Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 216
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(14): 7941-7949, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32179676

RESUMO

Late-onset sepsis (LOS) is a highly consequential complication of preterm birth and is defined by a positive blood culture obtained after 72 h of age. The causative bacteria can be found in patients' intestinal tracts days before dissemination, and cohort studies suggest reduced LOS risk in breastfed preterm infants through unknown mechanisms. Reduced concentrations of epidermal growth factor (EGF) of maternal origin within the intestinal tract of mice correlated to the translocation of a gut-resident human pathogen Escherichia coli, which spreads systemically and caused a rapid, fatal disease in pups. Translocation of Escherichia coli was associated with the formation of colonic goblet cell-associated antigen passages (GAPs), which translocate enteric bacteria across the intestinal epithelium. Thus, maternally derived EGF, and potentially other EGFR ligands, prevents dissemination of a gut-resident pathogen by inhibiting goblet cell-mediated bacterial translocation. Through manipulation of maternally derived EGF and alteration of the earliest gut defenses, we have developed an animal model of pathogen dissemination which recapitulates gut-origin neonatal LOS.


Assuntos
Translocação Bacteriana/imunologia , Receptores ErbB/metabolismo , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Microbioma Gastrointestinal/imunologia , Leite Humano/imunologia , Sepse Neonatal/imunologia , Animais , Animais Recém-Nascidos , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Aleitamento Materno , Colo/metabolismo , Colo/microbiologia , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Fezes/química , Fezes/microbiologia , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Transgênicos , Leite Humano/metabolismo , Sepse Neonatal/metabolismo , Sepse Neonatal/microbiologia , Transdução de Sinais/imunologia , Fatores de Tempo
2.
J Immunol ; 204(5): 1255-1262, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31941655

RESUMO

Gut bacteria-associated sepsis is a serious concern in patients with gastrointestinal acute radiation syndrome (GIARS). In our previous studies, gut bacteria-associated sepsis caused high mortality rates in mice exposed to 6-9 Gy of γ-rays. IL-12+CD38+ iNOS+ Mϕ (M1Mϕ) located in the bacterial translocation site (mesenteric lymph nodes [MLNs]) of unirradiated mice were characterized as host defense antibacterial effector cells. However, cells isolated from the MLNs of GIARS mice were mostly CCL1+IL-10+LIGHT+miR-27a+ Mϕ (M2bMϕ, inhibitor cells for the M1Mϕ polarization). Reduced long noncoding RNA Gas5 and increased miR-222 expression in MLN-Mϕ influenced by the irradiation were shown to be associated with M2bMϕ polarization. In this study, the mortality of mice exposed to 7 Gy of γ-rays (7 Gy GIARS mice) was completely controlled after the administration of glycyrrhizin (GL), a major active ingredient in licorice root (Glycyrrhiza glabra). Bacterial translocation and subsequent sepsis were minimal in 7 Gy GIARS mice treated with GL. Increased Gas5 RNA level and decreased miR-222 expression were shown in MLN-Mϕ isolated from 7 Gy GIARS mice treated with GL, and these macrophages did not display any properties of M2bMϕ. These results indicate that gut bacteria-associated sepsis in 7 Gy GIARS mice was controlled by the GL through the inhibition of M2bMϕ polarization at the bacteria translocation site. Expression of Ccl1, a gene required for M2bMϕ survival, is silenced in the MLNs of 7 Gy GIARS mice because of Gas5 RNA, which is increased in these cells after the suppression of miR-222 (a Gas5 RNA expression inhibitor) by the GL.


Assuntos
Bactérias/imunologia , Infecções Bacterianas , Fenômenos Fisiológicos Bacterianos , Translocação Bacteriana , Raios gama/efeitos adversos , Ácido Glicirrízico/farmacologia , Intestinos , Macrófagos , MicroRNAs/imunologia , RNA Longo não Codificante/imunologia , Lesões Experimentais por Radiação , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/patologia , Infecções Bacterianas/prevenção & controle , Fenômenos Fisiológicos Bacterianos/efeitos dos fármacos , Fenômenos Fisiológicos Bacterianos/imunologia , Fenômenos Fisiológicos Bacterianos/efeitos da radiação , Translocação Bacteriana/efeitos dos fármacos , Translocação Bacteriana/imunologia , Translocação Bacteriana/efeitos da radiação , Intestinos/imunologia , Intestinos/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Lesões Experimentais por Radiação/imunologia , Lesões Experimentais por Radiação/microbiologia , Lesões Experimentais por Radiação/patologia , Lesões Experimentais por Radiação/prevenção & controle , Sepse/imunologia , Sepse/microbiologia , Sepse/patologia , Sepse/prevenção & controle
3.
Gastroenterology ; 159(3): 849-863, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32569766

RESUMO

Intestinal barrier dysfunction and dysbiosis contribute to development of diseases in liver and other organs. Physical, immunologic, and microbiologic (bacterial, fungal, archaeal, viral, and protozoal) features of the intestine separate its nearly 100 trillion microbes from the rest of the human body. Failure of any aspect of this barrier can result in translocation of microbes into the blood and sustained inflammatory response that promote liver injury, fibrosis, cirrhosis, and oncogenic transformation. Alterations in intestinal microbial populations or their functions can also affect health. We review the mechanisms that regulate intestinal permeability and how changes in the intestinal microbiome contribute to development of acute and chronic liver diseases. We discuss individual components of the intestinal barrier and how these are disrupted during development of different liver diseases. Learning more about these processes will increase our understanding of the interactions among the liver, intestine, and its flora.


Assuntos
Translocação Bacteriana/imunologia , Disbiose/complicações , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/fisiopatologia , Hepatopatias/imunologia , Animais , Modelos Animais de Doenças , Disbiose/imunologia , Disbiose/microbiologia , Disbiose/fisiopatologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Fígado/imunologia , Fígado/patologia , Hepatopatias/microbiologia , Hepatopatias/patologia , Permeabilidade
4.
Immunity ; 37(3): 563-73, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22981539

RESUMO

Mice lacking junctional adhesion molecule A (JAM-A, encoded by F11r) exhibit enhanced intestinal epithelial permeability, bacterial translocation, and elevated colonic lymphocyte numbers, yet do not develop colitis. To investigate the contribution of adaptive immune compensation in response to increased intestinal epithelial permeability, we examined the susceptibility of F11r(-/-)Rag1(-/-) mice to acute colitis. Although negligible contributions of adaptive immunity in F11r(+/+)Rag1(-/-) mice were observed, F11r(-/-)Rag1(-/-) mice exhibited increased microflora-dependent colitis. Elimination of T cell subsets and cytokine analyses revealed a protective role for TGF-ß-producing CD4(+) T cells in F11r(-/-) mice. Additionally, loss of JAM-A resulted in elevated mucosal and serum IgA that was dependent upon CD4(+) T cells and TGF-ß. Absence of IgA in F11r(+/+)Igha(-/-) mice did not affect disease, whereas F11r(-/-)Igha(-/-) mice displayed markedly increased susceptibility to acute injury-induced colitis. These data establish a role for adaptive immune-mediated protection from acute colitis under conditions of intestinal epithelial barrier compromise.


Assuntos
Imunidade Adaptativa/imunologia , Colite/imunologia , Mucosa Intestinal/imunologia , Intestinos/imunologia , Imunidade Adaptativa/genética , Animais , Translocação Bacteriana/genética , Translocação Bacteriana/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Colite/induzido quimicamente , Colite/genética , Sulfato de Dextrana , Epitélio/imunologia , Epitélio/metabolismo , Feminino , Citometria de Fluxo , Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Imunoglobulina A/genética , Imunoglobulina A/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
5.
Int J Med Sci ; 18(14): 3050-3058, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34400875

RESUMO

To investigate the effect of zinc (Zn) supplementation on intestinal microflora changes and bacterial translocation in rats with severe acute pancreatitis (SAP), the rats were divided into the sham surgery (SS), SAP, SS + Zn, and SAP + Zn groups. Saline (0.1 mL/100g) and 5% sodium taurocholate were injected into the pancreaticobiliary duct of the rats in the SS and SAP + Zn groups, respectively. Intraperitoneal injection of 5 mg/kg Zn was performed immediately after injecting saline or 5% sodium taurocholate into the rats in both groups. Serum amylase and Zn levels, plasma endogenous endotoxin, intestinal permeability, and the positive rate of intestinal bacterial translocation were detected, haematoxylin and eosin (H&E) staining was performed, and the pancreatic tissue scores were calculated for each group. In addition, immunohistochemical (IHC) staining was performed to evaluate the expression of IL-1ß and TNF-α. Real-time fluorescence quantitative PCR was used to quantify the gene copy numbers of Escherichia, Bifidobacterium, and Lactobacillus in the cecum. The levels of amylase and plasma endotoxin in the SAP group were significantly higher than those in the SS and SS + Zn groups. Intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were increased in the SAP group. However, the levels of amylase and plasma endotoxin were decreased as a result of zinc supplementation in the SAP group. The expression of IL-1ß and TNF-α was also reduced to a greater degree in the SAP + Zn group than in the SAP group. Moreover, alleviated intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were found in the SAP + Zn group. The results of real-time quantitative PCR showed that the gene copy number of Escherichia increased with time, and the gene copy numbers of Lactobacillus and Bifidobacterium decreased over time. Zn supplementation prevented the release of TNF-α and IL-1ß, alleviated intestinal permeability and endotoxemia, reduced bacterial translocation, and inhibited changes in pathogenic intestinal flora in rats with SAP.


Assuntos
Translocação Bacteriana/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Pancreatite/tratamento farmacológico , Zinco/administração & dosagem , Animais , Translocação Bacteriana/imunologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/imunologia , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Pâncreas/imunologia , Pâncreas/patologia , Pancreatite/imunologia , Pancreatite/microbiologia , Pancreatite/patologia , Permeabilidade/efeitos dos fármacos , Ratos , Índice de Gravidade de Doença
6.
Gut ; 69(10): 1796-1806, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32317332

RESUMO

OBJECTIVE: Bacterial translocation to various organs including human adipose tissue (AT) due to increased intestinal permeability remains poorly understood. We hypothesised that: (1) bacterial presence is highly tissue specific and (2) related in composition and quantity to immune inflammatory and metabolic burden. DESIGN: We quantified and sequenced the bacterial 16S rRNA gene in blood and AT samples (omental, mesenteric and subcutaneous) of 75 subjects with obesity with or without type 2 diabetes (T2D) and used catalysed reporter deposition (CARD) - fluorescence in situ hybridisation (FISH) to detect bacteria in AT. RESULTS: Under stringent experimental and bioinformatic control for contaminants, bacterial DNA was detected in blood and omental, subcutaneous and mesenteric AT samples in the range of 0.1 to 5 pg/µg DNA isolate. Moreover, CARD-FISH allowed the detection of living, AT-borne bacteria. Proteobacteria and Firmicutes were the predominant phyla, and bacterial quantity was associated with immune cell infiltration, inflammatory and metabolic parameters in a tissue-specific manner. Bacterial composition differed between subjects with and without T2D and was associated with related clinical measures, including systemic and tissues-specific inflammatory markers. Finally, treatment of adipocytes with bacterial DNA in vitro stimulated the expression of TNFA and IL6. CONCLUSIONS: Our study provides contaminant aware evidence for the presence of bacteria and bacterial DNA in several ATs in obesity and T2D and suggests an important role of bacteria in initiating and sustaining local AT subclinical inflammation and therefore impacting metabolic sequelae of obesity.


Assuntos
Tecido Adiposo , Translocação Bacteriana/imunologia , DNA Bacteriano/isolamento & purificação , Diabetes Mellitus Tipo 2 , Firmicutes/isolamento & purificação , Obesidade , Proteobactérias/isolamento & purificação , RNA Ribossômico 16S/sangue , Tecido Adiposo/imunologia , Tecido Adiposo/microbiologia , Células Cultivadas , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/imunologia , Feminino , Humanos , Inflamação/imunologia , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/imunologia , Fator de Necrose Tumoral alfa/metabolismo
7.
Hepatology ; 70(3): 925-938, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30414342

RESUMO

In cirrhosis, intestinal dysbiosis, intestinal barrier impairment, and systemic immune system abnormalities lead to gut bacterial translocation (GBT) and bacterial infection. However, intestinal immune system dysfunction and its contribution to barrier damage are poorly understood. This study correlates immune system dysregulation in the intestines of rats at different stages of CCl4 -induced cirrhosis with barrier function and pathogenic microbiota. The following variables were addressed in the small intestine: intraepithelial lymphocyte (IEL) and lamina propria lymphocyte (LPL) activation status and cytokine production (flow cytometry), cytokine mRNA and protein expression (quantitative real-time PCR and immunofluorescence), microbiota composition of ileum content (16S recombinant DNA massive sequencing), permeability (fecal albumin loss), and epithelial junctions (immunohistochemistry and immunofluorescence). The intestinal mucosa in rats with cirrhosis showed a proinflammatory pattern of immune dysregulation in IELs and LPLs, which featured the expansion of activated lymphocytes, switch to a T helper 1 (Th1) regulatory pattern, and Th17 reduction. In rats with cirrhosis with ascites, this state was associated with epithelial junction protein disruption, fecal albumin loss, and GBT. Direct correlations (P < 0.01) were observed between elevated interferon gamma (IFNγ)-expressing T cytotoxic LPLs and fecal albumin and between inflammatory taxa abundance and IFNγ-producing immune cells in the ileum. Bowel decontamination led to redistributed microbiota composition, reduced proinflammatory activation of mucosal immune cells, normalized fecal albumin levels, and diminished GBT; but there were no modifications in Th17 depletion. Conclusion: The intestinal mucosa of rats with cirrhosis acquires a proinflammatory profile of immune dysregulation that parallels the severity of cirrhosis; this impaired intestinal immune response is driven by gut dysbiosis and leads to disrupted barrier function, promoting GBT.


Assuntos
Translocação Bacteriana/imunologia , Disbiose/imunologia , Interferon gama/metabolismo , Mucosa Intestinal/imunologia , Cirrose Hepática/patologia , Imunidade Adaptativa/fisiologia , Animais , Ascite/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Microbioma Gastrointestinal/imunologia , Humanos , Imunidade Inata/fisiologia , Mucosa Intestinal/microbiologia , Cirrose Hepática/microbiologia , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar
8.
BMC Nephrol ; 21(1): 141, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32316931

RESUMO

BACKGROUND: The relationships between digestive bacterial translocation, uremic toxins, oxidative stress and microinflammation in a population of chronic kidney disease (CKD) patients without metabolic nor inflammatory disease are unknown. METHODS: Bacterial translocation, uremic toxins, oxidative stress, and inflammation were assessed by measuring plasma levels of 16S ribosomal DNA (16S rDNA), p-cresyl sulfate (PCS), indoxyl sulfate (IS), indole acetic acid (IAA), F2-isoprostanes, hsCRP and receptor I of TNFα (RITNFα) in patients without metabolic nor inflammatory disease. 44 patients with CKD from stage IIIB to V and 14 controls with normal kidney function were included from the nephrology outpatients. 11 patients under hemodialysis (HD) were also included. Correlations between each factor and microinflammation markers were studied. RESULTS: 16S rDNA levels were not increased in CKD patients compared to controls but were decreased in HD compared to non-HD stage V patients (4.7 (3.9-5.3) vs 8.6 (5.9-9.7) copies/µl, p = 0.002). IS, PCS and IAA levels increased in HD compared to controls (106.3 (73.3-130.4) vs 3.17 (2.4-5.1) µmol/l, p < 0.0001 for IS; 174.2 (125-227.5) vs 23.7 (13.9-52.6) µmol/l, p = 0.006 for PCS; and 3.7 (2.6-4.6) vs 1.3 (1.0-1.9) µmol/l, p = 0.0002 for IAA). Urea increased in non-HD stage V patients compared to controls (27.6 (22.7-30.9) vs 5.4 (4.8-6.4) mmol/l, p < 0.0001) and was similar in HD and in non-HD stage V (19.3 (14.0-24.0) vs 27.6 (22.7-30.9) mmol/l, p = 0.7). RITNFα levels increased in HD patients compared to controls (12.6 (9.6-13.3) vs 1.1 (1.0-1.4) ng/ml, p < 0.0001); hsCRP levels increased in non-HD stage V patients compared to controls (2.9 (1.4-8.5) vs 0.8 (0.5-1.7) mg/l, p = 0.01) and remained stable in HD patients (2.9 (1.4-8.5) vs 5.1 (0.9-11.5) mg/l, p = 1). F2-isoprostanes did not differ in CKD patients compared to controls. Among uremic toxins, IS and urea were correlated to RITNFα (r = 0.8, p < 0.0001 for both). PCS, IS and urea were higher in patients with hsCRP≧5 mg/l (p = 0.01, 0.04 and 0.001 respectively). 16S rDNA, F2-isoprostanes were not correlated to microinflammation markers in our study. CONCLUSIONS: In CKD patients without any associated metabolic nor inflammatory disease, only PCS, IS, and urea were correlated with microinflammation. Bacterial translocation was decreased in patients under HD and was not correlated to microinflammation.


Assuntos
Translocação Bacteriana/imunologia , Biomarcadores/sangue , Microbioma Gastrointestinal/imunologia , Inflamação/metabolismo , Estresse Oxidativo , Diálise Renal/métodos , Insuficiência Renal Crônica , Proteína C-Reativa/análise , Feminino , Humanos , Indicã/sangue , Ácidos Indolacéticos/sangue , Testes de Função Renal/métodos , Masculino , Gravidade do Paciente , Projetos Piloto , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/terapia , Ésteres do Ácido Sulfúrico/sangue , Uremia/diagnóstico , Uremia/etiologia
9.
J Infect Dis ; 219(1): 89-100, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30107546

RESUMO

Objective: Immune activation is associated with morbidity and mortality during human immunodeficiency virus (HIV) infection, despite receipt of antiretroviral therapy (ART). We investigated whether microbial translocation drives immune activation in HIV-infected Ugandan children. Methods: Nineteen markers of immune activation and inflammation were measured over 96 weeks in HIV-infected Ugandan children in the CHAPAS-3 Trial and HIV-uninfected age-matched controls. Microbial translocation was assessed using molecular techniques, including next-generation sequencing. Results: Of 249 children included, 142 were infected with HIV; of these, 120 were ART naive, with a median age of 2.8 years (interquartile range [IQR], 1.7-4.0 years) and a median baseline CD4+ T-cell percentage of 20% (IQR, 14%-24%), and 22 were ART experienced, with a median age of 6.5 years (IQR, 5.9-9.2 years) and a median baseline CD4+ T-cell percentage of 35% (IQR, 31%-39%). The control group comprised 107 children without HIV infection. The median increase in the CD4+ T-cell percentage was 17 percentage points (IQR, 12-22 percentage points) at week 96 among ART-naive children, and the viral load was <100 copies/mL in 76% of ART-naive children and 91% of ART-experienced children. Immune activation decreased with ART use. Children could be divided on the basis of immune activation markers into the following 3 clusters: in cluster 1, the majority of children were HIV uninfected; cluster 2 comprised a mix of HIV-uninfected children and HIV-infected ART-naive or ART-experienced children; and in cluster 3, the majority were ART naive. Immune activation was low in cluster 1, decreased in cluster 3, and persisted in cluster 2. Blood microbial DNA levels were negative or very low across groups, with no difference between clusters except for Enterobacteriaceae organisms (the level was higher in cluster 1; P < .0001). Conclusion: Immune activation decreased with ART use, with marker clustering indicating different activation patterns according to HIV and ART status. Levels of bacterial DNA in blood were low regardless of HIV status, ART status, and immune activation status. Microbial translocation did not drive immune activation in this setting. Clinical Trials Registration: ISRCTN69078957.


Assuntos
Translocação Bacteriana/imunologia , Biomarcadores/sangue , Infecções por HIV/imunologia , Translocação Bacteriana/genética , Contagem de Linfócito CD4 , Criança , Pré-Escolar , DNA Bacteriano/sangue , DNA Ribossômico , Feminino , Infecções por HIV/sangue , Infecções por HIV/tratamento farmacológico , Infecções por HIV/microbiologia , Humanos , Lactente , Inflamação , Masculino , Uganda , Carga Viral
10.
J Neurovirol ; 25(4): 551-559, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31098925

RESUMO

Small intestinal bacterial overgrowth (SIBO) is common among patients with HIV-associated autonomic neuropathies (HIV-AN) and may be associated with increased bacterial translocation and elevated plasma inflammatory biomarkers. Pyridostigmine is an acetylcholinesterase inhibitor which has been used to augment autonomic signaling. We sought preliminary evidence as to whether pyridostigmine could improve proximal gastrointestinal motility, reduce SIBO, reduce plasma sCD14 (a marker of macrophage activation and indirect measure of translocation), and reduce the inflammatory cytokines IL-6 and TNFα in patients with HIV-AN. Fifteen participants with well-controlled HIV, HIV-AN, and SIBO were treated with 8 weeks of pyridostigmine (30 mg PO TID). Glucose breath testing for SIBO, gastric emptying studies (GES) to assess motility, plasma sCD14, IL-6, and TNFα, and gastrointestinal autonomic symptoms were compared before and after treatment. Thirteen participants (87%) experienced an improvement in SIBO following pyridostigmine treatment; with an average improvement of 50% (p = 0.016). There was no change in gastrointestinal motility; however, only two participants met GES criteria for gastroparesis at baseline. TNFα and sCD14 levels declined by 12% (p = 0.004) and 19% (p = 0.015), respectively; there was no significant change in IL-6 or gastrointestinal symptoms. Pyridostigmine may ameliorate SIBO and reduce levels of sCD14 and TNFα in patients with HIV-AN. Larger placebo-controlled studies are needed to definitively delineate how HIV-AN affects gastrointestinal motility, SIBO, and systemic inflammation in HIV, and whether treatment improves clinical outcomes.


Assuntos
Vias Autônomas/efeitos dos fármacos , Inibidores da Colinesterase/uso terapêutico , Infecções por HIV/tratamento farmacológico , Intestino Delgado/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Brometo de Piridostigmina/uso terapêutico , Vias Autônomas/imunologia , Vias Autônomas/microbiologia , Vias Autônomas/patologia , Translocação Bacteriana/efeitos dos fármacos , Translocação Bacteriana/imunologia , Esquema de Medicação , Feminino , Motilidade Gastrointestinal/efeitos dos fármacos , Expressão Gênica , Infecções por HIV/imunologia , Infecções por HIV/microbiologia , Infecções por HIV/patologia , Humanos , Interleucina-6/genética , Interleucina-6/imunologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
11.
Br J Nutr ; 121(4): 361-373, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30554574

RESUMO

Gluten is only partially digested by intestinal enzymes and can generate peptides that can alter intestinal permeability, facilitating bacterial translocation, thus affecting the immune system. Few studies addressed the role of diet with gluten in the development of colitis. Therefore, we investigate the effects of wheat gluten-containing diet on the evolution of sodium dextran sulphate (DSS)-induced colitis. Mice were fed a standard diet without (colitis group) or with 4·5 % wheat gluten (colitis + gluten) for 15 d and received DSS solution (1·5 %, w/v) instead of water during the last 7 d. Compared with the colitis group, colitis + gluten mice presented a worse clinical score, a larger extension of colonic injury area, and increased mucosal inflammation. Both intestinal permeability and bacterial translocation were increased, propitiating bacteria migration for peripheral organs. The mechanism by which diet with gluten exacerbates colitis appears to be related to changes in protein production and organisation in adhesion junctions and desmosomes. The protein α-E-catenin was especially reduced in mice fed gluten, which compromised the localisation of E-cadherin and ß-catenin proteins, weakening the structure of desmosomes. The epithelial damage caused by gluten included shortening of microvilli, a high number of digestive vacuoles, and changes in the endosome/lysosome system. In conclusion, our results show that wheat gluten-containing diet exacerbates the mucosal damage caused by colitis, reducing intestinal barrier function and increasing bacterial translocation. These effects are related to the induction of weakness and disorganisation of adhesion junctions and desmosomes as well as shortening of microvilli and modification of the endocytic vesicle route.


Assuntos
Translocação Bacteriana/imunologia , Colite/imunologia , Dieta/efeitos adversos , Glutens/efeitos adversos , Junções Íntimas/imunologia , Animais , Colite/induzido quimicamente , Colite/microbiologia , Colo , Sulfato de Dextrana , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade , Triticum/química
12.
Pediatr Nephrol ; 34(1): 45-59, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29497824

RESUMO

Acute kidney injury (AKI) is a common accompaniment in patients with liver disease. The causes, risk factors, manifestations and management of AKI in these patients vary according to the liver disease in question (acute liver failure, acute-on-chronic liver failure, post-liver transplantation or metabolic liver disease). There are multiple causes of AKI in patients with liver disease-pre-renal, acute tubular necrosis, post-renal, drug-induced renal failure and hepatorenal syndrome (HRS). Definitions of AKI in liver failure are periodically revised and updated, but pediatric definitions have still to see the light of the day. As our understanding of the pathophysiology of liver disease and renal involvement improves, treatment modalities have become more advanced and rationalized. Treatment includes reversing precipitating factors, such as infections and gastrointestinal bleeding, volume expansion, paracentesis and vasoconstrictors. This approach is tried and tested in adults. A pediatric tailored approach is still lacking due to inadequate numbers of patients, differences in causes of AKI and paucity of literature. In this review, we attempt to explore the pathophysiological basis, treatment modalities and controversies in the diagnosis and treatment of AKI in pediatric patients with chronic liver disease and discuss our own personal practice. We recognize that, although it is not a very commonly encountered entity in pediatric population, HRS has specific diagnostic criteria and treatment modalities that differ from other causes of AKI in patients with chronic liver disease; hence among the etiologies of kidney injury in patients with chronic liver disease, we focus here on HRS.


Assuntos
Injúria Renal Aguda/etiologia , Síndrome Hepatorrenal/etiologia , Cirrose Hepática/complicações , Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/epidemiologia , Injúria Renal Aguda/terapia , Antibacterianos/uso terapêutico , Antibioticoprofilaxia/métodos , Antagonistas dos Receptores de Hormônios Antidiuréticos/uso terapêutico , Translocação Bacteriana/efeitos dos fármacos , Translocação Bacteriana/imunologia , Biomarcadores/análise , Criança , Síndrome Hepatorrenal/diagnóstico , Síndrome Hepatorrenal/epidemiologia , Síndrome Hepatorrenal/terapia , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Transplante de Rim , Cirrose Hepática/imunologia , Cirrose Hepática/terapia , Transplante de Fígado , Paracentese , Terapia de Substituição Renal/métodos , Fatores de Risco , Vasoconstritores/uso terapêutico
13.
J Autoimmun ; 95: 47-57, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30340822

RESUMO

Gut microbiota and bacterial translocation have been implicated as significant contributors to mucosal immune responses and tolerance; alteration of microbial molecules, termed pathogen-associated molecular patterns (PAMP) and bacterial translocation are associated with immune pathology. However, the mechanisms by which dysregulated gut microbiota promotes autoimmunity is unclear. We have taken advantage of a well-characterized murine model of primary biliary cholangitis, dnTGFßRII mice, and an additional unique construct, toll-like receptor 2 (TLR2)-deficient dnTGFßRII mice coined dnTGFßRIITLR2-/- mice to investigate the influences of gut microbiota on autoimmune cholangitis. Firstly, we report that dnTGFßRII mice manifest altered composition of gut microbiota and that alteration of this gut microbiota by administration of antibiotics significantly alleviates T-cell-mediated infiltration and bile duct damage. Second, toll-like receptor 2 (TLR2)-deficient dnTGFßRII mice demonstrate significant exacerbation of autoimmune cholangitis when their epithelial barrier integrity was disrupted. Further, TLR2-deficiency mediates downregulated expression of tight junction-associated protein ZO-1 leading to increased gut permeability and bacterial translocation from gut to liver; use of antibiotics reduces microbiota translocation to liver and also decreases biliary pathology. In conclusion, our data demonstrates the important role of gut microbiota and bacterial translocation in the pathogenesis of murine autoimmune cholangitis.


Assuntos
Doenças Autoimunes/microbiologia , Translocação Bacteriana/imunologia , Ductos Biliares/imunologia , Cirrose Hepática Biliar/imunologia , Receptor do Fator de Crescimento Transformador beta Tipo II/imunologia , Receptor 2 Toll-Like/imunologia , Ampicilina/farmacologia , Animais , Antibacterianos/farmacologia , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/patologia , Translocação Bacteriana/efeitos dos fármacos , Ductos Biliares/efeitos dos fármacos , Ductos Biliares/microbiologia , Ductos Biliares/patologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/microbiologia , Colo/patologia , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Regulação da Expressão Gênica , Imunidade nas Mucosas/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Cirrose Hepática Biliar/tratamento farmacológico , Cirrose Hepática Biliar/microbiologia , Cirrose Hepática Biliar/patologia , Metronidazol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neomicina/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo II/deficiência , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Transdução de Sinais , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/imunologia
14.
J Surg Res ; 228: 14-19, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29907203

RESUMO

BACKGROUND: Obstructive jaundice (OJ) patients with cholangitis are prone to sepsis; however, the underlying mechanisms are still not clear and need to be clarified. METHODS: Analyzing all available published data related to the title of this article. RESULTS: OJ leads to absence of gut luminal bile and accumulation of hepatic and circulating bile acids. Absence of gut luminal bile deprives the gut from its antiinflammatory, endotoxin-binding, bacteriostatic, mucosal-trophic, epithelial tight-junction maintaining, and gut motility-regulating effects, leading to gut bacterial overgrowth, mucosal atrophy, mucosal tight-junction loss, and gut motility dysfunction. These alterations promote intestinal endotoxin and bacterial translocation (BT) into portal and systemic circulation. Gut BT triggers systemic inflammation, which can lead to multiple organ dysfunctions in OJ. The accumulation of hepatic and circulating bile acids kills/damages hepatocyte and Kupffer cells, and it also significantly decreases the number of liver natural killer T-cells in OJ. This results in impaired hepatic and systemic immune function, which facilitates BT. In addition, neutralizing bile HMGB1 can reverse endotoxemic bile-induced gut BT and mucosal injury in mice, suggesting that bile HMGB1 in OJ patients can be responsible for internal drainage-related clinical complications. Moreover, the elevated circulating HMGB1 level may contribute to multiple organ injuries, and it might also mediate gut BT in OJ. CONCLUSIONS: HMGB1 may significantly contribute to systemic inflammation and multiple organ dysfunctions in OJ.


Assuntos
Bile/imunologia , Colangite/imunologia , Proteína HMGB1/imunologia , Icterícia Obstrutiva/imunologia , Sepse/imunologia , Animais , Translocação Bacteriana/imunologia , Colangite/sangue , Colangite/microbiologia , Modelos Animais de Doenças , Endotoxinas/imunologia , Microbioma Gastrointestinal/imunologia , Proteína HMGB1/sangue , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Icterícia Obstrutiva/sangue , Icterícia Obstrutiva/complicações , Sepse/sangue , Sepse/microbiologia
15.
Georgian Med News ; (274): 152-158, 2018 Jan.
Artigo em Russo | MEDLINE | ID: mdl-29461245

RESUMO

This study investigates the morphological structure of lungs, liver and kidneys of the experimental animals. To reproduce the inflammation in maxilla-facial region under the periosteum of guinea pigs' mandible a suspension of S.aureus was inoculated. The infected site was exposed by a constant electric current of positive polarity of 5-10 µA strength. Allergotests were used to determine the level of sensitization. Structural changes in the organs were determined by microscopic examination. The results of the study showed that as the contact with the antigen increased, the number of positive allergic tests also increased. Histologically, a generalized damage to the microstructure of the internal organs, depending on the degree of bacterial sensitization of the animals was observed.


Assuntos
Rim/imunologia , Fígado/imunologia , Pulmão/imunologia , Maxila/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Alérgenos/farmacologia , Animais , Antígenos de Bactérias/farmacologia , Translocação Bacteriana/imunologia , Estimulação Elétrica , Cobaias , Imunização/métodos , Rim/microbiologia , Rim/patologia , Rim/ultraestrutura , Fígado/microbiologia , Fígado/patologia , Fígado/ultraestrutura , Pulmão/microbiologia , Pulmão/patologia , Pulmão/ultraestrutura , Masculino , Maxila/microbiologia , Maxila/patologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/patogenicidade
16.
Clin Immunol ; 183: 213-224, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28286112

RESUMO

Individuals with multiple sclerosis (MS) have a distinct intestinal microbial community (microbiota) and increased low-grade translocation of bacteria from the intestines into the circulation. The observed change of intestinal bacteria in MS patients regulate immune functions involved in MS pathogenesis. These functions include: systemic and central nervous system (CNS) immunity (including peripheral regulatory T cell function), the blood-brain barrier (BBB) permeability and CNS-resident cell activity. This review discusses the MS intestinal microbiota implication on MS systemic- and CNS-immunopathology. We introduce the possible contributions of MS low-grade microbial translocation (LG-MT) to the development of MS, and end on a discussion on microbiota therapies for MS patients.


Assuntos
Bactérias/classificação , Translocação Bacteriana/fisiologia , Microbioma Gastrointestinal/fisiologia , Esclerose Múltipla/microbiologia , Translocação Bacteriana/imunologia , Humanos , Esclerose Múltipla/imunologia
17.
Metab Brain Dis ; 32(5): 1571-1583, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28600633

RESUMO

Immunoglubulin (Ig)M responses directed to oxidative specific epitopes (OSEs) and nitric oxide (NO)-adducts are significantly associated with major depression and physio-somatic symptoms. End of term serum IgM responses to OSEs and NO-adducts were assayed in pregnant women with (n = 24) and without prenatal depression (n = 25) as well as in 24 non-pregnant women. Associations of IgM/IgA responses to Gram-negative gut commensal bacteria (leaky gut index) and IgA/IgM responses to tryptophan catabolites (TRYCATs) were analyzed. IgM responses to OSEs, but not NO-adducts, were significantly reduced at the end of term. There were no significant associations between IgM responses to OSEs and perinatal depression, whilst IgM responses to NO-adducts, especially NO-cysteinyl, were significantly associated with a lifetime major depression. IgM responses to OSEs and NO-cysteinyl were significantly associated with IgA/IgM responses to Gram-negative bacteria, especially Morganella morganii, Klebsiella pneumoniae and Citrobacter koseri. IgM responses to NO-adducts and OSEs, especially malondialdehyde and myristic acid, and C-reactive protein (CRP) were inversely associated with TRYCAT pathway activity, whilst a lifetime depression and Pseudomonas putida were positively associated. The attenuation of natural IgM-mediated responses to OSEs at the end of term may indicate lowered activity of this part of the compensatory (anti-)inflammatory reflex system and may be partly explained by lowered bacterial translocation. Increased IgM responses to NO-cysteinyl is a biomarker of lifetime depression and may be induced by bacterial translocation. Natural IgM-mediated autoimmune responses, increased nitrosylation and higher CRP levels may have negative regulatory effects on the TRYCAT pathway.


Assuntos
Autoimunidade/imunologia , Translocação Bacteriana/imunologia , Transtorno Depressivo Maior/imunologia , Epitopos/imunologia , Imunoglobulina M/imunologia , Gravidez/imunologia , Triptofano/metabolismo , Adulto , Biomarcadores , Citocinas/metabolismo , Depressão Pós-Parto/imunologia , Depressão Pós-Parto/psicologia , Transtorno Depressivo Maior/epidemiologia , Transtorno Depressivo Maior/psicologia , Feminino , Bactérias Gram-Negativas/imunologia , Humanos , Inflamação/imunologia , Cinurenina/metabolismo , Redes e Vias Metabólicas , Óxido Nítrico/metabolismo , Adulto Jovem
18.
Gut ; 65(7): 1100-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26045138

RESUMO

OBJECTIVE: Antibiotic use is associated with an increased risk of developing multiple inflammatory disorders, which in turn are linked to alterations in the intestinal microbiota. How these alterations in the intestinal microbiota translate into an increased risk for inflammatory responses is largely unknown. Here we investigated whether and how antibiotics promote inflammation via the translocation of live native gut commensal bacteria. DESIGN: Oral antibiotics were given to wildtype and induced mutant mouse strains, and the effects on bacterial translocation, inflammatory responses and the susceptibility to colitis were evaluated. The sources of the bacteria and the pathways required for bacterial translocation were evaluated using induced mutant mouse strains, 16s rRNA sequencing to characterise the microbial communities, and in vivo and ex vivo imaging techniques. RESULTS: Oral antibiotics induced the translocation of live native commensal bacteria across the colonic epithelium, promoting inflammatory responses, and predisposing to increased disease in response to coincident injury. Bacterial translocation resulted from decreased microbial signals delivered to colonic goblet cells (GCs), was associated with the formation of colonic GC-associated antigen passages, was abolished when GCs were depleted and required CX3CR1(+) dendritic cells. Bacterial translocation occurred following a single dose of most antibiotics tested, and the predisposition for increased inflammation was only associated with antibiotics inducing bacterial translocation. CONCLUSIONS: These findings reveal an unexpected outcome of antibiotic therapy and suggest that bacterial translocation as a result of alterations in the intestinal microflora may provide a link between increasing antibiotic use and the increased incidence of inflammatory disorders.


Assuntos
Antibacterianos/farmacologia , Translocação Bacteriana/efeitos dos fármacos , Colo/microbiologia , Células Caliciformes/metabolismo , Células Caliciformes/microbiologia , Inflamação/microbiologia , Linfonodos/microbiologia , Animais , Translocação Bacteriana/imunologia , Receptor 1 de Quimiocina CX3C , Colite/microbiologia , Citocinas/metabolismo , Células Dendríticas/química , Suscetibilidade a Doenças , Enterococcus faecalis/isolamento & purificação , Escherichia coli/isolamento & purificação , Células Caliciformes/imunologia , Intestino Delgado/microbiologia , Mesentério , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide , Receptores de Quimiocinas/análise
19.
Semin Liver Dis ; 36(4): 317-326, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27997971

RESUMO

The intestinal immune system recognizes and responds to the vast diversity of microbes present within the gut. Highly sophisticated cellular and molecular networks are continuously coordinated to tolerate the presence of a large number and diversity of bacteria on mucosal surfaces. Different types of bacteria induce different immune responses, and bacterial metabolism of dietary factors generates metabolites that have significant effects on host immune responses. Dendritic cells, epithelial cells, innate lymphoid cells, T-regulatory cells, effector lymphocytes, natural killer T cells, and B-cell responses can all be influenced by the microbiome. Many of the mechanisms being described are bacterial strain or metabolite-specific. A better understanding of the mechanisms governing microbiome-host immune responses will likely lead to novel therapeutics for inflammatory disorders.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunidade Inata/imunologia , Mucosa Intestinal/imunologia , Animais , Translocação Bacteriana/imunologia , Biomarcadores/análise , Células Epiteliais/imunologia , Humanos , Imunidade Celular , Mucosa Intestinal/microbiologia
20.
Semin Liver Dis ; 36(4): 327-330, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27997972

RESUMO

Chronic liver disease with progression to decompensated cirrhosis and its associated complications, including hepatic encephalopathy, spontaneous bacterial peritonitis, and sepsis, is a leading cause of mortality and morbidity. The pathophysiology of decompensated cirrhosis, which is being intensively studied, leads to the development of gut microbiome changes causing dysbiosis. This is likely related to altered bile acid composition, with a subsequent increase in the relative abundance of potentially pathogenic bacteria that contributes to hepatic encephalopathy and leads to their translocation and the development of spontaneous bacterial peritonitis and bacteremia. Treatments for these conditions have been found to target the gut microbiome, which has become a vital area of study in the treatment of cirrhosis.


Assuntos
Translocação Bacteriana/imunologia , Microbioma Gastrointestinal , Encefalopatia Hepática/etiologia , Cirrose Hepática/complicações , Peritonite/complicações , Sepse/etiologia , Animais , Encefalopatia Hepática/microbiologia , Encefalopatia Hepática/terapia , Humanos , Fígado/microbiologia , Cirrose Hepática/microbiologia , Cirrose Hepática/terapia , Peritonite/microbiologia , Inibidores da Bomba de Prótons/efeitos adversos , Sepse/microbiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa