1
|
Liao F, Zhou D, Cano M, Liu Z, Scozzi D, Tague LK, Byers DE, Li W, Sivapackiam J, Sharma V, Krupnick AS, Frank DW, Kreisel D, Kulkarni HS, Hachem RR, Gelman AE. Pseudomonas aeruginosa infection induces intragraft lymphocytotoxicity that triggers lung transplant antibody-mediated rejection. Sci Transl Med 2025; 17:eadp1349. [PMID: 39908350 DOI: 10.1126/scitranslmed.adp1349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
How pathogens inhibit transplant tolerance remains unclear. Here, we found that Pseudomonas aeruginosa infection, but not other common bacterial respiratory infections, increases antibody-mediated rejection (AMR) risk in recipients of lung transplants. To explore this relationship, we performed orthotopic lung transplants in mice, infected recipients with P. aeruginosa, and observed for the development of AMR. Intravital two-photon microscopy showed that P. aeruginosa rapidly invaded bronchial-associated lymphoid tissues, which resulted in acute lymphocytotoxicity, including the death of forkhead box P3 (Foxp3)+CD4+ T cells that are required to suppress AMR. P. aeruginosa-mediated AMR required expression of the type III secretion system (T3SS), which injects exotoxins into the cell cytoplasm. Through a combination of mutagenesis and epitope tagging experiments, we revealed that T3SS exotoxin T ADP ribosyl-transferase activity was sufficient for graft-resident Foxp3+CD4+ T cell apoptosis, leading to myeloid differentiation primary response 88 (Myd88)-dependent generation of T-box expressed in T cells (T-bet)- and C-X-C motif chemokine receptor 3 (CXCR3)-positive germinal center and memory B cells with high donor antigen avidity. We also found that T-bet+ and CXCR3+ B cells were elevated in biopsies from recipients of lung transplants who were diagnosed with AMR. In mice, CXCR3 deficiency restricted to B cells or CXCR3 blockade prevented AMR despite P. aeruginosa infection. Our work has identified a previously unrecognized role of bacterial virulence in lung allograft rejection and suggests potential strategies to prevent AMR for those at high risk of P. aeruginosa infection after transplant.
Collapse
Affiliation(s)
- Fuyi Liao
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dequan Zhou
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marlene Cano
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhiyi Liu
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Davide Scozzi
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laneshia K Tague
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Derek E Byers
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wenjun Li
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jothilingam Sivapackiam
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vijay Sharma
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander S Krupnick
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dara W Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel Kreisel
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hrishikesh S Kulkarni
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramsey R Hachem
- Department of Internal Medicine, Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT 84108, USA
| | - Andrew E Gelman
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Samiminemati A, Aprile D, Siniscalco D, Di Bernardo G. Methods to Investigate the Secretome of Senescent Cells. Methods Protoc 2024; 7:52. [PMID: 39051266 PMCID: PMC11270363 DOI: 10.3390/mps7040052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/27/2024] Open
Abstract
The word "secretome" was first used to describe the proteins that cells secrete under different circumstances; however, recent studies have proven the existence of other molecules such as RNA and chemical compounds in the secretome. The study of secretome has significance for the diagnosis and treatment of disease as it provides insight into cellular functions, including immune responses, development, and homeostasis. By halting cell division, cellular senescence plays a role in both cancer defense and aging by secreting substances known as senescence-associated secretory phenotypes (SASP). A variety of techniques could be used to analyze the secretome: protein-based approaches like mass spectrometry and protein microarrays, nucleic acid-based methods like RNA sequencing, microarrays, and in silico prediction. Each method offers unique advantages and limitations in characterizing secreted molecules. Top-down and bottom-up strategies for thorough secretome analysis are became possible by mass spectrometry. Understanding cellular function, disease causes, and proper treatment targets is aided by these methodologies. Their approaches, benefits, and drawbacks will all be discussed in this review.
Collapse
Affiliation(s)
- Afshin Samiminemati
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
| | - Domenico Aprile
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
| | - Dario Siniscalco
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
- Sbarro Health Research Organization, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
3
|
Gonzalez LE, Snyder DT, Casey H, Hu Y, Wang DM, Guetzloff M, Huckaby N, Dziekonski ET, Wells JM, Cooks RG. Machine-Learning Classification of Bacteria Using Two-Dimensional Tandem Mass Spectrometry. Anal Chem 2023; 95:17082-17088. [PMID: 37937965 DOI: 10.1021/acs.analchem.3c04016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Biothreat detection has continued to gain attention. Samples suspected to fall into any of the CDC's biothreat categories require identification by processes that require specialized expertise and facilities. Recent developments in analytical instrumentation and machine learning algorithms offer rapid and accurate classification of Gram-positive and Gram-negative bacterial species. This is achieved by analyzing the negative ions generated from bacterial cell extracts with a modified linear quadrupole ion-trap mass spectrometer fitted with two-dimensional tandem mass spectrometry capabilities (2D MS/MS). The 2D MS/MS data domain of a bacterial cell extract is recorded within five s using a five-scan average after sample preparation by a simple extraction. Bacteria were classified at the species level by their lipid profiles using the random forest, k-nearest neighbor, and multilayer perceptron machine learning models. 2D MS/MS data can also be treated as image data for use with image recognition algorithms such as convolutional neural networks. The classification accuracy of all models tested was greater than 99%. Adding to previously published work on the 2D MS/MS analysis of bacterial growth and the profiling of sporulating bacteria, this study demonstrates the utility and information-rich nature of 2D MS/MS in the identification of bacterial pathogens at the species level when coupled with machine learning.
Collapse
Affiliation(s)
- L Edwin Gonzalez
- Department of Chemistry, Purdue University, West Lafayette , Indiana 47907, United States
| | - Dalton T Snyder
- Teledyne FLIR Detection, West Lafayette, Indiana 47907, United States
| | - Harman Casey
- Teledyne FLIR Detection, West Lafayette, Indiana 47907, United States
| | - Yanyang Hu
- Department of Chemistry, Purdue University, West Lafayette , Indiana 47907, United States
| | - Donna M Wang
- Department of Chemistry, Purdue University, West Lafayette , Indiana 47907, United States
| | - Megan Guetzloff
- Teledyne FLIR Detection, West Lafayette, Indiana 47907, United States
| | - Nicole Huckaby
- Teledyne FLIR Detection, West Lafayette, Indiana 47907, United States
| | - Eric T Dziekonski
- Department of Chemistry, Purdue University, West Lafayette , Indiana 47907, United States
| | - J Mitchell Wells
- Teledyne FLIR Detection, West Lafayette, Indiana 47907, United States
| | - R Graham Cooks
- Department of Chemistry, Purdue University, West Lafayette , Indiana 47907, United States
| |
Collapse
|
4
|
Collars OA, Jones BS, Hu DD, Weaver SD, Sherman TA, Champion MM, Champion PA. An N-acetyltransferase required for ESAT-6 N-terminal acetylation and virulence in Mycobacterium marinum. mBio 2023; 14:e0098723. [PMID: 37772840 PMCID: PMC10653941 DOI: 10.1128/mbio.00987-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
IMPORTANCE N-terminal acetylation is a protein modification that broadly impacts basic cellular function and disease in higher organisms. Although bacterial proteins are N-terminally acetylated, little is understood how N-terminal acetylation impacts bacterial physiology and pathogenesis. Mycobacterial pathogens cause acute and chronic disease in humans and in animals. Approximately 15% of mycobacterial proteins are N-terminally acetylated, but the responsible enzymes are largely unknown. We identified a conserved mycobacterial protein required for the N-terminal acetylation of 23 mycobacterial proteins including the EsxA virulence factor. Loss of this enzyme from M. marinum reduced macrophage killing and spread of M. marinum to new host cells. Defining the acetyltransferases responsible for the N-terminal protein acetylation of essential virulence factors could lead to new targets for therapeutics against mycobacteria.
Collapse
Affiliation(s)
- Owen A. Collars
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Note Dame, Notre Dame, Indiana, USA
| | - Bradley S. Jones
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Note Dame, Notre Dame, Indiana, USA
| | - Daniel D. Hu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Simon D. Weaver
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Taylor A. Sherman
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Matthew M. Champion
- Eck Institute for Global Health, University of Note Dame, Notre Dame, Indiana, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Note Dame, Notre Dame, Indiana, USA
| |
Collapse
|
5
|
Collars OA, Jones BS, Hu DD, Weaver SD, Champion MM, Champion PA. An N-acetyltransferase required for EsxA N-terminal protein acetylation and virulence in Mycobacterium marinum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532585. [PMID: 36993388 PMCID: PMC10055061 DOI: 10.1101/2023.03.14.532585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
N-terminal protein acetylation is a ubiquitous post-translational modification that broadly impacts diverse cellular processes in higher organisms. Bacterial proteins are also N-terminally acetylated, but the mechanisms and consequences of this modification in bacteria are poorly understood. We previously quantified widespread N-terminal protein acetylation in pathogenic mycobacteria (C. R. Thompson, M. M. Champion, and P.A. Champion, J Proteome Res 17(9): 3246-3258, 2018, https:// doi: 10.1021/acs.jproteome.8b00373). The major virulence factor EsxA (ESAT-6, Early secreted antigen, 6kDa) was one of the first N-terminally acetylated proteins identified in bacteria. EsxA is conserved in mycobacterial pathogens, including Mycobacterium tuberculosis and Mycobacterium marinum, a non-tubercular mycobacterial species that causes tuberculosis-like disease in ectotherms. However, enzyme responsible for EsxA N-terminal acetylation has been elusive. Here, we used genetics, molecular biology, and mass-spectroscopy based proteomics to demonstrate that MMAR_1839 (renamed Emp1, ESX-1 modifying protein, 1) is the putative N-acetyl transferase (NAT) solely responsible for EsxA acetylation in Mycobacterium marinum. We demonstrated that ERD_3144, the orthologous gene in M. tuberculosis Erdman, is functionally equivalent to Emp1. We identified at least 22 additional proteins that require Emp1 for acetylation, demonstrating that this putative NAT is not dedicated to EsxA. Finally, we showed that loss of emp1 resulted in a significant reduction in the ability of M. marinum to cause macrophage cytolysis. Collectively, this study identified a NAT required for N-terminal acetylation in Mycobacterium and provided insight into the requirement of N-terminal acetylation of EsxA and other proteins in mycobacterial virulence in the macrophage.
Collapse
Affiliation(s)
- Owen A. Collars
- Department of Biological Sciences, University of Notre Dame, Notre Dame, USA
- Eck Institute for Global Health, University of Note Dame, Notre Dame, USA
| | - Bradley S. Jones
- Department of Biological Sciences, University of Notre Dame, Notre Dame, USA
- Eck Institute for Global Health, University of Note Dame, Notre Dame, USA
| | - Daniel D. Hu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, USA
| | - Simon D. Weaver
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, USA
| | - Matthew M. Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, USA
- Eck Institute for Global Health, University of Note Dame, Notre Dame, USA
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, USA
- Eck Institute for Global Health, University of Note Dame, Notre Dame, USA
| |
Collapse
|
6
|
Sterling FR, D'Amico J, Brumfield AM, Huegel KL, Vaughan PS, Morris K, Schwarz S, Joyce MV, Boggess B, Champion MM, Maciuba K, Allen P, Marasco E, Koch G, Gonzalez P, Hodges S, Leahy S, Gerstbauer E, Hinchcliffe EH, Vaughan KT. StARD9 is a novel lysosomal kinesin required for membrane tubulation, cholesterol transport and Purkinje cell survival. J Cell Sci 2023; 136:292582. [PMID: 36861884 DOI: 10.1242/jcs.260662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 01/18/2023] [Indexed: 03/03/2023] Open
Abstract
The pathological accumulation of cholesterol is a signature feature of Niemann-Pick type C (NPC) disease, in which excessive lipid levels induce Purkinje cell death in the cerebellum. NPC1 encodes a lysosomal cholesterol-binding protein, and mutations in NPC1 drive cholesterol accumulation in late endosomes and lysosomes (LE/Ls). However, the fundamental role of NPC proteins in LE/L cholesterol transport remains unclear. Here, we demonstrate that NPC1 mutations impair the projection of cholesterol-containing membrane tubules from the surface of LE/Ls. A proteomic survey of purified LE/Ls identified StARD9 as a novel lysosomal kinesin responsible for LE/L tubulation. StARD9 contains an N-terminal kinesin domain, a C-terminal StART domain, and a dileucine signal shared with other lysosome-associated membrane proteins. Depletion of StARD9 disrupts LE/L tubulation, paralyzes bidirectional LE/L motility and induces accumulation of cholesterol in LE/Ls. Finally, a novel StARD9 knock-out mouse recapitulates the progressive loss of Purkinje cells in the cerebellum. Together, these studies identify StARD9 as a microtubule motor protein responsible for LE/L tubulation and provide support for a novel model of LE/L cholesterol transport that becomes impaired in NPC disease.
Collapse
Affiliation(s)
- Felicity R Sterling
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jon D'Amico
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Kara L Huegel
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Patricia S Vaughan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Kathryn Morris
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shelby Schwarz
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michelle V Joyce
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.,University of Notre Dame Proteomics and Mass Spectrometry Facility, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Bill Boggess
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.,University of Notre Dame Proteomics and Mass Spectrometry Facility, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Matthew M Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.,University of Notre Dame Proteomics and Mass Spectrometry Facility, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Kevin Maciuba
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Philip Allen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Eric Marasco
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Grant Koch
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Peter Gonzalez
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shannon Hodges
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shannon Leahy
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Erica Gerstbauer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Kevin T Vaughan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.,Notre Dame Integrated Imaging Facility, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
7
|
Cronin RM, Ferrell MJ, Cahir CW, Champion MM, Champion PA. Proteo-genetic analysis reveals clear hierarchy of ESX-1 secretion in Mycobacterium marinum. Proc Natl Acad Sci U S A 2022; 119:e2123100119. [PMID: 35671426 PMCID: PMC9214503 DOI: 10.1073/pnas.2123100119] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
The ESX-1 (ESAT-6-system-1) system and the protein substrates it transports are essential for mycobacterial pathogenesis. The precise ways that ESX-1 substrates contribute to virulence remains unknown. Several known ESX-1 substrates are also required for the secretion of other proteins. We used a proteo-genetic approach to construct high-resolution dependency relationships for the roles of individual ESX-1 substrates in secretion and virulence in Mycobacterium marinum, a pathogen of humans and animals. Characterizing a collection of M. marinum strains with in-frame deletions in each of the known ESX-1 substrate genes and the corresponding complementation strains, we demonstrate that ESX-1 substrates are differentially required for ESX-1 activity and for virulence. Using isobaric-tagged proteomics, we quantified the degree of requirement of each substrate on protein secretion. We conclusively defined distinct contributions of ESX-1 substrates in protein secretion. Our data reveal a hierarchy of ESX-1 substrate secretion, which supports a model for the composition of the extracytoplasmic ESX-1 secretory machinery. Overall, our proteo-genetic analysis demonstrates discrete roles for ESX-1 substrates in ESX-1 function and secretion in M. marinum.
Collapse
Affiliation(s)
- Rachel M. Cronin
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| | - Micah J. Ferrell
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| | - Clare W. Cahir
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| | - Matthew M. Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| |
Collapse
|
8
|
Smith C, Canestrari JG, Wang AJ, Champion MM, Derbyshire KM, Gray TA, Wade JT. Pervasive translation in Mycobacterium tuberculosis. eLife 2022; 11:e73980. [PMID: 35343439 PMCID: PMC9094748 DOI: 10.7554/elife.73980] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Most bacterial ORFs are identified by automated prediction algorithms. However, these algorithms often fail to identify ORFs lacking canonical features such as a length of >50 codons or the presence of an upstream Shine-Dalgarno sequence. Here, we use ribosome profiling approaches to identify actively translated ORFs in Mycobacterium tuberculosis. Most of the ORFs we identify have not been previously described, indicating that the M. tuberculosis transcriptome is pervasively translated. The newly described ORFs are predominantly short, with many encoding proteins of ≤50 amino acids. Codon usage of the newly discovered ORFs suggests that most have not been subject to purifying selection, and hence are unlikely to contribute to cell fitness. Nevertheless, we identify 90 new ORFs (median length of 52 codons) that bear the hallmarks of purifying selection. Thus, our data suggest that pervasive translation of short ORFs in Mycobacterium tuberculosis serves as a rich source for the evolution of new functional proteins.
Collapse
Affiliation(s)
- Carol Smith
- Wadsworth Center, Division of Genetics, New York State Department of HealthAlbanyUnited States
| | - Jill G Canestrari
- Wadsworth Center, Division of Genetics, New York State Department of HealthAlbanyUnited States
| | - Archer J Wang
- Wadsworth Center, Division of Genetics, New York State Department of HealthAlbanyUnited States
| | - Matthew M Champion
- Department of Chemistry and Biochemistry, University of Notre DameNotre DameUnited States
| | - Keith M Derbyshire
- Wadsworth Center, Division of Genetics, New York State Department of HealthAlbanyUnited States
- Department of Biomedical Sciences, School of Public Health, University at AlbanyNew YorkUnited States
| | - Todd A Gray
- Wadsworth Center, Division of Genetics, New York State Department of HealthAlbanyUnited States
- Department of Biomedical Sciences, School of Public Health, University at AlbanyNew YorkUnited States
| | - Joseph T Wade
- Wadsworth Center, Division of Genetics, New York State Department of HealthAlbanyUnited States
- Department of Biomedical Sciences, School of Public Health, University at AlbanyNew YorkUnited States
| |
Collapse
|
9
|
Bao Y, Wang L, Sun J. Post-translational knockdown and post-secretional modification of EsxA determine contribution of EsxA membrane permeabilizing activity for mycobacterial intracellular survival. Virulence 2021; 12:312-328. [PMID: 33356823 PMCID: PMC7808419 DOI: 10.1080/21505594.2020.1867438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/10/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
Current genetic studies (e.g. gene knockout) have suggested that EsxA and EsxB function as secreted virulence factors that are essential for Mycobaterium tuberculosis (Mtb) intracellular survival, specifically in mediating phagosome rupture and translocation of Mtb to the cytosol of host cells, which further facilitates Mtb intracellular replicating and cell-to-cell spreading. The EsxA-mediated intracellular survival is presumably achieved by its pH-dependent membrane-permeabilizing activity (MPA). However, the data from other studies have generated a discrepancy regarding the role of EsxA MPA in mycobacterial intracellular survival, which has raised a concern that genetic manipulations, such as deletion of esxB-esxA operon or RD-1 locus, may affect other codependently secreted factors that could be also directly involved cytosolic translocation, or stimulate extended disturbance on other genes' expression. To avoid the drawbacks of gene knockout, we first engineered a Mycobacterium marinum (Mm) strain, in which a DAS4+ tag was fused to the C-terminus of EsxB to allow inducible knockdown of EsxB (also EsxA) at the post-translational level. We also engineered an Mm strain by fusing a SpyTag (ST) to the C-terminus of EsxA, which allowed inhibition of EsxA-ST MPA at the post-secretional level through a covalent linkage to SpyCatcher-GFP. Both post-translational knockdown and functional inhibition of EsxA resulted in attenuation of Mm intracellular survival in lung epithelial cells or macrophages, which unambiguously confirms the direct role of EsxA MPA in mycobacterial intracellular survival.
Collapse
Affiliation(s)
- Yanqing Bao
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, USA
| | - Lin Wang
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, USA
| | - Jianjun Sun
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, USA
| |
Collapse
|
10
|
Nicholson KR, Mousseau CB, Champion MM, Champion PA. The genetic proteome: Using genetics to inform the proteome of mycobacterial pathogens. PLoS Pathog 2021; 17:e1009124. [PMID: 33411813 PMCID: PMC7790235 DOI: 10.1371/journal.ppat.1009124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mycobacterial pathogens pose a sustained threat to human health. There is a critical need for new diagnostics, therapeutics, and vaccines targeting both tuberculous and nontuberculous mycobacterial species. Understanding the basic mechanisms used by diverse mycobacterial species to cause disease will facilitate efforts to design new approaches toward detection, treatment, and prevention of mycobacterial disease. Molecular, genetic, and biochemical approaches have been widely employed to define fundamental aspects of mycobacterial physiology and virulence. The recent expansion of genetic tools in mycobacteria has further increased the accessibility of forward genetic approaches. Proteomics has also emerged as a powerful approach to further our understanding of diverse mycobacterial species. Detection of large numbers of proteins and their modifications from complex mixtures of mycobacterial proteins is now routine, with efforts of quantification of these datasets becoming more robust. In this review, we discuss the “genetic proteome,” how the power of genetics, molecular biology, and biochemistry informs and amplifies the quality of subsequent analytical approaches and maximizes the potential of hypothesis-driven mycobacterial research. Published proteomics datasets can be used for hypothesis generation and effective post hoc supplementation to experimental data. Overall, we highlight how the integration of proteomics, genetic, molecular, and biochemical approaches can be employed successfully to define fundamental aspects of mycobacterial pathobiology.
Collapse
Affiliation(s)
- Kathleen R. Nicholson
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - C. Bruce Mousseau
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Matthew M. Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
- Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame Indiana, United States of America
- * E-mail: (MMC); (PAC)
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame Indiana, United States of America
- * E-mail: (MMC); (PAC)
| |
Collapse
|
11
|
Guo Q, Bi J, Wang H, Zhang X. Mycobacterium tuberculosis ESX-1-secreted substrate protein EspC promotes mycobacterial survival through endoplasmic reticulum stress-mediated apoptosis. Emerg Microbes Infect 2020; 10:19-36. [PMID: 33290182 PMCID: PMC7832037 DOI: 10.1080/22221751.2020.1861913] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
EsxA, secreted by the ESAT-6 secretion system 1 (ESX-1) secretion system, is considered the major Mycobacterium tuberculosis (Mtb) virulence determinant. However, the roles of the individual ESX-1 substrates, such as EspC, remain unclear due to their interdependency for secretion with EsxA. Here, we validated that EspC triggered ER stress-mediated apoptosis in macrophages. The EspC-mediated ER stress was involved in pro-inflammatory cytokines generation, intracellular Ca2+ release, and reactive oxygen species accumulation. Mitochondrial transmembrane potential dissipation and mitochondrial outer membrane permeabilization occurred in EspC-treated macrophages, causing apoptosis. Furthermore, ER stress-mediated apoptosis was effectively induced in EspC-overexpressing Mycobacterium smegmatis-infected macrophages and mice. EspC overexpression caused a significant increase in bacterial survival in the macrophages, spleens, and lungs, and accelerated mouse death was observed. Moreover, the increased viability of bacteria in the macrophages was significantly reduced by pretreatment with the apoptosis inhibitor. Overall, our results revealed that EspC is an essential ESX-1 protein for Mtb–host interactions and EspC-induced ER stress-mediated apoptosis may be employed by Mtb to establish and spread infection. Given the critical roles of the ESX systems in Mtb pathogenesis and immunity, our findings offer new perspectives on the complex host-pathogen interactions and mechanisms underlying ESX-1-mediated pathogenesis.
Collapse
Affiliation(s)
- Qinglong Guo
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China.,National Clinical Research Center for Infectious Disease (Tuberculosis), Shenzhen Third People's Hospital, South University of Science and Technology of China, Shenzhen, People's Republic of China
| | - Jing Bi
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China
| | - Xuelian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China.,Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Review on matrix-assisted laser desorption/ionization time-of-flight mass spectrometry for the rapid screening of microbial species: A promising bioanalytical tool. Microchem J 2020. [DOI: 10.1016/j.microc.2020.105387] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Modeling Tubercular ESX-1 Secretion Using Mycobacterium marinum. Microbiol Mol Biol Rev 2020; 84:84/4/e00082-19. [DOI: 10.1128/mmbr.00082-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pathogenic mycobacteria cause chronic and acute diseases ranging from human tuberculosis (TB) to nontubercular infections.
Mycobacterium tuberculosis
causes both acute and chronic human tuberculosis. Environmentally acquired nontubercular mycobacteria (NTM) cause chronic disease in humans and animals. Not surprisingly, NTM and
M. tuberculosis
often use shared molecular mechanisms to survive within the host. The ESX-1 system is a specialized secretion system that is essential for virulence and is functionally conserved between
M. tuberculosis
and
Mycobacterium marinum
.
Collapse
|
14
|
Jia W, Shi Q, Zhang R, Shi L, Chu X. Unraveling proteome changes of irradiated goat meat and its relationship to off-flavor analyzed by high-throughput proteomics analysis. Food Chem 2020; 337:127806. [PMID: 32799167 DOI: 10.1016/j.foodchem.2020.127806] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 01/06/2023]
Abstract
Irradiation has been regarded as a perspective preservation technology to prolong the shelf life of goat meat. In order to evaluate the effect of irradiation on goat meat, the high-throughput proteomics was applied to observe the proteome profiles changes and off-flavor formation of irradiated goat meat. A total of 207 proteins with significant changes (fold change > 2 or < 0.5, P < 0.05, VIP > 1) was determined in five irradiation doses (0, 1, 2, 4 and 6 kGy). Among them, 26 proteins involving 7 functional enrichments terms were found to be related to off-flavor. Particularly, proteins involved in protein oxidation, cysteine and methionine metabolism lead to irradiation off-flavor formation. However, the irradiation off-flavor will gradually dissipate due to metabolic process during storage. This study was designed to provide a proteomic insight into irradiation off-flavor of goat meat and suggest an underlying connection between off-flavor formation and biological processes.
Collapse
Affiliation(s)
- Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China.
| | - Qingyun Shi
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Rong Zhang
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Lin Shi
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Xiaogang Chu
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China; Chinese Academy of Inspection and Quarantine, Beijing 100123, China
| |
Collapse
|
15
|
Sanchez KG, Ferrell MJ, Chirakos AE, Nicholson KR, Abramovitch RB, Champion MM, Champion PA. EspM Is a Conserved Transcription Factor That Regulates Gene Expression in Response to the ESX-1 System. mBio 2020; 11:e02807-19. [PMID: 32019792 PMCID: PMC7002343 DOI: 10.1128/mbio.02807-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/18/2019] [Indexed: 01/01/2023] Open
Abstract
Pathogenic mycobacteria encounter multiple environments during macrophage infection. Temporally, the bacteria are engulfed into the phagosome, lyse the phagosomal membrane, and interact with the cytosol before spreading to another cell. Virulence factors secreted by the mycobacterial ESX-1 (ESAT-6-system-1) secretion system mediate the essential transition from the phagosome to the cytosol. It was recently discovered that the ESX-1 system also regulates mycobacterial gene expression in Mycobacterium marinum (R. E. Bosserman, T. T. Nguyen, K. G. Sanchez, A. E. Chirakos, et al., Proc Natl Acad Sci U S A 114:E10772-E10781, 2017, https://doi.org/10.1073/pnas.1710167114), a nontuberculous mycobacterial pathogen, and in the human-pathogenic species M. tuberculosis (A. M. Abdallah, E. M. Weerdenburg, Q. Guan, R. Ummels, et al., PLoS One 14:e0211003, 2019, https://doi.org/10.1371/journal.pone.0211003). It is not known how the ESX-1 system regulates gene expression. Here, we identify the first transcription factor required for the ESX-1-dependent transcriptional response in pathogenic mycobacteria. We demonstrate that the gene divergently transcribed from the whiB6 gene and adjacent to the ESX-1 locus in mycobacterial pathogens encodes a conserved transcription factor (MMAR_5438, Rv3863, now espM). We prove that EspM from both M. marinum and M. tuberculosis directly and specifically binds the whiB6-espM intergenic region. We show that EspM is required for ESX-1-dependent repression of whiB6 expression and for the regulation of ESX-1-associated gene expression. Finally, we demonstrate that EspM functions to fine-tune ESX-1 activity in M. marinum Taking the data together, this report extends the esx-1 locus, defines a conserved regulator of the ESX-1 virulence pathway, and begins to elucidate how the ESX-1 system regulates gene expression.IMPORTANCE Mycobacterial pathogens use the ESX-1 system to transport protein substrates that mediate essential interactions with the host during infection. We previously demonstrated that in addition to transporting proteins, the ESX-1 secretion system regulates gene expression. Here, we identify a conserved transcription factor that regulates gene expression in response to the ESX-1 system. We demonstrate that this transcription factor is functionally conserved in M. marinum, a pathogen of ectothermic animals; M. tuberculosis, the human-pathogenic species that causes tuberculosis; and M. smegmatis, a nonpathogenic mycobacterial species. These findings provide the first mechanistic insight into how the ESX-1 system elicits a transcriptional response, a function of this protein transport system that was previously unknown.
Collapse
Affiliation(s)
- Kevin G Sanchez
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Micah J Ferrell
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Alexandra E Chirakos
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Kathleen R Nicholson
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Robert B Abramovitch
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Matthew M Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Patricia A Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
16
|
A New ESX-1 Substrate in Mycobacterium marinum That Is Required for Hemolysis but Not Host Cell Lysis. J Bacteriol 2019; 201:JB.00760-18. [PMID: 30833360 DOI: 10.1128/jb.00760-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
The ESX-1 (ESAT-6 system 1) secretion system plays a conserved role in the virulence of diverse mycobacterial pathogens, including the human pathogen Mycobacterium tuberculosis and M. marinum, an environmental mycobacterial species. The ESX-1 system promotes the secretion of protein virulence factors to the extracytoplasmic environment. The secretion of these proteins triggers the host response by lysing the phagosome during macrophage infection. Using proteomic analyses of the M. marinum secretome in the presence and absence of a functional ESX-1 system, we and others have hypothesized that MMAR_2894, a PE family protein, is a potential ESX-1 substrate in M. marinum We used genetic and quantitative proteomic approaches to determine if MMAR_2894 is secreted by the ESX-1 system, and we defined the requirement of MMAR_2894 for ESX-1-mediated secretion and virulence. We show that MMAR_2894 is secreted by the ESX-1 system in M. marinum and is itself required for the optimal secretion of the known ESX-1 substrates in M. marinum Moreover, we found that MMAR_2894 was differentially required for hemolysis and cytolysis of macrophages, two lytic activities ascribed to the M. marinum ESX-1 system.IMPORTANCE Both Mycobacterium tuberculosis, the cause of human tuberculosis (TB), and Mycobacterium marinum, a pathogen of ectotherms, use the ESX-1 secretion system to cause disease. There are many established similarities between the ESX-1 systems in M. tuberculosis and in M. marinum Yet the two bacteria infect different hosts, hinting at species-specific functions of the ESX-1 system. Our findings demonstrate that MMAR_2894 is a PE protein secreted by the ESX-1 system of M. marinum We show that MMAR_2894 is required for the optimal secretion of mycobacterial proteins required for disease. Because the MMAR_2894 gene is not conserved in M. tuberculosis, our findings demonstrate that MMAR_2894 may contribute to a species-specific function of the ESX-1 system in M. marinum, providing new insight into how the M. marinum and M. tuberculosis systems differ.
Collapse
|
17
|
Thompson CR, Champion MM, Champion PA. Quantitative N-Terminal Footprinting of Pathogenic Mycobacteria Reveals Differential Protein Acetylation. J Proteome Res 2018; 17:3246-3258. [PMID: 30080413 DOI: 10.1021/acs.jproteome.8b00373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
N-terminal acetylation (NTA) is a post-transcriptional modification of proteins that is conserved from bacteria to humans. In bacteria, the enzymes that mediate protein NTA also promote antimicrobial resistance. In pathogenic mycobacteria, which cause human tuberculosis and other chronic infections, NTA has been linked to pathogenesis and stress response, yet the fundamental biology underlying NTA of mycobacterial proteins remains unclear. We enriched, defined, and quantified the NT-acetylated populations of both cell-associated and secreted proteins from both the human pathogen, Mycobacterium tuberculosis, and the nontuberculous opportunistic pathogen, Mycobacterium marinum. We used a parallel N-terminal enrichment strategy from proteolytic digests coupled to charge-based selection and stable isotope ratio mass spectrometry. We show that NTA of the mycobacterial proteome is abundant, diverse, and primarily on Thr residues, which is unique compared with other bacteria. We isolated both the acetylated and unacetylated forms of 256 proteins, indicating that NTA of mycobacterial proteins is homeostatic. We identified 16 mycobacterial proteins with differential levels of NTA on the cytoplasmic and secreted forms, linking protein modification and localization. Our findings reveal novel biology underlying the NTA of mycobacterial proteins, which may provide a basis to understand NTA in mycobacterial physiology, pathogenesis, and antimicrobial resistance.
Collapse
|
18
|
Abstract
Mycobacterial 6-kDa early secreted antigenic target (ESAT-6) system (ESX) exporters transport proteins across the cytoplasmic membrane. Many proteins transported by ESX systems are then translocated across the mycobacterial cell envelope and secreted from the cell. Although the mechanism underlying protein transport across the mycolate outer membrane remains elusive, the ESX systems are closely connected with and localize to the cell envelope. Links between ESX-associated proteins, cell wall synthesis, and the maintenance of cell envelope integrity have been reported. Genes encoding the ESX systems and those required for biosynthesis of the mycobacterial envelope are coregulated. Here, we review the interplay between ESX systems and the mycobacterial cell envelope.
Collapse
|
19
|
Bronchoalveolar Lavage Fluid Protein Expression in Acute Respiratory Distress Syndrome Provides Insights into Pathways Activated in Subjects with Different Outcomes. Sci Rep 2017; 7:7464. [PMID: 28785034 PMCID: PMC5547130 DOI: 10.1038/s41598-017-07791-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with high mortality. We sought to identify biological pathways in ARDS that differentiate survivors from non-survivors. We studied bronchoalveolar lavage fluid (BALF) from 36 patients with ARDS (20 survivors, 16 non-survivors). Each sample, obtained within seven days of ARDS onset, was depleted of high abundance proteins and labeled for iTRAQ LC-MS/MS separately. Protein identification and relative quantification was performed employing a target-decoy strategy. A variance weighted t-test was used to identify differential expression. Ingenuity Pathway Analysis was used to determine the canonical pathways that differentiated survivors from non-survivors. We identified 1115 high confidence proteins in the BALF out of which 142 were differentially expressed between survivors and non-survivors. These proteins mapped to multiple pathways distinguishing survivors from non-survivors, including several implicated in lung injury and repair such as coagulation/thrombosis, acute phase response signaling and complement activation. We also identified proteins assigned to fibrosis and ones involved in detoxification of lipid peroxide-mediated oxidative stress to be different in survivors and non-survivors. These results support our previous findings demonstrating early differences in the BALF protein expression in ARDS survivors vs. non-survivors, including proteins that counter oxidative stress and canonical pathways associated with fibrosis.
Collapse
|
20
|
Greco TM, Cristea IM. Proteomics Tracing the Footsteps of Infectious Disease. Mol Cell Proteomics 2017; 16:S5-S14. [PMID: 28163258 DOI: 10.1074/mcp.o116.066001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/25/2017] [Indexed: 01/20/2023] Open
Abstract
Every year, a major cause of human disease and death worldwide is infection with the various pathogens-viruses, bacteria, fungi, and protozoa-that are intrinsic to our ecosystem. In efforts to control the prevalence of infectious disease and develop improved therapies, the scientific community has focused on building a molecular picture of pathogen infection and spread. These studies have been aimed at defining the cellular mechanisms that allow pathogen entry into hosts cells, their replication and transmission, as well as the core mechanisms of host defense against pathogens. The past two decades have demonstrated the valuable implementation of proteomic methods in all these areas of infectious disease research. Here, we provide a perspective on the contributions of mass spectrometry and other proteomics approaches to understanding the molecular details of pathogen infection. Specifically, we highlight methods used for defining the composition of viral and bacterial pathogens and the dynamic interaction with their hosts in space and time. We discuss the promise of MS-based proteomics in supporting the development of diagnostics and therapies, and the growing need for multiomics strategies for gaining a systems view of pathogen infection.
Collapse
Affiliation(s)
- Todd M Greco
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Ileana M Cristea
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| |
Collapse
|
21
|
A Nonsense Mutation in Mycobacterium marinum That Is Suppressible by a Novel Mechanism. Infect Immun 2017; 85:IAI.00653-16. [PMID: 27789543 DOI: 10.1128/iai.00653-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/17/2016] [Indexed: 11/20/2022] Open
Abstract
Mycobacterial pathogens use the ESAT-6 system 1 (Esx-1) exporter to promote virulence. Previously, we used gene disruption and complementation to conclude that the MMAR_0039 gene in Mycobacterium marinum is required to promote Esx-1 export. Here we applied molecular genetics, proteomics, and whole-genome sequencing to demonstrate that the MMAR_0039 gene is not required for Esx-1 secretion or virulence. These findings suggest that we initially observed an indirect mechanism of genetic complementation. We identified a spontaneous nonsense mutation in a known Esx-1-associated gene which causes a loss of Esx-1 activity. We show that the Esx-1 function was restored by nonsense suppression. Moreover, we identified a polar mutation in the ppsC gene which reduced cellular impermeability but did not impact cytotoxicity in macrophages. Our studies reveal insight into Esx-1 export, nonsense suppression, and cell envelope lipid biogenesis.
Collapse
|
22
|
Rational engineering of a virulence gene from Mycobacterium tuberculosis facilitates proteomic analysis of a natural protein N-terminus. Sci Rep 2016; 6:33265. [PMID: 27625110 PMCID: PMC5021934 DOI: 10.1038/srep33265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/24/2016] [Indexed: 12/24/2022] Open
Abstract
Mass spectrometry (MS) for the detection of proteins is an indispensable tool for evaluating the biological processes of the proteome. Proteomics frequently requires proteolysis of proteins into peptide fragments. Proteins can be refractory to ideal proteolysis at the sequence level rendering them difficult to analyze by routine proteomics methods. EsxA (ESAT-6, Early Secreted Antigen, 6kDa) is a major virulence determinant of Mycobacterium tuberculosis, the cause of human tuberculosis. EsxA is routinely used to evaluate mycobacterial virulence in the laboratory and as a biomarker for tuberculosis in humans. The sequence of EsxA hinders deeper MS analysis beyond routine detection. Here we engineer the sequence of EsxA to add desirable tryptic properties aimed at improving complex MS analysis. We demonstrate that EsxA variants are amenable to MS analysis and remain functional in established in vitro and ex vivo assays of Esx-1-function. We provide the first demonstration of molecular engineering to specifically improve MS analysis of individual microbial proteins.
Collapse
|
23
|
Li X, Zhao Z, Pan C. Ionic liquid-assisted electrochemical exfoliation of carbon dots of different size for fluorescent imaging of bacteria by tuning the water fraction in electrolyte. Mikrochim Acta 2016; 183:2525-2532. [PMID: 27546906 PMCID: PMC4977338 DOI: 10.1007/s00604-016-1877-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/18/2016] [Indexed: 11/28/2022]
Abstract
An electrochemical approach is introduced for synthesis of carbon dots (CDs) by exfoliating graphite rods at a voltage of 15 V in an electrolyte consisting of a mixture of water and two ionic liquids. It is found that the size of the CDs can be tuned by varying the fraction of water in the mixed electrolyte; CDs in sizes of 4.9, 4.1 and 3.1 nm are obtained if the electrolyte contains water in fractions of 24, 38 and 56 %, respectively. The CDs have a quantum yield of almost 10 % and display the typical excitation wavelength-dependent maxima of photoluminescence, strongest at excitation/emission wavelengths of 360/440 nm. Fourier transform infrared and X-ray photoelectron spectroscopy show the CDs to have oxygen functional groups on their surface which strongly improve solubility. The CDs were applied to image cells of the electricity-producing bacteria Shewanellaoneidensis MR-1. Graphical AbstractAn electrochemical approach is introduced to synthesize carbon dots by exfoliating graphite rods in mixed electrolyte of water and ionic liquids. The increasing size of carbon dots was realized by reducing the volume of water in the mixed electrolyte. The carbon dots were used to fluorescently image the electricity-producing bacterium Shewanellaoneidensis MR-1.
Collapse
Affiliation(s)
- Xuehua Li
- School of Electronic Science and Engineering, Southeast University, Nanjing, 210096 People’s Republic of China
| | - Zhiwei Zhao
- School of Electronic Science and Engineering, Southeast University, Nanjing, 210096 People’s Republic of China
| | - Chen Pan
- School of Electronic Science and Engineering, Southeast University, Nanjing, 210096 People’s Republic of China
| |
Collapse
|
24
|
Bhargava M, Viken KJ, Dey S, Steinbach MS, Wu B, Jagtap PD, Higgins L, Panoskaltsis-Mortari A, Weisdorf DJ, Kumar V, Arora M, Bitterman PB, Ingbar DH, Wendt CH. Proteome Profiling in Lung Injury after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2016; 22:1383-1390. [PMID: 27155584 DOI: 10.1016/j.bbmt.2016.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 04/25/2016] [Indexed: 11/26/2022]
Abstract
Pulmonary complications due to infection and idiopathic pneumonia syndrome (IPS), a noninfectious lung injury in hematopoietic stem cell transplant (HSCT) recipients, are frequent causes of transplantation-related mortality and morbidity. Our objective was to characterize the global bronchoalveolar lavage fluid (BALF) protein expression of IPS to identify proteins and pathways that differentiate IPS from infectious lung injury after HSCT. We studied 30 BALF samples from patients who developed lung injury within 180 days of HSCT or cellular therapy transfusion (natural killer cell transfusion). Adult subjects were classified as having IPS or infectious lung injury by the criteria outlined in the 2011 American Thoracic Society statement. BALF was depleted of hemoglobin and 14 high-abundance proteins, treated with trypsin, and labeled with isobaric tagging for relative and absolute quantification (iTRAQ) 8-plex reagent for two-dimensional capillary liquid chromatography (LC) and data dependent peptide tandem mass spectrometry (MS) on an Orbitrap Velos system in higher-energy collision-induced dissociation activation mode. Protein identification employed a target-decoy strategy using ProteinPilot within Galaxy P. The relative protein abundance was determined with reference to a global internal standard consisting of pooled BALF from patients with respiratory failure and no history of HSCT. A variance weighted t-test controlling for a false discovery rate of ≤5% was used to identify proteins that showed differential expression between IPS and infectious lung injury. The biological relevance of these proteins was determined by using gene ontology enrichment analysis and Ingenuity Pathway Analysis. We characterized 12 IPS and 18 infectious lung injury BALF samples. In the 5 iTRAQ LC-MS/MS experiments 845, 735, 532, 615, and 594 proteins were identified for a total of 1125 unique proteins and 368 common proteins across all 5 LC-MS/MS experiments. When comparing IPS to infectious lung injury, 96 proteins were differentially expressed. Gene ontology enrichment analysis showed that these proteins participate in biological processes involved in the development of lung injury after HSCT. These include acute phase response signaling, complement system, coagulation system, liver X receptor (LXR)/retinoid X receptor (RXR), and farsenoid X receptor (FXR)/RXR modulation. We identified 2 canonical pathways modulated by TNF-α, FXR/RXR activation, and IL2 signaling in macrophages. The proteins also mapped to blood coagulation, fibrinolysis, and wound healing-processes that participate in organ repair. Cell movement was identified as significantly over-represented by proteins with differential expression between IPS and infection. In conclusion, the BALF protein expression in IPS differed significantly from infectious lung injury in HSCT recipients. These differences provide insights into mechanisms that are activated in lung injury in HSCT recipients and suggest potential therapeutic targets to augment lung repair.
Collapse
Affiliation(s)
- Maneesh Bhargava
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Minnesota Medical School, Minneapolis, Minnesota.
| | - Kevin J Viken
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Sanjoy Dey
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Michael S Steinbach
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Baolin Wu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Pratik D Jagtap
- Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - LeeAnn Higgins
- Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Angela Panoskaltsis-Mortari
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Daniel J Weisdorf
- Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Vipin Kumar
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Mukta Arora
- Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Peter B Bitterman
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - David H Ingbar
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Chris H Wendt
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Minnesota Medical School, Minneapolis, Minnesota; Pulmonary, Critical Care and Sleep Medicine, Minneapolis Veterans Affairs, Minneapolis, Minnesota
| |
Collapse
|
25
|
Abstract
Amphiphilic carbon dots employed for detection and visualization of bacterial cells and distinguishing among different bacterial strains.
Collapse
Affiliation(s)
- Sukhendu Nandi
- Department of Chemistry
- Ben Gurion University of the Negev
- Beer Sheva 84105
- Israel
| | - Margarita Ritenberg
- Department of Chemistry
- Ben Gurion University of the Negev
- Beer Sheva 84105
- Israel
| | - Raz Jelinek
- Department of Chemistry
- Ben Gurion University of the Negev
- Beer Sheva 84105
- Israel
- Ilse Katz Institute for Nanotechnology
| |
Collapse
|
26
|
Shi D, Yan F, Zheng T, Wang Y, Zhou X, Chen L. P-doped carbon dots act as a nanosensor for trace 2,4,6-trinitrophenol detection and a fluorescent reagent for biological imaging. RSC Adv 2015. [DOI: 10.1039/c5ra18800h] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A simple and rapid method for sensitive and selective detection of 2,4,6-trinitrophenol (TNP) was developed with the use of water-soluble carbon dots (CDs) as a nanosensor.
Collapse
Affiliation(s)
- Dechao Shi
- State Key Laboratory of Hollow Fiber Membrane Materials and Processes
- Key Lab of Fiber Modification & Functional Fiber of Tianjin
- Tianjin Polytechnic University
- Tianjin 300387
- PR China
| | - Fanyong Yan
- State Key Laboratory of Hollow Fiber Membrane Materials and Processes
- Key Lab of Fiber Modification & Functional Fiber of Tianjin
- Tianjin Polytechnic University
- Tianjin 300387
- PR China
| | - Tancheng Zheng
- State Key Laboratory of Hollow Fiber Membrane Materials and Processes
- Key Lab of Fiber Modification & Functional Fiber of Tianjin
- Tianjin Polytechnic University
- Tianjin 300387
- PR China
| | - Yinyin Wang
- TianJin Engineering Center for Safety Evaluation of Water Quality & Safeguards Technology
- PR China
| | - Xuguang Zhou
- State Key Laboratory of Hollow Fiber Membrane Materials and Processes
- Key Lab of Fiber Modification & Functional Fiber of Tianjin
- Tianjin Polytechnic University
- Tianjin 300387
- PR China
| | - Li Chen
- State Key Laboratory of Hollow Fiber Membrane Materials and Processes
- Key Lab of Fiber Modification & Functional Fiber of Tianjin
- Tianjin Polytechnic University
- Tianjin 300387
- PR China
| |
Collapse
|
27
|
Champion MM, Williams EA, Pinapati RS, Champion PAD. Correlation of phenotypic profiles using targeted proteomics identifies mycobacterial esx-1 substrates. J Proteome Res 2014; 13:5151-64. [PMID: 25106450 PMCID: PMC4227905 DOI: 10.1021/pr500484w] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
![]()
The
Esx/WXG-100 (ESAT-6/Wss) exporters are multiprotein complexes
that promote protein translocation across the cytoplasmic membrane
in a diverse range of pathogenic and nonpathogenic bacterial species.
The Esx-1 (ESAT-6 System-1) system mediates virulence factor translocation
in mycobacterial pathogens, including the human pathogen Mycobacterium
tuberculosis. Although several genes have been associated
with Esx-1-mediated transport and virulence, the contribution of individual
Esx-1 genes to export is largely undefined. A unique aspect of Esx-1
export is that several substrates require each other for export/stability.
We exploited substrate “codependency” to identify Esx-1
substrates. We simultaneously quantified changes in the levels of
13 Esx-1 proteins from both secreted and cytosolic protein fractions
generated from 16 Esx-1-deficient Mycobacterium marinum strains in a single experiment using MRM/SRM targeted mass spectrometry.
This expansion of measurable Esx-1 proteins allowed us to define statistical
rules for assigning novel substrates using phenotypic profiles of
known Esx-1 substrates. Using this approach, we identified three additional
Esx-1 substrates encoded by the esx-1 region. Our
studies begin to address how disruption of specific genes affects
several proteins in the Esx-1 complex. Overall, our findings illuminate
relationships between Esx-1 proteins and create a framework for the
identification of secreted substrates applicable to other protein
exporters and pathways.
Collapse
Affiliation(s)
- Matthew M Champion
- Department of Chemistry and Biochemistry, ‡Department of Biological Sciences, §Eck Institute for Global Health, and ∥Center for Rare and Neglected Diseases, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | | | | | | |
Collapse
|
28
|
Homeostasis of N-α-terminal acetylation of EsxA correlates with virulence in Mycobacterium marinum. Infect Immun 2014; 82:4572-86. [PMID: 25135684 DOI: 10.1128/iai.02153-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The mycobacterial Esx-1 (ESAT-6 system 1) exporter translocates virulence factors across the cytoplasmic membrane to the cell wall, cell surface, and the bacteriological medium in vitro. The mechanisms underlying substrate targeting to distinct locations are unknown. Several Esx-1 substrates are N-α-terminally acetylated. The role of this rare modification in bacteria is unclear. We sought to identify genes required for Esx-1 substrate modification, transport, and localization. Pathogenic mycobacteria lyse Acanthamoeba castellanii in an Esx-1-dependent manner. We conducted a genetic screen to identify Mycobacterium marinum strains which failed to lyse amoebae. We identified a noncytotoxic M. marinum strain with a transposon insertion in a predicted N-α-terminal acetyltransferase not previously linked to mycobacterial pathogenesis. Disruption of this gene led to attenuation of virulence, failure to induce a type I interferon response during macrophage infection, and loss of hemolytic activity. The major Esx-1 substrates, EsxA and EsxB, were exported to the cell surface, but only low levels were released into the bacteriological medium. The balance of EsxA N-α-terminal acetylation was disrupted, resulting in a mycobacterial strain in which surface-associated EsxA was hyperacetylated. Genetic complementation completely restored Esx-1 function and the levels of N-α-terminally acetylated EsxA on the surface but restored only low levels of Esx-1 substrates in the bacteriological medium. Our results reveal a novel gene required for mycobacterial Esx-1 export. Our findings indicate that maintaining the homeostasis of Esx-1 substrate N-α-terminal acetylation is essential for Esx-1-mediated virulence. We propose an inverse correlation between EsxA acetylation and virulence.
Collapse
|
29
|
Daily rhythms in antennal protein and olfactory sensitivity in the malaria mosquito Anopheles gambiae. Sci Rep 2014; 3:2494. [PMID: 23986098 PMCID: PMC3756343 DOI: 10.1038/srep02494] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/07/2013] [Indexed: 11/08/2022] Open
Abstract
We recently characterized 24-hr daily rhythmic patterns of gene expression in Anopheles gambiae mosquitoes. These include numerous odorant binding proteins (OBPs), soluble odorant carrying proteins enriched in olfactory organs. Here we demonstrate that multiple rhythmically expressed genes including OBPs and takeout proteins, involved in regulating blood feeding behavior, have corresponding rhythmic protein levels as measured by quantitative proteomics. This includes AgamOBP1, previously shown as important to An. gambiae odorant sensing. Further, electrophysiological investigations demonstrate time-of-day specific differences in olfactory sensitivity of antennae to major host-derived odorants. The pre-dusk/dusk peaks in OBPs and takeout gene expression correspond with peak protein abundance at night, and in turn coincide with the time of increased olfactory sensitivity to odorants requiring OBPs and times of increased blood-feeding behavior. This suggests an important role for OBPs in modulating temporal changes in odorant sensitivity, enabling the olfactory system to coordinate with the circadian niche of An. gambiae.
Collapse
|
30
|
Siegrist MS, Steigedal M, Ahmad R, Mehra A, Dragset MS, Schuster BM, Philips JA, Carr SA, Rubin EJ. Mycobacterial Esx-3 requires multiple components for iron acquisition. mBio 2014; 5:e01073-14. [PMID: 24803520 PMCID: PMC4010830 DOI: 10.1128/mbio.01073-14] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 03/28/2014] [Indexed: 12/16/2022] Open
Abstract
ABSTRACT The type VII secretion systems are conserved across mycobacterial species and in many Gram-positive bacteria. While the well-characterized Esx-1 pathway is required for the virulence of pathogenic mycobacteria and conjugation in the model organism Mycobacterium smegmatis, Esx-3 contributes to mycobactin-mediated iron acquisition in these bacteria. Here we show that several Esx-3 components are individually required for function under low-iron conditions but that at least one, the membrane-bound protease MycP3 of M. smegmatis, is partially expendable. All of the esx-3 mutants tested, including the ΔmycP3ms mutant, failed to export the native Esx-3 substrates EsxHms and EsxGms to quantifiable levels, as determined by targeted mass spectrometry. Although we were able to restore low-iron growth to the esx-3 mutants by genetic complementation, we found a wide range of complementation levels for protein export. Indeed, minute quantities of extracellular EsxHms and EsxGms were sufficient for iron acquisition under our experimental conditions. The apparent separation of Esx-3 function in iron acquisition from robust EsxGms and EsxHms secretion in the ΔmycP3ms mutant and in some of the complemented esx-3 mutants compels reexamination of the structure-function relationships for type VII secretion systems. IMPORTANCE Mycobacteria have several paralogous type VII secretion systems, Esx-1 through Esx-5. Whereas Esx-1 is required for pathogenic mycobacteria to grow within an infected host, Esx-3 is essential for growth in vitro. We and others have shown that Esx-3 is required for siderophore-mediated iron acquisition. In this work, we identify individual Esx-3 components that contribute to this process. As in the Esx-1 system, most mutations that abolish Esx-3 protein export also disrupt its function. Unexpectedly, however, ultrasensitive quantitation of Esx-3 secretion by multiple-reaction-monitoring mass spectrometry (MRM-MS) revealed that very low levels of export were sufficient for iron acquisition under similar conditions. Although protein export clearly contributes to type VII function, the relationship is not absolute.
Collapse
Affiliation(s)
- M. Sloan Siegrist
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | | | - Rushdy Ahmad
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Alka Mehra
- Division of Infectious Diseases, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | | | - Brian M. Schuster
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Jennifer A. Philips
- Division of Infectious Diseases, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Steven A. Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Eric J. Rubin
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Weigel KJ, Jakimenko A, Conti BA, Chapman SE, Kaliney WJ, Leevy WM, Champion MM, Schafer ZT. CAF-Secreted IGFBPs Regulate Breast Cancer Cell Anoikis. Mol Cancer Res 2014; 12:855-66. [DOI: 10.1158/1541-7786.mcr-14-0090] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
32
|
Geraci NS, Mukbel RM, Kemp MT, Wadsworth MN, Lesho E, Stayback GM, Champion MM, Bernard MA, Abo-Shehada M, Coutinho-Abreu IV, Ramalho-Ortigão M, Hanafi HA, Fawaz EY, El-Hossary SS, Wortmann G, Hoel DF, McDowell MA. Profiling of human acquired immunity against the salivary proteins of Phlebotomus papatasi reveals clusters of differential immunoreactivity. Am J Trop Med Hyg 2014; 90:923-938. [PMID: 24615125 PMCID: PMC4015589 DOI: 10.4269/ajtmh.13-0130] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Phlebotomus papatasi sand flies are among the primary vectors of Leishmania major parasites from Morocco to the Indian subcontinent and from southern Europe to central and eastern Africa. Antibody-based immunity to sand fly salivary gland proteins in human populations remains a complex contextual problem that is not yet fully understood. We profiled the immunoreactivities of plasma antibodies to sand fly salivary gland sonicates (SGSs) from 229 human blood donors residing in different regions of sand fly endemicity throughout Jordan and Egypt as well as 69 US military personnel, who were differentially exposed to P. papatasi bites and L. major infections in Iraq. Compared with plasma from control region donors, antibodies were significantly immunoreactive to five salivary proteins (12, 26, 30, 38, and 44 kDa) among Jordanian and Egyptian donors, with immunoglobulin G4 being the dominant anti-SGS isotype. US personnel were significantly immunoreactive to only two salivary proteins (38 and 14 kDa). Using k-means clustering, donors were segregated into four clusters distinguished by unique immunoreactivity profiles to varying combinations of the significantly immunogenic salivary proteins. SGS-induced cellular proliferation was diminished among donors residing in sand fly-endemic regions. These data provide a clearer picture of human immune responses to sand fly vector salivary constituents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mary Ann McDowell
- * Address correspondence to Mary Ann McDowell, Eck Institute for Global Health, University of Notre Dame, 215 Galvin Life Sciences, Notre Dame, IN 46556. E-mail:
| |
Collapse
|
33
|
A novel ESX-1 locus reveals that surface-associated ESX-1 substrates mediate virulence in Mycobacterium marinum. J Bacteriol 2014; 196:1877-88. [PMID: 24610712 DOI: 10.1128/jb.01502-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
EsxA (ESAT-6) and EsxB (CFP-10) are virulence factors exported by the ESX-1 system in mycobacterial pathogens. In Mycobacterium marinum, an established model for ESX-1 secretion in Mycobacterium tuberculosis, genes required for ESX-1 export reside at the extended region of difference 1 (RD1) locus. In this study, a novel locus required for ESX-1 export in M. marinum was identified outside the RD1 locus. An M. marinum strain bearing a transposon-insertion between the MMAR_1663 and MMAR_1664 genes exhibited smooth-colony morphology, was deficient for ESX-1 export, was nonhemolytic, and was attenuated for virulence. Genetic complementation revealed a restoration of colony morphology and a partial restoration of virulence in cell culture models. Yet hemolysis and the export of ESX-1 substrates into the bacteriological medium in vitro as measured by both immunoblotting and quantitative proteomics were not restored. We show that genetic complementation of the transposon insertion strain partially restored the translocation of EsxA and EsxB to the mycobacterial cell surface. Our findings indicate that the export of EsxA and EsxB to the cell surface, rather than secretion into the bacteriological medium, correlates with virulence in M. marinum. Together, these findings not only expand the known genetic loci required for ESX-1 secretion in M. marinum but also provide an explanation for the observed disparity between in vitro ESX-1 export and virulence.
Collapse
|
34
|
|
35
|
Armengaud J, Christie-Oleza JA, Clair G, Malard V, Duport C. Exoproteomics: exploring the world around biological systems. Expert Rev Proteomics 2013. [PMID: 23194272 DOI: 10.1586/epr.12.52] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term 'exoproteome' describes the protein content that can be found in the extracellular proximity of a given biological system. These proteins arise from cellular secretion, other protein export mechanisms or cell lysis, but only the most stable proteins in this environment will remain in abundance. It has been shown that these proteins reflect the physiological state of the cells in a given condition and are indicators of how living systems interact with their environments. High-throughput proteomic approaches based on a shotgun strategy, and high-resolution mass spectrometers, have modified the authors' view of exoproteomes. In the present review, the authors describe how these new approaches should be exploited to obtain the maximum useful information from a sample, whatever its origin. The methodologies used for studying secretion from model cell lines derived from eukaryotic, multicellular organisms, virulence determinants of pathogens and environmental bacteria and their relationships with their habitats are illustrated with several examples. The implication of such data, in terms of proteogenomics and the discovery of novel protein functions, is discussed.
Collapse
Affiliation(s)
- Jean Armengaud
- CEA, DSV, IBEB, Lab Biochim System Perturb, Bagnols-sur-Cèze, F-30207, France.
| | | | | | | | | |
Collapse
|
36
|
Low KO, Muhammad Mahadi N, Md. Illias R. Optimisation of signal peptide for recombinant protein secretion in bacterial hosts. Appl Microbiol Biotechnol 2013; 97:3811-26. [DOI: 10.1007/s00253-013-4831-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 03/03/2013] [Accepted: 03/04/2013] [Indexed: 10/27/2022]
|
37
|
Ngounou Wetie AG, Sokolowska I, Woods AG, Wormwood KL, Dao S, Patel S, Clarkson BD, Darie CC. Automated Mass Spectrometry–Based Functional Assay for the Routine Analysis of the Secretome. ACTA ACUST UNITED AC 2013; 18:19-29. [DOI: 10.1177/2211068212454738] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
van der Woude AD, Luirink J, Bitter W. Getting across the cell envelope: mycobacterial protein secretion. Curr Top Microbiol Immunol 2012; 374:109-34. [PMID: 23239236 DOI: 10.1007/82_2012_298] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Protein secretion is an essential determinant of mycobacterial virulence. Mycobacterium tuberculosis has a unique cell envelope consisting of two lipid bilayers, which requires dedicated protein secretion pathways. The conserved general Sec and Tat translocation systems are responsible for protein transport across the inner membrane and are both essential. Additionally, the accessory Sec pathway specifically contributes to virulence. How transport of Sec/Tat substrates across the outer membrane is accomplished is currently an enigma. In addition to these pathways, M. tuberculosis also developed specialized secretion systems for protein transport across both membranes, the type VII or ESX secretion systems. Here, we discuss our current knowledge about the mechanisms and substrates of these different protein translocation systems and their role in mycobacterial physiology and virulence.
Collapse
Affiliation(s)
- Aniek D van der Woude
- Department of Molecular Microbiology, Institute of Molecular Cell Biology, VU University, Amsterdam, The Netherlands
| | | | | |
Collapse
|