Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38698218

RESUMO

Our study specifically explores the biosynthesis of copper-silver bimetallic nanoparticles (Cu-Ag BMNPs) using Argyreia nervosa (AN) plant leaf green extract as a versatile agent for capping, reducing, and stabilizing. This biosynthesis method is characterized by its simplicity and cost-effectiveness, utilizing silver nitrate (AgNO3) and cupric oxide (CuO) as precursor materials. Our comprehensive characterization of the Cu-Ag BMNPs, employing techniques such as X-ray diffraction (XRD), UV-Vis spectrometry, scanning electron microscopy (SEM), Zetasizer, and Fourier transformed infrared spectrometry (FTIR). FTIR analysis reveals biofunctional groups and chemical bands, while SEM and XRD analyses provide morphological and structural details. To evaluate the antimicrobial properties of the Cu-Ag BMNPs, we conducted disc diffusion and minimum inhibitory concentration (MIC) assays against Escherichia coli (E. coli), with results compared to the standard gentamicin antibiotic. It is observed that the 2% and 5% CuO concentrations of AN Cu-Ag BMNPs exhibit substantial antibacterial activity in comparison to AN extract when tested on EPEC. Among these, the Cu-Ag BMNPs at a 2% concentration demonstrate higher antibacterial activity, potentially attributed to the enhanced dispersion of BMNPs facilitated by the lower CuO doping concentration. These two assays showcased the improved antimicrobial activity of Cu-Ag BMNPs, highlighting their synergistic effect, characterized by high MIC values and a broad zone of inhibition in the disc diffusion tests against E. coli. These results emphasize the significant antibacterial potential of the synthesized BMNPs, with a medicinal plant AN leaf extract playing a pivotal role in enhancing antibacterial activity.

2.
Front Immunol ; 9: 2211, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30319652

RESUMO

Background: Current ulcerative colitis (UC) treatments are focused on symptom management primarily via immune suppression. Despite the current arsenal of immunosuppressant treatments, the majority of patients with UC still experience disease progression. Importantly, aggressive long-term inhibition of immune function comes with consequent risk, such as serious infections and malignancy. There is thus a recognized need for new, safe and effective treatment strategies for people living with UC that work upstream of managing the symptoms of the disease. The objective of this study was to evaluate a microbial-based treatment, QBECO, that functions to productively activate rather than suppress mucosal immune function as a novel approach to treat UC. Methods: Two established models of experimental colitis, namely chemically-induced DSS colitis and the spontaneous colitis that develops in Muc2 deficient mice, were used to assess whether QBECO treatment could ameliorate gastrointestinal disease. A small exploratory 16-week QBECO open-label trial was subsequently conducted to test the safety and tolerability of this approach and also to determine whether similar improvements in clinical disease and histopathology could be demonstrated in patients with moderate-to-severe UC. Results: QBECO treatment successfully reduced inflammation and promoted mucosal and histological healing in both experimental models and in UC patients. The preclinical models of colitis showed that QBECO ameliorated mucosal pathology, in part by reducing inflammatory cell infiltration, primarily that induced by neutrophils and inflammatory T cells. The most rapid and noticeable change observed in QBECO treated UC patients was a marked reduction in rectal bleeding. Conclusion: Collectively, this work demonstrates for the first time that strategically activating immune function rather than suppressing it, not only does not worsen colitis induced-damage, but may lead to an objective reduction in UC disease pathology.


Assuntos
Colite Ulcerativa/terapia , Escherichia coli/imunologia , Microbioma Gastrointestinal/imunologia , Imunoterapia/métodos , Mucosa Intestinal/metabolismo , Adulto , Animais , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/genética , Colite Ulcerativa/imunologia , Colo/imunologia , Colo/metabolismo , Colo/microbiologia , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Feminino , Humanos , Injeções Subcutâneas , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucina-2/genética , Resultado do Tratamento , Adulto Jovem
3.
Am J Physiol Gastrointest Liver Physiol ; 314(3): G360-G377, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29122749

RESUMO

Goblet cells (GCs) are the predominant secretory epithelial cells lining the luminal surface of the mammalian gastrointestinal (GI) tract. Best known for their apical release of mucin 2 (Muc2), which is critical for the formation of the intestinal mucus barrier, GCs have often been overlooked for their active contributions to intestinal protection and host defense. In part, this oversight reflects the limited tools available to study their function but also because GCs have long been viewed as relatively passive players in promoting intestinal homeostasis and host defense. In light of recent studies, this perspective has shifted, as current evidence suggests that Muc2 as well as other GC mediators are actively released into the lumen to defend the host when the GI tract is challenged by noxious stimuli. The ability of GCs to sense and respond to danger signals, such as bacterial pathogens, has recently been linked to inflammasome signaling, potentially intrinsic to the GCs themselves. Moreover, further work suggests that GCs release Muc2, as well as other mediators, to modulate the composition of the gut microbiome, leading to both the expansion as well as the depletion of specific gut microbes. This review will focus on the mechanisms by which GCs actively defend the host from noxious stimuli, as well as describe advanced technologies and new approaches by which their responses can be addressed. Taken together, we will highlight current insights into this understudied, yet critical, aspect of intestinal mucosal protection and its role in promoting gut defense and homeostasis.


Assuntos
Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Microbioma Gastrointestinal , Células Caliciformes/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/microbiologia , Animais , Bactérias/crescimento & desenvolvimento , Infecções Bacterianas/metabolismo , Infecções Bacterianas/fisiopatologia , Células Caliciformes/metabolismo , Homeostase , Interações Hospedeiro-Patógeno , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/fisiopatologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatologia , Mucina-2/metabolismo , Muco/metabolismo , Transdução de Sinais
4.
Am J Physiol Endocrinol Metab ; 314(5): E418-E432, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29066462

RESUMO

Both type 2 diabetes (T2D) and nonalcoholic steatohepatitis (NASH) are associated with reduced hepatic mitochondrial respiratory capacity. Cholic acid (CA) is the predominant 12α-hydroxylated bile acid that regulates hepatic lipid metabolism, and its circulating levels are negatively correlated with insulin resistance. Abolishing CA synthesis via the genetic disruption of the enzyme sterol 12α-hydroxylase ( Cyp8b1-/-) leads in resistance to diabetes and hepatic steatosis. Here, we show that long-term stimulation of hepatic lipogenesis leads to a severe impairment in overall metabolic and respiratory function in control mice ( Cyp8b1+/+) but strikingly not in Cyp8b1-/- mice. Cyp8b1-/- mice are protected from such metabolic impairments associated with T2D and NASH by inhibiting hepatic de novo lipogenic gene and protein expression and altering gut microbiota composition. The protective phenotype is compromised when NASH induction is independent of impairment in de novo lipogenesis (DNL). Consequently, Cyp8b1-/- mice also show a reduction in hepatic inflammation and fibrosis along with a shift in antimicrobial dynamics in the small intestine. Our data show that the altered bile acid composition of Cyp8b1-/- mice preserves metabolic and respiratory function by repressing hepatic DNL and driving favorable changes in gut antimicrobial responses.


Assuntos
Microbioma Gastrointestinal/genética , Interações entre Hospedeiro e Microrganismos/genética , Metabolismo dos Lipídeos/genética , Hepatopatia Gordurosa não Alcoólica/genética , Esteroide 12-alfa-Hidroxilase/genética , Animais , Ácidos e Sais Biliares/metabolismo , Células Cultivadas , Metabolismo Energético/genética , Feminino , Deleção de Genes , Resistência à Insulina/genética , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Testes de Função Respiratória
5.
Infect Immun ; 85(1)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27795363

RESUMO

Enteropathogenic Escherichia coli (EPEC)-induced diarrhea is often associated with disruption of intestinal epithelial tight junctions. Although studies have shown alterations in the expression and localization of bicellular tight junction proteins during EPEC infections, little is known about whether tricellular tight junction proteins (tTJs) are affected. Using Caco-2 cell monolayers, we investigated if EPEC is capable of targeting the tTJ protein tricellulin. Our results demonstrated that at 4 h postinfection, EPEC induced a significant reduction in tricellulin levels, accompanied by a significant loss of transepithelial resistance (TEER) and a corresponding increase in paracellular permeability. Conversely, cells overexpressing tricellulin were highly resistant to EPEC-induced barrier disruption. Confocal microscopy revealed the distribution of tricellulin into the plasma membrane of infected epithelial cells and confirmed the localization of EPEC aggregates in close proximity to tTJs. Moreover, infections with EPEC strains lacking genes encoding specific type III secreted effector proteins demonstrated a crucial role for the effector EspG1 in modulating tricellulin expression. Complementation studies suggest that the EspG-induced depletion of tricellulin is microtubule dependent. Overall, our results show that EPEC-induced epithelial barrier dysfunction is mediated in part by EspG1-induced microtubule-dependent depletion of tricellulin.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteína 2 com Domínio MARVEL/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Junções Íntimas/metabolismo , Células CACO-2 , Linhagem Celular Tumoral , Diarreia/metabolismo , Diarreia/microbiologia , Escherichia coli Enteropatogênica/metabolismo , Infecções por Escherichia coli/microbiologia , Humanos , Microtúbulos/metabolismo , Microtúbulos/microbiologia , Permeabilidade , Junções Íntimas/microbiologia
6.
PLoS Pathog ; 11(8): e1005108, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26285214

RESUMO

Enterohemorrhagic Escherichia coli and related food and waterborne pathogens pose significant threats to human health. These attaching/effacing microbes infect the apical surface of intestinal epithelial cells (IEC), causing severe diarrheal disease. Colonizing the intestinal luminal surface helps segregate these microbes from most host inflammatory responses. Based on studies using Citrobacter rodentium, a related mouse pathogen, we speculate that hosts rely on immune-mediated changes in IEC, including goblet cells to defend against these pathogens. These changes include a CD4+ T cell-dependent increase in IEC proliferation to replace infected IEC, as well as altered production of the goblet cell-derived mucin Muc2. Another goblet cell mediator, REsistin-Like Molecule (RELM)-ß is strongly induced within goblet cells during C. rodentium infection, and was detected in the stool as well as serum. Despite its dramatic induction, RELM-ß's role in host defense is unclear. Thus, wildtype and RELM-ß gene deficient mice (Retnlb-/-) were orally infected with C. rodentium. While their C. rodentium burdens were only modestly elevated, infected Retnlb-/- mice suffered increased mortality and mucosal ulceration due to deep pathogen penetration of colonic crypts. Immunostaining for Ki67 and BrDU revealed Retnlb-/- mice were significantly impaired in infection-induced IEC hyper-proliferation. Interestingly, exposure to RELM-ß did not directly increase IEC proliferation, rather RELM-ß acted as a CD4+ T cell chemoattractant. Correspondingly, Retnlb-/- mice showed impaired CD4+ T cell recruitment to their infected colons, along with reduced production of interleukin (IL)-22, a multifunctional cytokine that directly increased IEC proliferation. Enema delivery of RELM-ß to Retnlb-/- mice restored CD4+ T cell recruitment, concurrently increasing IL-22 levels and IEC proliferation, while reducing mucosal pathology. These findings demonstrate that RELM-ß and goblet cells play an unexpected, yet critical role in recruiting CD4+ T cells to the colon to protect against an enteric pathogen, in part via the induction of increased IEC proliferation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Colite/imunologia , Células Caliciformes/imunologia , Hormônios Ectópicos/imunologia , Mucosa Intestinal/imunologia , Animais , Separação Celular , Citrobacter rodentium , Colite/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Células Caliciformes/metabolismo , Hormônios Ectópicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase
7.
PLoS One ; 10(5): e0125225, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25932952

RESUMO

Inflammatory bowel disease is a chronic gastrointestinal inflammatory disorder associated with changes in neuropeptide expression and function, including vasoactive intestinal peptide (VIP). VIP regulates intestinal vasomotor and secretomotor function and motility; however, VIP's role in development and maintenance of colonic epithelial barrier homeostasis is unclear. Using VIP deficient (VIPKO) mice, we investigated VIP's role in epithelial barrier homeostasis, and susceptibility to colitis. Colonic crypt morphology and epithelial barrier homeostasis were assessed in wildtype (WT) and VIPKO mice, at baseline. Colitic responses were evaluated following dinitrobenzene sulfonic acid (DNBS) or dextran-sodium sulfate (DSS) exposure. Mice were also treated with exogenous VIP. At baseline, VIPKO mice exhibited distorted colonic crypts, defects in epithelial cell proliferation and migration, increased apoptosis, and altered permeability. VIPKO mice also displayed reduced goblet cell numbers, and reduced expression of secreted goblet cell factors mucin 2 and trefoil factor 3. These changes were associated with reduced expression of caudal type homeobox 2 (Cdx2), a master regulator of intestinal function and homeostasis. DNBS and DSS-induced colitis were more severe in VIPKO than WT mice. VIP treatment rescued the phenotype, protecting VIPKO mice against DSS colitis, with results comparable to WT mice. In conclusion, VIP plays a crucial role in the development and maintenance of colonic epithelial barrier integrity under physiological conditions and promotes epithelial repair and homeostasis during colitis.


Assuntos
Colite/prevenção & controle , Homeostase/efeitos dos fármacos , Intestinos/patologia , Substâncias Protetoras/farmacologia , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Fator de Transcrição CDX2 , Contagem de Células , Colite/patologia , Dinitrofluorbenzeno/análogos & derivados , Suscetibilidade a Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Caliciformes/patologia , Proteínas de Homeodomínio/metabolismo , Intestinos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Peptídeo Intestinal Vasoativo/deficiência
8.
Infect Immun ; 82(9): 3753-63, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24958710

RESUMO

Intestinal epithelial cells (IECs), including secretory goblet cells, form essential physiochemical barriers that separate luminal bacteria from underlying immune cells in the intestinal mucosa. IECs are common targets for enteric bacterial pathogens, with hosts responding to these microbes through innate toll-like receptors that predominantly signal through the MyD88 adaptor protein. In fact, MyD88 signaling confers protection against several enteric bacterial pathogens, including Salmonella enterica serovar Typhimurium and Citrobacter rodentium. Since IECs are considered innately hyporesponsive, it is unclear whether MyD88 signaling within IECs contributes to this protection. We infected mice lacking MyD88 solely in their IECs (IEC-Myd88(-/-)) with S. Typhimurium. Compared to wild-type (WT) mice, infected IEC-Myd88(-/-) mice suffered accelerated tissue damage, exaggerated barrier disruption, and impaired goblet cell responses (Muc2 and RELMß). Immunostaining revealed S. Typhimurium penetrated the IECs of IEC-Myd88(-/-) mice, unlike in WT mice, where they were sequestered to the lumen. When isolated crypts were assayed for their antimicrobial actions, crypts from IEC-Myd88(-/-) mice were severely impaired in their antimicrobial activity against S. Typhimurium. We also examined whether MyD88 signaling in IECs impacted host defense against C. rodentium, with IEC-Myd88(-/-) mice again suffering exaggerated tissue damage, impaired goblet cell responses, and reduced antimicrobial activity against C. rodentium. These results demonstrate that MyD88 signaling within IECs plays an important protective role at early stages of infection, influencing host susceptibility to infection by controlling the ability of the pathogen to reach and survive at the intestinal mucosal surface.


Assuntos
Anti-Infecciosos/imunologia , Colite/imunologia , Células Caliciformes/imunologia , Mucosa Intestinal/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Animais , Citrobacter rodentium/imunologia , Colite/microbiologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Gastroenterite/imunologia , Gastroenterite/microbiologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Células Caliciformes/microbiologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/imunologia
9.
J Vis Exp ; (84): e51297, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24637969

RESUMO

Inflammatory Bowel Diseases (IBD), including Crohn's Disease and Ulcerative Colitis, have long been associated with a genetic basis, and more recently host immune responses to microbial and environmental agents. Dinitrobenzene sulfonic acid (DNBS)-induced colitis allows one to study the pathogenesis of IBD associated environmental triggers such as stress and diet, the effects of potential therapies, and the mechanisms underlying intestinal inflammation and mucosal injury. In this paper, we investigated the effects of dietary n-3 and n-6 fatty acids on the colonic mucosal inflammatory response to DNBS-induced colitis in rats. All rats were fed identical diets with the exception of different types of fatty acids [safflower oil (SO), canola oil (CO), or fish oil (FO)] for three weeks prior to exposure to intrarectal DNBS. Control rats given intrarectal ethanol continued gaining weight over the 5 day study, whereas, DNBS-treated rats fed lipid diets all lost weight with FO and CO fed rats demonstrating significant weight loss by 48 hr and rats fed SO by 72 hr. Weight gain resumed after 72 hr post DNBS, and by 5 days post DNBS, the FO group had a higher body weight than SO or CO groups. Colonic sections collected 5 days post DNBS-treatment showed focal ulceration, crypt destruction, goblet cell depletion, and mucosal infiltration of both acute and chronic inflammatory cells that differed in severity among diet groups. The SO fed group showed the most severe damage followed by the CO, and FO fed groups that showed the mildest degree of tissue injury. Similarly, colonic myeloperoxidase (MPO) activity, a marker of neutrophil activity was significantly higher in SO followed by CO fed rats, with FO fed rats having significantly lower MPO activity. These results demonstrate the use of DNBS-induced colitis, as outlined in this protocol, to determine the impact of diet in the pathogenesis of IBD.


Assuntos
Colite/induzido quimicamente , Colite/patologia , Gorduras na Dieta/administração & dosagem , Dinitrofluorbenzeno/análogos & derivados , Modelos Animais de Doenças , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/patologia , Ácido Trinitrobenzenossulfônico , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
10.
PLoS Pathog ; 9(8): e1003539, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23950714

RESUMO

Enteric bacterial pathogens such as enterohemorrhagic E. coli (EHEC) and Salmonella Typhimurium target the intestinal epithelial cells (IEC) lining the mammalian gastrointestinal tract. Despite expressing innate Toll-like receptors (TLRs), IEC are innately hypo-responsive to most bacterial products. This is thought to prevent maladaptive inflammatory responses against commensal bacteria, but it also limits antimicrobial responses by IEC to invading bacterial pathogens, potentially increasing host susceptibility to infection. One reason for the innate hypo-responsiveness of IEC is their expression of Single Ig IL-1 Related Receptor (SIGIRR), a negative regulator of interleukin (IL)-1 and TLR signaling. To address whether SIGIRR expression and the innate hypo-responsiveness of IEC impacts on enteric host defense, Sigirr deficient (-/-) mice were infected with the EHEC related pathogen Citrobacter rodentium. Sigirr -/- mice responded with accelerated IEC proliferation and strong pro-inflammatory and antimicrobial responses but surprisingly, Sigirr -/- mice proved dramatically more susceptible to infection than wildtype mice. Through haematopoietic transplantation studies, it was determined that SIGIRR expression by non-haematopoietic cells (putative IEC) regulated these responses. Moreover, the exaggerated responses were found to be primarily dependent on IL-1R signaling. Whilst exploring the basis for their susceptibility, Sigirr -/- mice were found to be unusually susceptible to intestinal Salmonella Typhimurium colonization, developing enterocolitis without the typical requirement for antibiotic based removal of competing commensal microbes. Strikingly, the exaggerated antimicrobial responses seen in Sigirr -/- mice were found to cause a rapid and dramatic loss of commensal microbes from the infected intestine. This depletion appears to reduce the ability of the microbiota to compete for space and nutrients (colonization resistance) with the invading pathogens, leaving the intestine highly susceptible to pathogen colonization. Thus, SIGIRR expression by IEC reflects a strategy that sacrifices maximal innate responsiveness by IEC in order to promote commensal microbe based colonization resistance against bacterial pathogens.


Assuntos
Infecções por Enterobacteriaceae/imunologia , Enterobacteriaceae/imunologia , Imunidade Inata , Receptores de Interleucina-1/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Animais , Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/genética , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Camundongos , Camundongos Knockout , Receptores de Interleucina-1/genética , Transdução de Sinais/genética , Receptores Toll-Like/genética
11.
J Vis Exp ; (72): e50222, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23462619

RESUMO

This protocol outlines the steps required to produce a robust model of infectious disease and colitis, as well as the methods used to characterize Citrobacter rodentium infection in mice. C. rodentium is a gram negative, murine specific bacterial pathogen that is closely related to the clinically important human pathogens enteropathogenic E. coli and enterohemorrhagic E. coli. Upon infection with C. rodentium, immunocompetent mice suffer from modest and transient weight loss and diarrhea. Histologically, intestinal crypt elongation, immune cell infiltration, and goblet cell depletion are observed. Clearance of infection is achieved after 3 to 4 weeks. Measurement of intestinal epithelial barrier integrity, bacterial load, and histological damage at different time points after infection, allow the characterization of mouse strains susceptible to infection. The virulence mechanisms by which bacterial pathogens colonize the intestinal tract of their hosts, as well as specific host responses that defend against such infections are poorly understood. Therefore the C. rodentium model of enteric bacterial infection serves as a valuable tool to aid in our understanding of these processes. Enteric bacteria have also been linked to Inflammatory Bowel Diseases (IBDs). It has been hypothesized that the maladaptive chronic inflammatory responses seen in IBD patients develop in genetically susceptible individuals following abnormal exposure of the intestinal mucosal immune system to enteric bacteria. Therefore, the study of models of infectious colitis offers significant potential for defining potentially pathogenic host responses to enteric bacteria. C. rodentium induced colitis is one such rare model that allows for the analysis of host responses to enteric bacteria, furthering our understanding of potential mechanisms of IBD pathogenesis; essential in the development of novel preventative and therapeutic treatments.


Assuntos
Citrobacter rodentium/patogenicidade , Colite/microbiologia , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/microbiologia , Animais , Interações Hospedeiro-Patógeno , Camundongos , Virulência
12.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G917-28, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23518681

RESUMO

Inflammatory bowel disease is an intestinal inflammatory disorder of multifactorial origin, in which diets that favor high n-6 and low n-3 fatty acids have been implicated. The present study addressed whether dietary n-6 and n-3 fatty acids alter colonic mucosal response to Citrobacter rodentium (C. rodentium) infection. Mice were fed diets identical except for fatty acids, with an energy percentage of 15% 18:2n-6 and <0.06% 18:3n-3, 4.2% 18:2n-6 and 1.9% 18:3n-3, or 1.44% 20:5n-3, 4.9% 22:6n-3, 0.32% 18:2n-6, and 0.12% 18:3n-3 from safflower, canola, or fish oil, respectively for 3 wk before infection. Dietary oils had no effect on colonic C. rodentium growth but altered colon 20:4n-6/(20:5n-3+22:6n-3) with 9.40 ± 0.06, 1.94 ± 0.08, and 0.32 ± 0.03% in colon phosphatidylcholine and 3.82 ± 0.18, 1.14 ± 0.02, and 0.30 ± 0.02% in phosphatidylethanolamine of mice fed safflower, canola, or fish oil, respectively. At 10 days postinfection, histological damage, F4/80-positive macrophages, and myeloperoxidase-positive neutrophils in colonic mucosa were higher in infected mice fed safflower than fish oil. Colon gene transcripts for macrophage inflammatory protein 2, keratinocyte cytokine, and monocyte chemoattractant protein 1 expression were significantly higher in infected mice fed safflower than canola or fish oil; IFN-γ, IL-6, and IL-17A expression were significantly elevated in mice fed safflower rather than fish oil; and IL-10 was significantly higher in mice fed fish oil rather than canola or safflower oil. This study demonstrates that oils high in 18:2n-6 with minimal n-3 fatty acids exacerbate mucosal immune response, whereas oils high in n-3 fatty acids attenuate mucosal immune response to C. rodentium. These studies implicate dietary oils as environmental modifiers of intestinal inflammation in response to infection.


Assuntos
Citrobacter rodentium , Colo/patologia , Gorduras Insaturadas na Dieta/farmacologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/patologia , Imunidade/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Carga Bacteriana , Proliferação de Células/efeitos dos fármacos , Colite/patologia , Colo/metabolismo , Citocinas/análise , Citocinas/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-6/farmacologia , Fezes/microbiologia , Fluoresceína-5-Isotiocianato , Imunofluorescência , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade , Fosfolipídeos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
13.
Infect Immun ; 79(5): 2043-50, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21383053

RESUMO

We investigated the early innate immune responses induced in human intestinal epithelial cells (IEC) by the three defined Toxoplasma gondii genotype strains. Transcriptome analysis revealed that among differentially expressed genes, ß-defensins distinguished the most IEC infected by fast- or slow-replicating T. gondii genotypes. Although ß-defensin 1 and 3 genes were not expressed in host cells at early time points postinfection, the slow-replicating type II and III parasites induced high levels of ß-defensin 2 gene expression. Notably, no ß-defensin 2 gene expression occurred early after infection with the fast-replicating type I parasite. However, activation of this gene in IEC by poly(I:C) treatment prior to infection substantially decreased parasite viability, and pretreatment of parasites with synthetic ß-defensin 2 significantly reduced their infectivity of IEC. These findings strongly support the modulation of early ß-defensin 2 expression as a mechanism used by type I T. gondii parasites to mediate immune evasion.


Assuntos
Evasão da Resposta Imune/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Toxoplasmose/imunologia , beta-Defensinas/biossíntese , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Toxoplasma/crescimento & desenvolvimento , Toxoplasma/imunologia , Toxoplasma/patogenicidade , Toxoplasmose/metabolismo , beta-Defensinas/imunologia
14.
Microbes Infect ; 13(4): 394-404, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21262371

RESUMO

CD4(+)CD25(+)Foxp3(+) T regulatory (Treg) cells, are known to regulate responses to infectious agents. Here we compared disease progression in BALB/c and C57BL/6(B6) mice infected perorally with Toxoplasma gondii for 7 days and examined the affect of partial depletion of Treg cells in these mice. BALB/c mice were seen to be resistant to peroral infection whereas B6 mice were susceptible in terms of mortality. Although the depletion of Treg cells before infection had no effect on the survival of B6 or BALB/c mice, it resulted in increased parasite burdens in BALB/c mice, especially in the lamina propria, but not in B6 mice. Pro-inflammatory cytokines were also increased in Treg cells depleted BALB/c mice as compared to B6 mice. In addition Treg cell depleted BALB/c mice displayed increased ileal histopathology compared to their non-treated counterparts. These findings provide evidence for the contribution of Treg cells, in the resistance of BALB/c mice against peroral T. gondii infection.


Assuntos
Antígenos CD/imunologia , Depleção Linfocítica , Linfócitos T Reguladores/imunologia , Toxoplasmose/imunologia , Doença Aguda , Animais , Anticorpos Monoclonais/imunologia , Células Cultivadas , Citocinas/imunologia , Progressão da Doença , Feminino , Íleo/imunologia , Íleo/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Toxoplasma/imunologia , Toxoplasmose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA