1
|
Woida PJ, Lamason RL. Pathogen-induced rerouting of host membrane trafficking. Curr Opin Cell Biol 2025; 94:102520. [PMID: 40262416 DOI: 10.1016/j.ceb.2025.102520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
Eukaryotic cell membranes are protective barriers that precisely control cargo import, trafficking, and export. In defiance of this control, intracellular bacterial pathogens forcefully invade host cells and establish intracellular niches. These pathogens require remarkable membrane remodeling events to support their large size, and a significant amount of work has examined how these pathogens co-opt cytoskeleton dynamics to remodel host membranes. Until recently, less attention was given to where the membranes came from to support remodeling around the pathogens at each stage of infection. In this review, we highlight recent examples of how bacterial pathogens reroute membrane trafficking to provide the membranes needed during invasion, intracellular growth, and eventual dissemination through host tissues. The examples discussed underscore emerging themes and areas for continued investigation rather than provide a survey of the entire field. We hope that highlighting these open questions will inspire researchers across disciplines to recognize the importance of pathogens as tools to understand both mechanisms of bacterial virulence and membrane trafficking.
Collapse
Affiliation(s)
- Patrick J Woida
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rebecca L Lamason
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
2
|
Khan ZA, Song SS, Xu H, Ahmad M, Wang A, Abdullah A, Jiang L, Ding X. Elimination of intracellular microbes using drug combination therapy and unveiling survival mechanism of host cells upon microbial invasion. Int J Antimicrob Agents 2025; 65:107471. [PMID: 39986398 DOI: 10.1016/j.ijantimicag.2025.107471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 01/16/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Intracellular microbes are actively present in various tumor types in low biomass and play a major role in metastasis. Eliminating intracellular microbes on a cellular level with precision remains a challenge. To address this issue, we designed a screening pipeline to characterize intracellular microbes and their interaction with host cells. We used host and microbial in vitro lab-based constant and reproducible model, host as (mammalian cancer HeLa), and microbial strain as (Escherichia coli 25922). To study the pharmacological impact on intracellular bacterial load, we used antibiotics (ampicillin, roxithromycin, and ciprofloxacin) and chemotherapy drugs (doxorubicin and cisplatin) as external stimuli for both host and microbes. We found that increasing pharmacological stress does not increase microbial load inside the host cells. Eliminations of intracellular bacteria was done by using permutation orthogonal arrays (POA), whereby we acquired optimal drug combination in particular sequence of drugs, which reduced 90%-95% of the intracellular microbial load. Proteomic analysis revealed that upon invasion of Escherichia coli 25922, HeLa cells enriched ATP production pathways to activate intermediate filaments, which should be investigated closely via in vivo models.
Collapse
Affiliation(s)
- Zara Ahmad Khan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China
| | - Sha-Sha Song
- Pathology Department, Yantai Fushan People's Hospital, Yantai, China
| | - Hongquan Xu
- Department of Statistics and Data Science, University of California, Los Angeles, California, USA
| | - Mashaal Ahmad
- Department of Anatomy, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Aiting Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China
| | - Aynur Abdullah
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Pathology, Wenling First People's Hospital, Wenling City, Zhejiang Province, China.
| |
Collapse
|
3
|
Marques PH, Rodrigues TCV, Santos EH, Bleicher L, Aburjaile FF, Martins FS, Oliveira CJF, Azevedo V, Tiwari S, Soares S. Design of a multi-epitope vaccine (vme-VAC/MST-1) against cholera and vibriosis based on reverse vaccinology and immunoinformatics approaches. J Biomol Struct Dyn 2025; 43:1788-1803. [PMID: 38112302 DOI: 10.1080/07391102.2023.2293256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023]
Abstract
Vibriosis and cholera are serious diseases distributed worldwide and caused by six marine bacteria of the Vibrio genus. Thousands of deaths occur each year due to these illnesses, necessitating the development of new preventive measures. Presently, the existing cholera vaccine demonstrates an effectiveness of approximately 60%. Here we describe a new multi-epitope vaccine, 'vme-VAC/MST-1' based on vaccine targets identified by reverse vaccinology and epitopes predicted by immunoinformatics, two currently effective tools for predicting new vaccines for bacterial pathogens. The vaccine was designed to combat vibriosis and cholera by incorporating epitopes predicted for CTL, HTL, and B cells. These epitopes were identified from six vaccine targets revealed through subtractive genomics, combined with reverse vaccinology, and were further filtered using immunoinformatics approaches based on their predicted immunogenicity. To construct the vaccine, 28 epitopes (24 CTL/B and 4 HTL/B) were linked to the sequence of the cholera toxin B subunit adjuvant. In silico analyses indicate that the resulting immunogen is stable, soluble, non-toxic, and non-allergenic. Furthermore, it exhibits no homology to the host and demonstrates a strong capacity to elicit innate, B-cell, and T-cell immune responses. Our analysis suggests that it is likely to elicit immune reactions mediated through the TLR5 pathway, as evidenced by the molecular docking of the vaccine with the receptor, which revealed high affinity and a favorable reaction. Thus, vme-VAC/MST-1 is predicted to be a safe and effective solution against pathogenic Vibrio spp. However, further experimental analyses are required to measure the vaccine's effects In vivo.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pedro Henrique Marques
- Institute of Biological Sciences, Post-graduate Interunits Program in Bioinformatics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thais Cristina Vilela Rodrigues
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Eduardo Horta Santos
- Institute of Biological Sciences, Post-graduate Interunits Program in Bioinformatics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lucas Bleicher
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flavia Figueira Aburjaile
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flaviano S Martins
- Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlo Jose Freire Oliveira
- Department of Microbiology, Immunology and Parasitology, Institute of Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Vasco Azevedo
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sandeep Tiwari
- Institute of Biology, Federal University of Bahia, Salvador, BA, Brazil
- Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil
| | - Siomar Soares
- Department of Microbiology, Immunology and Parasitology, Institute of Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| |
Collapse
|
4
|
Entrenas-García C, Suárez-Cárdenas JM, Fernández-Rodríguez R, Bautista R, Claros MG, Garrido JJ, Zaldívar-López S. miR-215 Modulates Ubiquitination to Impair Inflammasome Activation and Autophagy During Salmonella Typhimurium Infection in Porcine Intestinal Cells. Animals (Basel) 2025; 15:431. [PMID: 39943201 PMCID: PMC11815736 DOI: 10.3390/ani15030431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
The host response to S. Typhimurium infection can be post-transcriptionally regulated by miRNAs. In this study, we investigated the role of miR-215 using both in vivo porcine infection models and in vitro intestinal epithelial cell lines. Several miRNAs were found to be dysregulated in the porcine ileum during infection with wild-type and SPI2-defective mutant strains of S. Typhimurium, with some changes being SPI2-dependent. Notably, miR-215 was significantly downregulated during infection. To explore its functional role, gain-of-function experiments were performed by transfecting porcine intestinal epithelial cells (IPEC-J2) with a miR-215-5p mimic, followed by label-free quantitative (LFQ) proteomic analysis. This analysis identified 157 proteins, of which 35 were downregulated in response to miR-215 overexpression, suggesting they are potential targets of this miRNA. Among these, E2 small ubiquitin-like modifier (SUMO)-conjugating enzyme UBC9 and E3 ubiquitin-ligase HUWE1 were identified as key targets, both of which are upregulated during S. Typhimurium infection. The miR-215-mediated downregulation of these proteins resulted in a significant decrease in overall ubiquitination, a process crucial for regulating inflammasome activation and autophagy. Consistently, inflammasome markers caspase 1 (CASP1) and apoptosis-associated speck-like protein containing a CARD (ASC), as well as autophagy markers microtubule-associated protein 1A/1B-light chain 3 (LC3B) and Ras-related protein Rab-11 (RAB11A), showed decreased expression in miR-215 mimic-transfected and infected IPEC-J2 cells. To further validate these findings, human intestinal epithelial cells (HT29) were used as a complementary model, providing additional insights into conserved immune pathways and extending the observations made in the porcine system. Overall, our findings demonstrate that miR-215 plays a significant role in modulating host inflammasome activation and autophagy by targeting proteins involved in ubiquitination during S. Typhimurium infection.
Collapse
Affiliation(s)
- Carmen Entrenas-García
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
| | - José M. Suárez-Cárdenas
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Raúl Fernández-Rodríguez
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Rocío Bautista
- Plataforma Andaluza de Bioinformática, Supercomputing and Bioinnovation Center (SCBI), Universidad de Málaga, 29590 Malaga, Spain; (R.B.); (M.G.C.)
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, 29590 Malaga, Spain
| | - M. Gonzalo Claros
- Plataforma Andaluza de Bioinformática, Supercomputing and Bioinnovation Center (SCBI), Universidad de Málaga, 29590 Malaga, Spain; (R.B.); (M.G.C.)
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, 29590 Malaga, Spain
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM-UMA-CSIC), 29590 Malaga, Spain
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, 29010 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER) U741, 29071 Malaga, Spain
| | - Juan J. Garrido
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| | - Sara Zaldívar-López
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonosis and Emergent Diseases ENZOEM, Department of Genetics, University of Cordoba, 14014 Cordoba, Spain; (C.E.-G.); (J.M.S.-C.); (R.F.-R.)
- GA-14 Research Group, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), 14004 Cordoba, Spain
| |
Collapse
|
5
|
Tian JH, Huang S, Wang ZH, Li JJ, Song X, Jiang ZT, Shi BS, Zhao YY, Zhang HY, Wang KR, Hu XY, Zhang X, Guo DS. Supramolecular discrimination and diagnosis-guided treatment of intracellular bacteria. Nat Commun 2025; 16:1016. [PMID: 39863571 PMCID: PMC11762306 DOI: 10.1038/s41467-025-56308-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Pathogenic intracellular bacteria pose a significant threat to global public health due to the barriers presented by host cells hindering the timely detection of hidden bacteria and the effective delivery of therapeutic agents. To address these challenges, we propose a tandem diagnosis-guided treatment paradigm. A supramolecular sensor array is developed for simple, rapid, accurate, and high-throughput identification of intracellular bacteria. This diagnostic approach executes the significant guiding missions of screening a customized host-guest drug delivery system by disclosing the rationale behind the discrimination. We design eight azocalix[4]arenes with differential active targeting, cellular internalization, and hypoxia responsiveness to penetrate cells and interact with bacteria. Loaded with fluorescent indicators, these azocalix[4]arenes form a sensor array capable of discriminating eight intracellular bacterial species without cell lysis or separation. By fingerprinting specimens collected from bacteria-infected mice, the facilitated accurate diagnosis offers valuable guidance for selecting appropriate antibiotics. Moreover, mannose-modified azocalix[4]arene (ManAC4A) is screened as a drug carrier efficiently taken up by macrophages. Doxycycline loaded with ManAC4A exhibits improved efficacy against methicillin-resistant Staphylococcus aureus-infected peritonitis. This study introduces an emerging paradigm to intracellular bacterial diagnosis and treatment, offering broad potential in combating bacterial infectious diseases.
Collapse
Affiliation(s)
- Jia-Hong Tian
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Siyuan Huang
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Han Wang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Juan-Juan Li
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Xianhui Song
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Tao Jiang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Bing-Sen Shi
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ying-Ying Zhao
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Hui-Yan Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Xin-Yue Hu
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
| | - Xinge Zhang
- College of Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China.
| | - Dong-Sheng Guo
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, Kashi University, Kashi, China.
- College of Chemistry and Environmental Sciences, Kashi University, Kashi, China.
| |
Collapse
|
6
|
Bao P, Zhang XZ. Progress of tumor-resident intracellular bacteria for cancer therapy. Adv Drug Deliv Rev 2024; 214:115458. [PMID: 39383997 DOI: 10.1016/j.addr.2024.115458] [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: 07/12/2024] [Revised: 09/12/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Emerging studies have disclosed the pivotal role of cancer-associated microbiota in supporting cancer development, progression and dissemination, with the in-depth comprehending of tumor microenvironment. In particular, certain invasive bacteria that hide in various cells within the tumor tissues can render assistance to tumor growth and invasion through intricate mechanisms implicated in multiple branches of cancer biology. Thus, tumor-resident intracellular microbes are anticipated as next-generation targets for oncotherapy. This review is intended to delve into these internalized bacteria-driven cancer-promoting mechanisms and explore diversified antimicrobial therapeutic strategies to counteract the detrimental impact caused by these intruders, thereby improving therapeutic benefit of antineoplastic therapy.
Collapse
Affiliation(s)
- Peng Bao
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital, Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital, Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| |
Collapse
|
7
|
Yang J, Hai Z, Hou L, Liu Y, Zhang D, Zhou X. Baicalin Attenuates Panton-Valentine Leukocidin (PVL)-Induced Cytoskeleton Rearrangement via Regulating the RhoA/ROCK/LIMK and PI3K/AKT/GSK-3β Pathways in Bovine Mammary Epithelial Cells. Int J Mol Sci 2023; 24:14520. [PMID: 37833969 PMCID: PMC10572466 DOI: 10.3390/ijms241914520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Pore-forming toxins (PFTs) exert physiological effects by rearrangement of the host cell cytoskeleton. Staphylococcus aureus-secreted PFTs play an important role in bovine mastitis. In the study, we examined the effects of recombinant Panton-Valentine leukocidin (rPVL) on cytoskeleton rearrangement, and identified the signaling pathways involved in regulating the process in bovine mammary epithelial cells (BMECs) in vitro. Meanwhile, the underlying regulatory mechanism of baicalin for this process was investigated. The results showed that S. aureus induced cytoskeleton rearrangement in BMECs mainly through PVL. S. aureus and rPVL caused alterations in the cell morphology and layer integrity due to microfilament and microtubule rearrangement and focal contact inability. rPVL strongly induced the phosphorylation of cofilin at Ser3 mediating by the activation of the RhoA/ROCK/LIMK pathway, and resulted in the activation of loss of actin stress fibers, or the hyperphosphorylation of Tau at Ser396 inducing by the inhibition of the PI3K/AKT/GSK-3β pathways, and decreased the microtubule assembly. Baicalin significantly attenuated rPVL-stimulated cytoskeleton rearrangement in BMECs. Baicalin inhibited cofilin phosphorylation or Tau hyperphosphorylation via regulating the activation of RhoA/ROCK/LIMK and PI3K/AKT/GSK-3β signaling pathways. These findings provide new insights into the pathogenesis and potential treatment in S. aureus causing bovine mastitis.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuezhang Zhou
- Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan 750021, China; (J.Y.); (Z.H.)
| |
Collapse
|
8
|
Panera-Martínez S, Capita R, García-Fernández C, Alonso-Calleja C. Viability and Virulence of Listeria monocytogenes in Poultry. Microorganisms 2023; 11:2232. [PMID: 37764076 PMCID: PMC10538215 DOI: 10.3390/microorganisms11092232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
The prevalence of Listeria monocytogenes in 30 samples of poultry was determined using culture-dependent (isolation on OCLA and confirmation by conventional polymerase chain reaction -PCR-, OCLA&PCR) and culture-independent (real-time polymerase chain reaction, q-PCR) methods. L. monocytogenes was detected in 15 samples (50.0%) by OCLA&PCR and in 20 (66.7%) by q-PCR. The concentrations (log10 cfu/g) of L. monocytogenes (q-PCR) ranged from 2.40 to 5.22 (total cells) and from <2.15 to 3.93 (viable cells). The two methods, q-PCR using a viability marker (v-PCR) and OCLA&PCR (gold standard), were compared for their capacity to detect viable cells of L. monocytogenes, with the potential to cause human disease. The values for sensitivity, specificity and efficiency of the v-PCR were 100%, 66.7% and 83.3%, respectively. The agreement between the two methods (kappa coefficient) was 0.67. The presence of nine virulence genes (hlyA, actA, inlB, inlA, inlC, inlJ, prfA, plcA and iap) was studied in 45 L. monocytogenes isolates (three from each positive sample) using PCR. All the strains harbored between six and nine virulence genes. Fifteen isolates (33.3% of the total) did not show the potential to form biofilm on a polystyrene surface, as determined by a crystal violet assay. The remaining strains were classified as weak (23 isolates, 51.1% of the total), moderate (one isolate, 2.2%) or strong (six isolates, 13.3%) biofilm producers. The strains were tested for susceptibility to a panel of 15 antibiotics. An average of 5.11 ± 1.30 resistances per isolate was observed. When the values for resistance and for reduced susceptibility were taken jointly, this figure rose to 6.91 ± 1.59. There was a prevalence of resistance or reduced susceptibility of more than 50.0% for oxacillin, cefoxitin, cefotaxime, cefepime ciprofloxacin, enrofloxacin and nitrofurantoin. For the remaining antibiotics tested, the corresponding values ranged from 0.0% for chloramphenicol to 48.9% for rifampicin. The high prevalence and level of L. monocytogenes with numerous virulence factors in poultry underline how crucial it is to follow correct hygiene procedures during the processing of this foodstuff in order to reduce the risk of human listeriosis.
Collapse
Affiliation(s)
- Sarah Panera-Martínez
- Department of Food Hygiene and Technology, Veterinary Faculty, University of León, 24071 León, Spain
- Institute of Food Science and Technology, University of León, 24071 León, Spain
| | - Rosa Capita
- Department of Food Hygiene and Technology, Veterinary Faculty, University of León, 24071 León, Spain
- Institute of Food Science and Technology, University of León, 24071 León, Spain
| | | | - Carlos Alonso-Calleja
- Department of Food Hygiene and Technology, Veterinary Faculty, University of León, 24071 León, Spain
- Institute of Food Science and Technology, University of León, 24071 León, Spain
| |
Collapse
|
9
|
Carman PJ, Rebowski G, Dominguez R, Alqassim SS. Single particle cryo-EM analysis of Rickettsia conorii Sca2 reveals a formin-like core. J Struct Biol 2023; 215:107960. [PMID: 37028467 PMCID: PMC10200769 DOI: 10.1016/j.jsb.2023.107960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/16/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023]
Abstract
Spotted fever group Rickettsia undergo actin-based motility inside infected eukaryotic cells using Sca2 (surface cell antigen 2): an ∼ 1800 amino-acid monomeric autotransporter protein that is surface-attached to the bacterium and responsible for the assembly of long unbranched actin tails. Sca2 is the only known functional mimic of eukaryotic formins, yet it shares no sequence similarities to the latter. Using structural and biochemical approaches we have previously shown that Sca2 uses a novel actin assembly mechanism. The first ∼ 400 amino acids fold into helix-loop-helix repeats that form a crescent shape reminiscent of a formin FH2 monomer. Additionally, the N- and C- terminal halves of Sca2 display intramolecular interaction in an end-to-end manner and cooperate for actin assembly, mimicking a formin FH2 dimer. Towards a better structural understanding of this mechanism, we performed single-particle cryo-electron microscopy analysis of Sca2. While high-resolution structural details remain elusive, our model confirms the presence of a formin-like core: Sca2 indeed forms a doughnut shape, similar in diameter to a formin FH2 dimer and can accommodate two actin subunits. Extra electron density, thought to be contributed by the C-terminal repeat domain (CRD), covering one side is also observed. This structural analysis allows us to propose an updated model where nucleation proceeds by encircling two actin subunits, and elongation proceeds either by a formin-like mechanism that necessitates conformational changes in the observed Sca2 model, or via an insertional mechanism akin to that observed in the ParMRC system.
Collapse
Affiliation(s)
- Peter J Carman
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Grzegorz Rebowski
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Roberto Dominguez
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Saif S Alqassim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.
| |
Collapse
|
10
|
Hartland EL, Ghosal D, Giogha C. Manipulation of epithelial cell architecture by the bacterial pathogens Listeria and Shigella. Curr Opin Cell Biol 2022; 79:102131. [PMID: 36215855 DOI: 10.1016/j.ceb.2022.102131] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/22/2022] [Accepted: 09/06/2022] [Indexed: 01/31/2023]
Abstract
Subversion of the host cell cytoskeleton is a virulence attribute common to many bacterial pathogens. On mucosal surfaces, bacteria have evolved distinct ways of interacting with the polarised epithelium and manipulating host cell structure to propagate infection. For example, Shigella and Listeria induce cytoskeletal changes to induce their own uptake into enterocytes in order to replicate within an intracellular environment and then spread from cell-to-cell by harnessing the host actin cytoskeleton. In this review, we highlight some recent studies that advance our understanding of the role of the host cell cytoskeleton in the mechanical and molecular processes of pathogen invasion, cell-to-cell spread and the impact of infection on epithelial intercellular tension and innate mucosal defence.
Collapse
Affiliation(s)
- Elizabeth L Hartland
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
| | - Debnath Ghosal
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Cristina Giogha
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
11
|
Alqassim SS. Functional Mimicry of Eukaryotic Actin Assembly by Pathogen Effector Proteins. Int J Mol Sci 2022; 23:ijms231911606. [PMID: 36232907 PMCID: PMC9569871 DOI: 10.3390/ijms231911606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
The actin cytoskeleton lies at the heart of many essential cellular processes. There are hundreds of proteins that cells use to control the size and shape of actin cytoskeletal networks. As such, various pathogens utilize different strategies to hijack the infected eukaryotic host actin dynamics for their benefit. These include the control of upstream signaling pathways that lead to actin assembly, control of eukaryotic actin assembly factors, encoding toxins that distort regular actin dynamics, or by encoding effectors that directly interact with and assemble actin filaments. The latter class of effectors is unique in that, quite often, they assemble actin in a straightforward manner using novel sequences, folds, and molecular mechanisms. The study of these mechanisms promises to provide major insights into the fundamental determinants of actin assembly, as well as a deeper understanding of host-pathogen interactions in general, and contribute to therapeutic development efforts targeting their respective pathogens. This review discusses mechanisms and highlights shared and unique features of actin assembly by pathogen effectors that directly bind and assemble actin, focusing on eukaryotic actin nucleator functional mimics Rickettsia Sca2 (formin mimic), Burkholderia BimA (Ena/VASP mimic), and Vibrio VopL (tandem WH2-motif mimic).
Collapse
Affiliation(s)
- Saif S Alqassim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Health Care City, Dubai P.O. Box 505055, United Arab Emirates
| |
Collapse
|
12
|
Limosilactobacillus reuteri SLZX19-12 Protects the Colon from Infection by Enhancing Stability of the Gut Microbiota and Barrier Integrity and Reducing Inflammation. Microbiol Spectr 2022; 10:e0212421. [PMID: 35658572 PMCID: PMC9241593 DOI: 10.1128/spectrum.02124-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Limosilactobacillus reuteri plays an important role in regulating intestinal functions and maintaining barrier integrity in animals. In this study, Limosilactobacillus reuteri strain SLZX19-12 was isolated from the fecal microbiota of Tibetan pigs, and it was found that this strain is sensitive to common antibiotics and has strong resistance to stress. Upon being administered by gavage at different doses, including low, medium, and high doses, for 14 days, Limosilactobacillus reuteri SLZX19-12 may enhance the intestinal barrier. After administration of a high dose of SLZX19-12, mice were challenged with Salmonella enterica serovar Typhimurium SL1344. Infection with Salmonella Typhimurium SL1344 led to disordered colonic microbiotas, colonic inflammation through the S100A8/S100A9-NF-κB pathway and potential apoptosis, and translocation of pathogens to parenteral visceral organs in mice. However, the mice pretreated with Limosilactobacillus reuteri SLZX19-12 showed lower loads of Salmonella in visceral organs, less colonic inflammation, and higher barrier integrity. More importantly, the administration of strain SLZX19-12 resulted in a more stable microbiota structure of the colon, in which the abundance of Alloprevotella was greatly enhanced. Therefore, this study suggests that Limosilactobacillus reuteri SLZX19-12 can protect the colon from infection by enhancing the stability of gut microbiota and barrier integrity and reducing inflammation. IMPORTANCE The use of antibiotics to treat bacterial infections leads to a series of side effects. As an alternative method, the biocontrol strategy, which uses probiotics to suppress pathogens, is considered a potential way to deal with bacterial infections in gut. However, there are few probiotics that are currently safe and can protect against infection. In this study, Limosilactobacillus reuteri strain SLZX19-12 was obtained from Tibetan pigs, which have higher resistance to infection. This strain is sensitive to conventional antibiotics, secretes a wide spectrum of enzymes, and also promotes the intestinal barrier function in mice. In addition, Limosilactobacillus reuteri SLZX19-12 can promote the stability of the gut microbiota to avoid or alleviate the occurrence or development of foodborne infections.
Collapse
|
13
|
Lian L, Li W, Xue T, Ren J, Tang F, Liu Y, Xue F, Dai J. Comparative transcriptomic analysis provides insights into transcription mechanisms of Vibrio parahaemolyticus T3SS during interaction with HeLa cells. Braz J Microbiol 2022; 53:289-301. [PMID: 34652743 PMCID: PMC8882520 DOI: 10.1007/s42770-021-00627-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022] Open
Abstract
Vibrio parahaemolyticus is an important foodborne pathogenic bacterium that harbors the type III secretion system 1 (T3SS1) as an essential virulence factor. However, the pathogenesis and infection mechanism mediated by T3SS1 are not entirely clarified. Similar to previous studies on other T3SS-positive bacteria, the T3SS1 needle is a major extracellular component in V. parahaemolyticus. We recently showed that the needle gene-deletion mutant (ΔvscF) exhibited markedly decreased cytotoxicity and effector translocation during interaction with HeLa cells. To further elucidate the pathogenesis of T3SS1 during host cell infection, bacterial RNA was extracted from wild-type POR-1 and ΔvscF mutants under infected condition for comparative RNA sequencing analysis in HeLa cell. The results showed that 120 differentially expressed genes (DEGs) were identified in the ΔvscF-infected group. These encoded proteins of DEGs, such as VP2088, VP2089, and VP2091, were annotated as ABC transporter system, whereas VP0757, VP1123, and VP1289 may be new transcriptional regulators. In addition, the downregulation of T3SS1 had a positive influence on the expression of T3SS2. Moreover, the transcription of the basal body is unaffected by the needle, and there was a close relation among the tip, translocon, and needle, because bacterial adenylate cyclase two-hybrid system (BACTH system) assay indicated the interaction of VP1656, VP1670, VP1693, and VP1694 (VscF). This study provides insights into transcription mechanism of T3SS1 upon infecting HeLa cell, which is expected to better clarify the T3SS1 virulent mechanism.
Collapse
Affiliation(s)
- Lele Lian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wanjun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tingyue Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jianluan Ren
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Fang Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yongjie Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Feng Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Jianjun Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
14
|
Cross Talk between ARF1 and RhoA Coordinates the Formation of Cytoskeletal Scaffolds during Chlamydia Infection. mBio 2021; 12:e0239721. [PMID: 34903051 PMCID: PMC8669492 DOI: 10.1128/mbio.02397-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Chlamydia trachomatis is an obligate intracellular bacterium that has developed sophisticated mechanisms to survive inside its infectious compartment, the inclusion. Notably, Chlamydia weaves an extensive network of microtubules (MTs) and actin filaments to enable interactions with host organelles and enhance its stability. Despite the global health and economic burden caused by this sexually transmitted pathogen, little is known about how actin and MT scaffolds are integrated into an increasingly complex virulence system. Previously, we established that the chlamydial effector InaC interacts with ARF1 to stabilize MTs. We now demonstrate that InaC regulates RhoA to control actin scaffolds. InaC relies on cross talk between ARF1 and RhoA to coordinate MTs and actin, where the presence of RhoA downregulates stable MT scaffolds and ARF1 activation inhibits actin scaffolds. Understanding how Chlamydia hijacks complex networks will help elucidate how this clinically significant pathogen parasitizes its host and reveal novel cellular signaling pathways.
Collapse
|
15
|
Structural organization of erythrocyte membrane microdomains and their relation with malaria susceptibility. Commun Biol 2021; 4:1375. [PMID: 34880413 PMCID: PMC8655059 DOI: 10.1038/s42003-021-02900-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
Cholesterol-rich microdomains are membrane compartments characterized by specific lipid and protein composition. These dynamic assemblies are involved in several biological processes, including infection by intracellular pathogens. This work provides a comprehensive analysis of the composition of human erythrocyte membrane microdomains. Based on their floating properties, we also categorized the microdomain-associated proteins into clusters. Interestingly, erythrocyte microdomains include the vast majority of the proteins known to be involved in invasion by the malaria parasite Plasmodium falciparum. We show here that the Ecto-ADP-ribosyltransferase 4 (ART4) and Aquaporin 1 (AQP1), found within one specific cluster, containing the essential host determinant CD55, are recruited to the site of parasite entry and then internalized to the newly formed parasitophorous vacuole membrane. By generating null erythroid cell lines, we showed that one of these proteins, ART4, plays a role in P. falciparum invasion. We also found that genetic variants in both ART4 and AQP1 are associated with susceptibility to the disease in a malaria-endemic population.
Collapse
|
16
|
Campylobacter jejuni Triggers Signaling through Host Cell Focal Adhesions To Inhibit Cell Motility. mBio 2021; 12:e0149421. [PMID: 34425711 PMCID: PMC8406305 DOI: 10.1128/mbio.01494-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Campylobacter jejuni is a major foodborne pathogen that exploits the focal adhesions of intestinal cells to promote invasion and cause severe gastritis. Focal adhesions are multiprotein complexes involved in bidirectional signaling between the actin cytoskeleton and the extracellular matrix. We investigated the dynamics of focal adhesion structure and function in C. jejuni-infected cells using a comprehensive set of approaches, including confocal microscopy of live and fixed cells, immunoblotting, and superresolution interferometric photoactivated localization microscopy (iPALM). We found that C. jejuni infection of epithelial cells results in increased focal adhesion size and altered topology. These changes resulted in a persistent modulatory effect on the host cell focal adhesion, evidenced by an increase in cell adhesion strength, a decrease in individual cell motility, and a reduction in collective cell migration. We discovered that C. jejuni infection causes an increase in phosphorylation of paxillin and an alteration of paxillin turnover at the focal adhesion, which together represent a potential mechanistic basis for altered cell motility. Finally, we observed that infection of epithelial cells with the C. jejuni wild-type strain in the presence of a protein synthesis inhibitor, a C. jejuni CadF and FlpA fibronectin-binding protein mutant, or a C. jejuni flagellar export mutant blunts paxillin phosphorylation and partially reestablishes individual host cell motility and collective cell migration. These findings provide a potential mechanism for the restricted intestinal repair observed in C. jejuni-infected animals and raise the possibility that bacteria targeting extracellular matrix components can alter cell behavior after binding and internalization by manipulating focal adhesions.
Collapse
|
17
|
Lian L, Xue J, Li W, Ren J, Tang F, Liu Y, Xue F, Dai J. VscF in T3SS1 Helps to Translocate VPA0226 in Vibrio parahaemolyticus. Front Cell Infect Microbiol 2021; 11:652432. [PMID: 33869083 PMCID: PMC8047418 DOI: 10.3389/fcimb.2021.652432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
In Vibrio parahaemolyticus, type III secretion system 1 (T3SS1) is a major virulence factor that delivers effectors into the host eukaryotic cytoplasm; however, studies on its infection mechanism are currently limited. To determine the function of the vscF gene, we constructed the vscF deletion mutant ΔvscF and complementation strain CΔvscF. Compared with those of wild-type POR-1 and CΔvscF, the cytotoxic, adherent, and apoptotic abilities of ΔvscF in HeLa cells were significantly reduced (P < 0.01). Furthermore, in infected HeLa cells, the mutant strain reduced the translocation rates of VP1683 and VP1686 effectors compared to the wild-type and complementation strains. A BLAST search showed that vscF is homologous to the MixH needle protein of Shigella flexneri, indicating that the vscF gene encodes the needle protein of T3SS1 in V. parahaemolyticus. Additional translocation assays showed that VPA0226 translocated into the HeLa eukaryotic cytoplasm via T3SS1, secretion assays showed that VPA0226 can be secreted to supernatant by T3SS1, indicating that VPA0226 belongs to the unpublished class of T3SS1 effectors. In conclusion, our data indicate an essential role of vscF in V. parahaemolyticus T3SS1 and revealed that VPA0226 can be secreted into the host cell cytoplasm via T3SS1. This study provides insights into a previously unexplored aspect of T3SS1, which is expected to contribute to the understanding of its infection mechanism.
Collapse
Affiliation(s)
- Lele Lian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiao Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wanjun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jianluan Ren
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Fang Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yongjie Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Feng Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jianjun Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
18
|
Moraleda CP, Robledo D, Gutiérrez AP, Del-Pozo J, Yáñez JM, Houston RD. Investigating mechanisms underlying genetic resistance to Salmon Rickettsial Syndrome in Atlantic salmon using RNA sequencing. BMC Genomics 2021; 22:156. [PMID: 33676414 PMCID: PMC7936450 DOI: 10.1186/s12864-021-07443-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/11/2021] [Indexed: 12/29/2022] Open
Abstract
Background Salmon Rickettsial Syndrome (SRS), caused by Piscirickettsia salmonis, is one of the primary causes of morbidity and mortality in Atlantic salmon aquaculture, particularly in Chile. Host resistance is a heritable trait, and functional genomic studies have highlighted genes and pathways important in the response of salmon to the bacteria. However, the functional mechanisms underpinning genetic resistance are not yet well understood. In the current study, a large population of salmon pre-smolts were challenged with P. salmonis, with mortality levels recorded and samples taken for genotyping. In parallel, head kidney and liver samples were taken from animals of the same population with high and low genomic breeding values for resistance, and used for RNA-Sequencing to compare their transcriptome profile both pre and post infection. Results A significant and moderate heritability (h2 = 0.43) was shown for the trait of binary survival. Genome-wide association analyses using 38 K imputed SNP genotypes across 2265 animals highlighted that resistance is a polygenic trait. Several thousand genes were identified as differentially expressed between controls and infected samples, and enriched pathways related to the host immune response were highlighted. In addition, several networks with significant correlation with SRS resistance breeding values were identified, suggesting their involvement in mediating genetic resistance. These included apoptosis, cytoskeletal organisation, and the inflammasome. Conclusions While resistance to SRS is a polygenic trait, this study has highlighted several relevant networks and genes that are likely to play a role in mediating genetic resistance. These genes may be future targets for functional studies, including genome editing, to further elucidate their role underpinning genetic variation in host resistance. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07443-2.
Collapse
Affiliation(s)
- Carolina P Moraleda
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - Alejandro P Gutiérrez
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - Jorge Del-Pozo
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - José M Yáñez
- Faculty of Veterinary and Livestock Sciences, University of Chile, Santiago, Chile.
| | - Ross D Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
19
|
Cestero JJ, Castanheira S, Pucciarelli MG, García-Del Portillo F. A Novel Salmonella Periplasmic Protein Controlling Cell Wall Homeostasis and Virulence. Front Microbiol 2021; 12:633701. [PMID: 33679664 PMCID: PMC7933661 DOI: 10.3389/fmicb.2021.633701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Horizontal gene transfer has shaped the evolution of Salmonella enterica as pathogen. Some functions acquired by this mechanism include enzymes involved in peptidoglycan (PG) synthesis and remodeling. Here, we report a novel serovar Typhimurium protein that is absent in non-pathogenic bacteria and bears a LprI functional domain, first reported in a Mycobacterium tuberculosis lipoprotein conferring lysozyme resistance. Based on the presence of such domain, we hypothesized a role of this S. Typhimurium protein in PG metabolism. This protein, which we named ScwA for Salmonellacell wall-related regulator-A, controls positively the levels of the murein lytic transglycosylase MltD. In addition, the levels of other enzymes that cleave bonds in the PG lattice were affected in a mutant lacking ScwA, including a soluble lytic tranglycosylase (Slt), the amidase AmiC, and a few endo- and carboxypeptidases (NlpC, PBP4, and AmpH). The scwA gene has lower G+C content than the genomic average (43.1 vs. 52.2%), supporting acquisition by horizontal transfer. ScwA is located in the periplasm, stabilized by two disulfide bridges, produced preferentially in stationary phase and down-regulated following entry of the pathogen into eukaryotic cells. ScwA deficiency, however, results in a hypervirulent phenotype in the murine typhoid model. Based on these findings, we conclude that ScwA may be exploited by S. Typhimurium to ensure cell envelope homeostasis along the infection and to prevent host overt damage. This role could be accomplished by controlling the production or stability of a reduced number of peptidoglycan hydrolases whose activities result in the release of PG fragments.
Collapse
Affiliation(s)
- Juan J Cestero
- Laboratory of Intracellular Bacterial Pathogens, National Centre for Biotechnology (CNB)-CSIC, Madrid, Spain
| | - Sónia Castanheira
- Laboratory of Intracellular Bacterial Pathogens, National Centre for Biotechnology (CNB)-CSIC, Madrid, Spain
| | - M Graciela Pucciarelli
- Laboratory of Intracellular Bacterial Pathogens, National Centre for Biotechnology (CNB)-CSIC, Madrid, Spain.,Department of Molecular Biology, Autonomous University of Madrid, Madrid, Spain.,Center for Molecular Biology "Severo Ochoa" (CBMSO)-CSIC, Madrid, Spain
| | | |
Collapse
|
20
|
Yuan L, van der Mei HC, Busscher HJ, Peterson BW. Two-Stage Interpretation of Changes in TEER of Intestinal Epithelial Layers Protected by Adhering Bifidobacteria During E. coli Challenges. Front Microbiol 2020; 11:599555. [PMID: 33329490 PMCID: PMC7710611 DOI: 10.3389/fmicb.2020.599555] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Mechanisms of gastrointestinal protection by probiotic bacteria against infection involve amongst others, modulation of intestinal epithelial barrier function. Trans-epithelial electrical resistance (TEER) is widely used to evaluate cellular barrier functions. Here, we developed a two-stage interpretative model of the time-dependence of the TEER of epithelial layers grown in a transwell during Escherichia coli challenges in the absence or presence of adhering bifidobacteria. E. coli adhesion in absence or presence of adhering bifidobacteria was enumerated using selective plating. After 4-8 h, E. coli challenges increased TEER to a maximum due to bacterial adhesion and increased expression of a tight-junction protein [zonula occludens-1 (ZO-1)], concurrent with a less dense layer structure, that is indicative of mild epithelial layer damage. Before the occurrence of a TEER-maximum, decreases in electrical conductance (i.e., the reciprocal TEER) did not relate with para-cellular dextran-permeability, but after occurrence of a TEER-maximum, dextran-permeability and conductance increased linearly, indicative of more severe epithelial layer damage. Within 24 h after the occurrence of a TEER maximum, TEER decreased to below the level of unchallenged epithelial layers demonstrating microscopically observable holes and apoptosis. Under probiotic protection by adhering bifidobacteria, TEER-maxima were delayed or decreased in magnitude due to later transition from mild to severe damage, but similar linear relations between conductance and dextran permeability were observed as in absence of adhering bifidobacteria. Based on the time-dependence of the TEER and the relation between conductance and dextran-permeability, it is proposed that bacterial adhesion to epithelial layers first causes mild damage, followed by more severe damage after the occurrence of a TEER-maximum. The mild damage caused by E. coli prior to the occurrence of TEER maxima was reversible upon antibiotic treatment, but the severe damage after occurrence of TEER maxima could not be reverted by antibiotic treatment. Thus, single-time TEER is interpretable in two ways, depending whether increasing to or decreasing from its maximum. Adhering bifidobacteria elongate the time-window available for antibiotic treatment to repair initial pathogen damage to intestinal epithelial layers.
Collapse
Affiliation(s)
| | | | | | - Brandon W. Peterson
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
21
|
Listeria monocytogenes Interferes with Host Cell Mitosis through Its Virulence Factors InlC and ActA. Toxins (Basel) 2020; 12:toxins12060411. [PMID: 32575670 PMCID: PMC7354435 DOI: 10.3390/toxins12060411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023] Open
Abstract
Listeria monocytogenes is among the best-characterized intracellular pathogens. Its virulence factors, and the way they interfere with host cells to hijack host functions and promote the establishment and dissemination of the infection, have been the focus of multiple studies over the last 30 years. During cellular infection, L. monocytogenes was shown to induce host DNA damage and delay the host cell cycle to its own benefit. However, whether the cell cycle stage would interfere with the capacity of Listeria to infect human cultured cell lines was never assessed. We found here that L. monocytogenes preferentially infects cultured cells in G2/M phases. Inside G2/M cells, the bacteria lead to an increase in the overall mitosis duration by delaying the mitotic exit. We showed that L. monocytogenes infection causes a sustained activation of the spindle assembly checkpoint, which we correlated with the increase in the percentage of misaligned chromosomes detected in infected cells. Moreover, we demonstrated that chromosome misalignment in Listeria-infected cells required the function of two Listeria virulence factors, ActA and InlC. Our findings show the pleiotropic role of Listeria virulence factors and their cooperative action in successfully establishing the cellular infection.
Collapse
|
22
|
Araujo-Garrido JL, Baisón-Olmo F, Bernal-Bayard J, Romero F, Ramos-Morales F. Tubulin Folding Cofactor TBCB is a Target of the Salmonella Effector Protein SseK1. Int J Mol Sci 2020; 21:ijms21093193. [PMID: 32366039 PMCID: PMC7246435 DOI: 10.3390/ijms21093193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Salmonella enterica serovar Typhimurium is a human and animal pathogen that uses type III secretion system effectors to manipulate the host cell and fulfill infection. SseK1 is a Salmonella effector with glycosyltransferase activity. We carried out a yeast two-hybrid screen and have identified tubulin-binding cofactor B (TBCB) as a new binding partner for this effector. SseK1 catalyzed the addition of N-acetylglucosamine to arginine on TBCB, and its expression promoted the stabilization of the microtubule cytoskeleton of HEK293T cells. The conserved Asp-x-Asp (DxD) motif that is essential for the activity of SseK1 was required for the binding and modification of TBCB and for the effect on the cytoskeleton. Our study has identified a novel target for SseK1 and suggests that this effector may have a role in the manipulation of the host cell microtubule network to provide a safe niche for this pathogen.
Collapse
Affiliation(s)
- Juan Luis Araujo-Garrido
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain; (J.L.A.-G.); (F.B.-O.); (J.B.-B.)
| | - Fernando Baisón-Olmo
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain; (J.L.A.-G.); (F.B.-O.); (J.B.-B.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 1058, Chile
| | - Joaquín Bernal-Bayard
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain; (J.L.A.-G.); (F.B.-O.); (J.B.-B.)
| | - Francisco Romero
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - Francisco Ramos-Morales
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain; (J.L.A.-G.); (F.B.-O.); (J.B.-B.)
- Correspondence:
| |
Collapse
|
23
|
Chlamydia-induced curvature of the host-cell plasma membrane is required for infection. Proc Natl Acad Sci U S A 2020; 117:2634-2644. [PMID: 31964834 PMCID: PMC7007526 DOI: 10.1073/pnas.1911528117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During invasion of host cells, Chlamydia pneumoniae secretes the effector protein CPn0678, which facilitates internalization of the pathogen by remodeling the target cell's plasma membrane and recruiting sorting nexin 9 (SNX9), a central multifunctional endocytic scaffold protein. We show here that the strongly amphipathic N-terminal helix of CPn0678 mediates binding to phospholipids in both the plasma membrane and synthetic membranes, and is sufficient to induce extensive membrane tubulations. CPn0678 interacts via its conserved C-terminal polyproline sequence with the Src homology 3 domain of SNX9. Thus, SNX9 is found at bacterial entry sites, where C. pneumoniae is internalized via EGFR-mediated endocytosis. Moreover, depletion of human SNX9 significantly reduces internalization, whereas ectopic overexpression of CPn0678-GFP results in a dominant-negative effect on endocytotic processes in general, leading to the uptake of fewer chlamydial elementary bodies and diminished turnover of EGFR. Thus, CPn0678 is an early effector involved in regulating the endocytosis of C. pneumoniae in an EGFR- and SNX9-dependent manner.
Collapse
|
24
|
Epidemiology of Nucleus-Dwelling Holospora: Infection, Transmission, Adaptation, and Interaction with Paramecium. Results Probl Cell Differ 2020; 69:105-135. [PMID: 33263870 DOI: 10.1007/978-3-030-51849-3_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The chapter describes the exceptional symbiotic associations formed between the ciliate Paramecium and Holospora, highly infectious bacteria residing in the host nuclei. Holospora and Holospora-like bacteria (Alphaproteobacteria) are characterized by their ability for vertical and horizontal transmission in host populations, a complex biphasic life cycle, and pronounced preference for host species and colonized cell compartment. These bacteria are obligate intracellular parasites; thus, their metabolic repertoire is dramatically reduced. Nevertheless, they perform complex interactions with the host ciliate. We review ongoing efforts to unravel the molecular adaptations of these bacteria to their unusual lifestyle and the host's employment in the symbiosis. Furthermore, we summarize current knowledge on the genetic and genomic background of Paramecium-Holospora symbiosis and provide insights into the ecological and evolutionary consequences of this interaction. The diversity and occurrence of symbioses between ciliates and Holospora-like bacteria in nature is discussed in connection with transmission modes of symbionts, host specificity and compatibility of the partners. We aim to summarize 50 years of research devoted to these symbiotic systems and conclude trying to predict some perspectives for further studies.
Collapse
|
25
|
Andrade LO. Plasma membrane repair involvement in parasitic and other pathogen infections. CURRENT TOPICS IN MEMBRANES 2019; 84:217-238. [PMID: 31610864 DOI: 10.1016/bs.ctm.2019.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intracellular pathogens depend on specific mechanisms to be able to gain entry and survive into their host cells. For this, they subvert pathways involved in physiological cellular processes. Here we are going to focus on how two protozoan parasites, Trypanosoma cruzi and Leishmania sp, which may cause severe diseases in humans, use plasma membrane repair (PMR) mechanisms to gain entry in host intracellular environment. T. cruzi is the causative agent of Chagas disease, a disease originally endemic of central and South America, but that has become widespread around the globe. T. cruzi is able to invade any nucleated cell, but muscle cells are usually the main targets during chronic disease. During host cell contact, the parasite interacts with proteins at the host cell surface and may cause damage to their membrane, which has been shown to be responsible for inducing intracellular calcium increase and PMR-related events that culminate with parasite internalization. The same was recently observed for Leishmania sp, when infecting nonprofessional phagocytic cells, such as fibroblasts. Other pathogens, such as viruses or bacteria may also use PMR-related events for invasion and vacuole escape/maturation. In some cases, PMR may also be responsible to modulate pathogen intracellular development. These other PMR roles in pathogen infections will also be briefly discussed.
Collapse
Affiliation(s)
- Luciana O Andrade
- Department of Morphology, Federal University of Minas Gerais, Brazil.
| |
Collapse
|
26
|
Yang L, Lai F, He L, Lu Y, Zhong Q, Lai C, Dai Y. LI1035, a putative effector secreted by Lawsonia intracellularis, targets the MAPK pathway and regulates actin organizationin yeast and mammalian cells. Vet Microbiol 2019; 235:127-135. [PMID: 31282370 DOI: 10.1016/j.vetmic.2019.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/17/2019] [Accepted: 06/10/2019] [Indexed: 11/17/2022]
Abstract
Lawsonia intracellularis is an obligate intracellular Gram-negative bacterium that has been identified as the etiological agent of the contagious disease proliferative enteropathy (PE) in a wide range of animals, mainly pigs. The genome sequence of L. intracellularis indicates that this bacterium possess a type III secretion system (T3SS), which may assist the bacterium during cell invasion and host innate immune system evasion and could be a mechanism for inducing cellular proliferation. However, the effectors secreted by the T3SS (T3Es) of L. intracellularis have not been reported. T3Es often target conserved eukaryotic cellular processes, and yeast is an established and robust model system in which to reveal their function. By screening the growth inhibition of an ordered array of Saccharomyces cerevisiae strains expressing the hypothetical genes of L. intracellularis, LI1035 was identified as the first putative effector that inhibits yeast growth. The LI1035-induced growth inhibition was rescued in two of the 14 mitogen-activated protein kinase (MAPK) yeast haploid deletion strains, suggesting that LI1035 interacts with the components of the MAPK pathway in yeast. Phosphorylation assays confirmed that LI1035 inhibits MAPK signaling cascades in yeast and mammalian cells. Actin staining assays revealed that LI1035 regulates actin organization in yeast and mammalian cells. Taken together, these results indicate that LI1035 alters MAPK pathway activity and regulates actin organization in the host. These findings may contribute to the understanding the pathogenesis of L. intracellularis and support the use of yeast as a heterologous system for the functional analysis of pathogen-specific gene products in the laboratory.
Collapse
Affiliation(s)
- Lijuan Yang
- School of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Fenju Lai
- School of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Lei He
- School of Life Science, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yongjun Lu
- School of Life Science, Sun Yat-sen University, Guangzhou, 510275, China
| | - Qiwang Zhong
- School of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Chongde Lai
- School of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yimin Dai
- School of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
27
|
Ubiquitin, SUMO, and NEDD8: Key Targets of Bacterial Pathogens. Trends Cell Biol 2018; 28:926-940. [PMID: 30107971 DOI: 10.1016/j.tcb.2018.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 01/09/2023]
Abstract
Manipulation of host protein post-translational modifications (PTMs) is used by various pathogens to interfere with host cell functions. Among these modifications, ubiquitin (UBI) and ubiquitin-like proteins (UBLs) constitute key targets because they are regulators of pathways essential for the host cell. In particular, these PTM modifiers control pathways that have been described as crucial for infection such as pathogen entry, replication, propagation, or detection by the host. Although bacterial pathogens lack eucaryotic-like UBI or UBL systems, many of them produce proteins that specifically interfere with these host PTMs during infection. In this review we discuss the different mechanisms used by bacteria to interfere with host UBI and the two UBLs, SUMO and NEDD8.
Collapse
|
28
|
Cruz R, Pereira-Castro I, Almeida MT, Moreira A, Cabanes D, Sousa S. Epithelial Keratins Modulate cMet Expression and Signaling and Promote InlB-Mediated Listeria monocytogenes Infection of HeLa Cells. Front Cell Infect Microbiol 2018; 8:146. [PMID: 29868502 PMCID: PMC5960701 DOI: 10.3389/fcimb.2018.00146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
The host cytoskeleton is a major target for bacterial pathogens during infection. In particular, pathogens usurp the actin cytoskeleton function to strongly adhere to the host cell surface, to induce plasma membrane remodeling allowing invasion and to spread from cell to cell and disseminate to the whole organism. Keratins are cytoskeletal proteins that are the major components of intermediate filaments in epithelial cells however, their role in bacterial infection has been disregarded. Here we investigate the role of the major epithelial keratins, keratins 8 and 18 (K8 and K18), in the cellular infection by Listeria monocytogenes. We found that K8 and K18 are required for successful InlB/cMet-dependent L. monocytogenes infection, but are dispensable for InlA/E-cadherin-mediated invasion. Both K8 and K18 accumulate at InlB-mediated internalization sites following actin recruitment and modulate actin dynamics at those sites. We also reveal the key role of K8 and K18 in HGF-induced signaling which occurs downstream the activation of cMet. Strikingly, we show here that K18, and at a less extent K8, controls the expression of cMet and other surface receptors such TfR and integrin β1, by promoting the stability of their corresponding transcripts. Together, our results reveal novel functions for major epithelial keratins in the modulation of actin dynamics at the bacterial entry sites and in the control of surface receptors mRNA stability and expression.
Collapse
Affiliation(s)
- Rui Cruz
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Isabel Pereira-Castro
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Gene Regulation Group, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Maria T Almeida
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Alexandra Moreira
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Gene Regulation Group, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Didier Cabanes
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal
| | - Sandra Sousa
- Group of Molecular Microbiology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, Institute for Molecular and Cell Biology, Porto, Portugal
| |
Collapse
|
29
|
David DJ, Pagliuso A, Radoshevich L, Nahori MA, Cossart P. Lmo1656 is a secreted virulence factor of Listeria monocytogenes that interacts with the sorting nexin 6-BAR complex. J Biol Chem 2018; 293:9265-9276. [PMID: 29666193 PMCID: PMC6005434 DOI: 10.1074/jbc.ra117.000365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 04/09/2018] [Indexed: 12/14/2022] Open
Abstract
Listeria monocytogenes (Lm) is a facultative intracellular bacterial pathogen and the causative agent of listeriosis, a rare but fatal disease. During infection, Lm can traverse several physiological barriers; it can cross the intestine and placenta barrier and, in immunocompromised individuals, the blood–brain barrier. With the recent plethora of sequenced genomes available for Lm, it is clear that the complete repertoire of genes used by Lm to interact with its host remains to be fully explored. Recently, we focused on secreted Lm proteins because they are likely to interact with host cell components. Here, we investigated a putatively secreted protein of Lm, Lmo1656, that is present in most sequenced strains of Lm but absent in the nonpathogenic species Listeria innocua. lmo1656 gene is predicted to encode a small, positively charged protein. We show that Lmo1656 is secreted by Lm. Furthermore, deletion of the lmo1656 gene (Δlmo1656) attenuates virulence in mice infected orally but not intravenously, suggesting that Lmo1656 plays a role during oral listeriosis. We identified sorting nexin 6 (SNX6), an endosomal sorting component and BAR domain–containing protein, as a host cell interactor of Lmol656. SNX6 colocalizes with WT Lm during the early steps of infection. This colocalization depends on Lmo1656, and RNAi of SNX6 impairs infection in infected tissue culture cells, suggesting that SNX6 is utilized by Lm during infection. Our results reveal that Lmo1656 is a novel secreted virulence factor of Lm that facilitates recruitment of a specific member of the sorting nexin family in the mammalian host.
Collapse
Affiliation(s)
- Daryl Jason David
- From the Unité des Interactions Bactéries-Cellules, Department of Cell Biology and Infection, INSERM U604, Institut National de la Recherche Agronomique USC2020, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
| | - Alessandro Pagliuso
- From the Unité des Interactions Bactéries-Cellules, Department of Cell Biology and Infection, INSERM U604, Institut National de la Recherche Agronomique USC2020, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
| | - Lilliana Radoshevich
- From the Unité des Interactions Bactéries-Cellules, Department of Cell Biology and Infection, INSERM U604, Institut National de la Recherche Agronomique USC2020, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
| | - Marie-Anne Nahori
- From the Unité des Interactions Bactéries-Cellules, Department of Cell Biology and Infection, INSERM U604, Institut National de la Recherche Agronomique USC2020, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
| | - Pascale Cossart
- From the Unité des Interactions Bactéries-Cellules, Department of Cell Biology and Infection, INSERM U604, Institut National de la Recherche Agronomique USC2020, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
30
|
Abstract
Posttranslational modifications are covalent changes made to proteins that typically alter the function or location of the protein. AMPylation is an emerging posttranslational modification that involves the addition of adenosine monophosphate (AMP) to a protein. Like other, more well-studied posttranslational modifications, AMPylation is predicted to regulate the activity of the modified target proteins. However, the scope of this modification both in bacteria and in eukaryotes remains to be fully determined. In this review, we provide an up to date overview of the known AMPylating enzymes, the regulation of these enzymes, and the effect of this modification on target proteins.
Collapse
Affiliation(s)
- Amanda K. Casey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard NA5.120F, Dallas, Texas 75390-9148, United States
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard NA5.120F, Dallas, Texas 75390-9148, United States
- Howard Hughes Medical Institute, 6000 Harry Hines Boulevard NA5.120F, Dallas, Texas 75390-9148, United States
| |
Collapse
|
31
|
Amarachintha S, Harmel-Laws E, Steinbrecher KA. Guanylate cyclase C reduces invasion of intestinal epithelial cells by bacterial pathogens. Sci Rep 2018; 8:1521. [PMID: 29367634 PMCID: PMC5784150 DOI: 10.1038/s41598-018-19868-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/20/2017] [Indexed: 12/24/2022] Open
Abstract
The guanylate cyclase C (GC-C) receptor regulates electrolyte and water secretion into the gut following activation by the E. coli enterotoxin STa, or by weaker endogenous agonists guanylin and uroguanylin. Our previous work has demonstrated that GC-C plays an important role in controlling initial infection as well as carrying load of non-invasive bacterial pathogens in the gut. Here, we use Salmonella enterica serovar Typhimurium to determine whether GC-C signaling is important in host defense against pathogens that actively invade enterocytes. In vitro studies indicated that GC-C signaling significantly reduces Salmonella invasion into Caco2-BBE monolayers. Relative to controls, GC-C knockout mice develop severe systemic illness following oral Salmonella infection, characterized by disrupted intestinal mucus layer, elevated cytokines and organ CFUs, and reduced animal survival. In Salmonella-infected wildtype mice, oral gavage of GC-C agonist peptide reduced host/pathogen physical interaction and diminished bacterial translocation to mesenteric lymph nodes. These studies suggest that early life susceptibility to STa-secreting enterotoxigenic E. coli may be counter-balanced by a critical role of GC-C in protecting the mucosa from non-STa producing, invasive bacterial pathogens.
Collapse
Affiliation(s)
- Surya Amarachintha
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Eleana Harmel-Laws
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Kris A Steinbrecher
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229, USA.
| |
Collapse
|
32
|
Ecological fitness and virulence features of Vibrio parahaemolyticus in estuarine environments. Appl Microbiol Biotechnol 2017; 101:1781-1794. [PMID: 28144705 DOI: 10.1007/s00253-017-8096-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/22/2016] [Accepted: 01/02/2017] [Indexed: 12/11/2022]
Abstract
Vibrio parahaemolyticus is a commonly encountered and highly successful organism in marine ecosystems. It is a fast-growing, extremely versatile copiotroph that is active over a very broad range of conditions. It frequently occurs suspended in the water column (often attached to particles or zooplankton), and is a proficient colonist of submerged surfaces. This organism is an important pathogen of animals ranging from microcrustaceans to humans and is a causative agent of seafood-associated food poisoning. This review examines specific ecological adaptations of V. parahaemolyticus, including its broad tolerances to temperature and salinity, its utilization of a wide variety of organic carbon and energy sources, and its pervasive colonization of suspended and stationary materials that contribute to its success and ubiquity in temperate and tropical estuarine ecosystems. Several virulence-related features are examined, in particular the thermostable direct hemolysin (TDH), the TDH-related hemolysin (TRH), and the type 3 secretion system, and the possible importance of these features in V. parahaemolyticus pathogenicity is explored. The impact of new and much more effective PCR primers on V. parahaemolyticus detection and our views of virulent strain abundance are also described. It is clear that strains carrying the canonical virulence genes are far more common than previously thought, which opens questions regarding the role of these genes in pathogenesis. It is also clear that virulence is an evolving feature of V. parahaemolyticus and that novel combinations of virulence factors can lead to emergent virulence in which a strain that is markedly more pathogenic evolves and propagates to produce an outbreak. The effects of global climate change on the frequency of epidemic disease, the geographic distribution of outbreaks, and the human impacts of V. parahaemolyticus are increasing and this review provides information on why this ubiquitous human pathogen has increased its footprint and its significance so dramatically.
Collapse
|
33
|
Viala JP, Prima V, Puppo R, Agrebi R, Canestrari MJ, Lignon S, Chauvin N, Méresse S, Mignot T, Lebrun R, Bouveret E. Acylation of the Type 3 Secretion System Translocon Using a Dedicated Acyl Carrier Protein. PLoS Genet 2017; 13:e1006556. [PMID: 28085879 PMCID: PMC5279801 DOI: 10.1371/journal.pgen.1006556] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/30/2017] [Accepted: 12/29/2016] [Indexed: 12/18/2022] Open
Abstract
Bacterial pathogens often deliver effectors into host cells using type 3 secretion systems (T3SS), the extremity of which forms a translocon that perforates the host plasma membrane. The T3SS encoded by Salmonella pathogenicity island 1 (SPI-1) is genetically associated with an acyl carrier protein, IacP, whose role has remained enigmatic. In this study, using tandem affinity purification, we identify a direct protein-protein interaction between IacP and the translocon protein SipB. We show, by mass spectrometry and radiolabelling, that SipB is acylated, which provides evidence for a modification of the translocon that has not been described before. A unique and conserved cysteine residue of SipB is identified as crucial for this modification. Although acylation of SipB was not essential to virulence, we show that this posttranslational modification promoted SipB insertion into host-cell membranes and pore-forming activity linked to the SPI-1 T3SS. Cooccurrence of acyl carrier and translocon proteins in several γ- and β-proteobacteria suggests that acylation of the translocon is conserved in these other pathogenic bacteria. These results also indicate that acyl carrier proteins, known for their involvement in metabolic pathways, have also evolved as cofactors of new bacterial protein lipidation pathways. Acyl carrier proteins are small ubiquitous proteins involved in the synthesis of hydrocarbon based molecules. Notably, they are essential for the synthesis of fatty acids, which are the precursors of membrane phospholipids. They can also be involved in secondary metabolism, for example for the synthesis of molecules with antibacterial properties. Although acyl carrier proteins are widespread, the specific role of each individual protein seems comparatively poorly explored. In this study, we investigate the role of an acyl carrier protein genetically associated with a type 3 secretion system (T3SS). Many Gram-negative bacterial pathogens use T3SS to deliver effectors directly into the cytoplasm of eukaryotic host cells and to subvert host cellular pathways. For this purpose, the translocon, which is the terminal part of T3SS, forms a pore inserted into the host-cell membrane. Here we show that the acyl carrier protein associated with the T3SS has specialized to allow acylation of the translocon. The novel posttranslational modification of the translocon that we describe optimizes insertion into the host-cell membrane and pore-forming activity. This mechanism is likely to be conserved in other pathogenic bacteria given the conserved genetic association between T3SS and acyl carrier protein in several bacteria.
Collapse
Affiliation(s)
- Julie P. Viala
- Aix Marseille Univ, CNRS, IMM, LISM, Marseille, France
- * E-mail:
| | - Valérie Prima
- Aix Marseille Univ, CNRS, IMM, LISM, Marseille, France
| | - Rémy Puppo
- Aix Marseille Univ, CNRS, IMM, Proteomic Platform- IBISA, Marseille, France
| | - Rym Agrebi
- Aix Marseille Univ, CNRS, IMM, LCB, Marseille, France
| | | | - Sabrina Lignon
- Aix Marseille Univ, CNRS, IMM, Proteomic Platform- IBISA, Marseille, France
| | | | | | - Tâm Mignot
- Aix Marseille Univ, CNRS, IMM, LCB, Marseille, France
| | - Régine Lebrun
- Aix Marseille Univ, CNRS, IMM, Proteomic Platform- IBISA, Marseille, France
| | | |
Collapse
|
34
|
Mittal R, Lisi CV, Kumari H, Grati M, Blackwelder P, Yan D, Jain C, Mathee K, Weckwerth PH, Liu XZ. Otopathogenic Pseudomonas aeruginosa Enters and Survives Inside Macrophages. Front Microbiol 2016; 7:1828. [PMID: 27917157 PMCID: PMC5114284 DOI: 10.3389/fmicb.2016.01828] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
Otitis media (OM) is a broad term describing a group of infectious and inflammatory disorders of the middle ear. Despite antibiotic therapy, acute OM can progress to chronic suppurative otitis media (CSOM) characterized by ear drum perforation and purulent discharge. Pseudomonas aeruginosa is the most common pathogen associated with CSOM. Although, macrophages play an important role in innate immune responses but their role in the pathogenesis of P. aeruginosa-induced CSOM is not known. The objective of this study is to examine the interaction of P. aeruginosa with primary macrophages. We observed that P. aeruginosa enters and multiplies inside human and mouse primary macrophages. This bacterial entry in macrophages requires both microtubule and actin dependent processes. Transmission electron microscopy demonstrated that P. aeruginosa was present in membrane bound vesicles inside macrophages. Interestingly, deletion of oprF expression in P. aeruginosa abrogates its ability to survive inside macrophages. Our results suggest that otopathogenic P. aeruginosa entry and survival inside macrophages is OprF-dependent. The survival of bacteria inside macrophages will lead to evasion of killing and this lack of pathogen clearance by phagocytes contributes to the persistence of infection in CSOM. Understanding host-pathogen interaction will provide novel avenues to design effective treatment modalities against OM.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Christopher V Lisi
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Hansi Kumari
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami FL, USA
| | - M'hamed Grati
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Patricia Blackwelder
- Chemistry Department, Center for Advanced Microscopy, University of Miami, Coral GablesFL, USA; Rosenstiel School of Marine and Atmospheric Science, University of Miami, Key BiscayneFL, USA
| | - Denise Yan
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Chaitanya Jain
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, MiamiFL, USA; Global Health Consortium and Biomolecular Science Institute, Florida International University, MiamiFL, USA
| | - Paulo H Weckwerth
- Health Sciences Department, University of Sagrado Coração Bauru, Brazil
| | - Xue Z Liu
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| |
Collapse
|
35
|
Type I interferon promotes cell-to-cell spread ofListeria monocytogenes. Cell Microbiol 2016; 19. [DOI: 10.1111/cmi.12660] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 01/06/2023]
|
36
|
Popa C, Coll NS, Valls M, Sessa G. Yeast as a Heterologous Model System to Uncover Type III Effector Function. PLoS Pathog 2016; 12:e1005360. [PMID: 26914889 PMCID: PMC4767418 DOI: 10.1371/journal.ppat.1005360] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Type III effectors (T3E) are key virulence proteins that are injected by bacterial pathogens inside the cells of their host to subvert cellular processes and contribute to disease. The budding yeast Saccharomyces cerevisiae represents an important heterologous system for the functional characterisation of T3E proteins in a eukaryotic environment. Importantly, yeast contains eukaryotic processes with low redundancy and are devoid of immunity mechanisms that counteract T3Es and mask their function. Expression in yeast of effectors from both plant and animal pathogens that perturb conserved cellular processes often resulted in robust phenotypes that were exploited to elucidate effector functions, biochemical properties, and host targets. The genetic tractability of yeast and its amenability for high-throughput functional studies contributed to the success of this system that, in recent years, has been used to study over 100 effectors. Here, we provide a critical view on this body of work and describe advantages and limitations inherent to the use of yeast in T3E research. “Favourite” targets of T3Es in yeast are cytoskeleton components and small GTPases of the Rho family. We describe how mitogen-activated protein kinase (MAPK) signalling, vesicle trafficking, membrane structures, and programmed cell death are also often altered by T3Es in yeast and how this reflects their function in the natural host. We describe how effector structure–function studies and analysis of candidate targeted processes or pathways can be carried out in yeast. We critically analyse technologies that have been used in yeast to assign biochemical functions to T3Es, including transcriptomics and proteomics, as well as suppressor, gain-of-function, or synthetic lethality screens. We also describe how yeast can be used to select for molecules that block T3E function in search of new antibacterial drugs with medical applications. Finally, we provide our opinion on the limitations of S. cerevisiae as a model system and its most promising future applications.
Collapse
Affiliation(s)
- Crina Popa
- Genetics Department, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centre for Research in Agricultural Genomics (CSIC-IRTA-UAB-UB), Bellaterra, Catalonia, Spain
| | - Núria S. Coll
- Centre for Research in Agricultural Genomics (CSIC-IRTA-UAB-UB), Bellaterra, Catalonia, Spain
| | - Marc Valls
- Genetics Department, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centre for Research in Agricultural Genomics (CSIC-IRTA-UAB-UB), Bellaterra, Catalonia, Spain
- * E-mail: (GS); (MV)
| | - Guido Sessa
- Department of Molecular Biology and Ecology of Plants, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (GS); (MV)
| |
Collapse
|
37
|
Koch M, Baum J. The mechanics of malaria parasite invasion of the human erythrocyte - towards a reassessment of the host cell contribution. Cell Microbiol 2016; 18:319-29. [PMID: 26663815 PMCID: PMC4819681 DOI: 10.1111/cmi.12557] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 01/15/2023]
Abstract
Despite decades of research, we still know little about the mechanics of Plasmodium host cell invasion. Fundamentally, while the essential or non‐essential nature of different parasite proteins is becoming clearer, their actual function and how each comes together to govern invasion are poorly understood. Furthermore, in recent years an emerging world view is shifting focus away from the parasite actin–myosin motor being the sole force responsible for entry to an appreciation of host cell dynamics and forces and their contribution to the process. In this review, we discuss merozoite invasion of the erythrocyte, focusing on the complex set of pre‐invasion events and how these might prime the red cell to facilitate invasion. While traditionally parasite interactions at this stage have been viewed simplistically as mediating adhesion only, recent work makes it apparent that by interacting with a number of host receptors and signalling pathways, combined with secretion of parasite‐derived lipid material, that the merozoite may initiate cytoskeletal re‐arrangements and biophysical changes in the erythrocyte that greatly reduce energy barriers for entry. Seen in this light Plasmodium invasion may well turn out to be a balance between host and parasite forces, much like that of other pathogen infection mechanisms.
Collapse
Affiliation(s)
- Marion Koch
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| |
Collapse
|