Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 20(8): e1012495, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39178317

RESUMEN

There is a critical gap in knowledge about how Gram-negative bacterial pathogens, using survival strategies developed for other niches, cause lethal bacteremia. Facultative anaerobic species of the Enterobacterales order are the most common cause of Gram-negative bacteremia, including Escherichia coli, Klebsiella pneumoniae, Serratia marcescens, Citrobacter freundii, and Enterobacter hormaechei. Bacteremia often leads to sepsis, a life-threatening organ dysfunction resulting from unregulated immune responses to infection. Despite a lack of specialization for this host environment, Gram-negative pathogens cause nearly half of bacteremia cases annually. Based on our existing Tn-Seq fitness factor data from a murine model of bacteremia combined with comparative genomics of the five Enterobacterales species above, we prioritized 18 conserved fitness genes or operons for further characterization. Mutants were constructed for all genes in all five species. Each mutant was used to cochallenge C57BL/6 mice via tail vein injection along with each respective wild-type strain to determine competitive indices for each fitness gene. Five fitness factor genes, when mutated, attenuated mutants in four or five species in the spleen and liver (tatC, ruvA, gmhB, wzxE, arcA). Five additional fitness factor genes or operons were validated as outcompeted by wild-type in three, four, or five bacterial species in the spleen (xerC, prc, apaGH, atpG, aroC). Overall, 17 of 18 fitness factor mutants were attenuated in at least one species in the spleen or liver. Together, these findings allow for the development of a model of bacteremia pathogenesis that may include future targets of therapy against bloodstream infections.


Asunto(s)
Bacteriemia , Genoma Bacteriano , Animales , Bacteriemia/microbiología , Ratones , Ratones Endogámicos C57BL , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Enterobacteriaceae/genética , Enterobacteriaceae/patogenicidad , Proteínas Bacterianas/genética , Femenino , Modelos Animales de Enfermedad
2.
mBio ; 15(8): e0072124, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38958446

RESUMEN

Vibrio cholerae is a Gram-negative gastrointestinal pathogen responsible for the diarrheal disease cholera. Expression of key virulence factors, cholera toxin and toxin-coregulated pilus, is regulated directly by ToxT and indirectly by two transmembrane transcription regulators (TTRs), ToxR and TcpP, that promote the expression of toxT. TcpP abundance and activity are controlled by TcpH, a single-pass transmembrane protein, which protects TcpP from a two-step proteolytic process known as regulated intramembrane proteolysis (RIP). The mechanism of TcpH-mediated protection of TcpP represents a major gap in our understanding of V. cholerae pathogenesis. The absence of tcpH leads to unimpeded degradation of TcpP in vitro and a colonization defect in a neonate mouse model of V. cholerae colonization. Here, we show that TcpH protects TcpP from RIP via direct interaction. We also demonstrate that α-linolenic acid, a dietary fatty acid, promotes TcpH-dependent inhibition of RIP via co-association of TcpP and TcpH molecules within detergent-resistant membranes (DRMs) in a mechanism requiring the TcpH transmembrane domain. Taken together, our data support a model where V. cholerae cells use exogenous α-linolenic acid to remodel the phospholipid bilayer in vivo, leading to co-association of TcpP and TcpH within DRMs where RIP of TcpP is inhibited by TcpH, thereby promoting V. cholerae pathogenicity. IMPORTANCE: Vibrio cholerae continues to pose a significant global burden on health and an alternative therapeutic approach is needed, due to evolving multidrug resistance strains. Transcription of toxT, stimulated by TcpP and ToxR, is essential for V. cholerae pathogenesis. Our results show that TcpP, one of the major regulators of toxT gene expression, is protected from proteolysis by TcpH, via direct interaction. Furthermore, we identified a gut metabolite, α-linolenic acid, that stimulates the co-association of TcpP and TcpH within detergent-resistant membranes (also known as lipid-ordered membrane domains), thereby supporting TcpH-dependent antagonism of TcpP proteolysis. Data presented here extend our knowledge of RIP, virulence gene regulation in V. cholerae, and, to the best of our knowledge, provides the first evidence that lipid-ordered membranes exist within V. cholerae. The model presented here also suggests that TTRs, common among bacteria and archaea, and co-component signal transduction systems present in Enterobacteria, could also be influenced similarly.


Asunto(s)
Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica , Proteolisis , Factores de Transcripción , Vibrio cholerae , Factores de Virulencia , Vibrio cholerae/genética , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidad , Vibrio cholerae/efectos de los fármacos , Animales , Ratones , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Virulencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Cólera/microbiología
3.
Proc Natl Acad Sci U S A ; 121(2): e2316540120, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38170751

RESUMEN

How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using 6-wk-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni, ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within 2 to 3 d of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP, which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter (lctP) led to identification of a putative thiol-based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides better insights into the pathogenicity of C. jejuni.


Asunto(s)
Infecciones por Campylobacter , Campylobacter jejuni , Animales , Humanos , Ratones , Ácido Láctico/metabolismo , Campylobacter jejuni/genética , Hurones , Transportadores de Ácidos Monocarboxílicos
4.
bioRxiv ; 2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37873437

RESUMEN

How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using six-week-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni , ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within two-three days of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP , which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter ( lctP ) led to discovery of a putative thiol based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides new insights into the pathogenicity of C. jejuni . Significance: There is a gap in knowledge about the mechanisms by which C. jejuni populations expand during infection. Using an animal model which accurately reflects human infection without the need to alter the host microbiome or the immune system prior to infection, we explored pathophysiological alterations of the gut after C. jejuni infection. Our study identified the gut metabolite L-lactate as playing an important role as a growth substrate for C. jejuni during acute infection. We identified a DNA binding protein, LctR, that binds to the lctP promoter and may repress lctP expression, resulting in decreased lactate transport under low oxygen levels. This work provides new insights about C. jejuni pathogenicity.

5.
J Bacteriol ; 202(7)2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-31932316

RESUMEN

Campylobacter jejuni causes acute gastroenteritis worldwide and is transmitted primarily through poultry, in which it is often a commensal member of the intestinal microbiota. Previous transcriptome sequencing (RNA-Seq) experiment showed that transcripts from an operon encoding a high-affinity phosphate transporter (PstSCAB) of C. jejuni were among the most abundant when the bacterium was grown in chickens. Elevated levels of the pstSCAB mRNA were also identified in an RNA-Seq experiment from human infection studies. In this study, we explore the role of PstSCAB in the biology and colonization potential of C. jejuni Our results demonstrate that cells lacking PstSCAB survive poorly in stationary phase, in nutrient-limiting media, and under osmotic conditions reflective of those in the chicken. Polyphosphate levels in the mutant cells were elevated at stationary phase, consistent with alterations in expression of polyphosphate metabolism genes. The mutant strain was highly attenuated for colonization of newly hatched chicks, with levels of bacteria at several orders of magnitude below wild-type levels. Mutant and wild type grew similarly in complex media, but the pstS::kan mutant exhibited a significant growth defect in minimal medium supplemented with l-lactate, postulated as a carbon source in vivo Poor growth in lactate correlated with diminished expression of acetogenesis pathway genes previously demonstrated as important for colonizing chickens. The phosphate transport system is thus essential for diverse aspects of C. jejuni physiology and in vivo fitness and survival.IMPORTANCECampylobacter jejuni causes millions of human gastrointestinal infections annually, with poultry a major source of infection. Due to the emergence of multidrug resistance in C. jejuni, there is need to identify alternative ways to control this pathogen. Genes encoding the high-affinity phosphate transporter PstSCAB are highly expressed by C. jejuni in chickens and humans. In this study, we address the role of PstSCAB on chicken colonization and other C. jejuni phenotypes. PstSCAB is required for colonization in chicken, metabolism and survival under different stress responses, and during growth on lactate, a potential growth substrate in chickens. Our study highlights that PstSCAB may be an effective target to develop mechanisms for controlling bacterial burden in both chicken and human.


Asunto(s)
Infecciones por Campylobacter/veterinaria , Campylobacter jejuni/fisiología , Pollos/microbiología , Ácido Láctico/metabolismo , Proteínas de Transporte de Fosfato/genética , Enfermedades de las Aves de Corral/microbiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Metabolómica/métodos , Mutación , Proteínas de Transporte de Fosfato/metabolismo , Fosfatos/metabolismo , Estrés Fisiológico
6.
Microb Pathog ; 140: 103927, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31846743

RESUMEN

The use of traditional foods and beverages or their bioactive compounds as anti-virulence agents is a new alternative method to overcome the increased global emergence of antimicrobial resistance in enteric pathogens. In the present study, we investigated the anti-virulence activity of a polyphenolic fraction previously isolated from Kombucha, a 14-day fermented beverage of sugared black tea, against Vibrio cholerae O1. The isolated fraction was mainly composed of the polyphenols catechin and isorhamnetin. The fraction, the individual polyphenols and the combination of the individual polyphenols significantly inhibited bacterial swarming motility and expression of flagellar regulatory genes motY and flaC, even at sub-inhibitory concentrations. The polyphenolic compounds also decreased bacterial protease secretion and mucin penetration in vitro. In vivo study revealed that the polyphenolic fraction significantly inhibited V. cholerae induced fluid accumulation in the rabbit ileal loop model and intestinal colonization in suckling mice model. Therefore, the anti-virulence activity of the Kombucha polyphenolic fraction involved inhibition of motility and protease secretion of V. cholerae, thus preventing bacterial penetration through the mucin layer as well as fluid accumulation and bacterial colonization in the intestinal epithelial cells. The overall results implied that Kombucha might be considered as a potential alternative source of anti-virulence polyphenols against V. cholerae. To the best of our knowledge, this is the first report on the anti-virulence activity of Kombucha, mostly attributed to its polyphenolic content.


Asunto(s)
Té de Kombucha , Polifenoles/farmacología , Vibrio cholerae/efectos de los fármacos , Animales , Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/efectos de los fármacos , Catequina/farmacología , Movimiento Celular/efectos de los fármacos , Cólera/tratamiento farmacológico , Expresión Génica/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/microbiología , Ratones , Péptido Hidrolasas/efectos de los fármacos , Extractos Vegetales/farmacología , Quercetina/análogos & derivados , Quercetina/farmacología , Conejos , Vibrio cholerae/patogenicidad , Virulencia/efectos de los fármacos , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
7.
Microbes Infect ; 21(8-9): 368-376, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30853357

RESUMEN

Multiple diarrheagenic enteric bacterial infections cause global morbidity and mortality. A combination vaccine is needed to combat different diarrhea-causing organisms. In our present work, we formulated a combination of antigens from three different diarrheagenic Escherichia coli strains and three different Vibrio cholerae strains. We demonstrated that our newly formulated combination immunogen was able to raise species-specific immunogenicity. This formulation also gave protection against different diarrheagenic E. coli strains in the removable intestinal tie-adult rabbit diarrhea model. However, protective efficacy was not found against the V. cholerae El Tor Ogawa Haitian variant, but challenged with V. cholerae El Tor Inaba or O139 showed protection in rabbits. This is the first report of a single formulated nonliving heat-killed combination immunogen from different diarrheagenic E. coli and V. cholerae that could bestow protection against different bacteria in an animal model.


Asunto(s)
Vacunas contra el Cólera/inmunología , Cólera/prevención & control , Diarrea/prevención & control , Infecciones por Escherichia coli/prevención & control , Vacunas contra Escherichia coli/inmunología , Escherichia coli/inmunología , Vibrio cholerae/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Vacunas contra el Cólera/administración & dosificación , Protección Cruzada/inmunología , Diarrea/microbiología , Modelos Animales de Enfermedad , Escherichia coli/genética , Vacunas contra Escherichia coli/administración & dosificación , Inmunización , Inmunoglobulinas/sangre , Conejos , Vacunas Combinadas/inmunología , Vacunas de Productos Inactivados/inmunología , Vibrio cholerae/genética
8.
Front Microbiol ; 10: 111, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30804907

RESUMEN

Vibrio cholerae causes fatal diarrheal disease cholera in humans due to consumption of contaminated water and food. To instigate the disease, the bacterium must evade the host intestinal innate immune system; penetrate the mucus layer of the small intestine, adhere and multiply on the surface of microvilli and produce toxin(s) through the action of virulence associated genes. V. cholerae O1 that has caused a major cholera outbreak in Haiti contained several unique genetic signatures. These novel traits are used to differentiate them from the canonical El Tor strains. Several studies reported the spread of these Haitian variant strains in different parts of the world including Asia and Africa, but there is a paucity of information on the clinical consequence of these genetic changes. To understand the impact of these changes, we undertook a study involving mice and rabbit models to evaluate the pathogenesis. The colonization ability of Haitian variant strain in comparison to canonical El Tor strain was found to be significantly more in both suckling mice and rabbit model. Adult mice also displayed the same results. Besides that, infection patterns of Haitian variant strains showed a completely different picture. Increased mucosal damaging, colonization, and inflammatory changes were observed through hematoxylin-eosin staining and transmission electron microscopy. Fluid accumulation ability was also significantly higher in rabbit model. Our study indicated that these virulence features of the Haitian variant strain may have some association with the severe clinical outcome of the cholera patients in different parts of the world.

10.
PLoS One ; 13(9): e0203631, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30216367

RESUMEN

Salmonella Typhi and Salmonella Paratyphi A are the leading causative agents of enteric fever which cause morbidity and mortality worldwide. Currently, there is no combination vaccine which could protect infection from both the strains. In this paper, we are focusing on the development of a novel bivalent typhoidal Outer Membrane Vesicles (OMVs) based immunogen against enteric fever. We have isolated Salmonella Typhi and Paratyphi A OMVs and also characterized OMVs associated antigens. Then we immunized adult mice with three doses of our newly formulated bivalent immunogen orally (25 µg/200 µl). After three doses of oral immunization, we found our immunogen could significantly induce humoral response. We have also found serum IgG against LPS, Vi-polysaccharide etc. OMV immunization induces CD4, CD8 and CD19 population in immunized mice spleen. It also induces Th1 and Th17-cell mediated immunity. We also found bivalent OMVs immunization can prevent more than lethal dose of heterologous Salmonella strains mediated systemic infection in adult mice model. We determined that, the protective immune responses depend on the humoral and cell-mediated immune response. Furthermore, we have evaluated the mode of protective immune response carried out by anti-OMVs antibody by significantly inhibiting bacterial motility and mucin penetration ability. Taken together, these findings suggest that our bivalent immunogen could be used as a novel candidate vaccine against enteric fever.


Asunto(s)
Salmonella typhi/inmunología , Fiebre Tifoidea/inmunología , Vacunas Tifoides-Paratifoides/química , Animales , Anticuerpos Antibacterianos/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Fiebre Tifoidea/prevención & control
11.
PLoS Negl Trop Dis ; 11(7): e0005728, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28727722

RESUMEN

Few live attenuated vaccines protect against multiple serotypes of bacterial pathogen because host serotype-specific immune responses are limited to the serotype present in the vaccine strain. Here, immunization with a mutant of Shigella flexneri 2a protected guinea pigs against subsequent infection by S. dysenteriae type 1 and S. sonnei strains. This deletion mutant lacked the RNA-binding protein Hfq leading to increased expression of the type III secretion system via loss of regulation, resulting in attenuation of cell viability through repression of stress response sigma factors. Such increased antigen production and simultaneous attenuation were expected to elicit protective immunity against Shigella strains of heterologous serotypes. Thus, the vaccine potential of this mutant was tested in two guinea pig models of shigellosis. Animals vaccinated in the left eye showed fewer symptoms upon subsequent challenge via the right eye, and even survived subsequent intestinal challenge. In addition, oral vaccination effectively induced production of immunoglobulins without severe side effects, again protecting all animals against subsequent intestinal challenge with S. dysenteriae type 1 or S. sonnei strains. Antibodies against common virulence proteins and the O-antigen of S. flexneri 2a were detected by immunofluorescence microscopy. Reaction of antibodies with various strains, including enteroinvasive Escherichia coli, suggested that common virulence proteins induced protective immunity against a range of serotypes. Therefore, vaccination is expected to cover not only the most prevalent serotypes of S. sonnei and S. flexneri 2a, but also various Shigella strains, including S. dysenteriae type 1, which produces Shiga toxin.


Asunto(s)
Protección Cruzada , Disentería Bacilar/prevención & control , Eliminación de Gen , Proteína de Factor 1 del Huésped/deficiencia , Vacunas contra la Shigella/inmunología , Shigella/genética , Shigella/inmunología , Animales , Modelos Animales de Enfermedad , Disentería Bacilar/inmunología , Disentería Bacilar/patología , Cobayas , Masculino , Viabilidad Microbiana , Serogrupo , Vacunas contra la Shigella/administración & dosificación , Vacunas contra la Shigella/genética , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Virulencia
12.
Vaccine ; 35(28): 3534-3547, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28545924

RESUMEN

Bacterial outer membrane vesicles have been extensively investigated and considered as a next generation vaccine. Recently, we have demonstrated that the cholera pentavalent outer membrane vesicles (CPMVs) immunogen induced adaptive immunity and had a strong protective efficacy against the circulating V. cholerae strains in a mouse model. In this present study, we are mainly focusing on reducing outer membrane vesicle (OMV) -mediated toxicity without altering its antigenic property. Therefore, we have selected All-trans Retinoic Acid (ATRA), active metabolites of vitamin A, which have both anti-inflammatory and mucosal adjuvant properties. Pre-treatment of ATRA significantly reduced CPMVs induced TLR2 mediated pro-inflammatory responses in vitro and in vivo. Furthermore, we also found ATRA pre-treatment significantly induced mucosal immune response and protective efficacy after two doses of oral immunization with CPMVs (75µg). This study can help to reduce OMV based vaccine toxicity and induce better protective immunity where children and men suffered from malnutrition mainly in developing countries.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas contra el Cólera/inmunología , Inmunidad Mucosa , Inflamación/prevención & control , Tretinoina/administración & dosificación , Vibrio cholerae/inmunología , Administración Oral , Animales , Animales Recién Nacidos , Anticuerpos Antibacterianos/sangre , Proteínas de la Membrana Bacteriana Externa/toxicidad , Cólera/inmunología , Cólera/prevención & control , Vacunas contra el Cólera/administración & dosificación , Vacunas contra el Cólera/toxicidad , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Inmunogenicidad Vacunal , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos BALB C , Receptor Toll-Like 2/inmunología , Tretinoina/inmunología , Tretinoina/farmacología
13.
Cell Signal ; 35: 140-153, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28343946

RESUMEN

Cathelicidin antimicrobial peptide is a key component of the host innate immune system. It is constitutively expressed by the intestinal epithelial cells, but induced at further higher levels by different host-derived and microbial stimuli, including the ligands for Toll-like receptors (TLRs). While the underlying mechanisms of cathelicidin expression remain incompletely understood, altered expression may be associated with gastro-intestinal infections and inflammatory diseases. We demonstrate here that viral double-stranded RNA and its synthetic analog poly(I:C) are potent and tissue-specific inducers of cathelicidin mRNA and protein expression in the mouse as well as human intestinal epithelial cells. Reporter assays showed that poly(I:C) transcriptionally regulates murine cathelicidin-related antimicrobial peptide (mCRAMP) by recruiting Sp1 transcription factor to the GC-box cis-regulatory element at -71bp of the mCRAMP putative promoter. Sp1 recruitment to the endogenous mCRAMP promoter was confirmed by chromatin immunoprecipitation (ChIP) assays. Immunoblotting, qPCR, ChIP and siRNA-mediated gene knockdown studies revealed that the activation of phosphatidylinositol 3-kinase/protein kinase Cζ pathways in poly(I:C)-stimulated cells underlies Sp1 phosphorylation and recruitment to the mCRAMP promoter, leading to enhanced transcription. We further showed that intra-rectal poly(I:C) administration in mice reduces intestinal bacterial load and mucosal inflammation following Shigella flexneri 2a infection by inducing mCRAMP expression in the colonic epithelial cells. This study reports novel regulatory mechanisms of cathelicidin expression that may be targeted to treat gastro-intestinal infections.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Disentería Bacilar/genética , Proteína Quinasa C/genética , Factor de Transcripción Sp1/genética , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Disentería Bacilar/metabolismo , Disentería Bacilar/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Regulación de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Regiones Promotoras Genéticas , ARN Bicatenario/genética , Shigella flexneri/efectos de los fármacos , Shigella flexneri/patogenicidad , Transducción de Señal/genética , Catelicidinas
14.
Jpn J Infect Dis ; 70(1): 111-114, 2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-27169940

RESUMEN

Diabetes mellitus and diarrhea are becoming increasingly burdensome worldwide, particularly in developing countries such as India. Diabetic patients are susceptible to infection with pathogenic bacteria, particularly those causing invasive enteric infections. In this study, we observed changes in the pathophysiological features of mice with streptozotocin-induced hyperglycemia. In our experiments, both hyperglycemic and control mice were infected with pathogenic enteric bacteria-non-typhoidal Salmonella, Shigella flexneri, or Vibrio parahaemolyticus. Morbidity, mortality, and bacterial load were all higher in the diabetic mice than in the control mice, and the phagocytic and bactericidal activities of peritoneal macrophages isolated from hyperglycemic mice were lower than they were in the controls. We hypothesize that hyperglycemia leads to a downregulation of the innate immune response, which in turn increases vulnerability to enteric bacterial infection.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diarrea/epidemiología , Disentería Bacilar/epidemiología , Hiperglucemia/inducido químicamente , Hiperglucemia/complicaciones , Salmonelosis Animal/epidemiología , Vibriosis/epidemiología , Animales , Antibióticos Antineoplásicos , Carga Bacteriana , Diarrea/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Disentería Bacilar/inmunología , Macrófagos Peritoneales/inmunología , Ratones , Ratones Obesos , Salmonelosis Animal/inmunología , Estreptozocina/administración & dosificación , Análisis de Supervivencia , Vibriosis/inmunología
15.
Sci Rep ; 6: 39454, 2016 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-27991578

RESUMEN

Targeting bacterial virulence mechanisms without compromising bacterial growth is a promising strategy to prevent drug resistance. LysR-type transcriptional regulators (LTTRs) possess structural conservation across bacterial species and regulate virulence in numerous pathogens, making them attractive targets for antimicrobial agents. We targeted AphB, a Vibrio cholerae LTTR, which regulates the expression of genes encoding cholera toxin and toxin-co-regulated pilus for inhibitor designing. Since AphB ligand is unknown, we followed a molecular fragment-based approach for ligand designing using FDA-approved drugs and subsequent screen to identify molecules that exhibited high-affinity binding to AphB ligand-binding pocket. Among the identified compounds, ribavirin, an anti-viral drug, antagonized AphB functions. Ribavirin perturbed Vibrio cholerae pathogenesis in animal models. The inhibitory effects of the drug was limited to the bacteria expressing wild type AphB, but not its constitutively active mutant (AphBN100E), which represents the ligand-bound state, suggesting that ribavirin binds to the active site of AphB to exert its inhibitory role and there exists no AphB-independent mechanism of its action. Similarly, ribavirin suppressed the functions of Salmonella Typhi LTTR Hrg, indicating its broad spectrum efficacy. Moreover, ribavirin did not affect the bacterial viability in culture. This study cites an example of drug repurposing for anti-infective therapy.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Ribavirina/farmacología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Vibrio cholerae/metabolismo , Animales , Reposicionamiento de Medicamentos , Ligandos , Masculino , Ratones , Conformación Molecular , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Conejos , Salmonella typhi , Especificidad por Sustrato , Vibrio cholerae/genética , Virulencia
16.
Vaccine ; 34(42): 5099-5106, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27614779

RESUMEN

Salmonella-induced gastroenteritis causes massive morbidity and mortality in both adults and children of developing countries. However, it is difficult to study the mode of infection and vaccine efficacy due to inadequacies of current animal models. For this reason, we have explored using zebrafish as an improved model for non-typhoidal Salmonella (NTS) infection, including Salmonella enterica Typhimurium, Salmonella enterica Enteritidis and Salmonella enterica Weltevreden. In this study, we found that after infection of zebrafish with NTS, severe diarrhea like symptoms were observed and NTS significantly colonized the zebrafish intestine without any manipulation of the normal intestinal microbiota of the fish. Furthermore, these strains can colonize for longer than 72h and induce severe inflammation in the intestine, which may induce fish death. We also found that infected fish can transmit the pathogen into naïve fish. Moreover, we have established that zebrafish is an excellent model for vaccine study. Successive triple bath vaccination with heat-killed single serotype S. Typhimurium and S. Enteritidis immunogen induced protective efficacy against a high dose (10(8)CFU/ml) of infection with these pathogens. This study provides a natural infection model for the study of NTS infection, transmission and vaccine efficacy.


Asunto(s)
Modelos Animales de Enfermedad , Inmunogenicidad Vacunal , Infecciones por Salmonella/prevención & control , Vacunas contra la Salmonella/inmunología , Salmonella typhimurium/patogenicidad , Pez Cebra , Animales , Diarrea/microbiología , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/transmisión , Salmonelosis Animal/microbiología , Salmonella typhimurium/inmunología , Vacunas Tifoides-Paratifoides/inmunología
17.
Int J Med Microbiol ; 306(8): 657-665, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27670078

RESUMEN

Vibrio cholera survival in an aquatic environment depends on chitin utilization pathway that requires two factors, chitin binding protein and chitinases. The chitinases and the chitin utilization pathway are regulated by a two-component sensor histidine kinase ChiS in V. cholerae. In recent studies these two factors are also shown to be involved in V. cholerae pathogenesis. However, the role played by their upstream regulator ChiS in pathogenesis is yet to be known. In this study, we investigated the activation of ChiS in presence of mucin and its functional role in pathogenesis. We found ChiS is activated in mucin supplemented media. The isogenic chiS mutant (ChiS-) showed less growth compared to the wild type strain (ChiS+) in the presence of mucin supplemented media. The ChiS- strain also showed highly retarded motility as well as mucin layer penetration in vitro. Our result also showed that ChiS was important for adherence and survival in HT-29 cell. These observations indicate that ChiS is activated in presence of intestinal mucin and subsequently switch on the chitin utilization pathway. In animal models, our results also supported the in vitro observation. We found reduced fluid accumulation and colonization during infection with ChiS- strain. We also found ChiS- mutant with reduced expression of ctxA, toxT and tcpA. The cumulative effect of these events made V. cholerae ChiS- strain hypovirulent. Hence, we propose that ChiS plays a vital role in V. cholerae pathogenesis.


Asunto(s)
Histidina Quinasa/metabolismo , Vibrio cholerae/patogenicidad , Factores de Virulencia/metabolismo , Animales , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Línea Celular , Toxina del Cólera/metabolismo , Medios de Cultivo/química , Proteínas Fimbrias/metabolismo , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Células Caliciformes/microbiología , Histidina Quinasa/genética , Humanos , Locomoción , Ratones , Viabilidad Microbiana , Mucinas/metabolismo , Conejos , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Vibrio cholerae/crecimiento & desarrollo
18.
Immunobiology ; 221(8): 918-26, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27039316

RESUMEN

Diarrhea is a very common health problem in both developing and developed countries. Among the major entero-invasive bacteria, Shigella, Salmonella and Campylobacter cause serious problems in different geographic regions. Recently we have shown immunogenicity and protective efficacy of heat killed multi-serotype Shigella immunogen in different animal models. In our present study, we have advanced our research by preparing a combination heat-killed immunogen of three different entero-invasive bacteria, Shigella, Salmonella and Campylobacter. After three doses on 0th, 14th and 28th day of oral immunization with tri-valent heat-killed (TVHK) immunogen in rabbit model, the immunogenicity was determined by differential count of white blood cells and immunoglobulin assay at various time points. During oral immunization differential count of lymphocytes increased where as polymorphonuclear leucocytes (PMN) count decreased. Serum IgG and IgA showed significant elevation during oral immunization and remained at a detectable value upto 120 days. Protection study was performed in both, in vitro and in vivo conditions, using bacteriocidal assay and rabbit ligated ileal loop model, respectively, which conferred protection against homologous bacteria. Moreover, immunoblot assay against whole cell lysate and lipopolysaccharide exhibited significant amount of antigen-specific immunoglobulins raised against three different bacteria which proved that proteins along with lipopolysaccharides played a pivotal role in immunogenicity and protective efficacy. This trivalent heat-killed immunogen could be a low-cost, simple, oral, non-living vaccine candidate for future use against invasive diarrhea.


Asunto(s)
Campylobacter jejuni/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Inmunogenicidad Vacunal , Salmonella typhimurium/inmunología , Shigella flexneri/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Modelos Animales de Enfermedad , Infecciones por Bacterias Gramnegativas/prevención & control , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Conejos
19.
Infect Immun ; 84(5): 1478-1490, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26930702

RESUMEN

Proteases in Vibrio cholerae have been shown to play a role in its pathogenesis. V. cholerae secretes Zn-dependent hemagglutinin protease (HAP) and calcium-dependent trypsin-like serine protease (VesC) by using the type II secretion system (TIISS). Our present studies demonstrated that these proteases are also secreted in association with outer membrane vesicles (OMVs) and transported to human intestinal epithelial cells in an active form. OMV-associated HAP induces dose-dependent apoptosis in Int407 cells and an enterotoxic response in the mouse ileal loop (MIL) assay, whereas OMV-associated VesC showed a hemorrhagic fluid response in the MIL assay, necrosis in Int407 cells, and an increased interleukin-8 (IL-8) response in T84 cells, which were significantly reduced in OMVs from VesC mutant strain. Our results also showed that serine protease VesC plays a role in intestinal colonization of V. cholerae strains in adult mice. In conclusion, our study shows that V. cholerae OMVs secrete biologically active proteases which may play a role in cytotoxic and inflammatory responses.


Asunto(s)
Exosomas/enzimología , Inflamación/patología , Péptido Hidrolasas/metabolismo , Vibrio cholerae/enzimología , Vibrio cholerae/patogenicidad , Factores de Virulencia/metabolismo , Animales , Muerte Celular , Línea Celular , Células Epiteliales/patología , Humanos , Íleon/microbiología , Íleon/patología , Ratones Endogámicos BALB C
20.
Vaccine ; 34(15): 1839-46, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-26878295

RESUMEN

Our previous studies on outer membrane vesicles based vaccine development against shigellosis, revealed the inability of Shigella to release significant amount of vesicles naturally, during growth. Disruption of tolA, one of the genes of the Tol-Pal system of Gram negative bacterial membrane, has increased the vesicle release rate of a Shigella boydii type 4 strain to approximately 60% higher. We also noticed the vesicles, released from tolA-disrupted strain captured more OmpA protein and lipopolysaccharide, compared to the vesicles released from its wild type prototype. Six to seven weeks old BALB/c mice, immunized with 25 µg of three oral doses of the vesicles, released by tolA mutant, conferred 100% protection against lethal homologous challenge through nasal route, compared to only 60% protection after the same dose of wild type immunogen. Mice, immunized with the vesicles from tolA-mutant, manifested significant secretion of mucosal IgG and IgA. A sharp and significant response of pro-inflammatory cytokines (TNF-α, IL-6, IFN-γ) were also observed in the lung lavage of these groups of mice, within 6h post challenge; but at 24h, these inflammatory cytokines showed the sign of subsidence and the system was taken over by the release of anti-inflammatory cytokines (IL-4 and IL-10). Studies with naïve peritoneal macrophages, proved further, the potency of these vesicles to stimulate nitric oxide and TNF-α, IL-12p70, IL-6 and IL-10 productions in-vitro. The ability of these vesicles to trigger polarization of CD4(+) T cells toward Th1 adaptive immune response, had also been observed along with the presence of anti-inflammatory cytokines in the system. Our study demonstrated, the vesicles from tolA-disrupted Shigella were able to suppress Shigella-mediated inflammation in the host and could balance between inflammation and anti-inflammation, promoting better survival and health of the infected mice. Outer membrane vesicles from tolA-mutant, could be a potential cost-effective vaccine candidate against shigellosis.


Asunto(s)
Vesículas Extracelulares/inmunología , Vacunas contra la Shigella/inmunología , Shigella boydii , Administración Oral , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/química , Proteínas de la Membrana Bacteriana Externa/inmunología , Líquido del Lavado Bronquioalveolar/química , Citocinas/química , Disentería Bacilar/prevención & control , Técnicas de Inactivación de Genes , Pulmón/inmunología , Macrófagos Peritoneales/inmunología , Ratones Endogámicos BALB C , Shigella boydii/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA