Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Public Health ; 8: 558283, 2020.
Article in English | MEDLINE | ID: mdl-33194960

ABSTRACT

Anaplasma phagocytophilum subverts neutrophil function permitting intracellular survival, propagation and transmission. Sustained pro-inflammatory response, recruitment of new host cells for population expansion, and delayed apoptosis are associated with prolonged nuclear presence of NF-κB. We investigated NF-κB signaling and transcriptional activity with A. phagocytophilum infection using inhibitors of NF-κB signaling pathways, and through silencing of signaling pathway genes. How inhibitors or silencing affected A. phagocytophilum growth, inflammatory response (transcription of the κB-enhanced genes CXCL8 and MMP9), and NF-κB signaling pathway gene expression were tested. Among A. phagocytophilum-infected HL-60 cells, nuclear NF-κB p50, p65, and p52 were detected by immunoblots or iTRAQ proteomics. A. phagocytophilum growth was affected most by the IKKαß inhibitor wedelolactone (reductions of 96 to 99%) as compared with SC-514 that selectively inhibits IKKß, illustrating a role for the non-canonical pathway. Wedelolactone inhibited transcription of both CXCL8 (p = 0.001) and MMP9 (p = 0.002) in infected cells. Compared to uninfected THP-1 cells, A. phagocytophilum infection led to >2-fold down regulation of 64 of 92 NF-κB signaling pathway genes, and >2-fold increased expression in only 4. Wedelolactone and SC-514 reversed downregulation in all 64 and 45, respectively, of the genes down-regulated by infection, but decreased expression in 1 gene with SC-514 only. Silencing of 20 NF-κB signal pathway genes increased bacterial growth in 12 (IRAK1, MAP3K1, NFKB1B, MAP3K7, TICAM2, TLR3, TRADD, TRAF3, CHUK, IRAK2, LTBR, and MALT1). Most findings support canonical pathway activation; however, the presence of NFKB2 in infected cell nuclei, selective non-canonical pathway inhibitors that dampen CXCL8 and MMP9 transcription with infection, upregulation of non-canonical pathway target genes CCL13 and CCL19, enhanced bacterial growth with TRAF3 and LTBR silencing provide evidence for non-canonical pathway signaling. Whether this impacts distinct inflammatory processes that underlie disease, and whether and how A. phagocytophilum subverts NF-κB signaling via these pathways, need to be investigated.


Subject(s)
Anaplasma phagocytophilum , Ehrlichiosis/immunology , NF-kappa B , Signal Transduction , Anaplasma phagocytophilum/genetics , HL-60 Cells , Humans , I-kappa B Kinase/metabolism , NF-kappa B/genetics , TNF Receptor-Associated Factor 3
2.
BMC Biol ; 18(1): 70, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32560686

ABSTRACT

BACKGROUND: Fleas (Insecta: Siphonaptera) are small flightless parasites of birds and mammals; their blood-feeding can transmit many serious pathogens (i.e., the etiological agents of bubonic plague, endemic and murine typhus). The lack of flea genome assemblies has hindered research, especially comparisons to other disease vectors. Accordingly, we sequenced the genome of the cat flea, Ctenocephalides felis, an insect with substantial human health and veterinary importance across the globe. RESULTS: By combining Illumina and PacBio sequencing of DNA derived from multiple inbred female fleas with Hi-C scaffolding techniques, we generated a chromosome-level genome assembly for C. felis. Unexpectedly, our assembly revealed extensive gene duplication across the entire genome, exemplified by ~ 38% of protein-coding genes with two or more copies and over 4000 tRNA genes. A broad range of genome size determinations (433-551 Mb) for individual fleas sampled across different populations supports the widespread presence of fluctuating copy number variation (CNV) in C. felis. Similarly, broad genome sizes were also calculated for individuals of Xenopsylla cheopis (Oriental rat flea), indicating that this remarkable "genome-in-flux" phenomenon could be a siphonapteran-wide trait. Finally, from the C. felis sequence reads, we also generated closed genomes for two novel strains of Wolbachia, one parasitic and one symbiotic, found to co-infect individual fleas. CONCLUSION: Rampant CNV in C. felis has dire implications for gene-targeting pest control measures and stands to complicate standard normalization procedures utilized in comparative transcriptomics analysis. Coupled with co-infection by novel Wolbachia endosymbionts-potential tools for blocking pathogen transmission-these oddities highlight a unique and underappreciated disease vector.


Subject(s)
Ctenocephalides/genetics , DNA Copy Number Variations , Gene Duplication , Genome Size , Animals , Chromosomes , Female , Male
3.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-29084898

ABSTRACT

Rickettsia species are obligate intracellular bacteria with both conserved and lineage-specific strategies for invading and surviving within eukaryotic cells. One variable component of Rickettsia biology involves arthropod vectors: for instance, typhus group rickettsiae are principally vectored by insects (i.e., lice and fleas), whereas spotted fever group rickettsiae are exclusively vectored by ticks. For flea-borne Rickettsia typhi, the etiological agent of murine typhus, research on vertebrate host biology is facilitated using cell lines and animal models. However, due to the lack of any stable flea cell line or a published flea genome sequence, little is known regarding R. typhi biology in flea vectors that, importantly, do not suffer lethality due to R. typhi infection. To address if fleas combat rickettsial infection, we characterized the cat flea (Ctenocephalides felis) innate immune response to R. typhi Initially, we determined that R. typhi infects Drosophila cells and increases antimicrobial peptide (AMP) gene expression, indicating immune pathway activation. While bioinformatics analysis of the C. felis transcriptome identified homologs to all of the Drosophila immune deficiency (IMD) and Toll pathway components, an AMP gene expression profile in Drosophila cells indicated IMD pathway activation upon rickettsial infection. Accordingly, we assessed R. typhi-mediated flea IMD pathway activation in vivo using small interfering RNA (siRNA)-mediated knockdown. Knockdown of Relish and Imd increased R. typhi infection levels, implicating the IMD pathway as a critical regulator of R. typhi burden in C. felis These data suggest that targeting the IMD pathway could minimize the spread of R. typhi, and potentially other human pathogens, vectored by fleas.


Subject(s)
Ctenocephalides/immunology , Flea Infestations/immunology , Rickettsia Infections/immunology , Rickettsia typhi/immunology , Signal Transduction/immunology , Siphonaptera/immunology , Adenosine Monophosphate/metabolism , Animals , Cats , Cell Line , Chlorocebus aethiops , Ctenocephalides/microbiology , Drosophila/microbiology , Flea Infestations/microbiology , Gene Expression/immunology , Immunity, Innate/immunology , Insect Vectors/immunology , Insect Vectors/microbiology , Siphonaptera/microbiology , Typhus, Endemic Flea-Borne/immunology , Typhus, Endemic Flea-Borne/microbiology , Vero Cells
4.
Infect Immun ; 84(12): 3496-3506, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27698019

ABSTRACT

Rickettsiae are obligate intracellular pathogens that induce their uptake into nonphagocytic cells; however, the events instigating this process are incompletely understood. Importantly, diverse Rickettsia species are predicted to utilize divergent mechanisms to colonize host cells, as nearly all adhesins and effectors involved in host cell entry are differentially encoded in diverse Rickettsia species. One particular effector, RalF, a Sec7 domain-containing protein that functions as a guanine nucleotide exchange factor of ADP-ribosylation factors (Arfs), is critical for Rickettsia typhi (typhus group rickettsiae) entry but pseudogenized or absent from spotted fever group rickettsiae. Secreted early during R. typhi infection, RalF localizes to the host plasma membrane and interacts with host ADP-ribosylation factor 6 (Arf6). Herein, we demonstrate that RalF activates Arf6, a process reliant on a conserved Glu within the RalF Sec7 domain. Furthermore, Arf6 is activated early during infection, with GTP-bound Arf6 localized to the R. typhi entry foci. The regulation of phosphatidylinositol 4-phosphate 5-kinase (PIP5K), which generates PI(4,5)P2, by activated Arf6 is instrumental for bacterial entry, corresponding to the requirement of PI(4,5)P2 for R. typhi entry. PI(3,4,5)P3 is then synthesized at the entry foci, followed by the accumulation of PI(3)P on the short-lived vacuole. Inhibition of phosphoinositide 3-kinases, responsible for the synthesis of PI(3,4,5)P3 and PI(3)P, negatively affects R. typhi infection. Collectively, these results identify RalF as the first bacterial effector to directly activate Arf6, a process that initiates alterations in phosphoinositol metabolism critical for a lineage-specific Rickettsia entry mechanism.


Subject(s)
ADP-Ribosylation Factors/metabolism , Bacterial Proteins/metabolism , Phosphatidylinositols/metabolism , Rickettsia typhi/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Animals , Bacterial Proteins/genetics , Chlorocebus aethiops , Gene Expression Regulation/physiology , Guanosine Triphosphate , HeLa Cells , Humans , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Plasmids , Protein Binding , Rickettsia typhi/genetics , Vero Cells
5.
Pathog Dis ; 74(6)2016 08.
Article in English | MEDLINE | ID: mdl-27307105

ABSTRACT

Many prokaryotes utilize type IV secretion systems (T4SSs) to translocate substrates (e.g. nucleoprotein, DNA, protein) across the cell envelope, and/or to elaborate surface structures (i.e. pili or adhesins). Among eight distinct T4SS classes, P-T4SSs are typified by the Agrobacterium tumefaciens vir T4SS, which is comprised of 12 scaffold components (VirB1-VirB11, VirD4). While most P-T4SSs include all 12 Vir proteins, some differ from the vir archetype by either containing additional scaffold components not analogous to Vir proteins or lacking one or more of the Vir proteins. In a special case, the Rickettsiales vir homolog (rvh) P-T4SS comprises unprecedented gene family expansion. rvh contains three families of gene duplications (rvhB9, rvhB8, rvhB4): RvhB9,8,4-I are conserved relative to equivalents in other P-T4SSs, while RvhB9,8,4-II have evolved atypical features that deviate substantially from other homologs. Furthermore, rvh contains five VirB6-like genes (rvhB6a-e), which are tandemly arrayed and contain large N- and C-terminal extensions. Our work herein focuses on the complexity underpinned by rvh gene family expansion. Furthermore, we describe an RvhB10 insertion, which occurs in a region that forms the T4SS pore. The significance of these curious properties to rvh structure and function is evaluated, shedding light on a highly complex T4SS.


Subject(s)
Gene Duplication , Genes, Bacterial , Multigene Family , Rickettsia/physiology , Type IV Secretion Systems , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Gene Order , Models, Molecular , Mutagenesis, Insertional , Position-Specific Scoring Matrices , Protein Binding , Protein Conformation , Protein Domains , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Rickettsia Infections/microbiology
6.
mBio ; 6(6): e01867-15, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26646013

ABSTRACT

UNLABELLED: Prokaryotes use type IV secretion systems (T4SSs) to translocate substrates (e.g., nucleoprotein, DNA, and protein) and/or elaborate surface structures (i.e., pili or adhesins). Bacterial genomes may encode multiple T4SSs, e.g., there are three functionally divergent T4SSs in some Bartonella species (vir, vbh, and trw). In a unique case, most rickettsial species encode a T4SS (rvh) enriched with gene duplication. Within single genomes, the evolutionary and functional implications of cross-system interchangeability of analogous T4SS protein components remains poorly understood. To lend insight into cross-system interchangeability, we analyzed the VirB8 family of T4SS channel proteins. Crystal structures of three VirB8 and two TrwG Bartonella proteins revealed highly conserved C-terminal periplasmic domain folds and dimerization interfaces, despite tremendous sequence divergence. This implies remarkable structural constraints for VirB8 components in the assembly of a functional T4SS. VirB8/TrwG heterodimers, determined via bacterial two-hybrid assays and molecular modeling, indicate that differential expression of trw and vir systems is the likely barrier to VirB8-TrwG interchangeability. We also determined the crystal structure of Rickettsia typhi RvhB8-II and modeled its coexpressed divergent paralog RvhB8-I. Remarkably, while RvhB8-I dimerizes and is structurally similar to other VirB8 proteins, the RvhB8-II dimer interface deviates substantially from other VirB8 structures, potentially preventing RvhB8-I/RvhB8-II heterodimerization. For the rvh T4SS, the evolution of divergent VirB8 paralogs implies a functional diversification that is unknown in other T4SSs. Collectively, our data identify two different constraints (spatiotemporal for Bartonella trw and vir T4SSs and structural for rvh T4SSs) that mediate the functionality of multiple divergent T4SSs within a single bacterium. IMPORTANCE: Assembly of multiprotein complexes at the right time and at the right cellular location is a fundamentally important task for any organism. In this respect, bacteria that express multiple analogous type IV secretion systems (T4SSs), each composed of around 12 different components, face an overwhelming complexity. Our work here presents the first structural investigation on factors regulating the maintenance of multiple T4SSs within a single bacterium. The structural data imply that the T4SS-expressing bacteria rely on two strategies to prevent cross-system interchangeability: (i) tight temporal regulation of expression or (ii) rapid diversification of the T4SS components. T4SSs are ideal drug targets provided that no analogous counterparts are known from eukaryotes. Drugs targeting the barriers to cross-system interchangeability (i.e., regulators) could dysregulate the structural and functional independence of discrete systems, potentially creating interference that prevents their efficient coordination throughout bacterial infection.


Subject(s)
Bartonella/chemistry , Bartonella/metabolism , Rickettsia typhi/chemistry , Rickettsia typhi/metabolism , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Substrate Specificity , Two-Hybrid System Techniques
7.
PLoS Pathog ; 11(8): e1005115, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26291822

ABSTRACT

Bacterial Sec7-domain-containing proteins (RalF) are known only from species of Legionella and Rickettsia, which have facultative and obligate intracellular lifestyles, respectively. L. pneumophila RalF, a type IV secretion system (T4SS) effector, is a guanine nucleotide exchange factor (GEF) of ADP-ribosylation factors (Arfs), activating and recruiting host Arf1 to the Legionella-containing vacuole. In contrast, previous in vitro studies showed R. prowazekii (Typhus Group) RalF is a functional Arf-GEF that localizes to the host plasma membrane and interacts with the actin cytoskeleton via a unique C-terminal domain. As RalF is differentially encoded across Rickettsia species (e.g., pseudogenized in all Spotted Fever Group species), it may function in lineage-specific biology and pathogenicity. Herein, we demonstrate RalF of R. typhi (Typhus Group) interacts with the Rickettsia T4SS coupling protein (RvhD4) via its proximal C-terminal sequence. RalF is expressed early during infection, with its inactivation via antibody blocking significantly reducing R. typhi host cell invasion. For R. typhi and R. felis (Transitional Group), RalF ectopic expression revealed subcellular localization with the host plasma membrane and actin cytoskeleton. Remarkably, R. bellii (Ancestral Group) RalF showed perinuclear localization reminiscent of ectopically expressed Legionella RalF, for which it shares several structural features. For R. typhi, RalF co-localization with Arf6 and PI(4,5)P2 at entry foci on the host plasma membrane was determined to be critical for invasion. Thus, we propose recruitment of PI(4,5)P2 at entry foci, mediated by RalF activation of Arf6, initiates actin remodeling and ultimately facilitates bacterial invasion. Collectively, our characterization of RalF as an invasin suggests that, despite carrying a similar Arf-GEF unknown from other bacteria, different intracellular lifestyles across Rickettsia and Legionella species have driven divergent roles for RalF during infection. Furthermore, our identification of lineage-specific Arf-GEF utilization across some rickettsial species illustrates different pathogenicity factors that define diverse agents of rickettsial diseases.


Subject(s)
ADP-Ribosylation Factors/metabolism , Bacterial Proteins/metabolism , Rickettsia Infections/metabolism , Rickettsia/pathogenicity , Virus Internalization , ADP-Ribosylation Factors/genetics , Animals , Bacterial Proteins/genetics , Cell Line , Computational Biology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunoblotting , Phylogeny , Protein Conformation , Rickettsia/genetics , Rickettsia/metabolism , Rickettsia Infections/genetics , Transfection
8.
Cell Microbiol ; 17(11): 1640-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25996657

ABSTRACT

Control of host epigenetics is becoming evident as a mechanism by which symbionts and pathogens survive. Anaplasma phagocytophilum, an obligate intracellular bacterium, down-regulates multiple host defence genes where histone deacetylase 1 (HDAC1) binds and histone 3 is deacetylated at their promoters, including the NADPH oxidase component, CYBB. How HDAC1 is targeted to defence gene promoters is unknown. Ankyrin A (AnkA), an A. phagocytophilum type IV secretion system effector, enters the granulocyte nucleus, binds stretches of AT-rich DNA and alters transcription of antimicrobial defence genes, including down-regulation of CYBB. Here we found AnkA binds to a predicted matrix attachment region in the proximal CYBB promoter. Using the CYBB promoter as a model of cis-gene silencing, we interrogated the mechanism of AnkA-mediated CYBB repression. The N-terminus of AnkA was critical for nuclear localization, the central ANK repeats and C-terminus were important for DNA binding, and most promoter activity localized to the central ANK repeats. Furthermore, a direct interaction between AnkA and HDAC1 was detected at the CYBB promoter, and was critical for AnkA-mediated CYBB repression. This novel microbial manipulation of host chromatin and gene expression provides important evidence of the direct effects that prokaryotic nuclear effectors can exert over host transcription and function.


Subject(s)
Anaplasma phagocytophilum/physiology , Ankyrins/metabolism , Down-Regulation , Histone Deacetylase 1/metabolism , Host-Pathogen Interactions , Membrane Glycoproteins/biosynthesis , NADPH Oxidases/biosynthesis , Virulence Factors/metabolism , Cell Line , Chromatin , Humans , NADPH Oxidase 2 , Promoter Regions, Genetic , Protein Binding
9.
Front Genet ; 5: 274, 2014.
Article in English | MEDLINE | ID: mdl-25177343

ABSTRACT

Obligate intracellular pathogenic bacteria evolved to manipulate their host cells with a limited range of proteins constrained by their compact genomes. The harsh environment of a phagocytic defense cell is one that challenges the majority of commensal and pathogenic bacteria; yet, these are the obligatory vertebrate homes for important pathogenic species in the Anaplasmataceae family. Survival requires that the parasite fundamentally alter the native functions of the cell to allow its entry, intracellular replication, and transmission to a hematophagous arthropod. The small genomic repertoires encode several eukaryotic-like proteins, including ankyrin A (AnkA) of Anaplasma phagocytophilum and Ank200 and tandem-repeat containing proteins of Ehrlichia chaffeensis that localize to the host cell nucleus and directly bind DNA. As a model, A. phagocytophilum AnkA appears to directly alter host cell gene expression by recruiting chromatin modifying enzymes such as histone deacetylases and methyltransferases or by acting directly on transcription in cis. While cis binding could feasibly alter limited ranges of genes and cellular functions, the complex and dramatic alterations in transcription observed with infection are difficult to explain on the basis of individually targeted genes. We hypothesize that nucleomodulins can act broadly, even genome-wide, to affect entire chromosomal neighborhoods and topologically associating chromatin domains by recruiting chromatin remodeling complexes or by altering the folding patterns of chromatin that bring distant regulatory regions together to coordinate control of transcriptional reprogramming. This review focuses on the A. phagocytophilum nucleomodulin AnkA, how it impacts host cell transcriptional responses, and current investigations that seek to determine how these multifunctional eukaryotic-like proteins facilitate epigenetic alterations and cellular reprogramming at the chromosomal level.

10.
Pathog Dis ; 71(1): 55-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24376092

ABSTRACT

Anaplasma phagocytophilum, an obligate intracellular bacterium, modifies functions of its in vivo host, the neutrophil. The challenges of using neutrophils ex vivo necessitate cell line models. However, cell line infections do not currently mimic ex vivo neutrophil infection characteristics. To understand these discrepancies, we compared infection of cell lines to ex vivo human neutrophils and differentiated hematopoietic stem cells with regard to infection capacity, oxidative burst, host defense gene expression, and differentiation. Using established methods, marked ex vivo neutrophil infection heterogeneity was observed at 24-48 h necessitating cell sorting to obtain homogeneously infected cells at levels observed in vivo. Moreover, gene expression of infected cell lines differed markedly from the prior standard of unsorted infected neutrophils. Differentiated HL-60 cells sustained similar infection levels to neutrophils in vivo and closely mimicked functional and transcriptional changes of sorted infected neutrophils. Thus, care must be exercised using ex vivo neutrophils for A. phagocytophilum infection studies because a major determinant of transcriptional and functional changes among all cells was the intracellular bacteria quantity. Furthermore, comparisons of ex vivo neutrophils and the surrogate HL-60 cell model allowed the determination that specific cellular functions and transcriptional programs are targeted by the bacterium without significantly modifying differentiation.


Subject(s)
Anaplasma phagocytophilum/growth & development , Granulocytes/microbiology , Models, Theoretical , Cells, Cultured , Gene Expression Profiling , Host-Pathogen Interactions , Humans
11.
Infect Disord Drug Targets ; 12(5): 380-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23082961

ABSTRACT

Bacterial pathogens can alter global host gene expression via histone modifications and chromatin remodeling in order to subvert host responses, including those involved with innate immunity, allowing for bacterial survival. Shigella flexneri, Listeria monocytogenes, Chlamydia trachomatis, and Anaplasma phagocytophilum express effector proteins that modify host histones and chromatin structure. A. phagocytophilum modulates granulocyte respiratory burst in part by dampening transcription of several key phagocyte oxidase genes. The A. phagocytophilum protein AnkA localizes to the myeloid cell nucleus where it binds AT-rich regions in the CYBB promoter and decreases its transcription. AT-rich regions of DNA are characteristic of matrix attachment regions (MARs) which are critical for chromatin structure and transcription. MAR-binding proteins, such as SATB1, interact with histone modifying enzymes resulting in altered gene expression. With A. phagocytophilum infection, histone deacetylase 1 (HDAC1) expression is increased and histone H3 acetylation is decreased at the CYBB promoter, suggesting a role for AnkA in altering host epigenetics and modulating gene transcription, at this, and perhaps other loci. This review will focus on how bacterial pathogens alter host epigenetics, by specifically examining A. phagocytophilum AnkA cis-regulation of CYBB transcription and epigenetic changes associated with infection.


Subject(s)
Anaplasma phagocytophilum/pathogenicity , Chromatin Assembly and Disassembly , Ehrlichiosis/physiopathology , Animals , Ehrlichiosis/microbiology , Epigenesis, Genetic , Gene Expression Regulation , Humans , Immunity, Innate , Membrane Glycoproteins/genetics , NADPH Oxidase 2 , NADPH Oxidases/genetics , Transcription, Genetic
12.
Infect Immun ; 77(6): 2385-91, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19307214

ABSTRACT

Coevolution of intracellular bacterial pathogens and their host cells resulted in the appearance of effector molecules that when translocated into the host cell modulate its function, facilitating bacterial survival within the hostile host environment. Some of these effectors interact with host chromatin and other nuclear components. In this report, we show that the AnkA protein of Anaplasma phagocytophilum, which is translocated into the host cell nucleus, interacts with gene regulatory regions of host chromatin and is involved in downregulating expression of CYBB (gp91(phox)) and other key host defense genes. AnkA effector protein rapidly accumulated in nuclei of infected cells coincident with changes in CYBB transcription. AnkA interacted with transcriptional regulatory regions of the CYBB locus at sites where transcriptional regulators bind. AnkA binding to DNA occurred at regions with high AT contents. Mutation of AT stretches at these sites abrogated AnkA binding. Histone H3 acetylation decreased dramatically at the CYBB locus during A. phagocytophilum infection, particularly around AnkA binding sites. Transcription of CYBB and other defense genes was significantly decreased in AnkA-transfected HL-60 cells. These data suggest a mechanism by which intracellular pathogens directly regulate host cell gene expression mediated by nuclear effectors and changes in host chromatin structure.


Subject(s)
Anaplasma phagocytophilum/physiology , Bacterial Proteins/metabolism , Gene Silencing , Membrane Glycoproteins/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Cell Line , Cell Nucleus/chemistry , DNA/metabolism , Gene Expression Profiling , Humans , NADPH Oxidase 2 , Protein Binding
13.
J Virol ; 82(12): 5951-61, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400856

ABSTRACT

During retroviral maturation, the CA protein undergoes dramatic structural changes and establishes unique intermolecular interfaces in the mature capsid shell that are different from those that existed in the immature precursor. The most conserved region of CA, the major homology region (MHR), has been implicated in both immature and mature assembly, although the precise contribution of the MHR residues to each event has been largely undefined. To test the roles of specific MHR residues in mature capsid assembly, an in vitro system was developed that allowed for the first-time formation of Rous sarcoma virus CA into structures resembling authentic capsids. The ability of CA to assemble organized structures was destroyed by substitutions of two conserved hydrophobic MHR residues and restored by second-site suppressors, demonstrating that these MHR residues are required for the proper assembly of mature capsids in addition to any role that these amino acids may play in immature particle assembly. The defect caused by the MHR mutations was identified as an early step in the capsid assembly process. The results provide strong evidence for a model in which the hydrophobic residues of the MHR control a conformational reorganization of CA that is needed to initiate capsid assembly and suggest that the formation of an interdomain interaction occurs early during maturation.


Subject(s)
Capsid Proteins/chemistry , Capsid/physiology , Retroviridae/chemistry , Virus Assembly , Amino Acid Sequence , Amino Acid Substitution , Capsid/ultrastructure , Capsid Proteins/genetics , Capsid Proteins/isolation & purification , Capsid Proteins/ultrastructure , Circular Dichroism , Hydrophobic and Hydrophilic Interactions , Models, Chemical , Molecular Sequence Data , Protein Denaturation , Protein Structure, Secondary , Protein Structure, Tertiary , Retroviridae/genetics , Rous sarcoma virus/chemistry , Rous sarcoma virus/genetics , Sequence Homology, Amino Acid , Spectrophotometry, Ultraviolet , Thermodynamics , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...