1
|
Tang J, Song H, Li S, Lam SM, Ping J, Yang M, Li N, Chang T, Yu Z, Liu W, Lu Y, Zhu M, Tang Z, Liu Z, Guo YR, Shui G, Veillette A, Zeng Z, Wu N. TMEM16F Expressed in Kupffer Cells Regulates Liver Inflammation and Metabolism to Protect Against Listeria Monocytogenes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402693. [PMID: 39136057 PMCID: PMC11497084 DOI: 10.1002/advs.202402693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/20/2024] [Indexed: 10/25/2024]
Abstract
Infection by bacteria leads to tissue damage and inflammation, which need to be tightly controlled by host mechanisms to avoid deleterious consequences. It is previously reported that TMEM16F, a calcium-activated lipid scramblase expressed in various immune cell types including T cells and neutrophils, is critical for the control of infection by bacterium Listeria monocytogenes (Lm) in vivo. This function correlated with the capacity of TMEM16F to repair the plasma membrane (PM) damage induced in T cells in vitro, by the Lm toxin listeriolysin O (LLO). However, whether the protective effect of TMEM16F on Lm infection in vivo is mediated by an impact in T cells, or in other cell types, is not determined. Herein, the immune cell types and mechanisms implicated in the protective effect of TMEM16F against Lm in vivo are elucidated. Cellular protective effects of TMEM16F correlated with its capacity of lipid scrambling and augment PM fluidity. Using cell type-specific TMEM16F-deficient mice, the indication is obtained that TMEM16F expressed in liver Kupffer cells (KCs), but not in T cells or B cells, is key for protection against Listeria in vivo. In the absence of TMEM16F, Listeria induced PM rupture and fragmentation of KCs in vivo. KC death associated with greater liver damage, inflammatory changes, and dysregulated liver metabolism. Overall, the results uncovered that TMEM16F expressed in Kupffer cells is crucial to protect the host against Listeria infection. This influence is associated with the capacity of Kupffer cell-expressed TMEM16F to prevent excessive inflammation and abnormal liver metabolism.
Collapse
Affiliation(s)
- Jianlong Tang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Hua Song
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
| | - Shimin Li
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - Jieming Ping
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Mengyun Yang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Na Li
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Teding Chang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ze Yu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Weixiang Liu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Yan Lu
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Min Zhu
- Department of Thoracic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zhaohui Tang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Yusong R. Guo
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - André Veillette
- Laboratory of Molecular OncologyInstitut de recherches cliniques de Montréal (IRCM)MontréalQuébecH2W1R7Canada
- Department of MedicineUniversity of MontréalMontréalQuébecH3T 1J4Canada
- Department of MedicineMcGill UniversityMontréalQuébecH3G 1Y6Canada
| | - Zhutian Zeng
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Department of OncologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefei230001China
| | - Ning Wu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
2
|
Zhuang Y, Fischer JB, Nishanth G, Schlüter D. Cross-regulation of Listeria monocytogenes and the host ubiquitin system in listeriosis. Eur J Cell Biol 2024; 103:151401. [PMID: 38442571 DOI: 10.1016/j.ejcb.2024.151401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 01/30/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024] Open
Abstract
The facultative intracellular bacterium Listeria (L.) monocytogenes may cause severe diseases in humans and animals. The control of listeriosis/L. monocytogenes requires the concerted action of cells of the innate and adaptive immune systems. In this regard, cell-intrinsic immunity of infected cells, activated by the immune responses, is crucial for the control and elimination intracellular L. monocytogenes. Both the immune response against L. monocytogenes and cell intrinsic pathogen control are critically regulated by post-translational modifications exerted by the host ubiquitin system and ubiquitin-like modifiers (Ubls). In this review, we discuss our current understanding of the role of the ubiquitin system and Ubls in listeriosis, as well as future directions of research.
Collapse
Affiliation(s)
- Yuan Zhuang
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover 30625, Germany.
| | - Johanna B Fischer
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover 30625, Germany
| | - Gopala Nishanth
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover 30625, Germany
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover 30625, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| |
Collapse
|
3
|
Liu M, Tao M, Li J, Sang M, Wu X, Luo H, Zhang J. Functional of tongue sole (Cynoglossus semilaevis) gamma-interferon-inducible lysosomal thiol reductase with implications in innate immune reponse depend on CXXC active site. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104901. [PMID: 37531973 DOI: 10.1016/j.dci.2023.104901] [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: 05/06/2023] [Revised: 06/25/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
The enzyme gamma-interferon-inducible lysosomal thiol reductase (GILT) plays an important role in promoting the processing and presentation of major histocompatibility complex (MHC) class II-restricted antigens. It is also involved in MHC I-restricted antigens catalyzing disulfide bond reduction in fishes' adaptive immunity. The open reading frame of tongue sole (Cynoglossus semilaevis) GILT (tsGILT) gene is 771 bp long, encoding 257 amino acids, with a calculated molecular weight of 28.465 kDa and isoelectric point (pI) of 5.35. After induction with lipopolysaccharide, the expression of tsGILT mRNA was upregulated in spleen and kidney and recombinant tsGILT protein transferred to late endosomes and lysosomes in HeLa cells. The refolded tsGILT was capable of catalyzing the reduction of the interchain disulfide bonds against an IgG substrate depend on the active site CXXC motif at residues 75-78. The process of immune response to bacteria challenge needs GILT to catalyze the reduction of disulfide bond and unfolding native protein antigens, promoting their hydrolysis by proteases. Whether a single mutation or a double mutation of active site CXXC at residues75-78, the 3D structure of tsGILT protein has undergone major changes and lost its activity of catalyzing the reduction of the interchain disulfide bonds.
Collapse
Affiliation(s)
- Meiyan Liu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210046, China; School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China
| | - Mingxuan Tao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Jianfeng Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210046, China; Institute of Aging Research, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, Zhejiang Province, 311121, China
| | - Ming Sang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210046, China; Laboratory of Cellular and Molecular Biology Jiangsu Province Institute of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Xiaolong Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Haibo Luo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Jiaxin Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210046, China.
| |
Collapse
|
4
|
Listeria monocytogenes-How This Pathogen Uses Its Virulence Mechanisms to Infect the Hosts. Pathogens 2022; 11:pathogens11121491. [PMID: 36558825 PMCID: PMC9783847 DOI: 10.3390/pathogens11121491] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Listeriosis is a serious food-borne illness, especially in susceptible populations, including children, pregnant women, and elderlies. The disease can occur in two forms: non-invasive febrile gastroenteritis and severe invasive listeriosis with septicemia, meningoencephalitis, perinatal infections, and abortion. Expression of each symptom depends on various bacterial virulence factors, immunological status of the infected person, and the number of ingested bacteria. Internalins, mainly InlA and InlB, invasins (invasin A, LAP), and other surface adhesion proteins (InlP1, InlP4) are responsible for epithelial cell binding, whereas internalin C (InlC) and actin assembly-inducing protein (ActA) are involved in cell-to-cell bacterial spread. L. monocytogenes is able to disseminate through the blood and invade diverse host organs. In persons with impaired immunity, the elderly, and pregnant women, the pathogen can also cross the blood-brain and placental barriers, which results in the invasion of the central nervous system and fetus infection, respectively. The aim of this comprehensive review is to summarize the current knowledge on the epidemiology of listeriosis and L. monocytogenes virulence mechanisms that are involved in host infection, with a special focus on their molecular and cellular aspects. We believe that all this information is crucial for a better understanding of the pathogenesis of L. monocytogenes infection.
Collapse
|
5
|
Nan X, Zhao K, Qin Y, Song Y, Guo Y, Luo Z, Li W, Wang Q. Antibacterial responses and functional characterization of the interferon gamma inducible lysosomal thiol reductase (GILT) protein in Chinese mitten crab (Eriocheir sinensis). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 136:104514. [PMID: 35977559 DOI: 10.1016/j.dci.2022.104514] [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: 05/30/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
The inducible reductase of interferon gamma (IFN- γ), IFN-γ-induced lysosomal thiol reductase (GILT) is important in antiviral immunity, but its mechanism in invertebrate antimicrobial immunity is unclear. We determined that GILT protein was involved in the antibacterial immunity of Chinese mitten crab (Eriocheir sinensis). GILT protein was highly expressed in crab hemocytes and was significantly upregulated 6 h after bacterial stimulation. Recombinant E. sinensis GILT (rEsGILT) contained a CXXS active site that catalyzed disulfide bond reduction. Vibrio parahaemolyticus and Staphylococcus aureus were bound through interaction with peptidoglycan and lipopolysaccharide, respectively, and bacterial agglutination and clearance in the crabs was markedly promoted. Nevertheless, EsGILT exhibited no direct antibacterial or bactericidal activity. EsGILT also promoted crab hemocyte phagocytosis and played an anti-bacterial role, and inhibited hemocyte apoptosis. In summary, EsGILT promoted bacterial agglutination, clearance, and phagocytosis by recognizing and agglutinating pathogenic microorganisms and reduced the apoptosis level, indirectly participating in antibacterial reactions.
Collapse
Affiliation(s)
- Xingyu Nan
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ke Zhao
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yukai Qin
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yu Song
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yanan Guo
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhi Luo
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Weiwei Li
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense and Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
6
|
Amaro F, Martín-González A. Microbial warfare in the wild-the impact of protists on the evolution and virulence of bacterial pathogens. Int Microbiol 2021; 24:559-571. [PMID: 34365574 DOI: 10.1007/s10123-021-00192-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/03/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023]
Abstract
During the long history of co-evolution with protists, bacteria have evolved defense strategies to avoid grazing and survive phagocytosis. These mechanisms allow bacteria to exploit phagocytic cells as a protective niche in which to escape from environmental stress and even replicate. Importantly, these anti-grazing mechanisms can function as virulence factors when bacteria infect humans. Here, we discuss how protozoan predation exerts a selective pressure driving bacterial virulence and shaping their genomes, and how bacteria-protist interactions might contribute to the spread of antibiotic resistance as well. We provide examples to demonstrate that besides being voracious bacterial predators, protozoa can serve as melting pots where intracellular organisms exchange genetic information, or even "training grounds" where some pathogens become hypervirulent after passing through. In this special issue, we would like to emphasize the tremendous impact of bacteria-protist interactions on human health and the potential of amoebae as model systems to study biology and evolution of a variety of pathogens. Besides, a better understanding of bacteria-protist relationships will help us expand our current understanding of bacterial virulence and, likely, how pathogens emerge.
Collapse
Affiliation(s)
- Francisco Amaro
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Ana Martín-González
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University of Madrid, 28040, Madrid, Spain
| |
Collapse
|
7
|
Ewanchuk BW, Arnold CR, Balce DR, Premnath P, Orsetti TL, Warren AL, Olsen A, Krawetz RJ, Yates RM. A non-immunological role for γ-interferon-inducible lysosomal thiol reductase (GILT) in osteoclastic bone resorption. SCIENCE ADVANCES 2021; 7:7/17/eabd3684. [PMID: 33893096 PMCID: PMC8064644 DOI: 10.1126/sciadv.abd3684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
The extracellular bone resorbing lacuna of the osteoclast shares many characteristics with the degradative lysosome of antigen-presenting cells. γ-Interferon-inducible lysosomal thiol reductase (GILT) enhances antigen processing within lysosomes through direct reduction of antigen disulfides and maintenance of cysteine protease activity. In this study, we found the osteoclastogenic cytokine RANKL drove expression of GILT in osteoclast precursors in a STAT1-dependent manner, resulting in high levels of GILT in mature osteoclasts, which could be further augmented by γ-interferon. GILT colocalized with the collagen-degrading cysteine protease, cathepsin K, suggesting a role for GILT inside the osteoclastic resorption lacuna. GILT-deficient osteoclasts had reduced bone-resorbing capacity, resulting in impaired bone turnover and an osteopetrotic phenotype in GILT-deficient mice. We demonstrated that GILT could directly reduce the noncollagenous bone matrix protein SPARC, and additionally, enhance collagen degradation by cathepsin K. Together, this work describes a previously unidentified, non-immunological role for GILT in osteoclast-mediated bone resorption.
Collapse
Affiliation(s)
- Benjamin W Ewanchuk
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Corey R Arnold
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Dale R Balce
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Priyatha Premnath
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Tanis L Orsetti
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Amy L Warren
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Alexandra Olsen
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Roman J Krawetz
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Robin M Yates
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
8
|
Banerji R, Karkee A, Kanojiya P, Saroj SD. Pore-forming toxins of foodborne pathogens. Compr Rev Food Sci Food Saf 2021; 20:2265-2285. [PMID: 33773026 DOI: 10.1111/1541-4337.12737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 01/04/2023]
Abstract
Pore-forming toxins (PFTs) are water-soluble molecules that have been identified as the most crucial virulence factors during bacterial pathogenesis. PFTs disrupt the host cell membrane to internalize or to deliver other bacterial or virulence factors for establishing infections. Disruption of the host cell membrane by PFTs can lead to uncontrollable exchanges between the extracellular and the intracellular matrix, thereby disturbing the cellular homeostasis. Recent studies have provided insights into the molecular mechanism of PFTs during pathogenesis. Evidence also suggests the activation of several signal transduction pathways in the host cell on recognition of PFTs. Additionally, numerous distinctive host defense mechanisms as well as membrane repair mechanisms have been reported; however, studies reveal that PFTs aid in host immune evasion of the bacteria through numerous pathways. PFTs have been primarily associated with foodborne pathogens. Infection and death from diseases by consuming contaminated food are a constant threat to public health worldwide, affecting socioeconomic development. Moreover, the emergence of new foodborne pathogens has led to the rise of bacterial antimicrobial resistance affecting the population. Hence, this review focuses on the role of PFTs secreted by foodborne pathogens. The review highlights the molecular mechanism of foodborne bacterial PFTs, assisting bacterial survival from the host immune responses and understanding the downstream mechanism in the activation of various signaling pathways in the host upon PFT recognition. PFT research is a remarkable and an important field for exploring novel and broad applications of antimicrobial compounds as therapeutics.
Collapse
Affiliation(s)
- Rajashri Banerji
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Astha Karkee
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Poonam Kanojiya
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
9
|
The molecular mechanisms of listeriolysin O-induced lipid membrane damage. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183604. [PMID: 33722646 DOI: 10.1016/j.bbamem.2021.183604] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/22/2022]
Abstract
Listeria monocytogenes is an intracellular food-borne pathogen that causes listeriosis, a severe and potentially life-threatening disease. Listeria uses a number of virulence factors to proliferate and spread to various cells and tissues. In this process, three bacterial virulence factors, the pore-forming protein listeriolysin O and phospholipases PlcA and PlcB, play a crucial role. Listeriolysin O belongs to a family of cholesterol-dependent cytolysins that are mostly expressed by gram-positive bacteria. Its unique structural features in an otherwise conserved three-dimensional fold, such as the acidic triad and proline-glutamate-serine-threonine-like sequence, enable the regulation of its intracellular activity as well as distinct extracellular functions. The stability of listeriolysin O is pH- and temperature-dependent, and this provides another layer of control of its activity in cells. Moreover, many recent studies have demonstrated a unique mechanism of pore formation by listeriolysin O, i.e., the formation of arc-shaped oligomers that can subsequently fuse to form membrane defects of various shapes and sizes. During listerial invasion of host cells, these membrane defects can disrupt phagosome membranes, allowing bacteria to escape into the cytosol and rapidly multiply. The activity of listeriolysin O is profoundly dependent on the amount and accessibility of cholesterol in the lipid membrane, which can be modulated by the phospholipase PlcB. All these prominent features of listeriolysin O play a role during different stages of the L. monocytogenes life cycle by promoting the proliferation of the pathogen while mitigating excessive damage to its replicative niche in the cytosol of the host cell.
Collapse
|
10
|
Gao L, Li A, Lv Y, Huang M, Liu X, Deng H, Liu D, Zhao B, Liu B, Pang Q. Planarian gamma-interferon-inducible lysosomal thiol reductase (GILT) is required for gram-negative bacterial clearance. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 116:103914. [PMID: 33137392 DOI: 10.1016/j.dci.2020.103914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 06/11/2023]
Abstract
The powerful regenerative ability of planarians has long been a concern of scientists, but recently, their efficient immune system has attracted more and more attention from researchers. Gamma-interferon-inducible lysosomal thiol reductase (GILT) is related not only to antigen presentation but also to bacteria invasions. But the systematic studies are not yet to be conducted on the relationship between bacterial infection. Our study reveals for the first time that GILT of planarian (DjGILT) plays an essential role in the clearance of Gram-negative bacteria by conducting H2O2 concentration in planarians. In animals that DjGILT was silenced, it persisted for up to 9 days before all bacteria were cleared, compared with 6 days of the control group. When infected with E. coli and V. anguillarum, the level of H2O2 was significantly increased in DjGILT-silenced planarians, and concomitantly, mRNA level of C-type lectin DjCTL, which modulates agglutination and clearance efficiency of invading bacteria, was decreased. Further study showed that the decrease of H2O2 level led to a significant increase in DjCTL transcripts. Collectively, we proposed a mechanism model for the involvement of GILT gene in bacterial elimination. We have for the first time revealed the specific mechanism of GILT in innate immune response against bacterial infection.
Collapse
Affiliation(s)
- Lili Gao
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China; Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, 255049, PR China
| | - Ao Li
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China
| | - Yanhua Lv
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, China
| | - Mujie Huang
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China
| | - Xi Liu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China
| | - Hongkuan Deng
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China; Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, 255049, PR China
| | - Dongwu Liu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, 255049, PR China
| | - Baohua Liu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China; Shenzhen University of Health Science Center, District Shenzhen, 518060, China.
| | - Qiuxiang Pang
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, 255049, China; Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, 255049, PR China.
| |
Collapse
|
11
|
Innate immune responses to Listeria in vivo. Curr Opin Microbiol 2020; 59:95-101. [PMID: 33307408 DOI: 10.1016/j.mib.2020.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023]
Abstract
Listeria monocytogenes (Lm) is a foodborne bacterial pathogen that causes listeriosis, a severe infection that manifests as bacteremia and meningo-encephalitis mostly in immunocompromised individuals, and maternal-fetal infection. A critical pathogenic determinant of Lm relies on its ability to actively cross the intestinal barrier, disseminate systemically and cross the blood-brain and placental barriers. Here we illustrate how Lm both evades innate immunity, favoring its dissemination in host tissues, and triggers innate immune defenses that participate to its control.
Collapse
|
12
|
Anast JM, Bobik TA, Schmitz-Esser S. The Cobalamin-Dependent Gene Cluster of Listeria monocytogenes: Implications for Virulence, Stress Response, and Food Safety. Front Microbiol 2020; 11:601816. [PMID: 33240255 PMCID: PMC7677406 DOI: 10.3389/fmicb.2020.601816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Several genes of the eut, pdu, and cob/cbi operons are responsible for the metabolism of ethanolamine (EA) and 1,2-propanediol (PD) and are essential during the pathogenic lifecycles of various enteric pathogens. Studies concerning EA and PD metabolism have primarily focused on bacterial genera from the family Enterobacteriaceae, especially the genus Salmonella. Listeria monocytogenes is a member of the Firmicutes phylum and is the causative agent of the rare but highly fatal foodborne disease listeriosis. The eut, pdu, and cob/cbi operons are organized as a single large locus collectively referred to as the cobalamin-dependent gene cluster (CDGC). The CDGC is well conserved in L. monocytogenes; however, functional characterization of the genes in this cluster and how they may contribute to Listeria virulence and stress tolerance in food production environments is highly limited. Previous work suggests that the degradation pathway of PD is essential for L. monocytogenes establishment in the gastrointestinal tract. In contrast, EA metabolism may be more important during intracellular replication. Other studies indicate that the CDGC is utilized when L. monocytogenes is exposed to food and food production relevant stress conditions. Perhaps most noteworthy, L. monocytogenes exhibits attenuated growth at cold temperatures when a key EA utilization pathway gene was deleted. This review aims to summarize the current knowledge of these pathways in L. monocytogenes and their significance in virulence and stress tolerance, especially considering recent developments.
Collapse
Affiliation(s)
- Justin M Anast
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States.,Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Thomas A Bobik
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States.,Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Stephan Schmitz-Esser
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States.,Department of Animal Science, Iowa State University, Ames, IA, United States
| |
Collapse
|
13
|
Carey KL, Paulus GLC, Wang L, Balce DR, Luo JW, Bergman P, Ferder IC, Kong L, Renaud N, Singh S, Kost-Alimova M, Nyfeler B, Lassen KG, Virgin HW, Xavier RJ. TFEB Transcriptional Responses Reveal Negative Feedback by BHLHE40 and BHLHE41. Cell Rep 2020; 33:108371. [PMID: 33176151 DOI: 10.1016/j.celrep.2020.108371] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 12/26/2022] Open
Abstract
Transcription factor EB (TFEB) activates lysosomal biogenesis genes in response to environmental cues. Given implications of impaired TFEB signaling and lysosomal dysfunction in metabolic, neurological, and infectious diseases, we aim to systematically identify TFEB-directed circuits by examining transcriptional responses to TFEB subcellular localization and stimulation. We reveal that steady-state nuclear TFEB is sufficient to activate transcription of lysosomal, autophagy, and innate immunity genes, whereas other targets require higher thresholds of stimulation. Furthermore, we identify shared and distinct transcriptional signatures between mTOR inhibition and bacterial autophagy. Using a genome-wide CRISPR library, we find TFEB targets that protect cells from or sensitize cells to lysosomal cell death. BHLHE40 and BHLHE41, genes responsive to high, sustained levels of nuclear TFEB, act in opposition to TFEB upon lysosomal cell death induction. Further investigation identifies genes counter-regulated by TFEB and BHLHE40/41, adding this negative feedback to the current understanding of TFEB regulatory mechanisms.
Collapse
Affiliation(s)
- Kimberly L Carey
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Geraldine L C Paulus
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingfei Wang
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Dale R Balce
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jessica W Luo
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Phil Bergman
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Ianina C Ferder
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingjia Kong
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nicole Renaud
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Maria Kost-Alimova
- Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Beat Nyfeler
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Kara G Lassen
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramnik J Xavier
- Immunology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
14
|
Daussy CF, Wodrich H. "Repair Me if You Can": Membrane Damage, Response, and Control from the Viral Perspective. Cells 2020; 9:cells9092042. [PMID: 32906744 PMCID: PMC7564661 DOI: 10.3390/cells9092042] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Cells are constantly challenged by pathogens (bacteria, virus, and fungi), and protein aggregates or chemicals, which can provoke membrane damage at the plasma membrane or within the endo-lysosomal compartments. Detection of endo-lysosomal rupture depends on a family of sugar-binding lectins, known as galectins, which sense the abnormal exposure of glycans to the cytoplasm upon membrane damage. Galectins in conjunction with other factors orchestrate specific membrane damage responses such as the recruitment of the endosomal sorting complex required for transport (ESCRT) machinery to either repair damaged membranes or the activation of autophagy to remove membrane remnants. If not controlled, membrane damage causes the release of harmful components including protons, reactive oxygen species, or cathepsins that will elicit inflammation. In this review, we provide an overview of current knowledge on membrane damage and cellular responses. In particular, we focus on the endo-lysosomal damage triggered by non-enveloped viruses (such as adenovirus) and discuss viral strategies to control the cellular membrane damage response. Finally, we debate the link between autophagy and inflammation in this context and discuss the possibility that virus induced autophagy upon entry limits inflammation.
Collapse
|
15
|
Bianchi F, van den Bogaart G. Vacuolar escape of foodborne bacterial pathogens. J Cell Sci 2020; 134:134/5/jcs247221. [PMID: 32873733 DOI: 10.1242/jcs.247221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The intracellular pathogens Listeria monocytogenes, Salmonella enterica, Shigella spp. and Staphylococcus aureus are major causes of foodborne illnesses. Following the ingestion of contaminated food or beverages, pathogens can invade epithelial cells, immune cells and other cell types. Pathogens survive and proliferate intracellularly via two main strategies. First, the pathogens can remain in membrane-bound vacuoles and tailor organellar trafficking to evade host-cell defenses and gain access to nutrients. Second, pathogens can rupture the vacuolar membrane and proliferate within the nutrient-rich cytosol of the host cell. Although this virulence strategy of vacuolar escape is well known for L. monocytogenes and Shigella spp., it has recently become clear that S. aureus and Salmonella spp. also gain access to the cytosol, and that this is important for their survival and growth. In this Review, we discuss the molecular mechanisms of how these intracellular pathogens rupture the vacuolar membrane by secreting a combination of proteins that lyse the membranes or that remodel the lipids of the vacuolar membrane, such as phospholipases. In addition, we also propose that oxidation of the vacuolar membrane also contributes to cytosolic pathogen escape. Understanding these escape mechanisms could aid in the identification of new therapeutic approaches to combat foodborne pathogens.
Collapse
Affiliation(s)
- Frans Bianchi
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9722GR Groningen, The Netherlands
| | - Geert van den Bogaart
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9722GR Groningen, The Netherlands .,Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 9625GA Nijmegen, The Netherlands
| |
Collapse
|
16
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
17
|
La Pietra L, Hudel M, Pillich H, Abu Mraheil M, Berisha B, Aden S, Hodnik V, Lochnit G, Rafiq A, Perniss A, Anderluh G, Chakraborty T. Phosphocholine Antagonizes Listeriolysin O-Induced Host Cell Responses of Listeria monocytogenes. J Infect Dis 2020; 222:1505-1516. [DOI: 10.1093/infdis/jiaa022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/20/2020] [Indexed: 12/20/2022] Open
Abstract
Abstract
Background
Bacterial toxins disrupt plasma membrane integrity with multitudinous effects on host cells. The secreted pore-forming toxin listeriolysin O (LLO) of the intracellular pathogen Listeria monocytogenes promotes egress of the bacteria from vacuolar compartments into the host cytosol often without overt destruction of the infected cell. Intracellular LLO activity is tightly controlled by host factors including compartmental pH, redox, proteolytic, and proteostatic factors, and inhibited by cholesterol.
Methods
Combining infection studies of L. monocytogenes wild type and isogenic mutants together with biochemical studies with purified phospholipases, we investigate the effect of their enzymatic activities on LLO.
Results
Here, we show that phosphocholine (ChoP), a reaction product of the phosphatidylcholine-specific phospholipase C (PC-PLC) of L. monocytogenes, is a potent inhibitor of intra- and extracellular LLO activities. Binding of ChoP to LLO is redox-independent and leads to the inhibition of LLO-dependent induction of calcium flux, mitochondrial damage, and apoptosis. ChoP also inhibits the hemolytic activities of the related cholesterol-dependent cytolysins (CDC), pneumolysin and streptolysin.
Conclusions
Our study uncovers a strategy used by L. monocytogenes to modulate cytotoxic LLO activity through the enzymatic activity of its PC-PLC. This mechanism appears to be widespread and also used by other CDC pore-forming toxin-producing bacteria.
Collapse
Affiliation(s)
- Luigi La Pietra
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Martina Hudel
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Helena Pillich
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Mobarak Abu Mraheil
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Besim Berisha
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Saša Aden
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Vesna Hodnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Günter Lochnit
- Protein Analytics, Institute of Biochemistry, Justus-Liebig University Giessen, Giessen, Germany
| | - Amir Rafiq
- Institute of Anatomy and Cell Biology, Cardiopulmonary Institute, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Alexander Perniss
- Institute of Anatomy and Cell Biology, Cardiopulmonary Institute, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Trinad Chakraborty
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
18
|
Chen D, Hou Z, Jiang D, Zheng M, Li G, Zhang Y, Li R, Lin H, Chang J, Zeng H, Guo JT, Zhao X. GILT restricts the cellular entry mediated by the envelope glycoproteins of SARS-CoV, Ebola virus and Lassa fever virus. Emerg Microbes Infect 2020; 8:1511-1523. [PMID: 31631785 PMCID: PMC6818130 DOI: 10.1080/22221751.2019.1677446] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Interferons (IFNs) control viral infections by inducing expression of IFN-stimulated genes (ISGs) that restrict distinct steps of viral replication. We report herein that gamma-interferon-inducible lysosomal thiol reductase (GILT), a lysosome-associated ISG, restricts the infectious entry of selected enveloped RNA viruses. Specifically, we demonstrated that GILT was constitutively expressed in lung epithelial cells and fibroblasts and its expression could be further induced by type II interferon. While overexpression of GILT inhibited the entry mediated by envelope glycoproteins of SARS coronavirus (SARS-CoV), Ebola virus (EBOV) and Lassa fever virus (LASV), depletion of GILT enhanced the entry mediated by these viral envelope glycoproteins. Furthermore, mutations that impaired the thiol reductase activity or disrupted the N-linked glycosylation, a posttranslational modification essential for its lysosomal localization, largely compromised GILT restriction of viral entry. We also found that the induction of GILT expression reduced the level and activity of cathepsin L, which is required for the entry of these RNA viruses in lysosomes. Our data indicate that GILT is a novel antiviral ISG that specifically inhibits the entry of selected enveloped RNA viruses in lysosomes via disruption of cathepsin L metabolism and function and may play a role in immune control and pathogenesis of these viruses.
Collapse
Affiliation(s)
- Danying Chen
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| | - Zhifei Hou
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China.,Department of Pulmonary and Critical Care Medicine, General Hospital of Datong Coal Mine Group Co., Ltd. , People's Republic of China
| | - Dong Jiang
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| | - Mei Zheng
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| | - Guoli Li
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| | - Yue Zhang
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| | - Rui Li
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| | - Hanxin Lin
- Department of Pathology and Laboratory Medicine, Western University , London , Ontario , Canada
| | - Jinhong Chang
- Baruch S. Blumberg Institute, Hepatitis B Foundation , Doylestown , PA , USA
| | - Hui Zeng
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| | - Ju-Tao Guo
- Baruch S. Blumberg Institute, Hepatitis B Foundation , Doylestown , PA , USA
| | - Xuesen Zhao
- Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University , Beijing , People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Disease , Beijing , People's Republic of China
| |
Collapse
|
19
|
Jin J, Xu H, Wu R, Gao N, Wu N, Li S, Niu J. Identification of key genes and pathways associated with different immune statuses of hepatitis B virus infection. J Cell Mol Med 2019; 23:7474-7489. [PMID: 31565863 PMCID: PMC6815815 DOI: 10.1111/jcmm.14616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/18/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
We aimed to identify key genes and pathways associated with different immune statuses of hepatitis B virus (HBV) infection. The gene expression and DNA methylation profiles were analysed in different immune statuses of HBV infection. Differentially expressed genes (DEGs) and differentially methylated genes (DMGs) were identified, followed by their functional and integrative analyses. The differential expression of IgG Fc receptors (FcγRs) in chronic HBV-infected patients and immune cells during different stages of HBV infection was investigated. Toll-like receptor (TLR) signalling pathway (including TLR6) and leucocyte transendothelial migration pathway (including integrin subunit beta 1) were enriched during acute infection. Key DEGs, such as FcγR Ib and FcγR Ia, and interferon-alpha inducible protein 27 showed correlation with alanine aminotransferase levels, and they were differentially expressed between acute and immune-tolerant phases and between immune-tolerant and immune-clearance phases. The integrative analysis of DNA methylation profile showed that lowly methylated and highly expressed genes, including cytotoxic T lymphocyte-associated protein 4 and mitogen-activated protein kinase 3 were enriched in T cell receptor signalling pathway during acute infection. Highly methylated and lowly expressed genes, such as Ras association domain family member 1 and cyclin-dependent kinase inhibitor 2A were identified in chronic infection. Furthermore, differentially expressed FcγR Ia, FcγR IIa and FcγR IIb, CD3- CD56+ CD16+ natural killer cells and CD14high CD16+ monocytes were identified between immune-tolerant and immune-clearance phases by experimental validation. The above genes and pathways may be used to distinguish different immune statuses of HBV infection.
Collapse
MESH Headings
- DNA Methylation/genetics
- DNA Methylation/immunology
- Female
- Gene Expression/genetics
- Hepatitis B/genetics
- Hepatitis B/immunology
- Hepatitis B virus/immunology
- Hepatitis B, Chronic/genetics
- Hepatitis B, Chronic/immunology
- Hepatitis B, Chronic/virology
- Humans
- Killer Cells, Natural/immunology
- Male
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Signal Transduction/genetics
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Jinglan Jin
- Department of Hepatology, The First Hospital of Jilin UniversityJilin UniversityChangchunChina
| | - Hongqin Xu
- Department of Hepatology, The First Hospital of Jilin UniversityJilin UniversityChangchunChina
- Jilin Province Key Laboratory of Infectious DiseasesLaboratory of Molecular VirologyChangchunChina
| | - Ruihong Wu
- Department of Hepatology, The First Hospital of Jilin UniversityJilin UniversityChangchunChina
- Jilin Province Key Laboratory of Infectious DiseasesLaboratory of Molecular VirologyChangchunChina
| | - Na Gao
- Department of Infectious DiseaseThe Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhouChina
| | - Na Wu
- Lanshan People’s HospitalLinyiChina
| | - Shibo Li
- Department of Pediatrics, Genetics LaboratoryUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Junqi Niu
- Department of Hepatology, The First Hospital of Jilin UniversityJilin UniversityChangchunChina
- Jilin Province Key Laboratory of Infectious DiseasesLaboratory of Molecular VirologyChangchunChina
| |
Collapse
|
20
|
Cross Talk between SigB and PrfA in Listeria monocytogenes Facilitates Transitions between Extra- and Intracellular Environments. Microbiol Mol Biol Rev 2019; 83:83/4/e00034-19. [PMID: 31484692 DOI: 10.1128/mmbr.00034-19] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The foodborne pathogen Listeria monocytogenes can modulate its transcriptome and proteome to ensure its survival during transmission through vastly differing environmental conditions. While L. monocytogenes utilizes a large array of regulators to achieve survival and growth in different intra- and extrahost environments, the alternative sigma factor σB and the transcriptional activator of virulence genes protein PrfA are two key transcriptional regulators essential for responding to environmental stress conditions and for host infection. Importantly, emerging evidence suggests that the shift from extrahost environments to the host gastrointestinal tract and, subsequently, to intracellular environments requires regulatory interplay between σB and PrfA at transcriptional, posttranscriptional, and protein activity levels. Here, we review the current evidence for cross talk and interplay between σB and PrfA and their respective regulons and highlight the plasticity of σB and PrfA cross talk and the role of this cross talk in facilitating successful transition of L. monocytogenes from diverse extrahost to diverse extra- and intracellular host environments.
Collapse
|
21
|
Westman J, Hube B, Fairn GD. Integrity under stress: Host membrane remodelling and damage by fungal pathogens. Cell Microbiol 2019; 21:e13016. [DOI: 10.1111/cmi.13016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Johannes Westman
- Program in Cell Biology The Hospital for Sick Children Toronto Ontario Canada
| | - Bernhard Hube
- Department Microbial Pathogenicity Mechanisms Hans Knoell Institute Jena Germany
- Institute of Microbiology Microbial Pathogenicity Friedrich Schiller University Jena Germany
| | - Gregory D. Fairn
- Keenan Research Centre for Biomedical Sciences St. Michael's Hospital Toronto Ontario Canada
- Department of Surgery University of Toronto Toronto Ontario Canada
| |
Collapse
|
22
|
Nguyen BN, Peterson BN, Portnoy DA. Listeriolysin O: A phagosome-specific cytolysin revisited. Cell Microbiol 2019; 21:e12988. [PMID: 30511471 DOI: 10.1111/cmi.12988] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
Listeriolysin O (LLO) is an essential determinant of Listeria monocytogenes pathogenesis that mediates the escape of L. monocytogenes from host cell vacuoles, thereby allowing replication in the cytosol without causing appreciable cell death. As a member of the cholesterol-dependent cytolysin (CDC) family of pore-forming toxins, LLO is unique in that it is secreted by a facultative intracellular pathogen, whereas all other CDCs are produced by pathogens that are largely extracellular. Replacement of LLO with other CDCs results in strains that are extremely cytotoxic and 10,000-fold less virulent in mice. LLO has structural and regulatory features that allow it to function intracellularly without causing cell death, most of which map to a unique N-terminal region of LLO referred to as the proline, glutamic acid, serine, threonine (PEST)-like sequence. Yet, while LLO has unique properties required for its intracellular site of action, extracellular LLO, like other CDCs, affects cells in a myriad of ways. Because all CDCs form pores in cholesterol-containing membranes that lead to rapid Ca2+ influx and K+ efflux, they consequently trigger a wide range of host cell responses, including mitogen-activated protein kinase activation, histone modification, and caspase-1 activation. There is no debate that extracellular LLO, like all other CDCs, can stimulate multiple cellular activities, but the primary question we wish to address in this perspective is whether these activities contribute to L. monocytogenes pathogenesis.
Collapse
Affiliation(s)
- Brittney N Nguyen
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, California
| | - Bret N Peterson
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, California
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California.,Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California
| |
Collapse
|
23
|
Fu J, Chen S, Zhao X, Luo Z, Zou P, Liu Y. Identification and characterization of the interferon-γ-inducible lysosomal thiol reductase gene in Chinese soft-shelled turtle, Pelodiscus sinensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 90:55-59. [PMID: 30172908 DOI: 10.1016/j.dci.2018.08.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/29/2018] [Accepted: 08/29/2018] [Indexed: 06/08/2023]
Abstract
The reduction of disulfide bonds of exogenous antigens is crucial to the MHC-II class antigen processing and presenting pathway and is catalysed by interferon-γ-inducible lysosomal thiol reductase (GILT). In this study, a reptile GILT gene from Chinese soft-shelled turtle, Pelodiscus sinensis (PsGILT), was identified. The full-length cDNA of PsGILT is 1631 nucleotides (nt), including a 5'-untranslated region (UTR) of 3 nt, a 3'-UTR of 860 nt and an open reading frame (ORF) of 768 nt encoding 255 amino acids (aa). The conserved features in known GILTs, such as signal peptide, CXXC motif, GILT signature sequence, N-glycosylation site and conserved cysteines, were all found in the putative PsGILT protein. Genomic analysis revealed that PsGILT kept the "7 exons and 6 introns" structure of vertebrate GILT genes. PsGILT was expressed in all examined organs/tissues and was mainly expressed in spleen and blood. Increased mRNA expression levels of PsIFN-γ and PsGILT in PBLs were observed after induction with LPS, PolyI:C and recombinant IFN-γ (rIFN-γ). We also tested the reductase activity of rGILT in vitro and found that it could reduce intact human IgG into H chains and L chains. These above results implied that PsGILT may play an important role in resisting bacterial and viral infections, like other vertebrate GILTs.
Collapse
Affiliation(s)
- Jianping Fu
- College of Life Sciences, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi Province, 330022, China
| | - Shannan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Xin Zhao
- College of Life Sciences, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi Province, 330022, China
| | - Zhang Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Pengfei Zou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Yi Liu
- College of Life Sciences, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi Province, 330022, China.
| |
Collapse
|
24
|
Chen S, Wang Q, Shao X, Di G, Dai Y, Jiang X, Cheng L. Lentivirus mediated γ-interferon-inducible lysosomal thiol reductase (GILT) knockdown suppresses human glioma U373MG cell proliferation. Biochem Biophys Res Commun 2018; 509:182-187. [PMID: 30587343 DOI: 10.1016/j.bbrc.2018.12.099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 12/13/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Glioma is the most common malignancy in brain carcinoma with poor prognosis due to the lack of understanding of the mechanism underlying the disease. γ-interferon-inducible lysosomal thiol reductase (GILT) plays a critical role in the process of antigen processing. However, the role of GILT in the tumorigenesis of glioma remains unknown. MATERIALS AND METHODS The expression of GILT was analyzed by bioinformatics using the public database and by qPCR in three human glioma cell lines. Cell growth and viability were determined by Celigo and MTT assays, while cell cycle arrest and apoptosis were determined using flow cytometry. Giemsa staining was used to analyze the colony formation, while cell motility was assessed using transwell migration and invasion assays, as well as, using tumor growth in nude mice. RESULTS GILT was highly expressed as observed in the public database on human gliomas and two human glioma cell lines, U373MG and U87MG cells. The downregulation of GILT by lentiviral-mediated silencing inhibits the cell growth, colony formation, and migration but promotes apoptosis and results in cell cycle arrest at the G0/G1 phase in the U373MG cells. Also, the knockdown of GILT inhibits tumor growth in vivo. CONCLUSION Elevated GILT is positively associated with glioma progression. GILT silencing suppresses cell proliferation, colony formation, migration, and tumor growth, and induces apoptosis and cell cycle arrest. GILT may serve as a potential target for the treatment of glioma.
Collapse
Affiliation(s)
- Sansong Chen
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, No.2 Zheshan Road, Wuhu, 241001, Anhui, China
| | - Qifu Wang
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, No.2 Zheshan Road, Wuhu, 241001, Anhui, China
| | - Xuefei Shao
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, No.2 Zheshan Road, Wuhu, 241001, Anhui, China
| | - Guangfu Di
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, No.2 Zheshan Road, Wuhu, 241001, Anhui, China
| | - Yi Dai
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, No.2 Zheshan Road, Wuhu, 241001, Anhui, China
| | - Xiaochun Jiang
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, No.2 Zheshan Road, Wuhu, 241001, Anhui, China.
| | - Limin Cheng
- Central Laboratory of Microscopic Morphology, School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, Anhui, China.
| |
Collapse
|
25
|
Pizarro-Cerdá J, Cossart P. Listeria monocytogenes: cell biology of invasion and intracellular growth. Microbiol Spectr 2018; 6:10.1128/microbiolspec.gpp3-0013-2018. [PMID: 30523778 PMCID: PMC11633638 DOI: 10.1128/microbiolspec.gpp3-0013-2018] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Indexed: 12/26/2022] Open
Abstract
The Gram-positive pathogen Listeria monocytogenes is able to promote its entry into a diverse range of mammalian host cells by triggering plasma membrane remodeling, leading to bacterial engulfment. Upon cell invasion, L. monocytogenes disrupts its internalization vacuole and translocates to the cytoplasm, where bacterial replication takes place. Subsequently, L. monocytogenes uses an actin-based motility system that allows bacterial cytoplasmic movement and cell-to-cell spread. L. monocytogenes therefore subverts host cell receptors, organelles and the cytoskeleton at different infection steps, manipulating diverse cellular functions that include ion transport, membrane trafficking, post-translational modifications, phosphoinositide production, innate immune responses as well as gene expression and DNA stability.
Collapse
Affiliation(s)
- Javier Pizarro-Cerdá
- Unité Interactions Bactéries-Cellules, Institut Pasteur, Paris F-75015, FRANCE
- INSERM U604, Paris F-75015, FRANCE
- INRA USC2020, Paris F-75015, FRANCE
| | - Pascale Cossart
- Unité Interactions Bactéries-Cellules, Institut Pasteur, Paris F-75015, FRANCE
- INSERM U604, Paris F-75015, FRANCE
- INRA USC2020, Paris F-75015, FRANCE
| |
Collapse
|
26
|
Zhang B, Luo G, Zhao L, Huang L, Qin Y, Su Y, Yan Q. Integration of RNAi and RNA-seq uncovers the immune responses of Epinephelus coioides to L321_RS19110 gene of Pseudomonas plecoglossicida. FISH & SHELLFISH IMMUNOLOGY 2018; 81:121-129. [PMID: 30006040 DOI: 10.1016/j.fsi.2018.06.051] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/17/2018] [Accepted: 06/27/2018] [Indexed: 06/08/2023]
Abstract
Pseudomonas plecoglossicida is a threatening and important pathogen in aquaculture and causes heavy losses. Expression of L321_RS19110 was found significant up-regulated at 18 °C than at 28 °C, which was confirmed by quantitative real-time PCR. RNAi significantly reduced the content of L321_RS19110 mRNA of P. plecoglossicida, and exhibited the best efficiency of gene silencing with a reduction of 84.9%. Compared with the wild type strain, the infection of L321_RS19110-RNAi-1 strain resulted in the onset time delay, and 30% reduction in mortality of Epinephelus coioides, as well as alleviates in the symptoms of E. coioides spleen. Moreover, compared with wild type strain, the gene silence of L321_RS19110 in P. plecoglossicida resulted in a significant change in transcriptome of infected E. coioides. The results of KEGG analysis showed that genes of chemokine signaling pathway and cytokine-cytokine receptor interaction, digestion and absorption of proteins (non-specific immune) and antigen processing and presentation pathways were most affected by L321_RS19110-RNAi of P. plecoglossicida. In these immune pathways, the most affected immune genes were associated with different number of non-coding RNAs. Among them, most lncRNAs and miRNAs exerted negative regulation on immune genes. The results indicated that L321_RS19110 was an important virulent gene of P. plecoglossicida, the up-regulation of the immune pathways made E. coioides more likely to remove L321_RS19110-RNAi strain than the wild type strain of P. plecoglossicida, the expression of immune genes were regulated by miRNA and lncRNA in a complex way.
Collapse
Affiliation(s)
- Beibei Zhang
- Fisheries College, Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Jimei University, Xiamen, Fujian, 361021, China
| | - Gang Luo
- Fisheries College, Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Jimei University, Xiamen, Fujian, 361021, China
| | - Lingmin Zhao
- Fisheries College, Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Jimei University, Xiamen, Fujian, 361021, China
| | - Lixing Huang
- Fisheries College, Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Jimei University, Xiamen, Fujian, 361021, China
| | - Yingxue Qin
- Fisheries College, Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Jimei University, Xiamen, Fujian, 361021, China
| | - Yongquan Su
- State Key Laboratory of Large Yellow Croaker Breeding, Ningde, Fujian, 352000, China
| | - Qingpi Yan
- Fisheries College, Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Jimei University, Xiamen, Fujian, 361021, China; State Key Laboratory of Large Yellow Croaker Breeding, Ningde, Fujian, 352000, China.
| |
Collapse
|
27
|
The Ethanolamine Permease EutH Promotes Vacuole Adaptation of Salmonella enterica and Listeria monocytogenes during Macrophage Infection. Infect Immun 2018. [PMID: 29531136 DOI: 10.1128/iai.00172-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ethanolamine is a ubiquitous and essential molecule within a host. Significantly, bacterial pathogens exploit ethanolamine during infection to promote growth and regulate virulence. The ethanolamine permease EutH is dispensable for growth in vitro under standard conditions, whereas EutH is required for ethanolamine utilization at low pH. These findings suggested a model in which EutH facilitates diffusion of ethanolamine into the bacterial cell in acidic environments. To date, the ecological significance of this model has not been thoroughly investigated, and the importance of EutH to bacterial growth under physiologically relevant conditions is not known. During infection, immune cells internalize invading bacteria within an acidic, nutrient-depleted vacuole called the phagosome. Here, we investigated the hypothesis that EutH promotes bacterial survival following phagocytosis. Our findings indicate that EutH is important for survival and replication of the facultative intracellular pathogens Salmonella enterica serovar Typhimurium and Listeria monocytogenes during prolonged or transient exposure to the phagosome, respectively. Furthermore, in agreement with EutH being important in the acidic environment, neutralization of the vacuole abolished the requirement for EutH. Significantly, consistent with a role for EutH in promoting intramacrophage survival, EutH was not required during S Typhimurium local intestinal infection but specifically conferred an advantage upon dissemination to peripheral organs. These findings reveal a physiologically relevant and conserved role for EutH in spatiotemporal niche adaptation during infection.
Collapse
|
28
|
Osborne SE, Brumell JH. Listeriolysin O: from bazooka to Swiss army knife. Philos Trans R Soc Lond B Biol Sci 2018. [PMID: 28630160 DOI: 10.1098/rstb.2016.0222] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Listeria monocytogenes (Lm) is a Gram-positive facultative intracellular pathogen. Infections in humans can lead to listeriosis, a systemic disease with a high mortality rate. One important mechanism of Lm dissemination involves cell-to-cell spread after bacteria have entered the cytosol of host cells. Listeriolysin O (LLO; encoded by the hly gene) is a virulence factor present in Lm that plays a central role in the cell-to-cell spread process. LLO is a member of the cholesterol-dependent cytolysin (CDC) family of toxins that were initially thought to promote disease largely by inducing cell death and tissue destruction-essentially acting like a 'bazooka'. This view was supported by structural studies showing CDCs can form large pores in membranes. However, it is now appreciated that LLO has many subtle activities during Lm infection of host cells, and many of these likely do not involve large pores, but rather small membrane perforations. It is also appreciated that membrane repair pathways of host cells play a major role in limiting membrane damage by LLO and other toxins. LLO is now thought to represent a 'Swiss army knife', a versatile tool that allows Lm to induce many membrane alterations and cellular responses that promote bacterial dissemination during infection.This article is part of the themed issue 'Membrane pores: from structure and assembly, to medicine and technology'.
Collapse
Affiliation(s)
- Suzanne E Osborne
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
| | - John H Brumell
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada M5G 1X8 .,Sickkids IBD Centre, Hospital for Sick Children, Toronto, ON, Canada M5G 1X8.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada M5S 1A8.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
29
|
The eIF2α Kinase Heme-Regulated Inhibitor Protects the Host from Infection by Regulating Intracellular Pathogen Trafficking. Infect Immun 2018; 86:IAI.00707-17. [PMID: 29311243 DOI: 10.1128/iai.00707-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/19/2017] [Indexed: 12/20/2022] Open
Abstract
The host employs both cell-autonomous and system-level responses to limit pathogen replication in the initial stages of infection. Previously, we reported that the eukaryotic initiation factor 2α (eIF2α) kinases heme-regulated inhibitor (HRI) and protein kinase R (PKR) control distinct cellular and immune-related activities in response to diverse bacterial pathogens. Specifically for Listeria monocytogenes, there was reduced translocation of the pathogen to the cytosolic compartment in HRI-deficient cells and consequently reduced loading of pathogen-derived antigens on major histocompatibility complex class I (MHC-I) complexes. Here we show that Hri-/- mice, as well as wild-type mice treated with an HRI inhibitor, are more susceptible to listeriosis. In the first few hours of L. monocytogenes infection, there was much greater pathogen proliferation in the liver of Hri-/- mice than in the liver of Hri+/+ mice. Further, there was a rapid increase of serum interleukin-6 (IL-6) levels in Hri+/+ mice in the first few hours of infection whereas the increase in IL-6 levels in Hri-/- mice was notably delayed. Consistent with these in vivo findings, the rate of listeriolysin O (LLO)-dependent pathogen efflux from infected Hri-/- macrophages and fibroblasts was significantly higher than the rate seen with infected Hri+/+ cells. Treatment of cells with an eIF2α kinase activator enhanced both the HRI-dependent and PKR-dependent infection phenotypes, further indicating the pharmacologically malleability of this signaling pathway. Collectively, these results suggest that HRI mediates the cellular confinement and killing of virulent L. monocytogenes in addition to promoting a system-level cytokine response and that both are required to limit pathogen replication during the first few hours of infection.
Collapse
|
30
|
You X, Liu L, Li X, Du H, Nie D, Zhang X, Tong H, Wu M, Gao Y, Liao Z. Immune response of interferon-γ-inducible lysosomal thiol reductase (GILT) from Chinese sturgeon (Acipenser sinensis) to microbial invasion and its antioxdative activity in lipopolysaccharides-treated mammalian dentritic cells. FISH & SHELLFISH IMMUNOLOGY 2018; 72:356-366. [PMID: 29133251 DOI: 10.1016/j.fsi.2017.11.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 06/07/2023]
Abstract
Interferon-γ-inducible lysosomal thiol reductase (GILT) plays an important role in the major histocompatibility complex-restricted antigen processing of endocytosed proteins via catalyzing the disulfide bond reduction in the endocytic pathway. Here, the cDNA of Chinese sturgeon (Acipenser sinensis) GILT (CsGILT) was cloned. It contained an open reading frame of 762 nucleotides encoding a protein of 254 amino acids with an estimated molecular weight of 28.1 kDa. The characteristic structural features, including a signature sequence CQHGX2ECX2NX4C, a CXXC motif, two potential N-glycosylation sites, and eight conserved cysteines were detected in the deduced amino acid sequence of CsGILT. CsGILT was widely expressed in Chinese sturgeon with the highest expression in the spleen, and CsGILT mRNA expression was significantly up-regulated when Chinese sturgeons were challenged with polyinosinic polycytidylic acid or Vibrio anguillarum. The recombinant CsGILT displayed obvious thiol reductase activity demonstrated by catalyzing the reduction of mouse IgG(H+L) by dithiothreitol into heavy chain and light chain. CsGILT also displayed significant antioxidant activity in mouse dentritic cells as indicated by its increasing GSH level and GSH/GSSG ratio, decreasing intracellular reactive oxygen species and nitric oxide levels and lipid peroxidation, as well as enhancing the activities of the antioxidative redox enzymes including catalase and superoxide dismutase. Our results suggested an important role for CsGILT in the immune response in Chinese sturgeon to pathogen invasion possibly via a conserved functional mechanism throughout vertebrate evolution, contributing to our understanding the immune biology and protection of Chinese sturgeon.
Collapse
Affiliation(s)
- Xiuling You
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Liu Liu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Xiuyu Li
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Hejun Du
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Institute of Chinese Sturgeon, China Three Gorges Corporation, Yichang 443100, China
| | - Dongsong Nie
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China
| | - Xingguo Zhang
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Haibing Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Mingjiang Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yitian Gao
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Zhiyong Liao
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
31
|
Uribe-Querol E, Rosales C. Control of Phagocytosis by Microbial Pathogens. Front Immunol 2017; 8:1368. [PMID: 29114249 PMCID: PMC5660709 DOI: 10.3389/fimmu.2017.01368] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022] Open
Abstract
Phagocytosis is a fundamental process of cells to capture and ingest foreign particles. Small unicellular organisms such as free-living amoeba use this process to acquire food. In pluricellular organisms, phagocytosis is a universal phenomenon that all cells are able to perform (including epithelial, endothelial, fibroblasts, etc.), but some specialized cells (such as neutrophils and macrophages) perform this very efficiently and were therefore named professional phagocytes by Rabinovitch. Cells use phagocytosis to capture and clear all particles larger than 0.5 µm, including pathogenic microorganisms and cellular debris. Phagocytosis involves a series of steps from recognition of the target particle, ingestion of it in a phagosome (phagocytic vacuole), maturation of this phagosome into a phagolysosome, to the final destruction of the ingested particle in the robust antimicrobial environment of the phagolysosome. For the most part, phagocytosis is an efficient process that eliminates invading pathogens and helps maintaining homeostasis. However, several pathogens have also evolved different strategies to prevent phagocytosis from proceeding in a normal way. These pathogens have a clear advantage to perpetuate the infection and continue their replication. Here, we present an overview of the phagocytic process with emphasis on the antimicrobial elements professional phagocytes use. We also summarize the current knowledge on the microbial strategies different pathogens use to prevent phagocytosis either at the level of ingestion, phagosome formation, and maturation, and even complete escape from phagosomes.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
32
|
Anderson CJ, Kendall MM. Salmonella enterica Serovar Typhimurium Strategies for Host Adaptation. Front Microbiol 2017; 8:1983. [PMID: 29075247 PMCID: PMC5643478 DOI: 10.3389/fmicb.2017.01983] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022] Open
Abstract
Bacterial pathogens must sense and respond to newly encountered host environments to regulate the expression of critical virulence factors that allow for niche adaptation and successful colonization. Among bacterial pathogens, non-typhoidal serovars of Salmonella enterica, such as serovar Typhimurium (S. Tm), are a primary cause of foodborne illnesses that lead to hospitalizations and deaths worldwide. S. Tm causes acute inflammatory diarrhea that can progress to invasive systemic disease in susceptible patients. The gastrointestinal tract and intramacrophage environments are two critically important niches during S. Tm infection, and each presents unique challenges to limit S. Tm growth. The intestinal tract is home to billions of commensal microbes, termed the microbiota, which limits the amount of available nutrients for invading pathogens such as S. Tm. Therefore, S. Tm encodes strategies to manipulate the commensal population and side-step this nutritional competition. During subsequent stages of disease, S. Tm resists host immune cell mechanisms of killing. Host cells use antimicrobial peptides, acidification of vacuoles, and nutrient limitation to kill phagocytosed microbes, and yet S. Tm is able to subvert these defense systems. In this review, we discuss recently described molecular mechanisms that S. Tm uses to outcompete the resident microbiota within the gastrointestinal tract. S. Tm directly eliminates close competitors via bacterial cell-to-cell contact as well as by stimulating a host immune response to eliminate specific members of the microbiota. Additionally, S. Tm tightly regulates the expression of key virulence factors that enable S. Tm to withstand host immune defenses within macrophages. Additionally, we highlight the chemical and physical signals that S. Tm senses as cues to adapt to each of these environments. These strategies ultimately allow S. Tm to successfully adapt to these two disparate host environments. It is critical to better understand bacterial adaptation strategies because disruption of these pathways and mechanisms, especially those shared by multiple pathogens, may provide novel therapeutic intervention strategies.
Collapse
Affiliation(s)
- Christopher J Anderson
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine,, Charlottesville, VA, United States
| | - Melissa M Kendall
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine,, Charlottesville, VA, United States
| |
Collapse
|
33
|
Lou J, Li X, Huang W, Liang J, Zheng M, Xu T, Lyu J, Li D, Xu Q, Jin X, Fu G, Wang D, Lu L. SNX10 promotes phagosome maturation in macrophages and protects mice against Listeria monocytogenes infection. Oncotarget 2017; 8:53935-53947. [PMID: 28903313 PMCID: PMC5589552 DOI: 10.18632/oncotarget.19644] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/13/2017] [Indexed: 12/29/2022] Open
Abstract
Listeria monocytogenes (L. monocytogenes), which is a facultative intracellular bacterial pathogen that causes listeriosis, is widely used to study the mammalian immune response to infection. After phagocytosis by professional phagocytes, L. monocytogenes is initially contained within phagosomes, which mature into phagolysosomes, where the bacteria are degraded. Although phagocytosis and subsequent phagosome maturation is essential for the clearance of infectious microbial pathogens, the underlying regulatory mechanisms are still unclear. SNX10 (Sorting nexin 10) has the simplest structure of the SNX family and has been reported to regulate endosomal morphology, which might be crucial for macrophage function, including phagocytosis and digestion of pathogens, inflammatory response, and antigen presentation. Our results showed that SNX10 expression was upregulated following L. monocytogenes infection in macrophages. It was also revealed that SNX10 promoted phagosome maturation by recruiting the Mon1-Ccz1 complex to endosomes and phagosomes. As a result, SNX10 deficiency decreased the bacterial killing ability of macrophages, and SNX10-deficient mice showed increased susceptibility to L. monocytogenes infection in vivo. Thus, this study revealed an essential role of SNX10 in controlling bacterial infection.
Collapse
Affiliation(s)
- Jun Lou
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiawei Li
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Huang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Liang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingzhu Zheng
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Xu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Lyu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Li
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Xu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuexiao Jin
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Guotong Fu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Linrong Lu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Lee BC, Lee SG, Choo MK, Kim JH, Lee HM, Kim S, Fomenko DE, Kim HY, Park JM, Gladyshev VN. Selenoprotein MsrB1 promotes anti-inflammatory cytokine gene expression in macrophages and controls immune response in vivo. Sci Rep 2017; 7:5119. [PMID: 28698597 PMCID: PMC5506048 DOI: 10.1038/s41598-017-05230-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022] Open
Abstract
Post-translational redox modification of methionine residues often triggers a change in protein function. Emerging evidence points to this reversible protein modification being an important regulatory mechanism under various physiological conditions. Reduction of oxidized methionine residues is catalyzed by methionine sulfoxide reductases (Msrs). Here, we show that one of these enzymes, a selenium-containing MsrB1, is highly expressed in immune-activated macrophages and contributes to shaping cellular and organismal immune responses. In particular, lipopolysaccharide (LPS) induces expression of MsrB1, but not other Msrs. Genetic ablation of MsrB1 did not preclude LPS-induced intracellular signaling in macrophages, but resulted in attenuated induction of anti-inflammatory cytokines, such as interleukin (IL)-10 and the IL-1 receptor antagonist. This anomaly was associated with excessive pro-inflammatory cytokine production as well as an increase in acute tissue inflammation in mice. Together, our findings suggest that MsrB1 controls immune responses by promoting anti-inflammatory cytokine expression in macrophages. MsrB1-dependent reduction of oxidized methionine in proteins may be a heretofore unrecognized regulatory event underlying immunity and inflammatory disease, and a novel target for clinical applications.
Collapse
Affiliation(s)
- Byung Cheon Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea.
| | - Sang-Goo Lee
- Division of Genetics, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Min-Kyung Choo
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ji Hyung Kim
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Hae Min Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Sorah Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Dmitri E Fomenko
- Department of Biochemistry and Redox Biology Center, University of Nebraska, Lincoln, NE, 68588, USA
| | - Hwa-Young Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu, 42415, South Korea
| | - Jin Mo Park
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Cheng C, Dong Z, Han X, Wang H, Jiang L, Sun J, Yang Y, Ma T, Shao C, Wang X, Chen Z, Fang W, Freitag NE, Huang H, Song H. Thioredoxin A Is Essential for Motility and Contributes to Host Infection of Listeria monocytogenes via Redox Interactions. Front Cell Infect Microbiol 2017; 7:287. [PMID: 28702378 PMCID: PMC5487381 DOI: 10.3389/fcimb.2017.00287] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/12/2017] [Indexed: 12/17/2022] Open
Abstract
Microbes employ the thioredoxin system to defend against oxidative stress and ensure correct disulfide bonding to maintain protein function. Listeria monocytogenes has been shown to encode a putative thioredoxin, TrxA, but its biological roles and underlying mechanisms remain unknown. Here, we showed that expression of L. monocytogenes TrxA is significantly induced in bacteria treated with the thiol-specific oxidizing agent, diamide. Deletion of trxA markedly compromised tolerance of the pathogen to diamide, and mainly impaired early stages of infection in human intestinal epithelial Caco-2 cells. In addition, most trxA mutant bacteria were not associated with polymerized actin, and the rare bacteria that were associated with polymerized actin displayed very short tails or clouds during infection. Deletion or constitutive overexpression of TrxA, which was regulated by SigH, severely attenuated the virulence of the pathogen. Transcriptome analysis of L. monocytogenes revealed over 270 genes that were differentially transcribed in the ΔtrxA mutant compared to the wild-type, especially for the virulence-associated genes plcA, mpl, hly, actA, and plcB. Particularly, deletion of TrxA completely reduced LLO expression, and thereby led to a thoroughly impaired hemolytic activity. Expression of these virulence factors are positively regulated by the master regulator PrfA that was found here to use TrxA to maintain its reduced forms for activation. Interestingly, the trxA deletion mutant completely lacked flagella and was non-motile. We further confirmed that this deficiency is attributable to TrxA in maintaining the reduced intracellular monomer status of MogR, the key regulator for flagellar formation, to ensure correct dimerization. In summary, we demonstrated for the first time that L. monocytogenes thioredoxin A as a vital cellular reductase is essential for maintaining a highly reducing environment in the bacterial cytosol, which provides a favorable condition for protein folding and activation, and therefore contributes to bacterial virulence and motility.
Collapse
Affiliation(s)
- Changyong Cheng
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Zhimei Dong
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Xiao Han
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Hang Wang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Li Jiang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Jing Sun
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Yongchun Yang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Tiantian Ma
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Chunyan Shao
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Xiaodu Wang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Zhongwei Chen
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| | - Weihuan Fang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China.,Zhejiang University Institute of Preventive Veterinary Medicine and Zhejiang Provincial Key Laboratory of Preventive Veterinary MedicineHangzhou, China
| | - Nancy E Freitag
- Department of Microbiology and Immunology, University of Illinois at ChicagoChicago, IL, United States
| | - Huarong Huang
- Institute of Developmental and Regenerative Biology, College of Biological and Environmental Science, Hangzhou Normal UniversityZhejiang, China
| | - Houhui Song
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology of Zhejiang A&F UniversityLin'an, China
| |
Collapse
|
36
|
Johnston DGW, Kearney J, Zasłona Z, Williams MA, O'Neill LAJ, Corr SC. MicroRNA-21 Limits Uptake of Listeria monocytogenes by Macrophages to Reduce the Intracellular Niche and Control Infection. Front Cell Infect Microbiol 2017; 7:201. [PMID: 28589100 PMCID: PMC5440467 DOI: 10.3389/fcimb.2017.00201] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/05/2017] [Indexed: 12/19/2022] Open
Abstract
MiRNAs are important post-transcriptional regulators of gene expression. MiRNA expression is a crucial part of host responses to bacterial infection, however there is limited knowledge of their impact on the outcome of infections. We investigated the influence of miR-21 on macrophage responses during infection with Listeria monocytogenes, which establishes an intracellular niche within macrophages. MiR-21 is induced following infection of bone marrow-derived macrophages (BMDMs) with Listeria. MiR-21−/− macrophages display an increased bacterial burden with Listeria at 30 min and 2 h post-infection. This phenotype was reversed by the addition of synthetic miR-21 mimics to the system. To assess the immune response of wildtype (WT) and miR-21−/− macrophages, BMDMs were treated with bacterial LPS or infected with Listeria. There was no difference in IL-10 and IL-6 between WT and miR-21−/− BMDMs in response to LPS or Listeria. TNF-α was increased in miR-21−/− BMDMs stimulated with LPS or Listeria compared to WT macrophages. We next assessed the production of nitric oxide (NO), a key bactericidal factor in Listeria infection. There was no significant difference in NO production between WT and miR-21−/− cells, indicating that the increased bacterial burden may not be due to impaired killing. As the increased bacterial load was observed early following infection (30 min), we questioned whether this is due to differences in uptake of Listeria by WT and miR-21−/− macrophages. We show that miR-21-deficiency enhances uptake of FITC-dextran and FITC-Escherichia coli bioparticles by macrophages. The previously observed Listeria burden phenotype was ablated by pre-treatment of cells with the actin polymerization inhibitor cytochalasin-D. From analysis of miR-21 targets, we selected the pro-phagocytic regulators myristoylated alanine-rich C-kinase substrate (MARCKS) and Ras homolog gene family, member B (RhoB) for further investigation. MARCKS and RhoB are increased in miR-21−/− BMDMs, correlating with increased uptake of Listeria. Finally, intra-peritoneal infection of mice with Listeria led to increased bacterial burden in livers of miR-21−/− mice compared to WT mice. These findings suggest a possible role for miR-21 in regulation of phagocytosis during infection, potentially by repression of MARCKS and RhoB, thus serving to limit the availability of the intracellular niche of pathogens like L. monocytogenes.
Collapse
Affiliation(s)
- Daniel G W Johnston
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinDublin, Ireland.,Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College DublinDublin, Ireland
| | - Jay Kearney
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinDublin, Ireland
| | - Zbigniew Zasłona
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinDublin, Ireland
| | - Michelle A Williams
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College DublinDublin, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College DublinDublin, Ireland
| | - Sinéad C Corr
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College DublinDublin, Ireland
| |
Collapse
|
37
|
Activity of the Pore-Forming Virulence Factor Listeriolysin O Is Reversibly Inhibited by Naturally Occurring S-Glutathionylation. Infect Immun 2017; 85:IAI.00959-16. [PMID: 28138025 DOI: 10.1128/iai.00959-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/25/2017] [Indexed: 11/20/2022] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) represent a family of homologous pore-forming proteins secreted by many Gram-positive bacterial pathogens. CDCs mediate membrane binding partly through a conserved C-terminal undecapeptide, which contains a single cysteine residue. While mutational changes to other residues in the undecapeptide typically have severe effects, mutation of the cysteine residue to alanine has minor effects on overall protein function. Thus, the role of this highly conserved reactive cysteine residue remains largely unknown. We report here that the CDC listeriolysin O (LLO), secreted by the facultative intracellular pathogen Listeria monocytogenes, was posttranslationally modified by S-glutathionylation at this conserved cysteine residue and that either endogenously synthesized or exogenously added glutathione was sufficient to form this modification. When recapitulated with purified protein in vitro, this modification completely ablated the activity of LLO, and this inhibitory effect was fully reversible by treatment with reducing agents. A cysteine-to-alanine mutation in LLO rendered the protein completely resistant to inactivation by S-glutathionylation, and a mutant expressing this mutation retained full hemolytic activity. A mutant strain of L. monocytogenes expressing the cysteine-to-alanine variant of LLO was able to infect and replicate within bone marrow-derived macrophages indistinguishably from the wild type in vitro, yet it was attenuated 4- to 6-fold in a competitive murine infection model in vivo This study suggests that S-glutathionylation may represent a mechanism by which CDC-family proteins are posttranslationally modified and regulated and help explain an evolutionary pressure to retain the highly conserved undecapeptide cysteine.
Collapse
|
38
|
Quereda JJ, Sachse M, Balestrino D, Grenier T, Fredlund J, Danckaert A, Aulner N, Shorte S, Enninga J, Cossart P, Pizarro-Cerdá J. Assessing Vacuolar Escape of Listeria Monocytogenes. Methods Mol Biol 2017; 1535:173-195. [PMID: 27914079 DOI: 10.1007/978-1-4939-6673-8_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Listeria monocytogenes is a bacterial pathogen which invades and multiplies within non-professional phagocytes. Signaling cascades involved in cellular entry have been extensively analyzed, but the events leading to vacuolar escape remain less clear. In this chapter, we detail a microscopy FRET-based assay which allows quantitatively measuring L. monocytogenes infection and escape from its internalization vacuole, as well as a correlative light/electron microscopy method to investigate the morphological features of the vacuolar compartments containing L. monocytogenes.
Collapse
Affiliation(s)
- Juan J Quereda
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 25, rue du Docteur Roux, Paris, 75015, France
- INSERM, U604, Paris, 75015, France
- INRA, USC2020, Paris, 75015, France
| | - Martin Sachse
- Institut Pasteur, Ultrapole-CITech, Paris, 75015, France
| | - Damien Balestrino
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 25, rue du Docteur Roux, Paris, 75015, France
- INSERM, U604, Paris, 75015, France
- INRA, USC2020, Paris, 75015, France
- UMR CNRS 6023, Laboratoire Microorganismes: Génome Environnement, Université d'Auvergne, Clermont-Ferrand, 63000, France
| | - Théodore Grenier
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 25, rue du Docteur Roux, Paris, 75015, France
- INSERM, U604, Paris, 75015, France
- INRA, USC2020, Paris, 75015, France
| | - Jennifer Fredlund
- Institut Pasteur, Unité Dynamique des Interactions Hôte-Pathogène, Paris, 75015, France
| | - Anne Danckaert
- Institut Pasteur, Imagopole-CITech, Paris, 75015, France
| | | | - Spencer Shorte
- Institut Pasteur, Imagopole-CITech, Paris, 75015, France
| | - Jost Enninga
- Institut Pasteur, Unité Dynamique des Interactions Hôte-Pathogène, Paris, 75015, France
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 25, rue du Docteur Roux, Paris, 75015, France
- INSERM, U604, Paris, 75015, France
- INRA, USC2020, Paris, 75015, France
| | - Javier Pizarro-Cerdá
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 25, rue du Docteur Roux, Paris, 75015, France.
- INSERM, U604, Paris, 75015, France.
- INRA, USC2020, Paris, 75015, France.
| |
Collapse
|
39
|
Mu H, Niu H, Wang D, Sun F, Sun Y, Duan J. Chitosan conjugation enables intracellular bacteria susceptible to aminoglycoside antibiotic. Glycobiology 2016; 26:1190-1197. [DOI: 10.1093/glycob/cww079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 07/20/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022] Open
|
40
|
Manipulation of host membranes by the bacterial pathogens Listeria, Francisella, Shigella and Yersinia. Semin Cell Dev Biol 2016; 60:155-167. [PMID: 27448494 PMCID: PMC7082150 DOI: 10.1016/j.semcdb.2016.07.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 01/07/2023]
Abstract
Bacterial pathogens display an impressive arsenal of molecular mechanisms that allow survival in diverse host niches. Subversion of plasma membrane and cytoskeletal functions are common themes associated to infection by both extracellular and intracellular pathogens. Moreover, intracellular pathogens modify the structure/stability of their membrane-bound compartments and escape degradation from phagocytic or autophagic pathways. Here, we review the manipulation of host membranes by Listeria monocytogenes, Francisella tularensis, Shigella flexneri and Yersinia spp. These four bacterial model pathogens exemplify generalized strategies as well as specific features observed during bacterial infection processes.
Collapse
|
41
|
Shchennikova AV, Beletsky AV, Shulga OA, Mazur AM, Prokhortchouk EB, Kochieva EZ, Ravin NV, Skryabin KG. Deep-sequence profiling of miRNAs and their target prediction in Monotropa hypopitys. PLANT MOLECULAR BIOLOGY 2016; 91:441-458. [PMID: 27097902 DOI: 10.1007/s11103-016-0478-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/04/2016] [Indexed: 06/05/2023]
Abstract
Myco-heterotroph Monotropa hypopitys is a widely spread perennial herb used to study symbiotic interactions and physiological mechanisms underlying the development of non-photosynthetic plant. Here, we performed, for the first time, transcriptome-wide characterization of M. hypopitys miRNA profile using high throughput Illumina sequencing. As a result of small RNA library sequencing and bioinformatic analysis, we identified 55 members belonging to 40 families of known miRNAs and 17 putative novel miRNAs unique for M. hypopitys. Computational screening revealed 206 potential mRNA targets for known miRNAs and 31 potential mRNA targets for novel miRNAs. The predicted target genes were described in Gene Ontology terms and were found to be involved in a broad range of metabolic and regulatory pathways. The identification of novel M. hypopitys-specific miRNAs, some with few target genes and low abundances, suggests their recent evolutionary origin and participation in highly specialized regulatory mechanisms fundamental for non-photosynthetic biology of M. hypopitys. This global analysis of miRNAs and their potential targets in M. hypopitys provides a framework for further investigation of miRNA role in the evolution and establishment of non-photosynthetic myco-heterotrophs.
Collapse
Affiliation(s)
- Anna V Shchennikova
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071
| | - Alexey V Beletsky
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071
| | - Olga A Shulga
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071
| | - Alexander M Mazur
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071
| | - Egor B Prokhortchouk
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071
| | - Elena Z Kochieva
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071
| | - Nikolay V Ravin
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071
| | - Konstantin G Skryabin
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, Moscow, Russia, 119071.
| |
Collapse
|
42
|
Mitchell G, Chen C, Portnoy DA. Strategies Used by Bacteria to Grow in Macrophages. Microbiol Spectr 2016; 4:10.1128/microbiolspec.MCHD-0012-2015. [PMID: 27337444 PMCID: PMC4922531 DOI: 10.1128/microbiolspec.mchd-0012-2015] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Indexed: 12/24/2022] Open
Abstract
Intracellular bacteria are often clinically relevant pathogens that infect virtually every cell type found in host organisms. However, myeloid cells, especially macrophages, constitute the primary cells targeted by most species of intracellular bacteria. Paradoxically, macrophages possess an extensive antimicrobial arsenal and are efficient at killing microbes. In addition to their ability to detect and signal the presence of pathogens, macrophages sequester and digest microorganisms using the phagolysosomal and autophagy pathways or, ultimately, eliminate themselves through the induction of programmed cell death. Consequently, intracellular bacteria influence numerous host processes and deploy sophisticated strategies to replicate within these host cells. Although most intracellular bacteria have a unique intracellular life cycle, these pathogens are broadly categorized into intravacuolar and cytosolic bacteria. Following phagocytosis, intravacuolar bacteria reside in the host endomembrane system and, to some extent, are protected from the host cytosolic innate immune defenses. However, the intravacuolar lifestyle requires the generation and maintenance of unique specialized bacteria-containing vacuoles and involves a complex network of host-pathogen interactions. Conversely, cytosolic bacteria escape the phagolysosomal pathway and thrive in the nutrient-rich cytosol despite the presence of host cell-autonomous defenses. The understanding of host-pathogen interactions involved in the pathogenesis of intracellular bacteria will continue to provide mechanistic insights into basic cellular processes and may lead to the discovery of novel therapeutics targeting infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Gabriel Mitchell
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chen Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
43
|
Mellouk N, Enninga J. Cytosolic Access of Intracellular Bacterial Pathogens: The Shigella Paradigm. Front Cell Infect Microbiol 2016; 6:35. [PMID: 27092296 PMCID: PMC4820437 DOI: 10.3389/fcimb.2016.00035] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/14/2016] [Indexed: 12/20/2022] Open
Abstract
Shigella is a Gram-negative bacterial pathogen, which causes bacillary dysentery in humans. A crucial step of Shigella infection is its invasion of epithelial cells. Using a type III secretion system, Shigella injects several bacterial effectors ultimately leading to bacterial internalization within a vacuole. Then, Shigella escapes rapidly from the vacuole, it replicates within the cytosol and spreads from cell-to-cell. The molecular mechanism of vacuolar rupture used by Shigella has been studied in some detail during the recent years and new paradigms are emerging about the underlying molecular events. For decades, bacterial effector proteins were portrayed as main actors inducing vacuolar rupture. This includes the effector/translocators IpaB and IpaC. More recently, this has been challenged and an implication of the host cell in the process of vacuolar rupture has been put forward. This includes the bacterial subversion of host trafficking regulators, such as the Rab GTPase Rab11. The involvement of the host in determining bacterial vacuolar integrity has also been found for other bacterial pathogens, particularly for Salmonella. Here, we will discuss our current view of host factor and pathogen effector implications during Shigella vacuolar rupture and the steps leading to it.
Collapse
Affiliation(s)
- Nora Mellouk
- Dynamics of Host-Pathogen Interactions Unit, Institut Pasteur Paris, France
| | - Jost Enninga
- Dynamics of Host-Pathogen Interactions Unit, Institut Pasteur Paris, France
| |
Collapse
|
44
|
Kaufmann SH, Dorhoi A. Molecular Determinants in Phagocyte-Bacteria Interactions. Immunity 2016; 44:476-491. [DOI: 10.1016/j.immuni.2016.02.014] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 01/28/2016] [Accepted: 02/17/2016] [Indexed: 12/24/2022]
|
45
|
Barikbin P, Sallmon H, Hüseman D, Sarioglu N, Weichert A, von Weizsäcker K, Bührer C, Koehne P. Clinical, Laboratory, and Placental Findings in Perinatal Listeriosis. Fetal Pediatr Pathol 2016; 35:307-314. [PMID: 27212240 DOI: 10.1080/15513815.2016.1179822] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Clinical, laboratory, and placental manifestations of perinatal listeriosis are highly variable. Herein, we retrospectively analyzed all patients treated for neonatal listeriosis at the Charité University Medical Center in Berlin, Germany, 1999-2013. A total of 16 cases were identified. In 14 patients listeriosis was confirmed in neonatal specimens, while in two only the placenta tested positive. Elevated C-reactive protein and/or interleukin-6 levels were only inconsistently found, while a marked white blood cell left shift was present in all infants, if available. All but one infant manifested symptoms on the first day of life. Most patients required respiratory support, while none developed meningoencephalitis as evidenced by clinical or cerebrospinal fluid findings. Two patients died, all other patients survived without sequelae. In conclusion, perinatal listeriosis is still associated with significant morbidity and mortality. Clinical and laboratory findings are highly heterogeneous, but extreme leukocyte left shift seems to be a common feature.
Collapse
Affiliation(s)
- Payman Barikbin
- a Department of Neonatology , Charité University Medical Center , Berlin , Germany
| | - Hannes Sallmon
- a Department of Neonatology , Charité University Medical Center , Berlin , Germany
| | - Dieter Hüseman
- a Department of Neonatology , Charité University Medical Center , Berlin , Germany
| | - Nanette Sarioglu
- b Department of Pediatric Pathology and Placentology , Charité University Medical Center , Berlin , Germany
| | - Alexander Weichert
- c Department of Obstetrics , Charité University Medical Center , Berlin , Germany
| | | | - Christoph Bührer
- a Department of Neonatology , Charité University Medical Center , Berlin , Germany
| | - Petra Koehne
- a Department of Neonatology , Charité University Medical Center , Berlin , Germany
| |
Collapse
|
46
|
A Dual Microscopy-Based Assay To Assess Listeria monocytogenes Cellular Entry and Vacuolar Escape. Appl Environ Microbiol 2015; 82:211-7. [PMID: 26497455 DOI: 10.1128/aem.02302-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 10/14/2015] [Indexed: 01/11/2023] Open
Abstract
Listeria monocytogenes is a Gram-positive bacterium and a facultative intracellular pathogen that invades mammalian cells, disrupts its internalization vacuole, and proliferates in the host cell cytoplasm. Here, we describe a novel image-based microscopy assay that allows discrimination between cellular entry and vacuolar escape, enabling high-content screening to identify factors specifically involved in these two steps. We first generated L. monocytogenes and Listeria innocua strains expressing a β-lactamase covalently attached to the bacterial cell wall. These strains were then incubated with HeLa cells containing the Förster resonance energy transfer (FRET) probe CCF4 in their cytoplasm. The CCF4 probe was cleaved by the bacterial surface β-lactamase only in cells inoculated with L. monocytogenes but not those inoculated with L. innocua, thereby demonstrating bacterial access to the host cytoplasm. Subsequently, we performed differential immunofluorescence staining to distinguish extracellular versus total bacterial populations in samples that were also analyzed by the FRET-based assay. With this two-step analysis, bacterial entry can be distinguished from vacuolar rupture in a single experiment. Our novel approach represents a powerful tool for identifying factors that determine the intracellular niche of L. monocytogenes.
Collapse
|
47
|
Huang WS, Duan LP, Huang B, Zhou LH, Liang Y, Tu CL, Zhang FF, Nie P, Wang T. Identification of three IFN-γ inducible lysosomal thiol reductase ( GILT )-like genes in mud crab Scylla paramamosain with distinct gene organizations and patterns of expression. Gene 2015; 570:78-88. [DOI: 10.1016/j.gene.2015.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 05/17/2015] [Accepted: 06/02/2015] [Indexed: 12/22/2022]
|
48
|
Rausch MP, Hastings KT. Diverse cellular and organismal functions of the lysosomal thiol reductase GILT. Mol Immunol 2015; 68:124-8. [PMID: 26116226 DOI: 10.1016/j.molimm.2015.06.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/02/2015] [Indexed: 01/21/2023]
Abstract
Gamma-interferon-inducible lysosomal thiol reductase (GILT) is the only enzyme known to catalyze disulfide bond reduction in the endocytic pathway. GILT facilitates the presentation of a subset of epitopes from disulfide bond-containing antigens. Enhanced presentation of MHC class II-restricted epitopes alters central tolerance and modulates CD4+ T cell-mediated autoimmunity. Improved cross-presentation of viral epitopes results in improved cross-priming of viral-specific CD8+ T cells. GILT regulates the cellular redox state. In GILT-/- cells, there is a shift from the reduced to the oxidized form of glutathione, resulting in mitochondrial autophagy, decreased superoxide dismutase 2, and elevated superoxide levels. GILT expression diminishes cellular activation, including decreased phosphorylated ERK1/2, and decreases cellular proliferation. GILT enhances the activity of bacterial hemolysins, such as listeriolysin O, and increases bacterial replication and infection. GILT expression in cancer cells is associated with improved patient survival. These diverse roles of GILT are discussed.
Collapse
Affiliation(s)
- Matthew P Rausch
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, USA; University of Arizona Cancer Center, AZ, USA
| | - Karen Taraszka Hastings
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, USA; University of Arizona Cancer Center, AZ, USA; Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
49
|
Mellouk N, Weiner A, Aulner N, Schmitt C, Elbaum M, Shorte SL, Danckaert A, Enninga J. Shigella subverts the host recycling compartment to rupture its vacuole. Cell Host Microbe 2015; 16:517-30. [PMID: 25299335 DOI: 10.1016/j.chom.2014.09.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/21/2014] [Accepted: 08/12/2014] [Indexed: 01/01/2023]
Abstract
Shigella enters epithlial cells via internalization into a vacuole. Subsequent vacuolar membrane rupture allows bacterial escape into the cytosol for replication and cell-to-cell spread. Bacterial effectors such as IpgD, a PI(4,5)P2 phosphatase that generates PI(5)P and alters host actin, facilitate this internalization. Here, we identify host proteins involved in Shigella uptake and vacuolar membrane rupture by high-content siRNA screening and subsequently focus on Rab11, a constituent of the recycling compartment. Rab11-positive vesicles are recruited to the invasion site before vacuolar rupture, and Rab11 knockdown dramatically decreases vacuolar membrane rupture. Additionally, Rab11 recruitment is absent and vacuolar rupture is delayed in the ipgD mutant that does not dephosphorylate PI(4,5)P₂ into PI(5)P. Ultrastructural analyses of Rab11-positive vesicles further reveal that ipgD mutant-containing vacuoles become confined in actin structures that likely contribute to delayed vacular rupture. These findings provide insight into the underlying molecular mechanism of vacuole progression and rupture during Shigella invasion.
Collapse
Affiliation(s)
- Nora Mellouk
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, 25 Rue du Dr. Roux, 75724 Paris, France
| | - Allon Weiner
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, 25 Rue du Dr. Roux, 75724 Paris, France
| | - Nathalie Aulner
- Institut Pasteur, Imagopole, 28 Rue du Dr. Roux, 75724 Paris, France
| | - Christine Schmitt
- Institut Pasteur, Imagopole, 28 Rue du Dr. Roux, 75724 Paris, France
| | - Michael Elbaum
- Department of Materials and Interfaces, Weizmann Institute of Sciences, 234 Herzl Street, Rehovot 76100, Israel
| | - Spencer L Shorte
- Institut Pasteur, Imagopole, 28 Rue du Dr. Roux, 75724 Paris, France
| | - Anne Danckaert
- Institut Pasteur, Imagopole, 28 Rue du Dr. Roux, 75724 Paris, France.
| | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, 25 Rue du Dr. Roux, 75724 Paris, France.
| |
Collapse
|
50
|
Ren C, Chen T, Jiang X, Luo X, Wang Y, Hu C. The first echinoderm gamma-interferon-inducible lysosomal thiol reductase (GILT) identified from sea cucumber (Stichopus monotuberculatus). FISH & SHELLFISH IMMUNOLOGY 2015; 42:41-49. [PMID: 25449705 DOI: 10.1016/j.fsi.2014.10.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/23/2014] [Accepted: 10/17/2014] [Indexed: 06/04/2023]
Abstract
Gamma-interferon-inducible lysosomal thiol reductase (GILT) has been described as a key enzyme that facilitating the processing and presentation of major histocompatibility complex class II-restricted antigen in mammals. In this study, the first echinoderm GILT named StmGILT was identified from sea cucumber (Stichopus monotuberculatus). The StmGILT cDNA is 1529 bp in length, containing a 5'-untranslated region (UTR) of 87 bp, a 3'-UTR of 674 bp and an open reading frame (ORF) of 768 bp that encoding a protein of 255 amino acids with a deduced molecular weight of 27.82 kDa and a predicted isoelectric point of 4.73. The putative StmGILT protein possesses all the main characteristics of known GILT proteins, including a signature sequence, a reductase active site CXXC, twelve conserved cysteines, and two potential N-linked glycosylation sites. For the gene structure, StmGILT contains four exons separated by three introns. In the promoter region of StmGILT gene, an NF-κB binding site and an IFN-γ activation site were found. The thiol reductase activity of recombinant StmGILT protein was also demonstrated in this study. In addition, the highest level of mRNA expression was noticed in coelomocytes of S. monotuberculatus. In in vitro experiments performed in coelomocytes, the expression of StmGILT mRNA was significantly up-regulated by lipopolysaccharides (LPS), inactivated bacteria or polyriboinosinic polyribocytidylic acid [poly (I:C)] challenge, suggested that the sea cucumber GILT might play critical roles in the innate immune defending against bacterial and viral infections.
Collapse
Affiliation(s)
- Chunhua Ren
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Ting Chen
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Xiao Jiang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Xing Luo
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Yanhong Wang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Chaoqun Hu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| |
Collapse
|