Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Int J Mol Sci ; 23(5)2022 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-35269654

RESUMO

A chronic kidney disease (CKD) causes uremic toxin accumulation and gut dysbiosis, which further induces gut leakage and worsening CKD. Lipopolysaccharide (LPS) of Gram-negative bacteria and (1➔3)-ß-D-glucan (BG) of fungi are the two most abundant gut microbial molecules. Due to limited data on the impact of intestinal fungi in CKD mouse models, the influences of gut fungi and Lacticaseibacillus rhamnosus L34 (L34) on CKD were investigated using oral C. albicans-administered 5/6 nephrectomy (5/6Nx) mice. At 16 weeks post-5/6Nx, Candida-5/6Nx mice demonstrated an increase in proteinuria, serum BG, serum cytokines (tumor necrotic factor-α; TNF-α and interleukin-6), alanine transaminase (ALT), and level of fecal dysbiosis (Proteobacteria on fecal microbiome) when compared to non-Candida-5/6Nx. However, serum creatinine, renal fibrosis, or gut barrier defect (FITC-dextran assay and endotoxemia) remained comparable between Candida- versus non-Candida-5/6Nx. The probiotics L34 attenuated several parameters in Candida-5/6Nx mice, including fecal dysbiosis (Proteobacteria and Bacteroides), gut leakage (fluorescein isothiocyanate (FITC)-dextran), gut-derived uremic toxin (trimethylamine-N-oxide; TMAO) and indoxyl sulfate; IS), cytokines, and ALT. In vitro, IS combined with LPS with or without BG enhanced the injury on Caco-2 enterocytes (transepithelial electrical resistance and FITC-dextran permeability) and bone marrow-derived macrophages (supernatant cytokines (TNF-α and interleukin-1 ß; IL-1ß) and inflammatory genes (TNF-α, IL-1ß, aryl hydrocarbon receptor, and nuclear factor-κB)), compared with non-IS activation. These injuries were attenuated by the probiotics condition media. In conclusion, Candida administration worsens kidney damage in 5/6Nx mice through systemic inflammation, partly from gut dysbiosis-induced uremic toxins, which were attenuated by the probiotics. The additive effects on cell injury from uremic toxin (IS) and microbial molecules (LPS and BG) on enterocytes and macrophages might be an important underlying mechanism.


Assuntos
Lacticaseibacillus rhamnosus , Insuficiência Renal Crônica , Uremia , Animais , Células CACO-2 , Candida , Citocinas , Disbiose/microbiologia , Glucanos , Humanos , Lacticaseibacillus rhamnosus/fisiologia , Lipopolissacarídeos/toxicidade , Camundongos , Fator de Necrose Tumoral alfa/efeitos adversos , Toxinas Urêmicas
2.
Clin Sci (Lond) ; 135(12): 1467-1486, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34131711

RESUMO

Enterocyte damage and gut dysbiosis are caused by iron-overload in thalassemia (Thl), possibly making the gut vulnerable to additional injury. Hence, iron-overload in the heterozygous ß-globin deficient (Hbbth3/+) mice were tested with 3% dextran sulfate solution (DSS). With 4 months of iron-gavage, iron accumulation, gut-leakage (fluorescein isothiocyanate dextran (FITC-dextran), endotoxemia, and tight junction injury) in Thl mice were more prominent than WT mice. Additionally, DSS-induced mucositis in iron-overloaded mice from Thl group was also more severe than the WT group as indicated by mortality, liver enzyme, colon injury (histology and tissue cytokines), serum cytokines, and gut-leakage (FITC-dextran, endotoxemia, bacteremia, and the detection of Green-Fluorescent Producing Escherichia coli in the internal organs after an oral administration). However, Lactobacillus rhamnosus GG attenuated the disease severity of DSS in iron-overloaded Thl mice as indicated by mortality, cytokines (colon tissue and serum), gut-leakage (FITC-dextran, endotoxemia, and bacteremia) and fecal dysbiosis (microbiome analysis). Likewise, Lactobacillus conditioned media (LCM) decreased inflammation (supernatant IL-8 and cell expression of TLR-4, nuclear factor κB (NFκB), and cyclooxygenase-2 (COX-2)) and increased transepithelial electrical resistance (TEER) in enterocytes (Caco-2 cells) stimulated by lipopolysaccharide (LPS) and LPS plus ferric ion. In conclusion, in the case of iron-overloaded Thl, there was a pre-existing intestinal injury that wask more vulnerable to DSS-induced bacteremia (gut translocation). Hence, the prevention of gut-derived bacteremia and the monitoring on gut-leakage might be beneficial in patients with thalassemia.


Assuntos
Sulfato de Dextrana/farmacologia , Ferro/metabolismo , Mucosite/induzido quimicamente , Sepse/etiologia , Animais , Citocinas/sangue , Disbiose/induzido quimicamente , Microbioma Gastrointestinal/efeitos dos fármacos , Inflamação/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Camundongos Transgênicos , Sepse/metabolismo , Talassemia/etiologia
3.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G966-G979, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32308038

RESUMO

Iron overload induces intestinal-permeability defect (gut leakage), and gut translocation of organismal molecules might enhance systemic inflammation and sepsis severity in patients with thalassemia (Thal). Hence, iron administration in Hbbth3/+ mice, heterozygous ß-globin-deficient Thal mice, was explored. Oral iron administration induced more severe secondary hemochromatosis and gut leakage in Thal mice compared with wild-type (WT) mice. Gut leakage was determined by 1) FITC-dextran assay, 2) spontaneous serum elevation of endotoxin (LPS) and (1→3)-ß-d-glucan (BG), molecular structures of gut-organisms, and 3) reduction of tight-junction molecules with increased enterocyte apoptosis (activated caspase-3) by immunofluorescent staining. Iron overload also enhanced serum cytokines and increased Bacteroides spp. (gram-negative bacteria) in feces as analyzed by microbiome analysis. LPS injection in iron-overloaded Thal mice produced higher mortality and prominent cytokine responses. Additionally, stimulation with LPS plus iron in macrophage from Thal mice induced higher cytokines production with lower ß-globin gene expression compared with WT. Furthermore, possible gut leakage as determined by elevated LPS or BG (>60 pg/mL) in serum without systemic infection was demonstrated in 18 out of 41 patients with ß-thalassemia major. Finally, enhanced LPS-induced cytokine responses of mononuclear cells from these patients compared with cells from healthy volunteers were demonstrated. In conclusion, oral iron administration in Thal mice induced more severe gut leakage and increased fecal gram-negative bacteria, resulting in higher levels of endotoxemia and serum inflammatory cytokines compared with WT. Preexisting hyperinflammatory cytokines in iron-overloaded Thal enhanced susceptibility toward infection.NEW & NOTEWORTHY Although the impact of iron accumulation in several organs of patients with thalassemia is well known, the adverse effect of iron accumulation in gut is not frequently mentioned. Here, we demonstrated iron-induced gut-permeability defect, impact of organismal molecules from gut translocation of, and macrophage functional defect upon the increased sepsis susceptibility in thalassemia mice.


Assuntos
Citocinas/metabolismo , Duodeno/metabolismo , Microbioma Gastrointestinal , Hemocromatose/metabolismo , Mediadores da Inflamação/metabolismo , Ferro/metabolismo , Macrófagos/metabolismo , Sepse/metabolismo , Talassemia beta/metabolismo , Adulto , Animais , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Duodeno/imunologia , Duodeno/microbiologia , Feminino , Óxido de Ferro Sacarado , Hemocromatose/induzido quimicamente , Hemocromatose/imunologia , Hemocromatose/microbiologia , Heterozigoto , Humanos , Lipopolissacarídeos , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Sepse/induzido quimicamente , Sepse/imunologia , Sepse/microbiologia , Adulto Jovem , Globinas beta/genética , Talassemia beta/genética , Talassemia beta/imunologia , Talassemia beta/microbiologia
4.
PLoS One ; 16(12): e0261189, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34941893

RESUMO

Because of a possible impact of capsaicin in the high concentrations on enterocyte injury (cytotoxicity) and bactericidal activity on probiotics, Lactobacillus rhamnosus L34 (L34) and Lactobacillus rhamnosus GG (LGG), the probiotics derived from Thai and Caucasian population, respectively, were tested in the chili-extract administered C57BL/6 mice and in vitro experiments. In comparison with placebo, 2 weeks administration of the extract from Thai chili in mice caused loose feces and induced intestinal permeability defect as indicated by FITC-dextran assay and the reduction in tight junction molecules (occludin and zona occludens-1) using fluorescent staining and gene expression by quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, the chili extracts also induced the translocation of gut pathogen molecules; lipopolysaccharide (LPS) and (1→3)-ß-d-glucan (BG) and fecal dysbiosis (microbiome analysis), including reduced Firmicutes, increased Bacteroides, and enhanced total Gram-negative bacteria in feces. Both L34 and LGG attenuated gut barrier defect (FITC-dextran, the fluorescent staining and gene expression of tight junction molecules) but not improved fecal consistency. Additionally, high concentrations of capsaicin (0.02-2 mM) damage enterocytes (Caco-2 and HT-29) as indicated by cell viability test, supernatant cytokine (IL-8), transepithelial electrical resistance (TEER) and transepithelial FITC-dextran (4.4 kDa) but were attenuated by Lactobacillus condition media (LCM) from both probiotic-strains. The 24 h incubation with 2 mM capsaicin (but not the lower concentrations) reduced the abundance of LGG (but not L34) implying a higher capsaicin tolerance of L34. However, Lactobacillus rhamnosus fecal abundance, using qRT-PCR, of L34 or LGG after 3, 7, and 20 days of the administration in the Thai healthy volunteers demonstrated the similarity between both strains. In conclusion, high dose chili extracts impaired gut permeability and induced gut dysbiosis but were attenuated by probiotics. Despite a better capsaicin tolerance of L34 compared with LGG in vitro, L34 abundance in feces was not different to LGG in the healthy volunteers. More studies on probiotics with a higher intake of chili in human are interesting.


Assuntos
Capsaicina/efeitos adversos , Disbiose/prevenção & controle , Trato Gastrointestinal/efeitos dos fármacos , Inflamação/prevenção & controle , Lacticaseibacillus rhamnosus/química , Probióticos/administração & dosagem , Adolescente , Adulto , Idoso , Animais , Antibacterianos/administração & dosagem , Antipruriginosos/administração & dosagem , Antipruriginosos/efeitos adversos , Capsaicina/administração & dosagem , Citocinas/metabolismo , Disbiose/induzido quimicamente , Disbiose/microbiologia , Disbiose/patologia , Fezes/microbiologia , Feminino , Trato Gastrointestinal/microbiologia , Humanos , Inflamação/induzido quimicamente , Inflamação/microbiologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Extratos Vegetais/administração & dosagem , Extratos Vegetais/efeitos adversos , Probióticos/efeitos adversos , Junções Íntimas , Adulto Jovem
5.
Gut Microbes ; 11(3): 465-480, 2020 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-31530137

RESUMO

CANDIDA ALBICANS: is abundant in the human gut mycobiota but this species does not colonize the mouse gastrointestinal tract. C. albicans administration in dextran-sulfate solution (DSS) induced-colitis mouse model (DSS+Candida) might resemble more to human condition, therefore, a DSS colitis model with Candida administration was studied; first, to test the influence of fungi in DSS model and second, to test the efficacy of Lactobacillus rhamnosus L34. We demonstrated serum (1→3)-ß-D-glucan (BG) elevation in patients with IBD and endoscopic moderate colitis in clinical remission, supporting the possible influence of gut fungi toward IBD in human. Then, in mouse model, Candida gavage was found to worsen the DSS model indicated by higher mortality rate, more severe colon histology and enhanced gut-leakage (FITC-dextran assay, endotoxemia, serum BG and blood bacterial burdens) but did not affect weight loss and diarrhea. DSS+Candida induced higher pro-inflammatory cytokines both in blood and in intestinal tissue. Worsened systemic pro-inflammatory cytokine responses in DSS+Candida compared with DSS alone was possibly due to the more severe translocation of LPS, BG and bacteria (not fungemia) from gut into systemic circulation. Interestingly, bacteremia from Pseudomonas aeruginosa was more frequently isolated from DSS+Candida than DSS alone. In parallel, P. aeruginosa was also isolated from fecal culture in most of the mice in DSS+Candida group supported by prominent Gammaproteobacteria in fecal microbioata analysis. However, L. rhamnosus L34 attenuated both DSS+Candida and DSS model through the attenuation of gut local inflammation (cytokines and histology), gut-leakage severity, fecal dysbiosis (culture method and microbiome analysis) and systemic inflammation (serum cytokines). In conclusion, gut Candida in DSS model induced fecal bacterial dysbiosis and enhanced leaky-gut induced bacteremia. Probiotic treatment strategy aiming to reduce gut-fungi and fecal dysbiosis could attenuate disease severity. Investigation on gut fungi in patients with IBD is highly interesting.


Assuntos
Bacteriemia/microbiologia , Candida albicans/patogenicidade , Colite/microbiologia , Disbiose/microbiologia , Inflamação/microbiologia , Lacticaseibacillus rhamnosus/fisiologia , Adolescente , Adulto , Animais , Bacteriemia/induzido quimicamente , Translocação Bacteriana , Candida albicans/efeitos dos fármacos , Colite/induzido quimicamente , Colite/patologia , Citocinas/sangue , Sulfato de Dextrana , Modelos Animais de Doenças , Disbiose/induzido quimicamente , Fezes/microbiologia , Microbioma Gastrointestinal , Células HT29 , Humanos , Inflamação/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Probióticos/uso terapêutico , Adulto Jovem
6.
Shock ; 53(2): 189-198, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-30829903

RESUMO

The influence of gut fungi in chronic colitis was investigated by repeated oral administration of Candida albicans in a 3% dextran sulfate solution (DSS) induced-colitis mouse model. Candida administration in the DSS (DSS+Candida) model enhanced the mortality rate and induced bacteremia (without candidemia) resulting from a gut perm-selectivity defect despite similar diarrheal severity in mice treated with DSS alone. The dominant fecal bacteria in DSS+Candida and DSS alone mice were Pseudomonas spp. and Enterobacter spp., respectively, implying that Candida induced gut dysbiosis. Interestingly, chloramphenicol-resistant bacterial colonies, predominantly Pseudomonas spp., appeared in the feces and blood of DSS+Candida mice (not the DSS alone group) during fungal culture. These antibiotic-resistant bacteria were also isolated, ex vivo, by incubating mouse feces with DSS and heat-killed Candida or (1→3)-ß-D-glucan, suggesting bacterial fermentation on fungi. Administration of Pseudomonas aeruginosa isolated from chloramphenicol-resistant bacteria in the DSS+Candida model enhanced the severity of disease, and increased growth of isolated P aeruginosa in blood agar containing heat-killed Candida was demonstrated. These data suggested the selection of a highly virulent bacterial strain following fecal Candida presentation in the gut. Additionally, reduction of fecal fungi with fluconazole decreased the burden of chloramphenicol-resistant bacteria, attenuating the severity of DSS+Candida. In conclusion, gut Candida induced bacteremia in the DSS model through an inflammation-induced gut perm-selectivity defect and facilitated the growth of some gut bacteria. Treatment strategies aimed at reducing gut fungi could attenuate disease severity. Further investigation of gut fungi in inflammatory bowel disease is warranted.


Assuntos
Sulfato de Dextrana/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Sepse/microbiologia , Animais , Candida albicans/efeitos dos fármacos , Modelos Animais de Doenças , Disbiose/tratamento farmacológico , Disbiose/microbiologia , Fluconazol/farmacologia , Fluconazol/uso terapêutico , Células HT29 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pseudomonas aeruginosa/efeitos dos fármacos , Sepse/tratamento farmacológico
7.
Sci Rep ; 10(1): 777, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31964918

RESUMO

The influence of gut-leakage or gut-microbiota upon lupus progression was explored in 2 lupus mouse models. Pristane, administered in 4-wk-old wild-type (WT) female mice, induced lupus characteristics at 24-wk-old similar to the lupus-onset in FcGRIIb-/- mice. Gut-microbiota alteration was induced by co-housing together with the gavage of feces from 40-wk-old FcGRIIb-/- mice (symptomatic lupus). On the other hand,  gut-leakage was induced  by dextran sulfate solution (DSS). DSS and gut-microbiota alteration induced high serum anti-dsDNA immunoglobulin (Ig) as early as 30 days post-DSS only in FcGRIIb-/- mice. DSS, but not gut-microbiota alteration, enhanced lupus characteristics (serum creatinine and proteinuria) in both lupus models (but not in WT) at 60 days post-DSS. Indeed, DSS induced the translocation of molecular components of gut-pathogens as determined by bacterial burdens in mesenteric lymph node (MLN), endotoxemia (gut-bacterial molecule) and serum (1→3)-ß-D-glucan (BG) (gut-fungal molecule) as early as 15 days post-DSS together with enhanced MLN apoptosis in both WT and lupus mice. However, DSS induced spleen apoptosis in FcGRIIb-/- and WT mice at 30 and 60 days post-DSS, respectively, suggesting the higher impact of gut-leakage against spleen of lupus mice. In addition, macrophages preconditioning with LPS plus BG were susceptible to starvation-induced apoptosis, predominantly in FcGRIIb-/- cell, implying the influence of gut-leakage upon cell stress. In summary, gut-leakage induced gut-translocation of organismal-molecules then enhanced the susceptibility of stress-induced apoptosis, predominantly in lupus. Subsequently, the higher burdens of apoptosis in lupus mice increased anti-dsDNA Ig and worsen lupus severity through immune complex deposition. Hence, therapeutic strategies addressing gut-leakage in lupus are interesting.


Assuntos
Sulfato de Dextrana/efeitos adversos , Lúpus Eritematoso Sistêmico/genética , Receptores de IgG/deficiência , Terpenos/efeitos adversos , Animais , Citocinas/sangue , Modelos Animais de Doenças , Progressão da Doença , Fezes/microbiologia , Feminino , Lúpus Eritematoso Sistêmico/induzido quimicamente , Lúpus Eritematoso Sistêmico/microbiologia , Camundongos , beta-Glucanas/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA