Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Cells ; 12(22)2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37998332

RESUMEN

Activating inflammatory caspases and releasing pro-inflammatory mediators are two essential functions of inflammasomes which are triggered in response to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). The canonical inflammasome pathway involves the activation of inflammasome and its downstream pathway via the adaptor ASC protein, which causes caspase 1 activation and, eventually, the cleavage of pro-IL-1b and pro-IL-18. The non-canonical inflammasome pathway is induced upon detecting cytosolic lipopolysaccharide (LPS) by NLRP3 inflammasome in Gram-negative bacteria. The activation of NLRP3 triggers the cleavage of murine caspase 11 (human caspase 4 or caspase 5), which results in the formation of pores (via gasdermin) to cause pyroptosis. Ehrlichia is an obligately intracellular bacterium which is responsible for causing human monocytic ehrlichiosis (HME), a potentially lethal disease similar to toxic shock syndrome and septic shock syndrome. Several studies have indicated that canonical and non-canonical inflammasome activation is a crucial pathogenic mechanism that induces dysregulated inflammation and host cellular death in the pathophysiology of HME. Mechanistically, the activation of canonical and non-canonical inflammasome pathways affected by virulent Ehrlichia infection is due to a block in autophagy. This review aims to explore the significance of non-canonical inflammasomes in ehrlichiosis, and how the pathways involving caspases (with the exception of caspase 1) contribute to the pathophysiology of severe and fatal ehrlichiosis. Improving our understanding of the non-canonical inflammatory pathway that cause cell death and inflammation in ehrlichiosis will help the advancement of innovative therapeutic, preventative, and diagnostic approaches to the treatment of ehrlichiosis.


Asunto(s)
Ehrlichiosis , Inflamasomas , Animales , Ratones , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Caspasa 1 , Ehrlichiosis/metabolismo , Ehrlichiosis/patología , Caspasas/metabolismo , Inflamación , Piroptosis
2.
PLoS Pathog ; 18(5): e1010345, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35576232

RESUMEN

Ehrlichia chaffeensis (E. chaffeensis) has evolved eukaryotic ligand mimicry to repurpose multiple cellular signaling pathways for immune evasion. In this investigation, we demonstrate that TRP120 has a novel repetitive short linear motif (SLiM) that activates the evolutionarily conserved Hedgehog (Hh) signaling pathway to inhibit apoptosis. In silico analysis revealed that TRP120 has sequence and functional similarity with Hh ligands and a candidate Hh ligand SLiM was identified. siRNA knockdown of Hh signaling and transcriptional components significantly reduced infection. Co-immunoprecipitation and surface plasmon resonance demonstrated that rTRP120-TR interacted directly with Hh receptor Patched-2 (PTCH2). E. chaffeensis infection resulted in early upregulation of Hh transcription factor GLI-1 and regulation of Hh target genes. Moreover, soluble recombinant TRP120 (rTRP120) activated Hh and induced gene expression consistent with the eukaryotic Hh ligand. The TRP120-Hh-SLiM (NPEVLIKD) induced nuclear translocation of GLI-1 in THP-1 cells and primary human monocytes and induced a rapid and expansive activation of Hh pathway target genes. Furthermore, Hh activation was blocked by an α-TRP120-Hh-SLiM antibody. TRP120-Hh-SLiM significantly increased levels of Hh target, anti-apoptotic protein B-cell lymphoma 2 (BCL-2), and siRNA knockdown of BCL-2 dramatically inhibited infection. Blocking Hh signaling with the inhibitor Vismodegib, induced a pro-apoptotic cellular program defined by decreased mitochondria membrane potential, significant reductions in BCL-2, activation of caspase 3 and 9, and increased apoptotic cells. This study reveals a novel E. chaffeensis SLiM ligand mimetic that activates Hh signaling to maintain E. chaffeensis infection by engaging a BCL-2 anti-apoptotic cellular program.


Asunto(s)
Ehrlichia chaffeensis , Ehrlichiosis , Proteínas Bacterianas/metabolismo , Ehrlichia chaffeensis/genética , Ehrlichiosis/metabolismo , Proteínas Hedgehog/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Ligandos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
3.
mBio ; 12(5): e0229921, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34544283

RESUMEN

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.


Asunto(s)
Anaplasma phagocytophilum/crecimiento & desarrollo , Anaplasma phagocytophilum/metabolismo , Colesterol/metabolismo , Ehrlichiosis/microbiología , Ezetimiba/farmacología , Proteínas de la Membrana/metabolismo , Proteína Niemann-Pick C1/metabolismo , Anaplasma phagocytophilum/efectos de los fármacos , Anaplasma phagocytophilum/genética , Transporte Biológico , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Ehrlichiosis/genética , Ehrlichiosis/metabolismo , Interacciones Huésped-Patógeno , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/microbiología , Proteínas de la Membrana/genética , Proteína Niemann-Pick C1/genética , Unión Proteica , Transporte de Proteínas
4.
PLoS Pathog ; 16(4): e1008541, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32353058

RESUMEN

Ehrlichia chaffeensis (E. chaffeensis) exploits evolutionarily conserved Notch and Wnt host cell signaling pathways to downregulate innate immune host defenses and promote infection. The multifunctional E. chaffeensis TRP120 effector which has HECT E3 ubiquitin ligase activity, interacts with the host nuclear tumor suppressor F-BOX and WD domain repeating-containing 7 (FBW7). FBW7 is the substrate recognition subunit of the Skp1-cullin-1-FBOX E3 ubiquitin (Ub) ligase complex (SCF) known to negatively regulate a network of oncoproteins (Notch, cyclin E, c-Jun, MCL1 and cMYC). In this study, we demonstrate that TRP120 and FBW7 colocalize strongly in the nucleus by confocal immunofluorescent microscopy and interactions between TRP120 and FBW7 FBOX and WD40 domains were demonstrated by ectopic expression and co-immunoprecipitation. Although FBW7 gene expression increased during E. chaffeensis infection, FBW7 levels significantly decreased (>70%) by 72 h post infection. Moreover, an iRNA knockdown of FBW7 coincided with increased E. chaffeensis infection and levels of Notch intracellular domain (NICD), phosphorylated c-Jun, MCL-1 and cMYC, which are negatively regulated by FBW7. An increase in FBW7 K48 ubiquitination was detected during infection by co-IP, and FBW7 degradation was inhibited in infected cells treated with the proteasomal inhibitor bortezomib. Direct TRP120 ubiquitination of native and recombinant FBW7 was demonstrated in vitro and confirmed by ectopic expression of TRP120 HECT Ub ligase catalytic site mutant. This study identifies the tumor suppressor, FBW7, as a TRP120 HECT E3 Ub ligase substrate, and demonstrates that TRP120 ligase activity promotes ehrlichial infection by degrading FBW7 to maintain stability of Notch and other oncoproteins involved in cell survival and apoptosis.


Asunto(s)
Ehrlichia chaffeensis/metabolismo , Ehrlichiosis/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Apoptosis/fisiología , Proteínas Bacterianas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ehrlichia chaffeensis/genética , Ehrlichia chaffeensis/fisiología , Ehrlichiosis/metabolismo , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Interacciones Huésped-Patógeno , Humanos , Proteínas Oncogénicas/genética , Unión Proteica/fisiología , Células THP-1 , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
5.
mBio ; 11(1)2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31992623

RESUMEN

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.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Anaplasma phagocytophilum/fisiología , Ehrlichiosis/metabolismo , Ehrlichiosis/microbiología , Interacciones Huésped-Patógeno , Proteína Disulfuro Isomerasas/metabolismo , Adhesinas Bacterianas/química , Animales , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Ratones , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Tiorredoxinas/metabolismo
6.
Small GTPases ; 10(5): 343-349, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-28650718

RESUMEN

Intracellular pathogens often exploit RAB functions to establish a safe haven in which to survive and proliferate. Ehrlichia chaffeensis, an obligatory intracellular bacterium, resides in specialized membrane-bound inclusions that have early endosome-like characteristics, e.g., resident RAB5 GTPase and RAB5 effectors, including VPS34 (the catalytic subunit of class III phosphatidylinositol 3-kinase), but the inclusions lack late endosomal or lysosomal markers. Within inclusions, Ehrlichia obtains host-derived nutrients by inducing RAB5-regulated autophagy using Ehrlichia translocated factor-1 deployed by its type IV secretion system. This manipulation of RAB5 by a bacterial molecule offers a simple strategy for Ehrlichia to avoid destruction in lysosomes and obtain nutrients, membrane components, and a homeostatic intra-host-cell environment in which to grow.


Asunto(s)
Muerte Celular Autofágica , Ehrlichia chaffeensis/fisiología , Ehrlichiosis/metabolismo , Interacciones Huésped-Parásitos/fisiología , Sistemas de Secreción Tipo IV/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Animales , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Ehrlichiosis/patología , Endosomas/metabolismo , Endosomas/microbiología , Humanos , Lisosomas/metabolismo , Lisosomas/microbiología
7.
Sci Rep ; 8(1): 14224, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30242261

RESUMEN

The carbohydrate Galα1-3Galß1-(3)4GlcNAc-R (α-Gal) is produced in all mammals except for humans, apes and old world monkeys that lost the ability to synthetize this carbohydrate. Therefore, humans can produce high antibody titers against α-Gal. Anti-α-Gal IgE antibodies have been associated with tick-induced allergy (i.e. α-Gal syndrome) and anti-α-Gal IgG/IgM antibodies may be involved in protection against malaria, leishmaniasis and Chagas disease. The α-Gal on tick salivary proteins plays an important role in the etiology of the α-Gal syndrome. However, whether ticks are able to produce endogenous α-Gal remains currently unknown. In this study, the Ixodes scapularis genome was searched for galactosyltransferases and three genes were identified as potentially involved in the synthesis of α-Gal. Heterologous gene expression in α-Gal-negative cells and gene knockdown in ticks confirmed that these genes were involved in α-Gal synthesis and are essential for tick feeding. Furthermore, these genes were shown to play an important role in tick-pathogen interactions. Results suggested that tick cells increased α-Gal levels in response to Anaplasma phagocytophilum infection to control bacterial infection. These results provided the molecular basis of endogenous α-Gal production in ticks and suggested that tick galactosyltransferases are involved in vector development, tick-pathogen interactions and possibly the etiology of α-Gal syndrome in humans.


Asunto(s)
Anaplasma phagocytophilum/patogenicidad , Proteínas de Artrópodos/metabolismo , Galactosiltransferasas/metabolismo , Ixodes/microbiología , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Vectores de Enfermedades , Ehrlichiosis/genética , Ehrlichiosis/metabolismo , Genoma/genética , Células HL-60 , Interacciones Huésped-Patógeno/genética , Humanos
8.
Artículo en Inglés | MEDLINE | ID: mdl-29456968

RESUMEN

Eukaryotic proteome diversity exceeds that encoded within individual genes, and results in part from alternative splicing events of pre-messenger RNA. The diversity of these splicing events can shape the outcome in development and differentiation of normal tissues, and is important in pathogenic circumstances such as cancer and some heritable conditions. A role for alternative splicing of eukaryotic genes in response to viral and intracellular bacterial infections has only recently been recognized, and plays an important role in providing fitness for microbial survival, while potentially enhancing pathogenicity. Anaplasma phagocytophilum survives within mammalian neutrophils by reshaping transcriptional programs that govern cellular functions. We applied next generation RNAseq to ATRA-differentiated HL-60 cells established to possess transcriptional and functional responses similar to A. phagocytophilum-infected human neutrophils. This demonstrated an increase in transcripts with infection and high proportion of alternatively spliced transcript events (ASEs) for which predicted gene ontology processes were in part distinct from those identified by evaluation of single transcripts or gene-level analyses alone. The alternative isoforms are not on average shorter, and no alternative splicing in genes encoding spliceosome components is noted. Although not evident at gene-level analyses, individual spliceosome transcripts that impact nearly all spliceosome components were significantly upregulated. How the distinct GO processes predicted by ASEs are regulated by infection and whether they are relevant to fitness or pathogenicity of A. phagocytophilum should be addressed in more detailed studies.


Asunto(s)
Empalme Alternativo , Anaplasma phagocytophilum/fisiología , Ehrlichiosis/genética , Ehrlichiosis/microbiología , Células Mieloides/metabolismo , Transcriptoma , Diferenciación Celular/genética , Biología Computacional/métodos , Ehrlichiosis/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Células HL-60 , Humanos , Células Mieloides/citología , Empalmosomas/metabolismo , Activación Transcripcional
9.
Infect Immun ; 86(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29358333

RESUMEN

Ehrlichia chaffeensis has a group of well-characterized type I secreted tandem repeat protein (TRP) effectors that have moonlighting capabilities. TRPs modulate various cellular processes, reprogram host gene transcription as nucleomodulins, function as ubiquitin ligases, and directly activate conserved host cell signaling pathways to promote E. chaffeensis infection. One TRP-interacting host target is polycomb group ring finger protein 5 (PCGF5), a member of the polycomb group (PcG) protein family and a component of the polycomb repressive complex 1 (PRC1). The current study demonstrates that during early infection, PCGF5 strongly colocalizes with TRP120 in the nucleus and later dramatically redistributes to the ehrlichial vacuole along with other PCGF isoforms. Ectopic expression and immunoprecipitation of TRP120 confirmed the interaction of TRP120 with multiple different PCGF isoforms. At 48 h postinfection, a dramatic redistribution of PCGF isoforms from the nucleus to the ehrlichial vacuole was observed, which also temporally coincided with proteasomal degradation of PCGF isoforms and TRP120 expression on the vacuole. A decrease in PRC1-mediated repressive chromatin mark and an altered transcriptional activity in PRC1-associated Hox genes primarily from HOXB and HOXC clusters were observed along with the degradation of PCGF isoforms, suggesting disruption of the PRC1 in E. chaffeensis-infected cells. Notably, small interfering RNA (siRNA)-mediated knockdown of PCGF isoforms resulted in significantly increased E. chaffeensis infection. This study demonstrates a novel strategy in which E. chaffeensis manipulates PRC complexes through interactions between TRP120 and PCGF isoforms to promote infection.


Asunto(s)
Ehrlichia chaffeensis/fisiología , Ehrlichiosis/metabolismo , Ehrlichiosis/microbiología , Interacciones Huésped-Patógeno , Proteínas del Grupo Polycomb/metabolismo , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Rastreo Celular , Ehrlichiosis/genética , Genes Homeobox , Histonas/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas del Grupo Polycomb/genética , Isoformas de Proteínas , Proteolisis
10.
Microb Pathog ; 113: 34-39, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29042303

RESUMEN

The aim of this study was the identification of proteins differentially represented in the serum proteome of seropositive dogs with (Group 1) and without (Group 2) clinical-pathologic signs consistent with ehrlichiosis compared to healthy control dogs. Serum samples were collected from 20 dogs of various breeds with naturally occurring ehrlichiosis (10 dogs belonged to Group 1 and 10 to Group 2) and 10 healthy dogs. Two-dimensional electrophoresis (2DE) of pooled serum for each of the group of dogs were run in triplicate. 2D image analysis showed 39 spots differently expressed between Group 1 and Group 2 compared with healthy ones. Mass spectrometry analysis allowed identification of 6 proteins: albumin, haptoglobin (Hp), alpha-1-antitrypsin (AAT), Retinol Binding Protein 4 (RBP-4), alpha-1-acid glycoprotein (AGP) and vitamin D-binding protein (VDBP). When a confirmatory study was performed for albumin, Hp, AAT and RBP-4 by using different assays, significant differences (P < 0.05) between diseased and healthy groups were observed. It can be concluded that there are significant changes in the serum proteome of dogs with ehrlichiosis with modifications in proteins related with the acute phase response such as Hp, albumin and AGP, with vitamin A transport such as RBP-4, with inhibitors of serine proteases and anti-inflammatory proteins such as AAT, and vitamin D metabolism and actin scavengers such as VDBP.


Asunto(s)
Proteínas Sanguíneas/análisis , Enfermedades de los Perros/microbiología , Ehrlichia canis/metabolismo , Ehrlichiosis/metabolismo , Ehrlichiosis/veterinaria , Proteoma/análisis , Albúminas/análisis , Animales , Perros , Ehrlichiosis/microbiología , Femenino , Haptoglobinas/análisis , Masculino , Orosomucoide/análisis , Proteínas Plasmáticas de Unión al Retinol/análisis , Proteína de Unión a Vitamina D/sangre , alfa 1-Antitripsina/sangre
11.
PLoS Pathog ; 13(10): e1006644, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29049365

RESUMEN

Severe hepatic inflammation is a common cause of acute liver injury following systemic infection with Ehrlichia, obligate Gram-negative intracellular bacteria that lack lipopolysaccharide (LPS). We have previously shown that type I IFN (IFN-I) and inflammasome activation are key host-pathogenic mediators that promote excessive inflammation and liver damage following fatal Ehrlichia infection. However, the underlying signals and mechanisms that regulate protective immunity and immunopathology during Ehrlichia infection are not well understood. To address this issue, we compared susceptibility to lethal Ixodes ovatus Ehrlichia (IOE) infection between wild type (WT) and MyD88-deficient (MyD88-/-) mice. We show here that MyD88-/- mice exhibited decreased inflammasome activation, attenuated liver injury, and were more resistant to lethal infection than WT mice, despite suppressed protective immunity and increased bacterial burden in the liver. MyD88-dependent inflammasome activation was also dependent on activation of the metabolic checkpoint kinase mammalian target of rapamycin complex 1 (mTORC1), inhibition of autophagic flux, and defective mitophagy in macrophages. Blocking mTORC1 signaling in infected WT mice and primary macrophages enhanced bacterial replication and attenuated inflammasome activation, suggesting autophagy promotes bacterial replication while inhibiting inflammasome activation. Finally, our data suggest TLR9 and IFN-I are upstream signaling mechanisms triggering MyD88-mediated mTORC1 and inflammasome activation in macrophages following Ehrlichia infection. This study reveals that Ehrlichia-induced liver injury and toxic shock are mediated by MyD88-dependent inflammasome activation and autophagy inhibition.


Asunto(s)
Ehrlichiosis/inmunología , Inflamasomas/metabolismo , Fallo Hepático Agudo/microbiología , Factor 88 de Diferenciación Mieloide/metabolismo , Choque Séptico/metabolismo , Animales , Autofagia/inmunología , Western Blotting , Modelos Animales de Enfermedad , Ehrlichia/inmunología , Ehrlichiosis/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Procesamiento de Imagen Asistido por Computador , Etiquetado Corte-Fin in Situ , Inflamasomas/inmunología , Fallo Hepático Agudo/inmunología , Fallo Hepático Agudo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Factor 88 de Diferenciación Mieloide/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Choque Séptico/inmunología
12.
PLoS One ; 12(8): e0182898, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28797056

RESUMEN

Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis infects neutrophils and other cells from hematopoietic origin. Using human megakaryocytic cell line, MEG-01, we show that expression of cell cycle genes in these cells are altered upon A. phagocytophilum infection. Expression of several cell cycle genes in MEG-01 cells was significantly up regulated at early and then down regulated at later stages of A. phagocytophilum infection. Lactate dehydrogenase (LDH) assays revealed reduced cellular cytotoxicity in MEG-01 cells upon A. phagocytophilum infection. The levels of both PI3KCA (p110 alpha, catalytic subunit) and PI3KR1 (p85, regulatory subunit) of Class I PI3 kinases and phosphorylated protein kinase B (Akt/PKB) and inhibitory kappa B (IκB) were elevated at both early and late stages of A. phagocytophilum infection. Inhibition of PI3 kinases with LY294002 treatment resulted in significant reduction in the expression of tested cell cycle genes, A. phagocytophilum burden and phosphorylated Akt levels in these MEG-01 cells. Collectively, these results suggest a role for PI3K-Akt-NF-κB signaling pathway in the modulation of megakaryocyte cell cycle genes upon A. phagocytophilum infection.


Asunto(s)
Ciclo Celular/genética , Ehrlichiosis/metabolismo , Megacariocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Anaplasma phagocytophilum , Línea Celular , Humanos , Megacariocitos/microbiología , Fosforilación
13.
Artículo en Inglés | MEDLINE | ID: mdl-29376035

RESUMEN

Ehrlichia chaffeensis is an obligately intracellular bacterium that reprograms the mononuclear phagocyte through diverse effector-host interactions to modulate various host cell processes. In a previous study, we reported that the E. chaffeensis nucleomodulin TRP32 regulates transcription of host genes in several biologically relevant categories, including cell differentiation and proliferation. In this study, we investigate the effect of ubiquitination on TRP32 function and localization within the host cell. TRP32 is both mono- and polyubiquitinated on multiple lysine residues during infection and when ectopically expressed. Despite lacking a canonical PPxY motif, TRP32 interacted with, and was modified by the human HECT E3 ubiquitin (Ub) ligase NEDD4L. TRP32 ubiquitination was not by K48-linked polyUb chains, nor was it degraded by the proteasome; however, TRP32 was modified by K63-linked polyUb chains detected both in the cytosol and nucleus. HECT ligase inhibitor, heclin, altered the subnuclear localization of ectopically expressed TRP32 from a diffuse nuclear pattern to a lacy, punctate pattern with TRP32 distributed around the periphery of the nucleus and nucleoli. When a TRP32 lysine null (K-null) mutant was ectopically expressed, it exhibited a similar phenotype as single lysine mutants (K63R, K93R, and K123R). However, the K-null mutant showed increased amounts of cytoplasmic TRP32 compared to single lysine mutants or heclin-treated cells ectopically expressing TRP32. These alterations in localization corresponded to changes in TRP32 transcriptional repressor function with heclin-treated and single lysine mutants unable to repress transcription of a TRP32 target genes in a luciferase assay.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ehrlichia chaffeensis/fisiología , Ehrlichiosis/metabolismo , Ehrlichiosis/microbiología , Interacciones Huésped-Patógeno , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Línea Celular , Expresión Génica , Genes Reporteros , Humanos , Espacio Intracelular , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes , Factores de Transcripción/metabolismo , Ubiquitinación
14.
Mol Cell Proteomics ; 14(12): 3154-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26424601

RESUMEN

Anaplasma phagocytophilum is an emerging zoonotic pathogen that causes human granulocytic anaplasmosis. These intracellular bacteria establish infection by affecting cell function in both the vertebrate host and the tick vector, Ixodes scapularis. Previous studies have characterized the tick transcriptome and proteome in response to A. phagocytophilum infection. However, in the postgenomic era, the integration of omics datasets through a systems biology approach allows network-based analyses to describe the complexity and functionality of biological systems such as host-pathogen interactions and the discovery of new targets for prevention and control of infectious diseases. This study reports the first systems biology integration of metabolomics, transcriptomics, and proteomics data to characterize essential metabolic pathways involved in the tick response to A. phagocytophilum infection. The ISE6 tick cells used in this study constitute a model for hemocytes involved in pathogen infection and immune response. The results showed that infection affected protein processing in endoplasmic reticulum and glucose metabolic pathways in tick cells. These results supported tick-Anaplasma co-evolution by providing new evidence of how tick cells limit pathogen infection, while the pathogen benefits from the tick cell response to establish infection. Additionally, ticks benefit from A. phagocytophilum infection by increasing survival while pathogens guarantee transmission. The results suggested that A. phagocytophilum induces protein misfolding to limit the tick cell response and facilitate infection but requires protein degradation to prevent ER stress and cell apoptosis to survive in infected cells. Additionally, A. phagocytophilum may benefit from the tick cell's ability to limit bacterial infection through PEPCK inhibition leading to decreased glucose metabolism, which also results in the inhibition of cell apoptosis that increases infection of tick cells. These results support the use of this experimental approach to systematically identify cell pathways and molecular mechanisms involved in tick-pathogen interactions. Data are available via ProteomeXchange with identifier PXD002181.


Asunto(s)
Anaplasma phagocytophilum/fisiología , Ehrlichiosis/veterinaria , Metabolómica/métodos , Proteómica/métodos , Garrapatas/microbiología , Animales , Línea Celular , Ehrlichiosis/genética , Ehrlichiosis/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Glucosa/metabolismo , Interacciones Huésped-Patógeno , Redes y Vías Metabólicas , Biología de Sistemas/métodos
15.
Res Vet Sci ; 100: 18-20, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25956636

RESUMEN

Adrenocortical disturbances are expected in canine ehrlichiosis due to the immunological challenges caused by infection and consequent inflammation. Thus, this study aimed to evaluate the occurrence of adrenocortical hormonal alterations in dogs naturally infected with Ehrlichia canis (n = 21) as positively confirmed by the presence of anti-E. canis antibodies (Dot-ELISA) and nested PCR (nPCR). Serum dehydroepiandrosterone sulfate (DHEA-S) concentrations were assessed via ELISA before and one hour after ACTH stimulation. Another 10 healthy dogs were subjected to the same stimulation protocol and used as controls. The results revealed that baseline and post-ACTH DHEA-S concentrations were significantly greater in sick dogs, regardless of gender, and this finding illustrates the stress induced by naturally acquired ehrlichiosis in dogs.


Asunto(s)
Sulfato de Deshidroepiandrosterona/sangre , Enfermedades de los Perros/microbiología , Ehrlichia canis/fisiología , Ehrlichiosis/veterinaria , Hormona Adrenocorticotrópica/administración & dosificación , Animales , Enfermedades de los Perros/metabolismo , Perros , Ehrlichiosis/metabolismo , Ehrlichiosis/microbiología , Ensayo de Inmunoadsorción Enzimática/veterinaria , Femenino , Masculino , Reacción en Cadena de la Polimerasa/veterinaria
16.
PLoS Pathog ; 11(2): e1004669, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658707

RESUMEN

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.


Asunto(s)
Anaplasma phagocytophilum , Proteínas de la Membrana Bacteriana Externa , Ehrlichiosis , Simulación del Acoplamiento Molecular , Sustitución de Aminoácidos , Anaplasma phagocytophilum/química , Anaplasma phagocytophilum/genética , Anaplasma phagocytophilum/metabolismo , Anaplasma phagocytophilum/patogenicidad , Animales , Anticuerpos Antibacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Células CHO , Cricetinae , Cricetulus , Ehrlichiosis/genética , Ehrlichiosis/metabolismo , Células HL-60 , Humanos , Mutación Missense , Unión Proteica , Estructura Terciaria de Proteína
17.
Infect Immun ; 82(6): 2553-64, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24686067

RESUMEN

Saliva from arthropod vectors facilitates blood feeding by altering host inflammation. Whether arthropod saliva counters inflammasome signaling, a protein scaffold that regulates the activity of caspase-1 and cleavage of interleukin-1ß (IL-1ß) and IL-18 into mature molecules, remains elusive. In this study, we provide evidence that a tick salivary protein, sialostatin L2, inhibits inflammasome formation during pathogen infection. We show that sialostatin L2 targets caspase-1 activity during host stimulation with the rickettsial agent Anaplasma phagocytophilum. A. phagocytophilum causes macrophage activation and hemophagocytic syndrome features. The effect of sialostatin L2 in macrophages was not due to direct caspase-1 enzymatic inhibition, and it did not rely on nuclear factor κB or cathepsin L signaling. Reactive oxygen species from NADPH oxidase and the Loop2 domain of sialostatin L2 were important for the regulatory process. Altogether, our data expand the knowledge of immunoregulatory pathways of tick salivary proteins and unveil an important finding in inflammasome biology.


Asunto(s)
Anaplasma phagocytophilum/fisiología , Caspasa 1/metabolismo , Ehrlichiosis/microbiología , Cistatinas Salivales/fisiología , Análisis de Varianza , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ehrlichiosis/metabolismo , Ehrlichiosis/patología , Inflamasomas/metabolismo , Inflamación/fisiopatología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno
18.
J Comp Pathol ; 150(4): 351-6, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24602324

RESUMEN

Anaplasma phagocytophilum, the causative agent of tick-borne fever (TBF) in sheep and cattle and human granulocytic anaplasmosis, has the unique ability to selectively infect and multiply within the hostile environment of the neutrophil. Previous studies have shown that sheep with TBF are more susceptible to other infections and that infected neutrophils have reduced phagocytic ability and delayed apoptosis. This suggests that survival of A. phagocytophilum in these short-lived cells involves the ability to subvert or resist their bacterial killing, but also to modify the host cells such that the host cells survive long after infection. The present study shows that infection of sheep by A. phagocytophilum is characterized by up-regulation of some anti-apoptotic genes (BCL2, BIRC3 and CFLAR) in neutrophils and up-regulation of genes encoding the pro-inflammatory cytokines interferon-γ, interleukin (IL)-1ß and IL-6 in mononuclear cells during the period of bacteraemia. Infection with A. phagocytophilum was also characterized by significant up-regulation of CYBB, which is associated with the respiratory burst of neutrophils.


Asunto(s)
Anaplasma phagocytophilum , Ehrlichiosis/veterinaria , Leucocitos Mononucleares/metabolismo , Neutrófilos/metabolismo , Enfermedades de las Ovejas/genética , Regulación hacia Arriba/genética , Animales , Apoptosis/genética , Ehrlichiosis/genética , Ehrlichiosis/metabolismo , Ehrlichiosis/patología , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Leucocitos Mononucleares/patología , Neutrófilos/patología , Ovinos , Enfermedades de las Ovejas/metabolismo , Enfermedades de las Ovejas/patología
19.
PLoS Pathog ; 9(10): e1003666, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098122

RESUMEN

Ehrlichia chaffeensis, an obligatory intracellular rickettsial pathogen, enters and replicates in monocytes/macrophages and several non-phagocytic cells. E. chaffeensis entry into mammalian cells is essential not only for causing the emerging zoonosis, human monocytic ehrlichiosis, but also for its survival. It remains unclear if E. chaffeensis has evolved a specific surface protein that functions as an 'invasin' to mediate its entry. We report a novel entry triggering protein of Ehrlichia, EtpE that functions as an invasin. EtpE is an outer membrane protein and an antibody against EtpE (the C-terminal fragment, EtpE-C) greatly inhibited E. chaffeensis binding, entry and infection of both phagocytes and non-phagocytes. EtpE-C-immunization of mice significantly inhibited E. chaffeensis infection. EtpE-C-coated latex beads, used to investigate whether EtpE-C can mediate cell invasion, entered both phagocytes and non-phagocytes and the entry was blocked by compounds that block E. chaffeensis entry. None of these compounds blocked uptake of non-coated beads by phagocytes. Yeast two-hybrid screening revealed that DNase X, a glycosylphosphatidyl inositol-anchored mammalian cell-surface protein binds EtpE-C. This was confirmed by far-Western blotting, affinity pull-down, co-immunoprecipitation, immunofluorescence labeling, and live-cell image analysis. EtpE-C-coated beads entered bone marrow-derived macrophages (BMDMs) from wild-type mice, whereas they neither bound nor entered BMDMs from DNase X(-/-) mice. Antibody against DNase X or DNase X knock-down by small interfering RNA impaired E. chaffeensis binding, entry, and infection. E. chaffeensis entry and infection rates of BMDMs from DNase X(-/-) mice and bacterial load in the peripheral blood in experimentally infected DNase X(-/-) mice, were significantly lower than those from wild-type mice. Thus this obligatory intracellular pathogen evolved a unique protein EtpE that binds DNase X to enter and infect eukaryotic cells. This study is the first to demonstrate the invasin and its mammalian receptor, and their in vivo relevance in any ehrlichial species.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Desoxirribonucleasas/metabolismo , Ehrlichiosis/metabolismo , Proteínas Ligadas a GPI/metabolismo , Fagocitos/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Desoxirribonucleasas/genética , Perros , Ehrlichia chaffeensis , Ehrlichiosis/genética , Ehrlichiosis/patología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Fagocitos/microbiología , Fagocitos/patología , Unión Proteica
20.
Microbes Infect ; 15(14-15): 1005-16, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24141087

RESUMEN

Ehrlichia chaffeensis is an obligately intracellular gram negative bacterium with a small genome that thrives in mammalian mononuclear phagocytes by exploiting eukaryotic processes. Herein, we discuss the latest findings on moonlighting tandem repeat protein effectors and their secretion mechanisms, and novel molecular interkingdom interactions that provide insight into the intracellular pathobiology of ehrlichiae.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ehrlichia/fisiología , Ehrlichiosis/metabolismo , Ehrlichiosis/microbiología , Interacciones Huésped-Patógeno , Fagocitos/metabolismo , Fagocitos/microbiología , Animales , Ancirinas/metabolismo , Sistemas de Secreción Bacterianos , Núcleo Celular/metabolismo , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Interacciones Huésped-Patógeno/inmunología , Humanos , Espacio Intracelular/inmunología , Espacio Intracelular/microbiología , Unión Proteica , Transporte de Proteínas , Canales de Potencial de Receptor Transitorio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...