Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
mBio ; 15(4): e0029924, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38415594

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.


Subject(s)
Anaplasma phagocytophilum , Neoplasms , Animals , Humans , Mice , Anaplasma phagocytophilum/metabolism , Golgi Apparatus/metabolism , Ceramides , Mammals/metabolism
2.
Front Cell Infect Microbiol ; 12: 828605, 2022.
Article in English | MEDLINE | ID: mdl-35719343

ABSTRACT

Anaplasma phagocytophilum, a tick-borne obligately intracellular bacterium of neutrophils, causes human granulocytic anaplasmosis. Ankyrin A (AnkA), an effector protein with multiple ankyrin repeats (AR) is injected via type IV-secretion into the host neutrophil to gain access to the nucleus where it modifies the epigenome to promote microbial fitness and propagation. AR proteins transported into the host cell nucleus must use at least one of two known eukaryotic pathways, the classical importin ß-dependent pathway, and/or the RanGDP- and AR (ankyrin-repeat)-dependent importin ß-independent (RaDAR) pathway. Truncation of the first four AnkA N-terminal ARs (AR1-4), but not other regions, prevents AnkA nuclear accumulation. To investigate the mechanism of nuclear import, we created point mutations of AnkA N-terminal ARs, predicted to interfere with RaDAR protein import, and used importazole, a specific inhibitor of the importin α/ß, RanGTP-dependent pathway. Nuclear colocalization analysis shows that nuclear localization of AnkA is unaffected by single AR1-4 mutations but is significantly reduced by single mutations in consecutive ARs suggesting RaDAR protein nuclear import. However, AnkA nuclear localization was also decreased with importazole, and with GTPγS. Furthermore, A. phagocytophilum growth in HL-60 cells was completely suppressed with importazole, indicating that A. phagocytophilum propagation requires a ß-importin-dependent pathway. A typical classical NLS overlapping AR4 was subsequently identified suggesting the primacy of the importin-α/ß system in AnkA nuclear localization. Whether the mutational studies of putative key residues support RaDAR NLS function or simply reflect structural changes that diminish engagement of an AR-NLS-importin pathway needs to be resolved through careful structure-function studies.


Subject(s)
Anaplasma phagocytophilum , Active Transport, Cell Nucleus , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/metabolism , Animals , Ankyrins/metabolism , Cell Nucleus/metabolism , Humans , Karyopherins/metabolism , beta Karyopherins/genetics , beta Karyopherins/metabolism
3.
mBio ; 12(5): e0229921, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34544283

ABSTRACT

The intracellular cholesterol transport protein Niemann-Pick type C1 (NPC1) and lipid-raft protein flotillin (FLOT) are required for cholesterol uptake by the obligatory intracellular bacterium Anaplasma phagocytophilum and for infection, and each protein localizes to membrane-bound inclusions containing replicating bacteria. Here, we found striking localization of FLOT2 in NPC1-lined vesicles and a physical interaction between FLOT2 and NPC1. This interaction was cholesterol dependent, as a CRAC (cholesterol recognition/interaction amino acid cholesterol-binding) domain mutant of FLOT2 did not interact with NPC1, and the cholesterol-sequestering agent methyl-ß-cyclodextrin reduced the interaction. The stomatin-prohibitin-flotillin-HflC/K domain of FLOT2, FLOT21-183, was sufficient for the unique FLOT2 localization and interaction with NPC1. NPC1, FLOT2, and FLOT21-183 trafficked to the lumen of Anaplasma inclusions. A loss-of-function mutant, NPC1P691S (mutation in the sterol-sensing domain), did not colocalize or interact with FLOT2 or with Anaplasma inclusions and inhibited infection. Ezetimibe is a drug that blocks cholesterol absorption in the small intestine by inhibiting plasma membrane Niemann-Pick C1-like 1 interaction with FLOTs. Ezetimibe blocked the interaction between NPC1 and FLOT2 and inhibited Anaplasma infection. Ezetimibe did not directly inhibit Anaplasma proliferation but inhibited host membrane lipid and cholesterol traffic to the bacteria in the inclusion. These data suggest that Anaplasma hijacks NPC1 vesicles containing cholesterol bound to FLOT2 to deliver cholesterol into Anaplasma inclusions to assimilate cholesterol for its proliferation. These results provide insights into mechanisms of intracellular cholesterol transport and a potential approach to inhibit Anaplasma infection by blocking cholesterol delivery into the lumen of bacterial inclusions. IMPORTANCE Cholesterol influences membrane fluidity and forms membrane microdomains called lipid rafts that serve as organizing centers for the assembly of signaling molecules. Flotillin (FLOT) is a cholesterol-binding lipid-raft protein. The cholesterol-binding membrane glycoprotein Niemann-Pick type C1 (NPC1) is critical for managing cellular cholesterol level and its intracellular transport, and mutation of the gene encoding NPC1 causes the fatal cholesterol storage disease, Niemann-Pick disease, type C. Both FLOT and NPC1 are trafficked to inclusions created by the cholesterol-dependent bacterium Anaplasma phagocytophilum and required for cholesterol uptake by this bacterium for replication. Our novel findings that FLOT2 interacts physically with NPC1 and resides inside both bacterial inclusions and NPC1-containing vesicles underscore the important role for FLOT2 in infection, the intracellular transport of cholesterol in NPC1 vesicles, and cholesterol homeostasis. Both NPC1-FLOT2 interaction and A. phagocytophilum infection can be inhibited by ezetimibe, suggesting possible pharmacological intervention of intracellular cholesterol hijacking by Anaplasma.


Subject(s)
Anaplasma phagocytophilum/growth & development , Anaplasma phagocytophilum/metabolism , Cholesterol/metabolism , Ehrlichiosis/microbiology , Ezetimibe/pharmacology , Membrane Proteins/metabolism , Niemann-Pick C1 Protein/metabolism , Anaplasma phagocytophilum/drug effects , Anaplasma phagocytophilum/genetics , Biological Transport , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Ehrlichiosis/genetics , Ehrlichiosis/metabolism , Host-Pathogen Interactions , Humans , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Inclusion Bodies/microbiology , Membrane Proteins/genetics , Niemann-Pick C1 Protein/genetics , Protein Binding , Protein Transport
4.
Int J Biol Macromol ; 184: 497-508, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34126152

ABSTRACT

Anaplasma phagocytophilum is an obligate intracellular bacterium and a common tick-borne infectious pathogen that can cause human granulocytic anaplasmosis (HGA). Effector proteins play an important role in the pathogenic mechanism of A. phagocytophilum, but the specifics of the disease mechanism are unclear. We studied the effector protein AptA (A. phagocytophilum toxin A) using yeast two hybrid assays to screen its interacting protein proteasome assembly chaperone 3 (PSMG3, PAC3), and identified new mechanisms for the pathogenicity of A. phagocytophilum in HEK293T cells. After AptA enters the host cell, it interacts with PSMG3 to enhance the activity of the proteasome, causing ubiquitination and autophagy in the host cell and thereby increasing cross-talk between the ubiquitination-proteasome system (UPS) and autophagy. AptA also reduces the apoptotic efficiency of the host cells. These results offer new clues as to the pathogenic mechanism of A. phagocytophilum and support the hypothesis that AptA interacts with host PSMG3.


Subject(s)
Anaplasma phagocytophilum/pathogenicity , Bacterial Toxins/metabolism , Molecular Chaperones/metabolism , Anaplasma phagocytophilum/metabolism , Autophagy , HEK293 Cells , Host-Pathogen Interactions , Humans , Proteasome Endopeptidase Complex/metabolism , Two-Hybrid System Techniques , Ubiquitination
5.
J Bacteriol ; 202(23)2020 11 04.
Article in English | MEDLINE | ID: mdl-32928930

ABSTRACT

Many pathogenic bacteria translocate virulence factors into their eukaryotic hosts by means of type IV secretion systems (T4SS) spanning the inner and outer membranes. Genes encoding components of these systems have been identified within the order Rickettsiales based upon their sequence similarities to other prototypical systems. Anaplasma phagocytophilum strains are obligate intracellular, tick-borne bacteria that are members of this order. The organization of these components at the genomic level was determined in several Anaplasma phagocytophilum strains, showing overall conservation, with the exceptions of the virB2 and virB6 genes. The virB6 loci are characterized by the presence of four virB6 copies (virB6-1 through virB6-4) arranged in tandem within a gene cluster known as the sodB-virB operon. Interestingly, the virB6-4 gene varies significantly in length among different strains due to extensive tandem repeats at the 3' end. To gain an understanding of how these enigmatic virB6 genes function in A. phagocytophilum, we investigated their expression in infected human and tick cells. Our results show that these genes are expressed by A. phagocytophilum replicating in both cell types and that VirB6-3 and VirB6-4 proteins are surface exposed. Analysis of an A. phagocytophilum mutant carrying the Himar1 transposon within the virB6-4 gene demonstrated that the insertion not only disrupted its expression but also exerted a polar effect on the sodB-virB operon. Moreover, the altered expression of genes within this operon was associated with the attenuated in vitro growth of A. phagocytophilum in human and tick cells, indicating the importance of these genes in the physiology of this obligate intracellular bacterium in such different environments.IMPORTANCE Knowledge of the T4SS is derived from model systems, such as Agrobacterium tumefaciens The structure of the T4SS in Rickettsiales differs from the classical arrangement. These differences include missing and duplicated components with structural alterations. Particularly, two sequenced virB6-4 genes encode unusual C-terminal structural extensions resulting in proteins of 4,322 (GenBank accession number AGR79286.1) and 9,935 (GenBank accession number ANC34101.1) amino acids. To understand how the T4SS is used in A. phagocytophilum, we describe the expression of the virB6 paralogs and explore their role as the bacteria replicate within its host cell. Conclusions about the importance of these paralogs for colonization of human and tick cells are supported by the deficient phenotype of an A. phagocytophilum mutant isolated from a sequence-defined transposon insertion library.


Subject(s)
Anaplasma phagocytophilum/growth & development , Anaplasma phagocytophilum/genetics , Bacterial Proteins/genetics , Anaplasma phagocytophilum/metabolism , Bacterial Proteins/metabolism , Base Sequence , Cell Line , Ehrlichiosis/microbiology , Humans , Mutagenesis, Insertional , Operon , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
6.
Cell Microbiol ; 22(10): e13237, 2020 10.
Article in English | MEDLINE | ID: mdl-32562372

ABSTRACT

Reactive oxygen species (ROS) that are induced upon pathogen infection plays an important role in host defence. The rickettsial pathogen Anaplasma phagocytophilum, which is primarily transmitted by Ixodes scapularis ticks in the United States, has evolved many strategies to escape ROS and survive in mammalian cells. However, little is known on the role of ROS in A. phagocytophilum infection in ticks. Our results show that A. phagocytophilum and hemin induce activation of l-tryptophan pathway in tick cells. Xanthurenic acid (XA), a tryptophan metabolite, supports A. phagocytophilum growth in tick cells through inhibition of tryptophan dioxygenase (TDO) activity leading to reduced l-kynurenine levels that subsequently affects build-up of ROS. However, hemin supports A. phagocytophilum growth in tick cells by inducing TDO activity leading to increased l-kynurenine levels and ROS production. Our data reveal that XA and kynurenic acid (KA) chelate hemin. Furthermore, treatment of tick cells with 3-hydroxyl l-kynurenine limits A. phagocytophilum growth in tick cells. RNAi-mediated knockdown of kynurenine aminotransferase expression results in increased ROS production and reduced A. phagocytophilum burden in tick cells. Collectively, these results suggest that l-tryptophan pathway metabolites influence A. phagocytophilum survival by affecting build up of ROS levels in tick cells.


Subject(s)
Anaplasma phagocytophilum/metabolism , Ixodes/microbiology , Tryptophan/metabolism , Animals , Hemin/metabolism , Hemin/pharmacology , Host-Pathogen Interactions , Hydrolases/genetics , Hydrolases/metabolism , Ixodes/genetics , Ixodes/metabolism , Kynurenic Acid/metabolism , Kynurenic Acid/pharmacology , Kynurenine/analogs & derivatives , Kynurenine/metabolism , Kynurenine/pharmacology , NADP/biosynthesis , NADP/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Transaminases/genetics , Transaminases/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism , Up-Regulation , Xanthurenates/metabolism , Xanthurenates/pharmacology
7.
Sci Rep ; 9(1): 13236, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31520000

ABSTRACT

Aerobic organisms evolved conserved mechanisms controlling the generation of reactive oxygen species (ROS) to maintain redox homeostasis signaling and modulate signal transduction, gene expression and cellular functional responses under physiological conditions. The production of ROS by mitochondria is essential in the oxidative stress associated with different pathologies and in response to pathogen infection. Anaplasma phagocytophilum is an intracellular pathogen transmitted by Ixodes scapularis ticks and causing human granulocytic anaplasmosis. Bacteria multiply in vertebrate neutrophils and infect first tick midgut cells and subsequently hemocytes and salivary glands from where transmission occurs. Previous results demonstrated that A. phagocytophilum does not induce the production of ROS as part of its survival strategy in human neutrophils. However, little is known about the role of ROS during pathogen infection in ticks. In this study, the role of tick oxidative stress during A. phagocytophilum infection was characterized through the function of different pathways involved in ROS production. The results showed that tick cells increase mitochondrial ROS production to limit A. phagocytophilum infection, while pathogen inhibits alternative ROS production pathways and apoptosis to preserve cell fitness and facilitate infection. The inhibition of NADPH oxidase-mediated ROS production by pathogen infection appears to occur in both neutrophils and tick cells, thus supporting that A. phagocytophilum uses common mechanisms for infection of ticks and vertebrate hosts. However, differences in ROS response to A. phagocytophilum infection between human and tick cells may reflect host-specific cell tropism that evolved during pathogen life cycle.


Subject(s)
Anaplasma phagocytophilum/pathogenicity , Anaplasmosis/microbiology , Disease Vectors , Host-Pathogen Interactions , Ixodes/microbiology , Metabolic Networks and Pathways , Neutrophils/microbiology , Anaplasma phagocytophilum/metabolism , Anaplasmosis/metabolism , Anaplasmosis/transmission , Animals , Gene Expression Regulation , HL-60 Cells , Humans , Neutrophils/metabolism , Oxidation-Reduction , Oxidative Stress , Rabbits , Sheep , Signal Transduction
8.
Sci Rep ; 9(1): 4235, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862835

ABSTRACT

Anaplasma phagocytophilum, the aetiologic agent of human granulocytic anaplasmosis (HGA) is an obligate intracellular Gram-negative bacterium with the genome size of 1.47 megabases. The intracellular life style and small size of genome suggest that A. phagocytophilum has to modulate a multitude of host cell physiological processes to facilitate its replication. One strategy employed by A. phagocytophilum is through its type IV secretion system (T4SS), which translocates bacterial effectors into target cells to disrupt normal cellular activities. In this study we developed a TEM-1 ß-lactamase based protein translocation assay and applied this assay for identification of A. phagocytophilum T4SS effectors. An A. phagocytophilum hypothetical protein, APH0215 is identified as a T4SS effector protein and found interacting with trans-Golgi network in transfected cells. Hereby, this protein translocation assay developed in this study will facilitate the identification of A. phagocytophilum T4SS effectors and elucidation of HGA pathogenesis.


Subject(s)
Anaplasma phagocytophilum , Anaplasmosis , Bacterial Proteins , Biological Assay , beta-Lactamases , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/metabolism , Anaplasmosis/diagnosis , Anaplasmosis/genetics , Anaplasmosis/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CHO Cells , Cricetinae , Cricetulus , Humans , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , beta-Lactamases/genetics , beta-Lactamases/metabolism
9.
Sci Rep ; 7(1): 13256, 2017 10 16.
Article in English | MEDLINE | ID: mdl-29038575

ABSTRACT

The black-legged tick Ixodes scapularis transmits the human anaplasmosis agent, Anaplasma phagocytophilum. In this study, we show that A. phagocytophilum specifically up-regulates I. scapularis organic anion transporting polypeptide, isoatp4056 and kynurenine amino transferase (kat), a gene involved in the production of tryptophan metabolite xanthurenic acid (XA), for its survival in ticks. RNAi analysis revealed that knockdown of isoatp4056 expression had no effect on A. phagocytophilum acquisition from the murine host but affected the bacterial survival in tick cells. Knockdown of the expression of kat mRNA alone or in combination with isoatp4056 mRNA significantly affected A. phagocytophilum survival and isoatp4056 expression in tick cells. Exogenous addition of XA induces isoatp4056 expression and A. phagocytophilum burden in both tick salivary glands and tick cells. Electrophoretic mobility shift assays provide further evidence that A. phagocytophilum and XA influences isoatp4056 expression. Collectively, this study provides important novel information in understanding the interplay between molecular pathways manipulated by a rickettsial pathogen to survive in its arthropod vector.


Subject(s)
Arthropods/metabolism , Arthropods/pathogenicity , Organic Anion Transporters/metabolism , Peptides/metabolism , Transaminases/metabolism , Tryptophan/metabolism , Anaplasma phagocytophilum/metabolism , Animals , Humans , Mice , Organic Anion Transporters/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Salivary Glands/metabolism , Ticks/parasitology , Transaminases/genetics
10.
Article in English | MEDLINE | ID: mdl-28861402

ABSTRACT

The obligate intracellular pathogen, Anaplasma phagocytophilum, is the causative agent of life-threatening diseases in humans and animals. A. phagocytophilum is an emerging tick-borne pathogen in the United States, Europe, Africa and Asia, with increasing numbers of infected people and animals every year. It is increasingly recognized that intracellular pathogens modify host cell metabolic pathways to increase infection and transmission in both vertebrate and invertebrate hosts. Recent reports have shown that amino acids are central to the host-pathogen metabolic interaction. In this study, a genome-wide search for components of amino acid metabolic pathways was performed in Ixodes scapularis, the main tick vector of A. phagocytophilum in the United States, for which the genome was recently published. The enzymes involved in the synthesis and degradation pathways of the twenty amino acids were identified. Then, the available transcriptomics and proteomics data was used to characterize the mRNA and protein levels of I. scapularis amino acid metabolic pathway components in response to A. phagocytophilum infection of tick tissues and ISE6 tick cells. Our analysis was focused on the interplay between carbohydrate and amino acid metabolism during A. phagocytophilum infection in ISE6 cells. The results showed that tick cells increase the synthesis of phosphoenolpyruvate (PEP) from tyrosine to control A. phagocytophilum infection. Metabolic pathway analysis suggested that this is achieved by (i) increasing the transcript and protein levels of mitochondrial phosphoenolpyruvate carboxykinase (PEPCK-M), (ii) shunting tyrosine into the tricarboxylic acid (TCA) cycle to increase fumarate and oxaloacetate which will be converted into PEP by PEPCK-M, and (iii) blocking all the pathways that use PEP downstream gluconeogenesis (i.e., de novo serine synthesis pathway (SSP), glyceroneogenesis and gluconeogenesis). While sequestering host PEP may be critical for this bacterium because it cannot actively carry out glycolysis to produce PEP, excess of this metabolite may be toxic for A. phagocytophilum. The present work provides a more comprehensive view of the major amino acid metabolic pathways involved in the response to pathogen infection in ticks, and provides the basis for further studies to develop novel strategies for the control of granulocytic anaplasmosis.


Subject(s)
Anaplasma phagocytophilum/drug effects , Anaplasma phagocytophilum/metabolism , Host-Pathogen Interactions/physiology , Ixodes/microbiology , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate/pharmacology , Tyrosine/metabolism , Amino Acids/metabolism , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/pathogenicity , Anaplasmosis , Animals , Apoptosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbohydrate Metabolism , Cell Line , Citric Acid Cycle , Genome, Bacterial , Gluconeogenesis , Glycolysis , Metabolic Networks and Pathways/genetics , Mitochondria/metabolism , Oxaloacetic Acid/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Proteomics/methods , RNA, Messenger/genetics , Serine/metabolism , Transcriptome
11.
Article in English | MEDLINE | ID: mdl-28725639

ABSTRACT

Anaplasma phagocytophilum transmembrane and surface proteins play a role during infection and multiplication in host neutrophils and tick vector cells. Recently, A. phagocytophilum Major surface protein 4 (MSP4) and Heat shock protein 70 (HSP70) were shown to be localized on the bacterial membrane, with a possible role during pathogen infection in ticks. In this study, we hypothesized that A. phagocytophilum MSP4 and HSP70 have similar functions in tick-pathogen and host-pathogen interactions. To address this hypothesis, herein we characterized the role of these bacterial proteins in interaction and infection of vertebrate host cells. The results showed that A. phagocytophilum MSP4 and HSP70 are involved in host-pathogen interactions, with a role for HSP70 during pathogen infection. The analysis of the potential protective capacity of MSP4 and MSP4-HSP70 antigens in immunized sheep showed that MSP4-HSP70 was only partially protective against pathogen infection. This limited protection may be associated with several factors, including the recognition of non-protective epitopes by IgG in immunized lambs. Nevertheless, these antigens may be combined with other candidate protective antigens for the development of vaccines for the control of human and animal granulocytic anaplasmosis. Focusing on the characterization of host protective immune mechanisms and protein-protein interactions at the host-pathogen interface may lead to the discovery and design of new effective protective antigens.


Subject(s)
Anaplasma phagocytophilum/metabolism , Bacterial Proteins/metabolism , Ehrlichiosis/veterinary , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Sheep Diseases/microbiology , Anaplasma phagocytophilum/genetics , Animals , Bacterial Proteins/genetics , Ehrlichiosis/microbiology , HSP70 Heat-Shock Proteins/genetics , Host-Pathogen Interactions , Membrane Proteins/genetics , Sheep
12.
Mol Cell Probes ; 31: 76-84, 2017 02.
Article in English | MEDLINE | ID: mdl-27618775

ABSTRACT

Autophagy is crucial for maintaining physiological homeostasis, but its role in infectious diseases is not yet adequately understood. The binding of Anaplasma translocated substrate-1 (ATS1) to the human Beclin1 (BECN1) protein is responsible for the modulation of autophagy pathway. ATS1-BECN1 is a novel type of interaction that facilitates Anaplasma phagocytophilum proliferation, leading to intracellular infection via autophagosome induction and segregation from the lysosome. Currently, there is no report of post translational modifications (PTMs) of BECN1 or cross-talk required for ATS-BECN1 complex formation. Prediction/modeling of the cross-talk between phosphorylation and other PTMs (O-ß-glycosylation, sumoylation, methylation and palmitoylation) has been attempted in this study, which might be responsible for regulating function after the interaction of ATS1 with BECN1. PTMs were predicted computationally and mapped onto the interface of the docked ATS1-BECN1 complex. Results show that BECN1 phosphorylation at five residues (Thr91, Ser93, Ser96, Thr141 and Ser234), the interplay with O-ß-glycosylation at three sites (Thr91, Ser93 and Ser96) with ATS1 may be crucial for attachment and, hence, infection. No other PTM site at the BECN1 interface was predicted to associate with ATS1. These findings may have significant clinical implications for understanding the etiology of Anaplasma infection and for therapeutic studies.


Subject(s)
Anaplasma phagocytophilum/metabolism , Autophagosomes/metabolism , Computational Biology/methods , Host-Pathogen Interactions , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Beclin-1/chemistry , Beclin-1/metabolism , Humans , Mice , Models, Molecular , Protein Binding , Protein Structure, Secondary , Rats , Sequence Alignment
13.
Article in English | MEDLINE | ID: mdl-27713867

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.


Subject(s)
Anaplasma phagocytophilum/pathogenicity , Anaplasmosis/microbiology , Cytosol/microbiology , Host-Pathogen Interactions/physiology , Sumoylation/physiology , Vacuoles/microbiology , Anaplasma phagocytophilum/growth & development , Anaplasma phagocytophilum/metabolism , Animals , Bacterial Load , Bacterial Proteins/analysis , Bacterial Proteins/metabolism , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , DNA, Bacterial , Gene Expression , Genes, Bacterial , HEK293 Cells , HL-60 Cells , Humans , Microbial Viability , Microscopy, Confocal , Protein Processing, Post-Translational
14.
Ann Agric Environ Med ; 22(3): 414-7, 2015.
Article in English | MEDLINE | ID: mdl-26403105

ABSTRACT

The article focuses on the clinical and laboratory diagnosis of human granulocytic anaplasmosis (HGA) caused by Anaplasma phagocytophilum infection in one of 28 patients (3.6%; n=1/28 tested samples) with early Lyme borreliosis. The clinical and laboratory results of a 42-year-old patient fulfilled criteria of confirm anaplasmosis and suggest an acute stage of illness. The described case provides strong presumptive evidence that infection in this patient was acquired with a pathogenic strain of A. phagocytophilum through a tick bite. A positive DNA with PCR for A. phagocytophilum infection was sequenced and analyzed phylogenetically. Physicians should consider the possibility of anaplasmosis in patients with early Lyme borreliosis, and A. phagocytophilum should be considered as a differential diagnosis in all patients from an endemic region of potential high risk factors for tick-borne diseases.


Subject(s)
Anaplasma phagocytophilum/isolation & purification , Ehrlichiosis/diagnosis , Tick-Borne Diseases/diagnosis , Adult , Amoxicillin/therapeutic use , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/metabolism , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ceftriaxone/therapeutic use , Chaperonins/genetics , Chaperonins/metabolism , Coinfection/microbiology , Diagnosis, Differential , Ehrlichiosis/drug therapy , Ehrlichiosis/microbiology , Humans , Lyme Disease/microbiology , Male , Molecular Sequence Data , Phylogeny , Poland , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Sequence Analysis, DNA , Tick-Borne Diseases/drug therapy , Tick-Borne Diseases/microbiology , Treatment Outcome
15.
PLoS Pathog ; 11(2): e1004669, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25658707

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 (sLe(x)-capped P-selectin glycoprotein ligand 1. Molecular docking analyses predicted that OmpA residues G61 and K64 interact with the two sLe(x) 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 sLe(x) but express the structurally similar glycan, 6-sulfo-sLe(x), required α2,3-sialic acid and α1,3-fucose and was antagonized by 6-sulfo-sLe(x) antibody. Binding and uptake of OmpA-coated latex beads by myeloid cells was sensitive to sialidase, fucosidase, and sLe(x) 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.


Subject(s)
Anaplasma phagocytophilum , Bacterial Outer Membrane Proteins , Ehrlichiosis , Molecular Docking Simulation , Amino Acid Substitution , Anaplasma phagocytophilum/chemistry , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/metabolism , Anaplasma phagocytophilum/pathogenicity , Animals , Antibodies, Bacterial/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , CHO Cells , Cricetinae , Cricetulus , Ehrlichiosis/genetics , Ehrlichiosis/metabolism , HL-60 Cells , Humans , Mutation, Missense , Protein Binding , Protein Structure, Tertiary
16.
Exp Appl Acarol ; 66(1): 63-81, 2015 May.
Article in English | MEDLINE | ID: mdl-25721255

ABSTRACT

Anaplasma phagocytophilum and Babesia spp. are emerging tick-borne pathogens which can threaten human health. A duplex real-time PCR and qPCRs with primers and probes targeting 97 and 116 bp fragments of 16S rRNA and 18S rRNA genes, respectively, were used for qualitative and quantitative detection of both pathogens in Ixodes ricinus ticks. Altogether 1875 ticks (1084 adults and 791 nymphs) were collected from rural and urban habitats in northern Poland. Of them, at least 0.9% were found to be infected with A. phagocytophilum while 2.5% with Babesia spp. A comparison of the infection rates by the tick stage, the type of area, the collection site, habitats of different tick density and by the month of collection was done. The prevalence of pathogens was significantly lower in nymphs than in adult ticks (p = 0.02) and in rural areas than in urban areas (p = 0.007). Four different 16S rRNA gene variants of A. phagocytophilum were determine, however none of them showed 100% identity with compared sequences isolated from human patients. The dominant Babesia species was B. venatorum. Results of qPCRs with circular and linearized forms of plasmids used as the standards showed significant difference in the pathogen loads (p = 0.001). The copy numbers of A. phagocytophilum and Babesia spp. estimated from the linear plasmids were 28.7 and 5.1 times lower, respectively, when compared with their circular forms, and were accepted as more reliable. The average number of copies of 16S rRNA gene of A. phagocytophilum in the positive I. ricinus samples were 3.39 × 10(5) ± 6.09 × 10(5). The mean copy number of 18S rRNA gene of Babesia spp. was ~2.55 × 10(5) ± 1.04 × 10(6). We confirmed the presence of A. phagocytophilum and Babesia spp. in I. ricinus in both rural and urban environments. The determined low infection rates suggests, however, that the risk for local population and tourists to acquire infection is also low. Moreover, we confirmed recent findings that serious overestimation by circular plasmid DNA makes it less suitable as a standard and that the linear standards should be recommended for qPCR.


Subject(s)
Anaplasma phagocytophilum/isolation & purification , Babesia/isolation & purification , Ixodes/microbiology , Ixodes/parasitology , Anaplasma phagocytophilum/growth & development , Anaplasma phagocytophilum/metabolism , Animals , Babesia/genetics , Babesia/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA, Protozoan/genetics , DNA, Protozoan/metabolism , Environment , Molecular Sequence Data , Poland , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
17.
Methods Mol Biol ; 1197: 169-84, 2014.
Article in English | MEDLINE | ID: mdl-25172281

ABSTRACT

The modulation of host cell apoptosis by bacterial pathogens is critical for their intracellular survival. Several intracellular bacteria achieve this by secreting proteins that interact with apoptosis pathways to inhibit host cell apoptosis. Anaplasma phagocytophilum, which causes human granulocytic anaplasmosis, is such bacterium. The protein Ats-1, translocated from A. phagocytophilum by the bacterial type IV secretion system, localizes to host cell mitochondria, and interferes with apoptosis induction. In this chapter, we present a protocol applied to investigate an anti-apoptotic effect of Ats-1.


Subject(s)
Apoptosis/physiology , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Anaplasma phagocytophilum/metabolism , Cytoplasm/metabolism , Humans , Mitochondria/metabolism
18.
J Med Entomol ; 51(3): 694-701, 2014 May.
Article in English | MEDLINE | ID: mdl-24897864

ABSTRACT

Changes in the structure of managed red pine forests in Wisconsin caused by interacting root- and stem-colonizing insects are associated with increased abundance of the blacklegged tick, Ixodes scapularis Say, in comparison with nonimpacted stands. However, the frequency and variability of the occurrence of tick-borne pathogens in this coniferous forest type across Wisconsin is unknown. Red pine forests were surveyed from 2009 to 2013 to determine the prevalence of Borrelia burgdorferi and Anaplasma phagocytophilum in questing I. scapularis nymphs. Polymerase chain reaction analysis revealed geographical differences in the nymphal infection prevalence (NIP) of these pathogens in red pine forests. In the Kettle Moraine State Forest (KMSF) in southeastern Wisconsin, NIP of B. burgdorferi across all years was 35% (range of 14.5-53.0%). At the Black River State Forest (BRSF) in western Wisconsin, NIP of B. burgdorferi across all years was 26% (range of 10.9-35.5%). Differences in NIP of B. burgdorferi between KMSF and BRSF were statistically significant for 2010 and 2011 and for all years combined (P < 0.05). NIP ofA. phagocytophilum (human agent) averaged 9% (range of 4.6-15.8%) at KMSF and 3% (range of 0-6.4%) at BRSF, and was significantly different between the sites for all years combined (P < 0.05). Differences in coinfection of B. burgdorferi and A. phagocytophilum were not statistically significant between KMSF and BRSF, with an average of 3.4% (range of 1.7-10.5%) and 2.5% (range of 0-5.5%), respectively. In 2013, the density of infected nymphs in KMSF and BRSF was 14 and 30 per 1000m2, respectively, among the highest ever recorded for the state. Differences in the density of nymphs and NIP among sites were neither correlated with environmental factors nor time since tick colonization. These results document significant unexplained variation in tick-borne pathogens between coniferous forests in Wisconsin that warrants further study.


Subject(s)
Anaplasma phagocytophilum/isolation & purification , Borrelia burgdorferi/isolation & purification , Ehrlichiosis/epidemiology , Ixodes/microbiology , Lyme Disease/epidemiology , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/metabolism , Animals , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , Ecosystem , Ehrlichiosis/microbiology , Ixodes/growth & development , Lyme Disease/microbiology , Nymph/growth & development , Nymph/microbiology , Pinus , Polymerase Chain Reaction , Seasons , Wisconsin
19.
BMC Vet Res ; 9: 235, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24283328

ABSTRACT

BACKGROUND: Anaplasma phagocytophilum is a Gram-negative bacterium that replicates obligate intracellularly in neutrophils. It is transmitted by Ixodes spp. ticks and causes acute febrile disease in humans, dogs, horses, cats, and livestock. Because A. phagocytophilum is not transmitted transovarially in Ixodes spp., it is thought to depend on reservoir hosts to complete its life cycle. In Europe, A. phagocytophilum was detected in roe deer, red deer, wild boars, and small mammals. In contrast to roe deer, red deer and wild boars have been considered as reservoir hosts for granulocytic anaplasmosis in humans, dogs, and horses according to groESL- and ankA-based genotyping. A. phagocytophilum variants infecting small mammals in Europe have not been characterized extensively to date. RESULTS: We amplified the total ankA open reading frames of 27 strains from voles and shrews. The analysis revealed that they harboured A. phagocytophilum strains that belonged to a distinct newly described ankA gene cluster. Further, we provide evidence that the heterogeneity of ankA gene sequences might have arisen via recombination. CONCLUSIONS: Based on ankA-based genotyping voles and shrews are unlikely reservoir hosts for granulocytic anaplasmosis in humans, dogs, horses, and livestock in Europe.


Subject(s)
Anaplasma phagocytophilum/metabolism , Arvicolinae , Bacterial Proteins/metabolism , Ehrlichiosis/veterinary , Gene Expression Regulation, Bacterial/physiology , Shrews , Anaplasma phagocytophilum/genetics , Animals , Bacterial Proteins/genetics , Ehrlichiosis/epidemiology , Ehrlichiosis/microbiology , Europe/epidemiology
20.
Vet Microbiol ; 167(3-4): 484-93, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-23992798

ABSTRACT

Sheep infected with Anaplasma phagocytophilum, the causative agent of tick-borne fever (TBF), develop humoral immune responses 7-14 days after infection. Those individuals that survive acute TBF develop persistent infection, which may last for several months or even for life. The persistence of infection and recurrent bacteraemia is thought to be due to p44-mediated antigenic variation. The present study mapped linear B-cell epitopes within the hypervariable region (HVR) of the surface membrane protein P44 and investigated whether the development of antibodies against B cell epitopes within the HVR was preceded by the expression of p44 variants. Serum samples obtained from five sheep infected with the Old Sourhope strain of A. phagocytophilum (AP-OS) were used to detect antibody reactivity against 20-mer overlapping synthetic peptides spanning the HVR of two p44 variants which were expressed during primary bacteraemia and 3 variants expressed during secondary bacteraemia. The results showed that all five p44 variants of AP-OS have dominant B-cell epitopes residing mainly in the 3rd and 7th of the 10-11 peptides mapping each HVR. Antibody reactivity against peptides of the HVR of all the variants was characterised by a gradual rise, reaching peak levels in samples obtained 24 days post-inoculation (dpi) followed by a gradual decline. Anamnestic responses to whole cell antigens and to some of the dominant antigenic epitopes were detected in some of the animals, which were monitored for 52 weeks.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Ehrlichiosis/veterinary , Gene Expression Regulation/immunology , Sheep Diseases/immunology , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/immunology , Anaplasma phagocytophilum/metabolism , Animals , Antibodies, Bacterial/blood , Antigenic Variation , Bacteremia/immunology , Ehrlichiosis/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Molecular Sequence Data , Sheep/genetics , Sheep/immunology , Time
SELECTION OF CITATIONS
SEARCH DETAIL
...