1
|
Mahesh PP, Kolape J, Sultana H, Neelakanta G. McFarland Standards-Based Spectrophotometry Method for Calculating Approximate Multiplicity of Infection for an Obligate Intracellular Bacterium Anaplasma phagocytophilum. Microorganisms 2025; 13:662. [PMID: 40142553 PMCID: PMC11945594 DOI: 10.3390/microorganisms13030662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Anaplasma phagocytophilum is an obligate intracellular Gram-negative bacterium that causes human granulocytic anaplasmosis. Assessing the number of these bacteria is important for in vitro and in vivo infection studies. Colony count is used to set references for the multiplicity of infections in the case of culturable bacteria. However, the number of bacteria present inside the host cells, in which the bacteria are maintained, can be considered in the case of obligate intracellular bacteria. McFarland standards are a series of turbidity-based standards used to visually assess the approximate number of culturable bacteria. The turbidity of each standard can be related to their respective absorbances or optical densities (ODs). In this study, we describe a simple method to assess the approximate number of A. phagocytophilum based on McFarland standards. The ODs of cell-free crude extracts of A. phagocytophilum were used to assess the approximate number of bacteria while considering that the cell debris also contributes to the ODs. The consistency of this method was also tested using the bacterial cultures grown at different times. In summary, we provide a simple method to estimate the number of obligate intracellular bacteria for use in in vitro infection studies.
Collapse
Affiliation(s)
- P. P. Mahesh
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (P.P.M.); (H.S.)
| | - Jaydeep Kolape
- Advanced Microscopy and Imaging Center, College of Arts and Sciences, University of Tennessee, Knoxville, TN 37996, USA
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (P.P.M.); (H.S.)
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (P.P.M.); (H.S.)
| |
Collapse
|
2
|
Namjoshi P, Kolape J, Patel A, Sultana H, Neelakanta G. Rickettsial pathogen augments tick vesicular-associated membrane proteins for infection and survival in the vector host. mBio 2025; 16:e0354924. [PMID: 39950824 PMCID: PMC11898744 DOI: 10.1128/mbio.03549-24] [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: 11/18/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
Anaplasma phagocytophilum is an obligate intracellular rickettsial pathogen that infects humans and animals. The black-legged tick Ixodes scapularis acts as a vector and transmits this bacterium to the vertebrate host. Upon entry into a host cell, A. phagocytophilum resides and multiplies in a host-derived vacuole called morulae. There is not much information available on the molecules that play an important role(s) in A. phagocytophilum entry and formation of these morulae in tick cells. In this study, we provide evidence that tick vesicular-associated membrane proteins, VAMP3 and VAMP4, play important roles in this phenomenon. Quantitative real-time polymerase chain reaction (QRT-PCR) analysis showed that both vamp3 and vamp4 transcripts are significantly upregulated at early time points of A. phagocytophilum infection in tick cells. We noted that both VAMP3 and VAMP4 predominantly localized to the A. phagocytophilum-containing vacuole. RNAi-mediated silencing of vamp3 and/or vamp4 expression, followed by confocal microscopy and expression analysis, indicated an impairment in A. phagocytophilum morulae formation in tick cells. We also noted that VAMP3 and VAMP4 play a role in the A. phagocytophilum persistent infection of ticks and tick cells. Furthermore, RNAi-mediated silencing of expression of arthropod vamp3 and vamp4 affected bacterial acquisition from an infected murine host to ticks. Collectively, this study not only provides evidence on the role of arthropod vesicular-associated membrane proteins in A. phagocytophilum morulae formation in tick cells but also demonstrates that these proteins are important for bacterial acquisition from an infected vertebrate host into ticks. IMPORTANCE Anaplasma phagocytophilum is a tick-borne pathogen primarily transmitted by black-legged Ixodes scapularis ticks to humans and animals. This bacterium enters host cells, forms a host-derived vacuole, and multiplies within this vacuole. The molecules that are critical in the formation of host-derived vacuole in tick cells is currently not well-characterized. In this study, we provide evidence that arthropod vesicular-associated membrane proteins, VAMP3 and VAMP4, are critical for A. phagocytophilum early and persistent infection in tick cells. These arthropod proteins are important for the formation of host-derived vacuoles in tick cells. Our study also provides evidence that these proteins are important for A. phagocytophilum acquisition from the infected murine host into ticks. Characterization of tick molecules important in bacterial entry and/or survival in the vector host could lead to the development of strategies to target this and perhaps other rickettsial pathogens.
Collapse
Affiliation(s)
- Prachi Namjoshi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Jaydeep Kolape
- Advanced Microscopy and Imaging Center, College of Arts and Sciences, University of Tennessee, Knoxville, Tennessee, USA
| | - Avni Patel
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
3
|
Lind MCH, Naimi WA, Chiarelli TJ, Sparrer T, Ghosh M, Shapiro L, Carlyon JA. Anaplasma phagocytophilum invasin AipA interacts with CD13 to elicit Src kinase signaling that promotes infection. mBio 2024; 15:e0156124. [PMID: 39324816 PMCID: PMC11481542 DOI: 10.1128/mbio.01561-24] [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: 05/28/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
Host-microbe interactions that facilitate entry into mammalian cells are essential for obligate intracellular bacterial survival and pathogenesis. Anaplasma phagocytophilum is an obligate intracellular bacterium that invades neutrophils to cause granulocytic anaplasmosis. The invasin-receptor pairs and signaling events that induce Anaplasma uptake are inadequately defined. A. phagocytophilum invasion protein A orchestrates entry via residues 9-21 (AipA9-21) engaging an unknown receptor. Yeast two-hybrid screening suggested that AipA binds within C-terminal amino acids 851-967 of CD13 (aminopeptidase N), a multifunctional protein that, when crosslinked, initiates Src kinase and Syk signaling that culminates in endocytosis. Co-immunoprecipitation validated the interaction and confirmed that it requires the AipA N-terminus. CD13 ectopic expression on non-phagocytic cells increased susceptibility to A. phagocytophilum infection. Antibody blocking and enzymatic inhibition experiments found that the microbe exploits CD13 but not its ectopeptidase activity to infect myeloid cells. A. phagocytophilum induces Src and Syk phosphorylation during invasion. Inhibitor treatment established that Src is key for A. phagocytophilum infection, while Syk is dispensable and oriented the pathogen-invoked signaling pathway by showing that Src is activated before Syk. Disrupting the AipA-CD13 interaction with AipA9-21 or CD13781-967 antibody inhibited Src and Syk phosphorylation and also infection. CD13 crosslinking antibody that induces Src and Syk signaling restored infectivity of anti-AipA9-21-treated A. phagocytophilum. The bacterium poorly infected CD13 knockout mice, providing the first demonstration that CD13 is important for microbial infection in vivo. Overall, A. phagocytophilum AipA9-21 binds CD13 to induce Src signaling that mediates uptake into host cells, and CD13 is critical for infection in vivo. IMPORTANCE Diverse microbes engage CD13 to infect host cells. Yet invasin-CD13 interactions, the signaling they invoke for pathogen entry, and the relevance of CD13 to infection in vivo are underexplored. Dissecting these concepts would advance fundamental understanding of a convergently evolved infection strategy and could have translational benefits. Anaplasma phagocytophilum infects neutrophils to cause granulocytic anaplasmosis, an emerging disease for which there is no vaccine and few therapeutic options. We found that A. phagocytophilum uses its surface protein and recently identified protective immunogen, AipA, to bind CD13 to elicit Src kinase signaling, which is critical for infection. We elucidated the AipA CD13 binding domain, which CD13 region AipA engages, and established that CD13 is key for A. phagocytophilum infection in vivo. Disrupting the AipA-CD13 interaction could be utilized to prevent granulocytic anaplasmosis and offers a model that could be applied to protect against multiple infectious diseases.
Collapse
Affiliation(s)
- Mary Clark H. Lind
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Waheeda A. Naimi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Travis J. Chiarelli
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Tavis Sparrer
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Mallika Ghosh
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Linda Shapiro
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
4
|
Torres-Escobar A, Wilkins A, Juárez-Rodríguez MD, Circu M, Latimer B, Dragoi AM, Ivanov SS. Iron-depleting nutritional immunity controls extracellular bacterial replication in Legionella pneumophila infections. Nat Commun 2024; 15:7848. [PMID: 39245746 PMCID: PMC11381550 DOI: 10.1038/s41467-024-52184-x] [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: 02/02/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
The accidental human pathogen Legionella pneumophila (Lp) is the etiological agent for a severe atypical pneumonia known as Legionnaires' disease. In human infections and animal models of disease alveolar macrophages are the primary cellular niche that supports bacterial replication within a unique intracellular membrane-bound organelle. The Dot/Icm apparatus-a type IV secretion system that translocates ~300 bacterial proteins within the cytosol of the infected cell-is a central virulence factor required for intracellular growth. Mutant strains lacking functional Dot/Icm apparatus are transported to and degraded within the lysosomes of infected macrophages. The early foundational work from Dr. Horwitz's group unequivocally established that Legionella does not replicate extracellularly during infection-a phenomenon well supported by experimental evidence for four decades. Our data challenges this paradigm by demonstrating that macrophages and monocytes provide the necessary nutrients and support robust Legionella extracellular replication. We show that the previously reported lack of Lp extracellular replication is not a bacteria intrinsic feature but rather a result of robust restriction by serum-derived nutritional immunity factors. Specifically, the host iron-sequestering protein Transferrin is identified here as a critical suppressor of Lp extracellular replication in an iron-dependent manner. In iron-overload conditions or in the absence of Transferrin, Lp bypasses growth restriction by IFNγ-primed macrophages though extracellular replication. It is well established that certain risk factors associated with development of Legionnaires' disease, such as smoking, produce a chronic pulmonary environment of iron-overload. Our work indicates that iron-overload could be an important determinant of severe infection by allowing Lp to overcome nutritional immunity and replicate extracellularly, which in turn would circumvent intracellular cell intrinsic host defenses. Thus, we provide evidence for nutritional immunity as a key underappreciated host defense mechanism in Legionella pathogenesis.
Collapse
Affiliation(s)
- Ascención Torres-Escobar
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Ashley Wilkins
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
- Bacterial Physiology and Metabolism Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - María D Juárez-Rodríguez
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Magdalena Circu
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Brian Latimer
- Innovative North Louisiana Experimental Therapeutics program (INLET), Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Ana-Maria Dragoi
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
- Innovative North Louisiana Experimental Therapeutics program (INLET), Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Stanimir S Ivanov
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA.
| |
Collapse
|
5
|
Ranaweera CB, Shiva S, Madesh S, Chauhan D, Ganta RR, Zolkiewski M. Biochemical characterization of ClpB and DnaK from Anaplasma phagocytophilum. Cell Stress Chaperones 2024; 29:540-551. [PMID: 38908470 PMCID: PMC11268196 DOI: 10.1016/j.cstres.2024.06.003] [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: 05/13/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Anaplasma phagocytophilum is an intracellular tick-transmitted bacterial pathogen that infects neutrophils in mammals and causes granulocytic anaplasmosis. In this study, we investigated the molecular chaperones ClpB and DnaK from A. phagocytophilum. In Escherichia coli, ClpB cooperates with DnaK and its co-chaperones DnaJ and GrpE in ATP-dependent reactivation of aggregated proteins. Since ClpB is not produced in metazoans, it is a promising target for developing antimicrobial therapies, which generates interest in studies on that chaperone's role in pathogenic bacteria. We found that ClpB and DnaK are transcriptionally upregulated in A. phagocytophilum 3-5 days after infection of human HL-60 and tick ISE6 cells, which suggests an essential role of the chaperones in supporting the pathogen's intracellular life cycle. Multiple sequence alignments show that A. phagocytophilum ClpB and DnaK contain all structural domains that were identified in their previously studied orthologs from other bacteria. Both A. phagocytophilum ClpB and DnaK display ATPase activity, which is consistent with their participation in the ATP-dependent protein disaggregation system. However, despite a significant sequence similarity between the chaperones from A. phagocytophilum and those from E. coli, the former were not as effective as their E. coli orthologs during reactivation of aggregated proteins in vitro and in supporting the survival of E. coli cells under heat stress. We conclude that the A. phagocytophilum chaperones might have evolved with distinct biochemical properties to maintain the integrity of pathogenic proteins under unique stress conditions of an intracellular environment of host cells.
Collapse
Affiliation(s)
- Chathurange B Ranaweera
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA
| | - Sunitha Shiva
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA
| | - Swetha Madesh
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Deepika Chauhan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Roman R Ganta
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA; Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Michal Zolkiewski
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
6
|
Read CB, Ali AN, Stephenson DJ, Macknight HP, Maus KD, Cockburn CL, Kim M, Xie X, Carlyon JA, Chalfant CE. Ceramide-1-phosphate is a regulator of Golgi structure and is co-opted by the obligate intracellular bacterial pathogen Anaplasma phagocytophilum. mBio 2024; 15:e0029924. [PMID: 38415594 PMCID: PMC11005342 DOI: 10.1128/mbio.00299-24] [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: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Many intracellular pathogens structurally disrupt the Golgi apparatus as an evolutionarily conserved promicrobial strategy. Yet, the host factors and signaling processes involved are often poorly understood, particularly for Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis. We found that A. phagocytophilum elevated cellular levels of the bioactive sphingolipid, ceramide-1-phosphate (C1P), to promote Golgi fragmentation that enables bacterial proliferation, conversion from its non-infectious to infectious form, and productive infection. A. phagocytophilum poorly infected mice deficient in ceramide kinase, the Golgi-localized enzyme responsible for C1P biosynthesis. C1P regulated Golgi morphology via activation of a PKCα/Cdc42/JNK signaling axis that culminates in phosphorylation of Golgi structural proteins, GRASP55 and GRASP65. siRNA-mediated depletion of Cdc42 blocked A. phagocytophilum from altering Golgi morphology, which impaired anterograde trafficking of trans-Golgi vesicles into and maturation of the pathogen-occupied vacuole. Cells overexpressing phosphorylation-resistant versions of GRASP55 and GRASP65 presented with suppressed C1P- and A. phagocytophilum-induced Golgi fragmentation and poorly supported infection by the bacterium. By studying A. phagocytophilum, we identify C1P as a regulator of Golgi structure and a host factor that is relevant to disease progression associated with Golgi fragmentation.IMPORTANCECeramide-1-phosphate (C1P), a bioactive sphingolipid that regulates diverse processes vital to mammalian physiology, is linked to disease states such as cancer, inflammation, and wound healing. By studying the obligate intracellular bacterium Anaplasma phagocytophilum, we discovered that C1P is a major regulator of Golgi morphology. A. phagocytophilum elevated C1P levels to induce signaling events that promote Golgi fragmentation and increase vesicular traffic into the pathogen-occupied vacuole that the bacterium parasitizes. As several intracellular microbial pathogens destabilize the Golgi to drive their infection cycles and changes in Golgi morphology is also linked to cancer and neurodegenerative disorder progression, this study identifies C1P as a potential broad-spectrum therapeutic target for infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Curtis B. Read
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Anika N. Ali
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Daniel J. Stephenson
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - H. Patrick Macknight
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Kenneth D. Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Chelsea L. Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Xiujie Xie
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Charles E. Chalfant
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Cancer Biology, University of Virginia Cancer Center, Charlottesville, Virginia, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, Virginia, USA
| |
Collapse
|
7
|
Le Dortz LL, Rouxel C, Leroy Q, Ducongé F, Boulouis HJ, Haddad N, Deshuillers PL, Lagrée AC. Aptamer selection against cell extracts containing the zoonotic obligate intracellular bacterium, Anaplasma phagocytophilum. Sci Rep 2024; 14:2465. [PMID: 38291133 PMCID: PMC10828505 DOI: 10.1038/s41598-024-52808-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
A. phagocytophilum is a zoonotic and tick-borne bacterium, threatening human and animal health. Many questions persist concerning the variability of strains and the mechanisms governing the interactions with its different hosts. These gaps can be explained by the difficulty to cultivate and study A. phagocytophilum because of its strict intracellular location and the lack of specific tools, in particular monoclonal antibodies, currently unavailable. The objective of our study was to develop DNA aptamers against A. phagocytophilum, or molecules expressed during the infection, as new study and/or capture tools. Selecting aptamers was a major challenge due to the strict intracellular location of the bacterium. To meet this challenge, we set up a customized selection protocol against an enriched suspension of A. phagocytophilum NY18 strain, cultivated in HL-60 cells. The implementation of SELEX allowed the selection of three aptamers, characterized by a high affinity for HL-60 cells infected with A. phagocytophilum NY18 strain. Interestingly, the targets of these three aptamers are most likely proteins expressed at different times of infection. The selected aptamers could contribute to increase our understanding of the interactions between A. phagocytophilum and its hosts, as well as permit the development of new diagnostic, therapeutic or drug delivery appliances.
Collapse
Affiliation(s)
- Lisa Lucie Le Dortz
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France.
| | - Clotilde Rouxel
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Quentin Leroy
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Frédéric Ducongé
- CEA, Fundamental Research Division (DRF), Institute of Biology François Jacob, Molecular Imaging Research Center, CNRS UMR9199, Paris-Saclay University, 92265, Fontenay-Aux-Roses, France
| | - Henri-Jean Boulouis
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Nadia Haddad
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France.
| | - Pierre Lucien Deshuillers
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Anne-Claire Lagrée
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| |
Collapse
|
8
|
Clemente TM, Angara RK, Gilk SD. Establishing the intracellular niche of obligate intracellular vacuolar pathogens. Front Cell Infect Microbiol 2023; 13:1206037. [PMID: 37645379 PMCID: PMC10461009 DOI: 10.3389/fcimb.2023.1206037] [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: 04/14/2023] [Accepted: 07/21/2023] [Indexed: 08/31/2023] Open
Abstract
Obligate intracellular pathogens occupy one of two niches - free in the host cell cytoplasm or confined in a membrane-bound vacuole. Pathogens occupying membrane-bound vacuoles are sequestered from the innate immune system and have an extra layer of protection from antimicrobial drugs. However, this lifestyle presents several challenges. First, the bacteria must obtain membrane or membrane components to support vacuole expansion and provide space for the increasing bacteria numbers during the log phase of replication. Second, the vacuole microenvironment must be suitable for the unique metabolic needs of the pathogen. Third, as most obligate intracellular bacterial pathogens have undergone genomic reduction and are not capable of full metabolic independence, the bacteria must have mechanisms to obtain essential nutrients and resources from the host cell. Finally, because they are separated from the host cell by the vacuole membrane, the bacteria must possess mechanisms to manipulate the host cell, typically through a specialized secretion system which crosses the vacuole membrane. While there are common themes, each bacterial pathogen utilizes unique approach to establishing and maintaining their intracellular niches. In this review, we focus on the vacuole-bound intracellular niches of Anaplasma phagocytophilum, Ehrlichia chaffeensis, Chlamydia trachomatis, and Coxiella burnetii.
Collapse
Affiliation(s)
| | | | - Stacey D. Gilk
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
9
|
Muñoz-Callejas A, González-Sánchez E, Silván J, San Antonio E, González-Tajuelo R, Ramos-Manzano A, Sánchez-Abad I, González-Alvaro I, García-Pérez J, Tomero EG, de Vicuña RG, Vicente-Rabaneda EF, Castañeda S, Urzainqui A. Low P-Selectin Glycoprotein Ligand-1 Expression in Neutrophils Associates with Disease Activity and Deregulated NET Formation in Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:ijms24076144. [PMID: 37047117 PMCID: PMC10093849 DOI: 10.3390/ijms24076144] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease characterized by the generation of anti-DNA autoantibodies due to exposure of immune cells to excessive amounts of extracellular DNA. Lack of P-selectin in mice induces the development of a lupus-like syndrome and patients with cutaneous lupus have reduced P-selectin expression in skin vessels. Using flow cytometry we analyzed in healthy donors and patients the expression of P-selectin Glycoprotein Ligand-1 (PSGL-1) in circulating neutrophils and the implication of PSGL-1/P-selectin interaction in neutrophil extracellular traps (NETs) generation. We found a statistical significance that neutrophils from active SLE patients have a reduced expression of PSGL-1 and low levels of PSGL-1 in neutrophils from SLE patients associated with the presence of anti-dsDNA antibodies, clinical lung involvement, Raynaud's phenomenon, and positive lupus anticoagulant. PSGL-1 is present along the DNA in the NET. In healthy donors, neutrophil interaction with immobilized P-selectin triggers Syk activation, increases the NETs percentage and reduces the amount of DNA extruded in the NETs. In active SLE patients, neutrophil interaction with P-selectin does not activate Syk or reduce the amount of DNA extruded in the NETs, that might contribute to increase the extracellular level of DNA and hence, to disease pathogenesis.
Collapse
Affiliation(s)
- Antonio Muñoz-Callejas
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Elena González-Sánchez
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Javier Silván
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Esther San Antonio
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Rafael González-Tajuelo
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Alejandra Ramos-Manzano
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Inés Sánchez-Abad
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Isidoro González-Alvaro
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Javier García-Pérez
- Pulmonology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Eva G Tomero
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Rosario García de Vicuña
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Esther F Vicente-Rabaneda
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| | - Santos Castañeda
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
- Rheumatology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
- Catedra UAM-Roche, EPID-Future, Department of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Ana Urzainqui
- Immunology Department, Fundación de Investigación Biomédica (FIB), Instituto de Investigación Sanitaria-Princesa (IIS-Princesa), Hospital de la Princesa, 28006 Madrid, Spain
| |
Collapse
|
10
|
Read CB, Lind MCH, Chiarelli TJ, Izac JR, Adcox HE, Marconi RT, Carlyon JA. The Obligate Intracellular Bacterial Pathogen Anaplasma phagocytophilum Exploits Host Cell Multivesicular Body Biogenesis for Proliferation and Dissemination. mBio 2022; 13:e0296122. [PMID: 36409075 PMCID: PMC9765717 DOI: 10.1128/mbio.02961-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 11/23/2022] Open
Abstract
Anaplasma phagocytophilum is the etiologic agent of the emerging infection, granulocytic anaplasmosis. This obligate intracellular bacterium lives in a host cell-derived vacuole that receives membrane traffic from multiple organelles to fuel its proliferation and from which it must ultimately exit to disseminate infection. Understanding of these essential pathogenic mechanisms has remained poor. Multivesicular bodies (MVBs) are late endosomal compartments that receive biomolecules from other organelles and encapsulate them into intralumenal vesicles (ILVs) using endosomal sorting complexes required for transport (ESCRT) machinery and ESCRT-independent machinery. Association of the ESCRT-independent protein, ALIX, directs MVBs to the plasma membrane where they release ILVs as exosomes. We report that the A. phagocytophilum vacuole (ApV) is acidified and enriched in lysobisphosphatidic acid, a lipid that is abundant in MVBs. ESCRT-0 and ESCRT-III components along with ALIX localize to the ApV membrane. siRNA-mediated inactivation of ESCRT-0 and ALIX together impairs A. phagocytophilum proliferation and infectious progeny production. RNA silencing of ESCRT-III, which regulates ILV scission, pronouncedly reduces ILV formation in ApVs and halts infection by arresting bacterial growth. Rab27a and its effector Munc13-4, which drive MVB trafficking to the plasma membrane and subsequent exosome release, localize to the ApV. Treatment with Nexinhib20, a small molecule inhibitor that specifically targets Rab27a to block MVB exocytosis, abrogates A. phagocytophilum infectious progeny release. Thus, A. phagocytophilum exploits MVB biogenesis and exosome release to benefit each major stage of its intracellular infection cycle: intravacuolar growth, conversion to the infectious form, and exit from the host cell. IMPORTANCE Anaplasma phagocytophilum causes granulocytic anaplasmosis, a globally emerging zoonosis that can be severe, even fatal, and for which antibiotic treatment options are limited. A. phagocytophilum lives in an endosomal-like compartment that interfaces with multiple organelles and from which it must ultimately exit to spread within the host. How the bacterium accomplishes these tasks is poorly understood. Multivesicular bodies (MVBs) are intermediates in the endolysosomal pathway that package biomolecular cargo from other organelles as intralumenal vesicles for release at the plasma membrane as exosomes. We discovered that A. phagocytophilum exploits MVB biogenesis and trafficking to benefit all aspects of its intracellular infection cycle: proliferation, conversion to its infectious form, and release of infectious progeny. The ability of a small molecule inhibitor of MVB exocytosis to impede A. phagocytophilum dissemination indicates the potential of this pathway as a novel host-directed therapeutic target for granulocytic anaplasmosis.
Collapse
Affiliation(s)
- Curtis B. Read
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Mary Clark H. Lind
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Travis J. Chiarelli
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Jerilyn R. Izac
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Haley E. Adcox
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Richard T. Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
11
|
Levin ML, Troughton DR, Loftis AD. Duration of tick attachment necessary for transmission of Anaplasma phagocytophilum by Ixodes scapularis (Acari: Ixodidae) nymphs. Ticks Tick Borne Dis 2021; 12:101819. [PMID: 34520993 DOI: 10.1016/j.ttbdis.2021.101819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/19/2021] [Accepted: 08/19/2021] [Indexed: 11/27/2022]
Abstract
This study assessed the duration of tick attachment necessary for a successful transmission of Anaplasma phagocytophilum by an infected I. scapularis nymph. Individual nymphs were placed upon BALB/c mice and allowed to feed for predetermined time intervals of 4 to 72 h. Ticks removed from mice at predetermined intervals were tested by PCR for verification of infection and evaluation of the bacterial load. The success of pathogen transmission to mice was assessed by blood-PCR at 7, 14 and 21 days postinfestation, and IFA at 21 days postinfestation. Anaplasma phagocytophilum infection was documented in 10-30 % of mice, from which ticks were removed within the first 20 h of feeding. However, transmission success was ≥70% if ticks remained attached for 36 h or longer. Notably, none of the PCR-positive mice that were exposed to infected ticks for 4 to 8 h and only half of PCR-positive mice exposed for 24 h developed antibodies within 3 weeks postinfestation. On the other hand, all mice with detectable bacteremia after being infested for 36 h seroconverted. This suggests that although some of the ticks removed prior to 24 h of attachment succeed in injecting a small amount of A. phagocytophilum, this amount is insufficient for stimulating humoral immunity and perhaps for establishing disseminated infection in BALB/c mice. Although A. phagocytophilum may be present in salivary glands of unfed I. scapularis nymphs, the amount of A. phagocytophilum initially contained in saliva appears insufficient to cause sustainable infection in a host. Replication and, maybe, reactivation of the agent for 12-24 h in a feeding tick is required before a mouse can be consistently infected.
Collapse
Affiliation(s)
- Michael L Levin
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | - Danielle R Troughton
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Amanda D Loftis
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| |
Collapse
|
12
|
Patterson LL, Byerly CD, McBride JW. Anaplasmataceae: Dichotomous Autophagic Interplay for Infection. Front Immunol 2021; 12:642771. [PMID: 33912170 PMCID: PMC8075259 DOI: 10.3389/fimmu.2021.642771] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a vital conserved degradative process that maintains cellular homeostasis by recycling or eliminating dysfunctional cellular organelles and proteins. More recently, autophagy has become a well-recognized host defense mechanism against intracellular pathogens through a process known as xenophagy. On the host-microbe battlefield many intracellular bacterial pathogens have developed the ability to subvert xenophagy to establish infection. Obligately intracellular bacterial pathogens of the Anaplasmataceae family, including Ehrlichia chaffeensis, Anaplasma phaogocytophilium and Orientia tsutsugamushi have developed a dichotomous strategy to exploit the host autophagic pathway to obtain nutrients while escaping lysosomal destruction for intracellular survival within the host cell. In this review, the recent findings regarding how these master manipulators engage and inhibit autophagy for infection are explored. Future investigation to understand mechanisms used by Anaplasmataceae to exploit autophagy may advance novel antimicrobial therapies and provide new insights into how intracellular microbes exploit autophagy to survive.
Collapse
Affiliation(s)
- LaNisha L Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Caitlan D Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States.,Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
13
|
Protein and DNA Biosynthesis Demonstrated in Host Cell-Free Phagosomes Containing Anaplasma phagocytophilum or Ehrlichia chaffeensis in Axenic Media. Infect Immun 2021; 89:IAI.00638-20. [PMID: 33431703 PMCID: PMC8090944 DOI: 10.1128/iai.00638-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/23/2020] [Indexed: 11/20/2022] Open
Abstract
Rickettsiae belong to the Anaplasmataceae family, which includes mostly tick-transmitted pathogens causing human, canine, and ruminant diseases. Biochemical characterization of the pathogens remains a major challenge because of their obligate parasitism. Rickettsiae belong to the Anaplasmataceae family, which includes mostly tick-transmitted pathogens causing human, canine, and ruminant diseases. Biochemical characterization of the pathogens remains a major challenge because of their obligate parasitism. We investigated the use of an axenic medium for growth of two important pathogens—Anaplasma phagocytophilum and Ehrlichia chaffeensis—in host cell-free phagosomes. We recently reported that the axenic medium promotes protein and DNA biosynthesis in host cell-free replicating form of E. chaffeensis, although the bacterial replication is limited. We now tested the hypothesis that growth on axenic medium can be improved if host cell-free rickettsia-containing phagosomes are used. Purification of phagosomes from A. phagocytophilum- and E. chaffeensis-infected host cells was accomplished by density gradient centrifugation combined with magnet-assisted cell sorting. Protein and DNA synthesis was observed for both organisms in cell-free phagosomes with glucose-6-phosphate and/or ATP. The levels of protein and DNA synthesis were the highest for a medium pH of 7. The data demonstrate bacterial DNA and protein synthesis for the first time in host cell-free phagosomes for two rickettsial pathogens. The host cell support-free axenic growth of obligate pathogenic rickettsiae will be critical in advancing research goals in many important tick-borne diseases impacting human and animal health.
Collapse
|
14
|
Naimi WA, Gumpf JJ, Cockburn CL, Camus S, Chalfant CE, Li PL, Carlyon JA. Functional inhibition or genetic deletion of acid sphingomyelinase bacteriostatically inhibits Anaplasma phagocytophilum infection in vivo. Pathog Dis 2021; 79:ftaa072. [PMID: 33220685 PMCID: PMC7787905 DOI: 10.1093/femspd/ftaa072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
Anaplasma phagocytophilum infects neutrophils to cause granulocytic anaplasmosis. It poorly infects mice deficient in acid sphingomyelinase (ASM), a lysosomal enzyme critical for cholesterol efflux, and wild-type mice treated with desipramine that functionally inhibits ASM. Whether inhibition or genetic deletion of ASM is bacteriostatic or bactericidal for A. phagocytophilum and desipramine's ability to lower pathogen burden requires a competent immune system were unknown. Anaplasma phagocytophilum-infected severe combined immunodeficiency disorder (SCID) mice were administered desipramine or PBS, followed by the transfer of blood to naïve wild-type mice. Next, infected wild-type mice were given desipramine or PBS followed by transfer of blood to naïve SCID mice. Finally, wild-type or ASM-deficient mice were infected and blood transferred to naïve SCID mice. The percentage of infected neutrophils was significantly reduced in all desipramine-treated or ASM-deficient mice and in all recipients of blood from these mice. Infection was markedly lower in ASM-deficient and desipramine-treated wild-type mice versus desipramine-treated SCID mice. Yet, infection was never ablated. Thus, ASM activity contributes to optimal A. phagocytophilum infection in vivo, pharmacologic inhibition or genetic deletion of ASM impairs infection in a bacteriostatic and reversible manner and A. phagocytophilum is capable of co-opting ASM-independent lipid sources.
Collapse
Affiliation(s)
- Waheeda A Naimi
- Department of Microbiology and Immunology, Virginia Commonwealth University (VCU) Medical Center, VCU School of Medicine, Richmond, VA, 23398 USA
| | - Jacob J Gumpf
- Department of Microbiology and Immunology, Virginia Commonwealth University (VCU) Medical Center, VCU School of Medicine, Richmond, VA, 23398 USA
| | - Chelsea L Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University (VCU) Medical Center, VCU School of Medicine, Richmond, VA, 23398 USA
| | - Sarah Camus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University (VCU) Medical Center, VCU School of Medicine, Richmond, VA, 23298 USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL,33620 USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
- The Moffitt Cancer Center, Tampa, FL 33620, USA
- Research Service, James A. Haley Veterans' Hospital, Tampa, FL 33612, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University (VCU) Medical Center, VCU School of Medicine, Richmond, VA, 23298 USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University (VCU) Medical Center, VCU School of Medicine, Richmond, VA, 23398 USA
| |
Collapse
|
15
|
Taank V, Ramasamy E, Sultana H, Neelakanta G. An efficient microinjection method to generate human anaplasmosis agent Anaplasma phagocytophilum-infected ticks. Sci Rep 2020; 10:15994. [PMID: 32994497 PMCID: PMC7524789 DOI: 10.1038/s41598-020-73061-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/03/2020] [Indexed: 11/09/2022] Open
Abstract
Ticks are important vectors that transmit several pathogens including human anaplasmosis agent, Anaplasma phagocytophilum. This bacterium is an obligate intracellular rickettsial pathogen. An infected reservoir animal host is often required for maintenance of this bacterial colony and as a source for blood to perform needle inoculations in naïve animals for tick feeding studies. In this study, we report an efficient microinjection method to generate A. phagocytophilum-infected ticks in laboratory conditions. The dense-core (DC) form of A. phagocytophilum was isolated from in vitro cultures and injected into the anal pore of unfed uninfected Ixodes scapularis nymphal ticks. These ticks successfully transmitted A. phagocytophilum to the murine host. The bacterial loads were detected in murine blood, spleen, and liver tissues. In addition, larval ticks successfully acquired A. phagocytophilum from mice that were previously infected by feeding with DC-microinjected nymphal ticks. Transstadial transmission of A. phagocytophilum from larvae to nymphal stage was also evident in these ticks. Taken together, our study provides a timely, rapid, and an efficient method not only to generate A. phagocytophilum-infected ticks but also provides a tool to understand acquisition and transmission dynamics of this bacterium and perhaps other rickettsial pathogens from medically important vectors.
Collapse
Affiliation(s)
- Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Ellango Ramasamy
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA. .,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA. .,Department of Biological Sciences, Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, 23529, USA.
| |
Collapse
|
16
|
Green RS, Izac JR, Naimi WA, O'Bier N, Breitschwerdt EB, Marconi RT, Carlyon JA. Ehrlichia chaffeensis EplA Interaction With Host Cell Protein Disulfide Isomerase Promotes Infection. Front Cell Infect Microbiol 2020; 10:500. [PMID: 33072622 PMCID: PMC7538545 DOI: 10.3389/fcimb.2020.00500] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
Ehrlichia chaffeensis is an obligate intracellular bacterium that invades monocytes to cause the emerging and potentially severe disease, monocytic ehrlichiosis. Ehrlichial invasion of host cells, a process that is essential for the bacterium's survival and pathogenesis, is incompletely understood. In this study, we identified ECH_0377, henceforth designated as EplA (E. chaffeensis PDI ligand A) as an E. chaffeensis adhesin that interacts with host cell protein disulfide isomerase (PDI) to mediate bacterial entry into host cells. EplA is an outer membrane protein that E. chaffeensis expresses during growth in THP-1 monocytic cells. Canine sera confirmed to be positive for exposure to Ehrlichia spp. recognized recombinant EplA, indicating that it is expressed during infection in vivo. EplA antiserum inhibited the bacterium's ability to infect monocytic cells. The EplA-PDI interaction was confirmed via co-immunoprecipitation. Treating host cell surfaces with antibodies that inhibit PDI and/or thioredoxin-1 thiol reductase activity impaired E. chaffeensis infection. Chemical reduction of host cell surfaces, but not bacterial surfaces with tris(2-carboxyethyl)phosphine (TCEP) restored ehrlichial infectivity in the presence of the PDI-neutralizing antibody. Antisera specific for EplA C-terminal residues 95-104 (EplA95−104) or outer membrane protein A amino acids 53-68 (OmpA53−68) reduced E. chaffeensis infection of THP-1 cells. Notably, TCEP rescued ehrlichial infectivity of bacteria that had been treated with anti-EplA95−104, but not anti-EcOmpA53−68. These results demonstrate that EplA contributes to E. chaffeensis infection of monocytic cells by engaging PDI and exploiting the enzyme's reduction of host cell surface disulfide bonds in an EplA C-terminus-dependent manner and identify EplA95−104 and EcOmpA53−68 as novel ehrlichial receptor binding domains.
Collapse
Affiliation(s)
- Ryan S Green
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Jerilyn R Izac
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Waheeda A Naimi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Nathaniel O'Bier
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Edward B Breitschwerdt
- Department of Clinical Sciences and the Intracellular Pathogens Research Laboratory, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Richard T Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| |
Collapse
|
17
|
Immunization against Anaplasma phagocytophilum Adhesin Binding Domains Confers Protection against Infection in the Mouse Model. Infect Immun 2020; 88:IAI.00106-20. [PMID: 32661123 DOI: 10.1128/iai.00106-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 11/20/2022] Open
Abstract
Anaplasma phagocytophilum causes granulocytic anaplasmosis, a debilitating infection that can be fatal in the immunocompromised. It also afflicts animals, including dogs, horses, and sheep. No granulocytic anaplasmosis vaccine exists. Because A. phagocytophilum is an obligate intracellular bacterium, inhibiting microbe-host cell interactions that facilitate invasion can disrupt infection. The binding domains of A. phagocytophilum adhesins A. phagocytophilum invasion protein A (AipA), A. phagocytophilum surface protein (Asp14), and outer membrane protein A (OmpA) are essential for optimal bacterial entry into host cells, but their relevance to infection in vivo is undefined. In this study, C57BL/6 mice were immunized with a cocktail of keyhole limpet hemocyanin-conjugated peptides corresponding to the AipA, Asp14, and OmpA binding domains in alum followed by challenge with A. phagocytophilum The bacterial peripheral blood burden was pronouncedly reduced in immunized mice compared to controls. Examination of pre- and postchallenge sera from these mice revealed that immunization elicited antibodies against AipA and Asp14 peptides but not OmpA peptide. Nonetheless, pooled sera from pre- and postchallenge groups, but not from control groups, inhibited A. phagocytophilum infection of HL-60 cells. Adhesin domain immunization also elicited interferon gamma (IFN-γ)-producing CD8-positive (CD8+) T cells. A follow-up study confirmed that immunization against only the AipA or Asp14 binding domain was sufficient to reduce the bacterial peripheral blood load in mice following challenge and elicit antibodies that inhibit A. phagocytophilum cellular infection in vitro These data demonstrate that AipA and Asp14 are critical for A. phagocytophilum to productively infect mice, and immunization against their binding domains elicits a protective immune response.
Collapse
|
18
|
Green RS, Naimi WA, Oliver LD, O'Bier N, Cho J, Conrad DH, Martin RK, Marconi RT, Carlyon JA. Binding of Host Cell Surface Protein Disulfide Isomerase by Anaplasma phagocytophilum Asp14 Enables Pathogen Infection. mBio 2020; 11:e03141-19. [PMID: 31992623 PMCID: PMC6989111 DOI: 10.1128/mbio.03141-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 11/20/2022] Open
Abstract
Diverse intracellular pathogens rely on eukaryotic cell surface disulfide reductases to invade host cells. Pharmacologic inhibition of these enzymes is cytotoxic, making it impractical for treatment. Identifying and mechanistically dissecting microbial proteins that co-opt surface reductases could reveal novel targets for disrupting this common infection strategy. Anaplasma phagocytophilum invades neutrophils by an incompletely defined mechanism to cause the potentially fatal disease granulocytic anaplasmosis. The bacterium's adhesin, Asp14, contributes to invasion by virtue of its C terminus engaging an unknown receptor. Yeast-two hybrid analysis identified protein disulfide isomerase (PDI) as an Asp14 binding partner. Coimmunoprecipitation confirmed the interaction and validated it to be Asp14 C terminus dependent. PDI knockdown and antibody-mediated inhibition of PDI reductase activity impaired A. phagocytophilum infection of but not binding to host cells. Infection during PDI inhibition was rescued when the bacterial but not host cell surface disulfide bonds were chemically reduced with tris(2-carboxyethyl)phosphine-HCl (TCEP). TCEP also restored bacterial infectivity in the presence of an Asp14 C terminus blocking antibody that otherwise inhibits infection. A. phagocytophilum failed to productively infect myeloid-specific-PDI conditional-knockout mice, marking the first demonstration of in vivo microbial dependency on PDI for infection. Mutational analyses identified the Asp14 C-terminal residues that are critical for binding PDI. Thus, Asp14 binds and brings PDI proximal to A. phagocytophilum surface disulfide bonds that it reduces, which enables cellular and in vivo infection.IMPORTANCEAnaplasma phagocytophilum infects neutrophils to cause granulocytic anaplasmosis, an emerging potentially fatal disease and the second-most common tick-borne illness in the United States. Treatment options are limited, and no vaccine exists. Due to the bacterium's obligatory intracellular lifestyle, A. phagocytophilum survival and pathogenesis are predicated on its ability to enter host cells. Understanding its invasion mechanism will yield new targets for preventing bacterial entry and, hence, disease. We report a novel entry pathway in which the A. phagocytophilum outer membrane protein Asp14 binds host cell surface protein disulfide isomerase via specific C-terminal residues to promote reduction of bacterial surface disulfide bonds, which is critical for cellular invasion and productive infection in vivo Targeting the Asp14 C terminus could be used to prevent/treat granulocytic anaplasmosis. Our findings have broad implications, as a thematically similar approach could be applied to block infection by other intracellular microbes that exploit cell surface reductases.
Collapse
Affiliation(s)
- Ryan S Green
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Waheeda A Naimi
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Lee D Oliver
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Nathaniel O'Bier
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Jaehyung Cho
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Richard T Marconi
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| |
Collapse
|
19
|
Cockburn CL, Green RS, Damle SR, Martin RK, Ghahrai NN, Colonne PM, Fullerton MS, Conrad DH, Chalfant CE, Voth DE, Rucks EA, Gilk SD, Carlyon JA. Functional inhibition of acid sphingomyelinase disrupts infection by intracellular bacterial pathogens. Life Sci Alliance 2019; 2:e201800292. [PMID: 30902833 PMCID: PMC6431796 DOI: 10.26508/lsa.201800292] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022] Open
Abstract
Intracellular bacteria that live in host cell-derived vacuoles are significant causes of human disease. Parasitism of low-density lipoprotein (LDL) cholesterol is essential for many vacuole-adapted bacteria. Acid sphingomyelinase (ASM) influences LDL cholesterol egress from the lysosome. Using functional inhibitors of ASM (FIASMAs), we show that ASM activity is key for infection cycles of vacuole-adapted bacteria that target cholesterol trafficking-Anaplasma phagocytophilum, Coxiella burnetii, Chlamydia trachomatis, and Chlamydia pneumoniae. Vacuole maturation, replication, and infectious progeny generation by A. phagocytophilum, which exclusively hijacks LDL cholesterol, are halted and C. burnetii, for which lysosomal cholesterol accumulation is bactericidal, is killed by FIASMAs. Infection cycles of Chlamydiae, which hijack LDL cholesterol and other lipid sources, are suppressed but less so than A. phagocytophilum or C. burnetii A. phagocytophilum fails to productively infect ASM-/- or FIASMA-treated mice. These findings establish the importance of ASM for infection by intracellular bacteria and identify FIASMAs as potential host-directed therapies for diseases caused by pathogens that manipulate LDL cholesterol.
Collapse
Affiliation(s)
- Chelsea L Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Ryan S Green
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Sheela R Damle
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Naomi N Ghahrai
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Punsiri M Colonne
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Marissa S Fullerton
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Daniel E Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Elizabeth A Rucks
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stacey D Gilk
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| |
Collapse
|
20
|
Oliva Chávez AS, Herron MJ, Nelson CM, Felsheim RF, Oliver JD, Burkhardt NY, Kurtti TJ, Munderloh UG. Mutational analysis of gene function in the Anaplasmataceae: Challenges and perspectives. Ticks Tick Borne Dis 2018; 10:482-494. [PMID: 30466964 DOI: 10.1016/j.ttbdis.2018.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/13/2018] [Accepted: 11/09/2018] [Indexed: 01/10/2023]
Abstract
Mutational analysis is an efficient approach to identifying microbial gene function. Until recently, lack of an effective tool for Anaplasmataceae yielding reproducible results has created an obstacle to functional genomics, because surrogate systems, e.g., ectopic gene expression and analysis in E. coli, may not provide accurate answers. We chose to focus on a method for high-throughput generation of mutants via random mutagenesis as opposed to targeted gene inactivation. In our search for a suitable mutagenesis tool, we considered attributes of the Himar1 transposase system, i.e., random insertion into AT dinucleotide sites, which are abundant in Anaplasmataceae, and lack of requirement for specific host factors. We chose the Anaplasma marginale tr promoter, and the clinically irrelevant antibiotic spectinomycin for selection, and in addition successfully implemented non-antibiotic selection using an herbicide resistance gene. These constructs function reasonably well in Anaplasma phagocytophilum harvested from human promyelocyte HL-60 cells or Ixodes scapularis tick cells. We describe protocols developed in our laboratory, and discuss what likely makes them successful. What makes Anaplasmataceae electroporation competent is unknown and manipulating electroporation conditions has not improved mutational efficiency. A concerted effort is needed to resolve remaining problems that are inherent to the obligate intracellular bacteria. Finally, using this approach, we describe the discovery and characterization of a putative secreted effector necessary for Ap survival in HL-60 cells.
Collapse
Affiliation(s)
- Adela S Oliva Chávez
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Michael J Herron
- Department of Entomology, University of Minnesota, St. Paul, MN, USA
| | - Curtis M Nelson
- Department of Entomology, University of Minnesota, St. Paul, MN, USA
| | | | - Jonathan D Oliver
- School of Public Health, Division of Environmental Health Sciences, University of Minnesota, Minneapolis, MN, USA
| | | | - Timothy J Kurtti
- Department of Entomology, University of Minnesota, St. Paul, MN, USA
| | | |
Collapse
|
21
|
Nguyen Trong TP, Wang J, Majzoub-Altweck M, Fell S, Straubinger RK. Recovering host cell-free Anaplasma phagocytophilum from HL-60 cells by using rock tumbler grit in comparison to the syringe lysis method. Ticks Tick Borne Dis 2018; 10:280-285. [PMID: 30455057 DOI: 10.1016/j.ttbdis.2018.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/26/2018] [Accepted: 11/08/2018] [Indexed: 12/25/2022]
Abstract
Anaplasma phagocytophilum (Ap) is a tick-transmitted obligate intracellular bacterium and the causative agent of the granulocytic anaplasmosis in various species of domestic animals and in humans. During intracellular development Ap transforms from a dense-cored cell form into a reticulate cell form and vice versa. For isolation of intracellular bacteria, a range of different purification methods is used. However, unlike other Gram-negative bacteria Ap is considered to be sensitive to mechanical stress and osmolarity changes. An updated semi-purification method using rock tumbler grit is introduced here to increase the outcome of bacteria and to facilitate the procedure of host cell lysis. The objective of this study was to evaluate the structural integrity and infectivity of Ap after lysis of the host cells using rock tumbler grit and to compare the outcome to that of the frequently used method, syringe lysis. Human promyelocytic leukemia cell lines (HL-60) were infected with Ap and following host cell-free bacteria were assessed by transmission electron microscopy. The outcome of the different purification methods was compared using live/dead-staining based on immunofluorescence to count the number of viable bacteria and real-time PCR to compare the amount of DNA. Subsequently the isolated bacteria were tested to infect naive cell cultures. We observed that both Ap dense-cored cells and reticulate cells are preserved intact after the application of rock tumbler grit. The number of viable, host cell-free bacteria was higher by factor 1.7-2.4 compared to the syringe lysis protocol. Quantitative analysis based on real-time PCR showed an increase of bacterial DNA up to 1.6-2.9 times higher using the rock tumbler grit protocol. Bacteria released from the same number of infected host cells were used for new infections. Flow cytometric analysis of the cell cultures confirmed that the number of Ap organisms recovered by using the rock tumbler grit protocol resulted in higher infection rates than the number of Ap organisms recovered by using syringe lysis protocol. Our observations indicate that the rock tumbler grit protocol can be applied as a safe, robust and convenient method to recover Ap compared to syringe lysis.
Collapse
Affiliation(s)
- Thu Phong Nguyen Trong
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Faculty of Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539, Munich, Germany.
| | - Jinyong Wang
- Department of Microbiology & Immunology Chicago Medical School Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road 2-407, North Chicago, IL, 60064, USA.
| | - Monir Majzoub-Altweck
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine Faculty of Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539, Munich, Germany.
| | - Shari Fell
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Faculty of Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539, Munich, Germany.
| | - Reinhard K Straubinger
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Faculty of Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539, Munich, Germany.
| |
Collapse
|
22
|
Differential Susceptibility of Male Versus Female Laboratory Mice to Anaplasma phagocytophilum Infection. Trop Med Infect Dis 2018; 3:tropicalmed3030078. [PMID: 30274474 PMCID: PMC6161277 DOI: 10.3390/tropicalmed3030078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/07/2018] [Accepted: 07/18/2018] [Indexed: 12/20/2022] Open
Abstract
Human granulocytic anaplasmosis (HGA) is a debilitating, non-specific febrile illness caused by the granulocytotropic obligate intracellular bacterium called Anaplasma phagocytophilum. Surveillance studies indicate a higher prevalence of HGA in male versus female patients. Whether this discrepancy correlates with differential susceptibility of males and females to A. phagocytophilum infection is unknown. Laboratory mice have long been used to study granulocytic anaplasmosis. Yet, sex as a biological variable (SABV) in this model has not been evaluated. In this paper, groups of male and female C57Bl/6 mice that had been infected with A. phagocytophilum were assessed for the bacterial DNA load in the peripheral blood, the percentage of neutrophils harboring bacterial inclusions called morulae, and splenomegaly. Infected male mice exhibited as much as a 1.85-fold increase in the number of infected neutrophils, which is up to a 1.88-fold increase in the A. phagocytophilum DNA load, and a significant increase in spleen size when compared to infected female mice. The propensity of male mice to develop a higher level of A. phagocytophilum infection is relevant for studies utilizing the mouse model. This stresses the importance of including SABV and aligns with the observed higher incidence of infection in male versus female patients.
Collapse
|
23
|
Otten C, Brilli M, Vollmer W, Viollier PH, Salje J. Peptidoglycan in obligate intracellular bacteria. Mol Microbiol 2018; 107:142-163. [PMID: 29178391 PMCID: PMC5814848 DOI: 10.1111/mmi.13880] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2017] [Indexed: 01/08/2023]
Abstract
Peptidoglycan is the predominant stress-bearing structure in the cell envelope of most bacteria, and also a potent stimulator of the eukaryotic immune system. Obligate intracellular bacteria replicate exclusively within the interior of living cells, an osmotically protected niche. Under these conditions peptidoglycan is not necessarily needed to maintain the integrity of the bacterial cell. Moreover, the presence of peptidoglycan puts bacteria at risk of detection and destruction by host peptidoglycan recognition factors and downstream effectors. This has resulted in a selective pressure and opportunity to reduce the levels of peptidoglycan. In this review we have analysed the occurrence of genes involved in peptidoglycan metabolism across the major obligate intracellular bacterial species. From this comparative analysis, we have identified a group of predicted 'peptidoglycan-intermediate' organisms that includes the Chlamydiae, Orientia tsutsugamushi, Wolbachia and Anaplasma marginale. This grouping is likely to reflect biological differences in their infection cycle compared with peptidoglycan-negative obligate intracellular bacteria such as Ehrlichia and Anaplasma phagocytophilum, as well as obligate intracellular bacteria with classical peptidoglycan such as Coxiella, Buchnera and members of the Rickettsia genus. The signature gene set of the peptidoglycan-intermediate group reveals insights into minimal enzymatic requirements for building a peptidoglycan-like sacculus and/or division septum.
Collapse
Affiliation(s)
- Christian Otten
- The Centre for Bacterial Cell BiologyInstitute for Cell and Molecular Biosciences, Newcastle UniversityNewcastle upon TyneNE2 4AXUK
| | - Matteo Brilli
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE)University of Padova. Agripolis ‐ V.le dell'Università, 16 | 35020 Legnaro PadovaItaly
- Present address:
Department of BiosciencesUniversity of Milan, via Celoria 26(MI)Italy
| | - Waldemar Vollmer
- The Centre for Bacterial Cell BiologyInstitute for Cell and Molecular Biosciences, Newcastle UniversityNewcastle upon TyneNE2 4AXUK
| | - Patrick H. Viollier
- Department of Microbiology and Molecular MedicineInstitute of Genetics & Genomics in Geneva (iGE3), University of GenevaGenevaSwitzerland
| | - Jeanne Salje
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global HealthUniversity of OxfordOxfordUK
- Mahidol‐Oxford Tropical Medicine Research UnitMahidol UniversityBangkokThailand
| |
Collapse
|
24
|
Damle SR, Martin RK, Cockburn CL, Lownik JC, Carlyon JA, Smith AD, Conrad DH. ADAM10 and Notch1 on murine dendritic cells control the development of type 2 immunity and IgE production. Allergy 2018; 73:125-136. [PMID: 28745029 DOI: 10.1111/all.13261] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND Allergy and allergic asthma are significant health burdens in developed countries and are increasing in prevalence. Dendritic cells (DCs) initiate immune responses to common aeroallergens, and ADAM10 has been demonstrated to be important for the development of adaptive responses. This study's objective was to understand the role of ADAM10 on DCs in the development of allergic and anaphylactic responses. METHODS In this study, we used mouse models of allergic airway inflammation (house dust mice and Alternaria alternata) and OVA-induced models of active anaphylaxis to determine the DC-specific function of ADAM10 and Notch signaling. To examine TH 1 and TH 17 immunity infection with Anaplasma phagocytophilum and Citrobacter rodentium respectively, were used. RESULTS Mice, which have ADAM10 deleted from DCs, have dramatic reductions in IgE production and do not develop significant TH 2 immune responses. Further, ADAM10DC-/- mice are resistant to IgE-mediated anaphylaxis. This response is selective for TH 2 immunity as TH 1 and TH 17 immunity is largely unaffected. Notch1, a known ADAM10 substrate, when knocked out of DCs (Notch1DC-/- ) demonstrated a similar reduction in anaphylaxis and IgE. Without ADAM10 and Notch1 signaling, DCs were unable to make cytokines that stimulate TH 2 cells and cytokines. Anaphylaxis and allergic lung inflammation were restored in ADAM10DC-/- with the overexpression of the Notch1-intracellular domain, confirming the role of Notch signaling. CONCLUSIONS Targeting ADAM10 and Notch1 on DCs represent a novel strategy for modulating TH 2 immune responses and IgE production.
Collapse
Affiliation(s)
- S. R. Damle
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - R. K. Martin
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - C. L. Cockburn
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - J. C. Lownik
- Center for Clinical and Translational Research Virginia Commonwealth University Richmond VA USA
| | - J. A. Carlyon
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - A. D. Smith
- United States Department of Agriculture Diet, Genomics, and Immunology Laboratory, Agricultural Research Service Beltsville Human Nutrition Research Center Beltsville MD USA
| | - D. H. Conrad
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| |
Collapse
|
25
|
Abstract
Human ehrlichiosis and anaplasmosis are acute febrile tick-borne infectious diseases caused by various members from the genera Ehrlichia and Anaplasma. Ehrlichia chaffeensis is the major etiologic agent of human monocytotropic ehrlichiosis (HME), while Anaplasma phagocytophilum is the major cause of human granulocytic anaplasmosis (HGA). The clinical manifestations of HME and HGA ranges from subclinical to potentially life-threatening diseases associated with multi-organ failure. Macrophages and neutrophils are the major target cells for Ehrlichia and Anaplasma, respectively. The threat to public health is increasing with newly emerging ehrlichial and anaplasma agents, yet vaccines for human ehrlichioses and anaplasmosis are not available, and therapeutic options are limited. This article reviews recent advances in the understanding of HME and HGA.
Collapse
|
26
|
Oki AT, Huang B, Beyer AR, May LJ, Truchan HK, Walker NJ, Galloway NL, Borjesson DL, Carlyon JA. Anaplasma phagocytophilum APH0032 Is Exposed on the Cytosolic Face of the Pathogen-Occupied Vacuole and Co-opts Host Cell SUMOylation. Front Cell Infect Microbiol 2016; 6:108. [PMID: 27713867 PMCID: PMC5031783 DOI: 10.3389/fcimb.2016.00108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/05/2016] [Indexed: 12/31/2022] Open
Abstract
Anaplasma phagocytophilum, a member of the family Anaplasmataceae and the obligate intracellular bacterium that causes granulocytic anaplasmosis, resides in a host cell-derived vacuole. Bacterial proteins that localize to the A. phagocytophilum-occupied vacuole membrane (AVM) are critical host-pathogen interfaces. Of the few bacterial AVM proteins that have been identified, the domains responsible for AVM localization and the host cell pathways that they co-opt are poorly defined. APH0032 is an effector that is expressed and localizes to the AVM late during the infection cycle. Herein, the APH0032 domain that is essential for associating with host cell membranes was mapped. Immunofluorescent labeling of infected cells that had been differentially permeabilized confirmed that APH0032 is exposed on the AVM's cytosolic face, signifying its potential to interface with host cell processes. SUMOylation is the covalent attachment of a member of the small ubiquitin-like modifier (SUMO) family of proteins to lysines in target substrates. Previous work from our laboratory determined that SUMOylation is important for A. phagocytophilum survival and that SUMOylated proteins decorate the AVM. Algorithmic prediction analyses identified APH0032 as a candidate for SUMOylation. Endogenous APH0032 was precipitated from infected cells using a SUMO affinity matrix, confirming that the effector co-opts SUMOylation during infection. APH0032 pronouncedly colocalized with SUMO1, but not SUMO2/3 moieties on the AVM. Ectopic expression of APH0032 in A. phagocytophilum infected host cells significantly boosted the bacterial load. This study delineates the first domain of any Anaplasmataceae protein that is essential for associating with the pathogen-occupied vacuole membrane, demonstrates the importance of APH0032 to infection, and identifies it as the second A. phagocytophilum effector that co-opts SUMOylation, thus underscoring the relevance of this post-translational modification to infection.
Collapse
Affiliation(s)
- Aminat T Oki
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine Richmond, VA, USA
| | - Bernice Huang
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine Richmond, VA, USA
| | - Andrea R Beyer
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine Richmond, VA, USA
| | - Levi J May
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine Richmond, VA, USA
| | - Hilary K Truchan
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine Richmond, VA, USA
| | - Naomi J Walker
- Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine Davis, CA, USA
| | - Nathan L Galloway
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine Richmond, VA, USA
| | - Dori L Borjesson
- Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine Davis, CA, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine Richmond, VA, USA
| |
Collapse
|
27
|
Truchan HK, Cockburn CL, May LJ, VieBrock L, Carlyon JA. Anaplasma phagocytophilum-Occupied Vacuole Interactions with the Host Cell Cytoskeleton. Vet Sci 2016; 3:vetsci3030025. [PMID: 29056733 PMCID: PMC5606578 DOI: 10.3390/vetsci3030025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023] Open
Abstract
Anaplasma phagocytophilum is an obligate intracellular bacterial pathogen of humans and animals. The A. phagocytophium-occupied vacuole (ApV) is a critical host-pathogen interface. Here, we report that the intermediate filaments, keratin and vimentin, assemble on the ApV early and remain associated with the ApV throughout infection. Microtubules localize to the ApV to a lesser extent. Vimentin, keratin-8, and keratin-18 but not tubulin expression is upregulated in A. phagocytophilum infected cells. SUMO-2/3 but not SUMO-1 colocalizes with vimentin filaments that surround ApVs. PolySUMOylation of vimentin by SUMO-2/3 but not SUMO-1 decreases vimentin solubility. Consistent with this, more vimentin exists in an insoluble state in A. phagocytophilum infected cells than in uninfected cells. Knocking down the SUMO-conjugating enzyme, Ubc9, abrogates vimentin assembly at the ApV but has no effect on the bacterial load. Bacterial protein synthesis is dispensable for maintaining vimentin and SUMO-2/3 at the ApV. Withaferin A, which inhibits soluble vimentin, reduces vimentin recruitment to the ApV, optimal ApV formation, and the bacterial load when administered prior to infection but is ineffective once vimentin has assembled on the ApV. Thus, A. phagocytophilum modulates cytoskeletal component expression and co-opts polySUMOylated vimentin to aid construction of its vacuolar niche and promote optimal survival.
Collapse
Affiliation(s)
- Hilary K Truchan
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA.
| | - Chelsea L Cockburn
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA.
| | - Levi J May
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA.
| | - Lauren VieBrock
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA.
| | - Jason A Carlyon
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA.
| |
Collapse
|
28
|
PSGL-1 on Leukocytes is a Critical Component of the Host Immune Response against Invasive Pneumococcal Disease. PLoS Pathog 2016; 12:e1005500. [PMID: 26975045 PMCID: PMC4790886 DOI: 10.1371/journal.ppat.1005500] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/19/2016] [Indexed: 12/21/2022] Open
Abstract
Bacterial uptake by phagocytic cells is a vital event in the clearance of invading pathogens such as Streptococcus pneumoniae. A major role of the P-selectin glycoprotein ligand-1 (PSGL-1) on leukocytes against invasive pneumococcal disease is described in this study. Phagocytosis experiments using different serotypes demonstrated that PSGL-1 is involved in the recognition, uptake and killing of S. pneumoniae. Co-localization of several clinical isolates of S. pneumoniae with PSGL-1 was demonstrated, observing a rapid and active phagocytosis in the presence of PSGL-1. Furthermore, the pneumococcal capsular polysaccharide and the main autolysin of the bacterium ―the amidase LytA― were identified as bacterial ligands for PSGL-1. Experimental models of pneumococcal disease including invasive pneumonia and systemic infection showed that bacterial levels were markedly increased in the blood of PSGL-1−/− mice. During pneumonia, PSGL-1 controls the severity of pneumococcal dissemination from the lung to the bloodstream. In systemic infection, a major role of PSGL-1 in host defense is to clear the bacteria in the systemic circulation controlling bacterial replication. These results confirmed the importance of this receptor in the recognition and clearance of S. pneumoniae during invasive pneumococcal disease. Histological and cellular analysis demonstrated that PSGL-1−/− mice have increased levels of T cells migrating to the lung than the corresponding wild-type mice. In contrast, during systemic infection, PSGL-1−/− mice had increased numbers of neutrophils and macrophages in blood, but were less effective controlling the infection process due to the lack of this functional receptor. Overall, this study demonstrates that PSGL-1 is a novel receptor for S. pneumoniae that contributes to protection against invasive pneumococcal disease. S. pneumoniae is one of the most important and devastating human pathogens worldwide, mainly affecting young children, elderly people and immunocompromised patients. In terms of host immune defense against invasive pneumococcal isolates, professional phagocytes require receptor-mediated recognition of certain ligands on the bacterial surface for the uptake and clearance of the microorganism. In this study, we demonstrate that the P-selectin glycoprotein ligand-1 (PSGL-1) on leukocytes is involved in the phagocytosis process of S. pneumoniae by targeting the capsule and the surface protein LytA as pathogen-associated molecular patterns. To explore this process in more detail, we have used wild-type mice and mice deficient in PSGL-1 demonstrating that lack of PSGL-1 is detrimental for the host by increasing the susceptibility to the infection and the severity of the pneumococcal invasive disease. Overall, these data show the importance of PSGL-1 on leukocytes in host defense against S. pneumoniae and confirm that PSGL-1 plays a critical protective role against invasive bacterial disease.
Collapse
|
29
|
Truchan HK, Cockburn CL, Hebert KS, Magunda F, Noh SM, Carlyon JA. The Pathogen-Occupied Vacuoles of Anaplasma phagocytophilum and Anaplasma marginale Interact with the Endoplasmic Reticulum. Front Cell Infect Microbiol 2016; 6:22. [PMID: 26973816 PMCID: PMC4771727 DOI: 10.3389/fcimb.2016.00022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/08/2016] [Indexed: 11/13/2022] Open
Abstract
The genus Anaplasma consists of tick-transmitted obligate intracellular bacteria that invade white or red blood cells to cause debilitating and potentially fatal infections. A. phagocytophilum, a human and veterinary pathogen, infects neutrophils to cause granulocytic anaplasmosis. A. marginale invades bovine erythrocytes. Evidence suggests that both species may also infect endothelial cells in vivo. In mammalian and arthropod host cells, A. phagocytophilum and A. marginale reside in host cell derived pathogen-occupied vacuoles (POVs). While it was recently demonstrated that the A. phagocytophilum-occupied vacuole (ApV) intercepts membrane traffic from the trans-Golgi network, it is unclear if it or the A. marginale-occupied vacuole (AmV) interacts with other secretory organelles. Here, we demonstrate that the ApV and AmV extensively interact with the host endoplasmic reticulum (ER) in endothelial, myeloid, and/or tick cells. ER lumen markers, calreticulin, and protein disulfide isomerase, and the ER membrane marker, derlin-1, were pronouncedly recruited to the peripheries of both POVs. ApV association with the ER initiated early and continued throughout the infection cycle. Both the ApV and AmV interacted with the rough ER and smooth ER. However, only derlin-1-positive rough ER derived vesicles were delivered into the ApV lumen where they localized with intravacuolar bacteria. Transmission electron microscopy identified multiple ER-POV membrane contact sites on the cytosolic faces of both species' vacuoles that corresponded to areas on the vacuoles' lumenal faces where intravacuolar Anaplasma organisms closely associated. A. phagocytophilum is known to hijack Rab10, a GTPase that regulates ER dynamics and morphology. Yet, ApV-ER interactions were unhindered in cells in which Rab10 had been knocked down, demonstrating that the GTPase is dispensable for the bacterium to parasitize the ER. These data establish the ApV and AmV as pathogen-host interfaces that directly engage the ER in vertebrate and invertebrate host cells and evidence the conservation of ER parasitism between two Anaplasma species.
Collapse
Affiliation(s)
- Hilary K Truchan
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| | - Chelsea L Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| | - Kathryn S Hebert
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| | - Forgivemore Magunda
- Program in Vector Borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State UniversityPullman, WA, USA; The Paul G. Allen School for Global Animal Health, Washington State UniversityPullman, WA, USA
| | - Susan M Noh
- Program in Vector Borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State UniversityPullman, WA, USA; Animal Disease Research Unit, Agricultural Research Service, U. S. Department of AgriculturePullman, WA, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| |
Collapse
|
30
|
Truchan HK, VieBrock L, Cockburn CL, Ojogun N, Griffin BP, Wijesinghe DS, Chalfant CE, Carlyon JA. Anaplasma phagocytophilum Rab10-dependent parasitism of the trans-Golgi network is critical for completion of the infection cycle. Cell Microbiol 2016; 18:260-81. [PMID: 26289115 PMCID: PMC4891814 DOI: 10.1111/cmi.12500] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 08/03/2015] [Accepted: 08/18/2015] [Indexed: 02/01/2023]
Abstract
Anaplasma phagocytophilum is an emerging human pathogen and obligate intracellular bacterium. It inhabits a host cell-derived vacuole and cycles between replicative reticulate cell (RC) and infectious dense-cored (DC) morphotypes. Host-pathogen interactions that are critical for RC-to-DC conversion are undefined. We previously reported that A. phagocytophilum recruits green fluorescent protein (GFP)-tagged Rab10, a GTPase that directs exocytic traffic from the sphingolipid-rich trans-Golgi network (TGN) to its vacuole in a guanine nucleotide-independent manner. Here, we demonstrate that endogenous Rab10-positive TGN vesicles are not only routed to but also delivered into the A. phagocytophilum-occupied vacuole (ApV). Consistent with this finding, A. phagocytophilum incorporates sphingolipids while intracellular and retains them when naturally released from host cells. TGN vesicle delivery into the ApV is Rab10 dependent, up-regulates expression of the DC-specific marker, APH1235, and is critical for the production of infectious progeny. The A. phagocytophilum surface protein, uridine monophosphate kinase, was identified as a guanine nucleotide-independent, Rab10-specific ligand. These data delineate why Rab10 is important for the A. phagocytophilum infection cycle and expand the understanding of the benefits that exploiting host cell membrane traffic affords intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Hilary K. Truchan
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Lauren VieBrock
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Chelsea L. Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Nore Ojogun
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Brian P. Griffin
- Molecular Biology and Genetics Program, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Dayanjan S. Wijesinghe
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Charles E. Chalfant
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- The Victoria Johnson Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Institute for Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Research and Development, Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Molecular Biology and Genetics Program, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
31
|
Oliva Chávez AS, Fairman JW, Felsheim RF, Nelson CM, Herron MJ, Higgins L, Burkhardt NY, Oliver JD, Markowski TW, Kurtti TJ, Edwards TE, Munderloh UG. An O-Methyltransferase Is Required for Infection of Tick Cells by Anaplasma phagocytophilum. PLoS Pathog 2015; 11:e1005248. [PMID: 26544981 PMCID: PMC4636158 DOI: 10.1371/journal.ppat.1005248] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 10/03/2015] [Indexed: 12/16/2022] Open
Abstract
Anaplasma phagocytophilum, the causative agent of Human Granulocytic Anaplasmosis (HGA), is an obligately intracellular α-proteobacterium that is transmitted by Ixodes spp ticks. However, the pathogen is not transovarially transmitted between tick generations and therefore needs to survive in both a mammalian host and the arthropod vector to complete its life cycle. To adapt to different environments, pathogens rely on differential gene expression as well as the modification of proteins and other molecules. Random transposon mutagenesis of A. phagocytophilum resulted in an insertion within the coding region of an o-methyltransferase (omt) family 3 gene. In wild-type bacteria, expression of omt was up-regulated during binding to tick cells (ISE6) at 2 hr post-inoculation, but nearly absent by 4 hr p.i. Gene disruption reduced bacterial binding to ISE6 cells, and the mutant bacteria that were able to enter the cells were arrested in their replication and development. Analyses of the proteomes of wild-type versus mutant bacteria during binding to ISE6 cells identified Major Surface Protein 4 (Msp4), but also hypothetical protein APH_0406, as the most differentially methylated. Importantly, two glutamic acid residues (the targets of the OMT) were methyl-modified in wild-type Msp4, whereas a single asparagine (not a target of the OMT) was methylated in APH_0406. In vitro methylation assays demonstrated that recombinant OMT specifically methylated Msp4. Towards a greater understanding of the overall structure and catalytic activity of the OMT, we solved the apo (PDB_ID:4OA8), the S-adenosine homocystein-bound (PDB_ID:4OA5), the SAH-Mn2+ bound (PDB_ID:4PCA), and SAM- Mn2+ bound (PDB_ID:4PCL) X-ray crystal structures of the enzyme. Here, we characterized a mutation in A. phagocytophilum that affected the ability of the bacteria to productively infect cells from its natural vector. Nevertheless, due to the lack of complementation, we cannot rule out secondary mutations. Since its discovery in 1994, Human Granulocytic Anaplasmosis (HGA) has become the second most commonly diagnosed tick-borne disease in the US, and it is gaining importance in several countries in Europe. HGA is caused by Anaplasma phagocytophilum, a bacterium transmitted by black-legged ticks and their relatives. Whereas several of the molecules and processes leading to infection of human cells have been identified, little is known about their counterparts in the tick. We analyzed the effects of a mutation in a gene encoding an o-methyltransferase that is involved in methylation of an outer membrane protein. The mutation of the OMT appears to be important for the ability of A. phagocytophilum to adhere to, invade, and replicate in tick cells. Several tests including binding assays, microscopic analysis of the infection cycle within tick cells, gene expression assays, and biochemical assays using recombinant OMT strongly suggested that the mutation of the o-methyltransferase gene arrested the growth and development of this bacterium within tick cells. Proteomic analyses identified several possible OMT substrates, and in vitro methylation assays using recombinant o-methyltransferase identified an outer membrane protein, Msp4, as a specifically methyl-modified target. Our results indicated that methylation was important for infection of tick cells by A. phagocytophilum, and suggested possible strategies to block transmission of this emerging pathogen. The solved crystal structure of the o-methyltransferase will further stimulate the search for small molecule inhibitors that could break the tick transmission cycle of A. phagocytophilum in nature.
Collapse
Affiliation(s)
- Adela S. Oliva Chávez
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
- * E-mail:
| | - James W. Fairman
- Emerald Bio, Bainbridge Island, Washington, United States of America
- Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington, United States of America
| | - Roderick F. Felsheim
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Curtis M. Nelson
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Michael J. Herron
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Nicole Y. Burkhardt
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Jonathan D. Oliver
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Todd W. Markowski
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Timothy J. Kurtti
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Thomas E. Edwards
- Emerald Bio, Bainbridge Island, Washington, United States of America
- Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington, United States of America
| | - Ulrike G. Munderloh
- Department of Entomology, University of Minnesota, Saint Paul, Minnesota, United States of America
| |
Collapse
|
32
|
Oki AT, Seidman D, Lancina MG, Mishra MK, Kannan RM, Yang H, Carlyon JA. Dendrimer-enabled transformation of Anaplasma phagocytophilum. Microbes Infect 2015; 17:817-22. [PMID: 26369714 PMCID: PMC4666749 DOI: 10.1016/j.micinf.2015.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 12/22/2022]
Abstract
Anaplasma phagocytophilum is an obligate intracellular bacterium that causes the emerging infection, granulocytic anaplasmosis. While electroporation can transform A. phagocytophilum isolated from host cells, no method has been developed to transform it while growing inside the ApV (A. phagocytophilum-occupied vacuole). Polyamidoamine (PAMAM) dendrimers, well-defined tree-branched macromolecules used for gene therapy and nucleic acid delivery into mammalian cells, were recently shown to be effective in transforming Chlamydia spp. actively growing in host cells. We determined if we could adapt a similar system to transform A. phagocytophilum. Incubating fluorescently labeled PAMAM dendrimers with infected host cells resulted in fluorescein-positive ApVs. Incubating infected host cells or host cell-free A. phagocytophilum organisms with dendrimers complexed with pCis GFPuv-SS Himar A7 plasmid, which carries a Himar1 transposon cassette encoding GFPuv and spectinomycin/streptomycin resistance plus the Himar1 transposase itself, resulted in GFP-positive, antibiotic resistant bacteria. Yet, transformation efficiencies were low. The transformed bacterial populations could only be maintained for a few passages, likely due to random Himar1 cassette-mediated disruption of A. phagocytophilum genes required for fitness. Nonetheless, these results provide proof of principle that dendrimers can deliver exogenous DNA into A. phagocytophilum, both inside and outside of host cells.
Collapse
Affiliation(s)
- Aminat T Oki
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, PO Box 980678, Richmond, VA, 23298-0678, United States
| | - David Seidman
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, PO Box 980678, Richmond, VA, 23298-0678, United States
| | - Michael G Lancina
- Department of Biomedical Engineering, Virginia Commonwealth University School of Engineering, P.O. Box 843067, Richmond, VA 23284-3067, United States
| | - Manoj K Mishra
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21235, United States
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21235, United States
| | - Hu Yang
- Department of Biomedical Engineering, Virginia Commonwealth University School of Engineering, P.O. Box 843067, Richmond, VA 23284-3067, United States
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, PO Box 980678, Richmond, VA, 23298-0678, United States.
| |
Collapse
|
33
|
Walker DH, Dumler JS. The role of CD8 T lymphocytes in rickettsial infections. Semin Immunopathol 2015; 37:289-99. [PMID: 25823954 DOI: 10.1007/s00281-015-0480-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/15/2015] [Indexed: 12/01/2022]
Abstract
Arthropod-borne obligately intracellular bacteria pose a difficult challenge to the immune system. The genera Rickettsia, Orientia, Ehrlichia, and Anaplasma evolved mechanisms of immune evasion, and each interacts differently with the immune system. The roles of CD8 T cells include protective immunity and immunopathology. In Rickettsia infections, CD8 T cells are protective mediated in part by cytotoxicity toward infected cells. In contrast, TNF-α overproduction by CD8 T cells is pathogenic in lethal ehrlichiosis by induction of apoptosis/necrosis in hepatocytes. Yet, CD8 T cells, along with CD4 T cells and antibodies, also contribute to protective immunity in ehrlichial infections. In granulocytic anaplasmosis, CD8 T cells impact pathogen control modestly but could contribute to immunopathology by virtue of their dysfunction. While preliminary evidence indicates that CD8 T cells are important in protection against Orientia tsutsugamushi, mechanistic studies have been neglected. Valid animal models will enable experiments to elucidate protective and pathologic immune mechanisms. The public health need for vaccines against these agents of human disease, most clearly O. tsutsugamushi, and the veterinary diseases, canine monocytotropic ehrlichiosis (Ehrlichia canis), heartwater (Ehrlichia ruminantium), and bovine anaplasmosis (A. marginale), requires detailed immunity and immunopathology investigations, including the roles of CD8 T lymphocytes.
Collapse
Affiliation(s)
- David H Walker
- Department of Pathology, Director, UTMB Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555-0609, USA,
| | | |
Collapse
|
34
|
Essential domains of Anaplasma phagocytophilum invasins utilized to infect mammalian host cells. PLoS Pathog 2015; 11:e1004669. [PMID: 25658707 PMCID: PMC4450072 DOI: 10.1371/journal.ppat.1004669] [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: 08/21/2014] [Accepted: 01/07/2015] [Indexed: 12/01/2022] Open
Abstract
Anaplasma phagocytophilum causes granulocytic anaplasmosis, an emerging disease of humans and domestic animals. The obligate intracellular bacterium uses its invasins OmpA, Asp14, and AipA to infect myeloid and non-phagocytic cells. Identifying the domains of these proteins that mediate binding and entry, and determining the molecular basis of their interactions with host cell receptors would significantly advance understanding of A. phagocytophilum infection. Here, we identified the OmpA binding domain as residues 59 to 74. Polyclonal antibody generated against a peptide spanning OmpA residues 59 to 74 inhibited A. phagocytophilum infection of host cells and binding to its receptor, sialyl Lewis x (sLex-capped P-selectin glycoprotein ligand 1. Molecular docking analyses predicted that OmpA residues G61 and K64 interact with the two sLex sugars that are important for infection, α2,3-sialic acid and α1,3-fucose. Amino acid substitution analyses demonstrated that K64 was necessary, and G61 was contributory, for recombinant OmpA to bind to host cells and competitively inhibit A. phagocytophilum infection. Adherence of OmpA to RF/6A endothelial cells, which express little to no sLex but express the structurally similar glycan, 6-sulfo-sLex, required α2,3-sialic acid and α1,3-fucose and was antagonized by 6-sulfo-sLex antibody. Binding and uptake of OmpA-coated latex beads by myeloid cells was sensitive to sialidase, fucosidase, and sLex antibody. The Asp14 binding domain was also defined, as antibody specific for residues 113 to 124 inhibited infection. Because OmpA, Asp14, and AipA each contribute to the infection process, it was rationalized that the most effective blocking approach would target all three. An antibody cocktail targeting the OmpA, Asp14, and AipA binding domains neutralized A. phagocytophilum binding and infection of host cells. This study dissects OmpA-receptor interactions and demonstrates the effectiveness of binding domain-specific antibodies for blocking A. phagocytophilum infection. Anaplasma phagocytophilum causes the potentially deadly bacterial disease granulocytic anaplasmosis. The pathogen replicates inside white blood cells and, like all other obligate intracellular organisms, must enter host cells to survive. Multiple A. phagocytophilum surface proteins called invasins cooperatively orchestrate the entry process. Identifying these proteins’ domains that are required for function, and determining the molecular basis of their interaction with host cell receptors would significantly advance understanding of A. phagocytophilum pathogenesis. In this study, the binding domains of two A. phagocytophilum surface proteins, OmpA and Asp14, were identified. The specific OmpA residues that interact with its host cell receptor were also defined. An antibody cocktail generated against the binding domains of OmpA, Asp14, and a third invasin, AipA, blocked the ability of A. phagocytophilum to infect host cells. The data presented within suggest that binding domains of OmpA, Asp14, and AipA could be exploited to develop a vaccine for granulocytic anaplasmosis.
Collapse
|
35
|
Wang J, Dyachenko V, Munderloh UG, Straubinger RK. Transmission of Anaplasma phagocytophilum from endothelial cells to peripheral granulocytes in vitro under shear flow conditions. Med Microbiol Immunol 2015; 204:593-603. [PMID: 25618174 DOI: 10.1007/s00430-015-0387-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/13/2015] [Indexed: 01/06/2023]
Abstract
Anaplasma phagocytophilum (Ap) is a tick-borne pathogen, which can cause granulocytic anaplasmosis in humans and animals. In vivo this obligate intracellular pathogen is primarily located in circulating mature granulocytes, but it also infects endothelial cells. In order to study the interaction between Ap-infected endothelial cells and human granulocytes under conditions similar to those found naturally in the infected host, an in vitro model that mimics physiological flow conditions in the microvasculature was established. Cell-to-cell interactions were then visualized by microscopy, which showed that granulocytes adhered strongly to Ap-infected endothelial cells at a shear stress of 0.5 dyne/cm(2). In addition, Ap-transmission assays under flow conditions showed that the bacteria transferred from infected endothelial cells to circulating granulocytes and were able to establish infection in constantly moving granulocytes. Cell surface analysis showed that Ap induced up-regulation of the cell adhesion molecules ICAM-1 and VCAM-1 on infected endothelial cells in a dose-dependent manner. Furthermore, IL-8 secretion by endothelial cells indicated that the presence of Ap induced a pro-inflammatory response. In summary, the results of this study suggest that endothelial cells of the microvasculature (1) provide an excellent site for Ap dissemination to peripheral blood granulocytes under flow conditions and therefore may play a crucial role in the development of persistent infection, and (2) are stimulated by Ap to express surface molecules and cytokines that may lead to inflammatory responses at the site of the infection.
Collapse
Affiliation(s)
- Jinyong Wang
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Faculty of Veterinary Medicine, LMU Munich, Veterinärstr. 13, 80539, Munich, Germany
| | | | | | | |
Collapse
|
36
|
Proctor MC, Leiby DA. Do leukoreduction filters passively reduce the transmission risk of human granulocytic anaplasmosis? Transfusion 2014; 55:1242-8. [DOI: 10.1111/trf.12976] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/11/2014] [Accepted: 11/11/2014] [Indexed: 11/26/2022]
Affiliation(s)
- Melanie C. Proctor
- Transmissible Diseases Department; American Red Cross Holland Laboratory; Rockville Maryland
| | - David A. Leiby
- Transmissible Diseases Department; American Red Cross Holland Laboratory; Rockville Maryland
| |
Collapse
|
37
|
Beyer AR, Truchan HK, May LJ, Walker NJ, Borjesson DL, Carlyon JA. The Anaplasma phagocytophilum effector AmpA hijacks host cell SUMOylation. Cell Microbiol 2014; 17:504-19. [PMID: 25308709 DOI: 10.1111/cmi.12380] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 09/18/2014] [Accepted: 10/03/2014] [Indexed: 12/25/2022]
Abstract
SUMOylation, the covalent attachment of a member of the small ubiquitin-like modifier (SUMO) family of proteins to lysines in target substrates, is an essential post-translational modification in eukaryotes. Microbial manipulation of SUMOylation recently emerged as a key virulence strategy for viruses and facultative intracellular bacteria, the latter of which have only been shown to deploy effectors that negatively regulate SUMOylation. Here, we demonstrate that the obligate intracellular bacterium, Anaplasma phagocytophilum, utilizes an effector, AmpA (A. phagocytophilum post-translationally modified protein A) that becomes SUMOylated in host cells and this is important for the pathogen's survival. We previously discovered that AmpA (formerly APH1387) localizes to the A. phagocytophilum-occupied vacuolar membrane (AVM). Algorithmic prediction analyses denoted AmpA as a candidate for SUMOylation. We verified this phenomenon using a SUMO affinity matrix to precipitate both native AmpA and ectopically expressed green fluorescent protein (GFP)-tagged AmpA. SUMOylation of AmpA was lysine dependent, as SUMO affinity beads failed to precipitate a GFP-AmpA protein when its lysine residues were substituted with arginine. Ectopically expressed and endogenous AmpA were poly-SUMOylated, which was consistent with the observation that AmpA colocalizes with SUMO2/3 at the AVM. Only late during the infection cycle did AmpA colocalize with SUMO1, which terminally caps poly-SUMO2/3 chains. AmpA was also detected in the cytosol of infected host cells, further supporting its secretion and likely participation in interactions that aid pathogen survival. Indeed, whereas siRNA-mediated knockdown of Ubc9 - a necessary enzyme for SUMOylation - slightly bolstered A. phagocytophilum infection, pharmacologically inhibiting SUMOylation in infected cells significantly reduced the bacterial load. Ectopically expressed GFP-AmpA served as a competitive agonist against native AmpA in infected cells, while lysine-deficient GFP-AmpA was less effective, implying that modification of AmpA lysines is important for infection. Collectively, these data show that AmpA becomes directly SUMOylated during infection, representing a novel tactic for A. phagocytophilum survival.
Collapse
Affiliation(s)
- Andrea R Beyer
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | | | | | | | | | | |
Collapse
|
38
|
Seidman D, Ojogun N, Walker NJ, Mastronunzio J, Kahlon A, Hebert KS, Karandashova S, Miller DP, Tegels BK, Marconi RT, Fikrig E, Borjesson DL, Carlyon JA. Anaplasma phagocytophilum surface protein AipA mediates invasion of mammalian host cells. Cell Microbiol 2014; 16:1133-45. [PMID: 24612118 DOI: 10.1111/cmi.12286] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 01/11/2023]
Abstract
Anaplasma phagocytophilum, which causes granulocytic anaplasmosis in humans and animals, is a tick-transmitted obligate intracellular bacterium that mediates its own uptake into neutrophils and non-phagocytic cells. Invasins of obligate intracellular pathogens are attractive targets for protecting against or curing infection because blocking the internalization step prevents survival of these organisms. The complement of A. phagocytophilum invasins is incompletely defined. Here, we report the significance of a novel A. phagocytophilum invasion protein, AipA. A. phagocytophilum induced aipA expression during transmission feeding of infected ticks on mice. The bacterium upregulated aipA transcription when it transitioned from its non-infectious reticulate cell morphotype to its infectious dense-cored morphotype during infection of HL-60 cells. AipA localized to the bacterial surface and was expressed during in vivo infection. Of the AipA regions predicted to be surface-exposed, only residues 1 to 87 (AipA1-87 ) were found to be essential for host cell invasion. Recombinant AipA1-87 protein bound to and competitively inhibited A. phagocytophilum infection of mammalian cells. Antiserum specific for AipA1-87 , but not other AipA regions, antagonized infection. Additional blocking experiments using peptide-specific antisera narrowed down the AipA invasion domain to residues 9 to 21. An antisera combination targeting AipA1-87 together with two other A. phagocytophilum invasins, OmpA and Asp14, nearly abolished infection of host cells. This study identifies AipA as an A. phagocytophilum surface protein that is critical for infection, demarcates its invasion domain, and establishes a rationale for targeting multiple invasins to protect against granulocytic anaplasmosis.
Collapse
Affiliation(s)
- David Seidman
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dyachenko V, Geiger C, Pantchev N, Majzoub M, Bell-Sakyi L, Krupka I, Straubinger RK. Isolation of canine Anaplasma phagocytophilum strains from clinical blood samples using the Ixodes ricinus cell line IRE/CTVM20. Vet Microbiol 2013; 162:980-986. [PMID: 23146170 PMCID: PMC3757156 DOI: 10.1016/j.vetmic.2012.10.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/12/2012] [Accepted: 10/15/2012] [Indexed: 11/26/2022]
Abstract
Anaplasma phagocytophilum is an intracellular tick-borne rickettsial pathogen, which causes granulocytic anaplasmosis in various species of livestock and companion animals and also in humans. Previously A. phagocytophilum has been isolated and propagated in cell lines derived from the tick Ixodes scapularis and in the human promyelocytic cell line HL60. In this study we used the Ixodes ricinus-derived cell line IRE/CTVM20 to isolate and propagate two new canine strains of A. phagocytophilum. Blood samples were collected by veterinarians from two dogs, one from Germany and the other from Austria. Suspicion of clinical canine granulocytic anaplasmosis was raised by the treating veterinarians and after confirmation of A. phagocytophilum infection by real-time PCR, buffy coat cells were isolated and co-cultivated with IRE/CTVM20 cells maintained at 28 °C in L15/L15B medium. In the tick cells, rickettsial inclusions were first recognised after 86 days of incubation. Electron microscopic examination of tick cells infected with one of the isolates revealed cytoplasmic vacuoles containing pleomorphic organisms with individual bacteria enveloped by a bilayer membrane. Sequencing of 16S rRNA genes confirmed the isolation of A. phagocytophilum and showed the highest identity to the A. phagocytophilum human HZ strain. The two A. phagocytophilum isolates were passaged several times in IRE/CTVM20 cells and transferred to the I. scapularis cell line ISE6. This confirms for the first time the successful establishment and continuous cultivation of this pathogen in I. ricinus cells as well as infectivity of these canine strains for I. scapularis cells.
Collapse
Affiliation(s)
- Viktor Dyachenko
- Institute for Infectious Diseases and Zoonoses, Department for Veterinary Sciences, Faculty for Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539 Munich, Germany.
| | - Christine Geiger
- Institute for Infectious Diseases and Zoonoses, Department for Veterinary Sciences, Faculty for Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539 Munich, Germany
| | - Nikola Pantchev
- IDEXX Vet Med Lab, Moerikestr. 28/3, 71636 Ludwigsburg, Germany
| | - Monir Majzoub
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, Faculty for Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539 Munich, Germany
| | - Lesley Bell-Sakyi
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Inke Krupka
- Institute for Infectious Diseases and Zoonoses, Department for Veterinary Sciences, Faculty for Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539 Munich, Germany
| | - Reinhard K Straubinger
- Institute for Infectious Diseases and Zoonoses, Department for Veterinary Sciences, Faculty for Veterinary Medicine, LMU Munich, Veterinaerstr. 13, 80539 Munich, Germany
| |
Collapse
|
40
|
Ojogun N, Kahlon A, Ragland SA, Troese MJ, Mastronunzio JE, Walker NJ, VieBrock L, Thomas RJ, Borjesson DL, Fikrig E, Carlyon JA. Anaplasma phagocytophilum outer membrane protein A interacts with sialylated glycoproteins to promote infection of mammalian host cells. Infect Immun 2012; 80:3748-60. [PMID: 22907813 PMCID: PMC3486060 DOI: 10.1128/iai.00654-12] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 07/25/2012] [Indexed: 01/14/2023] Open
Abstract
Anaplasma phagocytophilum is the tick-transmitted obligate intracellular bacterium that causes human granulocytic anaplasmosis (HGA). A. phagocytophilum binding to sialyl Lewis x (sLe(x)) and other sialylated glycans that decorate P selectin glycoprotein 1 (PSGL-1) and other glycoproteins is critical for infection of mammalian host cells. Here, we demonstrate the importance of A. phagocytophilum outer membrane protein A (OmpA) APH_0338 in infection of mammalian host cells. OmpA is transcriptionally induced during transmission feeding of A. phagocytophilum-infected ticks on mice and is upregulated during invasion of HL-60 cells. OmpA is presented on the pathogen's surface. Sera from HGA patients and experimentally infected mice recognize recombinant OmpA. Pretreatment of A. phagocytophilum organisms with OmpA antiserum reduces their abilities to infect HL-60 cells. The OmpA N-terminal region is predicted to contain the protein's extracellular domain. Glutathione S-transferase (GST)-tagged versions of OmpA and OmpA amino acids 19 to 74 (OmpA(19-74)) but not OmpA(75-205) bind to, and competitively inhibit A. phagocytophilum infection of, host cells. Pretreatment of host cells with sialidase or trypsin reduces or nearly eliminates, respectively, GST-OmpA adhesion. Therefore, OmpA interacts with sialylated glycoproteins. This study identifies the first A. phagocytophilum adhesin-receptor pair and delineates the region of OmpA that is critical for infection.
Collapse
Affiliation(s)
- Nore Ojogun
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Amandeep Kahlon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Stephanie A. Ragland
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Matthew J. Troese
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Juliana E. Mastronunzio
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Naomi J. Walker
- Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California, USA
| | - Lauren VieBrock
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Rachael J. Thomas
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Dori L. Borjesson
- Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
41
|
Anaplasma phagocytophilum Asp14 is an invasin that interacts with mammalian host cells via its C terminus to facilitate infection. Infect Immun 2012; 81:65-79. [PMID: 23071137 DOI: 10.1128/iai.00932-12] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Anaplasma phagocytophilum, a member of the family Anaplasmataceae, is the tick-transmitted obligate intracellular bacterium that causes human granulocytic anaplasmosis. The life cycle of A. phagocytophilum is biphasic, transitioning between the noninfectious reticulate cell (RC) and infectious dense-cored (DC) forms. We analyzed the bacterium's DC surface proteome by selective biotinylation of surface proteins, NeutrAvidin affinity purification, and mass spectrometry. Transcriptional profiling of selected outer membrane protein candidates over the course of infection revealed that aph_0248 (designated asp14 [14-kDa A. phagocytophilum surface protein]) expression was upregulated the most during A. phagocytophilum cellular invasion. asp14 transcription was induced during transmission feeding of A. phagocytophilum-infected ticks on mice and was upregulated when the bacterium engaged its receptor, P-selectin glycoprotein ligand 1. Asp14 localized to the A. phagocytophilum surface and was expressed during in vivo infection. Treating DC organisms with Asp14 antiserum or preincubating mammalian host cells with glutathione S-transferase (GST)-Asp14 significantly inhibited infection of host cells. Moreover, preincubating host cells with GST-tagged forms of both Asp14 and outer membrane protein A, another A. phagocytophilum invasin, pronouncedly reduced infection relative to treatment with either protein alone. The Asp14 domain that is sufficient for cellular adherence and invasion lies within the C-terminal 12 to 24 amino acids and is conserved among other Anaplasma and Ehrlichia species. These results identify Asp14 as an A. phagocytophilum surface protein that is critical for infection, delineate its invasion domain, and demonstrate the potential of targeting Asp14 in concert with OmpA for protecting against infection by A. phagocytophilum and other Anaplasmataceae pathogens.
Collapse
|
42
|
Huang B, Ojogun N, Ragland SA, Carlyon JA. Monoubiquitinated proteins decorate the Anaplasma phagocytophilum-occupied vacuolar membrane. ACTA ACUST UNITED AC 2012; 64:32-41. [PMID: 22066989 DOI: 10.1111/j.1574-695x.2011.00873.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An emerging theme among vacuole-adapted bacterial pathogens is the ability to hijack ubiquitin machinery to modulate host cellular processes and secure pathogen survival. Mono- and polyubiquitination differentially dictate the subcellular localization, activity, and fate of protein substrates. Monoubiquitination directs membrane traffic from the plasma membrane to the endosome and has been shown to promote autophagy. Anaplasma phagocytophilum is an obligate intracellular bacterium that replicates within a host cell-derived vacuole that co-opts membrane traffic and numerous other host cell processes. Here, we show that monoubiquitinated proteins decorate the A. phagocytophilum-occupied vacuolar membrane (AVM) during infection of promyelocytic HL-60 cell, endothelial RF/6A cells, and to a lesser extent, embryonic tick ISE6 cells. Monoubiquitinated proteins are present on the AVM upon its formation and continue to accumulate throughout infection. Tetracycline-mediated inhibition of de novo bacterial protein synthesis promotes the loss of ubiquitinated proteins from the AVM. This effect is reversible, as removal of tetracycline restores AVM ubiquitination to pretreatment levels. These results demonstrate a novel mechanism by which A. phagocytophilum remodels the composition of its host cell-derived vacuolar membrane and present the first example of a Rickettsiales pathogen co-opting ubiquitin during intracellular residence.
Collapse
Affiliation(s)
- Bernice Huang
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298-0678, USA.
| | | | | | | |
Collapse
|
43
|
Postgenomic analyses reveal development of infectious Anaplasma phagocytophilum during transmission from ticks to mice. J Bacteriol 2012; 194:2238-47. [PMID: 22389475 DOI: 10.1128/jb.06791-11] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Obligate intracellular bacteria of the Rickettsiales order have evolved to colonize both arthropod and mammalian hosts, but few details are known about the bacterial adaptations that occur during transmission from blood-feeding arthropods to mammals. Here we apply proteomics and transcriptome sequencing to Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis, in Ixodes scapularis tick salivary glands, to detect proteins or genes expressed by the pathogen during transmission feeding by the tick. We detected expression of 139 genes, representing 11% of the open reading frames (ORFs) in the A. phagocytophilum genome. The predominant categories of proteins were ribosomal proteins, cell surface proteins, chaperones, and uncharacterized proteins. There was no evidence of DNA replication enzymes, suggesting that most of the A. phagocytophilum cells were no longer dividing. Instead, protein expression reflected conversion to the extracellular, infectious "dense-core" (DC) form. High expression of a DC-specific marker, APH_1235, further suggested this developmental transition in ticks. We showed that blocking APH_1235 with antibodies reduced A. phagocytophilum infection levels in mammalian cell culture. This work represents a starting point for clarifying essential proteins expressed by A. phagocytophilum during transmission from ticks to mammals and demonstrates that the abundantly expressed, DC-associated APH_1235 protein is important during in vivo infection by A. phagocytophilum.
Collapse
|
44
|
Infection with Anaplasma phagocytophilum activates the phosphatidylinositol 3-Kinase/Akt and NF-κB survival pathways in neutrophil granulocytes. Infect Immun 2012; 80:1615-23. [PMID: 22252875 DOI: 10.1128/iai.05219-11] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anaplasma phagocytophilum, a Gram-negative, obligate intracellular bacterium infects primarily neutrophil granulocytes. Infection with A. phagocytophilum leads to inhibition of neutrophil apoptosis and consequently contributes to the longevity of the host cells. Previous studies demonstrated that the infection inhibits the executionary apoptotic machinery in neutrophils. However, little attempt has been made to explore which survival signals are modulated by the pathogen. The aim of the present study was to clarify whether the phosphatidylinositol 3-kinase (PI3K)/Akt and NF-κB signaling pathways, which are considered as important survival pathways in neutrophils, are involved in A. phagocytophilum-induced apoptosis delay. Our data show that infection of neutrophils with A. phagocytophilum activates the PI3K/Akt pathway and suggest that this pathway, which in turn maintains the expression of the antiapoptotic protein Mcl-1, contributes to the infection-induced apoptosis delay. In addition, the PI3K/Akt pathway is involved in the activation of NF-κB in A. phagocytophilum-infected neutrophils. Activation of NF-κB leads to the release of interleukin-8 (IL-8) from infected neutrophils, which, in an autocrine manner, delays neutrophil apoptosis. In addition, enhanced expression of the antiapoptotic protein cIAP2 was observed in A. phagocytophilum-infected neutrophils. Taken together, the data indicate that upstream of the apoptotic cascade, signaling via the PI3K/Akt pathway plays a major role for apoptosis delay in A. phagocytophilum-infected neutrophils.
Collapse
|
45
|
Troese MJ, Kahlon A, Ragland SA, Ottens AK, Ojogun N, Nelson KT, Walker NJ, Borjesson DL, Carlyon JA. Proteomic analysis of Anaplasma phagocytophilum during infection of human myeloid cells identifies a protein that is pronouncedly upregulated on the infectious dense-cored cell. Infect Immun 2011; 79:4696-707. [PMID: 21844238 PMCID: PMC3257945 DOI: 10.1128/iai.05658-11] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/02/2011] [Indexed: 01/31/2023] Open
Abstract
Anaplasma phagocytophilum is an obligate intracellular bacterium that invades neutrophils to cause the emerging infectious disease human granulocytic anaplasmosis. A. phagocytophilum undergoes a biphasic developmental cycle, transitioning between an infectious dense-cored cell (DC) and a noninfectious reticulate cell (RC). To gain insights into the organism's biology and pathogenesis during human myeloid cell infection, we conducted proteomic analyses on A. phagocytophilum organisms purified from HL-60 cells. A total of 324 proteins were unambiguously identified, thereby verifying 23.7% of the predicted A. phagocytophilum proteome. Fifty-three identified proteins had been previously annotated as hypothetical or conserved hypothetical. The second most abundant gene product, after the well-studied major surface protein 2 (P44), was the hitherto hypothetical protein APH_1235. APH_1235 homologs are found in other Anaplasma and Ehrlichia species but not in other bacteria. The aph_1235 RNA level is increased 70-fold in the DC form relative to that in the RC form. Transcriptional upregulation of and our ability to detect APH_1235 correlate with RC to DC transition, DC exit from host cells, and subsequent DC binding and entry during the next round of infection. Immunoelectron microscopy pronouncedly detects APH_1235 on DC organisms, while detection on RC bacteria minimally, at best, exceeds background. This work represents an extensive study of the A. phagocytophilum proteome, discerns the complement of proteins that is generated during survival within human myeloid cells, and identifies APH_1235 as the first known protein that is pronouncedly upregulated on the infectious DC form.
Collapse
Affiliation(s)
| | | | | | - Andrew K. Ottens
- Anatomy and Neurobiology
- Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | | | - Kristina T. Nelson
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia
| | - Naomi J. Walker
- Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | - Dori L. Borjesson
- Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | | |
Collapse
|
46
|
Mechanisms of obligatory intracellular infection with Anaplasma phagocytophilum. Clin Microbiol Rev 2011; 24:469-89. [PMID: 21734244 PMCID: PMC3131063 DOI: 10.1128/cmr.00064-10] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Anaplasma phagocytophilum persists in nature by cycling between mammals and ticks. Human infection by the bite of an infected tick leads to a potentially fatal emerging disease called human granulocytic anaplasmosis. A. phagocytophilum is an obligatory intracellular bacterium that replicates inside mammalian granulocytes and the salivary gland and midgut cells of ticks. A. phagocytophilum evolved the remarkable ability to hijack the regulatory system of host cells. A. phagocytophilum alters vesicular traffic to create an intracellular membrane-bound compartment that allows replication in seclusion from lysosomes. The bacterium downregulates or actively inhibits a number of innate immune responses of mammalian host cells, and it upregulates cellular cholesterol uptake to acquire cholesterol for survival. It also upregulates several genes critical for the infection of ticks, and it prolongs tick survival at freezing temperatures. Several host factors that exacerbate infection have been identified, including interleukin-8 (IL-8) and cholesterol. Host factors that overcome infection include IL-12 and gamma interferon (IFN-γ). Two bacterial type IV secretion effectors and several bacterial proteins that associate with inclusion membranes have been identified. An understanding of the molecular mechanisms underlying A. phagocytophilum infection will foster the development of creative ideas to prevent or treat this emerging tick-borne disease.
Collapse
|
47
|
Anaplasma phagocytophilum infects mast cells via alpha1,3-fucosylated but not sialylated glycans and inhibits IgE-mediated cytokine production and histamine release. Infect Immun 2011; 79:2717-26. [PMID: 21536789 DOI: 10.1128/iai.00181-11] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mast cells are sentinels for infection. Upon exposure to pathogens, they release their stores of proinflammatory cytokines, chemokines, and histamine. Mast cells are also important for the control of certain tick-borne infections. Anaplasma phagocytophilum is an obligate intracellular tick-transmitted bacterium that infects neutrophils to cause the emerging disease granulocytic anaplasmosis. A. phagocytophilum adhesion to and infection of neutrophils depend on sialylated and α1,3-fucosylated glycans. We investigated the hypotheses that A. phagocytophilum invades mast cells and inhibits mast cell activation. We demonstrate that A. phagocytophilum binds and/or infects murine bone marrow-derived mast cells (BMMCs), murine peritoneal mast cells, and human skin-derived mast cells. A. phagocytophilum infection of BMMCs depends on α1,3-fucosylated, but not sialylated, glycans. A. phagocytophilum binding to and invasion of BMMCs do not elicit proinflammatory cytokine secretion. Moreover, A. phagocytophilum-infected cells are inhibited in the release of tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), IL-13, and histamine following stimulation with IgE or antigen. Thus, A. phagocytophilum mitigates mast cell activation. These findings potentially represent a novel means by which A. phagocytophilum usurps host defense mechanisms and shed light on the interplay between mast cells and vector-borne bacterial pathogens.
Collapse
|
48
|
Beare PA, Sandoz KM, Omsland A, Rockey DD, Heinzen RA. Advances in genetic manipulation of obligate intracellular bacterial pathogens. Front Microbiol 2011; 2:97. [PMID: 21833334 PMCID: PMC3153054 DOI: 10.3389/fmicb.2011.00097] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 04/19/2011] [Indexed: 11/22/2022] Open
Abstract
Infections by obligate intracellular bacterial pathogens result in significant morbidity and mortality worldwide. These bacteria include Chlamydia spp., which causes millions of cases of sexually transmitted disease and blinding trachoma annually, and members of the α-proteobacterial genera Anaplasma, Ehrlichia, Orientia, and Rickettsia, agents of serious human illnesses including epidemic typhus. Coxiella burnetii, the agent of human Q fever, has also been considered a prototypical obligate intracellular bacterium, but recent host cell-free (axenic) growth has rescued it from obligatism. The historic genetic intractability of obligate intracellular bacteria has severely limited molecular dissection of their unique lifestyles and virulence factors involved in pathogenesis. Host cell restricted growth is a significant barrier to genetic transformation that can make simple procedures for free-living bacteria, such as cloning, exceedingly difficult. Low transformation efficiency requiring long-term culture in host cells to expand small transformant populations is another obstacle. Despite numerous technical limitations, the last decade has witnessed significant gains in genetic manipulation of obligate intracellular bacteria including allelic exchange. Continued development of genetic tools should soon enable routine mutation and complementation strategies for virulence factor discovery and stimulate renewed interest in these refractory pathogens. In this review, we discuss the technical challenges associated with genetic transformation of obligate intracellular bacteria and highlight advances made with individual genera.
Collapse
Affiliation(s)
- Paul A Beare
- Coxiella Pathogenesis Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton, MT, USA
| | | | | | | | | |
Collapse
|
49
|
Huang B, Troese MJ, Howe D, Ye S, Sims JT, Heinzen RA, Borjesson DL, Carlyon JA. Anaplasma phagocytophilum APH_0032 is expressed late during infection and localizes to the pathogen-occupied vacuolar membrane. Microb Pathog 2010; 49:273-84. [PMID: 20600793 DOI: 10.1016/j.micpath.2010.06.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/18/2010] [Accepted: 06/23/2010] [Indexed: 01/16/2023]
Abstract
Anaplasma phagocytophilum infects neutrophils and myeloid, endothelial, and tick cell lines to reside within a host cell-derived vacuole that is indispensible for its survival. Here, we identify APH_0032 as an Anaplasma-derived protein that associates with the A. phagocytophilum-occupied vacuolar membrane (AVM). APH_0032 is a 66.1 kDa acidic protein that electrophoretically migrates with an apparent molecular weight of 130 kDa. It contains a predicted transmembrane domain and tandemly arranged direct repeats that comprise 46% of the protein. APH_0032 is undetectable on Anaplasma organisms bound to the surfaces of HL-60 cells, but is detected on the AVM and surfaces of intravacuolar bacteria beginning 24 h post-infection. APH_0032 localizes to the AVM in HL-60, THP-1, HMEC-1, and ISE6 cells. APH_0032, along with APH_1387, which encodes a confirmed AVM protein, is transcribed during A. phagocytophilum infection of tick salivary glands and murine neutrophils. APH_0032 localizes to the AVM in neutrophils recovered from infected mice. The Legionella pneumophila Dot/IcM type IV secretion system (T4SS) can heterologously secrete a CyaA-tagged version of the A. phagocytophilum VirB/D T4SS effector, AnkA, but fails to secrete CyaA-tagged APH_0032 or APH_1387. These data confirm APH_0032 as an Anaplasma-derived AVM protein and hint that neither it nor APH_1387 are T4SS effectors.
Collapse
Affiliation(s)
- Bernice Huang
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298-0678, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Huang B, Troese MJ, Ye S, Sims JT, Galloway NL, Borjesson DL, Carlyon JA. Anaplasma phagocytophilum APH_1387 is expressed throughout bacterial intracellular development and localizes to the pathogen-occupied vacuolar membrane. Infect Immun 2010; 78:1864-73. [PMID: 20212090 PMCID: PMC2863503 DOI: 10.1128/iai.01418-09] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 01/18/2010] [Accepted: 02/15/2010] [Indexed: 11/20/2022] Open
Abstract
Obligate vacuolar pathogens produce proteins that localize to the host cell-derived membranes of the vacuoles in which they reside, yielding unique organelles that are optimally suited for pathogen survival. Anaplasma phagocytophilum is an obligate vacuolar bacterium that infects neutrophils and causes the emerging and potentially fatal disease human granulocytic anaplasmosis. Here we identified APH_1387 as the first A. phagocytophilum-derived protein that associates with the A. phagocytophilum-occupied vacuolar membrane (AVM). APH_1387, also referred to as P100, is a 61.4-kDa acidic protein that migrates with an apparent molecular weight of 115 kDa on SDS-PAGE gels. It carries 3 tandem direct repeats that comprise 58% of the protein. Each APH_1387 repeat carries a bilobed hydrophobic alpha-helix domain, which is a structural characteristic that is consistent with the structure of chlamydia-derived proteins that traverse inclusion membranes. APH_1387 is not detectable on the surfaces of A. phagocytophilum dense core organisms bound at the HL-60 cell surface, but abundant APH_1387 is detected on the surfaces of intravacuolar reticulate cell and dense core organisms. APH_1387 accumulates on the AVM throughout infection. It associates with the AVM in human HL-60, THP-1, and HMEC-1 cells and tick ISE6 cells. APH_1387 is expressed and localizes to the AVM in neutrophils recovered from A. phagocytophilum-infected mice. This paper presents the first direct evidence that A. phagocytophilum actively modifies its host cell-derived vacuole.
Collapse
Affiliation(s)
- Bernice Huang
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40504, Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | - Matthew J. Troese
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40504, Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | - Shaojing Ye
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40504, Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | - Jonathan T. Sims
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40504, Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | - Nathan L. Galloway
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40504, Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | - Dori L. Borjesson
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40504, Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40504, Department of Pathology, Microbiology, and Immunology, University of California School of Veterinary Medicine, Davis, California 95616
| |
Collapse
|