1
|
Mehta P, Mazumder S. miR-146a is critical for orchestrating Mycobacterium fortuitum survival through anti-inflammatory and M2 macrophage responses in fish. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110271. [PMID: 40081436 DOI: 10.1016/j.fsi.2025.110271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
The significance of microRNAs (miRNAs) in host response to non-tuberculoid mycobacteria like Mycobacterium fortuitum remains nascent. Using zebrafish kidney macrophages (ZFKM), we elucidate a novel function of miR-146a, orchestrated by the TLR-2-PI3K-NF-κB pathway, in M. fortuitum pathogenesis. We demonstrate that miR-146a facilitates anti-inflammatory response by targeting IRAK-1 and TRAF-6 in M. fortuitum-infected ZFKM. Moreover, miR-146a mitigates M1 macrophage activity by suppressing the iNOS-NO axis while enhancing M2-specific TGF-β mRNA expression and subsequent inhibition of M. fortuitum eradication. These findings collectively suggest that miR-146a diminishes macrophage-mediate M. fortuitum clearance. Our study provides novel insights into the intricate interplay between miRNAs and mycobacterial infections. We propose a mechanistic model wherein the TLR-2/NF-κB axis initiates miR-146a expression, which, in turn, suppresses irak-1 and traf-6, fostering the development of M2 macrophages. Consequently, this creates an anti-inflammatory environment conducive to M. fortuitumsurvival. Our findings provide novel insights into the intricate interplay between miRNAs and mycobacterial persistence, a concerning aspect of pathogenesis.
Collapse
Affiliation(s)
- Priyanka Mehta
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
2
|
Guo Z, Zhou Y, Li N, Shen A, Jia Y, Yin R, Yang J, Yuan J, Yin R. The Molecular Mechanism by Which miR-129a-3p Targets the TLR4/NF-κB Signaling Pathway to Regulate Inflammatory Damage in 3D4/21 Cells Infected with Glaesserella parasuis. Animals (Basel) 2025; 15:1355. [PMID: 40427234 PMCID: PMC12108277 DOI: 10.3390/ani15101355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Glaesserella parasuis (G. parasuis) is the primary pathogen responsible for Glässer's disease and poses a significant threat to the global pig industry. MicroRNAs are a class of short, endogenous, single-stranded noncoding RNAs that play crucial roles in inflammation, apoptosis, proliferation, differentiation, and invasion in various organisms. This study analyzed the characteristics of porcine alveolar macrophage (PAM) cells infected with G. parasuis through the knockdown and overexpression of ssc-miR-129a-3p. We constructed a cellular model with ssc-miR-129a-3p knockdown invaded by G. parasuis strain XX0306, screening 160 differentially expressed genes via high-throughput sequencing. GO enrichment analysis revealed that 376 GO entries were enriched. KEGG enrichment analysis found that mRNA target genes were enriched in 17 cell signaling pathways, including G protein-coupled components, PPAR, and other signaling pathways that can mediate inflammatory pathways. By examining the expression of relevant inflammatory factors and signaling pathways, we elucidated the molecular mechanisms through which ssc-miR-129a-3p targets the TLR4/NF-κB signaling pathway to regulate inflammatory injury. This study establishes a foundation for further research into the role of miRNA in the pathogenesis of Glässer disease and is highly significant for the prevention and control of bacterial diseases within the pig industry.
Collapse
Affiliation(s)
- Zhongbo Guo
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| | - Yuanyuan Zhou
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| | - Na Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| | - Aobo Shen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| | - Yongchao Jia
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| | - Ronglan Yin
- Research Academy of Animal Husbandry and Veterinary Medicine Sciences of Jilin Province, Changchun 130062, China;
| | - Junjie Yang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| | - Jing Yuan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| | - Ronghuan Yin
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; (Z.G.); (Y.Z.); (N.L.); (A.S.); (Y.J.); (J.Y.)
| |
Collapse
|
3
|
Qiu GH, Fu M, Zheng X, Huang C. Protection of the genome and the central exome by peripheral non-coding DNA against DNA damage in health, ageing and age-related diseases. Biol Rev Camb Philos Soc 2025; 100:508-529. [PMID: 39327815 DOI: 10.1111/brv.13151] [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: 09/26/2023] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
DNA in eukaryotic genomes is under constant assault from both exogenous and endogenous sources, leading to DNA damage, which is considered a major molecular driver of ageing. Fortunately, the genome and the central exome are safeguarded against these attacks by abundant peripheral non-coding DNA. Non-coding DNA codes for small non-coding RNAs that inactivate foreign nucleic acids in the cytoplasm and physically blocks these attacks in the nucleus. Damage to non-coding DNA produced during such blockage is removed in the form of extrachromosomal circular DNA (eccDNA) through nucleic pore complexes. Consequently, non-coding DNA serves as a line of defence for the exome against DNA damage. The total amount of non-coding DNA/heterochromatin declines with age, resulting in a decrease in both physical blockage and eccDNA exclusion, and thus an increase in the accumulation of DNA damage in the nucleus during ageing and in age-related diseases. Here, we summarize recent evidence supporting a protective role of non-coding DNA in healthy and pathological states and argue that DNA damage is the proximate cause of ageing and age-related genetic diseases. Strategies aimed at strengthening the protective role of non-coding DNA/heterochromatin could potentially offer better systematic protection for the dynamic genome and the exome against diverse assaults, reduce the burden of DNA damage to the exome, and thus slow ageing, counteract age-related genetic diseases and promote a healthier life for individuals.
Collapse
Affiliation(s)
- Guo-Hua Qiu
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| | - Mingjun Fu
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| | - Xintian Zheng
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| | - Cuiqin Huang
- College of Life Sciences, Longyan University, Longyan, 364012, People's Republic of China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Engineering Research Center for the Prevention and Control of Animal-Origin Zoonosis, Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities, Longyan, People's Republic of China
| |
Collapse
|
4
|
Kaltenpoth M, Flórez LV, Vigneron A, Dirksen P, Engl T. Origin and function of beneficial bacterial symbioses in insects. Nat Rev Microbiol 2025:10.1038/s41579-025-01164-z. [PMID: 40148601 DOI: 10.1038/s41579-025-01164-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Beneficial bacterial symbionts are widespread in insects and affect the fitness of their hosts by contributing to nutrition, digestion, detoxification, communication or protection from abiotic stressors or natural enemies. Decades of research have formed our understanding of the identity, localization and functional benefits of insect symbionts, and the increasing availability of genome sequences spanning a diversity of pathogens and beneficial bacteria now enables comparative approaches of their metabolic features and their phylogenetic affiliations, shedding new light on the origin and function of beneficial symbioses in insects. In this Review, we explore the symbionts' metabolic traits that can provide benefits to insect hosts and discuss the evolutionary paths to the formation of host-beneficial symbiotic associations. Phylogenetic analyses and molecular studies reveal that extracellular symbioses colonizing cuticular organs or the digestive tract evolved from a broad diversity of bacterial partners, whereas intracellular beneficial symbionts appear to be restricted to a limited number of lineages within the Gram-negative bacteria and probably originated from parasitic ancestors. To unravel the general principles underlying host-symbiont interactions and recapitulate the early evolutionary steps leading towards beneficial symbioses, future efforts should aim to establish more symbiotic systems that are amenable to genetic manipulation and experimental evolution.
Collapse
Affiliation(s)
- Martin Kaltenpoth
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, Jena, Germany.
- Evolutionary Ecology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University, Mainz, Germany.
| | - Laura V Flórez
- Evolutionary Ecology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University, Mainz, Germany
- Section for Organismal Biology, Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Aurélien Vigneron
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, Jena, Germany
- Evolutionary Ecology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University, Mainz, Germany
- Universite Claude Bernard Lyon 1, Laboratoire d'Ecologie Microbienne, UMR CNRS 5557, UMR INRAE 1418, VetAgro Sup, Villeurbanne, France
| | - Philipp Dirksen
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, Jena, Germany
- Evolutionary Ecology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University, Mainz, Germany
| | - Tobias Engl
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, Jena, Germany
- Evolutionary Ecology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
5
|
Nai S, Song J, Su W, Liu X. Bidirectional Interplay Among Non-Coding RNAs, the Microbiome, and the Host During Development and Diseases. Genes (Basel) 2025; 16:208. [PMID: 40004537 PMCID: PMC11855195 DOI: 10.3390/genes16020208] [Citation(s) in RCA: 1] [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: 12/30/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
It is widely known that the dysregulation of non-coding RNAs (ncRNAs) and dysbiosis of the gut microbiome play significant roles in host development and the progression of various diseases. Emerging evidence has highlighted the bidirectional interplay between ncRNAs and the gut microbiome. This article aims to review the current understanding of the molecular mechanisms underlying the crosstalk between ncRNAs, especially microRNA (miRNA), and the gut microbiome in the context of development and diseases, such as colorectal cancer, inflammatory bowel diseases, neurological disorders, obesity, and cardiovascular disease. Ultimately, this review seeks to provide a foundation for exploring the potential roles of ncRNAs and gut microbiome interactions as biomarkers and therapeutic targets for clinical diagnosis and treatment, such as ncRNA mimics, antisense oligonucleotides, and small-molecule compounds, as well as probiotics, prebiotics, and diets.
Collapse
Affiliation(s)
| | | | | | - Xiaoqian Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (S.N.); (J.S.); (W.S.)
| |
Collapse
|
6
|
Wang L, Xi M, Cao W, Qin H, Qin D, Chen S, Zhou S, Hou Y, Chen Y, Xiao X, Zheng Q, Li D, Li Y. Electroacupuncture alleviates functional constipation by upregulating host-derived miR-205-5p to modulate gut microbiota and tryptophan metabolism. Front Microbiol 2025; 16:1517018. [PMID: 39973939 PMCID: PMC11835812 DOI: 10.3389/fmicb.2025.1517018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/07/2025] [Indexed: 02/21/2025] Open
Abstract
Electroacupuncture (EA) has shown promise as a treatment for Functional constipation (FC), with growing evidence suggesting it may enhance gut motility. MicroRNAs (miRNAs) serve as key regulatory molecules mediating host-microbiota interactions. However, the specific fecal miRNAs regulating microbiota composition and metabolism in EA-treated constipated mice, along with their key targets, remain unidentified. We examined fecal microbiome composition, metabolism, and colonic miRNA expression in loperamide-induced constipated mice and EA-treated mice to identify differentially expressed miRNAs and assess their relationships with microbial abundance, metabolism, and gut motility. An antibiotic cocktail and adeno-associated virus were employed to interfere with the gut microbiota and target miRNA in vivo, thereby validating the proposed mechanism. Our results indicate that miR-205-5p, significantly upregulated in fecal and colonic tissues of EA-treated constipated mice, promotes intestinal motility in a microbiome-dependent manner. Specifically, EA promoted the growth of Lactobacillus reuteri, enriched in the feces of constipation-recovered mice, through host-derived miR-205-5p regulation. Furthermore, Lactobacillus reuteri and its tryptophan metabolites (indole-3-acetamide, indole-3-acetic acid, and indole-3-carboxaldehyde) alleviated loperamide-induced constipation. These findings underscore the pivotal role of host-derived miR-205-5p in modulating microbial composition and tryptophan metabolites to enhance intestinal motility through EA.
Collapse
Affiliation(s)
- Lu Wang
- Department of Acupuncture, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Menghan Xi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Department of Acupuncture and Moxibustion, Chengdu Pidu District Hospital of TCM/The Third Clinical Medical College of Chengdu University of TCM, Chengdu, Sichuan, China
| | - Wei Cao
- Center of Preventive Medicine, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, Sichuan, China
| | - Haiyan Qin
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Di Qin
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuai Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Siyuan Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yujun Hou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ying Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xianjun Xiao
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qianhua Zheng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Dehua Li
- Department of Acupuncture, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ying Li
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Entrenas-García C, Suárez-Cárdenas JM, Fernández-Rodríguez R, Bautista R, Claros MG, Garrido JJ, Zaldívar-López S. miR-215 Modulates Ubiquitination to Impair Inflammasome Activation and Autophagy During Salmonella Typhimurium Infection in Porcine Intestinal Cells. Animals (Basel) 2025; 15:431. [PMID: 39943201 PMCID: PMC11815736 DOI: 10.3390/ani15030431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
The host response to S. Typhimurium infection can be post-transcriptionally regulated by miRNAs. In this study, we investigated the role of miR-215 using both in vivo porcine infection models and in vitro intestinal epithelial cell lines. Several miRNAs were found to be dysregulated in the porcine ileum during infection with wild-type and SPI2-defective mutant strains of S. Typhimurium, with some changes being SPI2-dependent. Notably, miR-215 was significantly downregulated during infection. To explore its functional role, gain-of-function experiments were performed by transfecting porcine intestinal epithelial cells (IPEC-J2) with a miR-215-5p mimic, followed by label-free quantitative (LFQ) proteomic analysis. This analysis identified 157 proteins, of which 35 were downregulated in response to miR-215 overexpression, suggesting they are potential targets of this miRNA. Among these, E2 small ubiquitin-like modifier (SUMO)-conjugating enzyme UBC9 and E3 ubiquitin-ligase HUWE1 were identified as key targets, both of which are upregulated during S. Typhimurium infection. The miR-215-mediated downregulation of these proteins resulted in a significant decrease in overall ubiquitination, a process crucial for regulating inflammasome activation and autophagy. Consistently, inflammasome markers caspase 1 (CASP1) and apoptosis-associated speck-like protein containing a CARD (ASC), as well as autophagy markers microtubule-associated protein 1A/1B-light chain 3 (LC3B) and Ras-related protein Rab-11 (RAB11A), showed decreased expression in miR-215 mimic-transfected and infected IPEC-J2 cells. To further validate these findings, human intestinal epithelial cells (HT29) were used as a complementary model, providing additional insights into conserved immune pathways and extending the observations made in the porcine system. Overall, our findings demonstrate that miR-215 plays a significant role in modulating host inflammasome activation and autophagy by targeting proteins involved in ubiquitination during S. Typhimurium infection.
Collapse
Affiliation(s)
- Carmen Entrenas-García
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
| | - José M. Suárez-Cárdenas
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Raúl Fernández-Rodríguez
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Rocío Bautista
- Plataforma Andaluza de Bioinformática, Supercomputing and Bioinnovation Center (SCBI), Universidad de Málaga, 29590 Malaga, Spain; (R.B.); (M.G.C.)
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, 29590 Malaga, Spain
| | - M. Gonzalo Claros
- Plataforma Andaluza de Bioinformática, Supercomputing and Bioinnovation Center (SCBI), Universidad de Málaga, 29590 Malaga, Spain; (R.B.); (M.G.C.)
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, 29590 Malaga, Spain
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM-UMA-CSIC), 29590 Malaga, Spain
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, 29010 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER) U741, 29071 Malaga, Spain
| | - Juan J. Garrido
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Sara Zaldívar-López
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| |
Collapse
|
8
|
Doghish AS, Elazazy O, Mohamed HH, Mansour RM, Ghanem A, Faraag AHI, Elballal MS, Elrebehy MA, Elesawy AE, Abdel Mageed SS, Saber S, Nassar YA, Abulsoud AI, Abdel-Reheim MA, Elawady AS, Ali MA, Basiouny MS, Hemdan M, Lutfy RH, Awad FA, El-Sayed SA, Ashour MM, El-Sayyad GS, Mohammed OA. A Review on miRNAs in Enteric Bacteria-mediated Host Pathophysiology: Mechanisms and Implications. J Biochem Mol Toxicol 2025; 39:e70160. [PMID: 39907181 DOI: 10.1002/jbt.70160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/22/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025]
Abstract
Recently, many studies focused on the billions of native bacteria found inside and all over the human body, commonly known as the microbiota, and its interactions with the eukaryotic host. One of the niches for such microbiota is the gastrointestinal tract (GIT), which harbors hundreds to thousands of bacterial species commonly known as enteric bacteria. Changes in the enteric bacterial populations were linked to various pathologies such as irritable bowel syndrome and obesity. The gut microbiome could affect the health status of individuals. MicroRNAs (miRNAs) are one of the extensively studied small-sized noncoding RNAs (ncRNAs) over the past decade to explore their multiple roles in health and disease. It was proven that miRNAs circulate in almost all body fluids and tissues, showing signature patterns of dysregulation associated with pathologies. Both cellular and circulating miRNAs participate in the posttranscriptional regulation of genes and are considered the potential key regulators of genes and participate in cellular communication. This manuscript explores the unique interplay between miRNAs and enteric bacteria in the gastrointestinal tract, emphasizing their dual role in shaping host-microbiota dynamics. It delves into the molecular mechanisms by which miRNAs influence bacterial colonization and host immune responses, linking these findings to gut-related diseases. The review highlights innovative therapeutic and diagnostic opportunities, offering insights for targeted treatments of dysbiosis-associated pathologies.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Hend H Mohamed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
- Biochemistry Department, Faculty of Science, Cairo University, Giza, 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, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Ahmed H I Faraag
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Medical Department, School of Biotechnology, Badr University in Cairo, Badr City, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Galala University, New Galala City, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Yara A Nassar
- Department of Botany, Faculty of Science, Biotechnology and Its Application Program, Mansoura University, Mansoura, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
| | | | - Alaa S Elawady
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Ali
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | | | - Mohamed Hemdan
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Radwa H Lutfy
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Farah A Awad
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Salma A El-Sayed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Mohamed M Ashour
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Gharieb S El-Sayyad
- Medical Laboratory Technology Department, Faculty of Applied Health Sciences Technology, Badr University in Cairo (BUC), Cairo, Egypt
- Microbiology and Immunology Department, Faculty of Pharmacy, Galala University, New Galala city, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
9
|
Zhang L, Ma D, Yu Y, Luo W, Jiang S, Feng S, Chen Z. Advances in biomacromolecule-functionalized magnetic particles for phytopathogen detection. Talanta 2025; 281:126876. [PMID: 39277940 DOI: 10.1016/j.talanta.2024.126876] [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: 04/05/2024] [Revised: 08/20/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Due to the increasing crop losses caused by common and newly emerging phytopathogens, there is a pressing need for the development of rapid and reliable methods for phytopathogen detection and analysis. Leveraging advancements in biochemical engineering technologies and nanomaterial sciences, researchers have put considerable efforts on utilizing biofunctionalized magnetic micro- and nanoparticles (MPs) to develop rapid and reliable systems for phytopathogen detection. MPs facilitate the rapid, high-throughput analysis and in-field applications, while the biomacromolecules, which play key roles in the biorecognitions, interactions and signal amplification, determine the specificity, sensitivity, reliability, and portability of pathogen detection systems. The integration of MPs and biomacromolecules provides dimensionality- and composition-dependent properties, representing a novel approach to develop phytopathogen detection systems. In this review, we summarize and discuss the general properties, synthesis and characterization of MPs, and focus on biomacromolecule-functionalized MPs as well as their representative applications for phytopathogen detection and analysis reported over the past decade. Extensively studied bioreceptors, such as antibodies, phages and phage proteins, nucleic acids, and glycans that are involved in the recognitions and interactions, are covered and discussed. Additionally, the integration of MPs-based detection system with portable microfluidic devices to facilitate their in-field applications is also discussed. Overall, this review focuses on biomacromolecule-functionalized MPs and their applications for phytopathogen detection, aiming to highlight their potential in developing advanced biosensing systems for effective plant protection.
Collapse
Affiliation(s)
- Libo Zhang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang, Guizhou, 550025, China.
| | - Dumei Ma
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Youbo Yu
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang, Guizhou, 550025, China
| | - Wiewei Luo
- The Ninth Medical Center of Chinese PLA General Hospital, Chaoyang District, Beijing, 100101, China
| | - Shilong Jiang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang, Guizhou, 550025, China
| | - Sheng Feng
- Department of Pathology and Laboratory Medicine, Boston University, Boston, MA, 02118, USA
| | - Zhuo Chen
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
10
|
Kreis V, Toffano-Nioche C, Denève-Larrazet C, Marvaud JC, Garneau JR, Dumont F, van Dijk EL, Jaszczyszyn Y, Boutserin A, D'Angelo F, Gautheret D, Kansau I, Janoir C, Soutourina O. Dual RNA-seq study of the dynamics of coding and non-coding RNA expression during Clostridioides difficile infection in a mouse model. mSystems 2024; 9:e0086324. [PMID: 39601557 DOI: 10.1128/msystems.00863-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Clostridioides difficile is the leading cause of healthcare-associated diarrhea in industrialized countries. Many questions remain to be answered about the mechanisms governing its interaction with the host during infection. Non-coding RNAs (ncRNAs) contribute to shape virulence in many pathogens and modulate host responses; however, their role in C. difficile infection (CDI) has not been explored. To better understand the dynamics of ncRNA expression contributing to C. difficile infectious cycle and host response, we used a dual RNA-seq approach in a conventional murine model. From the pathogen side, this transcriptomic analysis revealed the upregulation of virulence factors, metabolism, and sporulation genes, as well as the identification of 61 ncRNAs differentially expressed during infection that correlated with the analysis of available raw RNA-seq data sets from two independent studies. From these data, we identified 118 potential new transcripts in C. difficile, including 106 new ncRNA genes. From the host side, we observed the induction of several pro-inflammatory pathways, and among the 185 differentially expressed ncRNAs, the overexpression of microRNAs (miRNAs) previously associated to inflammatory responses or unknown long ncRNAs and miRNAs. A particular host gene expression profile could be associated to the symptomatic infection. In accordance, the metatranscriptomic analysis revealed specific microbiota changes accompanying CDI and specific species associated with symptomatic infection in mice. This first adaptation of in vivo dual RNA-seq to C. difficile contributes to unravelling the regulatory networks involved in C. difficile infectious cycle and host response and provides valuable resources for further studies of RNA-based mechanisms during CDI.IMPORTANCEClostridioides difficile is a major cause of nosocomial infections associated with antibiotic therapy classified as an urgent antibiotic resistance threat. This pathogen interacts with host and gut microbial communities during infection, but the mechanisms of these interactions remain largely to be uncovered. Noncoding RNAs contribute to bacterial virulence and host responses, but their expression has not been explored during C. difficile infection. We took advantage of the conventional mouse model of C. difficile infection to look simultaneously to the dynamics of gene expression in pathogen, its host, and gut microbiota composition, providing valuable resources for future studies. We identified a number of ncRNAs that could mediate the adaptation of C. difficile inside the host and the crosstalk with the host immune response. Promising inflammation markers and potential therapeutic targets emerged from this work open new directions for RNA-based and microbiota-modulatory strategies to improve the efficiency of C. difficile infection treatments.
Collapse
Affiliation(s)
- Victor Kreis
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Claire Toffano-Nioche
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | | | | | | | | | - Erwin L van Dijk
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Yan Jaszczyszyn
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anaïs Boutserin
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Francesca D'Angelo
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Daniel Gautheret
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Imad Kansau
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Orsay, France
| | - Claire Janoir
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Orsay, France
| | - Olga Soutourina
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
11
|
Zou J, Xu B, Luo P, Chen T, Duan H. Non-coding RNAs in bladder cancer, a bridge between gut microbiota and host? Front Immunol 2024; 15:1482765. [PMID: 39628486 PMCID: PMC11611751 DOI: 10.3389/fimmu.2024.1482765] [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: 08/18/2024] [Accepted: 10/30/2024] [Indexed: 12/06/2024] Open
Abstract
In recent years, the role of gut microbiota (GM) in bladder cancer has attracted significant attention. Research indicates that GM not only contributes to bladder carcinogenesis but also influences the efficacy of adjuvant therapies for bladder cancer. Despite this, interventions targeting GM have not been widely employed in the prevention and treatment of bladder cancer, mainly due to the incomplete understanding of the complex interactions between the host and gut flora. Simultaneously, aberrantly expressed non-coding RNAs (ncRNAs) have been frequently associated with bladder cancer, playing crucial roles in processes such as cell proliferation, invasion, and drug resistance. It is widely known that the regulation of GM-mediated host pathophysiological processes is partly regulated through epigenetic pathways. At the same time, ncRNAs are increasingly regarded as GM signaling molecules involved in GM-mediated epigenetic regulation. Accordingly, this review analyzes the ncRNAs that are closely related to the GM in the context of bladder cancer occurrence and treatment, and summarizes the role of their interaction with the GM in bladder cancer-related phenotypes. The aim is to delineate a regulatory network between GM and ncRNAs and provide a new perspective for the study and prevention of bladder cancer.
Collapse
Affiliation(s)
- Jun Zou
- Department of Otorhinolaryngology, The Affiliated Fengcheng Hospital of Yichun University, Fengcheng, Jiangxi, China
| | - Baisheng Xu
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| | - Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Huanglin Duan
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| |
Collapse
|
12
|
Ma Y, Zhou Z, Luo T, Meng Q, Wang H, Li X, Gu W, Zhou J, Meng Q. Rab7 GTPase, a direct target of miR-131-3p, limits intracellular Spiroplasma eriocheiris infection by modulating phagocytosis. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109879. [PMID: 39244074 DOI: 10.1016/j.fsi.2024.109879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Spiroplasma eriocheiris is a kind of intracellular pathogen without cell wall and the causative agent of Chinese mitten crab Eriocheir sinensis "tremor disease", which causes significant economic losses in the crustacean aquaculture. However, little is known about the intracellular transport of this pathogen and host innate immune response to this pathogen. Rab GTPases are key regulators for endocytosis and intracellular pathogen trafficking. In this study, we showed that S. eriocheiris infection upregulated the transcription of Rab7 through the downregulation of miR-131-3p. Subsequently, both hemocytes transfected with miR-131-3p mimics and hemocytes derived from Rab7 knockdown crabs exhibited reduced phagocytic activities and increased susceptibility to S. eriocheiris infection. Additionally, Rab7 could interact with the cell shape-determining protein MreB3 of S. eriocheiris, and its overexpression promoted S. eriocheiris internalization and fusion with lysosomes, thereby limiting S. eriocheiris replication in Drosophila S2 cells. Overall, these results demonstrated that Rab7 facilitated host cell phagocytosis and interacted with MreB3 of S. eriocheiris to prevent S. eriocheiris infection. Moreover, miR-131-3p was identified as a negative regulator of this process through its targeting of Rab7. Therefore, targeting miR-131-3p might be an effective strategy for controlling S. eriocheiris in crab aquaculture.
Collapse
Affiliation(s)
- Yubo Ma
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, 210023, China
| | - Zijie Zhou
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, 210023, China
| | - Tingyi Luo
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, 210023, China
| | - Qian Meng
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, 210023, China
| | - Hui Wang
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, 210023, China
| | - Xuguang Li
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, 210017, China
| | - Wei Gu
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Jun Zhou
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, 210017, China.
| | - Qingguo Meng
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China.
| |
Collapse
|
13
|
Wang M, Liu Z, Cheng A, Wang M, Wu Y, Yang Q, Tian B, Ou X, Sun D, Zhang S, Zhu D, Jia R, Chen S, Liu M, Zhao XX, Huang J. Host miRNA and mRNA profiles during in DEF and duck after DHAV-1 infection. Sci Rep 2024; 14:22575. [PMID: 39343789 PMCID: PMC11439951 DOI: 10.1038/s41598-024-72992-x] [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: 02/26/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
DHAV-1 is a highly infectious pathogen that can cause acute hepatitis in ducklings. MicroRNA (miRNA) plays an essential regulatory role in virus response. We characterized and compared miRNA and mRNA expression profiles in duck embryonic fibroblasts (DEF) and the liver of ducklings infected with DHAV-1. DHAV-1 infected DEF was divided into infection group (D group) and blank group (M group), and DHAV-1 infected duckling group was divided into infection group (H group) and blank group (N group). D vs. M have 130 differentially expressed (DE) miRNA (DEM) and 2204 differentially expressed (DE) mRNA (DEG), H vs. N have 72 DEM and 1976 DEG. By the intersection of D vs. M and H vs. N comparisons, 15 upregulated DEM, 5 downregulated DEM, 340 upregulated DEG and 50 downregulated DEG were found with both in vivo and in vitro DHAV-1 infection. In particular, we identified the same DE miRNA target genes and functional annotations of DE mRNA. We enriched with multiple gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, which may have important roles in viral virulence, host immunity, and metabolism. We selected miR-155, which is co-upregulated, and found that miR-155 targets SOCS1 to inhibit DHVA-1 replication.
Collapse
Affiliation(s)
- Meng Wang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Zezheng Liu
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Anchun Cheng
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China.
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China.
| | - Mingshu Wang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Ying Wu
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Qiao Yang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Bin Tian
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Xuming Ou
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Di Sun
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Shaqiu Zhang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Dekang Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Renyong Jia
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Shun Chen
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Mafeng Liu
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Xin Xin Zhao
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Juan Huang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, chengdu, China
- International Joint Research Center, Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| |
Collapse
|
14
|
Paksoy A, Meller S, Schwotzer F, Moroder P, Trampuz A, Imiolczyk JP, Perka C, Hackl M, Plachel F, Akgün D. MicroRNA expression analysis in peripheral blood and soft-tissue of patients with periprosthetic hip infection. Bone Jt Open 2024; 5:479-488. [PMID: 38839054 PMCID: PMC11152758 DOI: 10.1302/2633-1462.56.bjo-2023-0172.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024] Open
Abstract
Aims Current diagnostic tools are not always able to effectively identify periprosthetic joint infections (PJIs). Recent studies suggest that circulating microRNAs (miRNAs) undergo changes under pathological conditions such as infection. The aim of this study was to analyze miRNA expression in hip arthroplasty PJI patients. Methods This was a prospective pilot study, including 24 patients divided into three groups, with eight patients each undergoing revision of their hip arthroplasty due to aseptic reasons, and low- and high-grade PJI, respectively. The number of intraoperative samples and the incidence of positive cultures were recorded for each patient. Additionally, venous blood samples and periarticular tissue samples were collected from each patient to determine miRNA expressions between the groups. MiRNA screening was performed by small RNA-sequencing using the miRNA next generation sequencing (NGS) discovery (miND) pipeline. Results Overall, several miRNAs in plasma and tissue were identified to be progressively deregulated according to ongoing PJI. When comparing the plasma samples, patients with a high-grade infection showed significantly higher expression levels for hsa-miR-21-3p, hsa-miR-1290, and hsa-miR-4488, and lower expression levels for hsa-miR-130a-3p and hsa-miR-451a compared to the aseptic group. Furthermore, the high-grade group showed a significantly higher regulated expression level of hsa-miR-1260a and lower expression levels for hsa-miR-26a-5p, hsa-miR-26b-5p, hsa-miR-148b-5p, hsa-miR-301a-3p, hsa-miR-451a, and hsa-miR-454-3p compared to the low-grade group. No significant differences were found between the low-grade and aseptic groups. When comparing the tissue samples, the high-grade group showed significantly higher expression levels for 23 different miRNAs and lower expression levels for hsa-miR-2110 and hsa-miR-3200-3p compared to the aseptic group. No significant differences were found in miRNA expression between the high- and low-grade groups, as well as between the low-grade and aseptic groups. Conclusion With this prospective pilot study, we were able to identify a circulating miRNA signature correlating with high-grade PJI compared to aseptic patients undergoing hip arthroplasty revision. Our data contribute to establishing miRNA signatures as potential novel diagnostic and prognostic biomarkers for PJI.
Collapse
Affiliation(s)
- Alp Paksoy
- Charité University Hospital, Center for Musculoskeletal Surgery, Berlin, Germany
| | - Sebastian Meller
- Charité University Hospital, Center for Musculoskeletal Surgery, Berlin, Germany
| | - Florian Schwotzer
- Charité University Hospital, Center for Musculoskeletal Surgery, Berlin, Germany
| | | | - Andrej Trampuz
- Charité University Hospital, Center for Musculoskeletal Surgery, Berlin, Germany
| | | | - Carsten Perka
- Charité University Hospital, Center for Musculoskeletal Surgery, Berlin, Germany
| | | | | | - Doruk Akgün
- Charité University Hospital, Center for Musculoskeletal Surgery, Berlin, Germany
| |
Collapse
|
15
|
Louka E, Koumandou VL. The Emerging Role of Human Gut Bacteria Extracellular Vesicles in Mental Disorders and Developing New Pharmaceuticals. Curr Issues Mol Biol 2024; 46:4751-4767. [PMID: 38785554 PMCID: PMC11120620 DOI: 10.3390/cimb46050286] [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: 03/31/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
In recent years, further evidence has emerged regarding the involvement of extracellular vesicles in various human physiopathological conditions such as Alzheimer's disease, Parkinson's disease, irritable bowel syndrome, and mental disorders. The biogenesis and cargo of such vesicles may reveal their impact on human health nd disease and set the underpinnings for the development of novel chemical compounds and pharmaceuticals. In this review, we examine the link between bacteria-derived exosomes in the gastrointestinal tract and mental disorders, such as depression and anxiety disorders. Crucially, we focus on whether changes in the gut environment affect the human mental state or the other way around. Furthermore, the possibility of handling bacteria-derived exosomes as vectors of chemicals to treat such conditions is examined.
Collapse
Affiliation(s)
- Effrosyni Louka
- Genetics Laboratory, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - Vassiliki Lila Koumandou
- Genetics Laboratory, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| |
Collapse
|
16
|
Huang S, Xiao X, Wu H, Zhou F, Fu C. MicroRNA-582-3p knockdown alleviates non-alcoholic steatohepatitis by altering the gut microbiota composition and moderating TMBIM1. Ir J Med Sci 2024; 193:909-916. [PMID: 37823951 DOI: 10.1007/s11845-023-03529-w] [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: 05/10/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND The gut dysbiosis correlates with non-alcoholic steatohepatitis (NASH), involving the moderation of miRNAs. AIMS This study was aimed to investigate the correlation between gut microbiota and miR-582-3p in patients with non-alcoholic steatohepatitis (NASH) and to explore the possible regulation of miR-582-3p in the function of the activated hepatic stellate cells (HSCs). METHODS GSE69670 and GSE14435 datasets were analyzed by GEO2R. Plasma and fecal samples were obtained from the subjects, non-steatosis (n = 35), simple steatosis (n = 35), and NASH (n = 35). The variations in intestinal microbiota in the non-steatosis and NASH groups were analyzed using 16S rRNA sequencing. The expression of miR-582-3p among the groups was detected using RT-qPCR. Correlations between top-changed intestinal microbiota and miR-582-3p expression were analyzed using the Pearson correlation coefficient. Target gene identification was performed by prediction and dual-luciferase reporter assay. The effect of miR-582-3p on the cell function of TGF-β1-induced HSCs was assessed in vitro. RESULTS miR-582-3p was the common differentially expressed miRNA between GSE69670 and GSE14435. miR-582-3p was upregulated in NASH patients' plasma, as well as in TGF-β1-induced LX-2 cells. The non-steatosis and NASH groups showed significantly different intestinal microbiota distribution. miR-582-3p was positively correlated with specific microbiota populations. TMBIM1 was a target gene for miR-582-3p. Knockdown of miR-582-3p suppressed HSC proliferation and myofibroblast markers' expression but induced cell apoptosis, via TMBIM1. CONCLUSIONS This present study suggests that miR-582-3p promotes the progression of NASH. Knockdown of miR-582-3p may alleviate NASH by altering the gut microbiota composition and moderating TMBIM1.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Internal Medicine, Hunan Maternal and Child Health Hospital, Changsha, Hunan, 410013, China
| | - Xia Xiao
- Department of Internal Medicine, Hunan Maternal and Child Health Hospital, Changsha, Hunan, 410013, China
| | - Hongman Wu
- Department of Infection Control Center, Xiangya Hospital of Central South University, NO.87, Xiangya Road, Changsha, Hunan, 410008, China
| | - Feng Zhou
- Department of Infection Control Center, Xiangya Hospital of Central South University, NO.87, Xiangya Road, Changsha, Hunan, 410008, China
| | - Chenchao Fu
- Department of Infection Control Center, Xiangya Hospital of Central South University, NO.87, Xiangya Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
17
|
Cao Y, Xia H, Tan X, Shi C, Ma Y, Meng D, Zhou M, Lv Z, Wang S, Jin Y. Intratumoural microbiota: a new frontier in cancer development and therapy. Signal Transduct Target Ther 2024; 9:15. [PMID: 38195689 PMCID: PMC10776793 DOI: 10.1038/s41392-023-01693-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/20/2023] [Accepted: 10/24/2023] [Indexed: 01/11/2024] Open
Abstract
Human microorganisms, including bacteria, fungi, and viruses, play key roles in several physiological and pathological processes. Some studies discovered that tumour tissues once considered sterile actually host a variety of microorganisms, which have been confirmed to be closely related to oncogenesis. The concept of intratumoural microbiota was subsequently proposed. Microbiota could colonise tumour tissues through mucosal destruction, adjacent tissue migration, and hematogenic invasion and affect the biological behaviour of tumours as an important part of the tumour microenvironment. Mechanistic studies have demonstrated that intratumoural microbiota potentially promote the initiation and progression of tumours by inducing genomic instability and mutations, affecting epigenetic modifications, promoting inflammation response, avoiding immune destruction, regulating metabolism, and activating invasion and metastasis. Since more comprehensive and profound insights about intratumoral microbiota are continuously emerging, new methods for the early diagnosis and prognostic assessment of cancer patients have been under examination. In addition, interventions based on intratumoural microbiota show great potential to open a new chapter in antitumour therapy, especially immunotherapy, although there are some inevitable challenges. Here, we aim to provide an extensive review of the concept, development history, potential sources, heterogeneity, and carcinogenic mechanisms of intratumoural microorganisms, explore the potential role of microorganisms in tumour prognosis, and discuss current antitumour treatment regimens that target intratumoural microorganisms and the research prospects and limitations in this field.
Collapse
Affiliation(s)
- Yaqi Cao
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Xueyun Tan
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Chunwei Shi
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yanling Ma
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Daquan Meng
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Mengmeng Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Zhilei Lv
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Province Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Respiratory Diseases of National Health Commission, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Province Engineering Research Center for Tumour-Targeted Biochemotherapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Province Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|
18
|
Lian S, Liu P, Li X, Lv G, Song J, Zhang H, Wu R, Wang D, Wang J. BLV-miR-B1-5p Promotes Staphylococcus aureus Adhesion to Mammary Epithelial Cells by Targeting MUC1. Animals (Basel) 2023; 13:3811. [PMID: 38136848 PMCID: PMC10741194 DOI: 10.3390/ani13243811] [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: 10/12/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Bovine leukemia virus (BLV) is widely prevalent worldwide and can persistently infect mammary epithelial cells in dairy cows, leading to reduced cellular antimicrobial capacity. BLV-encoded microRNAs (BLV-miRNAs) can modify host genes and promote BLV replication. We previously showed that BLV-miR-B1-5p significantly promoted Staphylococcus aureus (S. aureus) adhesion to bovine mammary epithelial (MAC-T) cells; however, the pathway responsible for this effect remained unclear. This study aims to examine how BLV-miR-B1-5p promotes S. aureus adhesion to MAC-T cells via miRNA target gene prediction and validation. Target site prediction showed that BLV-miR-B1-5p could target the mucin family gene mucin 1 (MUC1). Real-time polymerase chain reaction, immunofluorescence, and dual luciferase reporter assay further confirmed that BLV-miR-B1-5p could target and inhibit the expression of MUC1 in bovine MAC-T cells while interfering with the expression of MUC1 promoted S. aureus adhesion to MAC-T cells. These results indicate that BLV-miR-B1-5p promotes S. aureus adhesion to mammary epithelial cells by targeting MUC1.
Collapse
Affiliation(s)
- Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Pengfei Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Xiao Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Guanxin Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Jiahe Song
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Han Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Rui Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
- College of Biology and Agriculture, Jiamusi University, Jiamusi 154007, China
| | - Di Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (S.L.); (P.L.); (X.L.); (G.L.); (J.S.); (H.Z.); (R.W.)
- China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| |
Collapse
|
19
|
Schorr L, Mathies M, Elinav E, Puschhof J. Intracellular bacteria in cancer-prospects and debates. NPJ Biofilms Microbiomes 2023; 9:76. [PMID: 37813921 PMCID: PMC10562400 DOI: 10.1038/s41522-023-00446-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
Recent evidence suggests that some human cancers may harbor low-biomass microbial ecosystems, spanning bacteria, viruses, and fungi. Bacteria, the most-studied kingdom in this context, are suggested by these studies to localize within cancer cells, immune cells and other tumor microenvironment cell types, where they are postulated to impact multiple cancer-related functions. Herein, we provide an overview of intratumoral bacteria, while focusing on intracellular bacteria, their suggested molecular activities, communication networks, host invasion and evasion strategies, and long-term colonization capacity. We highlight how the integration of sequencing-based and spatial techniques may enable the recognition of bacterial tumor niches. We discuss pitfalls, debates and challenges in decisively proving the existence and function of intratumoral microbes, while reaching a mechanistic elucidation of their impacts on tumor behavior and treatment responses. Together, a causative understanding of possible roles played by intracellular bacteria in cancer may enable their future utilization in diagnosis, patient stratification, and treatment.
Collapse
Affiliation(s)
- Lena Schorr
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Marius Mathies
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany.
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| | - Jens Puschhof
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
20
|
Zogg H, Singh R, Ha SE, Wang Z, Jin B, Ha M, Dafinone M, Batalon T, Hoberg N, Poudrier S, Nguyen L, Yan W, Layden BT, Dugas LR, Sanders KM, Ro S. miR-10b-5p rescues leaky gut linked with gastrointestinal dysmotility and diabetes. United European Gastroenterol J 2023; 11:750-766. [PMID: 37723933 PMCID: PMC10576606 DOI: 10.1002/ueg2.12463] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/31/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND/AIM Diabetes has substantive co-occurrence with disorders of gut-brain interactions (DGBIs). The pathophysiological and molecular mechanisms linking diabetes and DGBIs are unclear. MicroRNAs (miRNAs) are key regulators of diabetes and gut dysmotility. We investigated whether impaired gut barrier function is regulated by a key miRNA, miR-10b-5p, linking diabetes and gut dysmotility. METHODS We created a new mouse line using the Mb3Cas12a/Mb3Cpf1 endonuclease to delete mir-10b globally. Loss of function studies in the mir-10b knockout (KO) mice were conducted to characterize diabetes, gut dysmotility, and gut barrier dysfunction phenotypes in these mice. Gain of function studies were conducted by injecting these mir-10b KO mice with a miR-10b-5p mimic. Further, we performed miRNA-sequencing analysis from colonic mucosa from mir-10b KO, wild type, and miR-10b-5p mimic injected mice to confirm (1) deficiency of miR-10b-5p in KO mice, and (2) restoration of miR-10b-5p after the mimic injection. RESULTS Congenital loss of mir-10b in mice led to the development of hyperglycemia, gut dysmotility, and gut barrier dysfunction. Gut permeability was increased, but expression of the tight junction protein Zonula occludens-1 was reduced in the colon of mir-10b KO mice. Patients with diabetes or constipation- predominant irritable bowel syndrome, a known DGBI that is linked to leaky gut, had significantly reduced miR-10b-5p expression. Injection of a miR-10b-5p mimic in mir-10b KO mice rescued these molecular alterations and phenotypes. CONCLUSIONS Our study uncovered a potential pathophysiologic mechanism of gut barrier dysfunction that links both the diabetes and gut dysmotility phenotypes in mice lacking miR-10b-5p. Treatment with a miR-10b-5p mimic reversed the leaky gut, diabetic, and gut dysmotility phenotypes, highlighting the translational potential of the miR-10b-5p mimic.
Collapse
Affiliation(s)
- Hannah Zogg
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Rajan Singh
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Se Eun Ha
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Zhuqing Wang
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Byungchang Jin
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Mariah Ha
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Mirabel Dafinone
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Tylar Batalon
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Nicholas Hoberg
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Sandra Poudrier
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Linda Nguyen
- Division of Gastroenterology & HepatologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Wei Yan
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and MetabolismDepartment of MedicineThe University of Illinois at ChicagoChicagoIllinoisUSA
- Jesse Brown Veterans Affairs Medical CenterChicagoIllinoisUSA
| | - Lara R. Dugas
- Loyola University ChicagoPublic Health SciencesMaywoodIllinoisUSA
- Division of Epidemiology & BiostatisticsSchool of Public HealthFaculty of Health SciencesUniversity of Cape TownCape TownSouth Africa
| | - Kenton M. Sanders
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
| | - Seungil Ro
- Department of Physiology and Cell BiologySchool of MedicineUniversity of NevadaRenoNevadaUSA
- RosVivo TherapeuticsApplied Research FacilityRenoNevadaUSA
| |
Collapse
|
21
|
Bhati T, Ray A, Arora R, Siraj F, Parvez S, Rastogi S. Immunomodulation of cytokine signalling at feto-maternal interface by microRNA-223 and -150-5p in infection-associated spontaneous preterm birth. Mol Immunol 2023; 160:1-11. [PMID: 37285685 DOI: 10.1016/j.molimm.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/24/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Spontaneous preterm birth (sPTB) is a global health concern and it is the most prevalent cause of infant mortality and morbidity with occurrence rate of 5 - 18% worldwide. Studies suggest infection and infection-driven activation of inflammatory responses are the potential risk factors for sPTB. MicroRNAs (miRNAs) are thought to control the expression of several immune genes, making them crucial components of the intricate immune regulatory network and the dysregulation of miRNAs in placenta has been associated to several pregnancy-related complications. However, studies on possible role of miRNAs in immunomodulation of cytokine signalling in infection-associated sPTB are scarce. Present study aimed to investigate expression/ correlation of a few circulating miRNAs (miR-223, -150-5p, -185-5p, -191-5p), miRNA target genes and associated cytokines in sPTB women found infected with Chlamydia trachomatis/ Mycoplasma hominis/ Ureaplasma urealyticum. Non-heparinized blood and placental sample were collected from 140 sPTB and 140 term women visiting Safdarjung hospital, New Delhi (India) for conducting PCR and RT-PCR for pathogen detection and miRNA/ target gene/ cytokine expression, respectively. Common target genes of differentially expressed miRNAs were obtained from databases. The correlation between select target genes/ cytokines and serum miRNAs was determined by Spearman's rank correlation. 43 sPTB were infected with either pathogen and a significant upregulation of serum miRNAs was observed. However, miR-223 and 150-5p showed maximum fold-change (4.78 and 5.58, respectively) in PTB versus control group. IL-6ST, TGF-β R3 and MMP-14 were important target genes among 454 common targets, whereas, IL-6 and TGF-β were associated cytokines. miR-223 and 150-5p showed significant negative correlation with IL-6ST/ IL-6/ MMP-14 and positive correlation with TGF-β R3/ TGF-β. A significant positive correlation was found between IL-6ST and IL-6, TGF-β R3 and TGF-β. However, miR-185-5p and 191-5p were not significantly correlated. Although post-transcriptional validation is required, yet on the basis of mRNA findings, the study concludes that miR-223 and 150-5p are apparently of clinical importance in regulation of inflammatory processes during infection-associated sPTB.
Collapse
Affiliation(s)
- Tanu Bhati
- Molecular Microbiology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India; Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India
| | - Ankita Ray
- Molecular Microbiology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India; Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India
| | - Renu Arora
- Department of Obstetrics and Gynecology, Vardhman Mahavir Medical College (VMMC) and Safdarjung Hospital, New Delhi 110029, India
| | - Fouzia Siraj
- Pathology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India
| | - Sangita Rastogi
- Molecular Microbiology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India.
| |
Collapse
|
22
|
Tang B, Lu X, Tong Y, Feng Y, Mao Y, Dun G, Li J, Xu Q, Tang J, Zhang T, Deng L, He X, Lan Y, Luo H, Zeng L, Xiang Y, Li Q, Zeng D, Mao X. MicroRNA-31 induced by Fusobacterium nucleatum infection promotes colorectal cancer tumorigenesis. iScience 2023; 26:106770. [PMID: 37216106 PMCID: PMC10196571 DOI: 10.1016/j.isci.2023.106770] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/27/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Persistent Fusobacterium nucleatum infection is associated with the development of human colorectal cancer (CRC) and promotes tumorigenicity, but the underlying mechanisms remain unclear. Here, we reported that F. nucleatum promoted the tumorigenicity of CRC, which was associated with F. nucleatum-induced microRNA-31 (miR-31) expression in CRC tissues and cells. F. nucleatum infection inhibited autophagic flux by miR-31 through inhibiting syntaxin-12 (STX12) and was associated with the increased intracellular survival of F. nucleatum. Overexpression of miR-31 in CRC cells promoted their tumorigenicity by targeting eukaryotic initiation factor 4F-binding protein 1/2 (eIF4EBP1/2), whereas miR-31 knockout mice were resistant to the formation of colorectal tumors. In conclusion, F. nucleatum, miR-31, and STX12 form a closed loop in the autophagy pathway, and continuous F. nucleatum-induced miR-31 expression promotes the tumorigenicity of CRC cells by targeting eIF4EBP1/2. These findings reveal miR-31 as a potential diagnostic biomarker and therapeutic target in CRC patients with F. nucleatum infection.
Collapse
Affiliation(s)
- Bin Tang
- Department of Clinical Laboratory, Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin, Chongqing 402260, China
| | - Xiaoxue Lu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yanan Tong
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuyang Feng
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yilan Mao
- Class of 2021 undergraduate, Nursing College of Chongqing Medical University, Chongqing 400016, China
| | - Guodong Dun
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jing Li
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Qiaolin Xu
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Jie Tang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Tao Zhang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ling Deng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xiaoyi He
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuanzhi Lan
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Huaxing Luo
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Linghai Zeng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuanyuan Xiang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Dongzhu Zeng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xuhu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
23
|
Zhang Z, Guan X. Japanese Flounder pol-miR-155 Is Involved in Edwardsiella tarda Infection via ATG3. Genes (Basel) 2023; 14:genes14050958. [PMID: 37239318 DOI: 10.3390/genes14050958] [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: 03/10/2023] [Revised: 04/08/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that function in the post-transcriptionally regulation of the expression of diverse genes, including those involved in immune defense. Edwardsiella tarda can infect a broad range of hosts and cause severe disease in aquatic species, including Japanese flounder (Paralichthys olivaceus). In this study, we examined the regulation mechanism of a flounder miRNA, pol-miR-155, during the infection of E. tarda. Pol-miR-155 was identified to target flounder ATG3. Overexpression of pol-miR-155 or knockdown of ATG3 expression suppressed autophagy and promoted the intracellular replication of E. tarda in flounder cells. Overexpression of pol-miR-155 activated the NF-κB signaling pathway and further promoted the expression of downstream immune related genes of interleukin (IL)-6 and IL-8. These results unraveled the regulatory effect of pol-miR-155 in autophagy and in E. tarda infection.
Collapse
Affiliation(s)
- Zhanwei Zhang
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiaolu Guan
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
24
|
Global Molecular Response of Paracoccidioides brasiliensis to Zinc Deprivation: Analyses at Transcript, Protein and MicroRNA Levels. J Fungi (Basel) 2023; 9:jof9030281. [PMID: 36983449 PMCID: PMC10056003 DOI: 10.3390/jof9030281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Zinc is one of the main micronutrients for all organisms. One of the defense mechanisms used by the host includes the sequestration of metals used in fungal metabolism, such as iron and zinc. There are several mechanisms that maintain the balance in the intracellular zinc supply. MicroRNAs are effector molecules of responses between the pathogen and host, favoring or preventing infection in many microorganisms. Fungi of the Paracoccidioides genus are thermodimorphic and the etiological agents of paracoccidioidomycosis (PCM). In the current pandemic scenario world mycosis studies continue to be highly important since a significant number of patients with COVID-19 developed systemic mycoses, co-infections that complicated their clinical condition. The objective was to identify transcriptomic and proteomic adaptations in Paracoccidioides brasiliensis during zinc deprivation. Nineteen microRNAs were identified, three of which were differentially regulated. Target genes regulated by those microRNAs are elements of zinc homeostasis such as ZRT1, ZRT3 and COT1 transporters. Transcription factors that have zinc in their structure are also targets of those miRNAs. Transcriptional and proteomic data suggest that P. brasiliensis undergoes metabolic remodeling to survive zinc deprivation and that miRNAs may be part of the regulatory process.
Collapse
|
25
|
Lepcha TT, Kumar M, Sharma AK, Mal S, Majumder D, Jana K, Basu J, Kundu M. Uncovering the role of microRNA671-5p/CDCA7L/monoamine oxidase-A signaling in Helicobacter pylori mediated apoptosis in gastric epithelial cells. Pathog Dis 2023; 81:7143101. [PMID: 37140023 DOI: 10.1093/femspd/ftad006] [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: 02/09/2023] [Revised: 03/27/2023] [Accepted: 04/24/2023] [Indexed: 05/05/2023] Open
Abstract
Helicobacter pylori is a gram-negative microaerophilic bacterium and is associated with gastrointestinal diseases ranging from peptic ulcer and gastritis to gastric cancer and mucosa-associated lymphoid tissue lymphoma. In our laboratory, the transcriptomes and miRnomes of AGS cells infected with H. pylori have been profiled, and an miRNA-mRNA network has been constructed. MicroRNA 671-5p is upregulated during H. pylori infection of AGS cells or of mice. In this study, the role of miR-671-5p during infection has been investigated. It has been validated that miR-671-5p targets the transcriptional repressor CDCA7L, which is downregulated during infection (in vitro and in vivo) concomitant with miR-671-5p upregulation. Further, it has been established that the expression of monoamine oxidase A (MAO-A) is repressed by CDCA7L, and that MAO-A triggers the generation of reactive oxygen species (ROS). Consequently, miR-671-5p/CDCA7L signaling is linked to the generation of ROS during H. pylori infection. Finally, it has been demonstrated that ROS-mediated caspase 3 activation and apoptosis that occurs during H. pylori infection, is dependent on the miR-671-5p/CDCA7L/MAO-A axis. Based on the above reports, it is suggested that targeting miR-671-5p could offer a means of regulating the course and consequences of H. pylori infection.
Collapse
Affiliation(s)
- Thurbu Tshering Lepcha
- Department of Chemistry, Bose Institute, 93/1 Acharya Prafulla Chandra Road Kolkata 700009, India
| | - Manish Kumar
- Department of Chemistry, Bose Institute, 93/1 Acharya Prafulla Chandra Road Kolkata 700009, India
| | - Arun Kumar Sharma
- Department of Chemistry, Bose Institute, 93/1 Acharya Prafulla Chandra Road Kolkata 700009, India
| | - Soumya Mal
- Department of Chemistry, Bose Institute, 93/1 Acharya Prafulla Chandra Road Kolkata 700009, India
| | - Debayan Majumder
- Department of Chemistry, Bose Institute, 93/1 Acharya Prafulla Chandra Road Kolkata 700009, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, EN80 Sector V, Salt Lake City, Kolkata 700091, India
| | - Joyoti Basu
- Department of Chemistry, Bose Institute, 93/1 Acharya Prafulla Chandra Road Kolkata 700009, India
| | - Manikuntala Kundu
- Department of Chemistry, Bose Institute, 93/1 Acharya Prafulla Chandra Road Kolkata 700009, India
| |
Collapse
|
26
|
Nikolaieva N, Sevcikova A, Omelka R, Martiniakova M, Mego M, Ciernikova S. Gut Microbiota-MicroRNA Interactions in Intestinal Homeostasis and Cancer Development. Microorganisms 2022; 11:microorganisms11010107. [PMID: 36677399 PMCID: PMC9867529 DOI: 10.3390/microorganisms11010107] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Pre-clinical models and clinical studies highlight the significant impact of the host-microbiota relationship on cancer development and treatment, supporting the emerging trend for a microbiota-based approach in clinical oncology. Importantly, the presence of polymorphic microbes is considered one of the hallmarks of cancer. The epigenetic regulation of gene expression by microRNAs affects crucial biological processes, including proliferation, differentiation, metabolism, and cell death. Recent evidence has documented the existence of bidirectional gut microbiota-microRNA interactions that play a critical role in intestinal homeostasis. Importantly, alterations in microRNA-modulated gene expression are known to be associated with inflammatory responses and dysbiosis in gastrointestinal disorders. In this review, we summarize the current findings about miRNA expression in the intestine and focus on specific gut microbiota-miRNA interactions linked to intestinal homeostasis, the immune system, and cancer development. We discuss the potential clinical utility of fecal miRNA profiling as a diagnostic and prognostic tool in colorectal cancer, and demonstrate how the emerging trend of gut microbiota modulation, together with the use of personalized microRNA therapeutics, might bring improvements in outcomes for patients with gastrointestinal cancer in the era of precision medicine.
Collapse
Affiliation(s)
- Nataliia Nikolaieva
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Michal Mego
- National Cancer Institute and Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
- Correspondence: ; Tel.: +421-02-3229519
| |
Collapse
|
27
|
NUP160 knockdown inhibits the progression of diabetic nephropathy in vitro and in vivo. Regen Ther 2022; 21:87-95. [PMID: 35785044 PMCID: PMC9234011 DOI: 10.1016/j.reth.2022.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/02/2022] Open
Abstract
Diabetic nephropathy (DN) is a severe diabetic complication and podocyte damage is a hallmark of DN. The Nucleoporin 160 (NUP160) gene was demonstrated to regulate cell proliferation and apoptosis in mouse podocytes. This study explored the possible role and mechanisms of NUP160 in high glucose-triggered podocyte injury. A rat model of DN was established by intraperitoneal injection of 60 mg/kg streptozotocin (STZ). Podocytes were treated with 33 mM high glucose. The effects of the Nup160 on DN and its mechanisms were assessed using MTT, flow cytometry, Western blot, ELISA, RT-qPCR, and luciferase reporter assays. The in vivo effects of NUP160 were analyzed by HE, PAS, and MASSON staining assays. The NUP160 level was significantly upregulated in podocytes treated with 33 mM high glucose. Functionally, NUP160 knockdown alleviated high glucose-induced apoptosis and inflammation in podocytes. Mechanistically, miR-495-3p directly targeted NUP160, and lncRNA HCG18 upregulated NUP160 by sponging miR-495-3p by acting as a ceRNA. Additionally, NUP160 overexpression reversed the effects of HCG18 knockdown in high glucose treated-podocytes. The in vivo assays indicated that NUP160 knockdown alleviated the symptoms of DN rats. NUP160 knockdown plays a key role in preventing the progression of DN, suggesting that targeting NUP160 may be a potential therapeutic strategy for DN treatment.
Collapse
|
28
|
Ou J, Liu Q, Bian Y, Luan X, Meng Y, Dong H, Cao M, Zhang B, Wang Z, Zhao W. Integrated analysis of mRNA and microRNA transcriptome related to immunity and autophagy in shrimp hemocytes infected with Spiroplasma eriocheiris. FISH & SHELLFISH IMMUNOLOGY 2022; 130:436-452. [PMID: 36184970 DOI: 10.1016/j.fsi.2022.09.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
In recent years, the industry in charge of the cultivation of Macrobrachium nipponense (M.nipponense) has suffered significant economic losses due to an infectious pathogen called Spiroplasma eriocheiris (S.eriocheiris). There has therefore been a need to identify the key immune and autophagy genes that respond to M.nipponense's infection with S. eriocheiris to analyze its immune response mechanism and the regulation of related microRNAs (miRNAs). In this study, the mRNA and miRNA transcriptome of M.nipponense's hemocytes were analyzed at different stages of infection. This analysis employed the second and third-generation sequencing technologies. In the mRNA transcriptome, 1656 genes were expressed in healthy and susceptible M.nipponense. 892 of these were significantly up-regulated, while 764 were down-regulated. 118 genes with significant differences in autophagy, endocytosis, lysosome, Toll, IMD, and VEGF pathways were obtained from the transcriptome. In the miRNA transcriptome, 312 miRNAs (Conserved: 112, PN-type: 18, PC-type: 182) were sequenced. 74 were significantly up-regulated, and 57 were down-regulated. There were 25 miRNAs involved in regulating the Toll and IMD pathways, 41 in endocytosis, 30 in lysosome, and 12 in the VEGF pathway. An integrated analysis of immune-related miRNAs and mRNAs showed that miRNAs with significant differences (P < 0.05) such as ame-miR-29b-3p, dpu-miR-1and PC-3p-945_4074, had corresponding regulatory relationships with 118 important immune genes such as Relish, Dorsal, Caspase-3, and NF-κB. This study obtained the key immune and autophagy-related genes and corresponding regulatory miRNAs in M. nipponense's hemocytes in response to an infection by S.eriocheiris. The results can provide vital data that further reveals the defense mechanism of M.nipponense's immune system against S.eriocheiris. It can also help further comprehension and interpretation of M.nipponense's resistance mechanism to the invading S.eriocheiris, and provide molecular research information for the realization of host-directed therapies (HDT) for M.nipponense.
Collapse
Affiliation(s)
- Jiangtao Ou
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China.
| | - Qiao Liu
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China; The Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, Province Jiangsu, China
| | - Yunxia Bian
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Xiaoqi Luan
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China; Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, China
| | - Yusuo Meng
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Huizi Dong
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Miao Cao
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Benhou Zhang
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Zisheng Wang
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Weihong Zhao
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| |
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
|
Aminian-Dehkordi J, Valiei A, Mofrad MRK. Emerging computational paradigms to address the complex role of gut microbial metabolism in cardiovascular diseases. Front Cardiovasc Med 2022; 9:987104. [PMID: 36299869 PMCID: PMC9589059 DOI: 10.3389/fcvm.2022.987104] [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: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
The human gut microbiota and its associated perturbations are implicated in a variety of cardiovascular diseases (CVDs). There is evidence that the structure and metabolic composition of the gut microbiome and some of its metabolites have mechanistic associations with several CVDs. Nevertheless, there is a need to unravel metabolic behavior and underlying mechanisms of microbiome-host interactions. This need is even more highlighted when considering that microbiome-secreted metabolites contributing to CVDs are the subject of intensive research to develop new prevention and therapeutic techniques. In addition to the application of high-throughput data used in microbiome-related studies, advanced computational tools enable us to integrate omics into different mathematical models, including constraint-based models, dynamic models, agent-based models, and machine learning tools, to build a holistic picture of metabolic pathological mechanisms. In this article, we aim to review and introduce state-of-the-art mathematical models and computational approaches addressing the link between the microbiome and CVDs.
Collapse
Affiliation(s)
| | | | - Mohammad R. K. Mofrad
- Department of Bioengineering and Mechanical Engineering, University of California, Berkeley, Berkeley, CA, United States
| |
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: 7] [Impact Index Per Article: 2.3] [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
|
Zhang F, Zhou Y, Ding J. The current landscape of microRNAs (miRNAs) in bacterial pneumonia: opportunities and challenges. Cell Mol Biol Lett 2022; 27:70. [PMID: 35986232 PMCID: PMC9392286 DOI: 10.1186/s11658-022-00368-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/01/2022] [Indexed: 11/12/2022] Open
Abstract
MicroRNAs (miRNAs), which were initially discovered in Caenorhabditis elegans, can regulate gene expression by recognizing cognate sequences and interfering with the transcriptional or translational machinery. The application of bioinformatics tools for structural analysis and target prediction has largely driven the investigation of certain miRNAs. Notably, it has been found that certain miRNAs which are widely involved in the inflammatory response and immune regulation are closely associated with the occurrence, development, and outcome of bacterial pneumonia. It has been shown that certain miRNA techniques can be used to identify related targets and explore associated signal transduction pathways. This enhances the understanding of bacterial pneumonia, notably for "refractory" or drug-resistant bacterial pneumonia. Although these miRNA-based methods may provide a basis for the clinical diagnosis and treatment of this disease, they still face various challenges, such as low sensitivity, poor specificity, low silencing efficiency, off-target effects, and toxic reactions. The opportunities and challenges of these methods have been completely reviewed, notably in bacterial pneumonia. With the continuous improvement of the current technology, the miRNA-based methods may surmount the aforementioned limitations, providing promising support for the clinical diagnosis and treatment of "refractory" or drug-resistant bacterial pneumonia.
Collapse
Affiliation(s)
- Fan Zhang
- Beijing Key Laboratory of Basic Research With Traditional Chinese Medicine On Infectious Diseases, Beijing Institute of Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Yunxin Zhou
- Beijing Key Laboratory of Basic Research With Traditional Chinese Medicine On Infectious Diseases, Beijing Institute of Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Junying Ding
- Beijing Key Laboratory of Basic Research With Traditional Chinese Medicine On Infectious Diseases, Beijing Institute of Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| |
Collapse
|
33
|
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
|
34
|
Gòdia M, Brogaard L, Mármol-Sánchez E, Langhorn R, Nordang Kieler I, Jan Reezigt B, Nikolic Nielsen L, Rem Jessen L, Cirera S. Urinary microRNAome in healthy cats and cats with pyelonephritis or other urological conditions. PLoS One 2022; 17:e0270067. [PMID: 35857780 PMCID: PMC9299306 DOI: 10.1371/journal.pone.0270067] [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: 01/21/2022] [Accepted: 06/03/2022] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression at the post-transcriptional level. miRNAs have been found in urine and have shown diagnostic potential in human nephropathies. Here, we aimed to characterize, for the first time, the feline urinary miRNAome and explore the use of urinary miRNA profiles as non-invasive biomarkers for feline pyelonephritis (PN). Thirty-eight cats were included in a prospective case-control study and classified in five groups: healthy Control cats (n = 11), cats with PN (n = 10), cats with subclinical bacteriuria or cystitis (SB/C, n = 5), cats with ureteral obstruction (n = 7) and cats with chronic kidney disease (n = 5). By small RNA sequencing we identified 212 miRNAs in cat urine, including annotated (n = 137) and putative novel (n = 75) miRNAs. The 15 most highly abundant urinary miRNAs accounted for nearly 71% of all detected miRNAs, most of which were previously identified in feline kidney. Ninety-nine differentially abundant (DA) miRNAs were identified when comparing Control cats to cats with urological conditions and 102 DA miRNAs when comparing PN to other urological conditions. Tissue clustering analysis revealed that the majority of urine samples clustered close to kidney, which confirm the likely cellular origin of the secreted urinary miRNAs. Relevant DA miRNAs were verified by quantitative real-time PCR (qPCR). Eighteen miRNAs discriminated Control cats from cats with a urological condition. Of those, seven miRNAs were DA by both RNAseq and qPCR methods between Control and PN cats (miR-125b-5p, miR-27a-3p, miR-21-5p, miR-27b-3p, miR-125a-5p, miR-17-5p and miR-23a-3p) or DA between Control and SB/C cats (miR-125b-5p). Six additional miRNAs (miR-30b-5p, miR-30c, miR-30e-5p, miR-27a-3p, miR-27b-39 and miR-222) relevant for discriminating PN from other urological conditions were identified by qPCR alone (n = 4) or by both methods (n = 2) (P<0.05). This panel of 13 miRNAs has potential as non-invasive urinary biomarkers for diagnostic of PN and other urological conditions in cats.
Collapse
Affiliation(s)
- Marta Gòdia
- Department of Animal Medicine and Surgery, School of Veterinary Sciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Catalonia, Spain
- Centre for Research in Agricultural Genomics (CRAG) CSIC-IRTA-UAB-UB, Campus UAB, Cerdanyola del Vallès, Catalonia, Spain
| | - Louise Brogaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Emilio Mármol-Sánchez
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Centre for Paleogenetics, Stockholm University, Stockholm, Sweden
| | - Rebecca Langhorn
- Faculty of Health and Medical Sciences, Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ida Nordang Kieler
- Faculty of Health and Medical Sciences, Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Lise Nikolic Nielsen
- Faculty of Health and Medical Sciences, Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lisbeth Rem Jessen
- Faculty of Health and Medical Sciences, Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
- * E-mail: (LRJ); (SC)
| | - Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- * E-mail: (LRJ); (SC)
| |
Collapse
|
35
|
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
|
36
|
Li G, Zong X, Cheng Y, Xu J, Deng J, Huang Y, Ma C, Fu Q. miR-223-3p contributes to suppressing NLRP3 inflammasome activation in Streptococcus equi ssp. zooepidemicus infection. Vet Microbiol 2022; 269:109430. [PMID: 35427992 DOI: 10.1016/j.vetmic.2022.109430] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/21/2022] [Accepted: 04/03/2022] [Indexed: 11/20/2022]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an essential pathogen in a range of species, causing a worldwide variety of diseases, such as meningitis, endocarditis, and septicaemia. Studies have shown that microRNAs (miRNAs), which regulate target genes at the post-transcriptional level, play an important regulatory role in the organism. In this study, the infection of J774A.1 murine macrophages with SEZ up-regulated NLRP3 inflammasome and downstream pathways accompanied by miR-223-3p down-regulation. Through computational prediction and experimental confirmation, we have shown that miR-223-3p directly targets the NLRP3 mRNA. Consequently, overexpression of miR-223-3p suppressed NLRP3 inflammasome activation and downstream pathways in response to SEZ infection. The miR-223-3p inhibitor exhibited the opposite effect, causing hyperactivation of NLRP3 inflammation activation and downstream pathways. Additionally, we further demonstrated that miRNA-223-3p inhibited the secretion of IL-1β and IL-18 by regulating the NLRP3/caspase-1 pathway. Furthermore, intravenous administration of miR-223-3p significantly decreased inflammation in mice in response to SEZ. In conclusion, our results demonstrated that miR-223-3p contributes to suppressing the NLRP3 inflammasome activation in SEZ infection, contributing novel evidence to identify a therapeutic target for treating SEZ.
Collapse
Affiliation(s)
- Guochao Li
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Xueqing Zong
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Yun Cheng
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Jianqi Xu
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Jingfei Deng
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China
| | - Chunquan Ma
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Guangdong 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong 528225, China.
| |
Collapse
|
37
|
Herrera-Uribe J, Zaldívar-López S, Aguilar C, Entrenas-García C, Bautista R, Claros MG, Garrido JJ. Study of microRNA expression in Salmonella Typhimurium-infected porcine ileum reveals miR-194a-5p as an important regulator of the TLR4-mediated inflammatory response. Vet Res 2022; 53:35. [PMID: 35598011 PMCID: PMC9123658 DOI: 10.1186/s13567-022-01056-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Infection with Salmonella Typhimurium (S. Typhimurium) is a common cause of food-borne zoonosis leading to acute gastroenteritis in humans and pigs, causing economic losses to producers and farmers, and generating a food security risk. In a previous study, we demonstrated that S. Typhimurium infection produces a severe transcriptional activation of inflammatory processes in ileum. However, little is known regarding how microRNAs regulate this response during infection. Here, small RNA sequencing was used to identify 28 miRNAs differentially expressed (DE) in ileum of S. Typhimurium-infected pigs, which potentially regulate 14 target genes involved in immune system processes such as regulation of cytokine production, monocyte chemotaxis, or cellular response to interferon gamma. Using in vitro functional and gain/loss of function (mimics/CRISPR-Cas system) approaches, we show that porcine miR-194a-5p (homologous to human miR-194-5p) regulates TLR4 gene expression, an important molecule involved in pathogen virulence, recognition and activation of innate immunity in Salmonella infection.
Collapse
Affiliation(s)
- Juber Herrera-Uribe
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Viral Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Sara Zaldívar-López
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain. .,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Research Group GA-14, Córdoba, Spain.
| | - Carmen Aguilar
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Carmen Entrenas-García
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Research Group GA-14, Córdoba, Spain
| | - Rocío Bautista
- Andalusian Platform of Bioinformatics-SCBI, University of Málaga, Málaga, Spain
| | - M Gonzalo Claros
- Andalusian Platform of Bioinformatics-SCBI, University of Málaga, Málaga, Spain.,Department of Molecular Biology and Biochemistry, University of Málaga, Málaga, Spain
| | - Juan J Garrido
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Research Group GA-14, Córdoba, Spain
| |
Collapse
|
38
|
Zhuo R, Xu M, Wang X, Zhou B, Wu X, Leone V, Chang EB, Zhong X. The regulatory role of N 6 -methyladenosine modification in the interaction between host and microbes. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1725. [PMID: 35301791 DOI: 10.1002/wrna.1725] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/21/2022] [Accepted: 02/21/2022] [Indexed: 01/02/2023]
Abstract
N6 -methyladenosine (m6 A) is the most prevalent posttranscriptional modification in eukaryotic mRNAs. Dynamic and reversible m6 A modification regulates gene expression to control cellular processes and diverse biological functions. Growing evidence indicated that m6 A modification is involved in the homeostasis of host and microbes (mostly viruses and bacteria). Disturbance of m6 A modification affects the life cycles of viruses and bacteria, however, these microbes could in turn change host m6 A modification leading to human disease including autoimmune diseases and cancer. Thus, we raise the concept that m6 A could be a "messenger" molecule to participate in the interactions between host and microbes. In this review, we summarize the regulatory mechanisms of m6 A modification on viruses and commensal microbiota, highlight the roles of m6 A methylation in the interaction of host and microbes, and finally discuss drugs development targeting m6 A modification. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Ruhao Zhuo
- Joint International Research Laboratory of Animal Health & Food Safety, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Menghui Xu
- Joint International Research Laboratory of Animal Health & Food Safety, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiaoyun Wang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Bin Zhou
- Joint International Research Laboratory of Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xin Wu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Vanessa Leone
- Department of Animal Biologics and Metabolism, University of Wisconsin, Madison, Wisconsin, USA.,Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Eugene B Chang
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Xiang Zhong
- Joint International Research Laboratory of Animal Health & Food Safety, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
39
|
Ding C, Shi T, Wu G, Man J, Han H, Cui Y. The anti-cancer role of microRNA-143 in papillary thyroid carcinoma by targeting high mobility group AT-hook 2. Bioengineered 2022; 13:6629-6640. [PMID: 35213273 PMCID: PMC8973723 DOI: 10.1080/21655979.2022.2044277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Papillary thyroid carcinoma (PTC), a common thyroid cancer (TC) subtype, rapidly increases in occurrence. MicroRNAs (miRNAs), which are non-coding small RNAs, have been demonstrated to play a role in cancer pathogenic mechanisms. Although miR-143 is involved in suppressing certain malignant tumor progression, its biological role is unknown in PTC. The present study found that miR-143 levels were strongly lower in PTC patient samples and cell lines, implying that miR-143 may play a biological role in PTC. Down-regulation of miR-143 resulted in the increased expression of HMGA2. Furthermore, HMGA2 was found to be a direct target of miR-143. A dual-luciferase assay confirmed a direct binding site for miR-143 was confirmed on HMGA2 using a dual-luciferase assay. Next, over-expression of miR-143 suppressed PTC cell growth as analyzed by MTT, clone formation, and Ki-67 immunofluorescence staining assays. miR-143 mimics transfection downregulated the expression of PCNA, CDK4, CDK1, and Cyclin E1. In addition, wound healing and trans-well assays revealed that miR-143 up-regulation inhibited PTC cells invasion and migration. Co-transfection of HMGA2 expression vector restored HMGA2 expression and rescued PTC cells proliferation capability in miR-143 mimics transfected PTC cells, indicating that miR-143 inhibited PTC cells proliferation via HMGA2. These observations were also obtained in xenografts experiments in nude mice. Altogether, our study shed light on miR-143ʹs anti-cancer biological functions in PTC progression through targeting HMGA2, suggesting that restoration of miR-143 could be a potential therapeutic approach for PTC treatment.
Collapse
Affiliation(s)
- Chao Ding
- Departments of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tiefeng Shi
- Departments of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Gang Wu
- Departments of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianting Man
- Departments of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyu Han
- Departments of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yunfu Cui
- Departments of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
40
|
Monaghan TM, Polytarchou C, Kao D, Alexander C, Gurnani P. Therapeutic potential of miRNAs in Clostridioides difficile infection. Future Microbiol 2022; 17:315-318. [PMID: 35172603 DOI: 10.2217/fmb-2021-0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Tweetable abstract Treating Clostridioides difficile infection with miRNAs alone or combined with live biotherapeutic products may augment therapeutic efficacy and help counteract drug resistance in the future.
Collapse
Affiliation(s)
- Tanya M Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG7 2UH, UK.,Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Christos Polytarchou
- Department of Biosciences, John van Geest Cancer Research Centre, School of Science & Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK.,Centre for Health, Aging & Understanding Disease, School of Science & Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Dina Kao
- Department of Medicine, University of Alberta, Edmonton, AB, T6G 2G3, Canada
| | - Cameron Alexander
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Pratik Gurnani
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
41
|
Li N, Wang Z. Integrative Analysis of Deregulated miRNAs Reveals Candidate Molecular Mechanisms Linking H. pylori Infected Peptic Ulcer Disease with Periodontitis. DISEASE MARKERS 2022; 2022:1498525. [PMID: 35132337 PMCID: PMC8817886 DOI: 10.1155/2022/1498525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 01/17/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Periodontitis is a highly prevalent oral infectious disease and has been increasingly associated with H. pylori infection, gastric inflammation, and gastric cancer but little is known about epigenetic machinery underlying this potentially bidirectional association. The present study is aimed at identifying key deregulated miRNA, their associated genes, signaling pathways, and compounds linking periodontitis with H. pylori-associated peptic ulcer disease. METHODS miRNA expression datasets for periodontitis-affected and H. pylori-associated peptic ulcer disease-affected tissues were sought from the GEO database. Differentially expressed miRNA (DEmiRNAs) were identified and the overlapping, shared-DEmiRNA between both datasets were determined. Shared-DEmiRNA-target networks construction and functional analyses were constructed using miRNet 2.0, including shared-DEmiRNA-gene, shared-DEmiRNA-transcription factor (TF), and shared-DEmiRNA-compound networks. Functional enrichment analysis for shared DEmiRNA-gene and shared DEmiRNA-TF networks was performed using the KEGG, Reactome, and Geno Ontology (GO) pathways. RESULTS 11 shared-DEmiRNAs were identified, among which 9 showed similar expression patterns in both diseases, and 7 were overexpressed. miRNA hsa-hsa-mir-155-5p and hsa-mir-29a-3p were top miRNA nodes in both gene and TF networks. The topmost candidate miRNA-deregulated genes were PTEN, CCND1, MDM2, TNRC6A, and SCD while topmost deregulated TFs included STAT3, HIF1A, EZH2, CEBPA, and RUNX1. Curcumin, 5-fluorouracil, and the gallotanin 1,2,6-Tri-O-galloyl-beta-D-glucopyranose emerged as the most relevant linkage compound targets. Functional analyses revealed multiple cancer-associated pathways, PI3K pathways, kinase binding, and transcription factor binding among as enriched by the network-associated genes and TFs. CONCLUSION Integrative analysis of deregulated miRNAs revealed candidate molecular mechanisms comprising of top miRNA, their gene, and TF targets linking H. pylori-infected peptic ulcer disease with periodontitis and highlighted compounds targeting both diseases. These findings provide basis for directing future experimental research.
Collapse
Affiliation(s)
- Ning Li
- Department of Prosthetic Dentistry, The Affiliated Stomatological Hospital of Wenzhou Medical University, Longyao Avenue No. 1288, Yongzhong Street, Longwan District, Wenzhou 325000, Zhejiang Province, China
| | - Zhen Wang
- Department of Stomatology, The Quzhou Affiliated Hospital of Wenzhou Medical University (Quzhou People's Hospital), Kecheng District, Minjiang Avenue No. 100, Quzhou 332400, Zhejiang Province, China
| |
Collapse
|
42
|
Identification of human microRNAs targeting Pseudomonas aeruginosa genes by an in silico hybridization method. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
43
|
Zhou S, Zhu C, Jin S, Cui C, Xiao L, Yang Z, Wang X, Yu J. OUP accepted manuscript. FEMS Microbiol Lett 2022; 369:6607908. [PMID: 35712898 PMCID: PMC9199189 DOI: 10.1093/femsle/fnac023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/24/2022] [Accepted: 03/18/2022] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate the molecular mechanisms through which the intestinal microbiota and microRNAs (miRNAs) participate in colon cancer metastasis. Intestinal flora data, and the GSE29621 (messenger RNA/long non-coding RNA [mRNA/lncRNA]) and GSE29622 (miRNA) datasets, were downloaded from The Cancer Gene Atlas and Gene Expression Omnibus databases, respectively. Immune-related cells in M1 vs. M0 samples were analyzed using the Wilcoxon test. Furthermore, an lncRNA-miRNA-mRNA (competing endogenous RNA [ceRNA]) network was constructed, and survival analysis of RNAs in the network was performed. A total of 16 miRNA-genus co-expression pairs containing eight microbial genera and 15 miRNAs were screened; notably, Porphyromonas and Bifidobacterium spp. were found to be associated with most miRNAs, and has-miR-3943 was targeted by most microbial genera. Furthermore, five immune cell types, including activated natural killer cells, M1 macrophages, resting mast cells, activated mast cells and neutrophils, were differentially accumulated between the M1 and M0 groups. Enrichment analysis suggested that mRNAs related to colon cancer metastasis were mainly involved in pathways related to bacterial and immune responses. Survival analysis revealed that TMEM176A and PALM3 in the ceRNA network were significantly associated with the prognosis of patients with colon cancer. In conclusion, this study revealed a potential mechanism by which the intestinal microbiota influences the colon cancer microenvironment by targeting miRNAs.
Collapse
Affiliation(s)
| | | | | | - Chunhui Cui
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, 510000, China
| | - Linghui Xiao
- Department of Gastrointestinal Surgery, Huizhou First Hospital, Huizhou, Guangdong, 516003, China
| | - Zhi Yang
- The IVD Medical Marketing Department, 3D Medicines Inc., Shanghai, 201114, China
| | - Xi Wang
- Corresponding author: Department of Gastrointestinal Surgery, Huizhou First Hospital, Huizhou, Guangdong, 516003, China. E-mail:
| | - Jinlong Yu
- Corresponding author: Department of General Surgery, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Haizhu District, Guangzhou, 510000, Guangdong Province, China. E-mail:
| |
Collapse
|
44
|
Magryś A, Bogut A. MicroRNA hsa-let-7a facilitates staphylococcal small colony variants survival in the THP-1 macrophages by reshaping inflammatory responses. Int J Med Microbiol 2021; 311:151542. [PMID: 34864353 DOI: 10.1016/j.ijmm.2021.151542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/16/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022] Open
Abstract
Recent studies have provided emerging evidence of the critical involvement of microRNAs in host immune defence against bacterial infection and that likewise the expression of the miRNAs is profoundly impacted by a variety of pathogens to subvert the immune response. Here, we report the role of hsa-let-7a miRNA in response to Staphylococcus epidermidis Small Colony Variants infection. We also assessed whether the expression levels of inflammatory cytokines associated with the hsa-let-7a are manipulated by the pathogen and the effect of the IFN-γ priming on the expression of hsa-let-7a and the fate of SCVs/WTs in infected macrophages. A striking observation was the downregulation of the let-7a miRNA upon challenge of the THP-1 activated cells with the SCV isolates while no significant changes in expression were noticed after the infection of macrophages with their WT counterparts. Staphylococcus epidermidis WT and SCV strains were found to invade and survive in macrophages. A significant reduction in bacterial load for both phenotypes was observed in macrophages treated with let-7a mimic compared to untreated ones. Survival of WTs was augmented in cells treated with the inhibitor in 4 out of 5 strains as compared to the number of bacteria recovered from non-transfected cells. At the same time, let-7a inhibitor did not influence on the survival of SCVs in macrophages as their number was comparable to number recovered from non-transfected cells. When the ratio of both let-7a cytokine targets was compared, anti-inflammatory IL-10 cytokine was induced by SCVs predominantly, while the macrophage challenge with WTs was characterized by the inflammatory cytokine profile with high IL-6 and low IL-10 production. Moreover, the balance between pro-inflammatory and anti-inflammatory cytokines has been expectedly retrieved when macrophages were transfected with let-7a mimic before infection with WT or SCV strains. The results also show that IFN-γ likely regulates the macrophage environment contributing to the inflammatory response and elimination of bacteria from intracellular milieu by augmenting the synthesis of pro-inflammatory cytokines and supressing the anti-inflammatory IL-10. Our work has shown that SCVs have the potential to regulate the let-7a miRNA to balance the pro-inflammatory IL-6 with anti-inflammatory IL-10 and this mechanism is one of the ways in a complex regulatory network adopted by SCVs to promote their survival.
Collapse
Affiliation(s)
- Agnieszka Magryś
- Chair and Department of Medical Microbiology, Medical University of Lublin, Poland.
| | - Agnieszka Bogut
- Chair and Department of Medical Microbiology, Medical University of Lublin, Poland
| |
Collapse
|
45
|
Lisowski C, Dias J, Costa S, Silva RJ, Mano M, Eulalio A. Dysregulated endolysosomal trafficking in cells arrested in the G 1 phase of the host cell cycle impairs Salmonella vacuolar replication. Autophagy 2021; 18:1785-1800. [PMID: 34781820 DOI: 10.1080/15548627.2021.1999561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Modulation of the host cell cycle has emerged as a common theme among the pathways regulated by bacterial pathogens, arguably to promote host cell colonization. However, in most cases the exact benefit ensuing from such interference to the infection process remains unclear. Previously, we have shown that Salmonella actively induces G2/M arrest of host cells, and that infection is severely inhibited in cells arrested in G1. In this study, we demonstrate that Salmonella vacuolar replication is inhibited in host cells blocked in G1, whereas the cytosolic replication of the closely related pathogen Shigella is not affected. Mechanistically, we show that cells arrested in G1, but not cells arrested in G2, present dysregulated endolysosomal trafficking, displaying an abnormal accumulation of vesicles positive for late endosomal and lysosomal markers. In addition, the macroautophagic/autophagic flux and degradative lysosomal function are strongly impaired. This endolysosomal trafficking dysregulation results in sustained activation of the SPI-1 type III secretion system and lack of vacuole repair by the autophagy pathway, ultimately compromising the maturation and integrity of the Salmonella-containing vacuole. As such, Salmonella is released in the host cytosol. Collectively, our findings demonstrate that the modulation of the host cell cycle occurring during Salmonella infection is related to a disparity in the permissivity of cells arrested in G1 and G2/M, due to their intrinsic characteristics.
Collapse
Affiliation(s)
- Clivia Lisowski
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Jane Dias
- RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Susana Costa
- RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Ricardo Jorge Silva
- Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany.,RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
46
|
Identification of putative microRNAs in the complete genome of Mycobacterium avium and their possible interaction with human transcripts. J Appl Genet 2021; 63:169-182. [PMID: 34677783 DOI: 10.1007/s13353-021-00666-9] [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: 05/23/2021] [Revised: 08/05/2021] [Accepted: 10/14/2021] [Indexed: 10/20/2022]
Abstract
The grievous adversity regarding Mycobacterium avium is its ubiquitous nature. Isolation of the bacteria from drinking water, house dust, and soil, etc., is an alarming issue for the scientific community. The microRNAs are the molecular influencers of gene expression that act during the process of post transcription. A few reports claimed the existence of microRNAs or microRNA-like molecules in the prokaryotic species. Biogenesis of bacterial miRNAs requires their transport into the host cell. Subsequently, the host-encoded enzymes are exerted for the formation of bacterial mature miRNAs and their regulation. In our study, the screening of complete genome of Mycobacterium avium revealed six putative precursor microRNA sequences bearing typical secondary structures. The mature microRNAs were predicted in both arms of the secondary structures. A total of 12 possible mature microRNAs were identified in this study. The likely targets of the predicted mature miRNAs were searched in human 3' UTR. In the human transcriptome, 193 genes were possibly targeted by 12 mature miRNAs of Mycobacterium avium. The essential functionalities of the target genes included signal transduction, immune system, DNA binding, and response to stress.
Collapse
|
47
|
Prashar A, Capurro MI, Jones NL. Under the Radar: Strategies Used by Helicobacter pylori to Evade Host Responses. Annu Rev Physiol 2021; 84:485-506. [PMID: 34672717 DOI: 10.1146/annurev-physiol-061121-035930] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The body depends on its physical barriers and innate and adaptive immune responses to defend against the constant assault of potentially harmful microbes. In turn, successful pathogens have evolved unique mechanisms to adapt to the host environment and manipulate host defenses. Helicobacter pylori (Hp), a human gastric pathogen that is acquired in childhood and persists throughout life, is an example of a bacterium that is very successful at remodeling the host-pathogen interface to promote a long-term persistent infection. Using a combination of secreted virulence factors, immune subversion, and manipulation of cellular mechanisms, Hp can colonize and persist in the hostile environment of the human stomach. Here, we review the most recent and relevant information regarding how this successful pathogen overcomes gastric epithelial host defense responses to facilitate its own survival and establish a chronic infection. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Akriti Prashar
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Mariana I Capurro
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Nicola L Jones
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada; .,Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada.,Departments of Paediatrics and Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
48
|
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
|
49
|
Singh R, Zogg H, Ro S. Role of microRNAs in Disorders of Gut-Brain Interactions: Clinical Insights and Therapeutic Alternatives. J Pers Med 2021; 11:1021. [PMID: 34683162 PMCID: PMC8541612 DOI: 10.3390/jpm11101021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Disorders of gut-brain interactions (DGBIs) are heterogeneous in nature and intertwine with diverse pathophysiological mechanisms. Regular functioning of the gut requires complex coordinated interplay between a variety of gastrointestinal (GI) cell types and their functions are regulated by multiple mechanisms at the transcriptional, post-transcriptional, translational, and post-translational levels. MicroRNAs (miRNAs) are small non-coding RNA molecules that post-transcriptionally regulate gene expression by binding to specific mRNA targets to repress their translation and/or promote the target mRNA degradation. Dysregulation of miRNAs might impair gut physiological functions leading to DGBIs and gut motility disorders. Studies have shown miRNAs regulate gut functions such as visceral sensation, gut immune response, GI barrier function, enteric neuronal development, and GI motility. These biological processes are highly relevant to the gut where neuroimmune interactions are key contributors in controlling gut homeostasis and functional defects lead to DGBIs. Although extensive research has explored the pathophysiology of DGBIs, further research is warranted to bolster the molecular mechanisms behind these disorders. The therapeutic targeting of miRNAs represents an attractive approach for the treatment of DGBIs because they offer new insights into disease mechanisms and have great potential to be used in the clinic as diagnostic markers and therapeutic targets. Here, we review recent advances regarding the regulation of miRNAs in GI pacemaking cells, immune cells, and enteric neurons modulating pathophysiological mechanisms of DGBIs. This review aims to assess the impacts of miRNAs on the pathophysiological mechanisms of DGBIs, including GI dysmotility, impaired intestinal barrier function, gut immune dysfunction, and visceral hypersensitivity. We also summarize the therapeutic alternatives for gut microbial dysbiosis in DGBIs, highlighting the clinical insights and areas for further exploration. We further discuss the challenges in miRNA therapeutics and promising emerging approaches.
Collapse
Affiliation(s)
| | | | - Seungil Ro
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, 1664 North Virginia Street, Reno, NV 89557, USA; (R.S.); (H.Z.)
| |
Collapse
|
50
|
Wang S, Liu Y, Li J, Zhao L, Yan W, Lin B, Guo X, Wei Y. Fusobacterium nucleatum Acts as a Pro-carcinogenic Bacterium in Colorectal Cancer: From Association to Causality. Front Cell Dev Biol 2021; 9:710165. [PMID: 34490259 PMCID: PMC8417943 DOI: 10.3389/fcell.2021.710165] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a common cancer worldwide with complex etiology. Fusobacterium nucleatum (F. nucleatum), an oral symbiotic bacterium, has been linked with CRC in the past decade. A series of gut microbiota studies show that CRC patients carry a high abundance of F. nucleatum in the tumor tissue and fecal, and etiological studies have clarified the role of F. nucleatum as a pro-carcinogenic bacterium in various stages of CRC. In this review, we summarize the biological characteristics of F. nucleatum and the epidemiological associations between F. nucleatum and CRC, and then highlight the mechanisms by which F. nucleatum participates in CRC progression, metastasis, and chemoresistance by affecting cancer cells or regulating the tumor microenvironment (TME). We also discuss the research gap in this field and give our perspective for future studies. These findings will pave the way for manipulating gut F. nucleatum to deal with CRC in the future.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Liu
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun Li
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Zhao
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Yan
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baiqiang Lin
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Guo
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunwei Wei
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|