1
|
Datta A, Gupta D, Waryani D, C GPD. Decoding differentially expressed genes to identify potential immunity associated biomarkers in Tuberculosis: An integrative bioinformatics approach. Biochem Biophys Rep 2024; 40:101870. [PMID: 39649798 PMCID: PMC11625171 DOI: 10.1016/j.bbrep.2024.101870] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/09/2024] [Indexed: 12/11/2024] Open
Abstract
Tuberculosis (TB) poses a significant threat to the Indian population, with India accounting for 20 % of the global TB cases. The current study aims to identify molecular biomarkers for better diagnostics by comparing the transcriptome signatures of healthy individuals against TB-affected individuals. Next-generation sequencing (NGS) tools were used to identify critical differentially expressed genes (DEGs). 302 DEGs were identified based on a logFC threshold of |3| and adjusted p-value < 0.05. STRING database was used to plot the interactions amongst the 302 DEGs. The DEGs were functionally annotated, highlighting numerous physiological functions affected due to the dysregulation of the identified hub genes. TLR4, FCGR1A, ITGAM, LTF, and CXCR2 were the hub genes identified and observed to dysregulate crucial physiological functions. TLR4 has been implicated in the progression of TB in various populations, and the findings of this study will enable researchers to improve the current landscape of diagnostics for TB.
Collapse
Affiliation(s)
- Ankur Datta
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Divyanshi Gupta
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Diya Waryani
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| |
Collapse
|
2
|
Farvardin A, Llorens E, Liu-Xu L, Sánchez-Giménez L, Wong A, Biosca EG, Pedra JM, Falomir E, Camañes G, Scalschi L, Vicedo B. Solanum lycopersicum heme-binding protein 2 as a potent antimicrobial weapon against plant pathogens. Sci Rep 2023; 13:20336. [PMID: 37990046 PMCID: PMC10663603 DOI: 10.1038/s41598-023-47236-z] [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: 07/28/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
The rise in antibiotic-resistant bacteria caused by the excessive use of antibiotics has led to the urgent exploration of alternative antimicrobial solutions. Among these alternatives, antimicrobial proteins, and peptides (Apps) have garnered attention due to their wide-ranging antimicrobial effects. This study focuses on evaluating the antimicrobial properties of Solanum lycopersicum heme-binding protein 2 (SlHBP2), an apoplastic protein extracted from tomato plants treated with 1-Methyl tryptophan (1-MT), against Pseudomonas syringae pv. tomato DC3000 (Pst). Computational studies indicate that SlHBP2 is annotated as a SOUL heme-binding family protein. Remarkably, recombinant SlHBP2 demonstrated significant efficacy in inhibiting the growth of Pst within a concentration range of 3-25 μg/mL. Moreover, SlHBP2 exhibited potent antimicrobial effects against other microorganisms, including Xanthomonas vesicatoria (Xv), Clavibacter michiganensis subsp. michiganensis (Cmm), and Botrytis cinerea. To understand the mechanism of action employed by SlHBP2 against Pst, various techniques such as microscopy and fluorescence assays were employed. The results revealed that SlHBP2 disrupts the bacterial cell wall and causes leakage of intracellular contents. To summarize, the findings suggest that SlHBP2 has significant antimicrobial properties, making it a potential antimicrobial agent against a wide range of pathogens. Although further studies are warranted to explore the full potential of SlHBP2 and its suitability in various applications.
Collapse
Affiliation(s)
- Atefeh Farvardin
- Biochemistry and Biotechnology Group, Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| | - Eugenio Llorens
- Biochemistry and Biotechnology Group, Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| | - Luisa Liu-Xu
- Biochemistry and Biotechnology Group, Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| | - Lorena Sánchez-Giménez
- Biochemistry and Biotechnology Group, Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| | - Aloysius Wong
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang, China
| | - Elena G Biosca
- Department of Microbiology and Ecology, Universitat de Valencia, E-46100, Valencia, Spain
| | - José M Pedra
- Central Service of Scientific Instrumentation, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| | - Eva Falomir
- Department of Inorganic and Organic Chemistry, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| | - Gemma Camañes
- Biochemistry and Biotechnology Group, Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| | - Loredana Scalschi
- Biochemistry and Biotechnology Group, Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, 12071, Castellón de la Plana, Spain.
| | - Begonya Vicedo
- Biochemistry and Biotechnology Group, Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, 12071, Castellón de la Plana, Spain
| |
Collapse
|
3
|
Ma Y, Fei Y, Ding S, Jiang H, Fang J, Liu G. Trace metal elements: a bridge between host and intestinal microorganisms. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1976-1993. [PMID: 37528296 DOI: 10.1007/s11427-022-2359-4] [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: 02/15/2023] [Accepted: 04/23/2023] [Indexed: 08/03/2023]
Abstract
Trace metal elements, such as iron, copper, manganese, and zinc, are essential nutrients for biological processes. Although their intake demand is low, they play a crucial role in cell homeostasis as the cofactors of various enzymes. Symbiotic intestinal microorganisms compete with their host for the use of trace metal elements. Moreover, the metabolic processes of trace metal elements in the host and microorganisms affect the organism's health. Supplementation or the lack of trace metal elements in the host can change the intestinal microbial community structure and function. Functional changes in symbiotic microorganisms can affect the host's metabolism of trace metal elements. In this review, we discuss the absorption and transport processes of trace metal elements in the host and symbiotic microorganisms and the effects of dynamic changes in the levels of trace metal elements on the intestinal microbial community structure. We also highlight the participation of trace metal elements as enzyme cofactors in the host immune process. Our findings indicate that the host uses metal nutrition immunity or metal poisoning to resist pathogens and improve immunity.
Collapse
Affiliation(s)
- Yong Ma
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Yanquan Fei
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Hongmei Jiang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China.
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| |
Collapse
|
4
|
Gao M, Zhao T, Zhang C, Li P, Wang J, Han J, Zhang N, Pang B, Liu S. Ferritinophagy-mediated iron competition in RUTIs: Tug-of-war between UPEC and host. Biomed Pharmacother 2023; 163:114859. [PMID: 37167722 DOI: 10.1016/j.biopha.2023.114859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is the main pathogen of recurrent urinary tract infections (RUTIs). Urinary tract infection is a complicated interaction between UPEC and the host. During infection, UPEC can evade the host's immune response and retain in bladder epithelial cells, which requires adequate nutritional support. Iron is the first necessary trace element in life and a key nutritional factor, making it an important part of the competition between UPEC and the host. On the one hand, UPEC grabs iron to satisfy its reproduction, on the other hand, the host relies on iron to build nutritional immunity defenses against UPEC. Ferritinophagy is a selective autophagy of ferritin mediated by nuclear receptor coactivator 4, which is not only a way for the host to regulate iron metabolism to maintain iron homeostasis, but also a key point of competition between the host and UPEC. Although recent studies have confirmed the role of ferritinophagy in the progression of many diseases, the mechanism of potential interactions between ferritinophagy in UPEC and the host is poorly understood. In this paper, we reviewed the potential mechanisms of ferritinophagy-mediated iron competition in the UPEC-host interactions. This competitive relationship, like a tug-of-war, is a confrontation between the capability of UPEC to capture iron and the host's nutritional immunity defense, which could be the trigger for RUTIs. Therefore, understanding ferritinophagy-mediated iron competition may provide new strategies for exploring effective antibiotic alternative therapies to prevent and treat RUTIs.
Collapse
Affiliation(s)
- Mengqi Gao
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Tingting Zhao
- Department of Nephrology, Beijing Key Laboratory for Immune-Mediated Inflammatory 9 Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Chuanlong Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ping Li
- Department of Nephrology, Beijing Key Laboratory for Immune-Mediated Inflammatory 9 Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jiazhe Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiatong Han
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ning Zhang
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Bo Pang
- International Medical Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Shiwei Liu
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China.
| |
Collapse
|
5
|
Dai Y, Zhu C, Xiao W, Huang K, Wang X, Shi C, Lin D, Zhang H, Liu X, Peng B, Gao Y, Liu CH, Ge B, Kaufmann SH, Feng CG, Chen X, Cai Y. Mycobacterium tuberculosis hijacks host TRIM21- and NCOA4-dependent ferritinophagy to enhance intracellular growth. J Clin Invest 2023; 133:159941. [PMID: 37066876 PMCID: PMC10104892 DOI: 10.1172/jci159941] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 02/28/2023] [Indexed: 04/18/2023] Open
Abstract
Ferritin, a key regulator of iron homeostasis in macrophages, has been reported to confer host defenses against Mycobacterium tuberculosis (Mtb) infection. Nuclear receptor coactivator 4 (NCOA4) was recently identified as a cargo receptor in ferritin degradation. Here, we show that Mtb infection enhanced NCOA4-mediated ferritin degradation in macrophages, which in turn increased the bioavailability of iron to intracellular Mtb and therefore promoted bacterial growth. Of clinical relevance, the upregulation of FTH1 in macrophages was associated with tuberculosis (TB) disease progression in humans. Mechanistically, Mtb infection enhanced NCOA4-mediated ferritin degradation through p38/AKT1- and TRIM21-mediated proteasomal degradation of HERC2, an E3 ligase of NCOA4. Finally, we confirmed that NCOA4 deficiency in myeloid cells expedites the clearance of Mtb infection in a murine model. Together, our findings revealed a strategy by which Mtb hijacks host ferritin metabolism for its own intracellular survival. Therefore, this represents a potential target for host-directed therapy against tuberculosis.
Collapse
Affiliation(s)
- Youchao Dai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chuanzhi Zhu
- Laboratory of Molecular Biology, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Wei Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Kaisong Huang
- Zhuhai Center for Disease Control and Prevention, Zhuhai, China
| | - Xin Wang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chenyan Shi
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Dachuan Lin
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Huihua Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqian Liu
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
- Department of Infectious Disease, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Bin Peng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Yi Gao
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Stefan He Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - Carl G Feng
- Immunology and Host Defense Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
6
|
Nienaber A, Uyoga MA, Dolman-Macleod RC, Malan L. Iron Status and Supplementation during Tuberculosis. Microorganisms 2023; 11:microorganisms11030785. [PMID: 36985358 PMCID: PMC10055784 DOI: 10.3390/microorganisms11030785] [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: 02/14/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Tuberculosis (TB) is characterised by chronic non-resolving inflammation. The effects of the host immune and inflammatory response to reduce iron acquisition by the bacteria, together with other contributing factors, predispose TB patients to anaemia of infection and iron deficiency anaemia (IDA). The presence of anaemia in TB patients has been linked to poor clinical outcomes. However, due to the reliance of the bacteria on iron, the management of anaemia in TB is complicated, and anaemia of infection is likely to resolve with correct TB drug treatment. On the other hand, IDA may require iron supplementation. This review aims to describe iron metabolism in TB and how this contributes to the development of iron deficiency and anaemia. Additionally, we summarise the evidence on the association between iron status and clinical outcomes as well as the available preclinical and clinical trials on iron supplementation in TB.
Collapse
Affiliation(s)
- Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| | - Mary A Uyoga
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| | - Robin C Dolman-Macleod
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| |
Collapse
|
7
|
Immune cell interactions in tuberculosis. Cell 2022; 185:4682-4702. [PMID: 36493751 DOI: 10.1016/j.cell.2022.10.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 12/13/2022]
Abstract
Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.
Collapse
|
8
|
Cui Y, Gutierrez S, Ariai S, Öberg L, Thörn K, Gehrmann U, Cloonan SM, Naessens T, Olsson H. Non-heme iron overload impairs monocyte to macrophage differentiation via mitochondrial oxidative stress. Front Immunol 2022; 13:998059. [PMID: 36341326 PMCID: PMC9634638 DOI: 10.3389/fimmu.2022.998059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
Iron is a key element for systemic oxygen delivery and cellular energy metabolism. Thus regulation of systemic and local iron metabolism is key for maintaining energy homeostasis. Significant changes in iron levels due to malnutrition or hemorrhage, have been associated with several diseases such as hemochromatosis, liver cirrhosis and COPD. Macrophages are key cells in regulating iron levels in tissues as they sequester excess iron. How iron overload affects macrophage differentiation and function remains a subject of debate. Here we used an in vitro model of monocyte-to-macrophage differentiation to study the effect of iron overload on macrophage function. We found that providing excess iron as soluble ferric ammonium citrate (FAC) rather than as heme-iron complexes derived from stressed red blood cells (sRBC) interferes with macrophage differentiation and phagocytosis. Impaired macrophage differentiation coincided with increased expression of oxidative stress-related genes. Addition of FAC also led to increased levels of cellular and mitochondrial reactive oxygen species (ROS) and interfered with mitochondrial function and ATP generation. The effects of iron overload were reproduced by the mitochondrial ROS-inducer rotenone while treatment with the ROS-scavenger N-Acetylcysteine partially reversed FAC-induced effects. Finally, we found that iron-induced oxidative stress interfered with upregulation of M-CSFR and MAFB, two crucial determinants of macrophage differentiation and function. In summary, our findings suggest that high levels of non-heme iron interfere with macrophage differentiation by inducing mitochondrial oxidative stress. These findings might be important to consider in the context of diseases like chronic obstructive pulmonary disease (COPD) where both iron overload and defective macrophage function have been suggested to play a role in disease pathogenesis.
Collapse
Affiliation(s)
- Yue Cui
- Translational Science & Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- *Correspondence: Yue Cui, ; Saray Gutierrez,
| | - Saray Gutierrez
- Bioscience Cardiovascular, Early Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- *Correspondence: Yue Cui, ; Saray Gutierrez,
| | - Sheller Ariai
- Early Product Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lisa Öberg
- Translational Science & Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kristofer Thörn
- Translational Science & Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ulf Gehrmann
- Translational Science & Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, United States
- School of Medicine, Trinity Biomedical Sciences Institute and Tallaght University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Thomas Naessens
- Bioscience Cough & In vivo, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Henric Olsson
- Translational Science & Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
9
|
Kathamuthu GR, Rajamanickam A, Sridhar R, Baskaran D, Babu S. Strongyloidiasis stercoralis coinfection is associated with altered iron status biomarkers in tuberculous lymphadenitis. Front Immunol 2022; 13:999614. [PMID: 36341407 PMCID: PMC9632344 DOI: 10.3389/fimmu.2022.999614] [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: 07/21/2022] [Accepted: 10/07/2022] [Indexed: 11/26/2022] Open
Abstract
Soil-transmitted helminth [mainly Strongyloidiasis stercoralis (Ss)] and tuberculous lymphadenitis (TBL) coinfection in humans is a significant public health problem. We have previously shown that TBL+Ss+ coinfection significantly alters diverse cytokine, matrix metalloproteinase, and tissue inhibitors of metalloproteinase profiles. However, no data is available to understand the influence of Ss coinfection in TBL disease with respect to iron status biomarkers. Hence, we have studied the effect of Ss coinfection on the circulating levels of iron status (ferritin, transferrin [TF], apotransferrin [ApoT], hepcidin, hemopexin) biomarkers in TBL disease. Our results show that TBL+Ss+ and/or TBL+Ss- individuals are associated with significantly altered biochemical and hematological (red blood cell (RBC) counts, hemoglobin (Hb), hematocrit (HCT), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH) were decreased, and platelets were increased) parameters compared to TBL-Ss+ individuals. Our results also show that TBL+Ss+ coinfection is associated with diminished circulating levels of ferritin, ApoT, hepcidin, and hemopexin compared to TBL+Ss- individuals. TBL+Ss+ and TBL+Ss- groups are associated with altered iron status biomarkers (decreased ferritin [TBL+Ss+ alone] and increased TF, ApoT, hepcidin and hemopexin [TBL+Ss- alone]) compared to TBL-Ss+ group. The heat map expression profile and principal component analysis (PCA) analysis of iron status biomarkers were significantly altered in TBL+Ss+ compared to TBL+Ss- and/or TBL-Ss+ individuals. A significant correlation (positive/negative) was obtained among the biochemical and hematological parameters (white blood cells (WBC)/ferritin, TF, and hepcidin, mean corpuscular hemoglobin concentration (MCHC)/ferritin and hemopexin) with iron status biomarkers. Finally, receiver operating characteristic (ROC) analysis revealed that hemopexin was significantly associated with greater specificity and sensitivity in discriminating TBL+Ss+ and TBL+Ss- coinfected individuals. Thus, our data conclude that Ss coinfection is associated with altered iron status biomarkers indicating that coinfection might alter the host-Mtb interface and could influence the disease pathogenesis.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
- Indian Council of Medical Research-National Institute for Research in Tuberculosis (ICMR-NIRT), Chennai, India
- *Correspondence: Gokul Raj Kathamuthu,
| | - Anuradha Rajamanickam
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | | | - Dhanaraj Baskaran
- Indian Council of Medical Research-National Institute for Research in Tuberculosis (ICMR-NIRT), Chennai, India
| | - Subash Babu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
10
|
Fraschilla I, Evavold CL. Biting the hand that feeds: Metabolic determinants of cell fate during infection. Front Immunol 2022; 13:923024. [PMID: 36311735 PMCID: PMC9614662 DOI: 10.3389/fimmu.2022.923024] [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: 04/18/2022] [Accepted: 09/14/2022] [Indexed: 09/07/2024] Open
Abstract
Metabolic shifts can occur in cells of the innate immune system in response to microbial infection. Whether these metabolic shifts benefit host defense and propagation of an immune response appears to be context dependent. In an arms race, host-adapted microbes and mammalian cells vie for control of biosynthetic machinery, organelles, and metabolites. Herein, we discuss the intersection of host metabolism and cell-intrinsic immunity with implications for cell fate during infection. Sensation of microbial ligands in isolation results in host metabolic shifts that imbues normal innate immune function, such as cytokine secretion. However, living microbes have an arsenal of effectors and strategies to subvert cell-intrinsic immune responses by manipulating host metabolism. Consequently, host metabolism is monitored as an indicator of invasion or manipulation by a pathogen, primarily through the actions of guard proteins and inflammasome pathways. In this review, we frame initiation of cell-intrinsic immunity in the context of host metabolism to include a physiologic "Goldilocks zone" of allowable shifts with guard circuits monitoring wide perturbations away from this zone for the initiation of innate immune responses. Through comparison of studies with purified microbial ligands, dead microbes, and live pathogens we may begin to understand how shifts in metabolism determine the outcome of host-pathogen interactions.
Collapse
|
11
|
Lin Z, Yang X, Guan L, Qin L, Ding J, Zhou L. The link between ferroptosis and airway inflammatory diseases: A novel target for treatment. Front Mol Biosci 2022; 9:985571. [PMID: 36060261 PMCID: PMC9428508 DOI: 10.3389/fmolb.2022.985571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is an iron-dependent mode of cell death characterized by intracellular lipid peroxide accumulation and a redox reaction imbalance. Compared with other modes of cell death, ferroptosis has specific biological and morphological features. The iron-dependent lipid peroxidation accumulation is manifested explicitly in the abnormal metabolism of intracellular lipid oxides catalyzed by excessive iron ions with the production of many reactive oxygen species and over-oxidization of polyunsaturated fatty acids. Recent studies have shown that various diseases, which include intestinal diseases and cancer, are associated with ferroptosis, but few studies are related to airway inflammatory diseases. This review provides a comprehensive analysis of the primary damage mechanisms of ferroptosis and summarizes the relationship between ferroptosis and airway inflammatory diseases. In addition to common acute and chronic airway inflammatory diseases, we also focus on the progress of research on COVID-19 in relation to ferroptosis. New therapeutic approaches and current issues to be addressed in the treatment of inflammatory airway diseases using ferroptosis are further proposed.
Collapse
|
12
|
Iakobachvili N, Leon‐Icaza SA, Knoops K, Sachs N, Mazères S, Simeone R, Peixoto A, Bernard C, Murris‐Espin M, Mazières J, Cam K, Chalut C, Guilhot C, López‐Iglesias C, Ravelli RBG, Neyrolles O, Meunier E, Lugo‐Villarino G, Clevers H, Cougoule C, Peters P. Mycobacteria-host interactions in human bronchiolar airway organoids. Mol Microbiol 2022; 117:682-692. [PMID: 34605588 PMCID: PMC9298242 DOI: 10.1111/mmi.14824] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 02/04/2023]
Abstract
Respiratory infections remain a major global health concern. Tuberculosis is one of the top 10 causes of death worldwide, while infections with Non-Tuberculous Mycobacteria are rising globally. Recent advances in human tissue modeling offer a unique opportunity to grow different human "organs" in vitro, including the human airway, that faithfully recapitulates lung architecture and function. Here, we have explored the potential of human airway organoids (AOs) as a novel system in which to assess the very early steps of mycobacterial infection. We reveal that Mycobacterium tuberculosis (Mtb) and Mycobacterium abscessus (Mabs) mainly reside as extracellular bacteria and infect epithelial cells with very low efficiency. While the AO microenvironment was able to control, but not eliminate Mtb, Mabs thrives. We demonstrate that AOs responded to infection by modulating cytokine, antimicrobial peptide, and mucin gene expression. Given the importance of myeloid cells in mycobacterial infection, we co-cultured infected AOs with human monocyte-derived macrophages and found that these cells interact with the organoid epithelium. We conclude that adult stem cell (ASC)-derived AOs can be used to decipher very early events of mycobacteria infection in human settings thus offering new avenues for fundamental and therapeutic research.
Collapse
Affiliation(s)
- Nino Iakobachvili
- M4i Nanoscopy DivisionMaastricht UniversityMaastrichtThe Netherlands
| | - Stephen Adonai Leon‐Icaza
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Kèvin Knoops
- M4i Nanoscopy DivisionMaastricht UniversityMaastrichtThe Netherlands
| | - Norman Sachs
- Oncode Institute, Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences and University Medical CenterUtrechtThe Netherlands
| | - Serge Mazères
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Roxane Simeone
- Institut Pasteur, Unit for Integrated Mycobacterial PathogenomicsCNRS UMR3525ParisFrance
| | - Antonio Peixoto
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Célia Bernard
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | | | - Julien Mazières
- Service de PneumologieHôpital Larrey, CHU de ToulouseToulouseFrance
| | - Kaymeuang Cam
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Christian Chalut
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Christophe Guilhot
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | | | | | - Olivier Neyrolles
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
- International Associated Laboratory (LIA) CNRS “IM‐TB/HIV” (1167)ToulouseFrance
- International Associated Laboratory (LIA) CNRS “IM‐TB/HIV” (1167)Buenos AiresArgentina
| | - Etienne Meunier
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Geanncarlo Lugo‐Villarino
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
- International Associated Laboratory (LIA) CNRS “IM‐TB/HIV” (1167)ToulouseFrance
- International Associated Laboratory (LIA) CNRS “IM‐TB/HIV” (1167)Buenos AiresArgentina
| | - Hans Clevers
- Oncode Institute, Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences and University Medical CenterUtrechtThe Netherlands
| | - Céline Cougoule
- Institut de Pharmacologie et Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
- International Associated Laboratory (LIA) CNRS “IM‐TB/HIV” (1167)ToulouseFrance
- International Associated Laboratory (LIA) CNRS “IM‐TB/HIV” (1167)Buenos AiresArgentina
| | - Peter J. Peters
- M4i Nanoscopy DivisionMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
13
|
Tasinov O, Kiselova-Kaneva Y, Ivanova D, Pasheva M, Vankova D, Ivanova D. Ferrum phosphoricum D12 Treatment Affects J774A.1 Cell Proliferation, Transcription Levels of Iron Metabolism, Antioxidant Defense, and Inflammation-related Genes. HOMEOPATHY 2021; 111:113-120. [PMID: 34634834 DOI: 10.1055/s-0041-1731312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Ferrum phosphoricum (FP) is prescribed as a homeopathic remedy to treat the early stages of fever and inflammation in cases of colds or flu, muscle fatigue and anemia. We aimed to analyze the molecular mechanisms of action of FP D12 on cell proliferation and mRNA expression of iron metabolism, antioxidant defense and inflammation-related genes in mouse J774A.1 macrophages. METHODS Cell proliferation was examined using the MTT test. RT-qPCR analyses were performed to estimate gene expression changes. Relative gene expression levels were calculated using the 2-ΔΔCt method. The effect of treatment using FP D12 tablets was compared with that using placebo tablets (PT). RESULTS FP D12 in low concentrations (0.0125 mg/mL to 0.025 mg/mL) significantly stimulated proliferation of J774A.1 cells by up to 11% (p < 0.01) versus control untreated cells and by up to 40% (p < 0.01) versus PT-treated cells in the respective concentration. FP D12 versus PT induced a significant increase in mRNA expression of ferritin light chain (Ftl1) (by 8-fold, p < 0.01), β-2-microglobulin (B2m) (by 2.5-fold, p < 0.05) and iron-responsive element binding protein 2 (Ireb2) (by 4-fold, p < 0.05), and induced a slight decrease in myosin IE (Myo1e) mRNA expression levels (by 0.4-fold, p < 0.01) in macrophages. A highly significant (r2 = 0.99, p < 0.05) correlation was observed between Ireb2 and B2m transcription levels. Significant stimulation of antioxidant enzyme Gpx-1 (by 1.27-fold, p < 0.01) in cells by 0.025 mg/mL FP D12, but a slight decrease (by 0.12-fold, p < 0.05) in 0.0125 mg/mL-treated cells, was observed. A significant increase in the gene expression of IL-1β (by 3.5-fold, р < 0.05) in macrophages was also detected. CONCLUSION Ferrum phosphoricum in D12 dilution potentially exhibits iron retention, antioxidant and immunomodulation activities, possibly by modulating transcription levels of related genes in non-stimulated mouse macrophages.
Collapse
Affiliation(s)
- Oskan Tasinov
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University-Varna, Varna, Bulgaria
| | - Yoana Kiselova-Kaneva
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University-Varna, Varna, Bulgaria
| | - Desislava Ivanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University-Varna, Varna, Bulgaria
| | - Milena Pasheva
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University-Varna, Varna, Bulgaria
| | - Deyana Vankova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University-Varna, Varna, Bulgaria
| | - Diana Ivanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University-Varna, Varna, Bulgaria
| |
Collapse
|
14
|
Iron homeostasis during anemia of inflammation: a prospective study in patients with tuberculosis. Blood 2021; 138:1293-1303. [PMID: 33876222 DOI: 10.1182/blood.2020010562] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/02/2021] [Indexed: 11/20/2022] Open
Abstract
Anemia of inflammation is a hallmark of tuberculosis. Factors controlling iron metabolism during anemia of inflammation and its resolution are uncertain. Whether iron supplements should be given during anti-tuberculosis treatment to support Hb recovery is unclear. Before and during treatment of tuberculosis, we assessed iron kinetics, and changes in inflammation and iron metabolism indices. In a 26-wk prospective study, Tanzanian adults with tuberculosis (n=18) were studied before treatment and then every two weeks during treatment; oral and intravenous iron tracers were administered before treatment, after intensive phase (8/12 wk) and complete treatment (24 wk); no iron supplements were given. Before treatment, hepcidin and erythroferrone (ERFE) were greatly elevated, erythrocyte iron utilization was high (~80%) and iron absorption was negligible (<1%). During treatment, hepcidin and IL-6 decreased ~70% after only 2 wk (p<0.001); in contrast, ERFE did not significantly decrease until 8 wk (p<0.01). ERFE and IL-6 were the main opposing determinants of hepcidin (p<0.05) and greater ERFE was associated with reticulocytosis and hemoglobin (Hb) repletion (p<0.01). Dilution of baseline tracer concentration was 2.6-fold higher during intensive phase treatment (p<0.01) indicating enhanced erythropoiesis. After treatment completion, iron absorption increased ~20-fold (p<0.001); Hb increased ~25% (p<0.001). In tuberculosis-associated anemia of inflammation, our findings suggest elevated ERFE is unable to suppress hepcidin and iron absorption is negligible. During treatment, as inflammation resolves, ERFE may remain elevated, contributing to hepcidin suppression and Hb repletion. Iron is well-absorbed only after tuberculosis treatment and supplementation should be reserved for patients remaining anemic after treatment. (ClinicalTrials.gov Identifier:NCT02176772).
Collapse
|
15
|
Cahill C, O’Connell F, Gogan KM, Cox DJ, Basdeo SA, O’Sullivan J, Gordon SV, Keane J, Phelan JJ. The Iron Chelator Desferrioxamine Increases the Efficacy of Bedaquiline in Primary Human Macrophages Infected with BCG. Int J Mol Sci 2021; 22:ijms22062938. [PMID: 33805837 PMCID: PMC8001338 DOI: 10.3390/ijms22062938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022] Open
Abstract
For over 50 years, patients with drug-sensitive and drug-resistant tuberculosis have undergone long, arduous, and complex treatment processes with several antimicrobials. With the prevalence of drug-resistant strains on the rise and new therapies for tuberculosis urgently required, we assessed whether manipulating iron levels in macrophages infected with mycobacteria offered some insight into improving current antimicrobials that are used to treat drug-resistant tuberculosis. We investigated if the iron chelator, desferrioxamine, can support the function of human macrophages treated with an array of second-line antimicrobials, including moxifloxacin, bedaquiline, amikacin, clofazimine, linezolid and cycloserine. Primary human monocyte-derived macrophages were infected with Bacillus Calmette-Guérin (BCG), which is pyrazinamide-resistant, and concomitantly treated for 5 days with desferrioxamine in combination with each one of the second-line tuberculosis antimicrobials. Our data indicate that desferrioxamine used as an adjunctive treatment to bedaquiline significantly reduced the bacterial load in human macrophages infected with BCG. Our findings also reveal a link between enhanced bactericidal activity and increases in specific cytokines, as the addition of desferrioxamine increased levels of IFN-γ, IL-6, and IL-1β in BCG-infected human monocyte-derived macrophages (hMDMs) treated with bedaquiline. These results provide insight, and an in vitro proof-of-concept, that iron chelators may prove an effective adjunctive therapy in combination with current tuberculosis antimicrobials.
Collapse
Affiliation(s)
- Christina Cahill
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (C.C.); (K.M.G.); (D.J.C.); (S.A.B.); (J.K.)
| | - Fiona O’Connell
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (F.O.); (J.O.)
| | - Karl M. Gogan
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (C.C.); (K.M.G.); (D.J.C.); (S.A.B.); (J.K.)
| | - Donal J. Cox
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (C.C.); (K.M.G.); (D.J.C.); (S.A.B.); (J.K.)
| | - Sharee A. Basdeo
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (C.C.); (K.M.G.); (D.J.C.); (S.A.B.); (J.K.)
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (F.O.); (J.O.)
| | - Stephen V. Gordon
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, 4 Dublin, Ireland;
| | - Joseph Keane
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (C.C.); (K.M.G.); (D.J.C.); (S.A.B.); (J.K.)
| | - James J. Phelan
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, 8 Dublin, Ireland; (C.C.); (K.M.G.); (D.J.C.); (S.A.B.); (J.K.)
- Correspondence: ; Tel.: +353-18963265
| |
Collapse
|
16
|
Shao M, Wu F, Zhang J, Dong J, Zhang H, Liu X, Liang S, Wu J, Zhang L, Zhang C, Zhang W. Screening of potential biomarkers for distinguishing between latent and active tuberculosis in children using bioinformatics analysis. Medicine (Baltimore) 2021; 100:e23207. [PMID: 33592820 PMCID: PMC7870233 DOI: 10.1097/md.0000000000023207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 10/19/2020] [Indexed: 01/05/2023] Open
Abstract
Tuberculosis (TB) is one of the leading causes of childhood morbidity and death globally. Lack of rapid, effective non-sputum diagnosis and prediction methods for TB in children are some of the challenges currently faced. In recent years, blood transcriptional profiling has provided a fresh perspective on the diagnosis and predicting the progression of tuberculosis. Meanwhile, combined with bioinformatics analysis can help to identify the differentially expressed genes (DEGs) and functional pathways involved in the different clinical stages of TB. Therefore, this study investigated potential diagnostic markers for use in distinguishing between latent tuberculosis infection (LTBI) and active TB using children's blood transcriptome data.From the Gene Expression Omnibus database, we downloaded two gene expression profile datasets (GSE39939 and GSE39940) of whole blood-derived RNA sequencing samples, reflecting transcriptional signatures between latent and active tuberculosis in children. GEO2R tool was used to screen for DEGs in LTBI and active TB in children. Database for Annotation, Visualization and Integrated Discovery tools were used to perform Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analysis. STRING and Cytoscape analyzed the protein-protein interaction network and the top 15 hub genes respectively. Receiver operating characteristics curve was used to estimate the diagnostic value of the hub genes.A total of 265 DEGs were identified, including 79 upregulated and 186 downregulated DEGs. Further, 15 core genes were picked and enrichment analysis revealed that they were highly correlated with neutrophil activation and degranulation, neutrophil-mediated immunity and in defense response. Among them TLR2, FPR2, MMP9, MPO, CEACAM8, ELANE, FCGR1A, SELP, ARG1, GNG10, HP, LCN2, LTF, ADCY3 had significant discriminatory power between LTBI and active TB, with area under the curves of 0.84, 0.84, 0.84, 0.80, 0.87, 0.78, 0.88, 0.84, 0.86, 0.82, 0.85, 0.85, 0.79, and 0.88 respectively.Our research provided several genes with high potential to be candidate gene markers for developing non-sputum diagnostic tools for childhood Tuberculosis.
Collapse
Affiliation(s)
- Meng Shao
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| | - Fang Wu
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| | - Jie Zhang
- The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, XinJiang, PR China
| | - Jiangtao Dong
- The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, XinJiang, PR China
| | - Hui Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| | - Xiaoling Liu
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| | - Su Liang
- The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, XinJiang, PR China
| | - Jiangdong Wu
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| | - Le Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| | - Chunjun Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| | - Wanjiang Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/The Key Laboratory of Xinjiang Endemic and Ethnic Diseases
| |
Collapse
|
17
|
Holbein BE, Ang MTC, Allan DS, Chen W, Lehmann C. Iron-withdrawing anti-infectives for new host-directed therapies based on iron dependence, the Achilles' heel of antibiotic-resistant microbes. ENVIRONMENTAL CHEMISTRY LETTERS 2021; 19:2789-2808. [PMID: 33907538 PMCID: PMC8062846 DOI: 10.1007/s10311-021-01242-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/13/2021] [Indexed: 05/02/2023]
Abstract
The iron dependence of antibiotic-resistant microbes represents an Achilles' heel that can be exploited broadly. The growing global problem of antibiotic resistance of microbial pathogens wherein microbes become resistant to the very antibiotics used against them during infection is linked not only to our health uses but also to agribusiness practices and the changing environment. Here we review mechanisms of microbial iron acquisition and host iron withdrawal defense, and the influence of iron withdrawal on the antimicrobial activity of antibiotics. Antibiotic-resistant microbes are unaltered in their iron requirements, but iron withdrawal from microbes enhances the activities of various antibiotics and importantly suppresses outgrowth of antibiotic-exposed resistant microbial survivors. Of the three therapeutic approaches available to exploit microbial iron susceptibility, including (1) use of gallium as a non-functional iron analogue, (2) Trojan horse conjugates of microbial siderophores carrying antibiotics, and (3) new generation iron chelators, purposely designed as anti-microbials, the latter offers various advantages. For instance, these novel anti-microbial chelators overcome the limitations of conventional clinically-used hematological chelators which display host toxicity and are not useful antimicrobials. 3-Hydroxypyridin-4-one-containing polymeric chelators appear to have the highest potential. DIBI (developmental code name) is a well-developed lead candidate, being a low molecular weight, water-soluble copolymer with enhanced iron binding characteristics, strong anti-microbial and anti-inflammatory activities, low toxicity for animals and demonstrated freedom from microbial resistance development. DIBI has been shown to enhance antibiotic efficacy for antibiotic-resistant microbes during infection, and it also prevents recovery growth and resistance development during microbe exposure to various antibiotics. Because DIBI bolsters innate iron withdrawal defenses of the infected host, it has potential to provide a host-directed anti-infective therapy.
Collapse
Affiliation(s)
- Bruce E. Holbein
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
- Department of Microbiology and Immunology, Dalhousie University, 5859 College St., Halifax, NS B3H 1X5 Canada
| | - M. Trisha C. Ang
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
| | - David S. Allan
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6 Canada
| | - Christian Lehmann
- Department of Microbiology and Immunology, Dalhousie University, 5859 College St., Halifax, NS B3H 1X5 Canada
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
18
|
Jha V, Pal R, Kumar D, Mukhopadhyay S. ESAT-6 Protein of Mycobacterium tuberculosis Increases Holotransferrin-Mediated Iron Uptake in Macrophages by Downregulating Surface Hemochromatosis Protein HFE. THE JOURNAL OF IMMUNOLOGY 2020; 205:3095-3106. [PMID: 33148716 DOI: 10.4049/jimmunol.1801357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 10/01/2020] [Indexed: 11/19/2022]
Abstract
Iron is an essential element for Mycobacterium tuberculosis; it has at least 40 enzymes that require iron as a cofactor. Accessibility of iron at the phagosomal surface inside macrophage is crucial for survival and virulence of M. tuberculosis ESAT-6, a 6-kDa-secreted protein of region of difference 1, is known to play a crucial role in virulence and pathogenesis of M. tuberculosis In our earlier study, we demonstrated that ESAT-6 protein interacts with β-2-microglobulin (β2M) and affects class I Ag presentation through sequestration of β2M inside endoplasmic reticulum, which contributes toward inhibition of MHC class I:β2M:peptide complex formation. The 6 aa at C-terminal region of ESAT-6 are essential for ESAT6:β2M interaction. β2M is essential for proper folding of HFE, CD1, and MHC class I and their surface expression. It is known that M. tuberculosis recruit holotransferrin at the surface of the phagosome. But the upstream mechanism by which it modulates holotransferrin-mediated iron uptake at the surface of macrophage is not well understood. In the current study, we report that interaction of the ESAT-6 protein with β2M causes downregulation of surface HFE, a protein regulating iron homeostasis via interacting with transferrin receptor 1 (TFR1). We found that ESAT-6:β2M interaction leads to sequestration of HFE in endoplasmic reticulum, causing poorer surface expression of HFE and HFE:TFR1 complex (nonfunctional TFR1) in peritoneal macrophages from C57BL/6 mice, resulting in increased holotransferrin-mediated iron uptake in these macrophages. These studies suggest that M. tuberculosis probably targets the ESAT-6 protein to increase iron uptake.
Collapse
Affiliation(s)
- Vishwanath Jha
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India; and
| | - Ravi Pal
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India; and
| | - Dhiraj Kumar
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, Telangana, India;
| |
Collapse
|
19
|
Nienaber A, Baumgartner J, Dolman RC, Ozturk M, Zandberg L, Hayford FEA, Brombacher F, Blaauw R, Parihar SP, Smuts CM, Malan L. Omega-3 Fatty Acid and Iron Supplementation Alone, but Not in Combination, Lower Inflammation and Anemia of Infection in Mycobacterium tuberculosis-Infected Mice. Nutrients 2020; 12:E2897. [PMID: 32971969 PMCID: PMC7551947 DOI: 10.3390/nu12092897] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Progressive inflammation and anemia are common in tuberculosis (TB) and linked to poor clinical outcomes. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have inflammation-resolving properties, whereas iron supplementation in TB may have limited efficacy and enhance bacterial growth. We investigated effects of iron and EPA/DHA supplementation, alone and in combination, on inflammation, anemia, iron status markers and clinical outcomes in Mycobacterium tuberculosis-infected C3HeB/FeJ mice. One week post-infection, mice received the AIN-93 diet without (control) or with supplemental iron (Fe), EPA/DHA, or Fe+EPA/DHA for 3 weeks. Mice supplemented with Fe or EPA/DHA had lower soluble transferrin receptor, ferritin and hepcidin than controls, but these effects were attenuated in Fe+EPA/DHA mice. EPA/DHA increased inflammation-resolving lipid mediators and lowered lung IL-1α, IFN-γ, plasma IL-1β, and TNF-α. Fe lowered lung IL-1α, IL-1β, plasma IL-1β, TNF-α, and IL-6. However, the cytokine-lowering effects in the lungs were attenuated with Fe+EPA/DHA. Mice supplemented with EPA/DHA had lower lung bacterial loads than controls, but this effect was attenuated in Fe+EPA/DHA mice. Thus, individually, post-infection EPA/DHA and iron supplementation lowered systemic and lung inflammation and mitigated anemia of infection in TB, but not when combined. EPA/DHA also enhanced bactericidal effects and could support inflammation resolution and management of anemia.
Collapse
Affiliation(s)
- Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Jeannine Baumgartner
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
- Laboratory of Human Nutrition, ETH, 8092 Zurich, Switzerland
| | - Robin C. Dolman
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town 7925, South Africa; (M.O.); (F.B.); (S.P.P.)
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town 7925, South Africa
| | - Lizelle Zandberg
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Frank E. A. Hayford
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
- Department of Nutrition and Dietetics, School of biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra Box KB143, Ghana
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town 7925, South Africa; (M.O.); (F.B.); (S.P.P.)
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town 7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa
| | - Renee Blaauw
- Division of Human Nutrition, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa;
| | - Suraj P. Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town 7925, South Africa; (M.O.); (F.B.); (S.P.P.)
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town 7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa
- Division of Medical Microbiology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Cornelius M. Smuts
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| |
Collapse
|
20
|
Phelan JJ, McQuaid K, Kenny C, Gogan KM, Cox DJ, Basdeo SA, O’Leary S, Tazoll SC, Ó Maoldomhnaigh C, O’Sullivan MP, O’Neill LA, O’Sullivan MJ, Keane J. Desferrioxamine Supports Metabolic Function in Primary Human Macrophages Infected With Mycobacterium tuberculosis. Front Immunol 2020; 11:836. [PMID: 32477344 PMCID: PMC7237728 DOI: 10.3389/fimmu.2020.00836] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/14/2020] [Indexed: 02/02/2023] Open
Abstract
Tuberculosis is the single biggest infectious killer in the world and presents a major global health challenge. Antimicrobial therapy requires many months of multiple drugs and incidences of drug resistant tuberculosis continues to rise. Consequently, research is now focused on the development of therapies to support the function of infected immune cells. HIF1α-mediated induction of aerobic glycolysis is integral to the host macrophage response during infection with Mtb, as this promotes bacillary clearance. Some iron chelators have been shown to modulate cellular metabolism through the regulation of HIF1α. We examined if the iron chelator, desferrioxamine (DFX), could support the function of primary human macrophages infected with Mtb. Using RT-PCR, we found that DFX promoted the expression of key glycolytic enzymes in Mtb-infected primary human MDMs and human alveolar macrophages. Using Seahorse technology, we demonstrate that DFX enhances glycolytic metabolism in Mtb-stimulated human MDMs, while helping to enhance glycolysis during mitochondrial distress. Furthermore, the effect of DFX on glycolysis was not limited to Mtb infection as DFX also boosted glycolytic metabolism in uninfected and LPS-stimulated cells. DFX also supports innate immune function by inducing IL1β production in human macrophages during early infection with Mtb and upon stimulation with LPS. Moreover, using hypoxia, Western blot and ChIP-qPCR analyses, we show that DFX modulates IL1β levels in these cells in a HIF1α-mediated manner. Collectively, our data suggests that DFX exhibits potential to enhance immunometabolic responses and augment host immune function during early Mtb infection, in selected clinical settings.
Collapse
Affiliation(s)
- James Joseph Phelan
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate McQuaid
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Colin Kenny
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
| | - Karl Michael Gogan
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Dónal J. Cox
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Sharee Ann Basdeo
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Seónadh O’Leary
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Simone Christa Tazoll
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Cilian Ó Maoldomhnaigh
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Mary P. O’Sullivan
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Luke A. O’Neill
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Maureen J. O’Sullivan
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
| | - Joseph Keane
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Ferroptosis as an emerging target in inflammatory diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 155:20-28. [PMID: 32311424 DOI: 10.1016/j.pbiomolbio.2020.04.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/08/2020] [Accepted: 04/12/2020] [Indexed: 12/15/2022]
Abstract
Cell survival or death is one critical issue in inflammatory responses. Ferroptosis, which is characterized by iron-dependent lethal lipid peroxidation, has been found to participate in the development of cancers, degenerative brain diseases and ischemia-reperfusion injuries. Incorporation of polyunsaturated fatty acids (PUFAs) into cellular membranes represents a vulnerability to invasion of microbials and sterile stimuli. In addition, the competition for iron in the battle between microbials and host cells underlies infection development. Although host cells have been equipped with complex antioxidant systems to combat lethal accumulation of lipid peroxidation, emerging evidence suggests several pathogens may target PUFAs in the cell membrane, and manipulate ferroptosis as a way for pathogen propagation. Moreover, ferroptosis takes part in the progression of sterile inflammations, such as cigarette smoke-induced chronic obstructive pulmonary disease, stroke and ischemia-reperfusion injuries. As iron-dependent oxidative stress and lipid peroxidation are common features for ferroptosis and inflammatory diseases, underlying mechanisms linking such pathological conditions will be discussed in this review. Progress in the research of ferroptosis may shed more light on the etiology and treatment of inflammatory diseases.
Collapse
|
22
|
Moreira AC, Mesquita G, Gomes MS. Ferritin: An Inflammatory Player Keeping Iron at the Core of Pathogen-Host Interactions. Microorganisms 2020; 8:microorganisms8040589. [PMID: 32325688 PMCID: PMC7232436 DOI: 10.3390/microorganisms8040589] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
Iron is an essential element for virtually all cell types due to its role in energy metabolism, nucleic acid synthesis and cell proliferation. Nevertheless, if free, iron induces cellular and organ damage through the formation of free radicals. Thus, iron levels must be firmly controlled. During infection, both host and microbe need to access iron and avoid its toxicity. Alterations in serum and cellular iron have been reported as important markers of pathology. In this regard, ferritin, first discovered as an iron storage protein, has emerged as a biomarker not only in iron-related disorders but also in inflammatory diseases, or diseases in which inflammation has a central role such as cancer, neurodegeneration or infection. The basic research on ferritin identification and functions, as well as its role in diseases with an inflammatory component and its potential as a target in host-directed therapies, are the main considerations of this review.
Collapse
Affiliation(s)
- Ana C. Moreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.); (M.S.G.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Correspondence:
| | - Gonçalo Mesquita
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.); (M.S.G.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.); (M.S.G.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
23
|
Xu Z, Wang P, Wang H, Yu ZH, Au-Yeung HY, Hirayama T, Sun H, Yan A. Zinc excess increases cellular demand for iron and decreases tolerance to copper in Escherichia coli. J Biol Chem 2019; 294:16978-16991. [PMID: 31586033 DOI: 10.1074/jbc.ra119.010023] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Transition metals serve as an important class of micronutrients that are indispensable for bacterial physiology but are cytotoxic when they are in excess. Bacteria have developed exquisite homeostatic systems to control the uptake, storage, and efflux of each of biological metals and maintain a thermodynamically balanced metal quota. However, whether the pathways that control the homeostasis of different biological metals cross-talk and render cross-resistance or sensitivity in the host-pathogen interface remains largely unknown. Here, we report that zinc (Zn) excess perturbs iron (Fe) and copper (Cu) homeostasis in Escherichia coli, resulting in increased Fe and decreased Cu levels in the cell. Gene expression analysis revealed that Zn excess transiently up-regulates Fe-uptake genes and down-regulates Fe-storage genes and thereby increases the cellular Fe quota. In vitro and in vivo protein-DNA binding assays revealed that the elevated intracellular Fe poisons the primary Cu detoxification transcription regulator CueR, resulting in dysregulation of its target genes copA and cueO and activation of the secondary Cu detoxification system CusSR-cusCFBA Supplementation with the Fe chelator 2,2'-dipyridyl (DIP) or with the reducing agent GSH abolished the induction of cusCFBA during Zn excess. Consistent with the importance of this metal homeostatic network in cell physiology, combined metal treatment, including simultaneously overloading cells with both Zn (0.25 mm) and Cu (0.25 mm) and sequestering Fe with DIP (50 μm), substantially inhibited E. coli growth. These results advance our understanding of bacterial metallobiology and may inform the development of metal-based antimicrobial regimens to manage infectious diseases.
Collapse
Affiliation(s)
- Zeling Xu
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Pengchao Wang
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Haibo Wang
- Department of Chemistry, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zuo Hang Yu
- Department of Chemistry, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Ho Yu Au-Yeung
- Department of Chemistry, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4, Dairaku-nishi, Gifu, 501-1196, Japan
| | - Hongzhe Sun
- Department of Chemistry, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Aixin Yan
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
24
|
Kolloli A, Singh P, Rodriguez GM, Subbian S. Effect of Iron Supplementation on the Outcome of Non-Progressive Pulmonary Mycobacterium tuberculosis Infection. J Clin Med 2019; 8:jcm8081155. [PMID: 31382404 PMCID: PMC6722820 DOI: 10.3390/jcm8081155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
The human response to Mycobacterium tuberculosis (Mtb) infection is affected by the availability of iron (Fe), which is necessary for proper immune cell function and is essential for the growth and virulence of bacteria. Increase in host Fe levels promotes Mtb growth and tuberculosis (TB) pathogenesis, while Fe-supplementation to latently infected, asymptomatic individuals is a significant risk factor for disease reactivation. However, the effect of Fe-supplementation on the host immunity during latent Mtb infection remains unclear, due partly to the paucity in availability of animal models that recapitulate key pathophysiological features seen in humans. We have demonstrated that rabbits can develop non-progressive latency similar to infected humans. In this study, using this model we have evaluated the effect of Fe-supplementation on the bacterial growth, disease pathology, and immune response. Systemic and lung Fe parameters, gene expression profile, lung bacterial burden, and disease pathology were determined in the Mtb-infected/Fe- or placebo-supplemented rabbits. Results show that Fe-supplementation to Mtb-infected rabbits did not significantly change the hematocrit and Hb levels, although it elevated total Fe in the lungs. Expression of selected host iron- and immune-response genes in the blood and lungs was perturbed in Mtb-infected/Fe-supplemented rabbits. Iron-supplementation during acute or chronic stages of Mtb infection did not significantly affect the bacterial burden or disease pathology in the lungs. Data presented in this study is of significant relevance for current public health policies on Fe-supplementation therapy given to anemic patients with latent Mtb infection.
Collapse
Affiliation(s)
- Afsal Kolloli
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Pooja Singh
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - G Marcela Rodriguez
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Selvakumar Subbian
- The Public Health Research Institute Center of New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| |
Collapse
|
25
|
Pulmonary Iron Limitation Induced by Exogenous Type I IFN Protects Mice from Cryptococcus gattii Independently of T Cells. mBio 2019; 10:mBio.00799-19. [PMID: 31213551 PMCID: PMC6581853 DOI: 10.1128/mbio.00799-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cryptococcus neoformans and Cryptococcus gattii cause fatal infection in immunodeficient and immunocompetent individuals. While these fungi are sibling species, C. gattii infects very few AIDS patients, while C. neoformans infection is an AIDS-defining illness, suggesting that the host response to HIV selects C. neoformans over C. gattii. We used a viral mimic molecule (pICLC) to stimulate the immune response, and pICLC treatment improved mouse outcomes from both species. pICLC-induced action against C. neoformans was due to activation of well-defined immune pathways known to deter C. neoformans, whereas these immune pathways were dispensable for pICLC treatment of C. gattii. Since these immune pathways are eventually destroyed by HIV/AIDS, our data help explain why the antiviral immune response in AIDS patients is unable to control C. neoformans infection but is protective against C. gattii. Furthermore, pICLC induced tighter control of iron in the lungs of mice, which inhibited C. gattii, thus suggesting an entirely new mode of nutritional immunity activated by viral signals. Cryptococcus neoformans causes deadly mycosis primarily in AIDS patients, whereas Cryptococcus gattii infects mostly non-HIV patients, even in regions with high burdens of HIV/AIDS and an established environmental presence of C. gattii. As HIV induces type I IFN (t1IFN), we hypothesized that t1IFN would differentially affect the outcome of C. neoformans and C. gattii infections. Exogenous t1IFN induction using stabilized poly(I·C) (pICLC) improved murine outcomes in either cryptococcal infection. In C. neoformans-infected mice, pICLC activity was associated with C. neoformans containment and classical Th1 immunity. In contrast, pICLC activity against C. gattii did not require any immune factors previously associated with C. neoformans immunity: T, B, and NK cells, IFN-γ, and macrophages were all dispensable. Interestingly, C. gattii pICLC activity depended on β-2-microglobulin, which impacts iron levels among other functions. Iron supplementation reversed pICLC activity, suggesting C. gattii pICLC activity requires iron limitation. Also, pICLC induced a set of iron control proteins, some of which were directly inhibitory to cryptococcus in vitro, suggesting t1IFN regulates iron availability in the pulmonary air space fluids. Thus, exogenous induction of t1IFN significantly improves the outcome of murine infection by C. gattii and C. neoformans but by distinct mechanisms; the C. gattii effect was mediated by iron limitation, while the effect on C. neoformans infection was through induction of classical T-cell-dependent immunity. Together this difference in types of T-cell-dependent t1IFN immunity for different Cryptococcus species suggests a possible mechanism by which HIV infection may select against C. gattii but not C. neoformans.
Collapse
|
26
|
Iron Supplementation Therapy, A Friend and Foe of Mycobacterial Infections? Pharmaceuticals (Basel) 2019; 12:ph12020075. [PMID: 31108902 PMCID: PMC6630247 DOI: 10.3390/ph12020075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/21/2022] Open
Abstract
Iron is an essential element that is required for oxygen transfer, redox, and metabolic activities in mammals and bacteria. Mycobacteria, some of the most prevalent infectious agents in the world, require iron as growth factor. Mycobacterial-infected hosts set up a series of defense mechanisms, including systemic iron restriction and cellular iron distribution, whereas mycobacteria have developed sophisticated strategies to acquire iron from their hosts and to protect themselves from iron’s harmful effects. Therefore, it is assumed that host iron and iron-binding proteins, and natural or synthetic chelators would be keys targets to inhibit mycobacterial proliferation and may have a therapeutic potential. Beyond this hypothesis, recent evidence indicates a host protective effect of iron against mycobacterial infections likely through promoting remodeled immune response. In this review, we discuss experimental procedures and clinical observations that highlight the role of the immune response against mycobacteria under various iron availability conditions. In addition, we discuss the clinical relevance of our knowledge regarding host susceptibility to mycobacteria in the context of iron availability and suggest future directions for research on the relationship between host iron and the immune response and the use of iron as a therapeutic agent.
Collapse
|
27
|
Pal R, Hameed S, Kumar P, Singh S, Fatima Z. Understanding lipidomic basis of iron limitation induced chemosensitization of drug-resistant Mycobacterium tuberculosis. 3 Biotech 2019; 9:122. [PMID: 30863701 PMCID: PMC6401079 DOI: 10.1007/s13205-019-1645-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023] Open
Abstract
Under limited micronutrients condition, Mycobacterium tuberculosis (MTB) has to struggle for acquisition of the limited micronutrients available in the host. One such crucial micronutrient that MTB requires for the growth and sustenance is iron. The present study aimed to sequester the iron supply of MTB to control drug resistance in MTB. We found that iron restriction renders hypersensitivity to multidrug-resistant MTB strains against first-line anti-TB drugs. To decipher the effect of iron restriction on possible mechanisms of chemosensitization and altered cellular circuitry governing drug resistance and virulence of MTB, we explored MTB cellular architecture. We could identify non-intact cell envelope, tampered MTB morphology and diminished mycolic acid under iron restricted MDR-MTB cells. Deeper exploration unraveled altered lipidome profile observed through conventional TLC and advanced mass spectrometry-based LC-ESI-MS techniques. Lipidome analysis not only depicted profound alterations of various lipid classes which are crucial for pathogenecity but also exposed leads such as indispensability of iron to sustain metabolic, genotoxic and oxidative stresses. Furthermore, iron deprivation led to inhibited biofilm formation and capacity of MTB to adhere buccal epithelial cells. Lastly, we demonstrated enhanced survival of Mycobacterium-infected Caenorhabditis elegans model under iron limitation. The present study offers evidence and proposes alteration of lipidome profile and affected virulence traits upon iron chelation. Taken together, iron deprivation could be a potential strategy to rescue MDR and enhance the effectiveness of existing anti-TB drugs.
Collapse
Affiliation(s)
- Rahul Pal
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram, 122413 India
| | - Saif Hameed
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram, 122413 India
| | - Parveen Kumar
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Sarman Singh
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Zeeshan Fatima
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram, 122413 India
| |
Collapse
|
28
|
Richard KL, Kelley BR, Johnson JG. Heme Uptake and Utilization by Gram-Negative Bacterial Pathogens. Front Cell Infect Microbiol 2019; 9:81. [PMID: 30984629 PMCID: PMC6449446 DOI: 10.3389/fcimb.2019.00081] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Iron is a transition metal utilized by nearly all forms of life for essential cellular processes, such as DNA synthesis and cellular respiration. During infection by bacterial pathogens, the host utilizes various strategies to sequester iron in a process termed, nutritional immunity. To circumvent these defenses, Gram-negative pathogens have evolved numerous mechanisms to obtain iron from heme. In this review we outline the systems that exist in several Gram-negative pathogens that are associated with heme transport and utilization, beginning with hemolysis and concluding with heme degradation. In addition, Gram-negative pathogens must also closely regulate the intracellular concentrations of iron and heme, since high levels of iron can lead to the generation of toxic reactive oxygen species. As such, we also provide several examples of regulatory pathways that control heme utilization, showing that co-regulation with other cellular processes is complex and often not completely understood.
Collapse
Affiliation(s)
- Kaylie L Richard
- Department of Microbiology, University of Tennessee, Knoxville, TN, United States
| | - Brittni R Kelley
- Department of Microbiology, University of Tennessee, Knoxville, TN, United States
| | - Jeremiah G Johnson
- Department of Microbiology, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
29
|
Middha S, Yaeger R, Shia J, Stadler ZK, King S, Guercio S, Paroder V, Bates DDB, Rana S, Diaz LA, Saltz L, Segal N, Ladanyi M, Zehir A, Hechtman JF. Majority of B2M-Mutant and -Deficient Colorectal Carcinomas Achieve Clinical Benefit From Immune Checkpoint Inhibitor Therapy and Are Microsatellite Instability-High. JCO Precis Oncol 2019; 3. [PMID: 31008436 DOI: 10.1200/po.18.00321] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Microsatellite instability-high (MSI-H) colorectal carcinomas (CRCs) show high rates of response to immune checkpoint inhibitors (IOs). B2M mutations and protein loss have been proposed as causes of resistance to IOs, yet they are enriched in MSI-H CRC. We aimed to characterize B2M-mutant, IO-naive CRC. PATIENTS AND METHODS All CRCs with results for Memorial Sloan Kettering Integrated Mutation Profiling of Actionable Cancer Targets, a next-generation sequencing assay that interrogates > 400 genes for mutations as well as MSI status, were surveyed for B2M mutations. All B2M-mutant CRCs were assessed for expression of B2M, major histocompatibility complex class I, and programmed death-1 ligand (PD-L1) via immunohistochemistry and average CD3+ and CD8+ tumor-infiltrating lymphocyte counts against a control group of MSI-H B2M wild-type CRCs. RESULTS Fifty-nine (3.4%) of 1,751 patients with CRC harbored B2M mutations, with 84% (77 of 92) of the mutations predicted to be truncating. B2M mutations were significantly enriched in MSI-H CRCs, with 44 (24%) of 182 MSI-H CRCs harboring B2M mutations (P < .001). Thirty-two of 44 B2M-mutant CRCs with available material (73%) had complete loss of B2M expression, whereas all 26 CRCs with wild-type B2M retained expression (P < .001). B2M mutation status was not associated with major histocompatibility complex class I expression, KRAS or BRAF mutation, tumor-infiltrating lymphocyte level, or PD-L1 expression after adjustment for MSI status. Of 13 patients with B2M-mutant CRC who received programmed death-1 or PD-L1 IOs, 11 (85%) achieved clinical benefit, defined as stable disease or partial response using Response Evaluation Criteria in Solid Tumors criteria. CONCLUSION B2M mutations occur in approximately 24% of MSI-H CRCs and are usually associated with loss of B2M expression. Most patients with B2M-mutant MSI-H CRC with loss of protein expression obtain clinical benefit from IOs.
Collapse
Affiliation(s)
- Sumit Middha
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jinru Shia
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Sarah King
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | - Satshil Rana
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Luis A Diaz
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Leonard Saltz
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neil Segal
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
30
|
Sottile R, Federico G, Garofalo C, Tallerico R, Faniello MC, Quaresima B, Cristiani CM, Di Sanzo M, Cuda G, Ventura V, Wagner AK, Contrò G, Perrotti N, Gulletta E, Ferrone S, Kärre K, Costanzo FS, Carlomagno F, Carbone E. Iron and Ferritin Modulate MHC Class I Expression and NK Cell Recognition. Front Immunol 2019; 10:224. [PMID: 30873154 PMCID: PMC6404638 DOI: 10.3389/fimmu.2019.00224] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/25/2019] [Indexed: 12/30/2022] Open
Abstract
The ability of pathogens to sequester iron from their host cells and proteins affects their virulence. Moreover, iron is required for various innate host defense mechanisms as well as for acquired immune responses. Therefore, intracellular iron concentration may influence the interplay between pathogens and immune system. Here, we investigated whether changes in iron concentrations and intracellular ferritin heavy chain (FTH) abundance may modulate the expression of Major Histocompatibility Complex molecules (MHC), and susceptibility to Natural Killer (NK) cell cytotoxicity. FTH downregulation, either by shRNA transfection or iron chelation, led to MHC surface reduction in primary cancer cells and macrophages. On the contrary, mouse embryonic fibroblasts (MEFs) from NCOA4 null mice accumulated FTH for ferritinophagy impairment and displayed MHC class I cell surface overexpression. Low iron concentration, but not FTH, interfered with IFN-γ receptor signaling, preventing the increase of MHC-class I molecules on the membrane by obstructing STAT1 phosphorylation and nuclear translocation. Finally, iron depletion and FTH downregulation increased the target susceptibility of both primary cancer cells and macrophages to NK cell recognition. In conclusion, the reduction of iron and FTH may influence the expression of MHC class I molecules leading to NK cells activation.
Collapse
Affiliation(s)
- Rosa Sottile
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Department of Microbiology, Cell and Tumor Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Giorgia Federico
- Department of Molecular Medicine and Medical Biotechnologies Federico II University, Naples, Italy
| | - Cinzia Garofalo
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Rossana Tallerico
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Maria Concetta Faniello
- Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Barbara Quaresima
- Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Costanza Maria Cristiani
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Maddalena Di Sanzo
- Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Gianni Cuda
- Laboratory of Proteomics, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Valeria Ventura
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Division of Clinical Pathology, Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Arnika Kathleen Wagner
- Department of Microbiology, Cell and Tumor Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Gianluca Contrò
- Department of Health Sciences, University of Catanzaro Magna Graecia, Catanzaro, Italy
| | - Nicola Perrotti
- Department of Health Sciences, University of Catanzaro Magna Graecia, Catanzaro, Italy
| | - Elio Gulletta
- Division of Clinical Pathology, Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Klas Kärre
- Department of Microbiology, Cell and Tumor Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Francesco Saverio Costanzo
- Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy.,CIS for Genomics and Molecular Pathology, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Francesca Carlomagno
- Department of Molecular Medicine and Medical Biotechnologies Federico II University, Naples, Italy
| | - Ennio Carbone
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Department of Microbiology, Cell and Tumor Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Amaral EP, Costa DL, Namasivayam S, Riteau N, Kamenyeva O, Mittereder L, Mayer-Barber KD, Andrade BB, Sher A. A major role for ferroptosis in Mycobacterium tuberculosis-induced cell death and tissue necrosis. J Exp Med 2019; 216:556-570. [PMID: 30787033 PMCID: PMC6400546 DOI: 10.1084/jem.20181776] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/15/2018] [Accepted: 02/01/2019] [Indexed: 12/17/2022] Open
Abstract
Necrotic tissue damage is a major pathological feature of tuberculosis. Here, Amaral et al. show that ferroptosis, a newly described regulated cell death pathway, plays an important role in Mycobacterium tuberculosis–induced cellular necrosis both in vitro and in vivo. Necrotic cell death during Mycobacterium tuberculosis (Mtb) infection is considered host detrimental since it facilitates mycobacterial spread. Ferroptosis is a type of regulated necrosis induced by accumulation of free iron and toxic lipid peroxides. We observed that Mtb-induced macrophage necrosis is associated with reduced levels of glutathione and glutathione peroxidase-4 (Gpx4), along with increased free iron, mitochondrial superoxide, and lipid peroxidation, all of which are important hallmarks of ferroptosis. Moreover, necrotic cell death in Mtb-infected macrophage cultures was suppressed by ferrostatin-1 (Fer-1), a well-characterized ferroptosis inhibitor, as well as by iron chelation. Additional experiments in vivo revealed that pulmonary necrosis in acutely infected mice is associated with reduced Gpx4 expression as well as increased lipid peroxidation and is likewise suppressed by Fer-1 treatment. Importantly, Fer-1–treated infected animals also exhibited marked reductions in bacterial load. Together, these findings implicate ferroptosis as a major mechanism of necrosis in Mtb infection and as a target for host-directed therapy of tuberculosis.
Collapse
Affiliation(s)
- Eduardo P Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Nicolas Riteau
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,University of Orleans and CNRS, UMR7355, Orleans, France
| | - Olena Kamenyeva
- Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Lara Mittereder
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health Bethesda, MD
| | - Bruno B Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, José Silveira Foundation, Salvador, Brazil.,Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN.,Universidade Salvador, Laureate University, Salvador, Bahia, Brazil.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
32
|
Zygiel EM, Nelson CE, Brewer LK, Oglesby-Sherrouse AG, Nolan EM. The human innate immune protein calprotectin induces iron starvation responses in Pseudomonas aeruginosa. J Biol Chem 2019; 294:3549-3562. [PMID: 30622135 DOI: 10.1074/jbc.ra118.006819] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
Most microbial pathogens have a metabolic iron requirement, necessitating the acquisition of this nutrient in the host. In response to pathogen invasion, the human host limits iron availability. Although canonical examples of nutritional immunity are host strategies that limit pathogen access to Fe(III), little is known about how the host restricts access to another biologically relevant oxidation state of this metal, Fe(II). This redox species is prevalent at certain infection sites and is utilized by bacteria during chronic infection, suggesting that Fe(II) withholding by the host may be an effective but unrecognized form of nutritional immunity. Here, we report that human calprotectin (CP; S100A8/S100A9 or MRP8/MRP14 heterooligomer) inhibits iron uptake and induces an iron starvation response in Pseudomonas aeruginosa cells by sequestering Fe(II) at its unusual His6 site. Moreover, under aerobic conditions in which the Fe(III) oxidation state is favored, Fe(II) withholding by CP was enabled by (i) its ability to stabilize this redox state in solution and (ii) the production and secretion of redox-active, P. aeruginosa-produced phenazines, which reduce Fe(III) to Fe(II). Analyses of the interplay between P. aeruginosa secondary metabolites and CP indicated that Fe(II) withholding alters P. aeruginosa physiology and expression of virulence traits. Lastly, examination of the effect of CP on cell-associated metal levels in diverse human pathogens revealed that CP inhibits iron uptake by several bacterial species under aerobic conditions. This work implicates CP-mediated Fe(II) sequestration as a component of nutritional immunity in both aerobic and anaerobic milieus during P. aeruginosa infection.
Collapse
Affiliation(s)
- Emily M Zygiel
- From the Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139 and
| | | | - Luke K Brewer
- Department of Pharmaceutical Sciences, School of Pharmacy and
| | - Amanda G Oglesby-Sherrouse
- Department of Pharmaceutical Sciences, School of Pharmacy and .,Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Elizabeth M Nolan
- From the Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139 and
| |
Collapse
|
33
|
|
34
|
Dahl SL, Woodworth JS, Lerche CJ, Cramer EP, Nielsen PR, Moser C, Thomsen AR, Borregaard N, Cowland JB. Lipocalin-2 Functions as Inhibitor of Innate Resistance to Mycobacterium tuberculosis. Front Immunol 2018; 9:2717. [PMID: 30534124 PMCID: PMC6275245 DOI: 10.3389/fimmu.2018.02717] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/05/2018] [Indexed: 01/14/2023] Open
Abstract
Lipocalin-2 is a constituent of the neutrophil secondary granules and is expressed de novo by macrophages and epithelium in response to inflammation. Lipocalin-2 acts in a bacteriostatic fashion by binding iron-loaded siderophores required for bacterial growth. Mycobacterium tuberculosis (M.tb) produces siderophores that can be bound by lipocalin-2. The impact of lipocalin-2 in the innate immune response toward extracellular bacteria has been established whereas the effect on intracellular bacteria, such as M.tb, is less well-described. Here we show that lipocalin-2 surprisingly confers a growth advantage on M.tb in the early stages of infection (3 weeks post-challenge). Using mixed bone marrow chimeras, we demonstrate that lipocalin-2 derived from granulocytes, but not from epithelia and macrophages, leads to increased susceptibility to M.tb infection. In contrast, lipocalin-2 is not observed to promote mycobacterial growth at later stages of M.tb infection. We demonstrate co-localization of granulocytes and mycobacteria within the nascent granulomas at week 3 post-challenge, but not in the consolidated granulomas at week 5. We hypothesize that neutrophil-derived lipocalin-2 acts to supply a source of iron to M.tb in infected macrophages within the immature granuloma, thereby facilitating mycobacterial growth.
Collapse
Affiliation(s)
- Sara Louise Dahl
- Granulocyte Research Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Joshua S Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | | | | | - Pia Rude Nielsen
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Borregaard
- Granulocyte Research Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Jack Bernard Cowland
- Granulocyte Research Laboratory, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
35
|
Abstract
Transition metals are required cofactors for many proteins that are critical for life, and their concentration within cells is carefully maintained to avoid both deficiency and toxicity. To defend against bacterial pathogens, vertebrate immune proteins sequester metals, in particular zinc, iron, and manganese, as a strategy to limit bacterial acquisition of these necessary nutrients in a process termed "nutritional immunity." In response, bacteria have evolved elegant strategies to access metals and counteract this host defense. In mammals, metal abundance can drastically shift due to changes in dietary intake or absorption from the intestinal tract, disrupting the balance between host and pathogen in the fight for metals and altering susceptibility to disease. This review describes the current understanding of how dietary metals modulate host-microbe interactions and the subsequent impact on the outcome of disease.
Collapse
Affiliation(s)
- Christopher A Lopez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
36
|
Harrington-Kandt R, Stylianou E, Eddowes LA, Lim PJ, Stockdale L, Pinpathomrat N, Bull N, Pasricha J, Ulaszewska M, Beglov Y, Vaulont S, Drakesmith H, McShane H. Hepcidin deficiency and iron deficiency do not alter tuberculosis susceptibility in a murine M.tb infection model. PLoS One 2018; 13:e0191038. [PMID: 29324800 PMCID: PMC5764373 DOI: 10.1371/journal.pone.0191038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/27/2017] [Indexed: 12/20/2022] Open
Abstract
Tuberculosis (TB), caused by the macrophage-tropic pathogen Mycobacterium tuberculosis (M.tb) is a highly prevalent infectious disease. Since an immune correlate of protection or effective vaccine have yet to be found, continued research into host-pathogen interactions is important. Previous literature reports links between host iron status and disease outcome for many infections, including TB. For some extracellular bacteria, the iron regulatory hormone hepcidin is essential for protection against infection. Here, we investigated hepcidin (encoded by Hamp1) in the context of murine M.tb infection. Female C57BL/6 mice were infected with M.tb Erdman via aerosol. Hepatic expression of iron-responsive genes was measured by qRT-PCR and bacterial burden determined in organ homogenates. We found that hepatic Hamp1 mRNA levels decreased post-infection, and correlated with a marker of BMP/SMAD signalling pathways. Next, we tested the effect of Hamp1 deletion, and low iron diets, on M.tb infection. Hamp1 knockout mice did not have a significantly altered M.tb mycobacterial load in either the lungs or spleen. Up to 10 weeks of dietary iron restriction did not robustly affect disease outcome despite causing iron deficiency anaemia. Taken together, our data indicate that unlike with many other infections, hepcidin is decreased following M.tb infection, and show that hepcidin ablation does not influence M.tb growth in vivo. Furthermore, because even severe iron deficiency did not affect M.tb mycobacterial load, we suggest that the mechanisms M.tb uses to scavenge iron from the host must be extremely efficient, and may therefore represent potential targets for drugs and vaccines.
Collapse
Affiliation(s)
| | - Elena Stylianou
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Lucy A. Eddowes
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Pei Jin Lim
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Lisa Stockdale
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Naomi Bull
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Janet Pasricha
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Yulia Beglov
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Sophie Vaulont
- Institut Cochin, INSERM 567, CNRS 8104, Université Paris 5, Paris, France
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail: (HD); (HMcS)
| | - Helen McShane
- Jenner Institute, University of Oxford, Oxford, United Kingdom
- * E-mail: (HD); (HMcS)
| |
Collapse
|
37
|
Chow JK, Ganz T, Ruthazer R, Simpson MA, Pomfret EA, Gordon FD, Westerman ME, Snydman DR. Iron-related markers are associated with infection after liver transplantation. Liver Transpl 2017; 23:1541-1552. [PMID: 28703464 PMCID: PMC5696081 DOI: 10.1002/lt.24817] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/07/2017] [Accepted: 06/26/2017] [Indexed: 12/21/2022]
Abstract
Though serum iron has been known to be associated with an increased risk of infection, hepcidin, the major regulator of iron metabolism, has never been systematically explored in this setting. Finding early biomarkers of infection, such as hepcidin, could help identify patients in whom early empiric antimicrobial therapy would be beneficial. We prospectively enrolled consecutive patients (n = 128) undergoing first-time, single-organ orthotopic liver transplantation (OLT) without known iron overload disorders at 2 academic hospitals in Boston from August 2009 to November 2012. Cox regression compared the associations between different iron markers and the development of first infection at least 1 week after OLT; 47 (37%) patients developed a primary outcome of infection at least 1 week after OLT and 1 patient died. After adjusting for perioperative bleeding complications, number of hospital days, and hepatic artery thrombosis, changes in iron markers were associated with the development of infection post-OLT including increasing ferritin (hazard ratio [HR], 1.51; 95% confidence interval [CI], 1.12-2.05), rising ferritin slope (HR, 1.10; 95% CI, 1.03-1.17), and increasing hepcidin (HR, 1.43; 95% CI, 1.05-1.93). A decreasing iron (HR, 1.76; 95% CI, 1.20-2.57) and a decreasing iron slope (HR, 4.21; 95% CI, 2.51-7.06) were also associated with subsequent infections. In conclusion, hepcidin and other serum iron markers and their slope patterns or their combination are associated with infection in vulnerable patient populations. Liver Transplantation 23 1541-1552 2017 AASLD.
Collapse
Affiliation(s)
- Jennifer K.L Chow
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA, 02116, USA
| | - Tomas Ganz
- Departments of Medicine and Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Robin Ruthazer
- Tufts Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, 02116, USA
| | - Mary Ann Simpson
- Department of Transplantation, Lahey Hospital and Medical Center, Burlington, MA, 01805 USA
| | - Elizabeth A. Pomfret
- Department of Transplantation, Lahey Hospital and Medical Center, Burlington, MA, 01805 USA,Division of Transplant Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Fredric D. Gordon
- Department of Transplantation, Lahey Hospital and Medical Center, Burlington, MA, 01805 USA
| | | | - David R. Snydman
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA, 02116, USA
| |
Collapse
|
38
|
Wu Q, Shen Y, Tao Y, Wei J, Wang H, An P, Zhang Z, Gao H, Zhou T, Wang F, Min J. Hemojuvelin regulates the innate immune response to peritoneal bacterial infection in mice. Cell Discov 2017; 3:17028. [PMID: 28815056 PMCID: PMC5556331 DOI: 10.1038/celldisc.2017.28] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/13/2017] [Indexed: 12/20/2022] Open
Abstract
Hereditary hemochromatosis and iron imbalance are associated with susceptibility to bacterial infection; however, the underlying mechanisms are poorly understood. Here, we performed in vivo bacterial infection screening using several mouse models of hemochromatosis, including Hfe (Hfe−/−), hemojuvelin (Hjv−/−), and macrophage-specific ferroportin-1 (Fpn1fl/fl;LysM-Cre+) knockout mice. We found that Hjv−/− mice, but not Hfe−/− or Fpn1fl/fl;LysM-Cre+ mice, are highly susceptible to peritoneal infection by both Gram-negative and Gram-positive bacteria. Interestingly, phagocytic cells in the peritoneum of Hjv−/− mice have reduced bacterial clearance, IFN-γ secretion, and nitric oxide production; in contrast, both cell migration and phagocytosis are normal. Expressing Hjv in RAW264.7 cells increased the level of phosphorylated Stat1 and nitric oxide production. Moreover, macrophage-specific Hjv knockout mice are susceptible to bacterial infection. Finally, we found that Hjv facilitates the secretion of IFN-γ via the IL-12/Jak2/Stat4 signaling pathway. Together, these findings reveal a novel protective role of Hjv in the early stages of antimicrobial defense.
Collapse
Affiliation(s)
- Qian Wu
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Shen
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunlong Tao
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiayu Wei
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hao Wang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peng An
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhuzhen Zhang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Gao
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tianhua Zhou
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Agoro R, Benmerzoug S, Rose S, Bouyer M, Gozzelino R, Garcia I, Ryffel B, Quesniaux VFJ, Mura C. An Iron-Rich Diet Decreases the Mycobacterial Burden and Correlates With Hepcidin Upregulation, Lower Levels of Proinflammatory Mediators, and Increased T-Cell Recruitment in a Model of Mycobacterium bovis Bacille Calmette-Guerin Infection. J Infect Dis 2017; 216:907-918. [DOI: 10.1093/infdis/jix366] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 07/27/2017] [Indexed: 01/05/2023] Open
|
40
|
Stefanova D, Raychev A, Arezes J, Ruchala P, Gabayan V, Skurnik M, Dillon BJ, Horwitz MA, Ganz T, Bulut Y, Nemeth E. Endogenous hepcidin and its agonist mediate resistance to selected infections by clearing non-transferrin-bound iron. Blood 2017; 130:245-257. [PMID: 28465342 PMCID: PMC5520472 DOI: 10.1182/blood-2017-03-772715] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/29/2017] [Indexed: 12/27/2022] Open
Abstract
The iron-regulatory hormone hepcidin is induced early in infection, causing iron sequestration in macrophages and decreased plasma iron; this is proposed to limit the replication of extracellular microbes, but could also promote infection with macrophage-tropic pathogens. The mechanisms by which hepcidin and hypoferremia modulate host defense, and the spectrum of microbes affected, are poorly understood. Using mouse models, we show that hepcidin was selectively protective against siderophilic extracellular pathogens (Yersinia enterocolitica O9) by controlling non-transferrin-bound iron (NTBI) rather than iron-transferrin concentration. NTBI promoted the rapid growth of siderophilic but not nonsiderophilic bacteria in mice with either genetic or iatrogenic iron overload and in human plasma. Hepcidin or iron loading did not affect other key components of innate immunity, did not indiscriminately promote intracellular infections (Mycobacterium tuberculosis), and had no effect on extracellular nonsiderophilic Y enterocolitica O8 or Staphylococcus aureus Hepcidin analogs may be useful for treatment of siderophilic infections.
Collapse
Affiliation(s)
- Deborah Stefanova
- Molecular, Cellular, and Integrative Physiology Graduate Program and
| | - Antoan Raychev
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| | - Joao Arezes
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute for Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Piotr Ruchala
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| | - Victoria Gabayan
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland; and
| | - Barbara J Dillon
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| | - Marcus A Horwitz
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
- Department of Pathology and
| | - Yonca Bulut
- Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| |
Collapse
|
41
|
Argyropoulos CP, Chen SS, Ng YH, Roumelioti ME, Shaffi K, Singh PP, Tzamaloukas AH. Rediscovering Beta-2 Microglobulin As a Biomarker across the Spectrum of Kidney Diseases. Front Med (Lausanne) 2017; 4:73. [PMID: 28664159 PMCID: PMC5471312 DOI: 10.3389/fmed.2017.00073] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/26/2017] [Indexed: 12/28/2022] Open
Abstract
There is currently an unmet need for better biomarkers across the spectrum of renal diseases. In this paper, we revisit the role of beta-2 microglobulin (β2M) as a biomarker in patients with chronic kidney disease and end-stage renal disease. Prior to reviewing the numerous clinical studies in the area, we describe the basic biology of β2M, focusing in particular on its role in maintaining the serum albumin levels and reclaiming the albumin in tubular fluid through the actions of the neonatal Fc receptor. Disorders of abnormal β2M function arise as a result of altered binding of β2M to its protein cofactors and the clinical manifestations are exemplified by rare human genetic conditions and mice knockouts. We highlight the utility of β2M as a predictor of renal function and clinical outcomes in recent large database studies against predictions made by recently developed whole body population kinetic models. Furthermore, we discuss recent animal data suggesting that contrary to textbook dogma urinary β2M may be a marker for glomerular rather than tubular pathology. We review the existing literature about β2M as a biomarker in patients receiving renal replacement therapy, with particular emphasis on large outcome trials. We note emerging proteomic data suggesting that β2M is a promising marker of chronic allograft nephropathy. Finally, we present data about the role of β2M as a biomarker in a number of non-renal diseases. The goal of this comprehensive review is to direct attention to the multifaceted role of β2M as a biomarker, and its exciting biology in order to propose the next steps required to bring this recently rediscovered biomarker into the twenty-first century.
Collapse
Affiliation(s)
- Christos P Argyropoulos
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Shan Shan Chen
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Yue-Harn Ng
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Maria-Eleni Roumelioti
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Kamran Shaffi
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Pooja P Singh
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Antonios H Tzamaloukas
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States.,Raymond G. Murphy VA Medical Center Albuquerque, Albuquerque, NM, United States
| |
Collapse
|
42
|
Kyaw T, Peter K, Li Y, Tipping P, Toh BH, Bobik A. Cytotoxic lymphocytes and atherosclerosis: significance, mechanisms and therapeutic challenges. Br J Pharmacol 2017; 174:3956-3972. [PMID: 28471481 DOI: 10.1111/bph.13845] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 04/02/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023] Open
Abstract
Cytotoxic lymphocytes encompass natural killer lymphocytes (cells) and cytotoxic T cells that include CD8+ T cells, natural killer (NK) T cells, γ, δ (γδ)-T cells and human CD4 + CD28- T cells. These cells play critical roles in inflammatory diseases and in controlling cancers and infections. Cytotoxic lymphocytes can be activated via a number of mechanisms that may involve dendritic cells, macrophages, cytokines or surface proteins on stressed cells. Upon activation, they secrete pro-inflammatory cytokines as well as anti-inflammatory cytokines, chemokines and cytotoxins to promote inflammation and the development of atherosclerotic lesions including vulnerable lesions, which are strongly implicated in myocardial infarctions and strokes. Here, we review the mechanisms that activate and regulate cytotoxic lymphocyte activity, including activating and inhibitory receptors, cytokines, chemokine receptors-chemokine systems utilized to home to inflamed lesions and cytotoxins and cytokines through which they affect other cells within lesions. We also examine their roles in human and mouse models of atherosclerosis and the mechanisms by which they exert their pathogenic effects. Finally, we discuss strategies for therapeutically targeting these cells to prevent the development of atherosclerotic lesions and vulnerable plaques and the challenge of developing highly targeted therapies that only minimally affect the body's immune system, avoiding the complications, such as increased susceptibility to infections, which are currently associated with many immunotherapies for autoimmune diseases. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Tin Kyaw
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia
| | - Yi Li
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Peter Tipping
- Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Ban-Hock Toh
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Alex Bobik
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
43
|
Ali MK, Kim RY, Karim R, Mayall JR, Martin KL, Shahandeh A, Abbasian F, Starkey MR, Loustaud-Ratti V, Johnstone D, Milward EA, Hansbro PM, Horvat JC. Role of iron in the pathogenesis of respiratory disease. Int J Biochem Cell Biol 2017; 88:181-195. [PMID: 28495571 DOI: 10.1016/j.biocel.2017.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/01/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022]
Abstract
Iron is essential for many biological processes, however, too much or too little iron can result in a wide variety of pathological consequences, depending on the organ system, tissue or cell type affected. In order to reduce pathogenesis, iron levels are tightly controlled in throughout the body by regulatory systems that control iron absorption, systemic transport and cellular uptake and storage. Altered iron levels and/or dysregulated homeostasis have been associated with several lung diseases, including chronic obstructive pulmonary disease, lung cancer, cystic fibrosis, idiopathic pulmonary fibrosis and asthma. However, the mechanisms that underpin these associations and whether iron plays a key role in the pathogenesis of lung disease are yet to be fully elucidated. Furthermore, in order to survive and replicate, pathogenic micro-organisms have evolved strategies to source host iron, including freeing iron from cells and proteins that store and transport iron. To counter these microbial strategies, mammals have evolved immune-mediated defence mechanisms that reduce iron availability to pathogens. This interplay between iron, infection and immunity has important ramifications for the pathogenesis and management of human respiratory infections and diseases. An increased understanding of the role that iron plays in the pathogenesis of lung disease and respiratory infections may help inform novel therapeutic strategies. Here we review the clinical and experimental evidence that highlights the potential importance of iron in respiratory diseases and infections.
Collapse
Affiliation(s)
- Md Khadem Ali
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Richard Y Kim
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Rafia Karim
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Kristy L Martin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Ali Shahandeh
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Firouz Abbasian
- Global Centre for Environmental Remediation, Faculty of Science, the University of Newcastle, Callaghan, NSW 2308, Australia
| | - Malcolm R Starkey
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | | | - Daniel Johnstone
- Bosch Institute and Discipline of Physiology, The University of Sydney, Sydney NSW 2000, Australia
| | - Elizabeth A Milward
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia.
| |
Collapse
|
44
|
Tanner R, O’Shea MK, White AD, Müller J, Harrington-Kandt R, Matsumiya M, Dennis MJ, Parizotto EA, Harris S, Stylianou E, Naranbhai V, Bettencourt P, Drakesmith H, Sharpe S, Fletcher HA, McShane H. The influence of haemoglobin and iron on in vitro mycobacterial growth inhibition assays. Sci Rep 2017; 7:43478. [PMID: 28256545 PMCID: PMC5335253 DOI: 10.1038/srep43478] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/24/2017] [Indexed: 01/04/2023] Open
Abstract
The current vaccine against tuberculosis, live attenuated Mycobacterium bovis BCG, has variable efficacy, but development of an effective alternative is severely hampered by the lack of an immune correlate of protection. There has been a recent resurgence of interest in functional in vitro mycobacterial growth inhibition assays (MGIAs), which provide a measure of a range of different immune mechanisms and their interactions. We identified a positive correlation between mean corpuscular haemoglobin and in vitro growth of BCG in whole blood from healthy UK human volunteers. Mycobacterial growth in peripheral blood mononuclear cells (PBMC) from both humans and macaques was increased following the experimental addition of haemoglobin (Hb) or ferric iron, and reduced following addition of the iron chelator deferoxamine (DFO). Expression of Hb genes correlated positively with mycobacterial growth in whole blood from UK/Asian adults and, to a lesser extent, in PBMC from South African infants. Taken together our data indicate an association between Hb/iron levels and BCG growth in vitro, which may in part explain differences in findings between whole blood and PBMC MGIAs and should be considered when using such assays.
Collapse
Affiliation(s)
- Rachel Tanner
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | | | - Julius Müller
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | | | - Hal Drakesmith
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Sally Sharpe
- Public Health England, Porton Down, Salisbury, UK
| | | | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
45
|
Mycobacterium tuberculosis Rv1474c is a TetR-like transcriptional repressor that regulates aconitase, an essential enzyme and RNA-binding protein, in an iron-responsive manner. Tuberculosis (Edinb) 2017; 103:71-82. [PMID: 28237036 DOI: 10.1016/j.tube.2017.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 01/04/2017] [Accepted: 01/15/2017] [Indexed: 11/21/2022]
Abstract
Mycobacterium tuberculosis (M.tb), tuberculosis (TB) causing bacteria, employs several mechanisms to maintain iron homeostasis which is critical for its survival and pathogenesis. M.tb aconitase (Acn), a [4Fe-4S] cluster-containing essential protein, apart from participating in energy cycle, also binds to predicted iron-responsive RNA elements. In this study, we identified Rv1474c as a regulator of its operonic partner acn and carried out its biochemical and functional characterization. The binding motif for Rv1474c in the upstream region of acn (Rv1475c)-Rv1474c operon was verified by gel-shift assays. Reporter assays in E. coli followed by over-expression studies in mycobacteria, using both wild type and a DNA-binding defective mutant, demonstrated Rv1474c as a Tet-R like repressor of acn. Rv1474c, besides binding tetracycline, could also bind iron which negatively influenced its DNA binding activity. Further, a consistent decrease in the relative transcript levels of acn when M.tb was grown in iron-deficient conditions as compared to either normal or other stress conditions, indicated regulation of acn by Rv1474c in an iron-responsive manner in vivo. The absence of homologs in the human host and its association with indispensable iron homeostasis makes Rv1474c an attractive target for designing novel anti-mycobacterials.
Collapse
|
46
|
Abstract
The review highlights the intrinsic problems in the acquisition of ferric iron (FeIII) by pathogenic microorganisms, and bacteria in particular, during their infection of animals. Acquisition of iron from host sources, such as ferritin, transferrin, and heme compounds, is discussed. Acquisition can be by direct contact, via a surface receptor protein of the bacterium, with one of the iron-containing compounds, but more frequently iron is acquired by the production of a siderophore. Over 500 different siderophores are now known; they work by having a superior binding power to that of the host iron-containing materials. They literally strip the iron out of these molecules. They are low-molecular-weight (< 1,000 Da) compounds that are produced in response to iron deprivation, which is a primary host defense mechanism against infections. The iron–siderophore complex is small enough to be taken up into the bacterial cells, usually via an active transport process; the iron is removed from the siderophore, normally by a reductive process, and is then incorporated into the various apoproteins of the bacterial cell or is stored within the bacteria in the form of bacterioferritin. To combat the effectiveness of the siderophores, animals may synthesize specific proteins to bind and nullify their action. The role of one such protein, siderocalin (= lipocalin 2), is discussed. However, these countermeasures have, in turn, been thwarted by at least one bacterium, Salmonella, glycosylating its siderophore (enterobactin/enterochelin) so that binding of the modified siderophore (now termed salmochelin) with lipocalin can no longer occur.
Collapse
|
47
|
Iron Homeostasis in Mycobacterium tuberculosis: Mechanistic Insights into Siderophore-Mediated Iron Uptake. J Bacteriol 2016; 198:2399-409. [PMID: 27402628 DOI: 10.1128/jb.00359-16] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium tuberculosis requires iron for normal growth but faces a limitation of the metal ion due to its low solubility at biological pH and the withholding of iron by the mammalian host. The pathogen expresses the Fe(3+)-specific siderophores mycobactin and carboxymycobactin to chelate the metal ion from insoluble iron and the host proteins transferrin, lactoferrin, and ferritin. Siderophore-mediated iron uptake is essential for the survival of M. tuberculosis, as knockout mutants, which were defective in siderophore synthesis or uptake, failed to survive in low-iron medium and inside macrophages. But as excess iron is toxic due to its catalytic role in the generation of free radicals, regulation of iron uptake is necessary to maintain optimal levels of intracellular iron. The focus of this review is to present a comprehensive overview of iron homeostasis in M. tuberculosis that is discussed in the context of mycobactin biosynthesis, transport of iron across the mycobacterial cell envelope, and storage of excess iron. The clinical significance of the serum iron status and the expression of the iron-regulated protein HupB in tuberculosis (TB) patients is presented here, highlighting the potential of HupB as a marker, notably in extrapulmonary TB cases.
Collapse
|
48
|
Nonclassical MHC Ib-restricted CD8+ T Cells Recognize Mycobacterium tuberculosis-Derived Protein Antigens and Contribute to Protection Against Infection. PLoS Pathog 2016; 12:e1005688. [PMID: 27272249 PMCID: PMC4896622 DOI: 10.1371/journal.ppat.1005688] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/18/2016] [Indexed: 12/26/2022] Open
Abstract
MHC Ib-restricted CD8+ T cells have been implicated in host defense against Mycobacterium tuberculosis (Mtb) infection. However, the relative contribution of various MHC Ib-restricted T cell populations to anti-mycobacterial immunity remains elusive. In this study, we used mice that lack MHC Ia (Kb-/-Db-/-), MHC Ia/H2-M3 (Kb-/-Db-/-M3-/-), or β2m (β2m-/-) to study the role of M3-restricted and other MHC Ib-restricted T cells in immunity against Mtb. Unlike their dominant role in Listeria infection, we found that M3-restricted CD8+ T cells only represented a small proportion of the CD8+ T cells responding to Mtb infection. Non-M3, MHC Ib-restricted CD8+ T cells expanded preferentially in the lungs of Mtb-infected Kb-/-Db-/-M3-/- mice, exhibited polyfunctional capacities and conferred protection against Mtb. These MHC Ib-restricted CD8+ T cells recognized several Mtb-derived protein antigens at a higher frequency than MHC Ia-restricted CD8+ T cells. The presentation of Mtb antigens to MHC Ib-restricted CD8+ T cells was mostly β2m-dependent but TAP-independent. Interestingly, a large proportion of Mtb-specific MHC Ib-restricted CD8+ T cells in Kb-/-Db-/-M3-/- mice were Qa-2-restricted while no considerable numbers of MR1 or CD1-restricted Mtb-specific CD8+ T cells were detected. Our findings indicate that nonclassical CD8+ T cells other than the known M3, CD1, and MR1-restricted CD8+ T cells contribute to host immune responses against Mtb infection. Targeting these MHC Ib-restricted CD8+ T cells would facilitate the design of better Mtb vaccines with broader coverage across MHC haplotypes due to the limited polymorphism of MHC class Ib molecules.
Collapse
|
49
|
van Rensburg IC, Loxton AG. Transcriptomics: the key to biomarker discovery during tuberculosis? Biomark Med 2016; 9:483-95. [PMID: 25985177 DOI: 10.2217/bmm.15.16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tuberculosis is a global threat affecting millions of people and requires more efficient methods of diagnosis, monitoring treatment response and the development of more efficacious drug therapies and new vaccines. The use of transcriptomic approaches and gene expression techniques have contributed to the elucidation of these aspects concerning the study of tuberculosis, and more specifically, the utilization of transcriptional profiles to identify biomarkers. These markers are the key to developing tools required to improve diagnosis and treatment of tuberculosis. Several studies have led to the identification of markers able to distinguish between different infection states, as well as other pulmonary diseases. Utilizing a systems biology approach will assist in obtaining more reliable results, leading to the implementation of significant findings.
Collapse
|
50
|
Li L, Dong M, Wang XG. The Implication and Significance of Beta 2 Microglobulin: A Conservative Multifunctional Regulator. Chin Med J (Engl) 2016; 129:448-55. [PMID: 26879019 PMCID: PMC4800846 DOI: 10.4103/0366-6999.176084] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE This review focuses on the current knowledge on the implication and significance of beta 2 microglobulin (β2M), a conservative immune molecule in vertebrate. DATA SOURCES The data used in this review were obtained from PubMed up to October 2015. Terms of β2M, immune response, and infection were used in the search. STUDY SELECTIONS Articles related to β2M were retrieved and reviewed. Articles focusing on the characteristic and function of β2M were selected. The exclusion criteria of articles were that the studies on β2M-related molecules. RESULTS β2M is critical for the immune surveillance and modulation in vertebrate animals. The dysregulation of β2M is associated with multiple diseases, including endogenous and infectious diseases. β2M could directly participate in the development of cancer cells, and the level of β2M is deemed as a prognostic marker for several malignancies. It also involves in forming major histocompatibility complex (MHC class I or MHC I) or like heterodimers, covering from antigen presentation to immune homeostasis. CONCLUSIONS Based on the characteristic of β2M, it or its signaling pathway has been targeted as biomedical or therapeutic tools. Moreover, β2M is highly conserved among different species, and overall structures are virtually identical, implying the versatility of β2M on applications.
Collapse
Affiliation(s)
- Ling Li
- Department of Food Quality and Safety, College of Food Science and Biotechnology, Tianjin Agricultural University, Tianjin 300384, China
- Tianjin Engineering Research Center of Agricultural Products Processing, Tianjin 300384, China
| | - Mei Dong
- Department of Clinical Laboratory, Wangdu Hospital of Traditional Chinese Medicine, Baoding, Hebei 072450, China
| | - Xiao-Guang Wang
- Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado Denver, Aurora 80045, Colorado, USA
| |
Collapse
|