Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Elife ; 92020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32808593

RESUMO

Pathogens find diverse niches for survival including inside a host cell where replication occurs in a relatively protective environment. Vibrio parahaemolyticus is a facultative intracellular pathogen that uses its type 3 secretion system 2 (T3SS2) to invade and replicate inside host cells. Analysis of the T3SS2 pathogenicity island encoding the T3SS2 appeared to lack a mechanism for egress of this bacterium from the invaded host cell. Using a combination of molecular tools, we found that VPA0226, a constitutively secreted lipase, is required for escape of V. parahaemolyticus from the host cells. This lipase must be delivered into the host cytoplasm where it preferentially uses fatty acids associated with innate immune response to esterify cholesterol, weakening the plasma membrane and allowing egress of the bacteria. This study reveals the resourcefulness of microbes and the interplay between virulence systems and host cell resources to evolve an ingenious scheme for survival and escape.


Assuntos
Proteínas de Bactérias/metabolismo , Colesterol/metabolismo , Ácidos Graxos/metabolismo , Lipase/metabolismo , Vibrio parahaemolyticus/metabolismo , Esterificação , Ilhas Genômicas , Sistemas de Secreção Tipo III , Vibrio parahaemolyticus/enzimologia
2.
mBio ; 10(6)2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848276

RESUMO

The Gram-negative marine bacterium Vibrio parahaemolyticus is a common cause of infectious gastroenteritis due to the ingestion of contaminated seafood. Most virulent V. parahaemolyticus strains encode two type III secretion systems (T3SS1 and T3SS2); however, the roles they and their translocated effectors play in causing intestinal disease remain unclear. While studies have identified T3SS1 effectors as responsible for killing epithelial cells in culture, the T3SS2 effectors caused massive epithelial cell disruption in a rabbit ileal loop model. Additional models are thus needed to clarify the pathogen-host interactions that drive V. parahaemolyticus-associated gastroenteritis. Germfree mice were infected with a pathogenic clinical isolate of V. parahaemolyticus, RIMD2210633 (RIMD). The pathogen was found to adhere to as well as invade the cecal mucosa, accompanied by severe inflammation and dramatic mucosal damage, including widespread sloughing of infected epithelial cells. Mice infected with a V. parahaemolyticus strain lacking the T3SS1 (POR2) also developed severe pathology, similar to that seen with RIMD. In contrast, the ΔT3SS2 strain (POR3) appeared unable to invade the intestinal mucosa or cause any mucosal pathology. Confirming a role for TS332 effectors, a strain expressing the T3SS2 but lacking VopC (POR2ΔvopC), a T3SS2 effector implicated in epithelial cell invasion in culture, was strongly attenuated in invading the intestinal mucosa and in causing gastroenteritis, although infection with this mutant resulted in more pathology than the ΔT3SS2 strain. We thus present an experimental system that enables further characterization of T3SS effectors as well as the corresponding host inflammatory response involved in the gastroenteritis caused by invasive V. parahaemolyticusIMPORTANCEVibrio parahaemolyticus causes severe gastroenteritis following consumption of contaminated seafood. Global warming has allowed this pathogen to spread worldwide, contributing to recent outbreaks. Clinical isolates are known to harbor an array of virulence factors, including T3SS1 and T3SS2; however, the precise role these systems play in intestinal disease remains unclear. There is an urgent need to improve our understanding of how V. parahaemolyticus infects hosts and causes disease. We present a novel mouse model for this facultative intracellular pathogen and observe that the T3SS2 is essential to pathogenicity. Moreover, we show that the T3SS2 effector VopC, previously shown to be a Rac and Cdc42 deamidase that facilitates bacterial uptake by nonphagocytic cells, also plays a key role in the ability of V. parahaemolyticus to invade the intestinal mucosa and cause gastroenteritis. This experimental model thus provides a valuable tool for future elucidation of virulence mechanisms used by this facultative intracellular pathogen during in vivo infection.


Assuntos
Gastroenterite/microbiologia , Sistemas de Secreção Tipo III , Vibrioses/microbiologia , Vibrio parahaemolyticus/fisiologia , Fatores de Virulência/genética , Animais , Antibacterianos/farmacologia , Morte Celular , Proliferação de Células , Modelos Animais de Doenças , Farmacorresistência Bacteriana , Gastroenterite/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Estreptomicina/farmacologia , Vibrio parahaemolyticus/efeitos dos fármacos , Virulência
3.
Microbiol Spectr ; 7(3)2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31152523

RESUMO

Several pathogens have evolved to infect host cells from within, which requires subversion of many host intracellular processes. In the case of Gram-negative pathogenic bacteria, adaptation to an intracellular life cycle relies largely on the activity of type III secretion systems (T3SSs), an apparatus used to deliver effector proteins into the host cell, from where these effectors regulate important cellular functions such as vesicular trafficking, cytoskeleton reorganization, and the innate immune response. Each bacterium is equipped with a unique suite of these T3SS effectors, which aid in the development of an individual intracellular lifestyle for their respective pathogens. Some bacteria adapt to reside and propagate within a customized vacuole, while others establish a replicative niche in the host cytosol. In this article, we review the mechanisms by which T3SS effectors contribute to these different lifestyles. To illustrate the formation of a vacuolar and a cytosolic lifestyle, we discuss the intracellular habitats of the enteric pathogens Salmonella enterica serovar Typhimurium and Shigella flexneri, respectively. These represent well-characterized systems that function as informative models to contribute to our understanding of T3SS-dependent subversion of intracellular processes. Additionally, we present Vibrio parahaemolyticus, another enteric Gram-negative pathogen, as an emerging model for future studies of the cytosolic lifestyle.


Assuntos
Citoplasma/metabolismo , Citoplasma/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Citosol/microbiologia , Bactérias Gram-Negativas/patogenicidade , Bactérias Gram-Negativas/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Transporte Proteico , Salmonella typhimurium/patogenicidade , Salmonella typhimurium/fisiologia , Shigella flexneri/patogenicidade , Shigella flexneri/fisiologia , Vacúolos/microbiologia , Vibrio parahaemolyticus/patogenicidade , Vibrio parahaemolyticus/fisiologia
4.
PLoS One ; 14(5): e0213423, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31048915

RESUMO

Pore-forming proteins (PFPs) represent a functionally important protein family, that are found in organisms from viruses to humans. As a major branch of PFPs, bacteria pore-forming toxins (PFTs) permeabilize membranes and usually cause the death of target cells. E. coli hemolysin ClyA is the first member with the pore complex structure solved among α-PFTs, employing α-helices as transmembrane elements. ClyA is proposed to form pores composed of various numbers of protomers. With high-resolution cryo-EM structures, we observe that ClyA pore complexes can exist as newly confirmed oligomers of a tridecamer and a tetradecamer, at estimated resolutions of 3.2 Å and 4.3 Å, respectively. The 2.8 Å cryo-EM structure of a dodecamer dramatically improves the existing structural model. Structural analysis indicates that protomers from distinct oligomers resemble each other and neighboring protomers adopt a conserved interaction mode. We also show a stabilized intermediate state of ClyA during the transition process from soluble monomers to pore complexes. Unexpectedly, even without the formation of mature pore complexes, ClyA can permeabilize membranes and allow leakage of particles less than ~400 Daltons. In addition, we are the first to show that ClyA forms pore complexes in the presence of cholesterol within artificial liposomes. These findings provide new mechanistic insights into the dynamic process of pore assembly for the prototypical α-PFT ClyA.


Assuntos
Microscopia Crioeletrônica/métodos , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestrutura , Escherichia coli/metabolismo , Escherichia coli/ultraestrutura , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/ultraestrutura , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Proteínas de Escherichia coli/química , Proteínas Hemolisinas/química , Lipossomos/metabolismo , Lipossomos/ultraestrutura , Multimerização Proteica
5.
Elife ; 82019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31134894

RESUMO

Cancer evolves through a multistep process that occurs by the temporal accumulation of genetic mutations. Tumor-derived exosomes are emerging contributors to tumorigenesis. To understand how exosomes might contribute to cell transformation, we utilized the classic two-step NIH/3T3 cell transformation assay and observed that exosomes isolated from pancreatic cancer cells, but not normal human cells, can initiate malignant cell transformation and these transformed cells formed tumors in vivo. However, cancer cell exosomes are unable to transform cells alone or to act as a promoter of cell transformation. Utilizing proteomics and exome sequencing, we discovered cancer cell exosomes act as an initiator by inducing random mutations in recipient cells. Cells from the pool of randomly mutated cells are driven to transformation by a classic promoter resulting in foci, each of which encode a unique genetic profile. Our studies describe a novel molecular understanding of how cancer cell exosomes contribute to cell transformation. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that major issues remain unresolved (see decision letter).


Assuntos
Transformação Celular Neoplásica/patologia , Exossomos/metabolismo , Neoplasias Pancreáticas/patologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Exossomos/química , Genômica , Humanos , Camundongos , Células NIH 3T3 , Transplante de Neoplasias , Proteômica
6.
J Mol Biol ; 431(21): 4368-4379, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31002774

RESUMO

Urinary tract infections (UTIs) are the most commonly reported infections in adult women and have high rates of recurrence, especially in postmenopausal women. Recurrent UTI (RUTI) greatly reduces quality of life, places a significant burden on the healthcare system, and contributes to antimicrobial resistance. Because treatment of RUTI by long-term antibiotic therapy is often ineffective or poorly tolerated in elderly women, new therapies must be developed. The molecular basis of RUTI, especially in postmenopausal women, has remained unclear because modeling RUTI in mice is difficult, and human data are limited. Invasion of the urothelium and induction of host inflammation are hypothesized to be key mechanisms by which bacterial pathogens cause RUTI. To further our understanding of RUTI in humans, we performed a systematic analysis of urine and bladder biopsy samples from postmenopausal women undergoing cystoscopy with fulguration of trigonitis in the advanced management of antibiotic-refractory RUTI. We provide direct evidence that bacteria reside in the bladder wall of postmenopausal RUTI patients and that diverse bacterial species can be isolated from the bladder tissue. Histopathological scoring revealed significant edema and alterations of urothelial architecture in RUTI patient biopsies. Lymphocytes, including plasma B-cells, were detected within the mesenchyme, urothelium, and follicular aggregates in the majority of patients, indicating that the local adaptive immune response is active during human RUTI. These data provide conclusive evidence that bacteria invade the human urothelium and suggest that diverse bacterial species and the adaptive immune response play important roles in RUTI in humans.


Assuntos
Inflamação/microbiologia , Bexiga Urinária/microbiologia , Infecções Urinárias/microbiologia , Feminino , Humanos , Pós-Menopausa
7.
J Bacteriol ; 200(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29555695

RESUMO

The Gram-negative bacterium Vibrio parahaemolyticus is an opportunistic human pathogen and the leading cause of seafood-borne acute gastroenteritis worldwide. Recently, this bacterium was implicated as the etiologic agent of a severe shrimp disease with consequent devastating outcomes to shrimp farming. In both cases, acquisition of genetic material via horizontal transfer provided V. parahaemolyticus with new virulence tools to cause disease. Dissecting the molecular mechanisms of V. parahaemolyticus pathogenesis often requires manipulating its genome. Classically, genetic deletions in V. parahaemolyticus are performed using a laborious, lengthy, multistep process. Here, we describe a fast and efficient method to edit this bacterium's genome based on V. parahaemolyticus natural competence. Although this method is similar to one previously described, V. parahaemolyticus requires counterselection for curing of acquired plasmids due to its recalcitrant nature of retaining extrachromosomal DNA. We believe this approach will be of use to the Vibrio community.IMPORTANCE Spreading of vibrios throughout the world correlates with increased global temperatures. As they spread, they find new niches in which to survive, proliferate, and invade. Therefore, genetic manipulation of vibrios is of the utmost importance for studying these species. Here, we have delineated and validated a rapid method to create genetic deletions in Vibrio parahaemolyticus This study provides insightful methodology for studies with other Vibrio species.


Assuntos
Proteínas de Bactérias/metabolismo , Deleção de Genes , Regulação Bacteriana da Expressão Gênica/fisiologia , Vibrio parahaemolyticus/genética , Proteínas de Bactérias/genética , Plasmídeos , Transformação Genética , Vibrio parahaemolyticus/fisiologia
8.
EMBO Rep ; 18(11): 1978-1990, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28912123

RESUMO

Most type VI secretion systems (T6SSs) described to date are protein delivery apparatuses that mediate bactericidal activities. Several T6SSs were also reported to mediate virulence activities, although only few anti-eukaryotic effectors have been described. Here, we identify three T6SSs in the marine bacterium Vibrio proteolyticus and show that T6SS1 mediates bactericidal activities under warm marine-like conditions. Using comparative proteomics, we find nine potential T6SS1 effectors, five of which belong to the polymorphic MIX-effector class. Remarkably, in addition to six predicted bactericidal effectors, the T6SS1 secretome includes three putative anti-eukaryotic effectors. One of these is a MIX-effector containing a cytotoxic necrotizing factor 1 domain. We demonstrate that T6SS1 can use this MIX-effector to target phagocytic cells, resulting in morphological changes and actin cytoskeleton rearrangements. In conclusion, the V. proteolyticus T6SS1, a system homologous to one found in pathogenic vibrios, uses a suite of polymorphic effectors that target both bacteria and eukaryotic neighbors.


Assuntos
Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Cromossomos Bacterianos/química , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Sistemas de Secreção Tipo VI/genética , Vibrio/genética , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/ultraestrutura , Animais , Antibacterianos/metabolismo , Antibacterianos/toxicidade , Organismos Aquáticos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Mapeamento Cromossômico , Técnicas de Cocultura , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidade , Camundongos , Fagócitos/citologia , Fagócitos/efeitos dos fármacos , Domínios Proteicos , Células RAW 264.7 , Sistemas de Secreção Tipo VI/química , Sistemas de Secreção Tipo VI/metabolismo , Vibrio/metabolismo , Vibrio/patogenicidade , Virulência
9.
PLoS Pathog ; 13(6): e1006438, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28640881

RESUMO

The production of antimicrobial reactive oxygen species by the nicotinamide dinucleotide phosphate (NADPH) oxidase complex is an important mechanism for control of invading pathogens. Herein, we show that the gastrointestinal pathogen Vibrio parahaemolyticus counteracts reactive oxygen species (ROS) production using the Type III Secretion System 2 (T3SS2) effector VopL. In the absence of VopL, intracellular V. parahaemolyticus undergoes ROS-dependent filamentation, with concurrent limited growth. During infection, VopL assembles actin into non-functional filaments resulting in a dysfunctional actin cytoskeleton that can no longer mediate the assembly of the NADPH oxidase at the cell membrane, thereby limiting ROS production. This is the first example of how a T3SS2 effector contributes to the intracellular survival of V. parahaemolyticus, supporting the establishment of a protective intracellular replicative niche.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Sistemas de Secreção Tipo III/metabolismo , Vibrioses/metabolismo , Vibrio parahaemolyticus/metabolismo , Células CACO-2 , Humanos , Microscopia Confocal , Espécies Reativas de Oxigênio/metabolismo
10.
Sci Rep ; 6: 39341, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27996032

RESUMO

Classical antimicrobial drugs target proliferation and therefore place microbes under extreme selective pressure to evolve resistance. Alternative drugs that target bacterial virulence without impacting survival directly offer an attractive solution to this problem, but to date few such molecules have been discovered. We previously discovered a widespread group of bacterial adhesins, termed Multivalent Adhesion Molecules (MAMs) that are essential for initial binding of bacteria to host tissues and virulence. Thus, targeting MAM-based adherence is a promising strategy for displacing pathogens from host tissues and inhibiting infection. Here, we show that topical application of polymeric microbeads functionalized with the adhesin MAM7 to a burn infected with multidrug-resistant Pseudomonas aeruginosa substantially decreased bacterial loads in the wound and prevented the spread of the infection into adjacent tissues. As a consequence, the application of this adhesion inhibitor allowed for vascularization and wound healing, and maintained local and systemic inflammatory responses to the burn. We propose that MAM7-functionalized microbeads can be used as a topical treatment, to reduce bacterial attachment and hence prevent bacterial colonization and infection of wounds. As adhesion is not required for microbial survival, this anti-infective strategy has the potential to treat multidrug-resistant infections and limit the emergence of drug-resistant pathogens.


Assuntos
Adesinas Bacterianas/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Queimaduras/microbiologia , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/metabolismo , Infecção dos Ferimentos/prevenção & controle , Adesinas Bacterianas/metabolismo , Animais , Antibacterianos/farmacologia , Carga Bacteriana/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla , Humanos , Masculino , Microesferas , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Ratos , Ratos Sprague-Dawley , Cicatrização/efeitos dos fármacos , Infecção dos Ferimentos/tratamento farmacológico , Infecção dos Ferimentos/microbiologia
11.
mBio ; 7(4)2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27460800

RESUMO

UNLABELLED: Members of the genus Vibrio include many pathogens of humans and marine animals that share genetic information via horizontal gene transfer. Hence, the Vibrio pan-genome carries the potential to establish new pathogenic strains by sharing virulence determinants, many of which have yet to be characterized. Here, we investigated the virulence properties of Vibrio proteolyticus, a Gram-negative marine bacterium previously identified as part of the Vibrio consortium isolated from diseased corals. We found that V. proteolyticus causes actin cytoskeleton rearrangements followed by cell lysis in HeLa cells in a contact-independent manner. In search of the responsible virulence factor involved, we determined the V. proteolyticus secretome. This proteomics approach revealed various putative virulence factors, including active type VI secretion systems and effectors with virulence toxin domains; however, these type VI secretion systems were not responsible for the observed cytotoxic effects. Further examination of the V. proteolyticus secretome led us to hypothesize and subsequently demonstrate that a secreted hemolysin, belonging to a previously uncharacterized clan of the leukocidin superfamily, was the toxin responsible for the V. proteolyticus-mediated cytotoxicity in both HeLa cells and macrophages. Clearly, there remains an armory of yet-to-be-discovered virulence factors in the Vibrio pan-genome that will undoubtedly provide a wealth of knowledge on how a pathogen can manipulate host cells. IMPORTANCE: The pan-genome of the genus Vibrio is a potential reservoir of unidentified toxins that can provide insight into how members of this genus have successfully risen as emerging pathogens worldwide. We focused on Vibrio proteolyticus, a marine bacterium that was previously implicated in virulence toward marine animals, and characterized its interaction with eukaryotic cells. We found that this bacterium causes actin cytoskeleton rearrangements and leads to cell death. Using a proteomics approach, we identified a previously unstudied member of the leukocidin family of pore-forming toxins as the virulence factor responsible for the observed cytotoxicity in eukaryotic cells, as well as a plethora of additional putative virulence factors secreted by this bacterium. Our findings reveal a functional new clan of the leukocidin toxin superfamily and establish this pathogen as a reservoir of potential toxins that can be used for biomedical applications.


Assuntos
Proteínas Hemolisinas/análise , Proteômica , Vibrio/química , Fatores de Virulência/análise , Animais , Organismos Aquáticos/química , Sobrevivência Celular/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Células HeLa , Proteínas Hemolisinas/metabolismo , Humanos , Macrófagos/microbiologia , Macrófagos/fisiologia , Camundongos , Células RAW 264.7
12.
Cell Microbiol ; 17(2): 164-73, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25440316

RESUMO

Entry into host cells and intracellular persistence by invasive bacteria are tightly coupled to the ability of the bacterium to disrupt the eukaryotic cytoskeletal machinery. Herein we review the main strategies used by three intracellular pathogens to harness key modulators of the cytoskeleton. Two of these bacteria, namely Listeria monocytogenes and Salmonella enterica serovar Typhimurium, exhibit quite distinct intracellular lifestyles and therefore provide a comprehensive panel for the understanding of the intricate bacteria-cytoskeleton interplay during infections. The emerging intracellular pathogen Vibrio parahaemolyticus is depicted as a developing model for the uncovering of novel mechanisms used to hijack the cytoskeleton.


Assuntos
Citoesqueleto/metabolismo , Interações Hospedeiro-Patógeno , Listeria monocytogenes/fisiologia , Salmonella typhimurium/fisiologia , Vibrio parahaemolyticus/fisiologia , Endocitose , Listeria monocytogenes/crescimento & desenvolvimento , Salmonella typhimurium/crescimento & desenvolvimento , Vibrio parahaemolyticus/crescimento & desenvolvimento
13.
FEMS Microbiol Lett ; 361(2): 107-14, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25288215

RESUMO

The Gram-negative bacterium, Vibrio parahaemolyticus, is a major cause of seafood-derived food poisoning throughout the world. The pathogenicity of V. parahaemolyticus is attributed to several virulence factors, including two type III secretion systems (T3SS), T3SS1 and T3SS2. Herein, we compare the virulence of V. parahaemolyticus POR strains, which harbor a mutation in the T3SS needle apparatus of either system, to V. parahaemolyticus CAB strains, which harbor mutations in positive transcriptional regulators of either system. These strains are derived from the clinical RIMD 2210633 strain. We demonstrate that each mutation affects the virulence of the bacterium in a different manner. POR and CAB strains exhibited similar levels of swarming motility and T3SS effector production and secretion, but the CAB3 and CAB4 strains, which harbor a mutation in the T3SS2 master regulator gene, formed reduced biofilm growth under T3SS2 inducing conditions. Additionally, while the cytotoxicity of the POR and CAB strains was similar, the CAB2 (T3SS1 regulatory mutant) strain was strikingly more invasive than the comparable POR2 (T3SS1 structural mutant) strain. In summary, creating structural or regulatory mutations in either T3SS1 or T3SS2 causes differential downstream effects on other virulence systems. Understanding the biological differences of strains created from a clinical isolate is critical for interpreting and understanding the pathogenic nature of V. parahaemolyticus.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos , Mutação , Vibrioses/microbiologia , Vibrio parahaemolyticus/metabolismo , Vibrio parahaemolyticus/patogenicidade , Proteínas de Bactérias/química , Regulação Bacteriana da Expressão Gênica , Células HeLa , Humanos , Vibrio parahaemolyticus/química , Vibrio parahaemolyticus/genética , Virulência
14.
mBio ; 5(5): e01506-14, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25205094

RESUMO

UNLABELLED: Vibrio parahaemolyticus is a globally disseminated Gram-negative marine bacterium and the leading cause of seafood-borne acute gastroenteritis. Pathogenic bacterial isolates encode two type III secretion systems (T3SS), with the second system (T3SS2) considered the main virulence factor in mammalian hosts. For many decades, V. parahaemolyticus has been studied as an exclusively extracellular bacterium. However, the recent characterization of the T3SS2 effector protein VopC has suggested that this pathogen has the ability to invade, survive, and replicate within epithelial cells. Herein, we characterize this intracellular lifestyle in detail. We show that following internalization, V. parahaemolyticus is contained in vacuoles that develop into early endosomes, which subsequently mature into late endosomes. V. parahaemolyticus then escapes into the cytoplasm prior to vacuolar fusion with lysosomes. Vacuolar acidification is an important trigger for this escape. The cytoplasm serves as the pathogen's primary intracellular replicative niche; cytosolic replication is rapid and robust, with cells often containing over 150 bacteria by the time of cell lysis. These results show how V. parahaemolyticus successfully establishes an intracellular lifestyle that could contribute to its survival and dissemination during infection. IMPORTANCE: The marine bacterium V. parahaemolyticus is the leading cause worldwide of seafood-borne acute gastroenteritis. For decades, the pathogen has been studied exclusively as an extracellular bacterium. However, recent results have revealed the pathogen's ability to invade and replicate within host cells. The present study is the first characterization of the V. parahaemolyticus' intracellular lifestyle. Upon internalization, V. parahaemolyticus is contained in a vacuole that would in the normal course of events ultimately fuse with a lysosome, degrading the vacuole's contents. The bacterium subverts this pathway, escaping into the cytoplasm prior to lysosomal fusion. Once in the cytoplasm, it replicates prolifically. Our study provides new insights into the strategies used by this globally disseminated pathogen to survive and proliferate within its host.


Assuntos
Citosol/microbiologia , Células Epiteliais/microbiologia , Vacúolos/microbiologia , Vibrio parahaemolyticus/patogenicidade , Células Epiteliais/citologia , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Fatores de Virulência/metabolismo
15.
Arch Pharm Res ; 36(6): 731-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23519647

RESUMO

Due to the severity and high prevalence of allergic diseases, there is growing interest in the development of inhibitors of such conditions. 3-Arylcoumarin derivatives emerge as promising compounds for the treatment of allergic disorders, in particular due to their close structural similarity to flavonoids, whose anti-allergic activity has been extensively reported. The aim of this work was to perform a screening of a set of 3-arylcoumarins as potential inhibitors of mast cell degranulation, a key event for the development of allergic reactions. For that purpose, it was utilized a biosensor model based on mast cells, whose in vitro assay allows for such screening, in a high throughput fashion, and also permits bringing to attention some coumarin structural features that are important for their biological activity. The mast cell-based biosensor was shown to discriminate, with high sensitivity and reproducibility, between coumarins that did not affect or caused different degrees of inhibition of degranulation. Among active coumarins, some substituents could be accounted for their inhibitory activity, such as the hydroxylation of positions 6 and 2' of 3-phenylcoumarins, in addition to catechol, amino and thiophene moieties. In summary, 3-arylcoumarins could be suggested as potential candidates for the development of new anti-allergic drugs.


Assuntos
Antialérgicos/farmacologia , Degranulação Celular/efeitos dos fármacos , Cumarínicos/farmacologia , Mastócitos/efeitos dos fármacos , Animais , Antialérgicos/química , Técnicas Biossensoriais/métodos , Linhagem Celular , Cumarínicos/química , Ensaios de Triagem em Larga Escala , Mastócitos/metabolismo , Ratos , Reprodutibilidade dos Testes , Relação Estrutura-Atividade
16.
Int Immunopharmacol ; 15(3): 532-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23399420

RESUMO

Mast cells play a critical role during the development of an allergic response. Upon activation by an antigen and IgE, via FcεRI receptors, mast cells release histamine and other mediators that initiate and propagate immediate hypersensitivity reactions. Mast cells also secrete cytokines that regulate the immune responses. In this way, inhibitors of mast cell activity could work as promising therapeutics for allergic disorders. In the present work, we investigated the capacity of pyridovericin, a natural product isolated from the entomopathogenic fungus Beauveria bassiana, to inhibit mast cell degranulation and cytokine secretion. It was found that pyridovericin strongly decreased the release of ß-hexosaminidase, a marker for mast cell degranulation, when mast cells were stimulated by both FcεRI-dependent and independent pathways. In addition, pyridovericin strongly abrogated secretion of interleukin-4. Pyridovericin-mediated suppression of stimulated increase in intracellular Ca(2+) levels, a crucial signal for mounting of both degranulation and cytokine production responses, was ascribed as one of the inhibition targets of pyridovericin. Those initial studies identify pyridovericin as a potential new candidate for the development of new anti-allergic drugs.


Assuntos
Antialérgicos/farmacologia , Beauveria/química , Hipersensibilidade/tratamento farmacológico , Mastócitos/efeitos dos fármacos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Linhagem Celular , Regulação para Baixo , Hexosaminidases/genética , Hexosaminidases/metabolismo , Hipersensibilidade/imunologia , Interleucina-4/genética , Interleucina-4/metabolismo , Mastócitos/imunologia , Terapia de Alvo Molecular , Piridonas/farmacologia , Ratos , Receptores de IgE/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA