1
|
Acevedo-Román A, Pagán-Zayas N, Velázquez-Rivera LI, Torres-Ventura AC, Godoy-Vitorino F. Insights into Gut Dysbiosis: Inflammatory Diseases, Obesity, and Restoration Approaches. Int J Mol Sci 2024; 25:9715. [PMID: 39273662 PMCID: PMC11396321 DOI: 10.3390/ijms25179715] [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: 06/24/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024] Open
Abstract
The gut microbiota is one of the most critical factors in human health. It involves numerous physiological processes impacting host health, mainly via immune system modulation. A balanced microbiome contributes to the gut's barrier function, preventing the invasion of pathogens and maintaining the integrity of the gut lining. Dysbiosis, or an imbalance in the gut microbiome's composition and function, disrupts essential processes and contributes to various diseases. This narrative review summarizes key findings related to the gut microbiota in modern multifactorial inflammatory conditions such as ulcerative colitis or Crohn's disease. It addresses the challenges posed by antibiotic-driven dysbiosis, particularly in the context of C. difficile infections, and the development of novel therapies like fecal microbiota transplantation and biotherapeutic drugs to combat these infections. An emphasis is given to restoration of the healthy gut microbiome through dietary interventions, probiotics, prebiotics, and novel approaches for managing gut-related diseases.
Collapse
Affiliation(s)
- Andy Acevedo-Román
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Natalia Pagán-Zayas
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Liz I Velázquez-Rivera
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Aryanne C Torres-Ventura
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Filipa Godoy-Vitorino
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| |
Collapse
|
2
|
Zhang J, Zhang Y, Ren Z, Yan D, Li G. The role of TRIM family in metabolic associated fatty liver disease. Front Endocrinol (Lausanne) 2023; 14:1210330. [PMID: 37867509 PMCID: PMC10585262 DOI: 10.3389/fendo.2023.1210330] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
Metabolic associated fatty liver disease (MAFLD) ranks among the most prevalent chronic liver conditions globally. At present, the mechanism of MAFLD has not been fully elucidated. Tripartite motif (TRIM) protein is a kind of protein with E3 ubiquitin ligase activity, which participates in highly diversified cell activities and processes. It not only plays an important role in innate immunity, but also participates in liver steatosis, insulin resistance and other processes. In this review, we focused on the role of TRIM family in metabolic associated fatty liver disease. We also introduced the structure and functions of TRIM proteins. We summarized the TRIM family's regulation involved in the occurrence and development of metabolic associated fatty liver disease, as well as insulin resistance. We deeply discussed the potential of TRIM proteins as targets for the treatment of metabolic associated fatty liver disease.
Collapse
Affiliation(s)
- Jingyue Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Yingming Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Ze Ren
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Dongmei Yan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Guiying Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
3
|
Yu H, Jia X, Niu H, Xie L, Du B, Pang Y, Xu X, Li J. miR-23a regulates the disease resistance of grass carp (Ctenopharyngodon idella) by targeting autophagy-related genes, ATG3 and ATG12. FISH & SHELLFISH IMMUNOLOGY 2023; 138:108812. [PMID: 37172750 DOI: 10.1016/j.fsi.2023.108812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
miRNAs play a key role in the autophagy process. In recent years, the emerging role of autophagy in regulating immune response has attracted increasing attention. Since then, specific miRNAs have also been found to play an immune function indirectly by modulating autophagy as well. This study proved that miR-23a could downregulate grass carp autophagy simultaneously by targeting ATG3 and ATG12. Besides, both ATG3 and ATG12 mRNA levels were increased in kidney and intestine after being infected by Aeromonas hydrophila; yet almost at the same time, miR-23a was decreased. Besides, we illustrated that grass carp miR-23a could affect antimicrobial capacity, proliferation, migration, and antiapoptotic abilities of CIK cells. These results indicate that miR-23a was related to grass carp autophagy and plays an important role in antimicrobial immunity through targeting ATG3 and ATG12, which provides important information on autophagy-related miRNAs about the defense and immune mechanisms against pathogens in teleost.
Collapse
Affiliation(s)
- Hongyan Yu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Xuewen Jia
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Huiqin Niu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Lingli Xie
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Biao Du
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yifang Pang
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Xiaoyan Xu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| | - Jiale Li
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
4
|
Ellwanger JH, Kulmann-Leal B, Ziliotto M, Chies JAB. HIV Infection, Chromosome Instability, and Micronucleus Formation. Viruses 2023; 15:155. [PMID: 36680195 PMCID: PMC9867034 DOI: 10.3390/v15010155] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Genome integrity is critical for proper cell functioning, and chromosome instability can lead to age-related diseases, including cancer and neurodegenerative disorders. Chromosome instability is caused by multiple factors, including replication stress, chromosome missegregation, exposure to pollutants, and viral infections. Although many studies have investigated the effects of environmental or lifestyle genotoxins on chromosomal integrity, information on the effects of viral infections on micronucleus formation and other chromosomal aberrations is still limited. Currently, HIV infection is considered a chronic disease treatable by antiretroviral therapy (ART). However, HIV-infected individuals still face important health problems, such as chronic inflammation and age-related diseases. In this context, this article reviews studies that have evaluated genomic instability using micronucleus assays in the context of HIV infection. In brief, HIV can induce chromosome instability directly through the interaction of HIV proteins with host DNA and indirectly through chronic inflammation or as a result of ART use. Connections between HIV infection, immunosenescence and age-related disease are discussed in this article. The monitoring of HIV-infected individuals should consider the increased risk of chromosome instability, and lifestyle interventions, such as reduced exposure to genotoxins and an antioxidant-rich diet, should be considered. Therapies to reduce chronic inflammation in HIV infection are needed.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil
| | | | | | - José Artur Bogo Chies
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil
| |
Collapse
|
5
|
Huang N, Sun X, Li P, Liu X, Zhang X, Chen Q, Xin H. TRIM family contribute to tumorigenesis, cancer development, and drug resistance. Exp Hematol Oncol 2022; 11:75. [PMID: 36261847 PMCID: PMC9583506 DOI: 10.1186/s40164-022-00322-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/16/2022] [Indexed: 11/26/2022] Open
Abstract
The tripartite-motif (TRIM) family represents one of the largest classes of putative single protein RING-finger E3 ubiquitin ligases. TRIM family is involved in a variety of cellular signaling transductions and biological processes. TRIM family also contributes to cancer initiation, progress, and therapy resistance, exhibiting oncogenic and tumor-suppressive functions in different human cancer types. Moreover, TRIM family members have great potential to serve as biomarkers for cancer diagnosis and prognosis. In this review, we focus on the specific mechanisms of the participation of TRIM family members in tumorigenesis, and cancer development including interacting with dysregulated signaling pathways such as JAK/STAT, PI3K/AKT, TGF-β, NF-κB, Wnt/β-catenin, and p53 hub. In addition, many studies have demonstrated that the TRIM family are related to tumor resistance; modulate the epithelial–mesenchymal transition (EMT) process, and guarantee the acquisition of cancer stem cells (CSCs) phenotype. In the end, we havediscussed the potential of TRIM family members for cancer therapeutic targets.
Collapse
Affiliation(s)
- Ning Huang
- Department of Pharmacology, School of Pharmacy & General Surgery of Minhang Hospital, Fudan University, Shanghai, 201203, China.,PharmaLegacy Laboratories Co.,Ltd, Shengrong Road No.388, Zhangjiang High-tech Park, Pudong New Area, Shanghai, China
| | - Xiaolin Sun
- Department of Pharmacology, School of Pharmacy & General Surgery of Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Peng Li
- Department of Pharmacology, School of Pharmacy & General Surgery of Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Xin Liu
- Department of Pharmacology, School of Pharmacy & General Surgery of Minhang Hospital, Fudan University, Shanghai, 201203, China.,PharmaLegacy Laboratories Co.,Ltd, Shengrong Road No.388, Zhangjiang High-tech Park, Pudong New Area, Shanghai, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy & General Surgery of Minhang Hospital, Fudan University, Shanghai, 201203, China.
| | - Qian Chen
- Department of Pharmacology, School of Pharmacy & General Surgery of Minhang Hospital, Fudan University, Shanghai, 201203, China.
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy & General Surgery of Minhang Hospital, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
6
|
Mehto S, Jena KK, Yadav R, Priyadarsini S, Samal P, Krishna S, Dhar K, Jain A, Chauhan NR, Murmu KC, Bal R, Sahu R, Jaiswal P, Sahoo BS, Patnaik S, Kufer TA, Rusten TE, Chauhan S, Prasad P, Chauhan S. Selective autophagy of RIPosomes maintains innate immune homeostasis during bacterial infection. EMBO J 2022; 41:e111289. [PMID: 36221902 PMCID: PMC9713718 DOI: 10.15252/embj.2022111289] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 01/15/2023] Open
Abstract
The NOD1/2-RIPK2 is a key cytosolic signaling complex that activates NF-κB pro-inflammatory response against invading pathogens. However, uncontrolled NF-κB signaling can cause tissue damage leading to chronic diseases. The mechanisms by which the NODs-RIPK2-NF-κB innate immune axis is activated and resolved remain poorly understood. Here, we demonstrate that bacterial infection induces the formation of endogenous RIPK2 oligomers (RIPosomes) that are self-assembling entities that coat the bacteria to induce NF-κB response. Next, we show that autophagy proteins IRGM and p62/SQSTM1 physically interact with NOD1/2, RIPK2 and RIPosomes to promote their selective autophagy and limit NF-κB activation. IRGM suppresses RIPK2-dependent pro-inflammatory programs induced by Shigella and Salmonella. Consistently, the therapeutic inhibition of RIPK2 ameliorates Shigella infection- and DSS-induced gut inflammation in Irgm1 KO mice. This study identifies a unique mechanism where the innate immune proteins and autophagy machinery are recruited together to the bacteria for defense as well as for maintaining immune homeostasis.
Collapse
Affiliation(s)
- Subhash Mehto
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia
| | - Kautilya Kumar Jena
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia,Present address:
Division of Immunology, Boston Children's HospitalHarvard Medical SchoolBostonMAUSA
| | - Rina Yadav
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia,Regional Centre for Biotechnology, NCR Biotech Science ClusterFaridabadIndia
| | | | - Pallavi Samal
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia
| | - Sivaram Krishna
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia,Regional Centre for Biotechnology, NCR Biotech Science ClusterFaridabadIndia
| | - Kollori Dhar
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia,Regional Centre for Biotechnology, NCR Biotech Science ClusterFaridabadIndia
| | - Ashish Jain
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway,Department of Molecular Cell Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Nishant Ranjan Chauhan
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia
| | - Krushna C Murmu
- Epigenetic and Chromatin Biology UnitInstitute of Life SciencesBhubaneswarIndia
| | - Ramyasingh Bal
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia,School of BiotechnologyKIIT UniversityBhubaneswarIndia
| | - Rinku Sahu
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia,Regional Centre for Biotechnology, NCR Biotech Science ClusterFaridabadIndia
| | - Pundrik Jaiswal
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia
| | | | | | - Thomas A Kufer
- Department of Immunology, Institute of Nutritional MedicineUniversity of HohenheimStuttgartGermany
| | - Tor Erik Rusten
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway,Department of Molecular Cell Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Swati Chauhan
- Epigenetic and Chromatin Biology UnitInstitute of Life SciencesBhubaneswarIndia
| | - Punit Prasad
- Epigenetic and Chromatin Biology UnitInstitute of Life SciencesBhubaneswarIndia
| | - Santosh Chauhan
- Cell Biology and Infectious Diseases Unit, Department of Infectious Disease BiologyInstitute of Life SciencesBhubaneswarIndia,CSIR–Centre For Cellular And Molecular Biology (CCMB)HyderabadIndia
| |
Collapse
|
7
|
van der Vossen EWJ, de Goffau MC, Levin E, Nieuwdorp M. Recent insights into the role of microbiome in the pathogenesis of obesity. Therap Adv Gastroenterol 2022; 15:17562848221115320. [PMID: 35967920 PMCID: PMC9373125 DOI: 10.1177/17562848221115320] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 07/06/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity is a risk factor for many chronic diseases and its rising prevalence the last couple of decades is a healthcare concern in many countries. Obesity is a multifactorial problem that is not only limited in its causation by diet and lack of exercise. Genetics but also environmental factors such as the gut microbiome should similarly be taken into account. A plethora of articles have been published, that from various different angles, attempt to disentangle the complex interaction between gut microbiota and obesity. Examples range from the effect of the gut microbiota on the host immune system to the pathophysiological pathways in which microbial-derived metabolites affect obesity. Various discordant gut microbiota findings are a result of this complexity. In this review, in addition to summarizing the classical role of the gut microbiome in the pathogenesis of obesity, we attempt to view both the healthy and obesogenic effects of the gut microbiota as a consequence of the presence or absence of collective guilds/trophic networks. Lastly, we propose avenues and strategies for the future of gut microbiome research concerning obesity.
Collapse
Affiliation(s)
- Eduard W. J. van der Vossen
- Department of Experimental Vascular Medicine,
Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The
Netherlands
| | - Marcus C. de Goffau
- Department of Experimental Vascular Medicine,
Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The
Netherlands
| | - Evgeni Levin
- Department of Experimental Vascular Medicine,
Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The
Netherlands,Horaizon BV, Delft, The Netherlands
| | | |
Collapse
|
8
|
Kienes I, Johnston EL, Bitto NJ, Kaparakis-Liaskos M, Kufer TA. Bacterial subversion of NLR-mediated immune responses. Front Immunol 2022; 13:930882. [PMID: 35967403 PMCID: PMC9367220 DOI: 10.3389/fimmu.2022.930882] [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: 04/28/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
Members of the mammalian Nod-like receptor (NLR) protein family are important intracellular sensors for bacteria. Bacteria have evolved under the pressure of detection by host immune sensing systems, leading to adaptive subversion strategies to dampen immune responses for their benefits. These include modification of microbe-associated molecular patterns (MAMPs), interception of innate immune pathways by secreted effector proteins and sophisticated instruction of anti-inflammatory adaptive immune responses. Here, we summarise our current understanding of subversion strategies used by bacterial pathogens to manipulate NLR-mediated responses, focusing on the well-studied members NOD1/2, and the inflammasome forming NLRs NLRC4, and NLRP3. We discuss how bacterial pathogens and their products activate these NLRs to promote inflammation and disease and the range of mechanisms used by bacterial pathogens to evade detection by NLRs and to block or dampen NLR activation to ultimately interfere with the generation of host immunity. Moreover, we discuss how bacteria utilise NLRs to facilitate immunotolerance and persistence in the host and outline how various mechanisms used to attenuate innate immune responses towards bacterial pathogens can also aid the host by reducing immunopathologies. Finally, we describe the therapeutic potential of harnessing immune subversion strategies used by bacteria to treat chronic inflammatory conditions.
Collapse
Affiliation(s)
- Ioannis Kienes
- Department of Immunology, University of Hohenheim, Stuttgart, Germany
| | - Ella L. Johnston
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Natalie J. Bitto
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Thomas A. Kufer
- Department of Immunology, University of Hohenheim, Stuttgart, Germany
- *Correspondence: Thomas A. Kufer,
| |
Collapse
|
9
|
Afonina IS, Hoffmann E. Infection and immunity: ‘There Are Things Out There You (Don't) Need To Know About’. FEBS J 2022; 289:3920-3925. [DOI: 10.1111/febs.16569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Inna S. Afonina
- Unit of Molecular Signal Transduction in Inflammation VIB‐UGent Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology Ghent University Belgium
| | - Eik Hoffmann
- Institute Pasteur Lille Center for Infection and Immunity of Lille (CIIL) France
- European Regimen Accelerator for Tuberculosis (ERA4TB), Innovative Medicines Initiative 2 Joint Undertaking (JU no. 853989)
| |
Collapse
|
10
|
Role of GBP1 in innate immunity and potential as a tuberculosis biomarker. Sci Rep 2022; 12:11097. [PMID: 35773466 PMCID: PMC9247026 DOI: 10.1038/s41598-022-15482-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/24/2022] [Indexed: 01/19/2023] Open
Abstract
Tuberculosis (TB) is a global health problem of major concern. Identification of immune biomarkers may facilitate the early diagnosis and targeted treatment of TB. We used public RNA-sequencing datasets of patients with TB and healthy controls to identify differentially expressed genes and their associated functional networks. GBP1 expression was consistently significantly upregulated in TB, and 4492 differentially expressed genes were simultaneously associated with TB and high GBP1 expression. Weighted gene correlation analysis identified 12 functional modules. Modules positively correlated with TB and high GBP1 expression were associated with the innate immune response, neutrophil activation, neutrophil-mediated immunity, and NOD receptor signaling pathway. Eleven hub genes (GBP1, HLA-B, ELF4, HLA-E, IFITM2, TNFRSF14, CD274, AIM2, CFB, RHOG, and HORMAD1) were identified. The least absolute shrinkage and selection operator model based on hub genes accurately predicted the occurrence of TB (area under the receiver operating characteristic curve = 0.97). The GBP1-module-pathway network based on the STRING database showed that GBP1 expression correlated with the expression of interferon-stimulated genes (GBP5, BATF2, EPSTI1, RSAD2, IFI44L, IFIT3, and OAS3). Our study suggests GBP1 as an optimal diagnostic biomarker for TB, further indicating an association of the AIM2 inflammasome signaling pathway in TB pathology.
Collapse
|