Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 215
Filtrar
1.
mBio ; 12(6): e0239721, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34903051

RESUMO

Chlamydia trachomatis is an obligate intracellular bacterium that has developed sophisticated mechanisms to survive inside its infectious compartment, the inclusion. Notably, Chlamydia weaves an extensive network of microtubules (MTs) and actin filaments to enable interactions with host organelles and enhance its stability. Despite the global health and economic burden caused by this sexually transmitted pathogen, little is known about how actin and MT scaffolds are integrated into an increasingly complex virulence system. Previously, we established that the chlamydial effector InaC interacts with ARF1 to stabilize MTs. We now demonstrate that InaC regulates RhoA to control actin scaffolds. InaC relies on cross talk between ARF1 and RhoA to coordinate MTs and actin, where the presence of RhoA downregulates stable MT scaffolds and ARF1 activation inhibits actin scaffolds. Understanding how Chlamydia hijacks complex networks will help elucidate how this clinically significant pathogen parasitizes its host and reveal novel cellular signaling pathways. IMPORTANCE Chlamydia trachomatis is a major cause of human disease worldwide. The ability of Chlamydia to establish infection and cause disease depends on the maintenance of its parasitic niche, called the inclusion. To accomplish this feat, Chlamydia reorganizes host actin and microtubules around the inclusion membrane. How Chlamydia orchestrates these complex processes, however, is largely unknown. Here, we discovered that the chlamydial effector InaC activates Ras homolog family member A (RhoA) to control the formation of actin scaffolds around the inclusion, an event that is critical for inclusion stability. Furthermore, InaC directs the kinetics of actin and posttranslationally modified microtubule scaffolds by mediating cross talk between the GTPases that control these cytoskeletal elements, RhoA and ADP-ribosylation factor 1 (ARF1). The precise timing of these events is essential for the maintenance of the inclusion. Overall, this study provides the first evidence of ARF1-RhoA-mediated cross talk by a bacterial pathogen to coopt the host cytoskeleton.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/fisiologia , Citoesqueleto/microbiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Fator 1 de Ribosilação do ADP/genética , Actinas/genética , Actinas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/genética , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/genética , Citoesqueleto/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Corpos de Inclusão/microbiologia , Ligação Proteica , Virulência , Proteína rhoA de Ligação ao GTP/genética
2.
mBio ; 12(5): e0229921, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34544283

RESUMO

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.


Assuntos
Anaplasma phagocytophilum/crescimento & desenvolvimento , Anaplasma phagocytophilum/metabolismo , Colesterol/metabolismo , Ehrlichiose/microbiologia , Ezetimiba/farmacologia , Proteínas de Membrana/metabolismo , Proteína C1 de Niemann-Pick/metabolismo , Anaplasma phagocytophilum/efeitos dos fármacos , Anaplasma phagocytophilum/genética , Transporte Biológico , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Ehrlichiose/genética , Ehrlichiose/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Corpos de Inclusão/microbiologia , Proteínas de Membrana/genética , Proteína C1 de Niemann-Pick/genética , Ligação Proteica , Transporte Proteico
3.
Pathog Dis ; 79(2)2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33512479

RESUMO

Chlamydia trachomatis is the leading cause of infectious blindness and a sexually transmitted infection. All chlamydiae are obligate intracellular bacteria that replicate within a membrane-bound vacuole termed the inclusion. From the confines of the inclusion, the bacteria must interact with many host organelles to acquire key nutrients necessary for replication, all while promoting host cell viability and subverting host defense mechanisms. To achieve these feats, C. trachomatis delivers an arsenal of virulence factors into the eukaryotic cell via a type 3 secretion system (T3SS) that facilitates invasion, manipulation of host vesicular trafficking, subversion of host defense mechanisms and promotes bacteria egress at the conclusion of the developmental cycle. A subset of these proteins intercalate into the inclusion and are thus referred to as inclusion membrane proteins. Whereas others, referred to as conventional T3SS effectors, are released into the host cell where they localize to various eukaryotic organelles or remain in the cytosol. Here, we discuss the functions of T3SS effector proteins with a focus on how advances in chlamydial genetics have facilitated the identification and molecular characterization of these important factors.


Assuntos
Proteínas de Bactérias/fisiologia , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/fisiologia , Chlamydia trachomatis/patogenicidade , Interações Hospedeiro-Patógeno , Corpos de Inclusão/metabolismo , Sistemas de Secreção Tipo III/fisiologia , Células HeLa , Humanos , Corpos de Inclusão/microbiologia , Transporte Proteico , Vacúolos/metabolismo , Vacúolos/microbiologia , Fatores de Virulência
4.
Infect Immun ; 88(12)2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-32900818

RESUMO

The obligate intracellular pathogen Chlamydia trachomatis is the leading cause of noncongenital blindness and causative agent of the most common sexually transmitted infection of bacterial origin. With a reduced genome, C. trachomatis is dependent on its host for survival, in part due to a need for the host cell to compensate for incomplete bacterial metabolic pathways. However, relatively little is known regarding how C. trachomatis is able to hijack host cell metabolism. In this study, we show that two host glycolytic enzymes, aldolase A and pyruvate kinase, as well as lactate dehydrogenase, are enriched at the C. trachomatis inclusion membrane during infection. Inclusion localization was not species specific, since a similar phenotype was observed with C. muridarum Time course experiments showed that the number of positive inclusions increased throughout the developmental cycle. In addition, these host enzymes colocalized to the same inclusion, and their localization did not appear to be dependent on sustained bacterial protein synthesis or on intact host actin, vesicular trafficking, or microtubules. Depletion of the host glycolytic enzyme aldolase A resulted in decreased inclusion size and infectious progeny production, indicating a role for host glycolysis in bacterial growth. Finally, quantitative PCR analysis showed that expression of C. trachomatis glycolytic enzymes inversely correlated with host enzyme localization at the inclusion. We discuss potential mechanisms leading to inclusion localization of host glycolytic enzymes and how it could benefit the bacteria. Altogether, our findings provide further insight into the intricate relationship between host and bacterial metabolism during Chlamydia infection.


Assuntos
Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , Frutose-Bifosfato Aldolase/metabolismo , Glicólise , Interações entre Hospedeiro e Microrganismos , Corpos de Inclusão/metabolismo , L-Lactato Desidrogenase/metabolismo , Piruvato Quinase/metabolismo , Actinas/metabolismo , Membrana Externa Bacteriana/enzimologia , Membrana Externa Bacteriana/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/enzimologia , Infecções por Chlamydia/genética , Chlamydia muridarum/metabolismo , Chlamydia trachomatis/enzimologia , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/patogenicidade , Frutose-Bifosfato Aldolase/genética , Células HeLa , Humanos , Corpos de Inclusão/enzimologia , Corpos de Inclusão/microbiologia , L-Lactato Desidrogenase/genética , Microtúbulos/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Piruvato Quinase/genética
5.
Infect Immun ; 88(4)2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-31964750

RESUMO

Human genital Chlamydia infection is a major public health concern due to the serious reproductive system complications. Chlamydia binds several receptor tyrosine kinases (RTKs) on host cells, including the epidermal growth factor receptor (EGFR), and activates cellular signaling cascades for host invasion, cytoskeletal remodeling, optimal inclusion development, and induction of pathogenic epithelial-mesenchyme transition (EMT). Chlamydia also upregulates transforming growth factor beta (TGF-ß) expression, whose signaling pathway synergizes with the EGFR cascade, but its role in infectivity, inclusions, and EMT induction is unknown. We hypothesized that the EGFR and TGF-ß signaling pathways cooperate during chlamydial infection for optimal inclusion development and stable EMT induction. The results revealed that Chlamydia upregulated TGF-ß expression as early as 6 h postinfection of epithelial cells and stimulated both the EGFR and TGF-ß signaling pathways. Inhibition of either the EGFR or TGF-ßR1 signaling substantially reduced inclusion development; however, the combined inhibition of both EGFR and TGF-ßR1 signaling reduced inclusions by over 90% and prevented EMT induction. Importantly, EGFR inhibition suppressed TGF-ß expression, and an inhibitory thrombospondin-1 (Tsp1)-based peptide inhibited chlamydia-induced EMT, revealing a major source of active TGF-ß during infection. Finally, TGF-ßR signaling inhibition suppressed the expression of transforming acidic coiled-coil protein-3 (TACC3), which stabilizes EGFR signaling, suggesting reciprocal regulation between TGF-ß and EGFR signaling during chlamydial infection. Thus, RTK-mediated host invasion by chlamydia upregulated TGF-ß expression and signaling, which cooperated with other cellular signaling cascades and cytoskeletal remodeling to support optimal inclusion development and EMT induction. This finding may provide new targets for chlamydial disease biomarkers and prevention.


Assuntos
Infecções por Chlamydia/fisiopatologia , Chlamydia/crescimento & desenvolvimento , Células Epiteliais/microbiologia , Receptores ErbB/metabolismo , Interações Hospedeiro-Patógeno , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Endocitose , Transição Epitelial-Mesenquimal , Corpos de Inclusão/microbiologia , Camundongos , Modelos Biológicos
6.
Histol Histopathol ; 35(2): 177-184, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31298302

RESUMO

CONTEXT: Malakoplakia can be caused by incomplete digestion of Escherichia coli by lysosomes, leading to recurrent urinary tract infections and consequential mass-forming events that mimic tumors. OBJECTIVES: By using ultrastructural findings, we aimed to specify the process of phagolysosome to evoke malakoplakia. DESIGN: We observed a series of processes to form a peculiar Michaelis-Gutmann (MG) body in three patients with malakoplakia and compared with xanthogranulomatous pyelonephritis. RESULTS: The ultrastructural findings were realigned according to the sequence of events as pre-phagosomal, phagosomal, and post-phagosomal stages. For the mature MG body, numerous lysosomal aggregates targeting pathogens and subsequent incomplete digestion are prerequisite factors for the pre-phagosomal stage. Scattered lamellated residue is late evidence of the pre-phagosomal stage. Phagosomes can be formed by the fusion of multiple pathogens and multiple lysosomes. We utilized transmission and scanning electron microscopy to speculate on the process of phagolysosomal formation. CONCLUSION: The recognition of E. coli captured by phagosomes or partially damaged by lysosomal attack within the cell was recorded for the first time. Furthermore, SEM observation was performed on human tissue.


Assuntos
Infecções por Escherichia coli/patologia , Corpos de Inclusão/ultraestrutura , Malacoplasia/microbiologia , Malacoplasia/patologia , Idoso , Escherichia coli , Feminino , Humanos , Corpos de Inclusão/microbiologia , Corpos de Inclusão/patologia , Lisossomos/ultraestrutura , Masculino , Microscopia Eletrônica , Próstata/patologia , Próstata/ultraestrutura , Bexiga Urinária/patologia , Bexiga Urinária/ultraestrutura
7.
J Proteomics ; 212: 103595, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-31760040

RESUMO

The obligate intracellular bacterial pathogen, Chlamydia trachomatis, develops within a membrane-bound vacuole termed the inclusion. Affinity purification-mass spectrometry (AP-MS) experiments to study the interactions that occur at the chlamydial inclusion membrane have been performed and, more recently, combined with advances in C. trachomatis genetics. However, each of the four AP-MS published reports used either different experimental approaches or statistical tools to identify proteins that localize at the inclusion. We critically analyzed each experimental approach and performed a meta-analysis of the reported statistically significant proteins for each study, finding that only a few eukaryotic proteins were commonly identified between all four experimental approaches. The two similarly conducted in vivo labeling studies were compared using the same statistical analysis tool, Significance Analysis of INTeractome (SAINT), which revealed a disparity in the number of significant proteins identified by the original analysis. We further examined methods to identify potential background contaminant proteins that remain after statistical analysis. Overall, this meta-analysis highlights the importance of carefully controlling and analyzing the AP-MS data so that pertinent information can be obtained from these various AP-MS experimental approaches. This study provides important guidelines and considerations for using this methodology to study intracellular pathogens residing within a membrane-bound compartment. SIGNIFICANCE: Chlamydia trachomatis, an obligate intracellular pathogen, grows within a membrane-bound vacuole termed the inclusion. The inclusion is studded with bacterial membrane proteins that likely orchestrate numerous interactions with the host cell. Although maintenance of the intracellular niche is vital, an understanding of the host-pathogen interactions that occur at the inclusion membrane is limited by the difficulty in purifying membrane protein fractions from infected host cells. The experimental procedures necessary to solubilize hydrophobic proteins fail to maintain transient protein-protein interactions. Advances in C. trachomatis genetics has allowed us and others to use various experimental approaches in combination with affinity purification mass spectrometry (AP-MS) to study the interactions that occur at the chlamydial vacuolar, or inclusion, membrane. For the first time, two groups have published AP-MS studies using the same tool, the ascorbate peroxidase proximity labeling system (APEX2), which overcomes past experimental limitations because membrane protein interactions are labeled in vivo in the context of infection. The utility of this system is highlighted by its ability to study chlamydial type III secreted inclusion membrane protein (Inc) interactions. Incs act as the mediators of host-pathogen interactions at the inclusion during C. trachomatis infection. When carefully controlled and analyzed, the data obtained can yield copious amounts of useful information. Here, we critically analyzed four previously published studies, including statistical analysis of AP-MS datasets related to Chlamydia-host interactions, to contextualize the data and to identify the best practices in interpreting these types of complex outputs.


Assuntos
Proteínas de Bactérias/análise , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , Eucariotos/metabolismo , Corpos de Inclusão/metabolismo , Proteínas de Membrana/análise , Proteômica/métodos , Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/isolamento & purificação , Cromatografia de Afinidade/métodos , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/microbiologia , Espectrometria de Massas/métodos , Proteínas de Membrana/metabolismo , Vacúolos/química , Vacúolos/metabolismo , Vacúolos/microbiologia
8.
Methods Mol Biol ; 2042: 205-218, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31385278

RESUMO

Chlamydia trachomatis resides and replicates within a membranous vacuole, termed the inclusion. A group of Type III secreted effector proteins, the inclusion membrane proteins (Inc), are embedded within the inclusion membrane and facilitate the interaction of the inclusion with host cell organelles. These interactions are vital for bacterial replication and allow for the acquisition of essential nutrients from the host cell. However, it is not known if Inc proteins function independently or require interactions with other Inc proteins to function. This chapter describes a system to test the homotypic/heterotypic interactions of Inc proteins through the coinfection of Chlamydia strains expressing differently tagged inclusion membrane proteins. Our approach takes advantage of the natural homotypic fusion of inclusions and allows for the study of Inc protein interactions when they are embedded within the inclusion membrane.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/fisiologia , Mapeamento de Interação de Proteínas/métodos , Sistemas de Secreção Tipo III/metabolismo , Infecções por Chlamydia/microbiologia , Coinfecção/metabolismo , Coinfecção/microbiologia , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/microbiologia , Mapas de Interação de Proteínas , Vacúolos/metabolismo , Vacúolos/microbiologia
9.
Methods Mol Biol ; 2042: 245-278, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31385281

RESUMO

In the study of intracellular bacteria that reside within a membrane-bound vacuole, there are many questions related to how prokaryotic or eukaryotic transmembrane or membrane-associated proteins are organized and function within the membranes of these pathogen-containing vacuoles. Yet this host-pathogen interaction interface has proven difficult to experimentally resolve. For example, one method to begin to understand protein function is to determine the protein-binding partners; however, examining protein-protein interactions of hydrophobic transmembrane proteins is not widely successful using standard immunoprecipitation or coimmunoprecipitation techniques. In these scenarios, the lysis conditions that maintain protein-protein interactions are not compatible with solubilizing hydrophobic membrane proteins. In this chapter, we outline two proximity labeling systems to circumvent these issues to study (1) eukaryotic proteins that localize to the membrane-bound inclusion formed by Chlamydia trachomatis using BioID, and (2) chlamydial proteins that are inserted into the inclusion membrane using APEX2. BioID is a promiscuous biotin ligase to tag proximal proteins with biotin. APEX2 is an ascorbate peroxidase that creates biotin-phenoxyl radicals to label proximal proteins with biotin or 3,3'-diaminobenzidine intermediates for examination of APEX2 labeling of subcellular structures using transmission electron microscopy. We present how these methods were originally conceptualized and developed, so that the user can understand the strengths and limitations of each proximity labeling system. We discuss important considerations regarding experimental design, which include careful consideration of background conditions and statistical analysis of mass spectrometry results. When applied in the appropriate context with adequate controls, these methods can be powerful tools toward understanding membrane interfaces between intracellular pathogens and their hosts.


Assuntos
Infecções por Chlamydia/patologia , Chlamydia trachomatis/fisiologia , Interações Hospedeiro-Patógeno , Corpos de Inclusão/microbiologia , Ascorbato Peroxidases/análise , Proteínas de Bactérias/análise , Biotinilação , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/isolamento & purificação , Células HeLa , Humanos , Corpos de Inclusão/patologia , Coloração e Rotulagem/métodos
10.
Nat Commun ; 10(1): 2747, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31227715

RESUMO

Many intracellular bacteria, including Chlamydia, establish a parasitic membrane-bound organelle inside the host cell that is essential for the bacteria's survival. Chlamydia trachomatis forms inclusions that are decorated with poorly characterized membrane proteins known as Incs. The prototypical Inc, called IncA, enhances Chlamydia pathogenicity by promoting the homotypic fusion of inclusions and shares structural and functional similarity to eukaryotic SNAREs. Here, we present the atomic structure of the cytoplasmic domain of IncA, which reveals a non-canonical four-helix bundle. Structure-based mutagenesis, molecular dynamics simulation, and functional cellular assays identify an intramolecular clamp that is essential for IncA-mediated homotypic membrane fusion during infection.


Assuntos
Proteínas de Bactérias/ultraestrutura , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/patogenicidade , Corpos de Inclusão/microbiologia , Fusão de Membrana , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/genética , Chlamydia trachomatis/metabolismo , Cristalografia por Raios X , Técnicas de Inativação de Genes , Células HeLa , Humanos , Simulação de Dinâmica Molecular , Mutagênese , Conformação Proteica em alfa-Hélice , Domínios Proteicos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Proteínas SNARE/química
11.
mBio ; 10(2)2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30967464

RESUMO

Interferon-regulated immune defenses protect mammals from pathogenically diverse obligate intracellular bacterial pathogens of the genus Chlamydia Interferon gamma (IFN-γ) is especially important in controlling the virulence of Chlamydia species and thus impacts the modeling of human chlamydial infection and disease in mice. How IFN-γ contributes to cell-autonomous defenses against Chlamydia species and how these pathogens evade IFN-γ-mediated immunity in their natural hosts are not well understood. We conducted a genetic screen which identified 31 IFN-γ-sensitive (Igs) mutants of the mouse model pathogen Chlamydia muridarum Genetic suppressor analysis and lateral gene transfer were used to map the phenotype of one of these mutants, Igs4, to a missense mutation in a putative chlamydial inclusion membrane protein, TC0574. We observed the lytic destruction of Igs4-occupied inclusions and accompanying host cell death in response to IFN-γ priming or various proapoptotic stimuli. However, Igs4 was insensitive to IFN-γ-regulated cell-autonomous defenses previously implicated in anti-Chlamydia trachomatis host defense in mice. Igs4 inclusion integrity was restored by caspase inhibitors, indicating that the IFN-γ-mediated destruction of Igs4 inclusions is dependent upon the function of caspases or related prodeath cysteine proteases. We further demonstrated that the Igs4 mutant is immune restricted in an IFN-γ-dependent manner in a mouse infection model, thereby implicating IFN-γ-mediated inclusion destruction and host cell death as potent in vivo host defense mechanisms to which wild-type C. muridarum is resistant. Overall, our results suggest that C. muridarum evolved resistance mechanisms to counter IFN-γ-elicited programmed cell death and the associated destruction of intravacuolar pathogens.IMPORTANCE Multiple obligatory intracellular bacteria in the genus Chlamydia are important pathogens. In humans, strains of C. trachomatis cause trachoma, chlamydia, and lymphogranuloma venereum. These diseases are all associated with extended courses of infection and reinfection that likely reflect the ability of chlamydiae to evade various aspects of host immune responses. Interferon-stimulated genes, driven in part by the cytokine interferon gamma, restrict the host range of various Chlamydia species, but how these pathogens evade interferon-stimulated genes in their definitive host is poorly understood. Various Chlamydia species can inhibit death of their host cells and may have evolved this strategy to evade prodeath signals elicited by host immune responses. We present evidence that chlamydia-induced programmed cell death resistance evolved to counter interferon- and immune-mediated killing of Chlamydia-infected cells.


Assuntos
Apoptose , Chlamydia muridarum/imunologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata , Interferon gama/metabolismo , Animais , Infecções por Chlamydia/microbiologia , Infecções por Chlamydia/patologia , Chlamydia muridarum/genética , Modelos Animais de Doenças , Testes Genéticos , Corpos de Inclusão/microbiologia , Camundongos
12.
PLoS Pathog ; 15(4): e1007698, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30943267

RESUMO

Chlamydia trachomatis is the most common cause of bacterial sexually transmitted infection, responsible for millions of infections each year. Despite this high prevalence, the elucidation of the molecular mechanisms of Chlamydia pathogenesis has been difficult due to limitations in genetic tools and its intracellular developmental cycle. Within a host epithelial cell, chlamydiae replicate within a vacuole called the inclusion. Many Chlamydia-host interactions are thought to be mediated by the Inc family of type III secreted proteins that are anchored in the inclusion membrane, but their array of host targets are largely unknown. To investigate how the inclusion membrane proteome changes over the course of an infected cell, we have adapted the APEX2 system of proximity-dependent biotinylation. APEX2 is capable of specifically labeling proteins within a 20 nm radius in living cells. We transformed C. trachomatis to express the enzyme APEX2 fused to known inclusion membrane proteins, allowing biotinylation and purification of inclusion-associated proteins. Using quantitative mass spectrometry against APEX2 labeled samples, we identified over 400 proteins associated with the inclusion membrane at early, middle, and late stages of epithelial cell infection. This system was sensitive enough to detect inclusion interacting proteins early in the developmental cycle, at 8 hours post infection, a previously intractable time point. Mass spectrometry analysis revealed a novel, early association between C. trachomatis inclusions and endoplasmic reticulum exit sites (ERES), functional regions of the ER where COPII-coated vesicles originate. Pharmacological and genetic disruption of ERES function severely restricted early chlamydial growth and the development of infectious progeny. APEX2 is therefore a powerful in situ approach for identifying critical protein interactions on the membranes of pathogen-containing vacuoles. Furthermore, the data derived from proteomic mapping of Chlamydia inclusions has illuminated an important functional role for ERES in promoting chlamydial developmental growth.


Assuntos
Proteínas de Bactérias/análise , Infecções por Chlamydia/metabolismo , Retículo Endoplasmático/metabolismo , Corpos de Inclusão/metabolismo , Marcação por Isótopo/métodos , Proteínas de Membrana/análise , Proteoma/análise , Chlamydia/isolamento & purificação , Infecções por Chlamydia/microbiologia , Retículo Endoplasmático/microbiologia , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/microbiologia
13.
Cell Rep ; 26(12): 3380-3390.e5, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893609

RESUMO

Chlamydial infection requires the formation of a membrane-bound vacuole, termed the inclusion, that undergoes extensive interactions with select host organelles. The importance of the Inc protein CT229 in the formation and maintenance of the chlamydial inclusion was recently highlighted by studies demonstrating that its absence during infection results in reduced bacterial replication, premature inclusion lysis, and host cell death. Previous reports have indicated that CT229 binds Rab GTPases; however, the physiological implications of this interaction are unknown. Here, we show that CT229 regulates host multivesicular trafficking by recruiting multiple Rab GTPases and their cognate effectors to the inclusion. We demonstrate that CT229 specifically modulates clathrin-coated vesicle trafficking and regulates the trafficking of transferrin and the mannose-6-phosphate receptor, both of which are crucial for proper chlamydial development. This study highlights CT229 as a master regulator of multiple host vesicular trafficking pathways essential for chlamydial infection.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Vacúolos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas de Bactérias/genética , Transporte Biológico Ativo , Infecções por Chlamydia/genética , Infecções por Chlamydia/patologia , Chlamydia trachomatis/genética , Vesículas Revestidas por Clatrina/genética , Vesículas Revestidas por Clatrina/microbiologia , Células HeLa , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Corpos de Inclusão/microbiologia , Vacúolos/genética , Vacúolos/microbiologia , Proteínas rab de Ligação ao GTP/genética
14.
Microbiol Spectr ; 7(2)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30848236

RESUMO

Intravacuolar development has been adopted by several bacteria that grow inside a host cell. Remaining in a vacuole, as opposed to breaching the cytosol, protects the bacteria from some aspects of the cytosolic innate host defense and allows them to build an environment perfectly adapted to their needs. However, this raises new challenges: the host resources are separated from the bacteria by a lipid bilayer that is nonpermeable to most nutrients. In addition, the area of this lipid bilayer needs to expand to accommodate bacterial multiplication. This requires building material and energy that are not directly invested in bacterial growth. This article describes the strategies acquired by the obligate intracellular pathogen Chlamydia trachomatis to circumvent the difficulties raised by an intravacuolar lifestyle. We start with an overview of the origin and composition of the vacuolar membrane. Acquisition of host resources is largely, although not exclusively, mediated by interactions with membranous compartments of the eukaryotic cell, and we describe how the inclusion modifies the architecture of the cell and distribution of the neighboring compartments. The second part of this review describes the four mechanisms characterized so far by which the bacteria acquire resources from the host: (i) transport/diffusion across the vacuole membrane, (ii) fusion of this membrane with host compartments, (iii) direct transfer of lipids at membrane contact sites, and (iv) engulfment by the vacuole membrane of large cytoplasmic entities.


Assuntos
Chlamydia trachomatis/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno/fisiologia , Estilo de Vida , Vacúolos/microbiologia , Proteínas de Bactérias/metabolismo , Transporte Biológico , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/metabolismo , Chlamydia trachomatis/patogenicidade , Citosol/microbiologia , Células Eucarióticas , Interações Hospedeiro-Patógeno/imunologia , Humanos , Corpos de Inclusão/microbiologia , Bicamadas Lipídicas , Metabolismo dos Lipídeos
15.
Cell Microbiol ; 20(10): e12940, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30101516

RESUMO

Chlamydia belong to the group of obligate intracellular bacteria that reside in a membrane bound vacuole during the entire intracellular phase of their life cycle. This vacuole called inclusion shields the bacteria from adverse influences in the cytosol of the host cell like the destructive machinery of the cell-autonomous defence system. The inclusion thereby prevents the digestion and eradication in specialised compartments of the intact and viable cell called phagolysosomes or autophagolysosomes. It is becoming more and more evident that keeping the inclusion intact also prevents the onset of cell intrinsic cell death programmes that are activated upon damage of the inclusion and direct the cell to destruct itself and the pathogen inside. Chlamydia secrete numerous proteins into the inclusion membrane to protect and stabilise their unique niche inside the host cell. We will focus in this review on the diverse attack strategies of the host aiming at the destruction of the Chlamydia-containing inclusion and will summarise the current knowledge on the protection mechanisms elaborated by the bacteria to maintain the integrity of their replication niche.


Assuntos
Infecções por Chlamydia/imunologia , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/imunologia , Interações Hospedeiro-Patógeno/imunologia , Corpos de Inclusão/imunologia , Autofagossomos/imunologia , Infecções por Chlamydia/microbiologia , Infecções por Chlamydia/patologia , Chlamydia trachomatis/patogenicidade , Humanos , Corpos de Inclusão/microbiologia , Interferons/imunologia , Estágios do Ciclo de Vida/fisiologia , Vacúolos/microbiologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-30094225

RESUMO

Chlamydia trachomatis is an obligate intracellular human pathogen causing mainly ocular and genital infections of significant clinical and public health impact. C. trachomatis multiplies intracellularly in a membrane bound vacuole, known as inclusion. Both extracellularly and from within the inclusion, C. trachomatis uses a type III secretion system to deliver several effector proteins into the cytoplasm of host cells. A large proportion of these effectors, the inclusion membrane (Inc) proteins, are exposed to the host cell cytosol but possess a characteristic hydrophobic domain mediating their insertion in the inclusion membrane. By yeast two-hybrid, we found that C. trachomatis Inc CT288 interacts with the human centrosomal protein CCDC146 (coiled-coil domain-containing protein 146). The interaction was also detected by co-immunoprecipitation in mammalian cells either ectopically expressing CCDC146 and CT288 or ectopically expressing CCDC146 and infected by a C. trachomatis strain expressing epitope-tagged and inclusion membrane-localized CT288. In uninfected mammalian cells, ectopically expressed full-length CCDC146 (955 amino acid residues) localized at the centrosome; but in cells infected by wild-type C. trachomatis, its centrosomal localization was less evident and CCDC146 accumulated around the inclusion. Recruitment of CCDC146 to the inclusion periphery did not require intact host Golgi, microtubules or microfilaments, but was dependent on chlamydial protein synthesis. Full-length CCDC146 also accumulated at the periphery of the inclusion in cells infected by a C. trachomatis ct288 mutant; however, a C-terminal fragment of CCDC146 (residues 692-955), which interacts with CT288, showed differences in localization at the periphery of the inclusion in cells infected by wild-type or ct288 mutant C. trachomatis. This suggests a model in which chlamydial proteins other than CT288 recruit CCDC146 to the periphery of the inclusion, where the CT288-CCDC146 interaction might contribute to modulate the function of this host protein.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/fisiologia , Interações Hospedeiro-Patógeno , Corpos de Inclusão/microbiologia , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Vacúolos/microbiologia , Animais , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Imunoprecipitação , Corpos de Inclusão/química , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido , Vacúolos/química , Células Vero
17.
PLoS Pathog ; 14(3): e1006911, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29543918

RESUMO

Chlamydia trachomatis is an obligate intracellular bacterium that replicates within a vacuole termed an inclusion. At the end of their intracellular developmental cycle, chlamydiae are released either by lysis of the host cell or extrusion of the intact inclusion. The inclusion membrane is extensively modified by the insertion of type III secreted inclusion membrane proteins, Incs, which contribute to inclusion membrane structure and facilitate host-pathogen interactions. An interaction was identified between the inclusion membrane protein, MrcA, and the Ca2+ channel inositol-1,4,5-trisphosphate receptor, type 3 (ITPR3). ITPR3 was recruited and localized to active Src-family-kinase rich microdomains on the inclusion membrane as was the Ca2+ sensor, STIM1. Disruption of MrcA by directed mutagenesis resulted in loss of ITPR3 recruitment and simultaneous reduction of chlamydial release by extrusion. Complementation of MrcA restored ITPR3 recruitment and extrusion. Inhibition of extrusion was also observed following siRNA depletion of host ITPR3 or STIM1. Chlamydial extrusion was also inhibited by the calcium chelator BAPTA-AM. Each of these treatments resulted in a concomitant reduction in phosphorylation of the myosin regulatory light chain (MLC2) and a loss of myosin motor activity at the end of the developmental cycle which is consistent with the reduced extrusion formation. These studies suggest that Ca2+ signaling pathways play an important role in regulation of release mechanisms by C. trachomatis.


Assuntos
Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , Interações Hospedeiro-Patógeno , Corpos de Inclusão/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas de Membrana/metabolismo , Infecções por Chlamydia/genética , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/genética , Células HeLa , Humanos , Corpos de Inclusão/microbiologia , Receptores de Inositol 1,4,5-Trifosfato/genética , Proteínas de Membrana/genética , Fosforilação
18.
Int J Med Microbiol ; 308(1): 41-48, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28864236

RESUMO

Chlamydiales comprise important human and animal pathogens as well as endosymbionts of amoebae. Generally, these obligate intracellular living bacteria are characterized by a biphasic developmental cycle, a reduced genome and a restricted metabolic capacity. Because of their metabolic impairment, Chlamydiales essentially rely on the uptake of diverse metabolites from their hosts. Chlamydiales thrive in a special compartment, the inclusion, and hence are surrounded by an additional membrane. Solutes might enter the inclusion through pores and open channels or by redirection of host vesicles, which fuse with the inclusion membrane and release their internal cargo. Recent investigations shed new light on the chlamydia-host interaction and identified an additional way for nutrient uptake into the inclusion. Proteome studies and targeting analyses identified chlamydial and host solute carriers in inclusions of Chlamydia trachomatis infected cells. These transporters are involved in the provision of UDP-glucose and biotin, and probably deliver further metabolites to the inclusion. By the controlled recruitment of specific solute carriers to the inclusion, the chlamydial resident thus can actively manipulate the metabolite availability and composition in the inclusion. This review summarizes recent findings and new ideas on carrier mediated solute uptake into the chlamydial inclusion in the context of the bacterial and host metabolism.


Assuntos
Chlamydiales/fisiologia , Infecções por Bactérias Gram-Negativas/metabolismo , Corpos de Inclusão/metabolismo , Animais , Transporte Biológico , Proteínas de Transporte/metabolismo , Chlamydiales/crescimento & desenvolvimento , Chlamydiales/metabolismo , Infecções por Bactérias Gram-Negativas/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/microbiologia , Nutrientes/metabolismo , Vacúolos/metabolismo
19.
Int J Med Microbiol ; 308(1): 197-205, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29122514

RESUMO

Chlamydia trachomatis is an important human pathogen. This obligate intracellular bacterium grows inside the eukaryotic cell in a membrane-bound compartment, the inclusion. Recent global approaches describe the interactions of C. trachomatis with its host cell and indicate the inclusion is an intracellular trafficking hub embedded into the cellular vesicular trafficking pathways recruiting subunits of the retromer protein complex of the host cell. Here we review these recent developments in deciphering Chlamydia-host cell interactions with emphasis on the role of the retromer complex.


Assuntos
Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/metabolismo , Interações Hospedeiro-Patógeno , Animais , Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/crescimento & desenvolvimento , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/microbiologia , Transporte Proteico , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
20.
Artigo em Inglês | MEDLINE | ID: mdl-29067282

RESUMO

Chlamydia infection targets the mucosal epithelium, where squamous and columnar epithelia can be found. Research on Chlamydia-epithelia interaction has predominantly focused on columnar epithelia, with very little known on how Chlamydia interacts with the squamous epithelium. The stratification and differentiation processes found in the squamous epithelium might influence chlamydial growth and infection dissemination. For this reason, three-dimensional (3D) organotypic stratified squamous epithelial cultures were adapted to mimic the stratified squamous epithelium and chlamydial infection was characterized. Chlamydia trachomatis infection in monolayers and 3D cultures were monitored by immunofluorescence and transmission electron microscopy to evaluate inclusion growth and chlamydial interconversion between elementary and reticulate body. We observed that the stratified epithelium varied in susceptibility to C. trachomatis serovars L2 and D infection. The undifferentiated basal cells were susceptible to infection by both serovars, while the terminally differentiated upper layers were resistant. The differentiating suprabasal cells exhibited different susceptibilities to serovars L2 and D, with the latter unable to establish a successful infection in this layer. Mature elementary body-containing inclusions were much more prevalent in these permissive basal layers, while the uppermost differentiated layers consistently harbored very few reticulate bodies with no elementary bodies, indicative of severely limited bacterial replication and development. For serovar D, the differentiation state of the host cell was a determining factor, as calcium-induced differentiation of cells in a monolayer negatively affected growth of this serovar, in contrast to serovar L2. The apparent completion of the developmental cycle in the basal layers of the 3D cultures correlated with the greater degree of dissemination within and the level of disruption of the stratified epithelium. Our studies indicate that the squamous epithelium is a suboptimal environment for growth, and thus potentially contributing to the protection of the lower genital tract from infection. The relatively more fastidious serovar D exhibited more limited growth than the faster-growing and more invasive L2 strain. However, if given access to the more hospitable basal cell layer, both strains were able to produce mature inclusions, replicate, and complete their developmental cycle.


Assuntos
Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/crescimento & desenvolvimento , Células Epiteliais/microbiologia , Epitélio/microbiologia , Animais , Cálcio , Ciclo Celular , Diferenciação Celular , Técnicas de Cocultura , Células Alimentadoras/microbiologia , Células HeLa , Humanos , Corpos de Inclusão/microbiologia , Camundongos , Células NIH 3T3 , Sorogrupo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...