the most crucial miR in shaping the host immune defenses against M. tuberculosis. MiR-155 is remarkably downregulated in patients with clear clinical TB symptoms in comparison with latently infected patients and/or healthy individuals, thereby implicating its role in controlling M. tuberculosis infection. However, functional probing of miR-155 suggests dual effects in regulating the host's innate defenses in response to mycobacterial infection. This review provides comprehensive knowledge and future perspectives regarding complex signaling pathways that mediated miR-155 expression during M. tuberculosis infections. Moreover, miR-155-targeting signaling orchestrates inflammatory mediators' production, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Abualgasim Elgaili Abdalla
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia.
| | - Awadh Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Khalid Omer Abdalla Abosalif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Ayman Ali Mohammed Alameen
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Kashaf Junaid
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Emad Manni
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Albadawi Abdelbagi Talha
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia.
| |
Collapse
|
19
|
Peterson EJR, Brooks AN, Reiss DJ, Kaur A, Do J, Pan M, Wu WJ, Morrison R, Srinivas V, Carter W, Arrieta-Ortiz ML, Ruiz RA, Bhatt A, Baliga NS. MtrA modulates Mycobacterium tuberculosis cell division in host microenvironments to mediate intrinsic resistance and drug tolerance. Cell Rep 2023; 42:112875. [PMID: 37542718 PMCID: PMC10480492 DOI: 10.1016/j.celrep.2023.112875] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/21/2023] [Accepted: 07/11/2023] [Indexed: 08/07/2023] Open
Abstract
The success of Mycobacterium tuberculosis (Mtb) is largely attributed to its ability to physiologically adapt and withstand diverse localized stresses within host microenvironments. Here, we present a data-driven model (EGRIN 2.0) that captures the dynamic interplay of environmental cues and genome-encoded regulatory programs in Mtb. Analysis of EGRIN 2.0 shows how modulation of the MtrAB two-component signaling system tunes Mtb growth in response to related host microenvironmental cues. Disruption of MtrAB by tunable CRISPR interference confirms that the signaling system regulates multiple peptidoglycan hydrolases, among other targets, that are important for cell division. Further, MtrA decreases the effectiveness of antibiotics by mechanisms of both intrinsic resistance and drug tolerance. Together, the model-enabled dissection of complex MtrA regulation highlights its importance as a drug target and illustrates how EGRIN 2.0 facilitates discovery and mechanistic characterization of Mtb adaptation to specific host microenvironments within the host.
Collapse
Affiliation(s)
| | | | - David J Reiss
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Amardeep Kaur
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Julie Do
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Min Pan
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Wei-Ju Wu
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Robert Morrison
- Laboratory of Malaria, Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | | | - Warren Carter
- Institute for Systems Biology, Seattle, WA 98109, USA
| | | | - Rene A Ruiz
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Apoorva Bhatt
- School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Birmingham B15 2TT, UK
| | - Nitin S Baliga
- Institute for Systems Biology, Seattle, WA 98109, USA; Departments of Biology and Microbiology, University of Washington, Seattle, WA 98195, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA; Lawrence Berkeley National Lab, Berkeley, CA 94720, USA.
| |
Collapse
|
20
|
Shin E, Han SH, Park IS, Wee JH, Lee JS, Kim H. Does the Necrotic Portion of Metastatic Lymphadenopathy from Squamous Cell Carcinoma Still Have Tumoral Oncologic Information? Differential Diagnosis of Benign Necrotic Lymphadenopathy Using microRNA. Biomedicines 2023; 11:2407. [PMID: 37760848 PMCID: PMC10525664 DOI: 10.3390/biomedicines11092407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/14/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Neck necrotic lymph nodes commonly correspond to metastasis or benign inflammatory conditions such as Kikuchi disease and tuberculosis. Ultrasound-guided biopsy can be used for differential diagnosis, but results may be unclear. Therefore, this study aimed to identify target microRNAs (miRNAs) and genes for the differential diagnosis of inflammatory and malignant necrotic lymph nodes. We selected six inflammatory lymphadenitis formalin-fixed paraffin-embedded (FFPE) samples that showed internal necrosis and five cancer necrotic FFPE samples. Tissue microarray (TMA) was performed to separate the necrotic and cancerous portions. Total RNA was extracted from six pairs of separated inflammatory necrosis, five pairs of cancer necrosis, and cancer portions. Differentially expressed miRNAs were analyzed by comparing inflammatory necrosis, cancer, and cancer necrosis. Seventeen miRNAs were upregulated in cancer necrosis compared to inflammatory necrosis, and two miRNAs (hsa-miR-155-5p and hsa-miR-146b-5p) showed lower expression in cancer necrotic cells. Nineteen miRNAs that were differentially expressed between inflammatory and cancer necrosis were analyzed for target gene expression; these transcripts demonstrated a clear relationship with cancer. The differentially expressed miRNAs in inflammatory and tumor necrosis were associated with cancer-related pathways. These preliminary results might help in the differential diagnosis of cervical metastatic necrotic lymphadenopathy and avoiding unnecessary excisional biopsies.
Collapse
Affiliation(s)
- Eun Shin
- Department of Pathology, Dongtan Sacred Heart Hospital, Hwaseong 18450, Republic of Korea;
| | - Seung Hoon Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongtan Sacred Heart Hospital, Hwaseong 18450, Republic of Korea; (S.H.H.); (I.-S.P.)
| | - Il-Seok Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Dongtan Sacred Heart Hospital, Hwaseong 18450, Republic of Korea; (S.H.H.); (I.-S.P.)
| | - Jee Hye Wee
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea; (J.H.W.); (J.S.L.)
| | - Joong Seob Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea; (J.H.W.); (J.S.L.)
| | - Heejin Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea; (J.H.W.); (J.S.L.)
| |
Collapse
|
21
|
Alijani E, Rad FR, Katebi A, Ajdary S. Differential Expression of miR-146 and miR-155 in Active and Latent Tuberculosis Infection. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:1749-1757. [PMID: 37744552 PMCID: PMC10512130 DOI: 10.18502/ijph.v52i8.13414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/15/2022] [Indexed: 09/26/2023]
Abstract
Background Tuberculosis (TB) is one of the leading causes of death worldwide. Besides, one-third of the world population is infected with Mycobacterium tuberculosis (MTB) while staying clinically asymptomatic; the situation is called latent TB infection (LTBI). MiR-21, miR-31, miR-146a, and miR-155 play an important role in many immune and inflammatory pathways. In the present study the expression levels of MiR-21, miR-31, miR-146a, and miR-155 in peripheral blood mononuclear cells (PBMCs) from patients with active TB, latently infected individuals (LTBI), and healthy controls (HC) were investigated. Participants were recruited at the Bouali Hospital, Zahedan University of Medical Sciences, Zahedan, Iran from 2010 to 2011. Methods PBMCs were stimulated with PPD before RNA extraction. TaqMan RT-qPCR assay was used to analyze the expression levels of miRNAs. Results The results indicated no significant differences in the expression of miR-21 and miR-31 between different groups; however, in patients with active TB, the expression of miR-21 (P=0.03) and miR-31 (P=0.04) were significantly increased after stimulation with PPD compared to the unstimulated condition. The expression of miR-146 in response to PPD in both LTBI (P=0.02) and TB (P=0.03) groups compared to the HC group was increased. No significant differences were found in the expression level of miR-155 in response to PPD between LTBI and HC groups. However, the fold change was significantly higher in the TB group in comparison with the HC (P=0.03) and LTBI (P=0.05) groups. Conclusion The results confirm the main role of miR-146 and miR-155 in TB infection and suggest a role for miR-146 and miR-155 as infection and activation markers in tuberculosis infection, respectively.
Collapse
Affiliation(s)
- Ebrahim Alijani
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Farhad Riazi Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Asal Katebi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
22
|
MicroRNAs as Biomarkers of Active Pulmonary TB Course. Microorganisms 2023; 11:microorganisms11030626. [PMID: 36985200 PMCID: PMC10053298 DOI: 10.3390/microorganisms11030626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
The spread of drug-resistant forms of TB dictates the need for surgical treatment in the complex of anti-tuberculosis measures in Russia. Most often, surgical intervention is performed in the case of pulmonary tuberculoma or fibrotic cavitary tuberculosis (FCT). This study is devoted to the search for biomarkers that characterize the course of disease in surgical TB patients. It is assumed that such biomarkers will help the surgeon decide on the timing of the planned operation. A number of serum microRNAs, potential regulators of inflammation and fibrosis in TB, selected on the basis of PCR-Array analysis, were considered as biomarkers. Quantitative real time polymerase chain reaction and receiver operating curves (ROC) were used to verify Array data and to estimate the ability of microRNAs (miRNAs) to discriminate between healthy controls, tuberculoma patients, and FCT patients. The study showed that miR-155, miR-191 and miR-223 were differentially expressed in serum of tuberculoma with “decay” and tuberculoma without “decay” patients. Another combination (miR-26a, miR-191, miR-222 and miR-320) forms a set to differentiate between tuberculoma with “decay” and FCT. Patients with tuberculoma without “decay” diagnosis differ from those with FCT in serum expression of miR-26a, miR-155, miR-191, miR-222 and miR-223. Further investigations are required to evaluate these sets on a larger population so as to set cut-off values that could be applied in laboratory diagnosis.
Collapse
|
23
|
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
|
24
|
Qin W, Saris A, van ’t Veer C, Roelofs JJTH, Scicluna BP, de Vos AF, van der Poll T. Myeloid miR-155 plays a limited role in antibacterial defense during Klebsiella-derived pneumosepsis and is dispensable for lipopolysaccharide- or Klebsiella-induced inflammation in mice. Pathog Dis 2023; 81:ftad031. [PMID: 37858304 PMCID: PMC10636497 DOI: 10.1093/femspd/ftad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/02/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023] Open
Abstract
MicroRNA-155 (miR-155) plays a crucial role in regulating host inflammatory responses during bacterial infection. Previous studies have shown that constitutive miR-155 deficiency alleviates inflammation while having varying effects in different bacterial infection models. However, whether miR-155 in myeloid cells is involved in the regulation of inflammatory and antibacterial responses is largely elusive. Mice with myeloid cell specific miR-155 deficiency were generated to study the in vitro response of bone marrow-derived macrophages (BMDMs), alveolar macrophages (AMs) and peritoneal macrophages (PMs) to lipopolysaccharide (LPS), and the in vivo response after intranasal or intraperitoneal challenge with LPS or infection with Klebsiella (K.) pneumoniae via the airways. MiR-155-deficient macrophages released less inflammatory cytokines than control macrophages upon stimulation with LPS in vitro. However, the in vivo inflammatory cytokine response to LPS or K. pneumoniae was not affected by myeloid miR-155 deficiency. Moreover, bacterial outgrowth in the lungs was not altered in myeloid miR-155-deficient mice, but Klebsiella loads in the liver of these mice were significantly higher than in control mice. These data argue against a major role for myeloid miR-155 in host inflammatory responses during LPS-induced inflammation and K. pneumoniae-induced pneumosepsis but suggest that myeloid miR-155 contributes to host defense against Klebsiella infection in the liver.
Collapse
Affiliation(s)
- Wanhai Qin
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Anno Saris
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Cornelis van ’t Veer
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Brendon P Scicluna
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, MSD 2080, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, MSD 2080, Msida, Malta
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
25
|
Zhou H, Qian Y, Liu J. MicroRNA-127 promotes anti-microbial ability in porcine alveolar macrophages via S1PR3/TLR signaling pathway. J Vet Sci 2023; 24:e20. [PMID: 37012029 PMCID: PMC10071279 DOI: 10.4142/jvs.22110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND As Actinobacillus pleuropneumonniae (APP) infection causes considerable losses in the pig industry, there is a growing need to develop effective therapeutic interventions that leverage host immune defense mechanisms to combat these pathogens. OBJECTIVES To demonstrate the role of microRNA (miR)-127 in controlling bacterial infection against APP. Moreover, to investigate a signaling pathway in macrophages that controls the production of anti-microbial peptides. METHODS Firstly, we evaluated the effect of miR-127 on APP-infected pigs by cell count/enzyme-linked immunosorbent assay (ELISA). Then the impact of miR-127 on immune cells was detected. The cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6 were evaluated by ELISA. The expression of cytokines (anti-microbial peptides [AMPs]) was assessed using quantitative polymerase chain reaction. The expression level of IL-6, TNF-α and p-P65 were analyzed by western blot. The expression of p65 in the immune cells was investigated by immunofluorescence. RESULTS miR-127 showed a protective effect on APP-infected macrophage. Moreover, the protective effect might depend on its regulation of macrophage bactericidal activity and the generation of IL-22, IL-17 and AMPs by targeting sphingosine-1-phosphate receptor3 (SIPR3), the element involved in the Toll-like receptor (TLR) cascades. CONCLUSIONS Together, we identify that miR-127 is a regulator of S1PR3 and then regulates TLR/nuclear factor-κB signaling in macrophages with anti-bacterial acticity, and it might be a potential target for treating inflammatory diseases caused by APP.
Collapse
Affiliation(s)
- Honglei Zhou
- School of Pet Science and Technology, Jiangsu Agri-animal Husbandry Vocational College Jiangsu 225300, China
| | - Yujia Qian
- Taizhou Jianyouda Pharma Co., LTD, Jiangsu 225300, China
| | - Jing Liu
- School of Pet Science and Technology, Jiangsu Agri-animal Husbandry Vocational College Jiangsu 225300, China
| |
Collapse
|
26
|
Xia Q, Tang Y, Li W, Liang T, Zhou Y, Liu J, Liu F. Surface-Engineered Monocyte Immunotherapy Combined Graphene Quantum Dots Effective Against Solid Tumor Targets. Int J Nanomedicine 2023; 18:2127-2140. [PMID: 37122502 PMCID: PMC10145394 DOI: 10.2147/ijn.s404486] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/16/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction The immunosuppressive tumor microenvironment (TME) of solid tumors inhibits most drug delivery system-based nanomaterials from achieving deep penetration in tumor tissue and interferes with T cell activity in terms of differentiation and exhaustion, which is becoming a critical therapy hurdle for solid tumors. Therefore, developing a therapeutic strategy with abilities of rapid establishment of tumor-targeted cells, elimination of immune obstacles, and enhanced active immunization is very important, while is still a big challenge. Methods A new strategy was explored to enhance immune therapy via the conjugation of microRNA155 (miR) to the surface of therapeutic monocyte with graphene quantum dots (GQDs). Results TME was reversed using surface-engineered monocyte immunotherapy via reprogramming pro-tumoral M2 TAMs into antitumor M1, and thus tumor elimination was dramatically enhanced. Conclusion Such a surface-engineered monocyte immunotherapy has been demonstrated to be well tolerated to intravenous administration and bio-compatible, showing the potential to be extended for the solid tumor treatment.
Collapse
Affiliation(s)
- Qing Xia
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Yue Tang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Wang Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Tingting Liang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
- Correspondence: Feila Liu, School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China, Tel +86-15123002638, Fax +86 2362563190, Email
| |
Collapse
|
27
|
Iron: Not Just a Passive Bystander in AITD. Nutrients 2022; 14:nu14214682. [PMID: 36364944 PMCID: PMC9658435 DOI: 10.3390/nu14214682] [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: 09/30/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Autoimmune thyroid disease (AITD) is the most prevalent autoimmune disease all over the world and the most frequent cause of hypothyroidism in areas of iodine sufficiency. The pathogenesis of AITD is multifactorial and depends on complex interactions between genetic and environmental factors, with epigenetics being the crucial link. Iron deficiency (ID) can reduce the activities of thyroid peroxidase and 5′-deiodinase, inhibit binding of triiodothyronine to its nuclear receptor, and cause slower utilization of T3 from the serum pool. Moreover, ID can disturb the functioning of the immune system, increasing the risk of autoimmune disorders. ID can be responsible for residual symptoms that may persist in patients with AITD, even if their thyrometabolic status has been controlled. The human lifestyle in the 21st century is inevitably associated with exposure to chemical compounds, pathogens, and stress, which implies an increased risk of autoimmune disorders and thyroid dysfunction. To summarize, in our paper we discuss how iron deficiency can impair the functions of the immune system, cause epigenetic changes in human DNA, and potentiate tissue damage by chemicals acting as thyroid disruptors.
Collapse
|
28
|
Nisa A, Kipper FC, Panigrahy D, Tiwari S, Kupz A, Subbian S. Different modalities of host cell death and their impact on Mycobacterium tuberculosis infection. Am J Physiol Cell Physiol 2022; 323:C1444-C1474. [PMID: 36189975 PMCID: PMC9662802 DOI: 10.1152/ajpcell.00246.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/22/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is the pathogen that causes tuberculosis (TB), a leading infectious disease of humans worldwide. One of the main histopathological hallmarks of TB is the formation of granulomas comprised of elaborately organized aggregates of immune cells containing the pathogen. Dissemination of Mtb from infected cells in the granulomas due to host and mycobacterial factors induces multiple cell death modalities in infected cells. Based on molecular mechanism, morphological characteristics, and signal dependency, there are two main categories of cell death: programmed and nonprogrammed. Programmed cell death (PCD), such as apoptosis and autophagy, is associated with a protective response to Mtb by keeping the bacteria encased within dead macrophages that can be readily phagocytosed by arriving in uninfected or neighboring cells. In contrast, non-PCD necrotic cell death favors the pathogen, resulting in bacterial release into the extracellular environment. Multiple types of cell death in the PCD category, including pyroptosis, necroptosis, ferroptosis, ETosis, parthanatos, and PANoptosis, may be involved in Mtb infection. Since PCD pathways are essential for host immunity to Mtb, therapeutic compounds targeting cell death signaling pathways have been experimentally tested for TB treatment. This review summarizes different modalities of Mtb-mediated host cell deaths, the molecular mechanisms underpinning host cell death during Mtb infection, and its potential implications for host immunity. In addition, targeting host cell death pathways as potential therapeutic and preventive approaches against Mtb infection is also discussed.
Collapse
Affiliation(s)
- Annuurun Nisa
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Franciele C Kipper
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sangeeta Tiwari
- Department of Biological Sciences, Border Biomedical Research Center (BBRC), University of Texas, El Paso, Texas
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Townsville, Queensland, Australia
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
29
|
Davuluri KS, Chauhan DS. microRNAs associated with the pathogenesis and their role in regulating various signaling pathways during Mycobacterium tuberculosis infection. Front Cell Infect Microbiol 2022; 12:1009901. [PMID: 36389170 PMCID: PMC9647626 DOI: 10.3389/fcimb.2022.1009901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/03/2022] [Indexed: 11/22/2022] Open
Abstract
Despite more than a decade of active study, tuberculosis (TB) remains a serious health concern across the world, and it is still the biggest cause of mortality in the human population. Pathogenic bacteria recognize host-induced responses and adapt to those hostile circumstances. This high level of adaptability necessitates a strong regulation of bacterial metabolic characteristics. Furthermore, the immune reponse of the host virulence factors such as host invasion, colonization, and survival must be properly coordinated by the pathogen. This can only be accomplished by close synchronization of gene expression. Understanding the molecular characteristics of mycobacterial pathogenesis in order to discover therapies that prevent or resolve illness relies on the bacterial capacity to adjust its metabolism and replication in response to various environmental cues as necessary. An extensive literature details the transcriptional alterations of host in response to in vitro environmental stressors, macrophage infection, and human illness. Various studies have recently revealed the finding of several microRNAs (miRNAs) that are believed to play an important role in the regulatory networks responsible for adaptability and virulence in Mycobacterium tuberculosis. We highlighted the growing data on the existence and quantity of several forms of miRNAs in the pathogenesis of M. tuberculosis, considered their possible relevance to disease etiology, and discussed how the miRNA-based signaling pathways regulate bacterial virulence factors.
Collapse
|
30
|
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
|
31
|
Paik S, Kim KT, Kim IS, Kim YJ, Kim HJ, Choi S, Kim HJ, Jo EK. Mycobacterial acyl carrier protein suppresses TFEB activation and upregulates miR-155 to inhibit host defense. Front Immunol 2022; 13:946929. [PMID: 36248815 PMCID: PMC9559204 DOI: 10.3389/fimmu.2022.946929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022] Open
Abstract
Mycobacterial acyl carrier protein (AcpM; Rv2244), a key protein involved in Mycobacterium tuberculosis (Mtb) mycolic acid production, has been shown to suppress host cell death during mycobacterial infection. This study reports that mycobacterial AcpM works as an effector to subvert host defense and promote bacterial growth by increasing microRNA (miRNA)-155-5p expression. In murine bone marrow-derived macrophages (BMDMs), AcpM protein prevented transcription factor EB (TFEB) from translocating to the nucleus in BMDMs, which likely inhibited transcriptional activation of several autophagy and lysosomal genes. Although AcpM did not suppress autophagic flux in BMDMs, AcpM reduced Mtb and LAMP1 co-localization indicating that AcpM inhibits phagolysosomal fusion during Mtb infection. Mechanistically, AcpM boosted the Akt-mTOR pathway in BMDMs by upregulating miRNA-155-5p, a SHIP1-targeting miRNA. When miRNA-155-5p expression was inhibited in BMDMs, AcpM-induced increased intracellular survival of Mtb was suppressed. In addition, AcpM overexpression significantly reduced mycobacterial clearance in C3HeB/FeJ mice infected with recombinant M. smegmatis strains. Collectively, our findings point to AcpM as a novel mycobacterial effector to regulate antimicrobial host defense and a potential new therapeutic target for Mtb infection.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- *Correspondence: Seungwha Paik, ; Eun-Kyeong Jo,
| | - Kyeong Tae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - In Soo Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hyeon Ji Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Seunga Choi
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
- *Correspondence: Seungwha Paik, ; Eun-Kyeong Jo,
| |
Collapse
|
32
|
Daniel EA, Sathiyamani B, Thiruvengadam K, Vivekanandan S, Vembuli H, Hanna LE. MicroRNAs as diagnostic biomarkers for Tuberculosis: A systematic review and meta- analysis. Front Immunol 2022; 13:954396. [PMID: 36238288 PMCID: PMC9551313 DOI: 10.3389/fimmu.2022.954396] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe early diagnosis of tuberculosis using novel non-sputum-based biomarkers is of high priority in the End TB strategy. MicroRNAs (miRNAs) are significant regulators of TB pathogenesis and their differential expression pattern among healthy, latent, and active TB population has revealed their potentiality as biomarkers in recent studies. Thus, we systematically reviewed and performed a meta-analysis on the role of host miRNAs in TB diagnosis. We also reviewed the involvement of miRNAs in the immune response to Mycobacterium tuberculosis (Mtb).MethodsPubmed, Ovid and Cochrane databases were searched to retrieve published literature from 2000 to 2020 using predefined keywords. We screened relevant studies based on inclusion and exclusion criteria and the included studies were assessed for their quality using STARD guidelines and QUADAS-2 tool. Funnel plots were constructed to assess the publication bias. The heterogeneity of studies and overall pooled results of sensitivity, specificity and DOR were determined using forest plots.ResultsWe retrieved a total of 447 studies collectively from all the databases, out of which 21 studies were included for qualitative analysis. In these studies, miR-29, miR-31, miR-125b, miR146a and miR-155 were consistently reported. The overall sensitivity, specificity and DOR of these miRNAs were found to be 87.9% (81.7-92.2), 81.2% (74.5-86.5) and 43.1(20.3-91.3) respectively. Among these, miR-31 had the maximum diagnostic accuracy, with a sensitivity of 96% (89.7-98.5), specificity of 89% (81.2-93.8) and DOR of 345.9 (90.2-1326.3), meeting the minimal target product profile (TPP) for TB diagnostics.ConclusionmiRNAs can thus be exploited as potential biomarkers for rapid detection of tuberculosis as evident from their diagnostic performance. Systematic Review Registrationhttps://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021226559 PROSPERO (CRD42021226559).
Collapse
Affiliation(s)
- Evangeline Ann Daniel
- Department of Virology and Biotechnology, ICMR- National Institute for Research in Tuberculosis, Chennai, India
- University of Madras, Chennai, India
| | - Balakumaran Sathiyamani
- Department of Virology and Biotechnology, ICMR- National Institute for Research in Tuberculosis, Chennai, India
- University of Madras, Chennai, India
| | - Kannan Thiruvengadam
- Department of Statistics, Epidemiology Unit, ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - Sandhya Vivekanandan
- Department of Virology and Biotechnology, ICMR- National Institute for Research in Tuberculosis, Chennai, India
- University of Madras, Chennai, India
| | - Hemanathan Vembuli
- Department of Virology and Biotechnology, ICMR- National Institute for Research in Tuberculosis, Chennai, India
| | - Luke Elizabeth Hanna
- Department of Virology and Biotechnology, ICMR- National Institute for Research in Tuberculosis, Chennai, India
- *Correspondence: Luke Elizabeth Hanna,
| |
Collapse
|
33
|
Malekos E, Carpenter S. Short open reading frame genes in innate immunity: from discovery to characterization. Trends Immunol 2022; 43:741-756. [PMID: 35965152 PMCID: PMC10118063 DOI: 10.1016/j.it.2022.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/27/2022]
Abstract
Next-generation sequencing (NGS) technologies have greatly expanded the size of the known transcriptome. Many newly discovered transcripts are classified as long noncoding RNAs (lncRNAs) which are assumed to affect phenotype through sequence and structure and not via translated protein products despite the vast majority of them harboring short open reading frames (sORFs). Recent advances have demonstrated that the noncoding designation is incorrect in many cases and that sORF-encoded peptides (SEPs) translated from these transcripts are important contributors to diverse biological processes. Interest in SEPs is at an early stage and there is evidence for the existence of thousands of SEPs that are yet unstudied. We hope to pique interest in investigating this unexplored proteome by providing a discussion of SEP characterization generally and describing specific discoveries in innate immunity.
Collapse
Affiliation(s)
- Eric Malekos
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA; Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Susan Carpenter
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA; Department of Molecular Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
34
|
Ricci S, Petri RM, Pacífico C, Castillo-Lopez E, Rivera-Chacon R, Sener-Aydemir A, Reisinger N, Zebeli Q, Kreuzer-Redmer S. Characterization of presence and activity of microRNAs in the rumen of cattle hints at possible host-microbiota cross-talk mechanism. Sci Rep 2022; 12:13812. [PMID: 35970850 PMCID: PMC9378797 DOI: 10.1038/s41598-022-17445-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/26/2022] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs (miRNAs), as important post-transcriptional regulators, are ubiquitous in various tissues. The aim of this exploratory study was to determine the presence of miRNAs in rumen fluid, and to investigate the possibility of miRNA-mediated cross-talk within the ruminal ecosystem. Rumen fluid samples from four cannulated Holstein cows were collected during two feeding regimes (forage and high-grain diet) and DNA and RNA were extracted for amplicon and small RNA sequencing. Epithelial biopsies were simultaneously collected to investigate the co-expression of miRNAs in papillae and rumen fluid. We identified 377 miRNAs in rumen fluid and 638 in rumen papillae, of which 373 were shared. Analysis of microbiota revealed 20 genera to be differentially abundant between the two feeding regimes, whereas no difference in miRNAs expression was detected. Correlations with at least one genus were found for 170 miRNAs, of which, 39 were highly significant (r > |0.7| and P < 0.01). Both hierarchical clustering of the correlation matrix and WGCNA analysis identified two main miRNA groups. Putative target and functional prediction analysis for the two groups revealed shared pathways with the predicted metabolic activities of the microbiota. Hence, our study supports the hypothesis of a cross-talk within the rumen at least partly mediated by miRNAs.
Collapse
Affiliation(s)
- Sara Ricci
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine, Vienna, Austria.
| | - Renée M Petri
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC, Canada
| | - Cátia Pacífico
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine, Vienna, Austria
- Biome Diagnostics GmbH, Vienna, Austria
| | - Ezequias Castillo-Lopez
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine, Vienna, Austria
| | - Raul Rivera-Chacon
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine, Vienna, Austria
| | - Arife Sener-Aydemir
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine, Vienna, Austria
| | | | - Qendrim Zebeli
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine, Vienna, Austria
| | - Susanne Kreuzer-Redmer
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
35
|
Antonakos N, Gilbert C, Théroude C, Schrijver IT, Roger T. Modes of action and diagnostic value of miRNAs in sepsis. Front Immunol 2022; 13:951798. [PMID: 35990654 PMCID: PMC9389448 DOI: 10.3389/fimmu.2022.951798] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a clinical syndrome defined as a dysregulated host response to infection resulting in life-threatening organ dysfunction. Sepsis is a major public health concern associated with one in five deaths worldwide. Sepsis is characterized by unbalanced inflammation and profound and sustained immunosuppression, increasing patient susceptibility to secondary infections and mortality. microRNAs (miRNAs) play a central role in the control of many biological processes, and deregulation of their expression has been linked to the development of oncological, cardiovascular, neurodegenerative and metabolic diseases. In this review, we discuss the role of miRNAs in sepsis pathophysiology. Overall, miRNAs are seen as promising biomarkers, and it has been proposed to develop miRNA-based therapies for sepsis. Yet, the picture is not so straightforward because of the versatile and dynamic features of miRNAs. Clearly, more research is needed to clarify the expression and role of miRNAs in sepsis, and to promote the use of miRNAs for sepsis management.
Collapse
Affiliation(s)
| | | | | | | | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
36
|
MiR155 Disrupts the Intestinal Barrier by Inducing Intestinal Inflammation and Altering the Intestinal Microecology in Severe Acute Pancreatitis. Dig Dis Sci 2022; 67:2209-2219. [PMID: 34341909 DOI: 10.1007/s10620-021-07022-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/21/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Intestinal dysfunction is a common complication of acute pancreatitis. MiR155 may be involved in the occurrence and development of intestinal dysfunction mediated by acute pancreatitis, but the specific mechanism is not clear. AIMS To investigate the effect of miR155 on severe acute pancreatitis (SAP)-associated intestinal dysfunction and its possible mechanism in a mice model. METHODS In this study, SAP mice model was induced by intraperitoneal injection of caerulein and LPS in combination. Adeno-associated virus (AAV) was given by tail vein injection before the SAP model. The pancreatic and intestinal histopathology changes were analyzed. Cecal tissue was collected for 16S rRNA Gene Sequencing. Intestinal barrier proteins ZO-1 and E-cad were measured by Immunohistochemistry Staining and Western Blot, respectively. Intestinal tissue miR155 and inflammatory factors TNF-α, IL-1β, and IL-6 were detected by Q-PCR. The expression levels of protein associated with TNF-α and TLR4/MYD88 pathway in the intestinal were detected. RESULTS In miR155 overexpression SAP group, the levels of tissue inflammatory factor were significantly increased, intestinal barrier proteins were significantly decreased, and the injury of intestinal was aggravated. Bacterial 16S rRNA sequencing was performed, showing miR155 promotes gut microbiota dysbiosis. The levels of TNF-α, TLR4, and MYD88 in the intestinal were detected, suggesting that miR155 may regulate gut microbiota and activate the TLR4/MYD88 pathway, thereby affecting the release of inflammatory mediators and regulating SAP-related intestinal injury. After application of miR155-sponge, imbalance of intestinal flora and destruction of intestinal barrier-related proteins have been alleviated. The release of inflammatory mediators decreased, and the histopathology injury of intestinal was improved obviously. CONCLUSION MiR155 may play an important role in SAP-associated intestinal dysfunction. MiR155 can significantly alter the intestinal microecology, aggravated intestinal inflammation through TLR4/MYD88 pathway, and disrupts the intestinal barrier in SAP mice.
Collapse
|
37
|
Andrographolide Suppresses Pyroptosis in Mycobacterium tuberculosis-Infected Macrophages via the microRNA-155/Nrf2 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1885066. [PMID: 35528511 PMCID: PMC9072032 DOI: 10.1155/2022/1885066] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/11/2022] [Indexed: 11/17/2022]
Abstract
Tuberculosis (TB) remains a leading threat to public health worldwide with Mycobacterium tuberculosis (Mtb) infections causing long-term abnormal and excessive inflammatory responses, which in turn lead to lung damage and fibrosis, and ultimately death. Host-directed therapy (HDT) has been shown to be an effective anti-TB strategy in the absence of effective anti-TB drugs. Here, we used an in vitro macrophage model of Mtb infection to evaluate the effects of andrographolide (Andro), extracted from Andrographis paniculata, on pyroptosis in Mtb-infected macrophages. We evaluated the molecular mechanisms underlying these outcomes. These evaluations revealed that Andro downregulated the expression of proinflammatory miR-155-5p, which then promoted the expression of Nrf2 to suppress pyroptosis in Mtb-infected macrophages. Further study also demonstrated that siNrf2 could attenuate the inhibitory effect of Andro on TXNIP, validating our mechanistic studies. Thus, our data suggest that Andro may be a potential candidate adjuvant drug for anti-TB therapy as it inhibits pyroptosis in Mtb-infected macrophages, potentially improving clinical outcomes.
Collapse
|
38
|
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
|
39
|
Pattnaik B, Patnaik N, Mittal S, Mohan A, Agrawal A, Guleria R, Madan K. Micro RNAs as potential biomarkers in tuberculosis: A systematic review. Noncoding RNA Res 2022; 7:16-26. [PMID: 35128217 PMCID: PMC8792429 DOI: 10.1016/j.ncrna.2021.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB) remains a major infectious disease across the globe. With increasing TB infections and a rise in multi-drug resistance, rapid diagnostic modalities are required to achieve TB control. Radiological investigations and microbiological tests (microscopic examination, cartridge-based nucleic acid amplification tests, and cultures) are most commonly used to diagnose TB. Histopathological/cytopathological examinations are also required for an accurate diagnosis in many patients. The causative agent, Mycobacterium tuberculosis (Mtb), is known to circumvent the host's immune system. Circulating microRNAs (miRNAs) play a crucial role in biological pathways and can be used as a potential biomarker to detect tuberculosis. miRNAs are small non-coding RNAs and negatively regulate gene expression during post-transcriptional regulation. The differential expression of miRNAs in multiple clinical samples in tuberculosis patients may be helpful as potential disease biomarkers. This review summarizes the literature on miRNAs in various clinical samples as biomarkers for TB diagnosis.
Collapse
Affiliation(s)
- Bijay Pattnaik
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Niharika Patnaik
- Centre of Excellence in Asthma & Lung Disease, Molecular Immunogenetics Lab, CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Saurabh Mittal
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anant Mohan
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anurag Agrawal
- Centre of Excellence in Asthma & Lung Disease, Molecular Immunogenetics Lab, CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Randeep Guleria
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Karan Madan
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
40
|
Magdalena D, Magdalena G. Biological functions and diagnostic implications of microRNAs in Mycobacterium tuberculosis infection. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.333208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
41
|
Xu Y, Wu J, Yuan X, Liu W, Pan J, Xu B. MicroRNA-155 contributes to host immunity against Toxoplasma gondii. Parasite 2021; 28:83. [PMID: 34907898 PMCID: PMC8672677 DOI: 10.1051/parasite/2021082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 11/26/2021] [Indexed: 11/14/2022] Open
Abstract
Toxoplasma gondii is well known to infect almost all avian and mammalian species including humans, with worldwide distribution. This protozoan parasite can cause serious toxoplasmosis, posing with a risk to public health. The role of microRNAs in the pathogenesis of T. gondii has not been well described. The aim of the present study was to investigate the role of microRNA-155 (miR-155) in mediating innate and adaptive immune responses during T. gondii infection in mice models. The survival and parasite burden in T. gondii-infected miR-155−/− and wild-type (WT) C57BL6 mice were compared. In these two mouse models, ELISA tests were used for analysis of Th1-associated, Th2-associated, and Th17-associated cytokines, and flow cytometry was used for analysis of the subpopulations of NK, NKT, CD8+T, CD4+T cells and regulatory T cells (Tregs), as well as Ly6Chi inflammatory monocytes and dendritic cells. The lack of miR-155 led to increased parasite burden and decreased survival of infected mice in contrast to WT mice. Innate and adaptive immune responses were reduced in the absence of miR-155, along with decreased proinflammatory mediators, Th-1-associated and Th-2-associated cytokines and accumulation of lymphocyte subpopulations. Also, CD8+ T cell exhaustion was also worsened in the absence of miR-155 via targeting of SHIP-1 and SOCS1, showing as up-regulated recruitment of Tregs and expression of PD-1, and down-regulated expression of IFN-γ and TNF-α in CD8+ T cells. Our results show that miR-155 is a critical immune regulator for the control of T. gondii infection, suggesting that miR-155 can be explored as a potential molecular target for boosting immunity against T. gondii.
Collapse
Affiliation(s)
- Yanan Xu
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, PR China
| | - Junhua Wu
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, PR China
| | - Xiaoqi Yuan
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, PR China
| | - Wenyuan Liu
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, PR China
| | - Jiewen Pan
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, PR China
| | - Binbin Xu
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, PR China
| |
Collapse
|
42
|
Hou X, Zeng H, Chi X, Hu X. Pathogen Receptor Membrane-Coating Facet Structures Boost Nanomaterial Immune Escape and Antibacterial Performance. NANO LETTERS 2021; 21:9966-9975. [PMID: 34812644 DOI: 10.1021/acs.nanolett.1c03427] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nanomaterials show great potential for the treatment of bacterial infections, but their effects remain limited by low antibacterial efficiency and immune clearance. Facet-dependent nanozymes coated with pathogen receptor membranes were fabricated, providing an approach for producing superphotothermal antibacterial nanomaterials with high biocompatibility and low immune clearance. (100)- and (112)-Faceted CuFeSe2 presented excellent photothermal conversion efficiency (46%). However, the peroxidase-like activity of (100)-faceted CuFeSe2 enhanced the decomposition of H2O2 to hydroxyl radicals (•OH) and was markedly greater than that of (112)-faceted CuFeSe2, with nearly 100% of Staphylococcus aureus being killed under near-infrared (NIR) irradiation. Importantly, bacteria-pretreated immune membranes (i.e., pathogen receptor membranes) coated with CuFeSe2 exhibited superior S. aureus-binding ability, presented obvious immune-evading capability, and resulted in targeted delivery to infected sites. As a proof-of-principle demonstration, these findings hold promise for the use of pathogen receptor membrane-coated facet-dependent nanomaterials in clinical applications and the treatment of bacterial infections.
Collapse
Affiliation(s)
- Xuan Hou
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 30080, People's Republic of China
| | - Hui Zeng
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 30080, People's Republic of China
| | - Xue Chi
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 30080, People's Republic of China
| | - Xiangang Hu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 30080, People's Republic of China
| |
Collapse
|
43
|
Kazemi S, Afshar S, Karami M, Saidijam M, Keramat F, Hashemi SH, Alikhani MY. Association between risk of brucellosis and genetic variations in MicroRNA-146a. BMC Infect Dis 2021; 21:1070. [PMID: 34656082 PMCID: PMC8520608 DOI: 10.1186/s12879-021-06775-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 10/08/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) are the most common types of DNA changes in the human genome that leading to phenotypic differences in humans. MicroRNAs (miRNAs) are usually affected by various bacterial infections, and they are involved in controlling the immune responses. MicroRNA-146a (miR-146a) plays an essential role in the development of infectious and inflammatory diseases. The aim of the present study was to investigate the association between risk of brucellosis and genetic variations in miR-146a. METHODS This case-control study was conducted on 108 Brucellosis patients and 108 healthy controls. We genotyped two SNPs (rs2910164 and rs57095329) of the miR-146a using tetra-primer amplification refractory mutation system-polymerase chain reaction (T-ARMS-PCR) and restriction fragment length polymorphism-polymerase chain reaction (RFLP-PCR) methods. RESULTS The rs2910164 SNP was significantly associated with brucellosis in co-dominant [OR = 4.27, 95% CI = (2.35-7.79, P = 0.001] and dominant [OR = 3.52, 95% CI = (1.97-6.30, P = 0.001] models. Co-dominant (P = 0.047) and recessive (P = 0.018) models were significant at position rs57095329 between the two groups of patient and healthy. The A C haplotype (rs2910164 and rs57095329) was associated with brucellosis in the assessed population [OR (95% CI) = 1.98 (1.22-3.20), P = 0.0059]. CONCLUSIONS Consequently, our study demonstrated significant differences in genotype and haplotype frequencies of miR-146a variants between brucellosis patients and controls. Further studies on the larger sample sizes are required to verify the observed associations.
Collapse
Affiliation(s)
- Sima Kazemi
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Manoochehr Karami
- Department of Biostatistics and Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fariba Keramat
- Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Hamid Hashemi
- Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
44
|
Qu Y, Gao Q, Wu S, Xu T, Jiang D, Xu G. MicroRNA-142-3p inhibits autophagy and promotes intracellular survival of Mycobacterium tuberculosis by targeting ATG16L1 and ATG4c. Int Immunopharmacol 2021; 101:108202. [PMID: 34619495 DOI: 10.1016/j.intimp.2021.108202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/15/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Mycobacterium tuberculosis (M. tuberculosis), which parasitizes host macrophages and lead to cellular immunologic responses, such as autophagy and apoptosis. Several studies had indicated that autophagy played important roles in alleviating intracellular survival of M. tuberculosis by accelerating the maturation of phagosome. Previously, we found miR-142-3p was significantly down-regulated in the macrophages after infection with M. tuberculosis. However, the role of miR-142-3p in the regulation of autophagy and M. tuberculosis survival is elusive. METHODS Bioinformatics analysis was used to obtain target genes of miR-142-3p; the binding sites of ATG16L1 and ATG4c were further confirmed with dual luciferase reporter assay; RAW264.7 cells were infected with H37Ra and the expression of miR-142-3p was measured by qRT-PCR; the autophagic marker protein was detected by western blot as well as immunofluorescence microscopy and transmission electron microscopes analysis. RESULTS Overexpression of miR-142-3p significantly inhibited H37Ra-induced activation of autophagy, blocked the maturation of phagosome in macrophages and promoted M. tuberculosis survival in macrophages. Furthermore, miR-142-3p negatively-regulated expression of ATG16L1 and ATG4c by directly targeting its 3'-UTR, and meaningfully abated the level of autophagy. CONCLUSION These findings suggested that miR-142-3p inhibited M. tuberculosis-induced activation of autophagy and promoted H37Ra survival in RAW264.7 cells by targeting ATG16L1 and ATG4c.
Collapse
Affiliation(s)
- Yuliang Qu
- Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China; School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, PR China
| | - Qian Gao
- Department of Clinical Laboratory,Xijing Hospital,Air Force Military Medical University, Xi'an, 710032, PR China
| | - Shan Wu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, PR China
| | - Tao Xu
- Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Dan Jiang
- Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China; School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, PR China
| | - Guangxian Xu
- Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China; School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, PR China.
| |
Collapse
|
45
|
Llibre A, Grudzinska FS, O'Shea MK, Duffy D, Thickett DR, Mauro C, Scott A. Lactate cross-talk in host-pathogen interactions. Biochem J 2021; 478:3157-3178. [PMID: 34492096 PMCID: PMC8454702 DOI: 10.1042/bcj20210263] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Lactate is the main product generated at the end of anaerobic glycolysis or during the Warburg effect and its role as an active signalling molecule is increasingly recognised. Lactate can be released and used by host cells, by pathogens and commensal organisms, thus being essential for the homeostasis of host-microbe interactions. Infection can alter this intricate balance, and the presence of lactate transporters in most human cells including immune cells, as well as in a variety of pathogens (including bacteria, fungi and complex parasites) demonstrates the importance of this metabolite in regulating host-pathogen interactions. This review will cover lactate secretion and sensing in humans and microbes, and will discuss the existing evidence supporting a role for lactate in pathogen growth and persistence, together with lactate's ability to impact the orchestration of effective immune responses. The ubiquitous presence of lactate in the context of infection and the ability of both host cells and pathogens to sense and respond to it, makes manipulation of lactate a potential novel therapeutic strategy. Here, we will discuss the preliminary research that has been carried out in the context of cancer, autoimmunity and inflammation.
Collapse
Affiliation(s)
- Alba Llibre
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
- Translational Immunology Laboratory, Institut Pasteur, Paris, France
| | - Frances S Grudzinska
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Matthew K O'Shea
- Department of Infection, University Hospitals Birmingham NHS Foundation Trust, Birmingham, U.K
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, U.K
| | - Darragh Duffy
- Translational Immunology Laboratory, Institut Pasteur, Paris, France
| | - David R Thickett
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Aaron Scott
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| |
Collapse
|
46
|
Stimulus-specific responses in innate immunity: Multilayered regulatory circuits. Immunity 2021; 54:1915-1932. [PMID: 34525335 DOI: 10.1016/j.immuni.2021.08.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/07/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Immune sentinel cells initiate immune responses to pathogens and tissue injury and are capable of producing highly stimulus-specific responses. Insight into the mechanisms underlying such specificity has come from the identification of regulatory factors and biochemical pathways, as well as the definition of signaling circuits that enable combinatorial and temporal coding of information. Here, we review the multi-layered molecular mechanisms that underlie stimulus-specific gene expression in macrophages. We categorize components of inflammatory and anti-pathogenic signaling pathways into five layers of regulatory control and discuss unifying mechanisms determining signaling characteristics at each layer. In this context, we review mechanisms that enable combinatorial and temporal encoding of information, identify recurring regulatory motifs and principles, and present strategies for integrating experimental and computational approaches toward the understanding of signaling specificity in innate immunity.
Collapse
|
47
|
Fatima S, Kumari A, Agarwal M, Pahuja I, Yadav V, Dwivedi VP, Bhaskar A. Epigenetic code during mycobacterial infections: therapeutic implications for tuberculosis. FEBS J 2021; 289:4172-4191. [PMID: 34453865 DOI: 10.1111/febs.16170] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/23/2021] [Accepted: 08/26/2021] [Indexed: 12/21/2022]
Abstract
Epigenetics involves changing the gene function without any change in the sequence of the genes. In the case of tuberculosis (TB) infections, the bacilli, Mycobacterium tuberculosis (M.tb), uses epigenetics as a tool to protect itself from the host immune system. TB is a deadly disease-causing maximum death per year due to a single infectious agent. In the case of TB, there is an urgent need for novel host-directed therapies which can effectively target the survival and long-term persistence of the bacteria without developing drug resistance in the bacterial strains while also reducing the duration and toxicity associated with the mainstream anti-TB drugs. Recent studies have suggested that TB infection has a significant effect on the host epigenome thereby manipulating the host immune response in the favor of the pathogen. M.tb alters the activation status of key genes involved in the immune response against TB to promote its survival and subvert the antibacterial strategies of the host. These changes are reversible and can be exploited to design very efficient host-directed therapies to fight against TB. This review has been written with the purpose of discussing the role of epigenetic changes in TB pathogenesis and the therapeutic approaches involving epigenetics, which can be utilized for targeting the pathogen.
Collapse
Affiliation(s)
- Samreen Fatima
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Anjna Kumari
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Meetu Agarwal
- Department of Biosciences, Jamia Hamdard University, New Delhi, India
| | - Isha Pahuja
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Vinod Yadav
- Department of Microbiology, Central University of Haryana, Mahendragarh, India
| | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ashima Bhaskar
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
48
|
Yuan Z, Prasla Z, Lee FEH, Bedi B, Sutliff RL, Sadikot RT. MicroRNA-155 Modulates Macrophages' Response to Non-Tuberculous Mycobacteria through COX-2/PGE2 Signaling. Pathogens 2021; 10:920. [PMID: 34451384 PMCID: PMC8398909 DOI: 10.3390/pathogens10080920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/01/2021] [Accepted: 07/17/2021] [Indexed: 12/19/2022] Open
Abstract
Non-tuberculous mycobacteria (NTM) have been recognized as a causative agent of various human diseases, including severe infections in immunocompromised patients, such as people living with HIV. The most common species identified is the Mycobacterium avium-intracellulare complex (MAI/MAC), accounting for a majority of infections. Despite abundant information detailing the clinical significance of NTM, little is known about host-pathogen interactions in NTM infection. MicroRNAs (miRs) serve as important post-transcriptional regulators of gene expression. Using a microarray profile, we found that the expression of miR-155 and cyclo-oxygenase 2 (COX-2) is significantly increased in bone-marrow-derived macrophages from mice and human monocyte-derived macrophages from healthy volunteers that are infected with NTM. Antagomir against miR-155 effectively suppressed expression of COX-2 and reduced Prostaglandin E2(PGE2) secretion, suggesting that COX-2/PGE2 expression is dependent on miR-155. Mechanistically, we found that inhibition of NF-κB activity significantly reduced miR-155/COX-2 expression in infected macrophages. Most importantly, blockade of COX-2, E-prostanoid receptors (EP2 and EP4) enhanced killing of MAI in macrophages. These findings provide novel mechanistic insights into the role of miR-155/COX-2/PGE2 signalling and suggest that induction of these pathways enhances survival of mycobacteria in macrophages. Defining host-pathogen interactions can lead to novel immunomodulatory therapies for NTM infections which are difficult to treat.
Collapse
Affiliation(s)
- Zhihong Yuan
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zohra Prasla
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA; (Z.P.); (F.E.-H.L.); (B.B.); (R.L.S.)
| | - Frances Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA; (Z.P.); (F.E.-H.L.); (B.B.); (R.L.S.)
| | - Brahmchetna Bedi
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA; (Z.P.); (F.E.-H.L.); (B.B.); (R.L.S.)
| | - Roy L. Sutliff
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA; (Z.P.); (F.E.-H.L.); (B.B.); (R.L.S.)
| | - Ruxana T. Sadikot
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
49
|
Jiang Q, Shi L. Coordination of the Uptake and Metabolism of Amino Acids in Mycobacterium tuberculosis-Infected Macrophages. Front Immunol 2021; 12:711462. [PMID: 34326848 PMCID: PMC8315098 DOI: 10.3389/fimmu.2021.711462] [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: 05/18/2021] [Accepted: 06/25/2021] [Indexed: 01/31/2023] Open
Abstract
Macrophage polarization to the M1-like phenotype, which is critical for the pro-inflammatory and antimicrobial responses of macrophages against intracellular pathogens, is associated with metabolic reprogramming to the Warburg effect and a high output of NO from increased expression of NOS2. However, there is limited understanding about the uptake and metabolism of other amino acids during M1 polarization. Based on functional analysis of a group of upregulated transporters and enzymes involved in the uptake and/or metabolism of amino acids in Mycobacterium tuberculosis-infected macrophages, plus studies of immune cell activation, we postulate a coherent scheme for amino acid uptake and metabolism during macrophage polarization to the M1-like phenotype. We describe potential mechanisms that the increased arginine metabolism by NOS2 is metabolically coupled with system L transporters LAT1 and LAT2 for the uptake of neutral amino acids, including those that drive mTORC1 signaling toward the M1-like phenotype. We also discuss the underappreciated pleiotropic roles of glutamine metabolism in the metabolic reprogramming of M1-like macrophages. Collectively, our analyses argue that a coordinated amino acid uptake and metabolism constitutes an integral component of the broad metabolic scheme required for macrophage polarization to M1-like phenotype against M. tuberculosis infection. This idea could stimulate future experimental efforts to elucidate the metabolic map of macrophage activation for the development of anti-tuberculosis therapies.
Collapse
Affiliation(s)
- Qingkui Jiang
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers The State University of New Jersey, Newark, NJ, United States
| | - Lanbo Shi
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
50
|
Sampath P, Periyasamy KM, Ranganathan UD, Bethunaickan R. Monocyte and Macrophage miRNA: Potent Biomarker and Target for Host-Directed Therapy for Tuberculosis. Front Immunol 2021; 12:667206. [PMID: 34248945 PMCID: PMC8267585 DOI: 10.3389/fimmu.2021.667206] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
The end TB strategy reinforces the essentiality of readily accessible biomarkers for early tuberculosis diagnosis. Exploration of microRNA (miRNA) and pathway analysis opens an avenue for the discovery of possible therapeutic targets. miRNA is a small, non-coding oligonucleotide characterized by the mechanism of gene regulation, transcription, and immunomodulation. Studies on miRNA define their importance as an immune marker for active disease progression and as an immunomodulator for innate mechanisms, such as apoptosis and autophagy. Monocyte research is highly advancing toward TB pathogenesis and biomarker efficiency because of its innate and adaptive response connectivity. The combination of monocytes/macrophages and their relative miRNA expression furnish newer insight on the unresolved mechanism for Mycobacterium survival, exploitation of host defense, latent infection, and disease resistance. This review deals with miRNA from monocytes, their relative expression in different disease stages of TB, multiple gene regulating mechanisms in shaping immunity against tuberculosis, and their functionality as biomarker and host-mediated therapeutics. Future collaborative efforts involving multidisciplinary approach in various ethnic population with multiple factors (age, gender, mycobacterial strain, disease stage, other chronic lung infections, and inflammatory disease criteria) on these short miRNAs from body fluids and cells could predict the valuable miRNA biosignature network as a potent tool for biomarkers and host-directed therapy.
Collapse
Affiliation(s)
- Pavithra Sampath
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | | | - Uma Devi Ranganathan
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | | |
Collapse
|