Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(5): e1009630, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34048498

RESUMO

An intricate regulatory network controls the expression of Salmonella virulence genes. The transcriptional regulator HilD plays a central role in this network by controlling the expression of tens of genes mainly required for intestinal colonization. Accordingly, the expression/activity of HilD is highly regulated by multiple factors, such as the SirA/BarA two-component system and the Hcp-like protein HilE. SirA/BarA positively regulates translation of hilD mRNA through a regulatory cascade involving the small RNAs CsrB and CsrC, and the RNA-binding protein CsrA, whereas HilE inhibits HilD activity by protein-protein interaction. In this study, we show that SirA/BarA also positively regulates translation of hilE mRNA through the same mentioned regulatory cascade. Thus, our results reveal a paradoxical regulation exerted by SirA/BarA-Csr on HilD, which involves simultaneous opposite effects, direct positive control and indirect negative control through HilE. This kind of regulation is called an incoherent type-1 feedforward loop (I1-FFL), which is a motif present in certain regulatory networks and represents a complex biological problem to decipher. Interestingly, our results, together with those from a previous study, indicate that HilE, the repressor component of the I1-FFL reported here (I1-FFLSirA/BarA-HilE-HilD), is required to reduce the growth cost imposed by the expression of the genes regulated by HilD. Moreover, we and others found that HilE is necessary for successful intestinal colonization by Salmonella. Thus, these findings support that I1-FFLSirA/BarA-HilE-HilD cooperates to control the precise amount and activity of HilD, for an appropriate balance between the growth cost and the virulence benefit generated by the expression of the genes induced by this regulator. I1-FFLSirA/BarA-HilE-HilD represents a complex regulatory I1-FFL that involves multiple regulators acting at distinct levels of gene expression, as well as showing different connections to the rest of the regulatory network governing Salmonella virulence.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Redes Reguladoras de Genes , Infecções por Salmonella/microbiologia , Salmonella typhimurium/genética , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/patogenicidade , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Virulência , Fatores de Virulência/genética
2.
J Bacteriol ; 204(5): e0058521, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35404111

RESUMO

One important event for the divergence of Salmonella from Escherichia coli was the acquisition by horizontal transfer of the Salmonella pathogenicity island 1 (SPI-1), containing genes required for the invasion of host cells by Salmonella. HilD is an AraC-like transcriptional regulator in SPI-1 that induces the expression of the SPI-1 and many other acquired virulence genes located in other genomic regions of Salmonella. Additionally, HilD has been shown to positively control the expression of some ancestral genes (also present in E. coli and other bacteria), including phoH. In this study, we determined that both the gain of HilD and cis-regulatory evolution led to the integration of the phoH gene into the HilD regulon. Our results indicate that a HilD-binding sequence was generated in the regulatory region of the S. enterica serovar Typhimurium phoH gene, which mediates the activation of promoter 1 of this gene under SPI-1-inducing conditions. Furthermore, we found that repression by H-NS, a histone-like protein, was also adapted on the S. Typhimurium phoH gene and that HilD activates the expression of this gene in part by antagonizing H-NS. Additionally, our results revealed that the expression of the S. Typhmurium phoH gene is also activated in response to low phosphate but independently of the PhoB/R two-component system, known to regulate the E. coli phoH gene in response to low phosphate. Thus, our results indicate that cis-regulatory evolution has played a role in the expansion of the HilD regulon and illustrate the phenomenon of differential regulation of ortholog genes. IMPORTANCE Two mechanisms mediating differentiation of bacteria are well known: acquisition of genes by horizontal transfer events and mutations in coding DNA sequences. In this study, we found that the phoH ancestral gene is differentially regulated between Salmonella Typhimurium and Escherichia coli, two closely related bacterial species. Our results indicate that this differential regulation was generated by mutations in the regulatory sequence of the S. Typhimurium phoH gene and by the acquisition by S. Typhimurium of foreign DNA encoding the transcriptional regulator HilD. Thus, our results, together with those from an increasing number of studies, indicate that cis-regulatory evolution can lead to the rewiring and reprogramming of transcriptional regulation, which also plays an important role in the divergence of bacteria through time.


Assuntos
Regulação Bacteriana da Expressão Gênica , Salmonella typhimurium , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fosfatos/metabolismo , Salmonella typhimurium/metabolismo , Sorogrupo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Chemotherapy ; 61(1): 8-14, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26536333

RESUMO

BACKGROUND: Acinetobacter baumannii has emerged as a major cause of hospital-associated infections with increased morbidity and mortality among those affected. METHODS: A total of 85 isolates of a highly prevalent multidrug-resistant clone, identified during the period 2007-2011, were analyzed for biofilm formation on a polystyrene surface. The minimal inhibitory concentration was determined by the Sensititre System, the agar disk diffusion method and then read by means of the BIOMIC system and serial dilutions on Müller-Hinton agar. RESULTS: In this study, covering a period of 5 years (2007-2011), we demonstrate that a particular clone emerged as the most prevalent, with an associated lethality of 28.2%. We demonstrate that 92.9% of strains corresponding to this clone are biofilm producers. Our results also demonstrate that all isolates were 100% susceptible to polymyxin B. CONCLUSION: Our study suggests that the high prevalence and lethality of this multidrug-resistant clone of A. baumannii and its persistence over close to 5 years in a Mexican tertiary hospital environment can be explained in part by the ability of these clinical isolates of A. baumannii to form biofilms.


Assuntos
Infecções por Acinetobacter/epidemiologia , Acinetobacter baumannii/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Infecção Hospitalar/epidemiologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Polimixina B/farmacologia , Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/isolamento & purificação , Antibacterianos/farmacologia , Infecção Hospitalar/microbiologia , Humanos , México/epidemiologia , Testes de Sensibilidade Microbiana , Centros de Atenção Terciária
4.
J Immunol ; 190(3): 1201-9, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23284055

RESUMO

Salmonella infects and survives within B cells, but the mechanism used by the bacterium to promote its survival in these cells is unknown. In macrophages, flagellin secreted by Salmonella activates the Nod-like receptor (NLR) family CARD domain containing protein 4 (NLRC4) inflammasome, leading to the production of IL-1ß and pyroptosis of infected cells. In this study, we demonstrated that the NLRC4 inflammasome is functional in B cells; however, in Salmonella-infected B cells, IL-1ß secretion is prevented through the downregulation of NLRC4 expression. A functional Salmonella pathogenicity island 1 type III secretion system appears to be required for this process. Furthermore, infection induces Yap phosphorylation and promotes the interaction of Yap with Hck, thus preventing the transcriptional activation of NLRC4. The ability of Salmonella to inhibit IL-1ß production also prevents B cell death; thus, B cells represent an ideal niche in which Salmonella resides, thereby promoting its persistence and dissemination.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Apoptose/imunologia , Linfócitos B/microbiologia , Proteínas de Ligação ao Cálcio/biossíntese , Regulação para Baixo , Regulação da Expressão Gênica , Evasão da Resposta Imune/genética , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Salmonelose Animal/imunologia , Salmonella typhimurium/fisiologia , Transcrição Gênica , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular , Citocinas/metabolismo , Citotoxicidade Imunológica , Feminino , Flagelina/farmacologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-hck/metabolismo , Salmonelose Animal/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Virulência , Proteínas de Sinalização YAP
5.
mSphere ; 9(5): e0006024, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38647313

RESUMO

Enterobacter cloacae is an emerging pathogen isolated in healthcare-associated infections. A major virulence factor of this bacterium is the type VI secretion system (T6SS). The genome of E. cloacae harbors two T6SS gene clusters (T6SS-1 and T6SS-2), and the functional characterization of both systems showed that these two T6SSs are not expressed under the same conditions. Here, we report that the major histone-like protein HU positively regulates the expression of both T6SSs and, therefore, the function that each T6SS exerts in E. cloacae. Single deletions of the genes encoding the HU subunits (hupA and hupB) decreased mRNA levels of both T6SS. In contrast, the hupA hupB double mutant dramatically affected the T6SS expression, diminishing its transcription. The direct binding of HU to the promoter regions of T6SS-1 and T6SS-2 was confirmed by electrophoretic mobility shift assay. In addition, single and double mutations in the hup genes affected the ability of inter-bacterial killing, biofilm formation, adherence to epithelial cells, and intestinal colonization, but these phenotypes were restored when such mutants were trans-complemented. Our data broaden our understanding of the regulation of HU-mediated T6SS in these pathogenic bacteria. IMPORTANCE: T6SS is a nanomachine that functions as a weapon of bacterial destruction crucial for successful colonization in a specific niche. Enterobacter cloacae expresses two T6SSs required for bacterial competition, adherence, biofilm formation, and intestinal colonization. Expression of T6SS genes in pathogenic bacteria is controlled by multiple regulatory systems, including two-component systems, global regulators, and nucleoid proteins. Here, we reported that the HU nucleoid protein directly activates both T6SSs in E. cloacae, affecting the T6SS-related phenotypes. Our data describe HU as a new regulator involved in the transcriptional regulation of T6SS and its impact on E. cloacae pathogenesis.


Assuntos
Proteínas de Bactérias , Proteínas de Ligação a DNA , Enterobacter cloacae , Regulação Bacteriana da Expressão Gênica , Sistemas de Secreção Tipo VI , Enterobacter cloacae/genética , Enterobacter cloacae/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regiões Promotoras Genéticas , Família Multigênica
6.
Cell Microbiol ; 14(2): 255-73, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22023353

RESUMO

Burkholderia cenocepacia is a Gram-negative opportunistic pathogen of patients with cystic fibrosis and chronic granulomatous disease. The bacterium survives intracellularly in macrophages within a membrane-bound vacuole (BcCV) that precludes the fusion with lysosomes. The underlying cellular mechanisms and bacterial molecules mediating these phenotypes are unknown. Here, we show that intracellular B. cenocepacia expressing a type VI secretion system (T6SS) affects the activation of the Rac1 and Cdc42 RhoGTPase by reducing the cellular pool of GTP-bound Rac1 and Cdc42. The T6SS also increases the cellular pool of GTP-bound RhoA and decreases cofilin activity. These effects lead to abnormal actin polymerization causing collapse of lamellipodia and failure to retract the uropod. The T6SS also prevents the recruitment of soluble subunits of the NADPH oxidase complex including Rac1 to the BcCV membrane, but is not involved in the BcCV maturation arrest. Therefore, T6SS-mediated deregulation of Rho family GTPases is a common mechanism linking disruption of the actin cytoskeleton and delayed NADPH oxidase activation in macrophages infected with B. cenocepacia.


Assuntos
Citoesqueleto de Actina/metabolismo , Burkholderia cenocepacia/patogenicidade , Macrófagos/microbiologia , NADPH Oxidases/antagonistas & inibidores , Neuropeptídeos/antagonistas & inibidores , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac de Ligação ao GTP/antagonistas & inibidores , Animais , Linhagem Celular , Guanosina Trifosfato/metabolismo , Macrófagos/enzimologia , Macrófagos/metabolismo , Camundongos , Modelos Biológicos , Proteínas rac1 de Ligação ao GTP , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Proc Natl Acad Sci U S A ; 107(9): 4353-8, 2010 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-20176950

RESUMO

Salmonella enterica serovar Typhi can colonize the gallbladder and persist in an asymptomatic carrier state that is frequently associated with the presence of gallstones. We have shown that salmonellae form bile-mediated biofilms on human gallstones and cholesterol-coated surfaces in vitro. Here, we test the hypothesis that biofilms on cholesterol gallbladder stones facilitate typhoid carriage in mice and men. Naturally resistant (Nramp1(+/+)) mice fed a lithogenic diet developed cholesterol gallstones that supported biofilm formation during persistent serovar Typhimurium infection and, as a result, demonstrated enhanced fecal shedding and enhanced colonization of gallbladder tissue and bile. In typhoid endemic Mexico City, 5% of enrolled cholelithiasis patients carried serovar Typhi, and bacterial biofilms could be visualized on gallstones from these carriers whereas significant biofilms were not detected on gallstones from Escherichia coli infected gallbladders. These findings offer direct evidence that gallstone biofilms occur in humans and mice, which facilitate gallbladder colonization and shedding.


Assuntos
Cálculos Biliares/microbiologia , Salmonella typhimurium/crescimento & desenvolvimento , Animais , Biofilmes , Portador Sadio , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/fisiologia , Colesterol/metabolismo , Fezes/microbiologia , Cálculos Biliares/metabolismo , Humanos , Camundongos , Microscopia Eletrônica de Varredura
8.
Diagn Microbiol Infect Dis ; 105(2): 115866, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36525921

RESUMO

Burkholderia cepacia complex (Bcc) species are opportunistic pathogens widely distributed in the environment and often infect people with cystic fibrosis (CF). This study aims to determine which genomovars of the Bcc can cause infections in non-CF patients from a tertiary care hospital in Mexico and if they carry virulence factors that could increase their pathogenicity. We identified 23 clinical isolates that carry the recA gene. Twenty-two of them belongs to the genomovar V (B. vietnamiensis) and one to the genomovar II (B. multivorans). Thirteen pulsotypes were identified among 22 B. vietnamiensis isolates. All clinical isolates produced biofilm were motile and cytotoxic on murine macrophage-like RAW264.7 and in A549 human lung epithelial cells. In conclusion, B. vietnamiensis causes infections in non-CF patients in a tertiary care hospital in Mexico, rapid identification of this pathogen can help physicians to establish a better antimicrobial treatment.


Assuntos
Infecções por Burkholderia , Complexo Burkholderia cepacia , Burkholderia cepacia , Fibrose Cística , Humanos , Animais , Camundongos , Burkholderia cepacia/genética , Infecções por Burkholderia/epidemiologia , México/epidemiologia , Centros de Atenção Terciária , Reação em Cadeia da Polimerase , Complexo Burkholderia cepacia/genética , Fibrose Cística/complicações
9.
Microbiology (Reading) ; 158(Pt 9): 2315-2324, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22767545

RESUMO

Burkholderia cenocepacia is a member of the Burkholderia cepacia complex (Bcc), a group of Gram-negative opportunistic pathogens that cause severe lung infections in patients with cystic fibrosis and display extreme intrinsic resistance to antibiotics, including antimicrobial peptides. B. cenocepacia BCAL2157 encodes a protein homologous to SuhB, an inositol-1-monophosphatase from Escherichia coli, which was suggested to participate in post-transcriptional control of gene expression. In this work we show that a deletion of the suhB-like gene in B. cenocepacia (ΔsuhB(Bc)) was associated with pleiotropic phenotypes. The ΔsuhB(Bc) mutant had a growth defect manifested by an almost twofold increase in the generation time relative to the parental strain. The mutant also had a general defect in protein secretion, motility and biofilm formation. Further analysis of the type II and type VI secretion systems (T2SS and T6SS) activities revealed that these secretion systems were inactive in the ΔsuhB(Bc) mutant. In addition, the mutant exhibited increased susceptibility to polymyxin B but not to aminoglycosides such as gentamicin and kanamycin. Together, our results demonstrate that suhB(Bc) deletion compromises general protein secretion, including the activity of the T2SS and the T6SS, and affects polymyxin B resistance, motility and biofilm formation. The pleiotropic effects observed upon suhB(Bc) deletion demonstrate that suhB(Bc) plays a critical role in the physiology of B. cenocepacia.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Burkholderia cenocepacia/genética , Farmacorresistência Bacteriana , Locomoção , Polimixina B/farmacologia , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos , Burkholderia cenocepacia/efeitos dos fármacos , Burkholderia cenocepacia/metabolismo , Burkholderia cenocepacia/fisiologia , Deleção de Genes , Humanos
10.
Microb Pathog ; 52(6): 367-74, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22475626

RESUMO

We have previously reported that Salmonella infects B cells and survives within endosomal-lysosomal compartments. However, the mechanisms used by Salmonella to enter B cells remain unknown. In this study, we have shown that Salmonella induces its own entry by the induction of localized ruffling, macropinocytosis, and spacious phagosome formation. These events were associated with the rearrangement of actin and microtubule networks. The Salmonella pathogenesis island 1 (SPI-1) was necessary to invade B cells. In contrast to macrophages, B cells were highly resistant to cell death induced by Salmonella. These data demonstrate the ability of Salmonella to infect these non-professional phagocytic cells, where the bacterium can find an ideal intracellular niche to support persistence and the possible dissemination of infection.


Assuntos
Linfócitos B/microbiologia , Linfócitos B/fisiologia , Interações Hospedeiro-Patógeno , Fagossomos/microbiologia , Pinocitose , Salmonella/patogenicidade , Actinas/metabolismo , Animais , Proteínas de Bactérias , Células Cultivadas , Feminino , Macrófagos/microbiologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Microtúbulos/metabolismo , Salmonella/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
11.
Arch Med Res ; 53(6): 641-642, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36123225

RESUMO

Pseudomonas aeruginosa is a significant cause of lung infections in patients with cystic fibrosis (CF). Pseudomonas produces a chronic infection that increases the morbidity and mortality in affected individuals. The rapid identification of Pseudomonas in these individuals enables conventional antimicrobial treatment to be started. However, over the years, treatment of P. aeruginosa has become problematic and very challenging due to their intrinsic and acquired antibiotic resistance. Microbiology plays an essential role in determining the antimicrobial susceptibility/resistance profiles of isolated strains, helping to optimize antimicrobial treatment for affected patients. In addition to the conventional susceptibility analysis, whole genome sequencing has emerged as a powerful tool for determining specific genomic variants, both in specific geographic areas and globally. Thus, molecular epidemiologic surveillance could help to establish a better treatment strategy and counter the spread of high-risk, P. aeruginosa variants among CF individuals.


Assuntos
Fibrose Cística , Infecções por Pseudomonas , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Criança , Fibrose Cística/complicações , Fibrose Cística/genética , Genômica , Humanos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética
12.
Viruses ; 14(4)2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35458383

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent responsible for the coronavirus disease 2019 (COVID-19). The high rate of mutation of this virus is associated with a quick emergence of new viral variants that have been rapidly spreading worldwide. Several mutations have been documented in the receptor-binding domain (RBD) of the viral spike protein that increases the interaction between SARS-CoV-2 and its cellular receptor, the angiotensin-converting enzyme 2 (ACE2). Mutations in the spike can increase the viral spread rate, disease severity, and the ability of the virus to evade either the immune protective responses, monoclonal antibody treatments, or the efficacy of current licensed vaccines. This review aimed to highlight the functional virus classification used by the World Health Organization (WHO), Phylogenetic Assignment of Named Global Outbreak (PANGO), Global Initiative on Sharing All Influenza Data (GISAID), and Nextstrain, an open-source project to harness the scientific and public health potential of pathogen genome data, the chronological emergence of viral variants of concern (VOCs) and variants of interest (VOIs), the major findings related to the rate of spread, and the mutations in the spike protein that are involved in the evasion of the host immune responses elicited by prior SARS-CoV-2 infections and by the protection induced by vaccination.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Mutação , Filogenia , Ligação Proteica , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus
13.
Bol Med Hosp Infant Mex ; 79(4): 215-221, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36100204

RESUMO

Cystic fibrosis is an autosomal recessive inherited disease caused by mutations in the cystic fibrosis transmembrane conductance regulator gene (CFTR). CFTR is a protein that transports ions across the membrane of lung epithelial cells. Loss of its function leads to the production of thick sticky mucus, where various bacterial pathogens can establish and adapt, contributing to the gradual loss of lung function. In this review, evidence of the molecular mechanisms used by Pseudomonas aeruginosa and Burkholderia cenocepacia to survive and persist in the pulmonary environment will be provided. Additionally, new therapeutic strategies based on CFTR function modulators will be described.


La fibrosis quística es una enfermedad hereditaria autosómica recesiva que se origina por mutaciones en el gen regulador de conductancia transmembranal de la fibrosis quística (CFTR, cystic fibrosis transmembrane conductance regulator). El CFTR es una proteína que transporta iones a través de la membrana de las células epiteliales pulmonares. La pérdida de su función conlleva la producción de un moco pegajoso y espeso, donde se pueden establecer y adaptar diversos patógenos bacterianos que contribuyen a la pérdida gradual de la función pulmonar. En este artículo de revisión se dará evidencia de los mecanismos moleculares que utilizan Pseudomonas aeruginosa y Burkholderia cenocepacia para sobrevivir y persistir en el ambiente pulmonar. Adicionalmente, se describirán las nuevas estrategias de terapia a base de moduladores de la función del CFTR.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística , Fibrose Cística , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/uso terapêutico , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Fibrose , Humanos , Pseudomonas aeruginosa
14.
Microbiol Spectr ; 10(5): e0271022, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36073960

RESUMO

The acquisition of Salmonella pathogenicity island 2 (SPI-2) conferred on Salmonella the ability to survive and replicate within host cells. The ssrAB bicistronic operon, located in SPI-2, encodes the SsrAB two-component system (TCS), which is the central positive regulator that induces the expression of SPI-2 genes as well as other genes located outside this island. On the other hand, CpxRA is a two-component system that regulates expression of virulence genes in many bacteria in response to different stimuli that perturb the cell envelope. We previously reported that the CpxRA system represses the expression of SPI-1 and SPI-2 genes under SPI-1-inducing conditions by decreasing the stability of the SPI-1 regulator HilD. Here, we show that under SPI-2-inducing conditions, which mimic the intracellular environment, CpxRA represses the expression of SPI-2 genes by the direct action of phosphorylated CpxR (CpxR-P) on the ssrAB regulatory operon. CpxR-P recognized two sites located proximal and distal from the promoter located upstream of ssrA. Consistently, we found that CpxRA reduces the replication of Salmonella enterica serovar Typhimurium inside murine macrophages. Therefore, our results reveal CpxRA as an additional regulator involved in the intracellular lifestyle of Salmonella, which in turn adds a new layer to the intricate regulatory network controlling the expression of Salmonella virulence genes. IMPORTANCE SPI-2 encodes a type III secretion system (T3SS) that is a hallmark for the species Salmonella enterica, which is essential for the survival and replication within macrophages. Expression of SPI-2 genes is positively controlled by the two-component system SsrAB. Here, we determined a regulatory mechanism involved in controlling the overgrowth of Salmonella inside macrophages. In this mechanism, CpxRA, a two-component system that is activated by extracytoplasmic stress, directly represses expression of the ssrAB regulatory operon; as a consequence, expression of SsrAB target genes is decreased. Our findings reveal a novel mechanism involved in the intracellular lifestyle of Salmonella, which is expected to sense perturbations in the bacterial envelope that Salmonella faces inside host cells, as the synthesis of the T3SS-2 itself.


Assuntos
Regulação Bacteriana da Expressão Gênica , Ilhas Genômicas , Camundongos , Animais , Sistemas de Secreção Tipo III/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Óperon , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
15.
Bol Med Hosp Infant Mex ; 79(2): 115-122, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35073628

RESUMO

BACKGROUND: Cystic fibrosis (CF) is a genetic disease in which thick, sticky mucus is produced in the lungs (and other organs) that impairs ciliary clearance, leading to respiratory problems, increased chronic bacterial infections, and decreased lung function. Staphylococcus aureus is one of the primary bacterial pathogens colonizing the lungs of CF patients. This study aimed to characterize the genetic relatedness of S. aureus, its presence in children with CF, and its cytotoxic activity in THP1 cell-derived macrophages (THP1m). METHODS: Genetic relatedness of S. aureus isolates from a cohort of 50 children with CF was determined by pulsed-field gel electrophoresis (PFGE). The VITEK 2 automated system was used to determine antimicrobial susceptibility, and methicillin-resistance S. aureus (MRSA) was determined by diffusion testing using cefoxitin disk. The presence of mecA and lukPV genes was determined by the polymerase chain reaction and cytotoxic activity of S.aureus on THP1m by CytoTox 96® assay. RESULTS: From 51 S. aureus isolates from 50 children with CF, we identified 34pulsotypes by PFGE. Of the 50 children, 12 (24%) were colonized by more than one pulsotype, and 5/34 identified pulsotypes(14.7%) were shared between unrelated children. In addition, 3/34 pulsotypes (8.8%) were multidrug-resistant (MDR), and2/34 (5.9%) were MRSA. Notably, 30/34 pulsotypes (88.2%) exhibited cytotoxicity on THP1m cells and 14/34 (41.2%) alteredTHP1m monolayers. No isolate carried the lukPV gene. CONCLUSIONS: Although a low frequency of MRSA and MDR wasfound among clinical isolates, most of the S. aureus pulsotypes identified were cytotoxic on THP1m.


INTRODUCCIÓN: La fibrosis quística (FQ) es una enfermedad genética en la que se produce moco espeso y pegajoso en los pulmones (y otros órganos), lo que conduce a problemas respiratorios, incremento de las infecciones bacterianas crónicas y disminución de la función pulmonar. Staphylococcus aureus es uno de los principales patógenos que colonizan los pul-mones de los pacientes con FQ. El objetivo de este trabajo fue caracterizar la relación genética de S. aureus, su presencia en niños con FQ y su actividad citotóxica en macrófagos derivados de células THP1 (THP1m). MÉTODOS: La relación gené-tica de los aislados de S. aureus provenientes de una cohorte de 50 pacientes con FQ fue determinada por electroforesis en gel de campo pulsado (PFGE). La sensibilidad a los antimicrobianos se determinó mediante el sistema automatizado VITEK 2, y la resistencia a la meticilina (SARM) mediante la prueba de difusión utilizando discos de cefoxitina. La presen-cia de los genes mecA y lukPV se determinó mediante reacción en cadena de la polimerasa, y la actividad citotóxica de S. aureus sobre células THP1m mediante el ensayo CytoTox96®. RESULTADOS: A partir de 51 aislados de S. aureus provenientes de 50 niños con FQ se identificaron 34 pulsotipos por PFGE. De los 50 niños, 12 (24%) estaban colonizados por más de un pulsotipo y 5 de los 34 pulsotipos (14.7%) los compartían niños que no estaban relacionados. De los 34 pulsotipos, 3 (8.8%) presentaron multirresistencia (MDR) y 2 (5.9%) fueron SARM. Además, 30 pulsotipos (88.2%) fueron citotóxicos sobre células THP1m y 14 (41.2%) alteraron su monocapa. Ninguno de los pulsotipos presentó el gen lukPV. CONCLUSIONES: Aunque se encontró una baja frecuencia de SARM y MDR en los aislados, la mayoría de los pulsotipos de S. aureus identificados fueron citotóxicos para células THP1m.


Assuntos
Fibrose Cística , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Antibacterianos/farmacologia , Criança , Humanos , Staphylococcus aureus Resistente à Meticilina/genética , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética
16.
Arch Med Res ; 52(4): 357-361, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33309309

RESUMO

Cystic fibrosis (CF) is a progressive autosomal recessive genetic disease that principally affects the respiratory and digestive systems. It is a chronic disease that has no cure. Symptoms often include chronic cough, lung infections, and shortness of breath. Children with cystic fibrosis present failure to thrive as manifested by low weight and height for age. CF is caused by mutations in the cystic fibrosis transmembrane conductance regulator (cftr) gene that codes for a cell membrane protein of epithelial tissues and affects multiple organ systems in the human body. Mutations on the CFTR causes dysfunctional electrolyte regulation affecting intracellular water content. Defective CFTR function in airways produce a dehydrated and sticky mucus that leads the establishment of bacterial chronic infection that ultimate decrease the lung function. During the first decade of life, affected individuals are colonized principally by non typable Haemophilus influenzae and Staphylococcus aureus. During the second decade, Pseudomonas aeruginosa becomes the most dominant pathogen and persists throughout the remainder of their lives. In this work, we describe the mechanisms used by P. aeruginosa to adapt and persist in lungs of individuals with cystic fibrosis.


Assuntos
Fibrose Cística , Infecções por Pseudomonas , Pseudomonas aeruginosa , Fibrose Cística/genética , Humanos , Pulmão
17.
Front Cell Infect Microbiol ; 11: 760922, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34692569

RESUMO

Cystic fibrosis (CF) is a genetic disease affecting more than 70,000 people worldwide. It is caused by a mutation in the cftr gene, a chloride ion transporter localized in the plasma membrane of lung epithelial cells and other organs. The loss of CFTR function alters chloride, bicarbonate, and water transport through the plasma membrane, promoting the production of a thick and sticky mucus in which bacteria including Pseudomonas aeruginosa and Burkholderia cenocepacia can produce chronic infections that eventually decrease the lung function and increase the risk of mortality. Autophagy is a well-conserved lysosomal degradation pathway that mediates pathogen clearance and plays an important role in the control of bacterial infections. In this mini-review, we describe the principal strategies used by P. aeruginosa and B. cenocepacia to survive and avoid microbicidal mechanisms within the autophagic pathway leading to the establishment of chronic inflammatory immune responses that gradually compromise the lung function and the life of CF patients.


Assuntos
Burkholderia cenocepacia , Fibrose Cística , Infecções por Pseudomonas , Autofagia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Humanos , Pseudomonas aeruginosa
18.
Sci Rep ; 11(1): 20946, 2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34686741

RESUMO

Cell spreading and phagocytosis are notably regulated by small GTPases and GAP proteins. TBC1D10C is a dual inhibitory protein with GAP activity. In immune cells, TBC1D10C is one of the elements regulating lymphocyte activation. However, its specific role in macrophages remains unknown. Here, we show that TBC1D10C engages in functions dependent on the cytoskeleton and plasma membrane reorganization. Using ex vivo and in vitro assays, we found that elimination and overexpression of TBC1D10C modified the cytoskeletal architecture of macrophages by decreasing and increasing the spreading ability of these cells, respectively. In addition, TBC1D10C overexpression contributed to higher phagocytic activity against Burkholderia cenocepacia and to increased cell membrane tension. Furthermore, by performing in vitro and in silico analyses, we identified 27 TBC1D10C-interacting proteins, some of which were functionally classified as protein complexes involved in cytoskeletal dynamics. Interestingly, we identified one unreported TBC1D10C-intrinsically disordered region (IDR) with biological potential at the cytoskeleton level. Our results demonstrate that TBC1D10C shapes macrophage activity by inducing reorganization of the cytoskeleton-plasma membrane in cell spreading and phagocytosis. We anticipate our results will be the basis for further studies focused on TBC1D10C. For example, the specific molecular mechanism in Burkholderia cenocepacia phagocytosis and functional analysis of TBC1D10C-IDR are needed to further understand its role in health and disease.


Assuntos
Citoesqueleto/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiologia , Fagocitose/fisiologia , Animais , Burkholderia cenocepacia/patogenicidade , Membrana Celular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas rac1 de Ligação ao GTP/metabolismo
19.
Diagn Microbiol Infect Dis ; 101(1): 115454, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34271235

RESUMO

The emergence of high-risk clones of priority pathogens exhibiting convergence of antimicrobial resistance and virulence is a critical issue worldwide. In a previous study, an extensively drug-resistant Pseudomonas aeruginosa was isolated from a chronically colonized pediatric patient with cystic fibrosis (CF). In this study, we analyzed genomic data of this strain (CF023-Psa42), extracting clinically and epidemiologically relevant information (i.e., the antimicrobial resistome, virulome, and sequence type). In this regard, we report the emergence of GES-19 (extended-spectrum ß-lactamase)-producing P. aeruginosa with genotype exoU+. The CF023-Psa42 strain exhibited a broad resistome, belonging to the international high-risk clone sequence type ST235. The blaGES-19 gene was located on a class 1 integron, along to aac(6')-33, aac(6')-Ib-cr, blaOXA-2, aadA1, sul1, and qacEΔ1 resistance genes. Relevant virulence genes such as lasA (proteolysis and elastolysis), toxA (exotoxin A), alg (alginate biosynthesis operon), and exoU (toxin of type III secretion systems) were predicted. Our findings reveal the convergence of broad resistome and virulome in P. aeruginosa ST235. Genomic surveillance is essential to monitor the emergence and dissemination of priority pathogens with epidemiological success.


Assuntos
Fibrose Cística/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/isolamento & purificação , beta-Lactamases/metabolismo , Antibacterianos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Criança , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/genética , Feminino , Genoma Bacteriano/genética , Genótipo , Humanos , Integrons/genética , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Virulência/genética , beta-Lactamases/genética
20.
Front Microbiol ; 12: 711577, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489901

RESUMO

Klebsiella pneumoniae is recognized as a common cause of nosocomial infections and outbreaks causing pneumonia, septicemia, and urinary tract infections. This opportunistic bacterium shows an increasing acquisition of antibiotic-resistance genes, which complicates treatment of infections. Hence, fast reliable strain typing methods are paramount for the study of this opportunistic pathogen's multi-drug resistance genetic profiles. In this study, thirty-eight strains of K. pneumoniae isolated from the blood of pediatric patients were characterized by whole-genome sequencing and genomic clustering methods. Genes encoding ß-lactamase were found in all the bacterial isolates, among which the bla SHV variant was the most prevalent (53%). Moreover, genes encoding virulence factors such as fimbriae, capsule, outer membrane proteins, T4SS and siderophores were investigated. Additionally, a multi-locus sequence typing (MLST) analysis revealed 24 distinct sequence types identified within the isolates, among which the most frequently represented were ST76 (16%) and ST70 (11%). Based on LPS structure, serotypes O1 and O3 were the most prevalent, accounting for approximately 63% of all infections. The virulence capsular types K10, K136, and K2 were present in 16, 13, and 8% of the isolates, respectively. Phylogenomic analysis based on virtual genome fingerprints correlated with the MLST data. The phylogenomic reconstruction also denoted association between strains with a higher abundance of virulence genes and virulent serotypes compared to strains that do not possess these traits. This study highlights the value of whole-genomic sequencing in the surveillance of virulence attributes among clinical K. pneumoniae strains.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA