1
|
Carrara GMP, Souza-Silva GA, Reis TCBD, Alencar BCD, Boscardin SB, Kima PE, Stolf BS. Macrophage Protein Disulfide Isomerase Increases Infection by Leishmania amazonensis. Cell Biol Int 2025. [PMID: 40178024 DOI: 10.1002/cbin.70020] [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: 08/19/2024] [Revised: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Leishmania spp. are protozoans with a digenetic life cycle responsible for causing tegumentary and visceral leishmaniasis. Leishmania (L.) amazonensis is the second most prevalent dermotropic species in Brazil. Infection in humans and other mammals takes place when phagocytes, mainly macrophages, uptake the parasite. Many proteins on the phagocytic cell surface participate in Leishmania phagocytosis. In this study, we evaluated the role of surface protein disulfide isomerase (PDI) in phagocytosis and infection of macrophages by L. amazonensis. PDI is the second most abundant chaperone in the endoplasmic reticulum. A unique study in the literature associated the presence of PDI on the macrophage surface with increased phagocytosis by Leishmania (L.) infantum (syn L. chagasi), the species most frequently associated with visceral leishmaniasis in the Americas. In the present work we evaluated L. amazonensis infections in transgenic FVB/NJ mice overexpressing PDI (TgPDIA1). We validated the presence of PDI on their macrophages surface by flow cytometry. We demonstrated that infection of macrophages pretreated with anti-PDI antibodies was lower compared to control cells. Accordingly, we showed that the overexpression of PDI increased the adhesion of parasites and infection of macrophages. We also demonstrated that macrophages overexpressing PDI internalize more zymosan particles. In vivo imaging of infections with luciferase-expressing parasites in wild-type and TgPDIA1 mice indicated that the overexpression of PDI was not associated with significant differences in footpad lesions and parasite burden, probably due to the ubiquitous overexpression of PDI and the roles of this molecule in other immune system functions.
Collapse
Affiliation(s)
- Guilherme M P Carrara
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Guilherme A Souza-Silva
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Tania C B D Reis
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Bruna C D Alencar
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Silvia B Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Peter E Kima
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Beatriz S Stolf
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Licht P, Mailänder V. Multi-Omic Data Integration Suggests Putative Microbial Drivers of Aetiopathogenesis in Mycosis Fungoides. Cancers (Basel) 2024; 16:3947. [PMID: 39682136 DOI: 10.3390/cancers16233947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Mycosis fungoides (MF) represents the most prevalent entity of cutaneous T cell lymphoma (CTCL). The MF aetiopathogenesis is incompletely understood, due to significant transcriptomic heterogeneity and conflicting views on whether oncologic transformation originates in early thymocytes or mature effector memory T cells. Recently, using clinical specimens, our group showed that the skin microbiome aggravates disease course, mainly driven by an outgrowing, pathogenic S. aureus strain carrying the virulence factor spa, which was shown by others to activate the T cell signalling pathway NF-κB. METHODS To explore the role of the skin microbiome in MF aetiopathogenesis, we here performed RNA sequencing, multi-omic data integration of the skin microbiome and skin transcriptome using Multi-Omic Factor Analysis (MOFA), virome profiling, and T cell receptor (TCR) sequencing in 10 MF patients from our previous study group. RESULTS We observed that inter-patient transcriptional heterogeneity may be largely attributed to differential activation of T cell signalling pathways. Notably, the MOFA model resolved the heterogenous activation pattern of T cell signalling after denoising the transcriptome from microbial influence. The MOFA model suggested that the outgrowing S. aureus strain evoked signalling by non-canonical NF-κB and IL-1B, which in turn may have fuelled the aggravated disease course. Further, the MOFA model indicated aberrant pathways of early thymopoiesis alongside enrichment of antiviral innate immunity. In line with this, viral prevalence, particularly of Epstein-Barr virus (EBV), trended higher in both lesional skin and the blood compared to nonlesional skin. Additionally, TCRs in both MF skin lesions and the blood were significantly more likely to recognize EBV peptides involved in latent infection. CONCLUSIONS First, our findings suggest that S. aureus with its virulence factor spa fuels MF progression through non-canonical NF-κB and IL-1B signalling. Second, our data provide insights into the potential role of viruses in MF aetiology. Last, we propose a model of microbiome-driven MF aetiopathogenesis: Thymocytes undergo initial oncologic transformation, potentially caused by viruses. After maturation and skin infiltration, an outgrowing, pathogenic S. aureus strain evokes activation and maturation into effector memory T cells, resulting in aggressive disease. Further studies are warranted to verify and extend our data, which are based on computational analyses.
Collapse
Affiliation(s)
- Philipp Licht
- Department of Dermatology, University Medical Centre Mainz, 55131 Mainz, Germany
| | - Volker Mailänder
- Department of Dermatology, University Medical Centre Mainz, 55131 Mainz, Germany
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| |
Collapse
|
3
|
Wang F, Zhong ZR, Xie Q, Ou J, Xiong NX, Huang MZ, Li SY, Hu G, Qin ZL, Luo SW. Multiomics Analyses Explore the Immunometabolic Interplay in the Liver of White Crucian Carp (Carassius cuvieri) After Aeromonas veronii Challenge. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:790-809. [PMID: 39042324 DOI: 10.1007/s10126-024-10347-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/13/2024] [Indexed: 07/24/2024]
Abstract
Aeromonas veronii is one of the predominant pathogenic species that can imperil the survival of farmed fish. However, the interactive networks of immune regulation and metabolic response in A. veronii-infected fish are still unclear. In this investigation, we aimed to explore immunometabolic interplay in white crucian carp (WCC) after the A. veronii challenge. Elevated levels of immune-related genes were observed in various tissues after A. veronii infection, along with the sharp alteration of disease-related enzymatic activities. Besides, decreased levels of antioxidant status were observed in the liver, but most metabolic gene expressions increased dramatically. Multiomics analyses revealed that metabolic products of amino acids, such as formiminoglutamic acid (FIGLU), L-glutamate (L-Glu), and 4-hydroxyhippuric acid, were considered the crucial liver biomarkers in A. veronii-infected WCC. In addition, A. veronii infection may dysregulate endoplasmic reticulum (ER) function to affect the metabolic process of lipids, carbohydrates, and amino acids in the liver of WCC. These results may have a comprehensive implication for understanding immunometabolic response in WCC upon A. veronii infection.
Collapse
Affiliation(s)
- Fei Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Zi-Rou Zhong
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Qing Xie
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Jie Ou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Ning-Xia Xiong
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ming-Zhu Huang
- National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, 330022, China
| | - Shi-Yun Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Gang Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Zi-Le Qin
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China
| | - Sheng-Wei Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, Engineering Research Center of Polyploidy Fish Reproduction and Breeding of the State Education Ministry, College of Life Science, Hunan Normal University, Changsha, 410081, People's Republic of China.
| |
Collapse
|
4
|
Schroeder EA, Toro-Moreno M, Raphemot R, Sylvester K, Colón IC, Derbyshire ER. Toxoplasma and Plasmodium associate with host Arfs during infection. mSphere 2024; 9:e0077023. [PMID: 38349168 PMCID: PMC10964417 DOI: 10.1128/msphere.00770-23] [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: 12/18/2023] [Accepted: 01/17/2024] [Indexed: 03/27/2024] Open
Abstract
The apicomplexans Toxoplasma gondii and Plasmodium are intracellular parasites that reside within a host-derived compartment termed the parasitophorous vacuole (PV). During infection, the parasites must acquire critical host resources and transport them across their PV for development. However, the mechanism by which host resources are trafficked to and across the PV remains uncertain. Here, we investigated host ADP ribosylation factors (Arfs), a class of proteins involved in vesicular trafficking that may be exploited by T. gondii and Plasmodium berghei for nutrient acquisition. Using overexpressed Arf proteins coupled with immunofluorescence microscopy, we found that all Arfs were internalized into the T. gondii PV, with most vacuoles containing at least one punctum of Arf protein by the end of the lytic cycle. We further characterized Arf1, the most abundant Arf inside the T. gondii PV, and observed that active recycling between its GDP/GTP-bound state influenced Arf1 internalization independent of host guanine nucleotide exchange factors (GEFs). In addition, Arf1 colocalized with vesicle coat complexes and exogenous sphingolipids, suggesting a role in nutrient acquisition. While Arf1 and Arf4 were not observed inside the PV during P. berghei infection, our gene depletion studies showed that liver stage development and survival depended on the expression of Arf4 and the host GEF, GBF1. Collectively, these observations indicate that apicomplexans use distinct mechanisms to subvert the host vesicular trafficking network and efficiently replicate. The findings also pave the way for future studies to identify parasite proteins critical to host vesicle recruitment and the components of vesicle cargo. IMPORTANCE The parasites Toxoplasma gondii and Plasmodium live complex intracellular lifestyles where they must acquire essential host nutrients while avoiding recognition. Although previous work has sought to identify the specific nutrients scavenged by apicomplexans, the mechanisms by which host materials are transported to and across the parasite vacuole membrane are largely unknown. Here, we examined members of the host vesicular trafficking network to identify specific pathways subverted by T. gondii and Plasmodium berghei. Our results indicate that T. gondii selectively internalizes host Arfs, a class of proteins involved in intracellular trafficking. For P. berghei, host Arfs were restricted by the parasite's vacuole membrane, but proteins involved in vesicular trafficking were identified as essential for liver stage development. A greater exploration into how and why apicomplexans subvert host vesicular trafficking could help identify targets for host-directed therapeutics.
Collapse
Affiliation(s)
- Erin A. Schroeder
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Maria Toro-Moreno
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | - Rene Raphemot
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Isabel C. Colón
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | - Emily R. Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| |
Collapse
|
5
|
Cudjoe O, Afful R, Hagan TA. Toxoplasma-host endoplasmic reticulum interaction: How T. gondii activates unfolded protein response and modulates immune response. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 6:100223. [PMID: 38352129 PMCID: PMC10861954 DOI: 10.1016/j.crmicr.2024.100223] [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: 02/16/2024] Open
Abstract
Toxoplasma gondii is a neurotropic single-celled zoonotic parasite that can infect human beings and animals. Infection with T. gondii is usually asymptomatic in immune-competent individual, however, it can cause symptomatic and life-threatening conditions in immunocompromised individuals and in developing foetuses. Although the mechanisms that allow T. gondii to persist in host cells are poorly understood, studies in animal models have greatly improved our understanding of Toxoplasma-host cell interaction and how this interaction modulates parasite proliferation and development, host immune response and virulence of the parasite. T. gondii is capable of recruiting the host endoplasmic reticulum (ER), suggesting it may influence the host ER function. Herein, we provide an overview of T. gondii infection and the role of host ER during stressed conditions. Furthermore, we highlight studies that explore T. gondii's interaction with the host ER. We delve into how this interaction activates the unfolded protein response (UPR) and ER stress-mediated apoptosis. Additionally, we examine how T. gondii exploits these pathways to its advantage.
Collapse
Affiliation(s)
- Obed Cudjoe
- Department of Medical Laboratory Science, Klintaps College of Health and Allied Sciences, DTD TDC Plot 30A, Klagon, Tema, Ghana
- Department of Microbiology and Immunology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana
| | - Roger Afful
- Department of Medical Laboratory Science, Klintaps College of Health and Allied Sciences, DTD TDC Plot 30A, Klagon, Tema, Ghana
| | - Tonny Abraham Hagan
- Department of Biomedical Engineering, School of Life Science and Technology, University of Electronic Science and Technology of China, China
| |
Collapse
|
6
|
Ascari A, Frölich S, Zang M, Tran ENH, Wilson DW, Morona R, Eijkelkamp BA. Shigella flexneri remodeling and consumption of host lipids during infection. J Bacteriol 2023; 205:e0032023. [PMID: 37991380 PMCID: PMC10729657 DOI: 10.1128/jb.00320-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
IMPORTANCE Bacterial pathogens have vastly distinct sites that they inhabit during infection. This requires adaptation due to changes in nutrient availability and antimicrobial stress. The bacterial surface is a primary barrier, and here, we show that the bacterial pathogen Shigella flexneri increases its surface decorations when it transitions to an intracellular lifestyle. We also observed changes in bacterial and host cell fatty acid homeostasis. Specifically, intracellular S. flexneri increased the expression of their fatty acid degradation pathway, while the host cell lipid pool was significantly depleted. Importantly, bacterial proliferation could be inhibited by fatty acid supplementation of host cells, thereby providing novel insights into the possible link between human malnutrition and susceptibility to S. flexneri.
Collapse
Affiliation(s)
- Alice Ascari
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| | - Sonja Frölich
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia
| | - Maoge Zang
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| | - Elizabeth N. H. Tran
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - Danny W. Wilson
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia
| | - Renato Morona
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - Bart A. Eijkelkamp
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| |
Collapse
|
7
|
Kabir AR, Chaudhary AA, Aladwani MO, Podder S. Decoding the host-pathogen interspecies molecular crosstalk during oral candidiasis in humans: an in silico analysis. Front Genet 2023; 14:1245445. [PMID: 37900175 PMCID: PMC10603195 DOI: 10.3389/fgene.2023.1245445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction: The objective of this study is to investigate the interaction between Candida albicans and human proteins during oral candidiasis, with the aim of identifying pathways through which the pathogen subverts host cells. Methods: A comprehensive list of interactions between human proteins and C. albicans was obtained from the Human Protein Interaction Database using specific screening criteria. Then, the genes that exhibit differential expression during oral candidiasis in C. albicans were mapped with the list of human-Candida interactions to identify the corresponding host proteins. The identified host proteins were further compared with proteins specific to the tongue, resulting in a final list of 99 host proteins implicated in oral candidiasis. The interactions between host proteins and C. albicans proteins were analyzed using the STRING database, enabling the construction of protein-protein interaction networks. Similarly, the gene regulatory network of Candida proteins was reconstructed using data from the PathoYeastract and STRING databases. Core module proteins within the targeted host protein-protein interaction network were identified using ModuLand, a Cytoscape plugin. The expression levels of the core module proteins under diseased conditions were assessed using data from the GSE169278 dataset. To gain insights into the functional characteristics of both host and pathogen proteins, ontology analysis was conducted using Enrichr and YeastEnrichr, respectively. Result: The analysis revealed that three Candida proteins, HHT21, CYP5, and KAR2, interact with three core host proteins, namely, ING4 (in the DNMT1 module), SGTA, and TOR1A. These interactions potentially impair the immediate immune response of the host against the pathogen. Additionally, differential expression analysis of fungal proteins and their transcription factors in Candida-infected oral cell lines indicated that Rob1p, Tye7p, and Ume6p could be considered candidate transcription factors involved in instigating the pathogenesis of oral candidiasis during host infection. Conclusion: Our study provides a molecular map of the host-pathogen interaction during oral candidiasis, along with potential targets for designing regimens to overcome oral candidiasis, particularly in immunocompromised individuals.
Collapse
Affiliation(s)
- Ali Rejwan Kabir
- Computational and System Biology Lab, Department of Microbiology, Raiganj University, Raiganj, West Bengal, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Malak O Aladwani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Soumita Podder
- Computational and System Biology Lab, Department of Microbiology, Raiganj University, Raiganj, West Bengal, India
| |
Collapse
|
8
|
Liu M, Zhao Y, Shi Z, Zink JI, Yu Q. Virus-like Magnetic Mesoporous Silica Particles as a Universal Vaccination Platform against Pathogenic Infections. ACS NANO 2023; 17:6899-6911. [PMID: 36961475 DOI: 10.1021/acsnano.3c00644] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Vaccination is the most important way of population protection from life-threatening pathogenic infections. However, its efficiency is frequently compromised by a failure of strong antigen presentation and immune activation. Herein, we developed virus-like magnetic mesoporous silica nanoparticles as a universal vaccination platform (termed MagParV) for preventing pathogenic infections. This platform was constructed by integrating synthetic biology-based endoplasmic reticulum-targeting vesicles with magnetic mesoporous silica particles. This platform exhibited high antigen-loading capacity, strongly targeting the endoplasmic reticulum and promoting antigen presentation in dendritic cells. After prime-boost vaccination, the antigen-loading MagParV with AMF drastically elicited specific antibody production against corresponding antigens of fungal, bacterial, and viral pathogens. A systemic infection model further revealed that the platform effectively protected the mice from severe fungal systemic infections. This study realized synthetic biology-facilitated green manufacturing of vaccines, which is promising for magnetism-activated vaccination against different kinds of pathogenic infections.
Collapse
Affiliation(s)
- Mingyang Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, People's Republic of China
| | - Yan Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Zhishang Shi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- California Nano Systems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
9
|
Vinogradskaya GR, Ivanov AV, Kushch AA. Mechanisms of Survival of Cytomegalovirus-Infected Tumor Cells. Mol Biol 2022; 56:668-683. [PMID: 36217337 PMCID: PMC9534468 DOI: 10.1134/s0026893322050132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 11/04/2022]
Abstract
Human cytomegalovirus (HCMV) DNA and proteins are often detected in malignant tumors, warranting studies of the role that HCMV plays in carcinogenesis and tumor progression. HCMV proteins were shown to regulate the key processes involved in tumorigenesis. While HCMV as an oncogenic factor just came into focus, its ability to promote tumor progression is generally recognized. The review discusses the viral factors and cell molecular pathways that affect the resistance of cancer cells to therapy. CMV inhibits apoptosis of tumor cells, that not only promotes tumor progression, but also reduces the sensitivity of cells to antitumor therapy. Autophagy was found to facilitate either cell survival or cell death in different tumor cells. In leukemia cells, HCMV induces a "protective" autophagy that suppresses apoptosis. Viral factors that mediate drug resistance and their interactions with key cell death pathways are necessary to further investigate in order to develop agents that can restore the tumor sensitivity to anticancer drugs.
Collapse
Affiliation(s)
- G. R. Vinogradskaya
- Konstantinov St. Petersburg Institute of Nuclear Physics, National Research Center “Kurchatov Institute”, 188300 Gatchina, Leningrad oblast Russia
| | - A. V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A. A Kushch
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| |
Collapse
|
10
|
Mutations of γCOP Gene Disturb Drosophila melanogaster Innate Immune Response to Pseudomonas aeruginosa. Int J Mol Sci 2022; 23:ijms23126499. [PMID: 35742941 PMCID: PMC9223523 DOI: 10.3390/ijms23126499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 01/27/2023] Open
Abstract
Drosophila melanogaster (the fruit fly) is a valuable experimental platform for modeling host–pathogen interactions. It is also commonly used to define innate immunity pathways and to understand the mechanisms of both host tolerance to commensal microbiota and response to pathogenic agents. Herein, we investigate how the host response to bacterial infection is mirrored in the expression of genes of Imd and Toll pathways when D. melanogaster strains with different γCOP genetic backgrounds are infected with Pseudomonas aeruginosa ATCC 27853. Using microarray technology, we have interrogated the whole-body transcriptome of infected versus uninfected fruit fly males with three specific genotypes, namely wild-type Oregon, γCOPS057302/TM6B and γCOP14a/γCOP14a. While the expression of genes pertaining to Imd and Toll is not significantly modulated by P. aeruginosa infection in Oregon males, many of the components of these cascades are up- or downregulated in both infected and uninfected γCOPS057302/TM6B and γCOP14a/γCOP14a males. Thus, our results suggest that a γCOP genetic background modulates the gene expression profiles of Imd and Toll cascades involved in the innate immune response of D. melanogaster, inducing the occurrence of immunological dysfunctions in γCOP mutants.
Collapse
|
11
|
Jaldin-Fincati J, Moussaoui S, Gimenez MC, Ho CY, Lancaster CE, Botelho R, Ausar F, Brookes R, Terebiznik M. Aluminum hydroxide adjuvant diverts the uptake and trafficking of genetically detoxified pertussis toxin to lysosomes in macrophages. Mol Microbiol 2022; 117:1173-1195. [PMID: 35344242 PMCID: PMC9321756 DOI: 10.1111/mmi.14900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022]
Abstract
Aluminum salts have been successfully utilized as adjuvants to enhance the immunogenicity of vaccine antigens since the 1930s. However, the cellular mechanisms behind the immune adjuvanticity effect of these materials in antigen‐presenting cells are poorly understood. In this study, we investigated the uptake and trafficking of aluminum oxy‐hydroxide (AlOOH), in RAW 264.7 murine and U‐937 human macrophages‐like cells. Furthermore, we determined the impact that the adsorption to AlOOH particulates has on the trafficking of a Bordetella pertussis vaccine candidate, the genetically detoxified pertussis toxin (gdPT). Our results indicate that macrophages internalize AlOOH by constitutive macropinocytosis assisted by the filopodial protrusions that capture the adjuvant particles. Moreover, we show that AlOOH has the capacity to nonspecifically adsorb IgG, engaging opsonic phagocytosis, which is a feature that may allow for more effective capture and uptake of adjuvant particles by antigen‐presenting cells (APCs) at the site of vaccine administration. We found that AlOOH traffics to endolysosomal compartments that hold degradative properties. Importantly, while we show that gdPT escapes degradative endolysosomes and traffics toward the retrograde pathway, as reported for the wild‐type pertussis toxin, the adsorption to AlOOH diverts gdPT to traffic to the adjuvant’s lysosome‐type compartments, which may be key for MHC‐II‐driven antigen presentation and activation of CD4+ T cell. Thus, our findings establish a direct link between antigen adsorption to AlOOH and the intracellular trafficking of antigens within antigen‐presenting cells and bring to light a new potential mechanism for aluminum adjuvancy. Moreover, the in‐vitro single‐cell approach described herein provides a general framework and tools for understanding critical attributes of other vaccine formulations.
Collapse
Affiliation(s)
- Javier Jaldin-Fincati
- Department of Biological Sciences, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada
| | - Serene Moussaoui
- Department of Biological Sciences, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada.,Department of Cell and Systems Biology, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada
| | - Maria Cecilia Gimenez
- Department of Biological Sciences, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada
| | - Cheuk Y Ho
- Department of Biological Sciences, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada
| | - Charlene E Lancaster
- Department of Biological Sciences, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada.,Department of Cell and Systems Biology, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada
| | - Roberto Botelho
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Fernando Ausar
- BioProcess Research and Development, Sanofi Pasteur, 1755 Steeles Ave West, Toronto, Ontario M3R 3T4, Canada
| | - Roger Brookes
- BioProcess Research and Development, Sanofi Pasteur, 1755 Steeles Ave West, Toronto, Ontario M3R 3T4, Canada
| | - Mauricio Terebiznik
- Department of Biological Sciences, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada.,Department of Cell and Systems Biology, University of Toronto at Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada
| |
Collapse
|
12
|
Harapas CR, Idiiatullina E, Al-Azab M, Hrovat-Schaale K, Reygaerts T, Steiner A, Laohamonthonkul P, Davidson S, Yu CH, Booty L, Masters SL. Organellar homeostasis and innate immune sensing. Nat Rev Immunol 2022; 22:535-549. [PMID: 35197578 DOI: 10.1038/s41577-022-00682-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
A cell is delimited by numerous borders that define specific organelles. The walls of some organelles are particularly robust, such as in mitochondria or endoplasmic reticulum, but some are more fluid such as in phase-separated stress granules. Either way, all organelles can be damaged at times, leading their contents to leak out into the surrounding environment. Therefore, an elegant way to construct an innate immune defence system is to recognize host molecules that do not normally reside within a particular compartment. Here, we provide several examples where organellar homeostasis is lost, leading to the activation of a specific innate immune sensor; these include NLRP3 activation owing to a disrupted trans-Golgi network, Pyrin activation due to cytoskeletal damage, and cGAS-STING activation following the leakage of nuclear or mitochondrial DNA. Frequently, organelle damage is observed downstream of pathogenic infection but it can also occur in sterile settings as associated with auto-inflammatory disease. Therefore, understanding organellar homeostasis is central to efforts that will identify new innate immune pathways, and therapeutics that balance organellar homeostasis, or target the breakdown pathways that trigger innate immune sensors, could be useful treatments for infection and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Cassandra R Harapas
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Elina Idiiatullina
- Immunology Laboratory, Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong, China
| | - Mahmoud Al-Azab
- Immunology Laboratory, Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong, China
| | - Katja Hrovat-Schaale
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Thomas Reygaerts
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Annemarie Steiner
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Pawat Laohamonthonkul
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Sophia Davidson
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Chien-Hsiung Yu
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Lee Booty
- Immunology Network, Immunology Research Unit, GSK, Stevenage, UK
| | - Seth L Masters
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia. .,Immunology Laboratory, Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong, China.
| |
Collapse
|
13
|
Lin CY, Achor D, Levy A. Intracellular Life Cycle of ' Candidatus Liberibacter asiaticus' Inside Psyllid Gut Cells. PHYTOPATHOLOGY 2022; 112:145-153. [PMID: 34689612 DOI: 10.1094/phyto-07-21-0301-fi] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
'Candidatus Liberibacter asiaticus' (CLas), the devastating pathogen related to Huanglongbing (HLB), is a phloem-limited, fastidious, insect-borne bacterium. Rapid spread of HLB disease relies on CLas-efficient propagation in the vector, the Asian citrus psyllid Diaphorina citri, in a circulative manner. Understanding the intracellular lifecycle of CLas in psyllid midgut, the major organ for CLas transmission, is fundamental to improving current management strategies. Using a microscopic approach within CLas-infected insect midgut, we observed the entry of CLas into gut cells inside vesicles, termed Liberibacter-containing vacuoles (LCVs), by endocytosis. Endocytosis is followed by the formation of endoplasmic reticulum-related and replication permissive vacuoles (rLCVs). Additionally, we observed the formation of double membrane autophagosome-like structure, termed autophagy-related vacuole (aLCV). Vesicles containing CLas egress from aLCV and fuse with the cell membrane. Immunolocalization studies showed that CLas uses endocytosis- and exocytosis-like mechanisms that mediates bacterial invasion and egress. Upregulation of autophagy-related genes indicated subversion of host autophagy by CLas in psyllid vector to promote infection. These results indicate that CLas interacts with host cellular machineries to undergo a multistage intracellular cycle through endocytic, secretory, autophagic, and exocytic pathways via complex machineries. Potential tactics for HLB control can be made depending on further investigations on the knowledge of the molecular mechanisms of CLas intracellular cycle.
Collapse
Affiliation(s)
- Chun-Yi Lin
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL 33850
| | - Diann Achor
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL 33850
| | - Amit Levy
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL 33850
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611
| |
Collapse
|
14
|
Li J, Gao E, Xu C, Wang H, Wei Y. ER-Phagy and Microbial Infection. Front Cell Dev Biol 2021; 9:771353. [PMID: 34912806 PMCID: PMC8667338 DOI: 10.3389/fcell.2021.771353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum (ER) is an essential organelle in cells that synthesizes, folds and modifies membrane and secretory proteins. It has a crucial role in cell survival and growth, thus requiring strict control of its quality and homeostasis. Autophagy of the ER fragments, termed ER-phagy or reticulophagy, is an essential mechanism responsible for ER quality control. It transports stress-damaged ER fragments as cargo into the lysosome for degradation to eliminate unfolded or misfolded protein aggregates and membrane lipids. ER-phagy can also function as a host defense mechanism when pathogens infect cells, and its deficiency facilitates viral infection. This review briefly describes the process and regulatory mechanisms of ER-phagy, and its function in host anti-microbial defense during infection.
Collapse
Affiliation(s)
- Jiahui Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou, China
| | - Enfeng Gao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou, China
| | - Chenguang Xu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou, China
| | - Hongna Wang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou, China.,GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yongjie Wei
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou, China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| |
Collapse
|
15
|
Almeida C. A potential third-order role of the host endoplasmic reticulum as a contact site in interkingdom microbial endosymbiosis and viral infection. ENVIRONMENTAL MICROBIOLOGY REPORTS 2021; 13:255-271. [PMID: 33559322 DOI: 10.1111/1758-2229.12938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 06/12/2023]
Abstract
The normal functioning of eukaryotic cells depends on the compartmentalization of metabolic processes within specific organelles. Interactions among organelles, such as those between the endoplasmic reticulum (ER) - considered the largest single structure in eukaryotic cells - and other organelles at membrane contact sites (MCSs) have also been suggested to trigger synergisms, including intracellular immune responses against pathogens. In addition to the ER-endogenous functions and ER-organelle MCSs, we present the perspective of a third-order role of the ER as a host contact site for endosymbiotic microbial non-pathogens and pathogens, from endosymbiont bacteria to parasitic protists and viruses. Although understudied, ER-endosymbiont interactions have been observed in a range of eukaryotic hosts, including protists, plants, algae, and metazoans. Host ER interactions with endosymbionts could be an ER function built from ancient, conserved mechanisms selected for communicating with mutualistic endosymbionts in specific life cycle stages, and they may be exploited by pathogens and parasites. The host ER-'guest' interactome and traits in endosymbiotic biology are briefly discussed. The acknowledgment and understanding of these possible mechanisms might reveal novel evolutionary perspectives, uncover the causes of unexplained cellular disorders and suggest new pharmacological targets.
Collapse
Affiliation(s)
- Celso Almeida
- ENDOBIOS Biotech®, Praceta Progresso Clube n° 6, 2725-110 Mem-Martins, Portugal
| |
Collapse
|
16
|
De Niz M, Caldelari R, Kaiser G, Zuber B, Heo WD, Heussler VT, Agop-Nersesian C. Hijacking of the host cell Golgi by Plasmodium berghei liver stage parasites. J Cell Sci 2021; 134:jcs252213. [PMID: 34013963 PMCID: PMC8186485 DOI: 10.1242/jcs.252213] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 04/12/2021] [Indexed: 12/28/2022] Open
Abstract
The intracellular lifestyle represents a challenge for the rapidly proliferating liver stage Plasmodium parasite. In order to scavenge host resources, Plasmodium has evolved the ability to target and manipulate host cell organelles. Using dynamic fluorescence-based imaging, we here show an interplay between the pre-erythrocytic stages of Plasmodium berghei and the host cell Golgi during liver stage development. Liver stage schizonts fragment the host cell Golgi into miniaturized stacks, which increases surface interactions with the parasitophorous vacuolar membrane of the parasite. Expression of specific dominant-negative Arf1 and Rab GTPases, which interfere with the host cell Golgi-linked vesicular machinery, results in developmental delay and diminished survival of liver stage parasites. Moreover, functional Rab11a is critical for the ability of the parasites to induce Golgi fragmentation. Altogether, we demonstrate that the structural integrity of the host cell Golgi and Golgi-associated vesicular traffic is important for optimal pre-erythrocytic development of P. berghei. The parasite hijacks the Golgi structure of the hepatocyte to optimize its own intracellular development. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Gesine Kaiser
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Benoit Zuber
- Institute for Anatomy, University of Bern, CH-3012 Bern, Switzerland
| | - Won Do Heo
- Dept. of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Volker T. Heussler
- Institute of Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | | |
Collapse
|
17
|
González-Espinoza G, Arce-Gorvel V, Mémet S, Gorvel JP. Brucella: Reservoirs and Niches in Animals and Humans. Pathogens 2021; 10:pathogens10020186. [PMID: 33572264 PMCID: PMC7915599 DOI: 10.3390/pathogens10020186] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/18/2023] Open
Abstract
Brucella is an intracellular bacterium that causes abortion, reproduction failure in livestock and leads to a debilitating flu-like illness with serious chronic complications if untreated in humans. As a successful intracellular pathogen, Brucella has developed strategies to avoid recognition by the immune system of the host and promote its survival and replication. In vivo, Brucellae reside mostly within phagocytes and other cells including trophoblasts, where they establish a preferred replicative niche inside the endoplasmic reticulum. This process is central as it gives Brucella the ability to maintain replicating-surviving cycles for long periods of time, even at low bacterial numbers, in its cellular niches. In this review, we propose that Brucella takes advantage of the environment provided by the cellular niches in which it resides to generate reservoirs and disseminate to other organs. We will discuss how the favored cellular niches for Brucella infection in the host give rise to anatomical reservoirs that may lead to chronic infections or persistence in asymptomatic subjects, and which may be considered as a threat for further contamination. A special emphasis will be put on bone marrow, lymph nodes, reproductive and for the first time adipose tissues, as well as wildlife reservoirs.
Collapse
|
18
|
Long RKM, Moriarty KP, Cardoen B, Gao G, Vogl AW, Jean F, Hamarneh G, Nabi IR. Super resolution microscopy and deep learning identify Zika virus reorganization of the endoplasmic reticulum. Sci Rep 2020; 10:20937. [PMID: 33262363 PMCID: PMC7708840 DOI: 10.1038/s41598-020-77170-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023] Open
Abstract
The endoplasmic reticulum (ER) is a complex subcellular organelle composed of diverse structures such as tubules, sheets and tubular matrices. Flaviviruses such as Zika virus (ZIKV) induce reorganization of ER membranes to facilitate viral replication. Here, using 3D super resolution microscopy, ZIKV infection is shown to induce the formation of dense tubular matrices associated with viral replication in the central ER. Viral non-structural proteins NS4B and NS2B associate with replication complexes within the ZIKV-induced tubular matrix and exhibit distinct ER distributions outside this central ER region. Deep neural networks trained to distinguish ZIKV-infected versus mock-infected cells successfully identified ZIKV-induced central ER tubular matrices as a determinant of viral infection. Super resolution microscopy and deep learning are therefore able to identify and localize morphological features of the ER and allow for better understanding of how ER morphology changes due to viral infection.
Collapse
Affiliation(s)
- Rory K M Long
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Kathleen P Moriarty
- School of Computing Science, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Ben Cardoen
- School of Computing Science, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Guang Gao
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - A Wayne Vogl
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - François Jean
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Ghassan Hamarneh
- School of Computing Science, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
| | - Ivan R Nabi
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
19
|
Abstract
Brucellosis is a bacterial disease of domestic animals and humans. The pathogenic ability of Brucella organisms relies on their stealthy strategy and their capacity to replicate within host cells and to induce long-lasting infections. Brucella organisms barely induce neutrophil activation and survive within these leukocytes by resisting microbicidal mechanisms. Very few Brucella-infected neutrophils are found in the target organs, except for the bone marrow, early in infection. Still, Brucella induces a mild reactive oxygen species formation and, through its lipopolysaccharide, promotes the premature death of neutrophils, which release chemokines and express "eat me" signals. This effect drives the phagocytosis of infected neutrophils by mononuclear cells that become thoroughly susceptible to Brucella replication and vehicles for bacterial dispersion. The premature death of the infected neutrophils proceeds without NETosis, necrosis/oncosis, or classical apoptosis morphology. In the absence of neutrophils, the Th1 response exacerbates and promotes bacterial removal, indicating that Brucella-infected neutrophils dampen adaptive immunity. This modulatory effect opens a window for bacterial dispersion in host tissues before adaptive immunity becomes fully activated. However, the hyperactivation of immunity is not without a price, since neutropenic Brucella-infected animals develop cachexia in the early phases of the disease. The delay in the immunological response seems a sine qua non requirement for the development of long-lasting brucellosis. This property may be shared with other pathogenic alphaproteobacteria closely related to Brucella We propose a model in which Brucella-infected polymorphonuclear neutrophils (PMNs) function as "Trojan horse" vehicles for bacterial dispersal and as modulators of the Th1 adaptive immunity in infection.
Collapse
|
20
|
Pehote G, Vij N. Autophagy Augmentation to Alleviate Immune Response Dysfunction, and Resolve Respiratory and COVID-19 Exacerbations. Cells 2020; 9:cells9091952. [PMID: 32847034 PMCID: PMC7565665 DOI: 10.3390/cells9091952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
The preservation of cellular homeostasis requires the synthesis of new proteins (proteostasis) and organelles, and the effective removal of misfolded or impaired proteins and cellular debris. This cellular homeostasis involves two key proteostasis mechanisms, the ubiquitin proteasome system and the autophagy–lysosome pathway. These catabolic pathways have been known to be involved in respiratory exacerbations and the pathogenesis of various lung diseases, such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and coronavirus disease-2019 (COVID-19). Briefly, proteostasis and autophagy processes are known to decline over time with age, cigarette or biomass smoke exposure, and/or influenced by underlying genetic factors, resulting in the accumulation of misfolded proteins and cellular debris, elevating apoptosis and cellular senescence, and initiating the pathogenesis of acute or chronic lung disease. Moreover, autophagic dysfunction results in an impaired microbial clearance, post-bacterial and/or viral infection(s) which contribute to the initiation of acute and recurrent respiratory exacerbations as well as the progression of chronic obstructive and restrictive lung diseases. In addition, the autophagic dysfunction-mediated cystic fibrosis transmembrane conductance regulator (CFTR) immune response impairment further exacerbates the lung disease. Recent studies demonstrate the therapeutic potential of novel autophagy augmentation strategies, in alleviating the pathogenesis of chronic obstructive or restrictive lung diseases and exacerbations such as those commonly seen in COPD, CF, ALI/ARDS and COVID-19.
Collapse
Affiliation(s)
- Garrett Pehote
- Michigan State University College of Osteopathic Medicine, East Lansing, MI 48823, USA;
| | - Neeraj Vij
- Department of Pediatrics and Pulmonary Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- PRECISION THERANOSTICS INC, Baltimore, MD 21202, USA
- VIJ BIOTECH, Baltimore, MD 21202, USA
- Correspondence: or ; Tel.: +1-240-623-0757
| |
Collapse
|
21
|
Ali A, Ovais M, Cui X, Rui Y, Chen C. Safety Assessment of Nanomaterials for Antimicrobial Applications. Chem Res Toxicol 2020; 33:1082-1109. [DOI: 10.1021/acs.chemrestox.9b00519] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Arbab Ali
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, P.R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Muhammad Ovais
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - YuKui Rui
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510700, China
| |
Collapse
|
22
|
Ghosh S, Jassar O, Kontsedalov S, Lebedev G, Wang C, Turner D, Levy A, Ghanim M. A Transcriptomics Approach Reveals Putative Interaction of Candidatus Liberibacter Solanacearum with the Endoplasmic Reticulum of Its Psyllid Vector. INSECTS 2019; 10:insects10090279. [PMID: 31480697 PMCID: PMC6780682 DOI: 10.3390/insects10090279] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/21/2019] [Accepted: 08/28/2019] [Indexed: 12/14/2022]
Abstract
Candidatus Liberibacter solanacerum (CLso), transmitted by Bactericera trigonica in a persistent and propagative mode causes carrot yellows disease, inflicting hefty economic losses. Understanding the process of transmission of CLso by psyllids is fundamental to devise sustainable management strategies. Persistent transmission involves critical steps of adhesion, cell invasion, and replication before passage through the midgut barrier. This study uses a transcriptomic approach for the identification of differentially expressed genes with CLso infection in the midguts, adults, and nymphs of B. trigonica and their putative involvement in CLso transmission. Several genes related to focal adhesion and cellular invasion were upregulated after CLso infection. Interestingly, genes involved with proper functionality of the endoplasmic reticulum (ER) were upregulated in CLso infected samples. Notably, genes from the endoplasmic reticulum associated degradation (ERAD) and the unfolded protein response (UPR) pathway were overexpressed after CLso infection. Marker genes of the ERAD and UPR pathways were also upregulated in Diaphorina citri when infected with Candidatus Liberibacter asiaticus (CLas). Upregulation of the ERAD and UPR pathways indicate induction of ER stress by CLso/CLas in their psyllid vector. The role of ER in bacteria–host interactions is well-documented; however, the ER role following pathogenesis of CLso/CLas is unknown and requires further functional validation.
Collapse
Affiliation(s)
- Saptarshi Ghosh
- Department of Entomology, the Volcani Center, Rishon LeZion 7505101, Israel
| | - Ola Jassar
- Department of Entomology, the Volcani Center, Rishon LeZion 7505101, Israel
| | | | - Galina Lebedev
- Department of Entomology, the Volcani Center, Rishon LeZion 7505101, Israel
| | - Chunxia Wang
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL 33850, USA
| | - Donielle Turner
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL 33850, USA
| | - Amit Levy
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL 33850, USA
- Department of Plant Pathology, University of Florida, Gainesville, FL 32601, USA
| | - Murad Ghanim
- Department of Entomology, the Volcani Center, Rishon LeZion 7505101, Israel.
| |
Collapse
|
23
|
Kubelkova K, Macela A. Innate Immune Recognition: An Issue More Complex Than Expected. Front Cell Infect Microbiol 2019; 9:241. [PMID: 31334134 PMCID: PMC6616152 DOI: 10.3389/fcimb.2019.00241] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022] Open
Abstract
Primary interaction of an intracellular bacterium with its host cell is initiated by activation of multiple signaling pathways in response to bacterium recognition itself or as cellular responses to stress induced by the bacterium. The leading molecules in these processes are cell surface membrane receptors as well as cytosolic pattern recognition receptors recognizing pathogen-associated molecular patterns or damage-associated molecular patterns induced by the invading bacterium. In this review, we demonstrate possible sequences of events leading to recognition of Francisella tularensis, present findings on known mechanisms for manipulating cell responses to protect Francisella from being killed, and discuss newly published data from the perspective of early stages of host-pathogen interaction.
Collapse
Affiliation(s)
- Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| | | |
Collapse
|
24
|
Neutrophils Dampen Adaptive Immunity in Brucellosis. Infect Immun 2019; 87:IAI.00118-19. [PMID: 30804100 PMCID: PMC6479033 DOI: 10.1128/iai.00118-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 01/03/2023] Open
Abstract
Brucella organisms are intracellular stealth pathogens of animals and humans. The bacteria overcome the assault of innate immunity at early stages of an infection. Brucella organisms are intracellular stealth pathogens of animals and humans. The bacteria overcome the assault of innate immunity at early stages of an infection. Removal of polymorphonuclear neutrophils (PMNs) at the onset of adaptive immunity against Brucella abortus favored bacterial elimination in mice. This was associated with higher levels of interferon gamma (IFN-γ) and a higher proportion of cells expressing interleukin 6 (IL-6) and inducible nitric oxide synthase (iNOS), compatible with M1 macrophages, in PMN-depleted B. abortus-infected (PMNd-Br) mice. At later times in the acute infection phase, the amounts of IFN-γ fell while IL-6, IL-10, and IL-12 became the predominant cytokines in PMNd-Br mice. IL-4, IL-1β, and tumor necrosis factor alpha (TNF-α) remained at background levels at all times of the infection. Depletion of PMNs at the acute stages of infection promoted the premature resolution of spleen inflammation. The efficient removal of bacteria in the PMNd-Br mice was not due to an increase of antibodies, since the immunoglobulin isotype responses to Brucella antigens were dampened. Anti-Brucella antibodies abrogated the production of IL-6, IL-10, and IL-12 but did not affect the levels of IFN-γ at later stages of infection in PMNd-Br mice. These results demonstrate that PMNs have an active role in modulating the course of B. abortus infection after the adaptive immune response has already developed.
Collapse
|
25
|
Wong MY, DiChiara AS, Suen PH, Chen K, Doan ND, Shoulders MD. Adapting Secretory Proteostasis and Function Through the Unfolded Protein Response. Curr Top Microbiol Immunol 2018; 414:1-25. [PMID: 28929194 DOI: 10.1007/82_2017_56] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cells address challenges to protein folding in the secretory pathway by engaging endoplasmic reticulum (ER)-localized protective mechanisms that are collectively termed the unfolded protein response (UPR). By the action of the transmembrane signal transducers IRE1, PERK, and ATF6, the UPR induces networks of genes whose products alleviate the burden of protein misfolding. The UPR also plays instructive roles in cell differentiation and development, aids in the response to pathogens, and coordinates the output of professional secretory cells. These functions add to and move beyond the UPR's classical role in addressing proteotoxic stress. Thus, the UPR is not just a reaction to protein misfolding, but also a fundamental driving force in physiology and pathology. Recent efforts have yielded a suite of chemical genetic methods and small molecule modulators that now provide researchers with both stress-dependent and -independent control of UPR activity. Such tools provide new opportunities to perturb the UPR and thereby study mechanisms for maintaining proteostasis in the secretory pathway. Numerous observations now hint at the therapeutic potential of UPR modulation for diseases related to the misfolding and aggregation of ER client proteins. Growing evidence also indicates the promise of targeting ER proteostasis nodes downstream of the UPR. Here, we review selected advances in these areas, providing a resource to inform ongoing studies of secretory proteostasis and function as they relate to the UPR.
Collapse
Affiliation(s)
- Madeline Y Wong
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139-4307, USA
| | - Andrew S DiChiara
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139-4307, USA
| | - Patreece H Suen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139-4307, USA
| | - Kenny Chen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139-4307, USA
| | - Ngoc-Duc Doan
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139-4307, USA
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139-4307, USA.
| |
Collapse
|
26
|
Ghanim M, Achor D, Ghosh S, Kontsedalov S, Lebedev G, Levy A. 'Candidatus Liberibacter asiaticus' Accumulates inside Endoplasmic Reticulum Associated Vacuoles in the Gut Cells of Diaphorina citri. Sci Rep 2017; 7:16945. [PMID: 29208900 PMCID: PMC5717136 DOI: 10.1038/s41598-017-16095-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/06/2017] [Indexed: 01/10/2023] Open
Abstract
Citrus greening disease known also as Huanglongbing (HLB) caused by the phloem-limited bacterium 'Candidatus Liberibacter asiaticus' (CLas) has resulted in tremendous losses and the death of millions of trees worldwide. CLas is transmitted by the Asian citrus psyllid Diaphorina citri. The closely-related bacteria 'Candidatus Liberibacter solanacearum' (CLso), associated with vegetative disorders in carrots, is transmitted by the carrot psyllid Bactericera trigonica. A promising approach to prevent the transmission of these pathogens is to interfere with the vector-pathogen interactions, but our understanding of these processes is limited. It was recently reported that CLas induced changes in the nuclear architecture, and activated programmed cell death, in D. citri midgut cells. Here, we used electron and fluorescent microscopy and show that CLas induces the formation of endoplasmic reticulum (ER)-associated bodies. The bacterium recruits those ER structures into Liberibacter containing vacuoles (LCVs), in which bacterial cells seem to propagate. ER- associated LCV formation was unique to CLas, as we could not detect these bodies in B. trigonica infected with CLso. ER recruitment is hypothesized to generate a safe replicative body to escape cellular immune responses in the insect gut. Understanding the molecular interactions that undelay these responses will open new opportunities for controlling CLas.
Collapse
Affiliation(s)
- Murad Ghanim
- Department of Entomology, Volcani Center, Rishon LeZion, Israel.
| | - Diann Achor
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Saptarshi Ghosh
- Department of Entomology, Volcani Center, Rishon LeZion, Israel
| | | | - Galina Lebedev
- Department of Entomology, Volcani Center, Rishon LeZion, Israel
| | - Amit Levy
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA.
- Department of Plant Pathology, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
27
|
Radomski N, Rebbig A, Leonhardt RM, Knittler MR. Xenophagic pathways and their bacterial subversion in cellular self-defense - παντα ρει - everything is in flux. Int J Med Microbiol 2017; 308:185-196. [PMID: 29126745 DOI: 10.1016/j.ijmm.2017.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 01/09/2023] Open
Abstract
Autophagy is an evolutionarily ancient and highly conserved eukaryotic mechanism that targets cytoplasmic material for degradation. Autophagic flux involves the formation of autophagosomes and their degradation by lysosomes. The process plays a crucial role in maintaining cellular homeostasis and responds to various environmental conditions. While autophagy had previously been thought to be a non-selective process, it is now clear that it can also selectively target cellular organelles, such as mitochondria (referred to as mitophagy) and/or invading pathogens (referred to as xenophagy). Selective autophagy is characterized by specific substrate recognition and requires distinct cellular adaptor proteins. Here we review xenophagic mechanisms involved in the recognition and autolysosomal or autophagolysosomal degradation of different intracellular bacteria. In this context, we also discuss a recently discovered cellular self-defense pathway, termed mito-xenophagy, which occurs during bacterial infection of dendritic cells and depends on a TNF-α-mediated metabolic switch from oxidative phosphorylation to glycolysis.
Collapse
Affiliation(s)
- Nadine Radomski
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany
| | - Annica Rebbig
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany
| | - Ralf M Leonhardt
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Michael R Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany.
| |
Collapse
|
28
|
Reliance of Wolbachia on High Rates of Host Proteolysis Revealed by a Genome-Wide RNAi Screen of Drosophila Cells. Genetics 2017; 205:1473-1488. [PMID: 28159754 DOI: 10.1534/genetics.116.198903] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/27/2017] [Indexed: 11/18/2022] Open
Abstract
Wolbachia are gram-negative, obligate, intracellular bacteria carried by a majority of insect species worldwide. Here we use a Wolbachia-infected Drosophila cell line and genome-wide RNA interference (RNAi) screening to identify host factors that influence Wolbachia titer. By screening an RNAi library targeting 15,699 transcribed host genes, we identified 36 candidate genes that dramatically reduced Wolbachia titer and 41 that increased Wolbachia titer. Host gene knockdowns that reduced Wolbachia titer spanned a broad array of biological pathways including genes that influenced mitochondrial function and lipid metabolism. In addition, knockdown of seven genes in the host ubiquitin and proteolysis pathways significantly reduced Wolbachia titer. To test the in vivo relevance of these results, we found that drug and mutant inhibition of proteolysis reduced levels of Wolbachia in the Drosophila oocyte. The presence of Wolbachia in either cell lines or oocytes dramatically alters the distribution and abundance of ubiquitinated proteins. Functional studies revealed that maintenance of Wolbachia titer relies on an intact host Endoplasmic Reticulum (ER)-associated protein degradation pathway (ERAD). Accordingly, electron microscopy studies demonstrated that Wolbachia is intimately associated with the host ER and dramatically alters the morphology of this organelle. Given Wolbachia lack essential amino acid biosynthetic pathways, the reliance of Wolbachia on high rates of host proteolysis via ubiquitination and the ERAD pathways may be a key mechanism for provisioning Wolbachia with amino acids. In addition, the reliance of Wolbachia on the ERAD pathway and disruption of ER morphology suggests a previously unsuspected mechanism for Wolbachia's potent ability to prevent RNA virus replication.
Collapse
|
29
|
Qi X, Man SM, Malireddi RKS, Karki R, Lupfer C, Gurung P, Neale G, Guy CS, Lamkanfi M, Kanneganti TD. Cathepsin B modulates lysosomal biogenesis and host defense against Francisella novicida infection. J Exp Med 2016; 213:2081-97. [PMID: 27551156 PMCID: PMC5030800 DOI: 10.1084/jem.20151938] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 07/22/2016] [Indexed: 12/11/2022] Open
Abstract
Lysosomal cathepsins regulate an exquisite range of biological functions, and their deregulation is associated with inflammatory, metabolic, and degenerative diseases in humans. In this study, we identified a key cell-intrinsic role for cathepsin B as a negative feedback regulator of lysosomal biogenesis and autophagy. Mice and macrophages lacking cathepsin B activity had increased resistance to the cytosolic bacterial pathogen Francisella novicida Genetic deletion or pharmacological inhibition of cathepsin B down-regulated mechanistic target of rapamycin activity and prevented cleavage of the lysosomal calcium channel TRPML1. These events drove transcription of lysosomal and autophagy genes via transcription factor EB, which increased lysosomal biogenesis and activation of autophagy initiation kinase ULK1 for clearance of the bacteria. Our results identified a fundamental biological function of cathepsin B in providing a checkpoint for homeostatic maintenance of lysosome populations and basic recycling functions in the cell.
Collapse
Affiliation(s)
- Xiaopeng Qi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Si Ming Man
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | - Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Christopher Lupfer
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Prajwal Gurung
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Clifford S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Mohamed Lamkanfi
- Inflammation Research Center, VIB, B-9052 Zwijnaarde-Ghent, Belgium Department of Internal Medicine, Ghent University, B-9000 Ghent, Belgium
| | | |
Collapse
|
30
|
Martins AS, Alves I, Helguero L, Domingues MR, Neves BM. The Unfolded Protein Response in Homeostasis and Modulation of Mammalian Immune Cells. Int Rev Immunol 2016; 35:457-476. [PMID: 27119724 DOI: 10.3109/08830185.2015.1110151] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The endoplasmic reticulum (ER) plays important roles in eukaryotic protein folding and lipid biosynthesis. Several exogenous and endogenous cellular sources of stress can perturb ER homeostasis leading to the accumulation of unfolded proteins in the lumen. Unfolded protein accumulation triggers a signal-transduction cascade known as the unfolded protein response (UPR), an adaptive mechanism which aims to protect cells from protein aggregates and to restore ER functions. Further to this protective mechanism, in immune cells, UPR molecular effectors have been shown to participate in a wide range of biological processes such as cell differentiation, survival and immunoglobulin and cytokine production. Recent findings also highlight the involvement of the UPR machinery in the maturational program and antigen presentation capacities of dendritic cells. UPR is therefore a key element in immune system homeostasis with direct implications on both adaptive and innate immune responses. The present review summarizes the knowledge on the emerging roles of UPR signaling cascades in mammalian immune cells as well as the consequences of their dysregulation in relation to the pathogenesis of several diseases.
Collapse
Affiliation(s)
- Ana Sofia Martins
- a Mass Spectrometry Centre, Department of Chemistry and QOPNA , University of Aveiro, Campus Universitário de Santiago , Aveiro , Portugal
| | - Inês Alves
- a Mass Spectrometry Centre, Department of Chemistry and QOPNA , University of Aveiro, Campus Universitário de Santiago , Aveiro , Portugal
| | - Luisa Helguero
- a Mass Spectrometry Centre, Department of Chemistry and QOPNA , University of Aveiro, Campus Universitário de Santiago , Aveiro , Portugal.,b Institute for Research in Biomedicine - iBiMED, Health Sciences Program, Universidade de Aveiro , Portugal
| | - Maria Rosário Domingues
- a Mass Spectrometry Centre, Department of Chemistry and QOPNA , University of Aveiro, Campus Universitário de Santiago , Aveiro , Portugal
| | - Bruno Miguel Neves
- a Mass Spectrometry Centre, Department of Chemistry and QOPNA , University of Aveiro, Campus Universitário de Santiago , Aveiro , Portugal.,c Faculty of Pharmacy and Centre for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal
| |
Collapse
|
31
|
Truchan HK, Cockburn CL, Hebert KS, Magunda F, Noh SM, Carlyon JA. The Pathogen-Occupied Vacuoles of Anaplasma phagocytophilum and Anaplasma marginale Interact with the Endoplasmic Reticulum. Front Cell Infect Microbiol 2016; 6:22. [PMID: 26973816 PMCID: PMC4771727 DOI: 10.3389/fcimb.2016.00022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/08/2016] [Indexed: 11/13/2022] Open
Abstract
The genus Anaplasma consists of tick-transmitted obligate intracellular bacteria that invade white or red blood cells to cause debilitating and potentially fatal infections. A. phagocytophilum, a human and veterinary pathogen, infects neutrophils to cause granulocytic anaplasmosis. A. marginale invades bovine erythrocytes. Evidence suggests that both species may also infect endothelial cells in vivo. In mammalian and arthropod host cells, A. phagocytophilum and A. marginale reside in host cell derived pathogen-occupied vacuoles (POVs). While it was recently demonstrated that the A. phagocytophilum-occupied vacuole (ApV) intercepts membrane traffic from the trans-Golgi network, it is unclear if it or the A. marginale-occupied vacuole (AmV) interacts with other secretory organelles. Here, we demonstrate that the ApV and AmV extensively interact with the host endoplasmic reticulum (ER) in endothelial, myeloid, and/or tick cells. ER lumen markers, calreticulin, and protein disulfide isomerase, and the ER membrane marker, derlin-1, were pronouncedly recruited to the peripheries of both POVs. ApV association with the ER initiated early and continued throughout the infection cycle. Both the ApV and AmV interacted with the rough ER and smooth ER. However, only derlin-1-positive rough ER derived vesicles were delivered into the ApV lumen where they localized with intravacuolar bacteria. Transmission electron microscopy identified multiple ER-POV membrane contact sites on the cytosolic faces of both species' vacuoles that corresponded to areas on the vacuoles' lumenal faces where intravacuolar Anaplasma organisms closely associated. A. phagocytophilum is known to hijack Rab10, a GTPase that regulates ER dynamics and morphology. Yet, ApV-ER interactions were unhindered in cells in which Rab10 had been knocked down, demonstrating that the GTPase is dispensable for the bacterium to parasitize the ER. These data establish the ApV and AmV as pathogen-host interfaces that directly engage the ER in vertebrate and invertebrate host cells and evidence the conservation of ER parasitism between two Anaplasma species.
Collapse
Affiliation(s)
- Hilary K Truchan
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| | - Chelsea L Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| | - Kathryn S Hebert
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| | - Forgivemore Magunda
- Program in Vector Borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State UniversityPullman, WA, USA; The Paul G. Allen School for Global Animal Health, Washington State UniversityPullman, WA, USA
| | - Susan M Noh
- Program in Vector Borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State UniversityPullman, WA, USA; Animal Disease Research Unit, Agricultural Research Service, U. S. Department of AgriculturePullman, WA, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| |
Collapse
|
32
|
Wallqvist A, Memišević V, Zavaljevski N, Pieper R, Rajagopala SV, Kwon K, Yu C, Hoover TA, Reifman J. Using host-pathogen protein interactions to identify and characterize Francisella tularensis virulence factors. BMC Genomics 2015; 16:1106. [PMID: 26714771 PMCID: PMC4696196 DOI: 10.1186/s12864-015-2351-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 12/21/2015] [Indexed: 11/10/2022] Open
Abstract
Background Francisella tularensis is a select bio-threat agent and one of the most virulent intracellular pathogens known, requiring just a few organisms to establish an infection. Although several virulence factors are known, we lack an understanding of virulence factors that act through host-pathogen protein interactions to promote infection. To address these issues in the highly infectious F. tularensis subsp. tularensis Schu S4 strain, we deployed a combined in silico, in vitro, and in vivo analysis to identify virulence factors and their interactions with host proteins to characterize bacterial infection mechanisms. Results We initially used comparative genomics and literature to identify and select a set of 49 putative and known virulence factors for analysis. Each protein was then subjected to proteome-scale yeast two-hybrid (Y2H) screens with human and murine cDNA libraries to identify potential host-pathogen protein-protein interactions. Based on the bacterial protein interaction profile with both hosts, we selected seven novel putative virulence factors for mutant construction and animal validation experiments. We were able to create five transposon insertion mutants and used them in an intranasal BALB/c mouse challenge model to establish 50 % lethal dose estimates. Three of these, ΔFTT0482c, ΔFTT1538c, and ΔFTT1597, showed attenuation in lethality and can thus be considered novel F. tularensis virulence factors. The analysis of the accompanying Y2H data identified intracellular protein trafficking between the early endosome to the late endosome as an important component in virulence attenuation for these virulence factors. Furthermore, we also used the Y2H data to investigate host protein binding of two known virulence factors, showing that direct protein binding was a component in the modulation of the inflammatory response via activation of mitogen-activated protein kinases and in the oxidative stress response. Conclusions Direct interactions with specific host proteins and the ability to influence interactions among host proteins are important components for F. tularensis to avoid host-cell defense mechanisms and successfully establish an infection. Although direct host-pathogen protein-protein binding is only one aspect of Francisella virulence, it is a critical component in directly manipulating and interfering with cellular processes in the host cell. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2351-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, 21702, USA.
| | - Vesna Memišević
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, 21702, USA.
| | - Nela Zavaljevski
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, 21702, USA.
| | | | | | - Keehwan Kwon
- J. Craig Venter Institute, Rockville, MD, 20850, USA.
| | - Chenggang Yu
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, 21702, USA.
| | - Timothy A Hoover
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, USA.
| | - Jaques Reifman
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, 21702, USA.
| |
Collapse
|
33
|
Zhou MT, Qin Y, Li M, Chen C, Chen X, Shu HB, Guo L. Quantitative Proteomics Reveals the Roles of Peroxisome-associated Proteins in Antiviral Innate Immune Responses. Mol Cell Proteomics 2015; 14:2535-49. [PMID: 26124285 DOI: 10.1074/mcp.m115.048413] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
Compared with whole-cell proteomic analysis, subcellular proteomic analysis is advantageous not only for the increased coverage of low abundance proteins but also for generating organelle-specific data containing information regarding dynamic protein movement. In the present study, peroxisome-enriched fractions from Sendai virus (SeV)-infected or uninfected HepG2 cells were obtained and subjected to quantitative proteomics analysis. We identified 311 proteins that were significantly changed by SeV infection. Among these altered proteins, 25 are immune response-related proteins. Further bioinformatic analysis indicated that SeV infection inhibits cell cycle-related proteins and membrane attack complex-related proteins, all of which are beneficial for the survival and replication of SeV within host cells. Using Luciferase reporter assays on several innate immune-related reporters, we performed functional analysis on 11 candidate proteins. We identified LGALS3BP and CALU as potential negative regulators of the virus-induced activation of the type I interferons.
Collapse
Affiliation(s)
- Mao-Tian Zhou
- From the ‡State Key Laboratory of Virology, College of Life Sciences
| | - Yue Qin
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University
| | - Mi Li
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University
| | - Chen Chen
- From the ‡State Key Laboratory of Virology, College of Life Sciences
| | - Xi Chen
- ¶Wuhan Institute of Biotechnology, Wuhan, China
| | - Hong-Bing Shu
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University;
| | - Lin Guo
- From the ‡State Key Laboratory of Virology, College of Life Sciences;
| |
Collapse
|
34
|
D’Osualdo A, Anania VG, Yu K, Lill JR, Kaufman RJ, Matsuzawa SI, Reed JC. Transcription Factor ATF4 Induces NLRP1 Inflammasome Expression during Endoplasmic Reticulum Stress. PLoS One 2015; 10:e0130635. [PMID: 26086088 PMCID: PMC4472728 DOI: 10.1371/journal.pone.0130635] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/21/2015] [Indexed: 12/30/2022] Open
Abstract
Perturbation of endoplasmic reticulum (ER) homeostasis triggers the ER stress response (also known as Unfolded Protein Response), a hallmark of many pathological disorders. However the connection between ER stress and inflammation remains largely unexplored. Recent data suggest that ER stress controls the activity of inflammasomes, key signaling platforms that mediate innate immune responses. Here we report that expression of NLRP1, a core inflammasome component, is specifically up-regulated during severe ER stress conditions in human cell lines. Both IRE1α and PERK, but not the ATF6 pathway, modulate NLRP1 gene expression. Furthermore, using mutagenesis, chromatin immunoprecipitation and CRISPR-Cas9-mediated genome editing technology, we demonstrate that ATF4 transcription factor directly binds to NLRP1 promoter during ER stress. Although involved in different types of inflammatory responses, XBP-1 splicing was not required for NLRP1 induction. This study provides further evidence that links ER stress with innate
Collapse
Affiliation(s)
- Andrea D’Osualdo
- Cell Death and Survival Networks Program, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Veronica G. Anania
- Department of Protein Chemistry, Genentech Inc, South San Francisco, California, United States of America
| | - Kebing Yu
- Department of Protein Chemistry, Genentech Inc, South San Francisco, California, United States of America
| | - Jennie R. Lill
- Department of Protein Chemistry, Genentech Inc, South San Francisco, California, United States of America
| | - Randal J. Kaufman
- Degenerative Diseases Research Program, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Shu-ichi Matsuzawa
- Cell Death and Survival Networks Program, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- * E-mail: (JCR); (SM)
| | - John C. Reed
- Cell Death and Survival Networks Program, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- Pharma Research and Early Development (pRED), Roche, Basel, Switzerland
- * E-mail: (JCR); (SM)
| |
Collapse
|
35
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with uncertain pathogenesis. Endoplasmic reticulum (ER) stress has close correlations with inflammation and/or immune diseases. However, it is unknown whether aberrant ER stress is involved in SLE pathogenesis. We aimed to characterize the ER stress-related genes in patients with SLE and analyzed their correlations with the disease. Peripheral blood leucocytes were isolated from 76 well-characterized patients with SLE and 69 healthy controls. ER stress-related genes were determined at transcription level by absolute quantitative real-time polymerase chain reaction. Stepwise regression and correlation analysis were used to analyze the relationships between SLE disease and ER stress. Abnormal unfolded protein responses were found in patients with SLE with the downregulation of inositol-requiring enzyme 1 (IRE1), pancreatic ER kinase (PERK) and CCAAT/enhancer-binding protein homologous protein (CHOP) and upregulation of XBP1, XBP1s and MANF. In the patients with SLE disease activity index (SLEDAI) <12, PERK and MANF expressions were significantly decreased, compared with the patients with severe SLE (SLEDAI ≥ 12). However, there was no significant change in ATF6 mRNA expression in the patients with SLE. Negative correlation between IRE1/XBP1 and SLEDAI was observed in lower SLEDAI score group. Negative correlations between CHOP and anti-dsDNA antibody, MANF and antinuclear antibody were observed in high-SLEDAI score group. We also found that antinuclear antibody and anti-dsDNA antibodies correlated with SLEDAI in a weak positive manner. SLEDAI was negatively related with C3 level. SLEDAI and anti-dsDNA antibody showed modestly positive correlation with urine protein. These findings suggest that the abnormal unfolded protein responses, especially IRE1/XBP1 and PERK/CHOP axes, may contribute to SLE pathogenesis, which may be potential diagnosis indicators or treatment targets.
Collapse
|
36
|
Emerging functions of the unfolded protein response in immunity. Nat Immunol 2014; 15:910-9. [PMID: 25232821 DOI: 10.1038/ni.2991] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/18/2014] [Indexed: 12/14/2022]
Abstract
The unfolded protein response (UPR) has traditionally been viewed as an adaptive response triggered by the accumulation of unfolded proteins in the endoplasmic reticulum (ER) and aimed at restoring ER function. The UPR can also be an anticipatory response that is activated well before the disruption of protein homeostasis. UPR signaling intersects at many levels with the innate and adaptive immune responses. In some types of cells of the immune system, such as dendritic cells (DCs) and B cells, particular sensors that detect the UPR seem to be constitutively active in the absence of induction of the traditional UPR gene program and are necessary for antigen presentation and immunoglobulin synthesis. The UPR also influences signaling via Toll-like receptors (TLRs) and activation of the transcription factor NF-κB, and some pathogens subvert the UPR. This Review summarizes these emerging noncanonical functions of the UPR in immunity.
Collapse
|
37
|
Vorwerk S, Krieger V, Deiwick J, Hensel M, Hansmeier N. Proteomes of host cell membranes modified by intracellular activities of Salmonella enterica. Mol Cell Proteomics 2014; 14:81-92. [PMID: 25348832 DOI: 10.1074/mcp.m114.041145] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Intracellular pathogens need to establish a growth-stimulating host niche for survival and replication. A unique feature of the gastrointestinal pathogen Salmonella enterica serovar Typhimurium is the creation of extensive membrane networks within its host. An understanding of the origin and function of these membranes is crucial for the development of new treatment strategies. However, the characterization of this compartment is very challenging, and only fragmentary knowledge of its composition and biogenesis exists. Here, we describe a new proteome-based approach to enrich and characterize Salmonella-modified membranes. Using a Salmonella mutant strain that does not form this unique membrane network as a reference, we identified a high-confidence set of host proteins associated with Salmonella-modified membranes. This comprehensive analysis allowed us to reconstruct the interactions of Salmonella with host membranes. For example, we noted that Salmonella redirects endoplasmic reticulum (ER) membrane trafficking to its intracellular niche, a finding that has not been described for Salmonella previously. Our system-wide approach therefore has the potential to rapidly close gaps in our knowledge of the infection process of intracellular pathogens and demonstrates a hitherto unrecognized complexity in the formation of Salmonella host niches.
Collapse
Affiliation(s)
- Stephanie Vorwerk
- From the ‡Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Viktoria Krieger
- From the ‡Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Jörg Deiwick
- From the ‡Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Michael Hensel
- From the ‡Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Nicole Hansmeier
- From the ‡Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| |
Collapse
|
38
|
Intracellular Vibrio parahaemolyticus escapes the vacuole and establishes a replicative niche in the cytosol of epithelial cells. mBio 2014; 5:e01506-14. [PMID: 25205094 PMCID: PMC4173779 DOI: 10.1128/mbio.01506-14] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED Vibrio parahaemolyticus is a globally disseminated Gram-negative marine bacterium and the leading cause of seafood-borne acute gastroenteritis. Pathogenic bacterial isolates encode two type III secretion systems (T3SS), with the second system (T3SS2) considered the main virulence factor in mammalian hosts. For many decades, V. parahaemolyticus has been studied as an exclusively extracellular bacterium. However, the recent characterization of the T3SS2 effector protein VopC has suggested that this pathogen has the ability to invade, survive, and replicate within epithelial cells. Herein, we characterize this intracellular lifestyle in detail. We show that following internalization, V. parahaemolyticus is contained in vacuoles that develop into early endosomes, which subsequently mature into late endosomes. V. parahaemolyticus then escapes into the cytoplasm prior to vacuolar fusion with lysosomes. Vacuolar acidification is an important trigger for this escape. The cytoplasm serves as the pathogen's primary intracellular replicative niche; cytosolic replication is rapid and robust, with cells often containing over 150 bacteria by the time of cell lysis. These results show how V. parahaemolyticus successfully establishes an intracellular lifestyle that could contribute to its survival and dissemination during infection. IMPORTANCE The marine bacterium V. parahaemolyticus is the leading cause worldwide of seafood-borne acute gastroenteritis. For decades, the pathogen has been studied exclusively as an extracellular bacterium. However, recent results have revealed the pathogen's ability to invade and replicate within host cells. The present study is the first characterization of the V. parahaemolyticus' intracellular lifestyle. Upon internalization, V. parahaemolyticus is contained in a vacuole that would in the normal course of events ultimately fuse with a lysosome, degrading the vacuole's contents. The bacterium subverts this pathway, escaping into the cytoplasm prior to lysosomal fusion. Once in the cytoplasm, it replicates prolifically. Our study provides new insights into the strategies used by this globally disseminated pathogen to survive and proliferate within its host.
Collapse
|
39
|
Baruch M, Hertzog BB, Ravins M, Anand A, Cheng CY, Biswas D, Tirosh B, Hanski E. Induction of endoplasmic reticulum stress and unfolded protein response constitutes a pathogenic strategy of group A streptococcus. Front Cell Infect Microbiol 2014; 4:105. [PMID: 25136516 PMCID: PMC4120759 DOI: 10.3389/fcimb.2014.00105] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/14/2014] [Indexed: 11/30/2022] Open
Abstract
The connection between bacterial pathogens and unfolded protein response (UPR) is poorly explored. In this review we highlight the evidence showing that group A streptococcus (GAS) induces endoplasmic reticulum (ER) stress and UPR through which it captures the amino acid asparagine (ASN) from the host. GAS acts extracellularly and during adherence to host cells it delivers the hemolysin toxins; streptolysin O (SLO) and streptolysin S (SLS). By poorly understood pathways, these toxins trigger UPR leading to the induction of the transcriptional regulator ATF4 and consequently to the upregulation of asparagine synthetase (ASNS) transcription leading to production and release of ASN. GAS senses ASN and alters gene expression profile accordingly, and increases the rate of multiplication. We suggest that induction of UPR by GAS and by other bacterial pathogens represent means through which bacterial pathogens gain nutrients from the host, obviating the need to become internalized or inflict irreversible cell damage.
Collapse
Affiliation(s)
- Moshe Baruch
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University of Jerusalem (HUJI) Jerusalem, Israel
| | - Baruch B Hertzog
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University of Jerusalem (HUJI) Jerusalem, Israel
| | - Miriam Ravins
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University of Jerusalem (HUJI) Jerusalem, Israel
| | - Aparna Anand
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University of Jerusalem (HUJI) Jerusalem, Israel
| | - Catherine Youting Cheng
- Department of Microbiology, Center for Research Excellence and Technological Enterprise (CREATE), National University of Singapore (NUS) and NUS-HUJI Singapore
| | - Debabrata Biswas
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University of Jerusalem (HUJI) Jerusalem, Israel ; Department of Microbiology, Center for Research Excellence and Technological Enterprise (CREATE), National University of Singapore (NUS) and NUS-HUJI Singapore
| | - Boaz Tirosh
- The School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem Jerusalem, Israel
| | - Emanuel Hanski
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Hebrew University of Jerusalem (HUJI) Jerusalem, Israel ; Department of Microbiology, Center for Research Excellence and Technological Enterprise (CREATE), National University of Singapore (NUS) and NUS-HUJI Singapore
| |
Collapse
|
40
|
Mehlitz A, Karunakaran K, Herweg JA, Krohne G, van de Linde S, Rieck E, Sauer M, Rudel T. The chlamydial organism Simkania negevensis forms ER vacuole contact sites and inhibits ER-stress. Cell Microbiol 2014; 16:1224-43. [PMID: 24528559 DOI: 10.1111/cmi.12278] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/28/2014] [Accepted: 02/05/2014] [Indexed: 01/01/2023]
Abstract
Most intracellular bacterial pathogens reside within membrane-surrounded host-derived vacuoles. Few of these bacteria exploit membranes from the host's endoplasmic reticulum (ER) to form a replicative vacuole. Here, we describe the formation of ER-vacuole contact sites as part of the replicative niche of the chlamydial organism Simkania negevensis. Formation of ER-vacuole contact sites is evolutionary conserved in the distantly related protozoan host Acanthamoeba castellanii. Simkania growth is accompanied by mitochondria associating with the Simkania-containing vacuole (SCV). Super-resolution microscopy as well as 3D reconstruction from electron micrographs of serial ultra-thin sections revealed a single vacuolar system forming extensive ER-SCV contact sites on the Simkania vacuolar surface. Simkania infection induced an ER-stress response, which was later downregulated. Induction of ER-stress with Thapsigargin or Tunicamycin was strongly inhibited in cells infected with Simkania. Inhibition of ER-stress was required for inclusion formation and efficient growth, demonstrating a role of ER-stress in the control of Simkania infection. Thus, Simkania forms extensive ER-SCV contact sites in host species evolutionary as diverse as human and amoeba. Moreover, Simkania is the first bacterial pathogen described to interfere with ER-stress induced signalling to promote infection.
Collapse
Affiliation(s)
- Adrian Mehlitz
- University of Wuerzburg, Biocenter, Department of Microbiology, Am Hubland, D-97074, Wuerzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Marianecci C, Rinaldi F, Di Marzio L, Mastriota M, Pieretti S, Celia C, Paolino D, Iannone M, Fresta M, Carafa M. Ammonium glycyrrhizinate-loaded niosomes as a potential nanotherapeutic system for anti-inflammatory activity in murine models. Int J Nanomedicine 2014; 9:635-51. [PMID: 24493924 PMCID: PMC3908944 DOI: 10.2147/ijn.s55066] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Liquorice extracts demonstrate therapeutic efficacy in treating dermatitis, eczema, and psoriasis when compared with corticosteroids. In this work, nonionic surfactant vesicles (niosomes, NSVs) containing polysorbate 20 (Tween 20), cholesterol, and cholesteryl hemisuccinate at different molar concentrations were used to prepare monoammonium glycyrrhizinate (AG)-loaded NSVs. The anti-inflammatory properties of AG-loaded NSVs were investigated in murine models. METHODS The physicochemical properties of the NSVs were characterized using dynamic light scattering. The fluidity of the lipid bilayer was evaluated by measuring the fluorescence intensity of diphenylhexatriene. The drug entrapment efficiency of AG was assessed using high-performance liquid chromatography. The physicochemical stability of the NSVs was evaluated as a function of time using dynamic light scattering combined with Turbiscan Lab Expert analysis. Serum stability was determined by incubating the NSVs with 10% v/v fetal bovine serum. The cytotoxic effects of the NSVs were investigated in human dermal fibroblasts using the Trypan blue dye exclusion assay (for cell mortality) and an MTT assay (for cell viability). Release profiles for the AG-loaded NSVs were studied in vitro using cellulose membranes. NSVs showing the most desirable physicochemical properties were selected to test for in vivo anti-inflammatory activity in murine models. The anti-inflammatory activity of the NSVs was investigated by measuring edema and nociception in mice stimulated with chemical agents. RESULTS NSVs showed favorable physicochemical properties for in vitro and in vivo administration. In addition, they demonstrated long-term stability based on Turbiscan Lab Expert analysis. The membrane fluidity of the NSVs was not affected by self-assembling of the surfactants into colloidal structures. Fluorescence anisotropy was found to be independent of the molar ratios of cholesteryl hemisuccinate and/or cholesterol during preparation of the NSVs. The anti-inflammatory AG drug showed no effect on the stability of the NSVs. In vivo experiments demonstrated that AG-loaded NSVs decreased edema and nociceptive responses when compared with AG alone and empty NSVs. In vitro and in vivo results demonstrated that pH sensitive and neutral NSVs show no statistical significant difference. CONCLUSION NSVs were nontoxic and showed features favorable for potential administration in vivo. In addition, neutral NSVs showed signs of increased anti-inflammatory and antinociceptive responses when compared with AG.
Collapse
Affiliation(s)
- Carlotta Marianecci
- Department of Drug Chemistry and Technologies, University Sapienza of Rome, Rome, Italy
| | - Federica Rinaldi
- Department of Drug Chemistry and Technologies, University Sapienza of Rome, Rome, Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University G d’Annunzio of Chieti of Pescara, Chieti, Italy
| | - Marica Mastriota
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Pieretti
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Christian Celia
- Department of Pharmacy, University G d’Annunzio of Chieti of Pescara, Chieti, Italy
- Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, TX, USA
| | - Donatella Paolino
- Department of Health Sciences, University Magna Graecia of Catanzaro, University Campus S Venuta, Building of BioSciences, Germaneto, Italy
| | - Michelangelo Iannone
- ARPA Calabria, Environmental Epidemiology Center, Italy
- CNR, Neuroscience Institute, Pharmacology Section, Complesso “Nini Barbieri”, Roccelletta di Borgia, Italy
| | - Massimo Fresta
- Department of Health Sciences, University Magna Graecia of Catanzaro, University Campus S Venuta, Building of BioSciences, Germaneto, Italy
| | - Maria Carafa
- Department of Drug Chemistry and Technologies, University Sapienza of Rome, Rome, Italy
| |
Collapse
|
42
|
Smith JA, Khan M, Magnani DD, Harms JS, Durward M, Radhakrishnan GK, Liu YP, Splitter GA. Brucella induces an unfolded protein response via TcpB that supports intracellular replication in macrophages. PLoS Pathog 2013; 9:e1003785. [PMID: 24339776 PMCID: PMC3855547 DOI: 10.1371/journal.ppat.1003785] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 10/08/2013] [Indexed: 01/18/2023] Open
Abstract
Brucella melitensis is a facultative intracellular bacterium that causes brucellosis, the most prevalent zoonosis worldwide. The Brucella intracellular replicative niche in macrophages and dendritic cells thwarts immune surveillance and complicates both therapy and vaccine development. Currently, host-pathogen interactions supporting Brucella replication are poorly understood. Brucella fuses with the endoplasmic reticulum (ER) to replicate, resulting in dramatic restructuring of the ER. This ER disruption raises the possibility that Brucella provokes an ER stress response called the Unfolded Protein Response (UPR). In this study, B. melitensis infection up regulated expression of the UPR target genes BiP, CHOP, and ERdj4, and induced XBP1 mRNA splicing in murine macrophages. These data implicate activation of all 3 major signaling pathways of the UPR. Consistent with previous reports, XBP1 mRNA splicing was largely MyD88-dependent. However, up regulation of CHOP, and ERdj4 was completely MyD88 independent. Heat killed Brucella stimulated significantly less BiP, CHOP, and ERdj4 expression, but induced XBP1 splicing. Although a Brucella VirB mutant showed relatively intact UPR induction, a TcpB mutant had significantly compromised BiP, CHOP and ERdj4 expression. Purified TcpB, a protein recently identified to modulate microtubules in a manner similar to paclitaxel, also induced UPR target gene expression and resulted in dramatic restructuring of the ER. In contrast, infection with the TcpB mutant resulted in much less ER structural disruption. Finally, tauroursodeoxycholic acid, a pharmacologic chaperone that ameliorates the UPR, significantly impaired Brucella replication in macrophages. Together, these results suggest Brucella induces a UPR, via TcpB and potentially other factors, that enables its intracellular replication. Thus, the UPR may provide a novel therapeutic target for the treatment of brucellosis. These results also have implications for other intracellular bacteria that rely on host physiologic stress responses for replication.
Collapse
Affiliation(s)
- Judith A. Smith
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Mike Khan
- Cellular and Molecular Pathology Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Diogo D. Magnani
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| | - Jerome S. Harms
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| | - Marina Durward
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| | | | - Yi-Ping Liu
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Gary A. Splitter
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| |
Collapse
|
43
|
Gorbatyuk M, Gorbatyuk O. Review: retinal degeneration: focus on the unfolded protein response. Mol Vis 2013; 19:1985-98. [PMID: 24068865 PMCID: PMC3782367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 09/18/2013] [Indexed: 11/08/2022] Open
Abstract
Recently published literature has provided evidence that the unfolded protein response (UPR) is involved in the development of retinal degeneration. The scope of these studies encompassed diabetic retinopathy, retinopathy of prematurity, glaucoma, retinal detachment, light-induced retinal degeneration, age-related macular degeneration, and inherited retinal degeneration. Subsequent studies investigating the role of individual UPR markers in retinal pathogenesis and examining the therapeutic potential of reprogramming the UPR as a method for modulating the rate of retinal degeneration have been initiated. Manipulation of UPR markers has been made possible by the use of knockout mice, pharmacological agents, and viral vector-mediated augmentation of gene expression. Future research will aim at identifying specific inhibitors and/or inducers of UPR regulatory markers as well as expand the list of UPR-related animal models. Additionally, adeno-associated virus-mediated gene delivery is a safe and effective method for modulating gene expression, and thus is a useful research tool for manipulating individual UPR markers in affected retinas and a promising delivery vector for gene therapy in retinal degenerative disorders.
Collapse
Affiliation(s)
- Marina Gorbatyuk
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Oleg Gorbatyuk
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610
| |
Collapse
|
44
|
Escoll P, Rolando M, Gomez-Valero L, Buchrieser C. From amoeba to macrophages: exploring the molecular mechanisms of Legionella pneumophila infection in both hosts. Curr Top Microbiol Immunol 2013; 376:1-34. [PMID: 23949285 DOI: 10.1007/82_2013_351] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Legionella pneumophila is a Gram-negative bacterium and the causative agent of Legionnaires' disease. It replicates within amoeba and infects accidentally human macrophages. Several similarities are seen in the L. pneumophila-infection cycle in both hosts, suggesting that the tools necessary for macrophage infection may have evolved during co-evolution of L. pneumophila and amoeba. The establishment of the Legionella-containing vacuole (LCV) within the host cytoplasm requires the remodeling of the LCV surface and the hijacking of vesicles and organelles. Then L. pneumophila replicates in a safe intracellular niche in amoeba and macrophages. In this review we will summarize the existing knowledge of the L. pneumophila infection cycle in both hosts at the molecular level and compare the factors involved within amoeba and macrophages. This knowledge will be discussed in the light of recent findings from the Acanthamoeba castellanii genome analyses suggesting the existence of a primitive immune-like system in amoeba.
Collapse
Affiliation(s)
- Pedro Escoll
- Institut Pasteur, Biologie des Bactéries Intracellulaires and CNRS UMR, 3525, Paris, France
| | | | | | | |
Collapse
|
45
|
Schönthal AH. Endoplasmic reticulum stress: its role in disease and novel prospects for therapy. SCIENTIFICA 2012; 2012:857516. [PMID: 24278747 PMCID: PMC3820435 DOI: 10.6064/2012/857516] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/12/2012] [Indexed: 05/19/2023]
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle required for lipid biosynthesis, calcium storage, and protein folding and processing. A number of physiological and pathological conditions, as well as a variety of pharmacological agents, are able to disturb proper ER function and thereby cause ER stress, which severely impairs protein folding and therefore poses the risk of proteotoxicity. Specific triggers for ER stress include, for example, particular intracellular alterations (e.g., calcium or redox imbalances), certain microenvironmental conditions (e.g., hypoglycemia, hypoxia, and acidosis), high-fat and high-sugar diet, a variety of natural compounds (e.g., thapsigargin, tunicamycin, and geldanamycin), and several prescription drugs (e.g., bortezomib/Velcade, celecoxib/Celebrex, and nelfinavir/Viracept). The cell reacts to ER stress by initiating a defensive process, called the unfolded protein response (UPR), which is comprised of cellular mechanisms aimed at adaptation and safeguarding cellular survival or, in cases of excessively severe stress, at initiation of apoptosis and elimination of the faulty cell. In recent years, this dichotomic stress response system has been linked to several human diseases, and efforts are underway to develop approaches to exploit ER stress mechanisms for therapy. For example, obesity and type 2 diabetes have been linked to ER stress-induced failure of insulin-producing pancreatic beta cells, and current research efforts are aimed at developing drugs that ameliorate cellular stress and thereby protect beta cell function. Other studies seek to pharmacologically aggravate chronic ER stress in cancer cells in order to enhance apoptosis and achieve tumor cell death. In the following, these principles will be presented and discussed.
Collapse
Affiliation(s)
- Axel H. Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR-405, Los Angeles, CA 90033, USA
| |
Collapse
|
46
|
Dumoux M, Clare DK, Saibil HR, Hayward RD. Chlamydiae assemble a pathogen synapse to hijack the host endoplasmic reticulum. Traffic 2012; 13:1612-27. [PMID: 22901061 PMCID: PMC3533787 DOI: 10.1111/tra.12002] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 08/15/2012] [Accepted: 08/17/2012] [Indexed: 12/22/2022]
Abstract
Chlamydiae are obligate intracellular bacterial pathogens that replicate within a specialized membrane-bound compartment, termed an 'inclusion'. The inclusion membrane is a critical host-pathogen interface, yet the extent of its interaction with cellular organelles and the origin of this membrane remain poorly defined. Here we show that the host endoplasmic reticulum (ER) is specifically recruited to the inclusion, and that key rough ER (rER) proteins are enriched on and translocated into the inclusion. rER recruitment is a Chlamydia-orchestrated process that occurs independently of host trafficking. Generation of infectious progeny requires an intact ER, since ER vacuolation early during infection stalls inclusion development, whereas disruption post ER recruitment bursts the inclusion. Electron tomography and immunolabelling of Chlamydia-infected cells reveal 'pathogen synapses' at which ordered arrays of chlamydial type III secretion complexes connect to the inclusion membrane only at rER contact sites. Our data show a supramolecular assembly involved in pathogen hijack of a key host organelle.
Collapse
Affiliation(s)
- Maud Dumoux
- Institute of Structural and Molecular Biology, Birkbeck & University College LondonMalet Street, London, WC1E 7HX, UK
| | - Daniel K Clare
- Department of Crystallography, Institute of Structural and Molecular BiologyBirkbeck, Malet Street, London, WC1E 7HX, UK
| | - Helen R Saibil
- Department of Crystallography, Institute of Structural and Molecular BiologyBirkbeck, Malet Street, London, WC1E 7HX, UK
| | - Richard D Hayward
- Institute of Structural and Molecular Biology, Birkbeck & University College LondonMalet Street, London, WC1E 7HX, UK
| |
Collapse
|
47
|
Teng TS, Foo SS, Simamarta D, Lum FM, Teo TH, Lulla A, Yeo NKW, Koh EGL, Chow A, Leo YS, Merits A, Chin KC, Ng LFP. Viperin restricts chikungunya virus replication and pathology. J Clin Invest 2012; 122:4447-60. [PMID: 23160199 DOI: 10.1172/jci63120] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 09/20/2012] [Indexed: 12/15/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne arthralgia arbovirus that is reemergent in sub-Saharan Africa and Southeast Asia. CHIKV infection has been shown to be self-limiting, but the molecular mechanisms of the innate immune response that control CHIKV replication remain undefined. Here, longitudinal transcriptional analyses of PBMCs from a cohort of CHIKV-infected patients revealed that type I IFNs controlled CHIKV infection via RSAD2 (which encodes viperin), an enigmatic multifunctional IFN-stimulated gene (ISG). Viperin was highly induced in monocytes, the major target cell of CHIKV in blood. Anti-CHIKV functions of viperin were dependent on its localization in the ER, and the N-terminal amphipathic α-helical domain was crucial for its antiviral activity in controlling CHIKV replication. Furthermore, mice lacking Rsad2 had higher viremia and severe joint inflammation compared with wild-type mice. Our data demonstrate that viperin is a critical antiviral host protein that controls CHIKV infection and provide a preclinical basis for the design of effective control strategies against CHIKV and other reemerging arthrogenic alphaviruses.
Collapse
Affiliation(s)
- Terk-Shin Teng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Canton J, Kima PE. Interactions of pathogen-containing compartments with the secretory pathway. Cell Microbiol 2012; 14:1676-86. [PMID: 22862745 DOI: 10.1111/cmi.12000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 07/20/2012] [Accepted: 07/23/2012] [Indexed: 02/03/2023]
Abstract
A subgroup of intracellular pathogens reside and replicate within membrane-bound compartments often termed pathogen-containing compartments (PCC). PCCs navigate around a wide range of host cell vesicles and organelles. In light of the perils of engaging with vesicles of the endocytic pathway, most PCCs modulate their interactions with endocytic vesicles while a few avoid those interactions. The secretory pathway constitutes another important grouping of vesicles and organelles in host cells. Although the negative consequences of engaging with the secretory pathway are not known, there is evidence that PCCs interact differentially with vesicles and organelles in this pathway as well. In this review, we consider three prokaryote pathogens and two protozoan parasites for which there is information on the interactions of their PCCs with the secretory pathway. Current understandings of the molecular interactions as well as the metabolic benefits that accompany those interactions are discussed. Not unexpectedly, our understanding of the extent of these interactions is variable. An underlying theme that is brought to the fore is that PCCs establish preferential interactions with distinct compartments of the secretory pathway.
Collapse
Affiliation(s)
- Johnathan Canton
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | | |
Collapse
|
49
|
Spielmann T, Montagna GN, Hecht L, Matuschewski K. Molecular make-up of the Plasmodium parasitophorous vacuolar membrane. Int J Med Microbiol 2012; 302:179-86. [PMID: 22898489 DOI: 10.1016/j.ijmm.2012.07.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Plasmodium, the causative agent of malaria, is an obligate, intracellular, eukaryotic cell that invades, replicates, and differentiates within hepatocytes and erythrocytes. Inside a host cell, a second membrane delineates the developing pathogen in addition to the parasite plasma membrane, resulting in a distinct cellular compartment, termed parasitophorous vacuole (PV). The PV membrane (PVM) constitutes the parasite-host cell interface and is likely central to nutrient acquisition, host cell remodeling, waste disposal, environmental sensing, and protection from innate defense. Over the past two decades, a number of parasite-encoded PVM proteins have been identified. They include multigene families and protein complexes, such as early-transcribed membrane proteins (ETRAMPs) and the Plasmodium translocon for exported proteins (PTEX). Nearly all Plasmodium PVM proteins are restricted to this genus and display transient and stage-specific expression. Here, we provide an overview of the PVM proteins of Plasmodium blood and liver stages. Biochemical and experimental genetics data suggest that some PVM proteins are ideal targets for novel anti-malarial intervention strategies.
Collapse
Affiliation(s)
- Tobias Spielmann
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| | | | | | | |
Collapse
|
50
|
Martinon F. The endoplasmic reticulum: a sensor of cellular stress that modulates immune responses. Microbes Infect 2012; 14:1293-300. [PMID: 22800981 PMCID: PMC7110899 DOI: 10.1016/j.micinf.2012.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 07/03/2012] [Accepted: 07/04/2012] [Indexed: 12/25/2022]
Abstract
Many inflammatory and infectious diseases are characterized by the activation of signaling pathways steaming from the endoplasmic reticulum (ER). These pathways, primarily associated with loss of ER homeostasis, are emerging as key regulators of inflammation and infection. Recent advances shed light on the mechanisms linking ER-stress and immune responses.
Collapse
Affiliation(s)
- Fabio Martinon
- Dept. of Biochemistry, University of Lausanne, 155 Ch. Des Boveresses, Epalinges 1066, Switzerland.
| |
Collapse
|