1
|
Li F, Zhang X, Xu J, Zhang Y, Li G, Yang X, Deng G, Dai Y, Liu B, Kosan C, Chen X, Cai Y. SIRT7 remodels the cytoskeleton via RAC1 to enhance host resistance to Mycobacterium tuberculosis. mBio 2024; 15:e0075624. [PMID: 39287444 PMCID: PMC11481912 DOI: 10.1128/mbio.00756-24] [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: 03/14/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Phagocytosis of Mycobacterium tuberculosis (Mtb) followed by its integration into the matured lysosome is critical in the host defense against tuberculosis. How Mtb escapes this immune attack remains elusive. In this study, we unveiled a novel regulatory mechanism by which SIRT7 regulates cytoskeletal remodeling by modulating RAC1 activation. We discovered that SIRT7 expression was significantly reduced in CD14+ monocytes of TB patients. Mtb infection diminished SIRT7 expression by macrophages at both the mRNA and protein levels. SIRT7 deficiency impaired actin cytoskeleton-dependent macrophage phagocytosis, LC3II expression, and bactericidal activity. In a murine tuberculosis model, SIRT7 deficiency detrimentally impacted host resistance to Mtb, while Sirt7 overexpression significantly increased the host defense against Mtb, as determined by bacterial burden and inflammatory-histopathological damage in the lung. Mechanistically, we demonstrated that SIRT7 limits Mtb infection by directly interacting with and activating RAC1, through which cytoskeletal remodeling is modulated. Therefore, we concluded that SIRT7, in its role regulating cytoskeletal remodeling through RAC1, is critical for host responses during Mtb infection and proposes a potential target for tuberculosis treatment.IMPORTANCETuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant global health issue. Critical to macrophages' defense against Mtb is phagocytosis, governed by the actin cytoskeleton. Previous research has revealed that Mtb manipulates and disrupts the host's actin network, though the specific mechanisms have been elusive. Our study identifies a pivotal role for SIRT7 in this context: Mtb infection leads to reduced SIRT7 expression, which, in turn, diminishes RAC1 activation and consequently impairs actin-dependent phagocytosis. The significance of our research is that SIRT7 directly engages with and activates Rac Family Small GTPase 1 (RAC1), thus promoting effective phagocytosis and the elimination of Mtb. This insight into the dynamic between host and pathogen in TB not only broadens our understanding but also opens new avenues for therapeutic development.
Collapse
Affiliation(s)
- Fuxiang Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Ximeng Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Jinjin Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yue Zhang
- School of Pharmaceutical Sciences, Shenzhen University Medical School, Shenzhen, China
| | - Guo Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Xirui Yang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Guofang Deng
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Youchao Dai
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, China
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
2
|
Guallar-Garrido S, Soldati T. Exploring host-pathogen interactions in the Dictyostelium discoideum-Mycobacterium marinum infection model of tuberculosis. Dis Model Mech 2024; 17:dmm050698. [PMID: 39037280 PMCID: PMC11552500 DOI: 10.1242/dmm.050698] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Mycobacterium tuberculosis is a pathogenic mycobacterium that causes tuberculosis. Tuberculosis is a significant global health concern that poses numerous clinical challenges, particularly in terms of finding effective treatments for patients. Throughout evolution, host immune cells have developed cell-autonomous defence strategies to restrain and eliminate mycobacteria. Concurrently, mycobacteria have evolved an array of virulence factors to counteract these host defences, resulting in a dynamic interaction between host and pathogen. Here, we review recent findings, including those arising from the use of the amoeba Dictyostelium discoideum as a model to investigate key mycobacterial infection pathways. D. discoideum serves as a scalable and genetically tractable model for human phagocytes, providing valuable insights into the intricate mechanisms of host-pathogen interactions. We also highlight certain similarities between M. tuberculosis and Mycobacterium marinum, and the use of M. marinum to more safely investigate mycobacteria in D. discoideum.
Collapse
Affiliation(s)
- Sandra Guallar-Garrido
- Department of Biochemistry, Faculty of Science, University of Geneva, 30 quai Ernest-Ansermet, Science II, 1211 Geneva-4, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, 30 quai Ernest-Ansermet, Science II, 1211 Geneva-4, Switzerland
| |
Collapse
|
3
|
MIDESP: Mutual Information-Based Detection of Epistatic SNP Pairs for Qualitative and Quantitative Phenotypes. BIOLOGY 2021; 10:biology10090921. [PMID: 34571798 PMCID: PMC8469369 DOI: 10.3390/biology10090921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary The interactions between SNPs, which are known as epistasis, can strongly influence the phenotype. Their detection is still a challenge, which is made even more difficult through the existence of background associations that can hide correct epistatic interactions. To address the limitations of existing methods, we present in this study our novel method MIDESP for the detection of epistatic SNP pairs. It is the first mutual information-based method that can be applied to both qualitative and quantitative phenotypes and which explicitly accounts for background associations in the dataset. Abstract The interactions between SNPs result in a complex interplay with the phenotype, known as epistasis. The knowledge of epistasis is a crucial part of understanding genetic causes of complex traits. However, due to the enormous number of SNP pairs and their complex relationship to the phenotype, identification still remains a challenging problem. Many approaches for the detection of epistasis have been developed using mutual information (MI) as an association measure. However, these methods have mainly been restricted to case–control phenotypes and are therefore of limited applicability for quantitative traits. To overcome this limitation of MI-based methods, here, we present an MI-based novel algorithm, MIDESP, to detect epistasis between SNPs for qualitative as well as quantitative phenotypes. Moreover, by incorporating a dataset-dependent correction technique, we deal with the effect of background associations in a genotypic dataset to separate correct epistatic interaction signals from those of false positive interactions resulting from the effect of single SNP×phenotype associations. To demonstrate the effectiveness of MIDESP, we apply it on two real datasets with qualitative and quantitative phenotypes, respectively. Our results suggest that by eliminating the background associations, MIDESP can identify important genes, which play essential roles for bovine tuberculosis or the egg weight of chickens.
Collapse
|
4
|
Gilpin TE, Walter FR, Herbath M, Sandor M, Fabry Z. Mycobacterium bovis Bacillus Calmette-Guérin-Infected Dendritic Cells Induce TNF-α-Dependent Cell Cluster Formation That Promotes Bacterial Dissemination through an In Vitro Model of the Blood-Brain Barrier. THE JOURNAL OF IMMUNOLOGY 2021; 207:1065-1077. [PMID: 34321229 DOI: 10.4049/jimmunol.2001094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 06/11/2021] [Indexed: 11/19/2022]
Abstract
CNS tuberculosis (CNSTB) is the most severe manifestation of extrapulmonary tuberculosis infection, but the mechanism of how mycobacteria cross the blood-brain barrier (BBB) is not well understood. In this study, we report a novel murine in vitro BBB model combining primary brain endothelial cells, Mycobacterium bovis bacillus Calmette-Guérin-infected dendritic cells (DCs), PBMCs, and bacterial Ag-specific CD4+ T cells. We show that mycobacterial infection limits DC mobility and also induces cellular cluster formation that has a similar composition to pulmonary mycobacterial granulomas. Within the clusters, infection from DCs disseminates to the recruited monocytes, promoting bacterial expansion. Mycobacterium-induced in vitro granulomas have been described previously, but this report shows that they can form on brain endothelial cell monolayers. Cellular cluster formation leads to cluster-associated damage of the endothelial cell monolayer defined by mitochondrial stress, disorganization of the tight junction proteins ZO-1 and claudin-5, upregulation of the adhesion molecules VCAM-1 and ICAM-1, and increased transmigration of bacteria-infected cells across the BBB. TNF-α inhibition reduces cluster formation on brain endothelial cells and mitigates cluster-associated damage. These data describe a model of bacterial dissemination across the BBB shedding light on a mechanism that might contribute to CNS tuberculosis infection and facilitate treatments.
Collapse
Affiliation(s)
- Trey E Gilpin
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI; and.,Graduate Training Program of Cellular and Molecular Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Fruzsina R Walter
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI; and
| | - Melinda Herbath
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI; and
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI; and
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI; and .,Graduate Training Program of Cellular and Molecular Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
5
|
Agnew A, Nulty C, Creagh EM. Regulation, Activation and Function of Caspase-11 during Health and Disease. Int J Mol Sci 2021; 22:ijms22041506. [PMID: 33546173 PMCID: PMC7913190 DOI: 10.3390/ijms22041506] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 02/04/2023] Open
Abstract
Caspase-11 is a pro-inflammatory enzyme that is stringently regulated during its expression and activation. As caspase-11 is not constitutively expressed in cells, it requires a priming step for its upregulation, which occurs following the stimulation of pathogen and cytokine receptors. Once expressed, caspase-11 activation is triggered by its interaction with lipopolysaccharide (LPS) from Gram-negative bacteria. Being an initiator caspase, activated caspase-11 functions primarily through its cleavage of key substrates. Gasdermin D (GSDMD) is the primary substrate of caspase-11, and the GSDMD cleavage fragment generated is responsible for the inflammatory form of cell death, pyroptosis, via its formation of pores in the plasma membrane. Thus, caspase-11 functions as an intracellular sensor for LPS and an immune effector. This review provides an overview of caspase-11—describing its structure and the transcriptional mechanisms that govern its expression, in addition to its activation, which is reported to be regulated by factors such as guanylate-binding proteins (GBPs), high mobility group box 1 (HMGB1) protein, and oxidized phospholipids. We also discuss the functional outcomes of caspase-11 activation, which include the non-canonical inflammasome, modulation of actin dynamics, and the initiation of blood coagulation, highlighting the importance of inflammatory caspase-11 during infection and disease.
Collapse
|
6
|
Chauhan P, Dandapat J, Sarkar A, Saha B. March of Mycobacterium: miRNAs intercept host cell CD40 signalling. Clin Transl Immunology 2020; 9:e1179. [PMID: 33072321 PMCID: PMC7541823 DOI: 10.1002/cti2.1179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/22/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
The disease tuberculosis is fatal if untreated. It is caused by the acid-fast bacilli Mycobacterium tuberculosis. Mycobacterium resides and replicates within the alveolar macrophages, causing inflammation and granuloma, wherein macrophage-T cell interactions enhance the inflammation-causing pulmonary caseous lesions. The first interactions between Mycobacterium and the receptors on macrophages decide the fate of Mycobacterium because of phagolysosomal impairments and the expression of several miRNAs, which may regulate CD40 expression on macrophages. While the altered phagolysosomal functions impede antigen presentation to the T cell-expressed antigen receptor, the interactions between the macrophage-expressed CD40 and the T cell-expressed CD40-ligand (CD40L or CD154) provide signals to T cells and Mycobacterium-infected macrophages. These two functions significantly influence the resolution or persistence of Mycobacterium infection. CD40 controls T-cell polarisation and host-protective immunity by eliciting interleukin-12p40, nitric oxide, reactive oxygen species and IFN-γ production. Indeed, CD40-deficient mice succumb to low-dose aerosol infection with Mycobacterium because of deficient interleukin (IL)-12 production leading to impaired IFN-γ-secreting T-cell response. In contrast, despite generating fewer granulomas, the CD40L-deficient mice developed anti-mycobacterial T-cell responses to the levels observed in the wild-type mice. These host-protective responses are significantly subdued by the Mycobacterium-infected macrophage produced TGF-β and IL-10, which promote pro-mycobacterial T-cell responses. The CD40-CD40L-induced counteractive immune responses against Mycobacterium thus present a conundrum that we explain here with a reconciliatory hypothesis. Experimental validation of the hypothesis will provide a rationale for designing anti-tubercular immunotherapy.
Collapse
Affiliation(s)
| | | | - Arup Sarkar
- Trident Academy of Creative TechnologyBhubaneswarIndia
| | - Bhaskar Saha
- National Centre for Cell Science (NCCS)PuneIndia
- Trident Academy of Creative TechnologyBhubaneswarIndia
| |
Collapse
|
7
|
Mishra M, Dadhich R, Mogha P, Kapoor S. Mycobacterium Lipids Modulate Host Cell Membrane Mechanics, Lipid Diffusivity, and Cytoskeleton in a Virulence-Selective Manner. ACS Infect Dis 2020; 6:2386-2399. [PMID: 32786287 DOI: 10.1021/acsinfecdis.0c00128] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Microbial lipids play a critical role in the pathogenesis of infectious diseases by modulating the host cell membrane properties, including lipid/protein diffusion and membrane organization. Mycobacterium tuberculosis (Mtb) synthesizes various chemically distinct lipids that are exposed on its outer membrane and interact with host cell membranes. However, the effects of the structurally diverse Mtb lipids on the host cell membrane properties to fine-tune the host cellular response remain unknown. In this study, we employed membrane biophysics and cell biology to assess the effects of different Mtb lipids on cell membrane mechanics, lipid diffusion, and the cytoskeleton of THP-1 macrophages. We found that Mtb lipids modulate macrophage membrane properties, actin cytoskeleton, and biochemical processes, such as protein phosphorylation and lipid peroxidation, in a virulence lipid-selective manner. These results emphasize that Mtb can fine-tune its interactions with the host cells governed by modulating the lipid profile on its surface. These observations provide a novel lipid-centric paradigm of Mtb pathogenesis that is amenable to pharmacological inhibition and could promote the development of robust biomarkers of Mtb infection and pathogenesis.
Collapse
Affiliation(s)
- Manjari Mishra
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Pankaj Mogha
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
8
|
To K, Cao R, Yegiazaryan A, Owens J, Venketaraman V. General Overview of Nontuberculous Mycobacteria Opportunistic Pathogens: Mycobacterium avium and Mycobacterium abscessus. J Clin Med 2020; 9:E2541. [PMID: 32781595 PMCID: PMC7463534 DOI: 10.3390/jcm9082541] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/02/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022] Open
Abstract
Nontuberculous mycobacteria (NTM) are emerging human pathogens, causing a wide range of clinical diseases affecting individuals who are immunocompromised and who have underlying health conditions. NTM are ubiquitous in the environment, with certain species causing opportunistic infection in humans, including Mycobacterium avium and Mycobacterium abscessus. The incidence and prevalence of NTM infections are rising globally, especially in developed countries with declining incidence rates of M. tuberculosis infection. Mycobacterium avium, a slow-growing mycobacterium, is associated with Mycobacterium avium complex (MAC) infections that can cause chronic pulmonary disease, disseminated disease, as well as lymphadenitis. M. abscessus infections are considered one of the most antibiotic-resistant mycobacteria and are associated with pulmonary disease, especially cystic fibrosis, as well as contaminated traumatic skin wounds, postsurgical soft tissue infections, and healthcare-associated infections (HAI). Clinical manifestations of diseases depend on the interaction of the host's immune response and the specific mycobacterial species. This review will give a general overview of the general characteristics, vulnerable populations most at risk, pathogenesis, treatment, and prevention for infections caused by Mycobacterium avium, in the context of MAC, and M. abscessus.
Collapse
Affiliation(s)
- Kimberly To
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA; (K.T.); (A.Y.)
| | - Ruoqiong Cao
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA; (R.C.); (J.O.)
| | - Aram Yegiazaryan
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA; (K.T.); (A.Y.)
| | - James Owens
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA; (R.C.); (J.O.)
| | - Vishwanath Venketaraman
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA; (R.C.); (J.O.)
| |
Collapse
|
9
|
Budzik JM, Swaney DL, Jimenez-Morales D, Johnson JR, Garelis NE, Repasy T, Roberts AW, Popov LM, Parry TJ, Pratt D, Ideker T, Krogan NJ, Cox JS. Dynamic post-translational modification profiling of Mycobacterium tuberculosis-infected primary macrophages. eLife 2020; 9:e51461. [PMID: 31951200 PMCID: PMC7030789 DOI: 10.7554/elife.51461] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/16/2020] [Indexed: 12/23/2022] Open
Abstract
Macrophages are highly plastic cells with critical roles in immunity, cancer, and tissue homeostasis, but how these distinct cellular fates are triggered by environmental cues is poorly understood. To uncover how primary murine macrophages respond to bacterial pathogens, we globally assessed changes in post-translational modifications of proteins during infection with Mycobacterium tuberculosis, a notorious intracellular pathogen. We identified hundreds of dynamically regulated phosphorylation and ubiquitylation sites, indicating that dramatic remodeling of multiple host pathways, both expected and unexpected, occurred during infection. Most of these cellular changes were not captured by mRNA profiling, and included activation of ubiquitin-mediated autophagy, an evolutionarily ancient cellular antimicrobial system. This analysis also revealed that a particular autophagy receptor, TAX1BP1, mediates clearance of ubiquitylated Mtb and targets bacteria to LC3-positive phagophores. These studies provide a new resource for understanding how macrophages shape their proteome to meet the challenge of infection.
Collapse
Affiliation(s)
- Jonathan M Budzik
- Department of MedicineUniversity of California, San FranciscoSan FranciscoUnited States
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Danielle L Swaney
- Department of Cellular and Molecular PharmacologyUniversity of California, San FranciscoSan FranciscoUnited States
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoUnited States
- Gladstone InstitutesSan FranciscoUnited States
| | - David Jimenez-Morales
- Department of Cellular and Molecular PharmacologyUniversity of California, San FranciscoSan FranciscoUnited States
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoUnited States
- Gladstone InstitutesSan FranciscoUnited States
- Department of Medicine, Division of Cardiovascular MedicineStanford UniversityStanfordUnited States
| | - Jeffrey R Johnson
- Department of Cellular and Molecular PharmacologyUniversity of California, San FranciscoSan FranciscoUnited States
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoUnited States
- Gladstone InstitutesSan FranciscoUnited States
| | - Nicholas E Garelis
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Teresa Repasy
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Allison W Roberts
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Lauren M Popov
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Trevor J Parry
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Dexter Pratt
- Department of MedicineUniversity of California, San DiegoLa JollaUnited States
| | - Trey Ideker
- Department of MedicineUniversity of California, San DiegoLa JollaUnited States
| | - Nevan J Krogan
- Department of Cellular and Molecular PharmacologyUniversity of California, San FranciscoSan FranciscoUnited States
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoUnited States
- Gladstone InstitutesSan FranciscoUnited States
| | - Jeffery S Cox
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
10
|
Jati S, Sarraf TR, Naskar D, Sen M. Wnt Signaling: Pathogen Incursion and Immune Defense. Front Immunol 2019; 10:2551. [PMID: 31736969 PMCID: PMC6828841 DOI: 10.3389/fimmu.2019.02551] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022] Open
Abstract
Wnt ligands interact with the transmembrane cell surface receptors Frizzled and ROR/RYK to initiate complex signaling cascades that are crucial for cell physiology and the proper functioning of the immune system. Wnt signaling is instrumental in maintaining immune surveillance and during infections by pathogenic microbes helps mount host resistance to infection. Some pathogens, however, utilize Wnt signaling to build a niche for their survival. The goal of this review is to summarize current and developing concepts about the tug of war between Wnt signaling and pathogens for deployment of host resources, focusing mostly on macrophages and cytoskeletal actin dynamics. An additional objective is to outline the interrelation between Wnt signaling and the host microbiota, which is vital for immune defense, discussing in the same perspective, how Wnt signaling could be differentiating pathogen from non-pathogen.
Collapse
Affiliation(s)
- Suborno Jati
- Division of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| | - Tresa Rani Sarraf
- Division of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| | - Debdut Naskar
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | - Malini Sen
- Division of Cancer Biology and Inflammatory Disorder, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
11
|
Mishra M, Adhyapak P, Dadhich R, Kapoor S. Dynamic Remodeling of the Host Cell Membrane by Virulent Mycobacterial Sulfoglycolipid-1. Sci Rep 2019; 9:12844. [PMID: 31492926 PMCID: PMC6731295 DOI: 10.1038/s41598-019-49343-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 08/23/2019] [Indexed: 12/31/2022] Open
Abstract
Lipids dictate membrane properties to modulate lateral membrane organization, lipid/protein diffusion and lipid-protein interactions, thereby underpinning proper functioning of cells. Mycobacterium tuberculosis harnesses the power of its atypical cell wall lipids to impact immune surveillance machinery centered at the host cell membrane. However, the role of specific virulent lipids in altering host cellular functions by modulating membrane organization and the associated signaling response are still pertinent unresolved questions. Here, combining membrane biophysics and cell biology, we elucidate how virulent Mtb sulfoglycolipids hijack the host cell membrane, affecting its order, fluidity, and stiffness along with manipulating the linked cytoskeleton. The functional outcome of this perturbation was assayed by monitoring membrane-associated autophagy signaling. These actions form a part of the overall response to commandeer host membrane-associated immune processes during infection. The findings on the mechanism of action of Mtb lipids on host cell membrane structure and downstream signaling will deepen the collective understanding of their functional aspects in membrane-dictated bacterial survival, pathogenesis and drug resistance and reveal suitable membrane driven-therapeutic intervention points and diagnostic tools.
Collapse
Affiliation(s)
- Manjari Mishra
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India
| | - Pranav Adhyapak
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India
| | - Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India.
| |
Collapse
|
12
|
Bussi C, Gutierrez MG. Mycobacterium tuberculosis infection of host cells in space and time. FEMS Microbiol Rev 2019; 43:341-361. [PMID: 30916769 PMCID: PMC6606852 DOI: 10.1093/femsre/fuz006] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/26/2019] [Indexed: 12/16/2022] Open
Abstract
Tuberculosis (TB) caused by the bacterial pathogen Mycobacterium tuberculosis (Mtb) remains one of the deadliest infectious diseases with over a billion deaths in the past 200 years (Paulson 2013). TB causes more deaths worldwide than any other single infectious agent, with 10.4 million new cases and close to 1.7 million deaths in 2017. The obstacles that make TB hard to treat and eradicate are intrinsically linked to the intracellular lifestyle of Mtb. Mtb needs to replicate within human cells to disseminate to other individuals and cause disease. However, we still do not completely understand how Mtb manages to survive within eukaryotic cells and why some cells are able to eradicate this lethal pathogen. Here, we summarise the current knowledge of the complex host cell-pathogen interactions in TB and review the cellular mechanisms operating at the interface between Mtb and the human host cell, highlighting the technical and methodological challenges to investigating the cell biology of human host cell-Mtb interactions.
Collapse
Affiliation(s)
- Claudio Bussi
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Maximiliano G Gutierrez
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| |
Collapse
|
13
|
Carranza C, Chavez-Galan L. Several Routes to the Same Destination: Inhibition of Phagosome-Lysosome Fusion by Mycobacterium tuberculosis. Am J Med Sci 2019; 357:184-194. [DOI: 10.1016/j.amjms.2018.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/29/2018] [Accepted: 12/06/2018] [Indexed: 02/04/2023]
|
14
|
Chaurasiya SK. Tuberculosis: Smart manipulation of a lethal host. Microbiol Immunol 2018; 62:361-379. [PMID: 29687912 DOI: 10.1111/1348-0421.12593] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/21/2018] [Accepted: 04/16/2018] [Indexed: 11/28/2022]
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis remains a global threat to human health. Development of drug resistance and co-infection with HIV has increased the morbidity and mortality caused by TB. Macrophages serve as primary defense against microbial infections, including TB. Upon recognition and uptake of mycobacteria, macrophages initiate a series of events designed to lead to generation of effective immune responses and clearance of infection. However, pathogenic mycobacteria utilize multiple mechanisms for manipulating macrophage responses to protect itself from being killed and to survive within these cells that are designed to kill them. The outcomes of mycobacterial infection are determined by several host- and pathogen-related factors. Significant advancements in understanding mycobacterial pathogenesis have been made in recent years. In this review, some of the important factors/mechanisms regulating mycobacterial survival inside macrophages are discussed.
Collapse
Affiliation(s)
- Shivendra K Chaurasiya
- Host-pathogen Interaction and Signal Transduction Laboratory, Department of Microbiology, School of Biological Sciences, Dr. Hari Singh Gour University, Sagar, MP-470003, India
| |
Collapse
|
15
|
CD82 hypomethylation is essential for tuberculosis pathogenesis via regulation of RUNX1-Rab5/22. Exp Mol Med 2018; 50:1-15. [PMID: 29760437 PMCID: PMC5951854 DOI: 10.1038/s12276-018-0091-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 01/23/2023] Open
Abstract
The tumor suppressor gene CD82/KAI1 is a member of the tetraspanin superfamily and organizes various membrane-based processes. Mycobacterium tuberculosis (MTB) persists in host macrophages by interfering with phagolysosome biogenesis and inflammatory responses, but the role of CD82 in controlling the intracellular survival of pathogenic mycobacteria within macrophages remains poorly understood. In this study, we demonstrated that the virulent MTB strain H37Rv (MTB Rv) induced CD82 promoter hypomethylation, resulting in CD82 expression. Targeting of the runt-related transcription factor 1 (RUNX1) by CD82 is essential for phagosome arrest via interacting with Rab5/22. This arrest is required for the intracellular growth of MTB in vitro and in vivo, but not for that of MTB H37Ra (MTB Ra) in macrophages. In addition, knockdown or knockout of CD82 or RUNX1 increased antibacterial host defense via phagolysosome biogenesis, inflammatory cytokine production, and subsequent antimicrobial activity both in vitro and in vivo. Notably, the levels of CD82 and RUNX1 in granulomas were elevated in tuberculosis (TB) patients, indicating that CD82 and RUNX1 have clinical significance in human TB. Our findings identify a previously unrecognized role of CD82 hypomethylation in the regulation of phagosome maturation, enhanced intracellular survival, and the innate host immune response to MTB. Thus, the CD82-RUNX1-Rab5/22 axis may be a previously unrecognized virulence mechanism of MTB pathogenesis.
Collapse
|
16
|
Song OR, Queval CJ, Iantomasi R, Delorme V, Marion S, Veyron-Churlet R, Werkmeister E, Popoff M, Ricard I, Jouny S, Deboosere N, Lafont F, Baulard A, Yeramian E, Marsollier L, Hoffmann E, Brodin P. ArfGAP1 restricts Mycobacterium tuberculosis entry by controlling the actin cytoskeleton. EMBO Rep 2017; 19:29-42. [PMID: 29141986 DOI: 10.15252/embr.201744371] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 10/03/2017] [Accepted: 10/23/2017] [Indexed: 11/09/2022] Open
Abstract
The interaction of Mycobacterium tuberculosis (Mtb) with pulmonary epithelial cells is critical for early stages of bacillus colonization and during the progression of tuberculosis. Entry of Mtb into epithelial cells has been shown to depend on F-actin polymerization, though the molecular mechanisms are still unclear. Here, we demonstrate that mycobacterial uptake into epithelial cells requires rearrangements of the actin cytoskeleton, which are regulated by ADP-ribosylation factor 1 (Arf1) and phospholipase D1 (PLD1), and is dependent on the M3 muscarinic receptor (M3R). We show that this pathway is controlled by Arf GTPase-activating protein 1 (ArfGAP1), as its silencing has an impact on actin cytoskeleton reorganization leading to uncontrolled uptake and replication of Mtb. Furthermore, we provide evidence that this pathway is critical for mycobacterial entry, while the cellular infection with other pathogens, such as Shigella flexneri and Yersinia pseudotuberculosis, is not affected. Altogether, these results reveal how cortical actin plays the role of a barrier to prevent mycobacterial entry into epithelial cells and indicate a novel role for ArfGAP1 as a restriction factor of host-pathogen interactions.
Collapse
Affiliation(s)
- Ok-Ryul Song
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France.,Equipe ATIP AVENIR, CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Institute Pasteur Korea, Seongnam-si Gyeonggi-do, South Korea
| | - Christophe J Queval
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Raffaella Iantomasi
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Vincent Delorme
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France.,Institute Pasteur Korea, Seongnam-si Gyeonggi-do, South Korea
| | - Sabrina Marion
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Romain Veyron-Churlet
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Elisabeth Werkmeister
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Michka Popoff
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France.,CNRS, UMR8520, Institut d'électronique, de microélectronique et de nanotechnologie, Villeneuve d'Ascq, France
| | - Isabelle Ricard
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Samuel Jouny
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Nathalie Deboosere
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Frank Lafont
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Alain Baulard
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Edouard Yeramian
- Unité de Microbiologie Structurale, CNRS UMR3528, Institut Pasteur, Paris, France
| | - Laurent Marsollier
- Equipe ATIP AVENIR, CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Eik Hoffmann
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Priscille Brodin
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France .,Institute Pasteur Korea, Seongnam-si Gyeonggi-do, South Korea
| |
Collapse
|
17
|
Peddireddy V, Doddam SN, Ahmed N. Mycobacterial Dormancy Systems and Host Responses in Tuberculosis. Front Immunol 2017; 8:84. [PMID: 28261197 PMCID: PMC5309233 DOI: 10.3389/fimmu.2017.00084] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/18/2017] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB) caused by the intracellular pathogen, Mycobacterium tuberculosis (Mtb), claims more than 1.5 million lives worldwide annually. Despite promulgation of multipronged strategies to prevent and control TB, there is no significant downfall occurring in the number of new cases, and adding to this is the relapse of the disease due to the emergence of antibiotic resistance and the ability of Mtb to remain dormant after primary infection. The pathology of Mtb is complex and largely attributed to immune-evading strategies that this pathogen adopts to establish primary infection, its persistence in the host, and reactivation of pathogenicity under favorable conditions. In this review, we present various biochemical, immunological, and genetic strategies unleashed by Mtb inside the host for its survival. The bacterium enables itself to establish a niche by evading immune recognition via resorting to masking, establishment of dormancy by manipulating immune receptor responses, altering innate immune cell fate, enhancing granuloma formation, and developing antibiotic tolerance. Besides these, the regulatory entities, such as DosR and its regulon, encompassing various putative effector proteins play a vital role in maintaining the dormant nature of this pathogen. Further, reactivation of Mtb allows relapse of the disease and is favored by the genes of the Rtf family and the conditions that suppress the immune system of the host. Identification of target genes and characterizing the function of their respective antigens involved in primary infection, dormancy, and reactivation would likely provide vital clues to design novel drugs and/or vaccines for the control of dormant TB.
Collapse
Affiliation(s)
- Vidyullatha Peddireddy
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad , Hyderabad , India
| | - Sankara Narayana Doddam
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad , Hyderabad , India
| | - Niyaz Ahmed
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India; Laboratory Sciences and Services Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
18
|
Sen R, Nayak L, De RK. A review on host-pathogen interactions: classification and prediction. Eur J Clin Microbiol Infect Dis 2016; 35:1581-99. [PMID: 27470504 DOI: 10.1007/s10096-016-2716-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/22/2016] [Indexed: 01/01/2023]
Abstract
The research on host-pathogen interactions is an ever-emerging and evolving field. Every other day a new pathogen gets discovered, along with comes the challenge of its prevention and cure. As the intelligent human always vies for prevention, which is better than cure, understanding the mechanisms of host-pathogen interactions gets prior importance. There are many mechanisms involved from the pathogen as well as the host sides while an interaction happens. It is a vis-a-vis fight of the counter genes and proteins from both sides. Who wins depends on whether a host gets an infection or not. Moreover, a higher level of complexity arises when the pathogens evolve and become resistant to a host's defense mechanisms. Such pathogens pose serious challenges for treatment. The entire human population is in danger of such long-lasting persistent infections. Some of these infections even increase the rate of mortality. Hence there is an immediate emergency to understand how the pathogens interact with their host for successful invasion. It may lead to discovery of appropriate preventive measures, and the development of rational therapeutic measures and medication against such infections and diseases. This review, a state-of-the-art updated scenario of host-pathogen interaction research, has been done by keeping in mind this urgency. It covers the biological and computational aspects of host-pathogen interactions, classification of the methods by which the pathogens interact with their hosts, different machine learning techniques for prediction of host-pathogen interactions, and future scopes of this research field.
Collapse
Affiliation(s)
- R Sen
- Machine Intelligence Unit, Indian Statistical Institute, 203, Barrackpore Trunk Road, Kolkata, 700108, India
| | - L Nayak
- Machine Intelligence Unit, Indian Statistical Institute, 203, Barrackpore Trunk Road, Kolkata, 700108, India
| | - R K De
- Machine Intelligence Unit, Indian Statistical Institute, 203, Barrackpore Trunk Road, Kolkata, 700108, India.
| |
Collapse
|
19
|
Galbadage T, Shepherd TF, Cirillo SLG, Gumienny TL, Cirillo JD. The Caenorhabditis elegans p38 MAPK Gene plays a key role in protection from mycobacteria. Microbiologyopen 2016; 5:436-52. [PMID: 26919641 PMCID: PMC4905996 DOI: 10.1002/mbo3.341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 12/17/2022] Open
Abstract
Mitogen-activated protein kinases (MAPK) are critical mediators of cellular responses to pathogens and are activated in response to infection, but investigation is difficult in multi-cell hosts due to developmental lethality of mutations. Mycobacterium marinum (Mm) is an established model for tuberculosis, a disease afflicting nearly one-third of the world's population. We found that Mm-infected Caenorhabditis elegans display >80% mortality, but nonpathogenic M. smegmatis cause <15% mortality. C. elegans display pathological changes when infected with Mm, whereas Mm mutants produce lower mortality, suggesting that C. elegans is a promising virulence model for detailed genetic analysis. C. elegans MAPK mutants are hypersusceptible to mycobacterial infection; however, the C. elegans TOL-like, TGF-β and insulin-like pathway genes do not play important roles in susceptibility. We show that pathogenic mycobacteria inhibit MAPK-mediated protection through the MAPK phosphatase gene and demonstrate that C. elegans provide a genetically tractable pathogenicity model of both the host and pathogen.
Collapse
Affiliation(s)
- Thushara Galbadage
- Departments of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas, 77807-3260
| | - Tonya F Shepherd
- Departments of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas, 77807-3260
| | - Suat L G Cirillo
- Departments of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas, 77807-3260
| | - Tina L Gumienny
- Department of Biology, Texas Woman's University, Denton, Texas, 76204-5799
| | - Jeffrey D Cirillo
- Departments of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas, 77807-3260
| |
Collapse
|
20
|
Wang J, Yao Y, Wu J, Deng Z, Gu T, Tang X, Cheng Y, Li G. The mechanism of cytoskeleton protein β-actin and cofilin-1 of macrophages infected by Mycobacterium avium. Am J Transl Res 2016; 8:1055-1063. [PMID: 27158391 PMCID: PMC4846948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/12/2016] [Indexed: 06/05/2023]
Abstract
Cytoskeleton proteins and their regulation proteins could be influenced seriously in Mycobacterium tuberculosis infection host cells leading to the apoptosis of host cells. Macrophages infected by Mycobacterium avium were detected from cell morphology and genome levels to analyze changes of the cytoskeleton of M. avium infection macrophages. Then the expression of β-actin, cofilin-1 proteins in M. avium infected macrophages were analyzed by western blotting, and the apoptosis of M. avium infection macrophages were tested by flow cytometry. Results indicated that the morphology and genomic DNA of M. avium infection macrophages were not damaged significantly. Meanwhile, β-actin gene and its proteins in M. avium infection macrophages were both decreased, but its regulatory protein cofilin-1 was expressed conversely. Furthermore, macrophages could be induced to apoptosis due to M. avium infection by cytoskeleton changes. These findings contributed us to understand that macrophages infected by M. avium could be lead to apoptosis by regulating cytoskeleton protein β-actin or its regulatory protein cofilin-1.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu UniversityKunshan 215300, People’s Republic of China
| | - Yongliang Yao
- Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu UniversityKunshan 215300, People’s Republic of China
| | - Jianhong Wu
- Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu UniversityKunshan 215300, People’s Republic of China
| | - Zhiyong Deng
- Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu UniversityKunshan 215300, People’s Republic of China
| | - Tao Gu
- Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu UniversityKunshan 215300, People’s Republic of China
| | - Xin Tang
- Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu UniversityKunshan 215300, People’s Republic of China
| | - Yang Cheng
- Department of Clinical Laboratory, Kunshan First People’s Hospital, Affiliated to Jiangsu UniversityKunshan 215300, People’s Republic of China
| | - Guangxin Li
- Department of Pathology, Chong Qing Cancer InstituteChongqing 400030, People’s Republic of China
| |
Collapse
|
21
|
Caution K, Gavrilin MA, Tazi M, Kanneganti A, Layman D, Hoque S, Krause K, Amer AO. Caspase-11 and caspase-1 differentially modulate actin polymerization via RhoA and Slingshot proteins to promote bacterial clearance. Sci Rep 2015; 5:18479. [PMID: 26686473 PMCID: PMC4685268 DOI: 10.1038/srep18479] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/18/2015] [Indexed: 01/19/2023] Open
Abstract
Inflammasomes are multiprotein complexes that include members of the NOD-like receptor family and caspase-1. Caspase-1 is required for the fusion of the Legionella vacuole with lysosomes. Caspase-11, independently of the inflammasome, also promotes phagolysosomal fusion. However, it is unclear how these proteases alter intracellular trafficking. Here, we show that caspase-11 and caspase-1 function in opposing manners to phosphorylate and dephosphorylate cofilin, respectively upon infection with Legionella. Caspase-11 targets cofilin via the RhoA GTPase, whereas caspase-1 engages the Slingshot phosphatase. The absence of either caspase-11 or caspase-1 maintains actin in the polymerized or depolymerized form, respectively and averts the fusion of pathogen-containing vacuoles with lysosomes. Therefore, caspase-11 and caspase-1 converge on the actin machinery with opposing effects to promote vesicular trafficking.
Collapse
Affiliation(s)
- Kyle Caution
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, Columbus OH 43210.,Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| | - Mikhail A Gavrilin
- Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| | - Mia Tazi
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, Columbus OH 43210.,Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| | - Apurva Kanneganti
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, Columbus OH 43210.,Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| | - Daniel Layman
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, Columbus OH 43210.,Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| | - Sheshadri Hoque
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, Columbus OH 43210.,Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, Columbus OH 43210.,Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| | - Amal O Amer
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, Columbus OH 43210.,Dorothy M. Davis Heart and Lung Research Institute, and The Ohio State University, Columbus OH 43210
| |
Collapse
|
22
|
Zheng K, Kitazato K, Wang Y, He Z. Pathogenic microbes manipulate cofilin activity to subvert actin cytoskeleton. Crit Rev Microbiol 2015; 42:677-95. [PMID: 25853495 DOI: 10.3109/1040841x.2015.1010139] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Actin-depolymerizing factor (ADF)/cofilin proteins are key players in controlling the temporal and spatial extent of actin dynamics, which is crucial for mediating host-pathogen interactions. Pathogenic microbes have evolved molecular mechanisms to manipulate cofilin activity to subvert the actin cytoskeletal system in host cells, promoting their internalization into the target cells, modifying the replication niche and facilitating their intracellular and intercellular dissemination. The study of how these pathogens exploit cofilin pathways is crucial for understanding infectious disease and providing potential targets for drug therapies.
Collapse
Affiliation(s)
- Kai Zheng
- a Department of Pharmacy, School of Medicine , Shenzhen University , Shenzhen , Guangdong , People's Republic of China .,c Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University , Guangzhou , China
| | - Kaio Kitazato
- b Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology , Nagasaki University , Nagasaki , Japan , and
| | - Yifei Wang
- c Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University , Guangzhou , China
| | - Zhendan He
- a Department of Pharmacy, School of Medicine , Shenzhen University , Shenzhen , Guangdong , People's Republic of China
| |
Collapse
|
23
|
Fu YR, Gao KS, Ji R, Yi ZJ. Differential transcriptional response in macrophages infected with cell wall deficient versus normal Mycobacterium Tuberculosis. Int J Biol Sci 2015; 11:22-30. [PMID: 25552926 PMCID: PMC4278251 DOI: 10.7150/ijbs.10217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/13/2014] [Indexed: 02/06/2023] Open
Abstract
Host-pathogen interactions determine the outcome following infection by mycobacterium tuberculosis (Mtb). Under adverse circumstances, normal Mtb can form cell-wall deficient (CWD) variants within macrophages, which have been considered an adaptive strategy for facilitating bacterial survival inside macrophages. However, the molecular mechanism by which infection of macrophages with different phenotypic Mtb elicits distinct responses of macrophages is not fully understood. To explore the molecular events triggered upon Mtb infection of macrophages, differential transcriptional responses of RAW264.7 cells infected with two forms of Mtb, CWD-Mtb and normal Mtb, were studied by microarray analysis. Some of the differentially regulated genes were confirmed by RT-qPCR in both RAW264.7 cells and primary macrophages. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway was used to analyze functions of differentially expressed genes. Distinct gene expression patterns were observed between CWD-Mtb and normal Mtb group. Mapt was up-regulated, while NOS2 and IL-11 were down-regulated in CWD-Mtb infected RAW264.7 cells and primary macrophages compared with normal Mtb infected ones. Many deregulated genes were found to be related to macrophages activation, immune response, phagosome maturation, autophagy and lipid metabolism. KEGG analysis showed that the differentially expressed genes were mainly involved in MAPK signaling pathway, nitrogen metabolism, cytokine-cytokine receptor interaction and focal adhesion. Taken together, the present study showed that differential macrophage responses were induced by intracellular CWD-Mtb an normal Mtb infection, which suggested that interactions between macrophages and different phenotypic Mtb are very complex. The results provide evidence for further understanding of pathogenesis of CWD-Mtb and may help in improving strategies to eliminate intracellular CWD-Mtb.
Collapse
Affiliation(s)
- Yu-Rong Fu
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China; ; 2. Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Kun-Shan Gao
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China
| | - Rui Ji
- 2. Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Zheng-Jun Yi
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China
| |
Collapse
|
24
|
Talmale S, Bhujade A, Patil M. Anti-allergic and anti-inflammatory properties of Zizyphus mauritiana root bark. Food Funct 2015; 6:2975-83. [DOI: 10.1039/c5fo00270b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The MAF, a fraction with potent anti-allergic and anti-inflammatory compounds, is isolated fromZizyphus mauritianaroot bark. The MAF has an excellent ability to inhibit the complement system, COX-1, COX-2 and 5-LOX and has the potential to prevent anaphylactic shock and the Arthus reaction.
Collapse
Affiliation(s)
- Suhas Talmale
- University Department of Biochemistry
- RTM Nagpur University
- Nagpur 440033
- India
| | - Arti Bhujade
- University Department of Biochemistry
- RTM Nagpur University
- Nagpur 440033
- India
| | - Mandakini Patil
- University Department of Biochemistry
- RTM Nagpur University
- Nagpur 440033
- India
| |
Collapse
|
25
|
Zhou H, Gao S, Nguyen NN, Fan M, Jin J, Liu B, Zhao L, Xiong G, Tan M, Li S, Wong L. Stringent homology-based prediction of H. sapiens-M. tuberculosis H37Rv protein-protein interactions. Biol Direct 2014; 9:5. [PMID: 24708540 PMCID: PMC4022245 DOI: 10.1186/1745-6150-9-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 03/26/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND H. sapiens-M. tuberculosis H37Rv protein-protein interaction (PPI) data are essential for understanding the infection mechanism of the formidable pathogen M. tuberculosis H37Rv. Computational prediction is an important strategy to fill the gap in experimental H. sapiens-M. tuberculosis H37Rv PPI data. Homology-based prediction is frequently used in predicting both intra-species and inter-species PPIs. However, some limitations are not properly resolved in several published works that predict eukaryote-prokaryote inter-species PPIs using intra-species template PPIs. RESULTS We develop a stringent homology-based prediction approach by taking into account (i) differences between eukaryotic and prokaryotic proteins and (ii) differences between inter-species and intra-species PPI interfaces. We compare our stringent homology-based approach to a conventional homology-based approach for predicting host-pathogen PPIs, based on cellular compartment distribution analysis, disease gene list enrichment analysis, pathway enrichment analysis and functional category enrichment analysis. These analyses support the validity of our prediction result, and clearly show that our approach has better performance in predicting H. sapiens-M. tuberculosis H37Rv PPIs. Using our stringent homology-based approach, we have predicted a set of highly plausible H. sapiens-M. tuberculosis H37Rv PPIs which might be useful for many of related studies. Based on our analysis of the H. sapiens-M. tuberculosis H37Rv PPI network predicted by our stringent homology-based approach, we have discovered several interesting properties which are reported here for the first time. We find that both host proteins and pathogen proteins involved in the host-pathogen PPIs tend to be hubs in their own intra-species PPI network. Also, both host and pathogen proteins involved in host-pathogen PPIs tend to have longer primary sequence, tend to have more domains, tend to be more hydrophilic, etc. And the protein domains from both host and pathogen proteins involved in host-pathogen PPIs tend to have lower charge, and tend to be more hydrophilic. CONCLUSIONS Our stringent homology-based prediction approach provides a better strategy in predicting PPIs between eukaryotic hosts and prokaryotic pathogens than a conventional homology-based approach. The properties we have observed from the predicted H. sapiens-M. tuberculosis H37Rv PPI network are useful for understanding inter-species host-pathogen PPI networks and provide novel insights for host-pathogen interaction studies.
Collapse
Affiliation(s)
- Hufeng Zhou
- NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
- Department of Medicine, Brigham and Women’s Hospital, Boston, USA
- Department of Microbiology and Immunobiology, Harvard University, Cambridge, USA
| | - Shangzhi Gao
- Department of Environmental Health, Harvard School of Public Health, Harvard University, Cambridge, USA
| | - Nam Ninh Nguyen
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Mengyuan Fan
- NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Jingjing Jin
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Bing Liu
- Computer Science Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Liang Zhao
- Bioinformatics Research Center, & School of Computer Engineering, Nanyang Technological University, Singapore, Singapore
| | - Geng Xiong
- Department of Medicine, Brigham and Women’s Hospital, Boston, USA
| | - Min Tan
- Department of Medicine, Brigham and Women’s Hospital, Boston, USA
- Department of Microbiology and Immunobiology, Harvard University, Cambridge, USA
| | - Shijun Li
- Department of Medicine, Brigham and Women’s Hospital, Boston, USA
- Department of Microbiology and Immunobiology, Harvard University, Cambridge, USA
| | - Limsoon Wong
- School of Computing, National University of Singapore, Singapore, Singapore
| |
Collapse
|
26
|
Zhou H, Rezaei J, Hugo W, Gao S, Jin J, Fan M, Yong CH, Wozniak M, Wong L. Stringent DDI-based prediction of H. sapiens-M. tuberculosis H37Rv protein-protein interactions. BMC SYSTEMS BIOLOGY 2013; 7 Suppl 6:S6. [PMID: 24564941 PMCID: PMC4029759 DOI: 10.1186/1752-0509-7-s6-s6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND H. sapiens-M. tuberculosis H37Rv protein-protein interaction (PPI) data are very important information to illuminate the infection mechanism of M. tuberculosis H37Rv. But current H. sapiens-M. tuberculosis H37Rv PPI data are very scarce. This seriously limits the study of the interaction between this important pathogen and its host H. sapiens. Computational prediction of H. sapiens-M. tuberculosis H37Rv PPIs is an important strategy to fill in the gap. Domain-domain interaction (DDI) based prediction is one of the frequently used computational approaches in predicting both intra-species and inter-species PPIs. However, the performance of DDI-based host-pathogen PPI prediction has been rather limited. RESULTS We develop a stringent DDI-based prediction approach with emphasis on (i) differences between the specific domain sequences on annotated regions of proteins under the same domain ID and (ii) calculation of the interaction strength of predicted PPIs based on the interacting residues in their interaction interfaces. We compare our stringent DDI-based approach to a conventional DDI-based approach for predicting PPIs based on gold standard intra-species PPIs and coherent informative Gene Ontology terms assessment. The assessment results show that our stringent DDI-based approach achieves much better performance in predicting PPIs than the conventional approach. Using our stringent DDI-based approach, we have predicted a small set of reliable H. sapiens-M. tuberculosis H37Rv PPIs which could be very useful for a variety of related studies. We also analyze the H. sapiens-M. tuberculosis H37Rv PPIs predicted by our stringent DDI-based approach using cellular compartment distribution analysis, functional category enrichment analysis and pathway enrichment analysis. The analyses support the validity of our prediction result. Also, based on an analysis of the H. sapiens-M. tuberculosis H37Rv PPI network predicted by our stringent DDI-based approach, we have discovered some important properties of domains involved in host-pathogen PPIs. We find that both host and pathogen proteins involved in host-pathogen PPIs tend to have more domains than proteins involved in intra-species PPIs, and these domains have more interaction partners than domains on proteins involved in intra-species PPI. CONCLUSIONS The stringent DDI-based prediction approach reported in this work provides a stringent strategy for predicting host-pathogen PPIs. It also performs better than a conventional DDI-based approach in predicting PPIs. We have predicted a small set of accurate H. sapiens-M. tuberculosis H37Rv PPIs which could be very useful for a variety of related studies.
Collapse
|
27
|
Kolonko M, Geffken AC, Blumer T, Hagens K, Schaible UE, Hagedorn M. WASH-driven actin polymerization is required for efficient mycobacterial phagosome maturation arrest. Cell Microbiol 2013; 16:232-46. [PMID: 24119059 DOI: 10.1111/cmi.12217] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/21/2013] [Accepted: 09/19/2013] [Indexed: 12/24/2022]
Abstract
Pathogenic mycobacteria survive in phagocytic host cells primarily as a result of their ability to prevent fusion of their vacuole with lysosomes, thereby avoiding a bactericidal environment. The molecular mechanisms to establish and maintain this replication compartment are not well understood. By combining molecular and microscopical approaches we show here that after phagocytosis the actin nucleation-promoting factor WASH associates and generates F-actin on the mycobacterial vacuole. Disruption of WASH or depolymerization of F-actin leads to the accumulation of the proton-pumping V-ATPase around the mycobacterial vacuole, its acidification and reduces the viability of intracellular mycobacteria. This effect is observed for M. marinum in the model phagocyte Dictyostelium but also for M. marinum and M. tuberculosis in mammalian phagocytes. This demonstrates an evolutionarily conserved mechanism by which pathogenic mycobacteria subvert the actin-polymerization activity of WASH to prevent phagosome acidification and maturation, as a prerequisite to generate and maintain a replicative niche.
Collapse
Affiliation(s)
- Margot Kolonko
- Section Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Ignatov D, Malakho S, Majorov K, Skvortsov T, Apt A, Azhikina T. RNA-Seq analysis of Mycobacterium avium non-coding transcriptome. PLoS One 2013; 8:e74209. [PMID: 24066122 PMCID: PMC3774663 DOI: 10.1371/journal.pone.0074209] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/29/2013] [Indexed: 12/25/2022] Open
Abstract
Deep sequencing was implemented to study the transcriptional landscape of Mycobacterium avium. High-resolution transcriptome analysis identified the transcription start points for 652 genes. One third of these genes represented leaderless transcripts, whereas the rest of the transcripts had 5' UTRs with the mean length of 83 nt. In addition, the 5' UTRs of 6 genes contained SAM-IV and Ykok types of riboswitches. 87 antisense RNAs and 10 intergenic small RNAs were mapped. 6 intergenic small RNAs, including 4.5S RNA and rnpB, were transcribed at extremely high levels. Although several intergenic sRNAs are conserved in M. avium and M. tuberculosis, both of these species have unique intergenic sRNAs. Moreover, we demonstrated that even conserved small RNAs are regulated differently in these species. Different sets of intergenic sRNAs may underlie differences in physiology between conditionally pathogenic M. avium and highly specialized pathogen M. tuberculosis.
Collapse
Affiliation(s)
- Dmitriy Ignatov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- * E-mail:
| | - Sofia Malakho
- Center of Innovations and Technologies “Biologically Active Compounds and their Applications”, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Timofey Skvortsov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | | | - Tatyana Azhikina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
29
|
Basler T, Brumshagen C, Beineke A, Goethe R, Bäumer W. Mycobacterium avium subspecies impair dendritic cell maturation. Innate Immun 2013; 19:451-61. [DOI: 10.1177/1753425912470291] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Mycobacterium avium ssp. paratuberculosis (MAP) causes Johne’s disease, a chronic, granulomatous enteritis of ruminants. Dendritic cells (DC) of the gut are ideally placed to combat invading mycobacteria; however, little is known about their interaction with MAP. Here, we investigated the interaction of MAP and the closely related M. avium ssp. avium (MAA) with murine DC and the effect of infected macrophages on DC maturation. The infection of DC with MAP or MAA induced DC maturation, which differed to that of LPS as maturation was accompanied by higher production of IL-10 and lower production of IL-12. Treatment of maturing DC with supernatants from mycobacteria-infected macrophages resulted in impaired DC maturation, leading to a semi-mature, tolerogenic DC phenotype expressing low levels of MHCII, CD86 and TNF-α after LPS stimulation. Though the cells were not completely differentiated they responded with an increased IL-10 and a decreased IL-12 production. Using recombinant cytokines we provide evidence that the semi-mature DC phenotype results from a combination of secreted cytokines and released antigenic mycobacterial components of the infected macrophage. Our results indicate that MAP and MAA are able to subvert DC function directly by infecting and indirectly via the milieu created by infected macrophages.
Collapse
Affiliation(s)
- Tina Basler
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Christina Brumshagen
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ralph Goethe
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wolfgang Bäumer
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
30
|
Pan F, Zhao Y, Zhu S, Sun C, Lei L, Feng X, Han WY. Different transcriptional profiles of RAW264.7 infected with Mycobacterium tuberculosis H37Rv and BCG identified via deep sequencing. PLoS One 2012; 7:e51988. [PMID: 23284841 PMCID: PMC3526534 DOI: 10.1371/journal.pone.0051988] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 11/13/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The Mycobacterium tuberculosis H37Rv and BCG effects on the host cell transcriptional profile consider a main research point. In the present study the transcriptome profiling analysis of RAW264.7 either infected with Mycobacterium tuberculosis H37Rv or BCG have been reported using Solexa/Illumina digital gene expression (DGE). RESULTS The DGE analysis showed 1,917 different expressed genes between the BCG and H37Rv group. In addition, approximately 5% of the transcripts appeared to be predicted genes that have never been described before. KEGG Orthology (KO) annotations showed more than 71% of these transcripts are possibly involved in approximately 210 known metabolic or signaling pathways. The gene of the 28 pathways about pathogen recognition receptors and Mycobacterium tuberculosis interaction with macrophages were analyzed using the CLUSTER 3.0 available, the Tree View tool and Gene Orthology (GO). Some genes were randomly selected to confirm their altered expression levels by quantitative real-time PCR (qRT-PCR). CONCLUSION The present study used DGE from pathogen recognition receptors and Mycobacterium tuberculosis interaction with macrophages to understand the interplay between Mycobacterium tuberculosis and RAW264.7. Meanwhile find some important host protein which was affected by Mycobacterium tuberculosis to provide evidence for the further improvement of the present efficacy of existing Mycobacterium tuberculosis therapy and vaccine.
Collapse
Affiliation(s)
- Fengguang Pan
- Laboratory of Food Safety and Hygienic Inspection, Jilin University, Changchun, Jilin, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yaya Zhao
- Laboratory of Food Safety and Hygienic Inspection, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Seng Zhu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Changjiang Sun
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
- * E-mail: (CS); (WyH)
| | - Liancheng Lei
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xin Feng
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Wen yu Han
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
- * E-mail: (CS); (WyH)
| |
Collapse
|
31
|
Akhter A, Caution K, Abu Khweek A, Tazi M, Abdulrahman BA, Abdelaziz DHA, Voss OH, Doseff AI, Hassan H, Azad AK, Schlesinger LS, Wewers MD, Gavrilin MA, Amer AO. Caspase-11 promotes the fusion of phagosomes harboring pathogenic bacteria with lysosomes by modulating actin polymerization. Immunity 2012; 37:35-47. [PMID: 22658523 DOI: 10.1016/j.immuni.2012.05.001] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 03/14/2012] [Accepted: 05/02/2012] [Indexed: 01/24/2023]
Abstract
Inflammasomes are multiprotein complexes that include members of the NLR (nucleotide-binding domain leucine-rich repeat containing) family and caspase-1. Once bacterial molecules are sensed within the macrophage, the inflammasome is assembled, mediating the activation of caspase-1. Caspase-11 mediates caspase-1 activation in response to lipopolysaccharide and bacterial toxins, and yet its role during bacterial infection is unknown. Here, we demonstrated that caspase-11 was dispensable for caspase-1 activation in response to Legionella, Salmonella, Francisella, and Listeria. We also determined that active mouse caspase-11 was required for restriction of L. pneumophila infection. Similarly, human caspase-4 and caspase-5, homologs of mouse caspase-11, cooperated to restrict L. pneumophila infection in human macrophages. Caspase-11 promoted the fusion of the L. pneumophila vacuole with lysosomes by modulating actin polymerization through cofilin. However, caspase-11 was dispensable for the fusion of lysosomes with phagosomes containing nonpathogenic bacteria, uncovering a fundamental difference in the trafficking of phagosomes according to their cargo.
Collapse
Affiliation(s)
- Anwari Akhter
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Fine KL, Metcalfe MG, White E, Virji M, Karls RK, Quinn FD. Involvement of the autophagy pathway in trafficking of Mycobacterium tuberculosis bacilli through cultured human type II epithelial cells. Cell Microbiol 2012; 14:1402-14. [PMID: 22519722 DOI: 10.1111/j.1462-5822.2012.01804.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 03/26/2012] [Accepted: 04/15/2012] [Indexed: 11/29/2022]
Abstract
Interactions between Mycobacterium tuberculosis bacilli and alveolar macrophages have been extensively characterized, while similar analyses in epithelial cells have not been performed. In this study, we microscopically examined endosomal trafficking of M. tuberculosis strain Erdman in A549 cells, a human type II pneumocyte cell line. Immuno-electron microscopic (IEM) analyses indicate that M. tuberculosis bacilli are internalized to a compartment labelled first with Rab5 and then with Rab7 small GTPase proteins. This suggests that, unlike macrophages, M. tuberculosis bacilli traffic to late endosomes in epithelial cells. However, fusion of lysosomes with the bacteria-containing compartment appears to be inhibited, as illustrated by IEM studies employing LAMP-2 and cathepsin-L antibodies. Examination by transmission electron microscopy and IEM revealed M. tuberculosis-containing compartments surrounded by double membranes and labelled with antibodies against the autophagy marker Lc3, providing evidence for involvement and intersection of the autophagy and endosomal pathways. Interestingly, inhibition of the autophagy pathway using 3-methyladenine improved host cell viability and decreased numbers of viable intracellular bacteria recovered after 72 h post infection. Collectively, these data suggest that trafficking patterns for M. tuberculosis bacilli in alveolar epithelial cells differ from macrophages, and that autophagy is involved this process.
Collapse
Affiliation(s)
- Kari L Fine
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | |
Collapse
|
33
|
Ignatov D, Kondratieva E, Azhikina T, Apt A. Mycobacterium avium-triggered diseases: pathogenomics. Cell Microbiol 2012; 14:808-18. [DOI: 10.1111/j.1462-5822.2012.01776.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Requirement for invariant chain in macrophages for Mycobacterium tuberculosis replication and CD1d antigen presentation. Infect Immun 2011; 79:3053-63. [PMID: 21576321 DOI: 10.1128/iai.01108-10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mycobacterium tuberculosis is an intracellular bacterium that persists in phagosomes of myeloid cells. M. tuberculosis-encoded factors support pathogen survival and reduce fusion of phagosomes with bactericidal lysosomal compartments. It is, however, not entirely understood if host factors that mediate endosomal fusion affect M. tuberculosis intracellular localization and survival. Neither is it known if endosomal fusion influences induction of host immune reactivity by M. tuberculosis-infected cells. Lysosomal degradation of M. tuberculosis appears to be pivotal for making available lipid substrates for assembly into lipid-CD1d complexes to allow activation of CD1d-restricted invariant natural killer T (iNKT) cells. To clarify the role for endosomal fusion in M. tuberculosis survival and induction of host CD1d-mediated immune defense, we focused our studies on the invariant chain (Ii). Ii regulates endosome docking and fusion and thereby controls endosomal transport. Through direct binding, Ii also directs intracellular transport of the class II major histocompatibility complex and CD1d. Our findings demonstrate that upon infection of Ii-knockout (Ii(-/-)) macrophages, M. tuberculosis is initially retained in early endosomal antigen 1-positive lysosomal-associated membrane protein 1-negative phagosomes, which results in slightly impaired pathogen replication. The absence of Ii did not affect the ability of uninfected and infected macrophages to produce nitric oxide, tumor necrosis factor alpha, or interleukin-12. However, induction of cell surface CD1d was impaired in infected Ii(-/-) macrophages, and CD1d-restricted iNKT cells were unable to suppress bacterial replication when they were cocultured with M. tuberculosis-infected Ii(-/-) macrophages. Thus, while the host factor Ii is not essential for the formation of the M. tuberculosis-containing vacuole, its presence is crucial for iNKT cell recognition of infected macrophages.
Collapse
|
35
|
Fettucciari K, Quotadamo F, Noce R, Palumbo C, Modesti A, Rosati E, Mannucci R, Bartoli A, Marconi P. Group B Streptococcus (GBS) disrupts by calpain activation the actin and microtubule cytoskeleton of macrophages. Cell Microbiol 2011; 13:859-84. [PMID: 21414124 DOI: 10.1111/j.1462-5822.2011.01584.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Group B Streptococcus (GBS) has evolved several strategies to avoid host defences where macrophages are one of main targets. Since pathogens frequently target the cytoskeleton to evade immune defences, we investigated if GBS manipulates macrophage cytoskeleton. GBS-III-COH31 in a time- and infection ratio-dependent manner induces great macrophage cytoskeleton alterations, causing degradation of several structural and regulatory cytoskeletal proteins. GBS β-haemolysin is involved in cytoskeleton alterations causing plasma membrane permeability defects which allow calcium influx and calpain activation. In fact, cytoskeleton alterations are not induced by GBS-III-COH31 in conditions that suppress β-haemolysin expression/activity and in presence of dipalmitoylphosphatidylcholine (β-haemolysin inhibitor). Calpains, particularly m-calpain, are responsible for GBS-III-COH31-induced cytoskeleton disruption. In fact, the calpain inhibitor PD150606, m-calpain small-interfering-RNA and EGTA which inhibit calpain activation prevented cytoskeleton degradation whereas µ-calpain and other protease inhibitors did not. Finally, calpain inhibition strongly increased the number of viable intracellular GBS-III-COH31, showing that cytoskeleton alterations reduced macrophage phagocytosis. Marked macrophage cytoskeleton alterations are also induced by GBS-III-NEM316 and GBS-V-10/84 through β-haemolysin-mediated plasma membrane permeability defects which allow calpain activation. This study suggests a new GBS strategy to evade macrophage antimicrobial responses based on cytoskeleton disruption by an unusual mechanism mediated by calcium influx and calpain activation.
Collapse
Affiliation(s)
- Katia Fettucciari
- General Pathology and Immunology Section, Perugia University, Perugia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Early J, Fischer K, Bermudez LE. Mycobacterium avium uses apoptotic macrophages as tools for spreading. Microb Pathog 2011; 50:132-9. [PMID: 21167273 PMCID: PMC3030681 DOI: 10.1016/j.micpath.2010.12.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 12/04/2010] [Accepted: 12/07/2010] [Indexed: 02/04/2023]
Abstract
BACKGROUND Mycobacterium avium (MAC) lives and replicates in macrophages and causes disseminated disease in immunocompromised individuals. As a host response to control disease, many macrophages become apoptotic a few days after MAC infection. In this study, we hypothesized that MAC can survive autophagic and apoptotic macrophages and spread. METHODS Electron, time-lapse video, fluorescence microscopy. Apoptosis was determined by ELISA and TUNEL assays. Autophagy was seen by migration of LC3-1. RESULTS Apoptotic macrophages harbor chiefly viable MAC. MAC escapes both the vacuole and the macrophage once apoptosis is triggered, leaving the bacteria free to infect nearby macrophages in the process of spreading. In addition, some MAC species will have apoptotic bodies and are released in healthy macrophages following apoptotic body ingestion. Because autophagy precedes apoptosis, it was established that heat-killed MAC, and viable MAC induces autophagy in macrophages at similar rates, but MAC still survives. CONCLUSION MAC spreading from cell-to-cell is triggered by the macrophage's attempt to kill the bacterium, undergoing apoptosis.
Collapse
Affiliation(s)
- Julie Early
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | | | | |
Collapse
|
37
|
Actin dynamics and Rho GTPases regulate the size and formation of parasitophorous vacuoles containing Coxiella burnetii. Infect Immun 2009; 77:4609-20. [PMID: 19635823 DOI: 10.1128/iai.00301-09] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Q fever is a disease caused by Coxiella burnetii. In the host cell, this pathogen generates a large parasitophorous vacuole (PV) with lysosomal characteristics. Here we show that F-actin not only is recruited to but also is involved in the formation of the typical PV. Treatment of infected cells with F-actin-depolymerizing agents alters PV development. The small PVs formed in latrunculin B-treated cells were loaded with transferrin and Lysotracker and labeled with an antibody against cathepsin D, suggesting that latrunculin B did not affect vacuole cargo and its lysosomal characteristics. Nevertheless, the vacuoles were unable to fuse with latex bead phagosomes. It is known that actin dynamics are regulated by the Rho family GTPases. To assess the role of these GTPases in PV formation, infected cells were transfected with pEGFP expressing wild-type and mutant Rac1, Cdc42, and RhoA proteins. Rac1 did not show significant PV association. In contrast, PVs were decorated by both the wild types and constitutively active mutants of Cdc42 and RhoA. This association was inhibited by treatment of infected cells with chloramphenicol, suggesting a role for bacterial protein synthesis in the recruitment of these proteins. Interestingly, a decrease in vacuole size was observed in cells expressing dominant-negative RhoA; however, these small vacuoles accumulated transferrin, Lysotracker, and DQ-BSA. In summary, these results suggest that actin, likely modulated by the GTPases RhoA and Cdc42 and by bacterial proteins, is involved in the formation of the typical PV.
Collapse
|
38
|
Jayachandran R, Gatfield J, Massner J, Albrecht I, Zanolari B, Pieters J. RNA interference in J774 macrophages reveals a role for coronin 1 in mycobacterial trafficking but not in actin-dependent processes. Mol Biol Cell 2007; 19:1241-51. [PMID: 18162581 DOI: 10.1091/mbc.e07-07-0640] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Macrophages are crucial for innate immunity, apoptosis, and tissue remodeling, processes that rely on the capacity of macrophages to internalize and process cargo through phagocytosis. Coronin 1, a member of the WD repeat protein family of coronins specifically expressed in leukocytes, was originally identified as a molecule that is recruited to mycobacterial phagosomes and prevents the delivery of mycobacteria to lysosomes, allowing these to survive within phagosomes. However, a role for coronin 1 in mycobacterial pathogenesis has been disputed in favor for its role in mediating phagocytosis and cell motility. In this study, a role for coronin 1 in actin-mediated cellular processes was addressed using RNA interference in the murine macrophage cell line J774. It is shown that the absence of coronin 1 in J774 macrophages expressing small interfering RNA constructs specific for coronin 1 does not affect phagocytosis, macropinocytosis, cell locomotion, or regulation of NADPH oxidase activity. However, in coronin 1-negative J774 cells, internalized mycobacteria were rapidly transferred to lysosomes and killed. Therefore, these results show that in J774 cells coronin 1 has a specific role in modulating phagosome-lysosome transport upon mycobacterial infection and that it is dispensable for most F-actin-mediated cytoskeletal rearrangements.
Collapse
|
39
|
Davis FP, Barkan DT, Eswar N, McKerrow JH, Sali A. Host pathogen protein interactions predicted by comparative modeling. Protein Sci 2007; 16:2585-96. [PMID: 17965183 DOI: 10.1110/ps.073228407] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Pathogens have evolved numerous strategies to infect their hosts, while hosts have evolved immune responses and other defenses to these foreign challenges. The vast majority of host-pathogen interactions involve protein-protein recognition, yet our current understanding of these interactions is limited. Here, we present and apply a computational whole-genome protocol that generates testable predictions of host-pathogen protein interactions. The protocol first scans the host and pathogen genomes for proteins with similarity to known protein complexes, then assesses these putative interactions, using structure if available, and, finally, filters the remaining interactions using biological context, such as the stage-specific expression of pathogen proteins and tissue expression of host proteins. The technique was applied to 10 pathogens, including species of Mycobacterium, apicomplexa, and kinetoplastida, responsible for "neglected" human diseases. The method was assessed by (1) comparison to a set of known host-pathogen interactions, (2) comparison to gene expression and essentiality data describing host and pathogen genes involved in infection, and (3) analysis of the functional properties of the human proteins predicted to interact with pathogen proteins, demonstrating an enrichment for functionally relevant host-pathogen interactions. We present several specific predictions that warrant experimental follow-up, including interactions from previously characterized mechanisms, such as cytoadhesion and protease inhibition, as well as suspected interactions in hypothesized networks, such as apoptotic pathways. Our computational method provides a means to mine whole-genome data and is complementary to experimental efforts in elucidating networks of host-pathogen protein interactions.
Collapse
Affiliation(s)
- Fred P Davis
- Department of Biopharmaceutical Sciences, University of California at San Francisco, San Francisco, California 94158, USA.
| | | | | | | | | |
Collapse
|
40
|
Appelberg R. Pathogenesis of Mycobacterium avium infection: typical responses to an atypical mycobacterium? Immunol Res 2007; 35:179-90. [PMID: 17172645 DOI: 10.1385/ir:35:3:179] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/02/2023]
Abstract
Studying infections with Mycobacterium avium in mouse models has allowed the dissection of the antimycobacterial pathways of the mammalian host. Whereas the paradigm of cell-mediated immunity to intracellular pathogens has been confirmed, namely with regard to the pivotal roles of CD4+ T cells, macrophages, and the IL12-IFNgamma cytokine axis, atypical features have been uncovered such as the resistance to NO, the involvement of minor players in the induction of type 1 protective immunity (such as TLR2, CD40, and CD30), and the development of immunopathology during the infection with highly virulent strains such as the development of caseous necrosis of granulomas or the progressive emergence of severe lymphopenia.
Collapse
Affiliation(s)
- Rui Appelberg
- Laboratory of Microbiology and Immunology of Infection, IBMC-Institute for Molecular and Cell Biology and ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, University of Porto, Portugal.
| |
Collapse
|
41
|
Robinson N, Wolke M, Ernestus K, Plum G. A mycobacterial gene involved in synthesis of an outer cell envelope lipid is a key factor in prevention of phagosome maturation. Infect Immun 2006; 75:581-91. [PMID: 17088345 PMCID: PMC1828500 DOI: 10.1128/iai.00997-06] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Virulent mycobacteria cause arrest of phagosome maturation as a part of their survival strategy in hosts. This process is mediated through multiple virulence factors, whose molecular nature remains elusive. Using Mycobacterium marinum as a model, we performed a genome-wide screen to identify mutants whose ability to inhibit phagosome maturation was impaired, and we succeeded in isolating a comprehensive set of mutants that were not able to occupy an early endosome-like phagosomal compartment in mammalian macrophages. Categorizing and ordering the multiple mutations according to their gene families demonstrated that the genes modulating the cell envelope are the principal factors in arresting phagosome maturation. In particular, we identified a novel gene, pmiA, which is capable of influencing the constitution of the cell envelope lipids, thereby leading to the phagosome maturation block. The pmiA mutant was not able to resist phagosome maturation and was severely attenuated in mice. Complementing the mutant with the wild-type gene restored the attenuated virulence to wild-type levels in mice.
Collapse
MESH Headings
- Animals
- Bacterial Proteins/genetics
- Bacterial Proteins/physiology
- Chromatography, Thin Layer
- DNA, Bacterial/chemistry
- DNA, Bacterial/genetics
- Disease Models, Animal
- Gene Deletion
- Genes, Bacterial
- Genetic Complementation Test
- Histocytochemistry
- Immunohistochemistry
- Liver/microbiology
- Liver/pathology
- Macrophages/microbiology
- Membrane Lipids/analysis
- Membrane Lipids/isolation & purification
- Mice
- Mice, Inbred C57BL
- Microscopy, Fluorescence
- Molecular Sequence Data
- Mutagenesis, Insertional
- Mycobacterium Infections, Nontuberculous/microbiology
- Mycobacterium marinum/genetics
- Mycobacterium marinum/immunology
- Mycobacterium marinum/pathogenicity
- Phagosomes/chemistry
- Phagosomes/microbiology
- Phagosomes/physiology
- Sequence Analysis, DNA
- Spleen/microbiology
- Spleen/pathology
- Virulence Factors/genetics
- Virulence Factors/physiology
Collapse
Affiliation(s)
- Nirmal Robinson
- Institute for Medical Microbiology, University of Cologne, 50935 Cologne, Germany
| | | | | | | |
Collapse
|
42
|
Nepal RM, Mampe S, Shaffer B, Erickson AH, Bryant P. Cathepsin L maturation and activity is impaired in macrophages harboring M. avium and M. tuberculosis. Int Immunol 2006; 18:931-9. [PMID: 16636015 DOI: 10.1093/intimm/dxl029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium tuberculosis-infected macrophages demonstrate diminished capacity to present antigens via class II MHC molecules. Since successful class II MHC-restricted antigen presentation relies on the actions of endocytic proteases, we asked whether the activities of cathepsins (Cat) B, S and L-three major lysosomal cysteine proteases-are modulated in macrophages infected with pathogenic Mycobacterium spp. Infection of murine bone marrow-derived macrophages with either Mycobacterium avium or M. tuberculosis had no obvious effect on Cat B or Cat S activity. In contrast, the activity of Cat L was altered in infected cells. Specifically, whereas the 24-kDa two-chain mature form of active Cat L predominated in uninfected cells, we observed an increase in the steady-state activity of the precursor single-chain (30 kDa) and 25-kDa two-chain forms of the enzyme in cells infected with either M. avium or M. tuberculosis. Pulse-chase analyses revealed that maturation of nascent, single-chain Cat L into the 25-kDa two-chain form was impaired in infected macrophages, and that maturation into the 24-kDa two-chain form did not occur. Consistent with these data, M. avium infection inhibited the IFNgamma-induced secretion of active two-chain Cat L by macrophages. Viable bacilli were not required to disrupt Cat L maturation, suggesting that a constitutively expressed mycobacterial component was responsible. The absence of the major active form of lysosomal Cat L in M. avium- and M. tuberculosis-infected macrophages may influence the types of T cell epitopes generated in these antigen-presenting cells, and/or the rate of class II MHC peptide loading.
Collapse
Affiliation(s)
- Rajeev M Nepal
- Department of Microbiology, Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
43
|
Hestvik ALK, Hmama Z, Av-Gay Y. Mycobacterial manipulation of the host cell. FEMS Microbiol Rev 2005; 29:1041-50. [PMID: 16040149 DOI: 10.1016/j.femsre.2005.04.013] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Revised: 03/12/2005] [Accepted: 04/18/2005] [Indexed: 11/20/2022] Open
Abstract
Phagosome biogenesis, the process by which macrophages neutralize ingested pathogens and initiate antigen presentation, has entered the field of cellular mycobacteriology research largely owing to the discovery 30 years ago that phagosomes harboring mycobacteria are refractory to fusion with lysosomes. In the past decade, the use of molecular genetics and biology in different model systems to study phagosome biogenesis have made significant advances in understanding subtle mechanisms by which mycobacteria inhibit the maturation of its phagosome. Thus, we are beginning to appreciate the extent to which these pathogens are able to interfere with innate immune responses and manipulate defense mechanisms to enhance their survival within the human host cell. Here, we summarize current knowledge about phagosome maturation arrest in infected macrophages and the subsequent attenuation of the macrophage-initiated adaptive anti-mycobacterial immune defenses.
Collapse
Affiliation(s)
- Anne Lise K Hestvik
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia, 2733 Heather St. Vancouver, BC, Canada V5Z 3J5
| | | | | |
Collapse
|
44
|
Castandet J, Prost JF, Peyron P, Astarie-Dequeker C, Anes E, Cozzone AJ, Griffiths G, Maridonneau-Parini I. Tyrosine phosphatase MptpA of Mycobacterium tuberculosis inhibits phagocytosis and increases actin polymerization in macrophages. Res Microbiol 2005; 156:1005-13. [PMID: 16085396 DOI: 10.1016/j.resmic.2005.05.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2005] [Revised: 05/12/2005] [Accepted: 05/18/2005] [Indexed: 11/23/2022]
Abstract
Protein tyrosine phosphatases from several microorganisms have been shown to play a role as virulence factors by modifying the phosphorylation/dephosphorylation equilibrium in cells of their host. Two tyrosine phosphatases, MptpA and MptpB, secreted by Mycobacterium tuberculosis, have been identified. Expression of MptpA is upregulated upon infection of monocytes, but its role in host cells has not been elucidated. A eukaryotic expression vector containing the mptpA cDNA has been transfected into macrophages. We report that MptpA reduced phagocytosis of mycobacteria, opsonized zymosan or zymosan, but had no effect on phagocytosis of IgG-coated particles. We also noted that the presence of F-actin at the surface of phagosomes containing opsonized zymosan was significantly increased in cells expressing MptpA. In the presence of recombinant MptpA, the process of actin polymerization at the surface of isolated phagosomes was increased; this was not the case in the presence of the phosphatase-dead mutant MptpA(C11S). MptpA had no effect when IgG-coated particles were present inside isolated phagosomes. These results indicate that, like other tyrosine phosphatases of pathogens, MptpA plays a role in phagocytosis and actin polymerization. However, MptpA had no effect on IgG particles, suggesting that its putative substrate(s) is not linked to the signaling pathways of Fcgamma receptors.
Collapse
Affiliation(s)
- Jérôme Castandet
- Institut de Pharmacologie et de Biologie Structurale, UMR CNRS 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Melo TG, Almeida DS, de Meirelles MDNSL, Pereira MC. Trypanosoma cruzi infection disrupts vinculin costameres in cardiomyocytes. Eur J Cell Biol 2005; 83:531-40. [PMID: 15679099 DOI: 10.1078/0171-9335-00419] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chagas' disease cardiomyopathy is an important manifestation of Trypanosoma cruzi infection, leading to cardiac dysfunction and serious arrhythmias. We have here investigated by indirect immunofluorescence assay the distribution of vinculin, a focal adhesion protein with a major role in the transmission of contraction force, during the T. cruzi-cardiomyocyte infection in vitro and in vivo. No change in vinculin distribution was observed after 24 h of infection, where control and T. cruzi-infected cardiomyocytes displayed vinculin localized at costameres and intercalated discs. On the other hand, a clear disruption of vinculin costameric distribution was noted after 72 h of infection. A significant reduction in the levels of vinculin expression was observed at all times of infection. In murine experimental Chagas' disease, alteration in the vinculin distribution was also detected in the infected myocardium, with no costameric staining in infected myocytes and irregular alignment of intercalated discs in cardiac fibers. These data suggest that the disruption of costameric vinculin distribution and the enlargement of interstitial space due to inflammatory infiltration may contribute to the reduction of transmission of cardiac contraction force, leading to alterations in the heart function in Chagas' disease.
Collapse
Affiliation(s)
- Tatiana G Melo
- Departamento de Ultra-estrutura e Biologia Celular, Laboratório de Ultra-estrutura Celular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
46
|
Abstract
Phagocytosis and phagolysosome biogenesis represent fundamental biological processes essential for proper tissue homeostasis, development, elimination of invading microorganisms, and antigen processing and presentation. Phagosome formation triggers a preprogrammed pathway of maturation into the phagolysosome, a process controlled by Ca2+ and the regulators of organellar trafficking centered around the small GTP-binding proteins Rabs and their downstream effectors, including lipid kinases, organellar tethering molecules, and membrane fusion apparatus. Mycobacterium tuberculosis is a potent human pathogen parasitizing macrophages. It interferes with the Rab-controlled membrane trafficking and arrests the maturing phagosome at a stage where no harm can be done to the pathogen while the delivery of nutrients and membrane to the vacuole harboring the microorganism continues. This process, referred to as the M. tuberculosis phagosome maturation arrest or inhibition of phagosome-lysosome fusion, is critical for M. tuberculosis persistence in human populations. It also provides a general model system for dissecting the phagolysosome biogenesis pathways. Here we review the fundamental trafficking processes targeted by M. tuberculosis and the mycobacterial products that interfere with phagosomal maturation.
Collapse
Affiliation(s)
- Isabelle Vergne
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-001, USA.
| | | | | | | |
Collapse
|
47
|
Anes E, Kühnel MP, Bos E, Moniz-Pereira J, Habermann A, Griffiths G. Selected lipids activate phagosome actin assembly and maturation resulting in killing of pathogenic mycobacteria. Nat Cell Biol 2003; 5:793-802. [PMID: 12942085 DOI: 10.1038/ncb1036] [Citation(s) in RCA: 202] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2003] [Accepted: 07/30/2003] [Indexed: 01/02/2023]
Abstract
Pathogenic mycobacteria such as Mycobacterium tuberculosis and Mycobacterium avium facilitate disease by surviving intracellularly within a potentially hostile environment: the macrophage phagosome. They inhibit phagosome maturation processes, including fusion with lysosomes, acidification and, as shown here, membrane actin assembly. An in vitro assay developed for latex bead phagosomes (LBPs) provided insights into membrane signalling events that regulate phagosome actin assembly, a process linked to membrane fusion. Different lipids were found to stimulate or inhibit actin assembly by LBPs and mycobacterial phagosomes in vitro. In addition, selected lipids activated actin assembly and phagosome maturation in infected macrophages, resulting in a significant killing of M. tuberculosis and M. avium. In contrast, the polyunsaturated sigma-3 lipids behaved differently and stimulated pathogen growth. Thus, lipids can be involved in both stimulatory and inhibitory signalling networks in the phagosomal membrane.
Collapse
Affiliation(s)
- Elsa Anes
- Molecular Pathogenesis Centre, Faculty of Pharmacy, University of Lisbon, Av. Forcas Armadas, 1600-085 Lisbon, Portugal
| | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Velasco-Velázquez MA, Barrera D, González-Arenas A, Rosales C, Agramonte-Hevia J. Macrophage--Mycobacterium tuberculosis interactions: role of complement receptor 3. Microb Pathog 2003; 35:125-131. [PMID: 12927520 DOI: 10.1016/s0882-4010(03)00099-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tuberculosis is the leading infectious disease in the world. Mycobacterium tuberculosis, the causal agent of this disease, invades macrophages and can replicate inside them. Because invasion of macrophages is a critical step for establishing a mycobacterial infection, there is much interest in understanding the mechanisms for M. tuberculosis entry into macrophages. Complement receptor 3 (CR3) is a heterodimeric surface receptor with multiple binding sites, which can mediate complement-opsonized as well as nonopsonic entrance of M. tuberculosis into macrophages. Here, we describe and discuss the role of CR3 in macrophage[bond]M. tuberculosis interactions. The actual information suggests that CR3 mediates a substantial amount of M. tuberculosis binding to macrophages, but CR3 is not related to the mechanisms that allow mycobacteria to survive and replicate intracellularly. Understanding the mechanisms of macrophage[bond]M. tuberculosis interaction will help developing more effective methods to prevent and treat tuberculosis in the future.
Collapse
|
50
|
Abstract
Phagocytosis is the process that cells have evolved to internalise large particles such as mineral debris, which they store, or apoptotic cells and pathogens, which they have the capacity to kill and degrade. However, several important pathogens can suppress these killing functions and survive and multiply within phagosomes, causing disease. Recent advances in phagosome biology have been made possible largely by a model system that uses inert latex beads. The ability to purify latex bead-containing phagosomes has opened the door to allow comprehensive biochemical analyses and functional assays to study the molecular mechanisms governing phagosome function. These approaches have led to unique insights directly relevant for the understanding of the biology of intracellular pathogens and the ways by which they subvert their hosts.
Collapse
Affiliation(s)
- Michel Desjardins
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, CP 6128, Succ. centre ville, H3C 3J7, Montreal, Canada.
| | | |
Collapse
|