Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Cell Dev Biol ; 30: 79-109, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25103867

RESUMO

Pathogens use a vast number of strategies to alter host membrane dynamics. Targeting the host membrane machinery is important for the survival and pathogenesis of several extracellular, vacuolar, and cytosolic bacteria. Membrane manipulation promotes bacterial replication while suppressing host responses, allowing the bacterium to thrive in a hostile environment. This review provides a comprehensive summary of various strategies used by both extracellular and intracellular bacteria to hijack host membrane trafficking machinery. We start with mechanisms used by bacteria to alter the plasma membrane, delve into the hijacking of various vesicle trafficking pathways, and conclude by summarizing bacterial adaptation to host immune responses. Understanding bacterial manipulation of host membrane trafficking provides insights into bacterial pathogenesis and uncovers the molecular mechanisms behind various processes within a eukaryotic cell.


Assuntos
Fenômenos Fisiológicos Bacterianos , Membrana Celular/metabolismo , Células/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Animais , Autofagia/fisiologia , Proteínas de Bactérias/fisiologia , Toxinas Bacterianas/farmacologia , Transporte Biológico , Permeabilidade da Membrana Celular , Células/ultraestrutura , Citosol/microbiologia , Endocitose/fisiologia , Humanos , Lisossomos/fisiologia , Lipídeos de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Fagossomos/fisiologia , Transporte Proteico , Vacúolos/microbiologia , Vacúolos/fisiologia
2.
Proc Natl Acad Sci U S A ; 120(33): e2303942120, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37549300

RESUMO

Legionella pneumophila grows intracellularly within the membrane-bound Legionella-containing vacuole (LCV) established by proteins translocated via the bacterial type IV secretion system (T4SS). The Sde family, one such group of translocated proteins, catalyzes phosphoribosyl-ubiquitin (pR-Ub) modification of target substrates. Mutational loss of the entire Sde family results in small defects in intracellular growth, making it difficult to identify a clear role for this posttranslational modification in supporting the intracellular lifestyle. Therefore, mutations that aggravate the loss of sde genes and caused intracellular growth defects were identified, providing a mechanistic connection between Sde function and vacuole biogenesis. These double mutants drove the formation of LCVs that showed vacuole disintegration within 2 h of bacterial contact. Sde proteins appeared critical for blocking access of membrane-disruptive early endosomal membrane material to the vacuole, as RNAi depletion of endosomal pathway components partially restored LCV integrity. The role of Sde proteins in preventing host degradation of the LCV was limited to the earliest stages of infection. The time that Sde proteins could prevent vacuole disruption, however, was extended by deletion of sidJ, which encodes a translocated protein that inactivates Sde protein active sites. These results indicate that Sde proteins act as temporally regulated vacuole guards during the establishment of the replication niche, possibly by constructing a physical barrier that blocks access of disruptive host compartments during the earliest steps of LCV biogenesis.


Assuntos
Legionella pneumophila , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Vacúolos/metabolismo , Ubiquitina/metabolismo , Endossomos/metabolismo , Membranas/metabolismo , Proteínas de Bactérias/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(8): e2215237120, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36787358

RESUMO

Acinetobacter baumannii is a gram-negative bacterial pathogen that causes challenging nosocomial infections. ß-lactam targeting of penicillin-binding protein (PBP)-mediated cell wall peptidoglycan (PG) formation is a well-established antimicrobial strategy. Exposure to carbapenems or zinc (Zn)-deprived growth conditions leads to a rod-to-sphere morphological transition in A. baumannii, an effect resembling that caused by deficiency in the RodA-PBP2 PG synthesis complex required for cell wall elongation. While it is recognized that carbapenems preferentially acylate PBP2 in A. baumannii and therefore block the transpeptidase function of the RodA-PBP2 system, the molecular details underpinning cell wall elongation inhibition upon Zn starvation remain undefined. Here, we report the X-ray crystal structure of A. baumannii PBP2, revealing an unexpected Zn coordination site in the transpeptidase domain required for protein stability. Mutations in the Zn-binding site of PBP2 cause a loss of bacterial rod shape and increase susceptibility to ß-lactams, therefore providing a direct rationale for cell wall shape maintenance and Zn homeostasis in A. baumannii. Furthermore, the Zn-coordinating residues are conserved in various ß- and γ-proteobacterial PBP2 orthologs, consistent with a widespread Zn-binding requirement for function that has been previously unknown. Due to the emergence of resistance to virtually all marketed antibiotic classes, alternative or complementary antimicrobial strategies need to be explored. These findings offer a perspective for dual inhibition of Zn-dependent PG synthases and metallo-ß-lactamases by metal chelating agents, considered the most sought-after adjuvants to restore ß-lactam potency against gram-negative bacteria.


Assuntos
Acinetobacter baumannii , Peptidil Transferases , Acinetobacter baumannii/metabolismo , Peptidil Transferases/metabolismo , Zinco/metabolismo , Forma Celular , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Proteínas de Ligação às Penicilinas/metabolismo , beta-Lactamas/farmacologia , Carbapenêmicos/farmacologia , Quelantes/farmacologia , Sítios de Ligação , Proteínas de Bactérias/metabolismo
4.
Annu Rev Microbiol ; 73: 481-506, 2019 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-31206345

RESUMO

Acinetobacter baumannii has emerged as an important nosocomial pathogen, particularly for patients in intensive care units and with invasive indwelling devices. The most recent clinical isolates are resistant to several classes of clinically important antibiotics, greatly restricting the ability to effectively treat critically ill patients. The bacterial envelope is an important driver of A. baumannii disease, both at the level of battling against antibiotic therapy and at the level of protecting from host innate immune function. This review provides a comprehensive overview of key features of the envelope that interface with both the host and antimicrobial therapies. Carbohydrate structures that contribute to protecting from the host are detailed, and mutations that alter these structures, resulting in increased antimicrobial resistance, are explored. In addition, protein complexes involved in both intermicrobial and host-microbe interactions are described. Finally we discuss regulatory mechanisms that control the nature of the cell envelope and its impact on host innate immune function.


Assuntos
Acinetobacter baumannii , Parede Celular/imunologia , Farmacorresistência Bacteriana Múltipla/genética , Glicolipídeos , Virulência/genética , Acinetobacter baumannii/citologia , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/genética , Acinetobacter baumannii/imunologia , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Biofilmes , Parede Celular/microbiologia , Infecção Hospitalar , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Genes Bacterianos , Glicolipídeos/imunologia , Glicolipídeos/metabolismo , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Canais Iônicos/genética , Canais Iônicos/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Interações Microbianas , Polissacarídeos Bacterianos , Porinas/genética , Porinas/metabolismo , Sistemas de Secreção Tipo II/genética , Sistemas de Secreção Tipo II/metabolismo , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/metabolismo , beta-Glucanas/imunologia , beta-Glucanas/metabolismo
5.
Immunity ; 41(3): 347-349, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25238090

RESUMO

The hallmark of bubonic plague is the presence of grotesquely swollen lymph nodes, called buboes. This frenzied inflammatory response to Yersinia pestis is poorly understood. In this issue of Immunity, St. John et al. (2014) explore the mechanism by which Y. pestis spreads and thus leads to this striking lymphadenopathy.


Assuntos
Linfonodos/patologia , Lisofosfolipídeos/genética , Peste/patologia , Receptores de Lisoesfingolipídeo/imunologia , Esfingosina/análogos & derivados , Yersinia pestis/patogenicidade , Animais , Feminino , Esfingosina/genética
6.
Cell ; 134(6): 907-10, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18805081

RESUMO

The 2008 Lasker-Koshland Award will be presented to Stanley Falkow, one of the legendary figures in the history of microbiology research. Falkow's many contributions remade the way we think about bacterial pathogens, antibiotic resistance, and infectious disease.


Assuntos
Distinções e Prêmios , Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Pesquisa Biomédica/história , Microbiologia/história , Bactérias/genética , Clonagem Molecular , Farmacorresistência Bacteriana , História do Século XX , Plasmídeos/genética , Fatores de Virulência/genética
7.
Infect Immun ; 90(11): e0017922, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36321832

RESUMO

Legionella pneumophila grows within membrane-bound vacuoles in alveolar macrophages during human disease. Pathogen manipulation of the host cell is driven by bacterial proteins translocated through a type IV secretion system (T4SS). Although host protein synthesis during infection is arrested by the action of several of these translocated effectors, translation of a subset of host proteins predicted to restrict the pathogen is maintained. To identify the spectrum of host proteins selectively synthesized after L. pneumophila challenge, macrophages infected with the pathogen were allowed to incorporate the amino acid analog azidohomoalanine (AHA) during a 2-h time window, and newly synthesized macrophage proteins were isolated by orthogonal chemistry followed by mass spectrometry. Among the proteins isolated were interferon-stimulated genes as well as proteins translated from highly abundant transcripts. Surprisingly, a large number of the identified proteins were from low-abundance transcripts. These proteins were predicted to be among the most efficiently translated per unit transcript in the cell based on ribosome profiling data sets. To determine if high ribosome loading was a consequence of efficient translation initiation, the 5' untranslated regions (5' UTR) of transcripts having the highest and lowest predicted loading levels were inserted upstream of a reporter, and translation efficiency was determined in response to L. pneumophila challenge. The efficiency of reporter expression largely correlated with predicted ribosome loading and lack of secondary structure. Therefore, determinants in the 5' UTR allow selected host cell transcripts to overcome a pathogen-driven translation blockade.


Assuntos
Legionella pneumophila , Humanos , Legionella pneumophila/fisiologia , Regiões 5' não Traduzidas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Interações Hospedeiro-Patógeno/genética , Vacúolos/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
8.
Proc Natl Acad Sci U S A ; 116(8): 3221-3228, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30718423

RESUMO

The cell cycle machinery controls diverse cellular pathways and is tightly regulated. Misregulation of cell division plays a central role in the pathogenesis of many disease processes. Various microbial pathogens interfere with the cell cycle machinery to promote host cell colonization. Although cell cycle modulation is a common theme among pathogens, the role this interference plays in promoting diseases is unclear. Previously, we demonstrated that the G1 and G2/M phases of the host cell cycle are permissive for Legionella pneumophila replication, whereas S phase provides a toxic environment for bacterial replication. In this study, we show that L. pneumophila avoids host S phase by blocking host DNA synthesis and preventing cell cycle progression into S phase. Cell cycle arrest upon Legionella contact is dependent on the Icm/Dot secretion system. In particular, we found that cell cycle arrest is dependent on the intact enzymatic activity of translocated substrates that inhibits host translation. Moreover, we show that, early in infection, the presence of these translation inhibitors is crucial to induce the degradation of the master regulator cyclin D1. Our results demonstrate that the bacterial effectors that inhibit translation are associated with preventing entry of host cells into a phase associated with restriction of L. pneumophila Furthermore, control of cyclin D1 may be a common strategy used by intracellular pathogens to manipulate the host cell cycle and promote bacterial replication.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Ciclina D1/genética , Interações Hospedeiro-Patógeno/genética , Legionella pneumophila/genética , Replicação do DNA/genética , Humanos , Imunidade Inata/genética , Legionella pneumophila/patogenicidade , Doença dos Legionários/genética , Doença dos Legionários/microbiologia , Macrófagos/metabolismo , Translocação Genética/genética
9.
Proc Natl Acad Sci U S A ; 116(36): 17775-17785, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31431530

RESUMO

Legionella pneumophila causes a potentially fatal form of pneumonia by replicating within macrophages in the Legionella-containing vacuole (LCV). Bacterial survival and proliferation within the LCV rely on hundreds of secreted effector proteins comprising high functional redundancy. The vacuolar membrane-localized MavN, hypothesized to support iron transport, is unique among effectors because loss-of-function mutations result in severe intracellular growth defects. We show here an iron starvation response by L. pneumophila after infection of macrophages that was prematurely induced in the absence of MavN, consistent with MavN granting access to limiting cellular iron stores. MavN cysteine accessibilities to a membrane-impermeant label were determined during macrophage infections, revealing a topological pattern supporting multipass membrane transporter models. Mutations to several highly conserved residues that can take part in metal recognition and transport resulted in defective intracellular growth. Purified MavN and mutant derivatives were directly tested for transporter activity after heterologous purification and liposome reconstitution. Proteoliposomes harboring MavN exhibited robust transport of Fe2+, with the severity of defect of most mutants closely mimicking the magnitude of defects during intracellular growth. Surprisingly, MavN was equivalently proficient at transporting Fe2+, Mn2+, Co2+, or Zn2+ Consequently, flooding infected cells with either Mn2+ or Zn2+ allowed collaboration with iron to enhance intracellular growth of L. pneumophila ΔmavN strains, indicating a clear role for MavN in transporting each of these ions. These findings reveal that MavN is a transition-metal-ion transporter that plays a critical role in response to iron limitation during Legionella infection.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte de Cátions , Legionella pneumophila , Metais/metabolismo , Vacúolos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Doença dos Legionários/genética , Doença dos Legionários/metabolismo , Doença dos Legionários/patologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Células U937 , Vacúolos/genética , Vacúolos/metabolismo
10.
J Bacteriol ; 203(12): e0056520, 2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-33782056

RESUMO

Acinetobacter baumannii is a poorly understood bacterium capable of life-threatening infections in hospitals. Few antibiotics remain effective against this highly resistant pathogen. Development of rationally designed antimicrobials that can target A. baumannii requires improved knowledge of the proteins that carry out essential processes allowing growth of the organism. Unfortunately, studying essential genes has been challenging using traditional techniques, which usually require time-consuming recombination-based genetic manipulations. Here, we performed saturating mutagenesis with dual transposon systems to identify essential genes in A. baumannii, and we developed a CRISPR interference (CRISPRi) system for facile analysis of these genes. We show that the CRISPRi system enables efficient transcriptional silencing in A. baumannii. Using these tools, we confirmed the essentiality of the novel cell division protein AdvA and discovered a previously uncharacterized AraC family transcription factor (ACX60_RS03245) that is necessary for growth. In addition, we show that capsule biosynthesis is a conditionally essential process, with mutations in late-acting steps causing toxicity in strain ATCC 17978 that can be bypassed by blocking early-acting steps or activating the BfmRS stress response. These results open new avenues for analysis of essential pathways in A. baumannii. IMPORTANCE New approaches are urgently needed to control A. baumannii, one of the most drug-resistant pathogens known. To facilitate the development of novel targets that allow inhibition of the pathogen, we performed a large-scale identification of genes whose products the bacterium needs for growth. We also developed a CRISPR-based gene knockdown tool that operates efficiently in A. baumannii, allowing rapid analysis of these essential genes. We used these methods to define multiple processes vital to the bacterium, including a previously uncharacterized gene regulatory factor and export of a protective polymeric capsule. These tools will enhance our ability to investigate processes critical for the essential biology of this challenging hospital-acquired pathogen.


Assuntos
Acinetobacter baumannii/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Elementos de DNA Transponíveis/fisiologia , Cápsulas Bacterianas , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Elementos de DNA Transponíveis/genética , Regulação Bacteriana da Expressão Gênica , Técnicas de Silenciamento de Genes , Mutagênese
11.
Infect Immun ; 89(12): e0043021, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34543120

RESUMO

Despite the maintenance of YopP/J alleles throughout the human-pathogenic Yersinia lineage, the benefit of YopP/J-induced phagocyte death for Yersinia pathogenesis in animals is not obvious. To determine how the sequence divergence of YopP/J has impacted Yersinia virulence, we examined protein polymorphisms in this type III secreted effector protein across 17 Yersinia species and tested the consequences of polymorphism in a murine model of subacute systemic yersiniosis. Our evolutionary analysis revealed that codon 177 has been subjected to positive selection; the Yersinia enterocolitica residue had been altered from a leucine to a phenylalanine in nearly all Yersinia pseudotuberculosis and Yersinia pestis strains examined. Despite this change being minor, as both leucine and phenylalanine have hydrophobic side chains, reversion of YopJF177 to the ancestral YopJL177 variant yielded a Y. pseudotuberculosis strain with enhanced cytotoxicity toward macrophages, consistent with previous findings. Surprisingly, expression of YopJF177L in the mildly attenuated ksgA- background rendered the strain completely avirulent in mice. Consistent with this hypothesis that YopJ activity relates indirectly to Yersinia pathogenesis in vivo, ksgA- strains lacking functional YopJ failed to kill macrophages but actually regained virulence in animals. Also, treatment with the antiapoptosis drug suramin prevented YopJ-mediated macrophage cytotoxicity and enhanced Y. pseudotuberculosis virulence in vivo. Our results demonstrate that Yersinia-induced cell death is detrimental for bacterial pathogenesis in this animal model of illness and indicate that positive selection has driven YopJ/P and Yersinia evolution toward diminished cytotoxicity and increased virulence, respectively.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Yersiniose/microbiologia , Yersinia/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Suscetibilidade a Doenças , Humanos , Mutação , Virulência/genética , Fatores de Virulência , Yersinia/patogenicidade
12.
Cell Microbiol ; 22(4): e13151, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32096265

RESUMO

Legionella pneumophila requires the Dot/Icm translocation system to replicate in a vacuolar compartment within host cells. Strains lacking the translocated substrate SdhA form a permeable vacuole during residence in the host cell, exposing bacteria to the host cytoplasm. In primary macrophages, mutants are defective for intracellular growth, with a pyroptotic cell death response mounted due to bacterial exposure to the cytosol. To understand how SdhA maintains vacuole integrity during intracellular growth, we performed high-throughput RNAi screens against host membrane trafficking genes to identify factors that antagonise vacuole integrity in the absence of SdhA. Depletion of host proteins involved in endocytic uptake and recycling resulted in enhanced intracellular growth and lower levels of permeable vacuoles surrounding the ΔsdhA mutant. Of interest were three different Rab GTPases involved in these processes: Rab11b, Rab8b and Rab5 isoforms, that when depleted resulted in enhanced vacuole integrity surrounding the sdhA mutant. Proteins regulated by these Rabs are responsible for interfering with proper vacuole membrane maintenance, as depletion of the downstream effectors EEA1, Rab11FIP1, or VAMP3 rescued vacuole integrity and intracellular growth of the sdhA mutant. To test the model that specific vesicular components associated with these effectors could act to destabilise the replication vacuole, EEA1 and Rab11FIP1 showed increased density about the sdhA mutant vacuole compared with the wild type (WT) vacuole. Depletion of Rab5 isoforms or Rab11b reduced this aberrant redistribution. These findings are consistent with SdhA interfering with both endocytic and recycling membrane trafficking events that act to destabilise vacuole integrity during infection.


Assuntos
Citosol/microbiologia , Endocitose , Interações Hospedeiro-Patógeno , Legionella pneumophila/crescimento & desenvolvimento , Vacúolos/microbiologia , Vacúolos/patologia , Animais , Proteínas de Bactérias/genética , Transporte Biológico , Feminino , Flavoproteínas/genética , Macrófagos/microbiologia , Camundongos , Transporte Proteico , Células RAW 264.7 , Interferência de RNA
13.
PLoS Pathog ; 14(5): e1007030, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29795704

RESUMO

The nosocomial pathogen Acinetobacter baumannii is a significant threat due to its ability to cause infections refractory to a broad range of antibiotic treatments. We show here that a highly conserved sensory-transduction system, BfmRS, mediates the coordinate development of both enhanced virulence and resistance in this microorganism. Hyperactive alleles of BfmRS conferred increased protection from serum complement killing and allowed lethal systemic disease in mice. BfmRS also augmented resistance and tolerance against an expansive set of antibiotics, including dramatic protection from ß-lactam toxicity. Through transcriptome profiling, we showed that BfmRS governs these phenotypes through global transcriptional regulation of a post-exponential-phase-like program of gene expression, a key feature of which is modulation of envelope biogenesis and defense pathways. BfmRS activity defended against cell-wall lesions through both ß-lactamase-dependent and -independent mechanisms, with the latter being connected to control of lytic transglycosylase production and proper coordination of morphogenesis and division. In addition, hypersensitivity of bfmRS knockouts could be suppressed by unlinked mutations restoring a short, rod cell morphology, indicating that regulation of drug resistance, pathogenicity, and envelope morphogenesis are intimately linked by this central regulatory system in A. baumannii. This work demonstrates that BfmRS controls a global regulatory network coupling cellular physiology to the ability to cause invasive, drug-resistant infections.


Assuntos
Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Farmacorresistência Bacteriana/genética , Infecções por Acinetobacter/patologia , Alelos , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Farmacorresistência Bacteriana/imunologia , Farmacorresistência Bacteriana/fisiologia , Farmacorresistência Bacteriana Múltipla/genética , Feminino , Regulação Bacteriana da Expressão Gênica/genética , Regulação Bacteriana da Expressão Gênica/imunologia , Homeostase/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/genética , Transcriptoma/imunologia , Virulência/efeitos dos fármacos , Virulência/imunologia , Resistência beta-Lactâmica/genética , beta-Lactamases/metabolismo
14.
Infect Immun ; 87(10)2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31331956

RESUMO

To successfully colonize host tissues, bacteria must respond to and detoxify many different host-derived antimicrobial compounds, such as nitric oxide (NO). NO has direct antimicrobial activity through attack on iron-sulfur (Fe-S) cluster-containing proteins. NO detoxification plays an important role in promoting bacterial survival, but it remains unclear if repair of Fe-S clusters is also important for bacterial survival within host tissues. Here we show that the Fe-S cluster repair protein YtfE contributes to the survival of Yersinia pseudotuberculosis within the spleen following nitrosative stress. Y. pseudotuberculosis forms clustered centers of replicating bacteria within deep tissues, where peripheral bacteria express the NO-detoxifying gene hmp. ytfE expression also occurred specifically within peripheral cells at the edges of microcolonies. In the absence of ytfE, the area of microcolonies was significantly smaller than that of the wild type (WT), consistent with ytfE contributing to the survival of peripheral cells. The loss of ytfE did not alter the ability of cells to detoxify NO, which occurred within peripheral cells in both WT and ΔytfE microcolonies. In the absence of NO-detoxifying activity by hmp, NO diffused across ΔytfE microcolonies, and there was a significant decrease in the area of microcolonies lacking ytfE, indicating that ytfE also contributes to bacterial survival in the absence of NO detoxification. These results indicate a role for Fe-S cluster repair in the survival of Y. pseudotuberculosis within the spleen and suggest that extracellular bacteria may rely on this pathway for survival within host tissues.


Assuntos
Proteínas de Bactérias/genética , Proteínas Ferro-Enxofre/genética , NADH NADPH Oxirredutases/genética , Óxido Nítrico/metabolismo , Infecções por Yersinia pseudotuberculosis/microbiologia , Yersinia pseudotuberculosis/genética , Animais , Proteínas de Bactérias/metabolismo , Feminino , Deleção de Genes , Expressão Gênica , Interações Hospedeiro-Patógeno , Proteínas Ferro-Enxofre/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , NADH NADPH Oxirredutases/metabolismo , Óxido Nítrico/antagonistas & inibidores , Baço/microbiologia , Yersinia pseudotuberculosis/enzimologia
15.
BMC Genomics ; 20(1): 961, 2019 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-31823727

RESUMO

BACKGROUND: During infection by intracellular pathogens, a highly complex interplay occurs between the infected cell trying to degrade the invader and the pathogen which actively manipulates the host cell to enable survival and proliferation. Many intracellular pathogens pose important threats to human health and major efforts have been undertaken to better understand the host-pathogen interactions that eventually determine the outcome of the infection. Over the last decades, the unicellular eukaryote Dictyostelium discoideum has become an established infection model, serving as a surrogate macrophage that can be infected with a wide range of intracellular pathogens. In this study, we use high-throughput RNA-sequencing to analyze the transcriptional response of D. discoideum when infected with Mycobacterium marinum and Legionella pneumophila. The results were compared to available data from human macrophages. RESULTS: The majority of the transcriptional regulation triggered by the two pathogens was found to be unique for each bacterial challenge. Hallmark transcriptional signatures were identified for each infection, e.g. induction of endosomal sorting complexes required for transport (ESCRT) and autophagy genes in response to M. marinum and inhibition of genes associated with the translation machinery and energy metabolism in response to L. pneumophila. However, a common response to the pathogenic bacteria was also identified, which was not induced by non-pathogenic food bacteria. Finally, comparison with available data sets of regulation in human monocyte derived macrophages shows that the elicited response in D. discoideum is in many aspects similar to what has been observed in human immune cells in response to Mycobacterium tuberculosis and L. pneumophila. CONCLUSIONS: Our study presents high-throughput characterization of D. discoideum transcriptional response to intracellular pathogens using RNA-seq. We demonstrate that the transcriptional response is in essence distinct to each pathogen and that in many cases, the corresponding regulation is recapitulated in human macrophages after infection by mycobacteria and L. pneumophila. This indicates that host-pathogen interactions are evolutionary conserved, derived from the early interactions between free-living phagocytic cells and bacteria. Taken together, our results strengthen the use of D. discoideum as a general infection model.


Assuntos
Infecções Bacterianas/microbiologia , Dictyostelium/microbiologia , Modelos Biológicos , Proteínas de Protozoários/genética , Células Cultivadas , Citoplasma/microbiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Legionella pneumophila/fisiologia , Macrófagos/microbiologia , Mycobacterium marinum/fisiologia , Proteínas de Protozoários/metabolismo , Especificidade da Espécie , Transcrição Gênica
16.
Bioessays ; 38(8): 782-90, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27273675

RESUMO

Bacterial populations are heterogeneous, which in many cases can provide a selective advantage during changes in environmental conditions. In some instances, heterogeneity exists at the genetic level, in which significant allelic variation occurs within a population seeded by a single cell. In other cases, heterogeneity exists due to phenotypic differences within a clonal, genetically identical population. A variety of mechanisms can drive this latter strategy. Stochastic fluctuations can drive differential gene expression, but heterogeneity in gene expression can also be driven by environmental changes sensed by individual cells residing in distinct locales. Utilizing multiple single cell approaches, workers have started to uncover the extent of heterogeneity within bacterial populations. This review will first describe several examples of phenotypic and genetic heterogeneity, and then discuss many single cell approaches that have recently been applied to define heterogeneity within bacterial populations.


Assuntos
Fenômenos Fisiológicos Bacterianos , Interações Hospedeiro-Patógeno , Análise de Célula Única , Adaptação Fisiológica , Animais , Bactérias/genética , Células Clonais , Meio Ambiente , Regulação Bacteriana da Expressão Gênica , Variação Genética , Humanos
17.
Proc Natl Acad Sci U S A ; 112(49): E6790-7, 2015 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-26598709

RESUMO

Cells of the innate immune system recognize bacterial pathogens by detecting common microbial patterns as well as pathogen-specific activities. One system that responds to these stimuli is the IRE1 branch of the unfolded protein response (UPR), a sensor of endoplasmic reticulum (ER) stress. Activation of IRE1, in the context of Toll-like receptor (TLR) signaling, induces strong proinflammatory cytokine induction. We show here that Legionella pneumophila, an intravacuolar pathogen that replicates in an ER-associated compartment, blocks activation of the IRE1 pathway despite presenting pathogen products that stimulate this response. L. pneumophila TLR ligands induced the splicing of mRNA encoding XBP1s, the main target of IRE1 activity. L. pneumophila was able to inhibit both chemical and bacterial induction of XBP1 splicing via bacterial translocated proteins that interfere with host protein translation. A strain lacking five translocated translation elongation inhibitors was unable to block XBP1 splicing, but this could be rescued by expression of a single such inhibitor, consistent with limitation of the response by translation elongation inhibitors. Chemical inhibition of translation elongation blocked pattern recognition receptor-mediated XBP1 splicing, mimicking the effects of the bacterial translation inhibitors. In contrast, host cell-promoted inhibition of translation initiation in response to the pathogen was ineffective in blocking XBP1 splicing, demonstrating the need for the elongation inhibitors for protection from the UPR. The inhibition of host translation elongation may be a common strategy used by pathogens to limit the innate immune response by interfering with signaling via the UPR.


Assuntos
Legionella pneumophila/fisiologia , Biossíntese de Proteínas , Resposta a Proteínas não Dobradas , Cicloeximida/farmacologia , Proteínas de Ligação a DNA/genética , Endorribonucleases/fisiologia , Humanos , Proteínas Serina-Treonina Quinases/fisiologia , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição/genética , Proteína 1 de Ligação a X-Box
18.
Proc Natl Acad Sci U S A ; 112(37): E5208-17, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26330609

RESUMO

Iron is essential for the growth and virulence of most intravacuolar pathogens. The mechanisms by which microbes bypass host iron restriction to gain access to this metal across the host vacuolar membrane are poorly characterized. In this work, we identify a unique intracellular iron acquisition strategy used by Legionella pneumophila. The bacterial Icm/Dot (intracellular multiplication/defect in organelle trafficking) type IV secretion system targets the bacterial-derived MavN (more regions allowing vacuolar colocalization N) protein to the surface of the Legionella-containing vacuole where this putative transmembrane protein facilitates intravacuolar iron acquisition. The ΔmavN mutant exhibits a transcriptional iron-starvation signature before its growth is arrested during the very early stages of macrophage infection. This intracellular growth defect is rescued only by the addition of excess exogenous iron to the culture medium and not a variety of other metals. Consistent with MavN being a translocated substrate that plays an exclusive role during intracellular growth, the mutant shows no defect for growth in broth culture, even under severe iron-limiting conditions. Putative iron-binding residues within the MavN protein were identified, and point mutations in these residues resulted in defects specific for intracellular growth that are indistinguishable from the ΔmavN mutant. This model of a bacterial protein inserting into host membranes to mediate iron transport provides a paradigm for how intravacuolar pathogens can use virulence-associated secretion systems to manipulate and acquire host iron.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas de Transporte de Cátions/fisiologia , Legionella pneumophila/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Proliferação de Células , Meios de Cultura/química , Citoplasma/metabolismo , Dictyostelium/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Sistema Imunitário , Ferro/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Dados de Sequência Molecular , Fagossomos/metabolismo , Mutação Puntual , Estrutura Secundária de Proteína , Transporte Proteico , Células RAW 264.7 , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Células U937 , Vacúolos/metabolismo , Virulência
19.
J Infect Dis ; 215(suppl_1): S9-S17, 2017 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-28375515

RESUMO

Diseases caused by antibiotic-resistant bacteria in hospitals are the outcome of complex relationships between several dynamic factors, including bacterial pathogenicity, the fitness costs of resistance in the human host, and selective forces resulting from interventions such as antibiotic therapy. The emergence and fate of mutations that drive antibiotic resistance are governed by these interactions. In this review, we will examine how different forms of antibiotic resistance modulate bacterial fitness and virulence potential, thus influencing the ability of pathogens to evolve in the context of nosocomial infections. We will focus on 3 important multidrug-resistant pathogens that are notoriously problematic in hospitals: Pseudomonas aeruginosa, Acinetobacter baumannii, and Staphylococcus aureus. An understanding of how antibiotic resistance mutations shape the pathobiology of multidrug-resistant infections has the potential to drive novel strategies that can control the development and spread of drug resistance.


Assuntos
Acinetobacter baumannii/genética , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla/genética , Pseudomonas aeruginosa/genética , Staphylococcus aureus/genética , Infecções por Acinetobacter/tratamento farmacológico , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/patogenicidade , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecção Hospitalar/tratamento farmacológico , Regulação Bacteriana da Expressão Gênica , Humanos , Testes de Sensibilidade Microbiana , Mutação , Porinas/genética , Porinas/metabolismo , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/patogenicidade , Fatores de Virulência/genética , Resistência beta-Lactâmica/genética , beta-Lactamases/genética , beta-Lactamases/metabolismo
20.
PLoS Pathog ; 11(2): e1004691, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25679516

RESUMO

Acinetobacter baumannii is an opportunistic pathogen of increasing importance due to its propensity for intractable multidrug-resistant infections in hospitals. All clinical isolates examined contain a conserved gene cluster, the K locus, which determines the production of complex polysaccharides, including an exopolysaccharide capsule known to protect against killing by host serum and to increase virulence in animal models of infection. Whether the polysaccharides determined by the K locus contribute to intrinsic defenses against antibiotics is unknown. We demonstrate here that mutants deficient in the exopolysaccharide capsule have lowered intrinsic resistance to peptide antibiotics, while a mutation affecting sugar precursors involved in both capsule and lipopolysaccharide synthesis sensitizes the bacterium to multiple antibiotic classes. We observed that, when grown in the presence of certain antibiotics below their MIC, including the translation inhibitors chloramphenicol and erythromycin, A. baumannii increases production of the K locus exopolysaccharide. Hyperproduction of capsular exopolysaccharide is reversible and non-mutational, and occurs concomitantly with increased resistance to the inducing antibiotic that is independent of the presence of the K locus. Strikingly, antibiotic-enhanced capsular exopolysaccharide production confers increased resistance to killing by host complement and increases virulence in a mouse model of systemic infection. Finally, we show that augmented capsule production upon antibiotic exposure is facilitated by transcriptional increases in K locus gene expression that are dependent on a two-component regulatory system, bfmRS. These studies reveal that the synthesis of capsule, a major pathogenicity determinant, is regulated in response to antibiotic stress. Our data are consistent with a model in which gene expression changes triggered by ineffectual antibiotic treatment cause A. baumannii to transition between states of low and high virulence potential, which may contribute to the opportunistic nature of the pathogen.


Assuntos
Infecções por Acinetobacter/patologia , Acinetobacter baumannii/patogenicidade , Cápsulas Bacterianas/genética , Farmacorresistência Bacteriana Múltipla/fisiologia , Polissacarídeos Bacterianos/genética , Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Animais , Antibacterianos/farmacologia , Modelos Animais de Doenças , Farmacorresistência Bacteriana Múltipla/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA