Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 4697, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170216

ABSTRACT

Previous studies demonstrate an association between activation of the maternal immune system during pregnancy and increased risk of neurodevelopmental psychiatric conditions, such as schizophrenia and autism, in the offspring. Relatively recent findings also suggest that the gut microbiota plays an important role in shaping brain development and behavior. Here we show that maternal immune activation (MIA) accomplished by infection with a mouse-adapted influenza virus during pregnancy induced up-regulation of frontal cortex serotonin 5-HT2A receptor (5-HT2AR) density in the adult offspring, a phenotype previously observed in postmortem frontal cortex of schizophrenic subjects. 5-HT2AR agonist-induced head-twitch behavior was also augmented in this preclinical mouse model. Using the novel object recognition (NOR) test to evaluate cognitive performance, we demonstrate that MIA induced NOR deficits in adult offspring. Oral antibiotic treatment of prepubertal mice prevented this cognitive impairment, but not increased frontal cortex 5-HT2AR density or psychedelic-induced head-twitch behavior in adult MIA offspring. Additionally, gut microbiota transplantation from MIA mice produced behavioral deficits in antibiotic-treated mock mice. Adult MIA offspring displayed altered gut microbiota, and relative abundance of specific components of the gut microbiota, including Ruminococcaceae, correlated with frontal cortex 5-HT2AR density. Together, these findings provide a better understanding of basic mechanisms by which prenatal insults impact offspring brain function, and suggest gut-brain axis manipulation as a potential therapeutic approach for neurodevelopmental psychiatric conditions.


Subject(s)
Behavior, Animal , Disease Susceptibility , Gastrointestinal Microbiome , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/psychology , Problem Behavior , Sexual Maturation , Age Factors , Animals , Anti-Bacterial Agents/administration & dosage , Disease Models, Animal , Memory , Mice , Phenotype , Recognition, Psychology , Schizophrenia/etiology
2.
Alcohol Clin Exp Res ; 41(11): 1857-1865, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28925102

ABSTRACT

BACKGROUND: Cirrhosis and alcohol can independently affect the gut-liver axis with systemic inflammation. However, their concurrent impact in humans is unclear. METHODS: Our aim was to determine the effect of continued alcohol misuse on the gut-liver axis in cirrhotic patients. Age- and MELD-balanced cirrhotic patients who were currently drinking (Alc) or abstinent (NAlc) and healthy controls underwent serum and stool collection. A subset underwent upper endoscopy and colonoscopy for biopsies and duodenal fluid collection. The groups were compared regarding (i) inflammation/intestinal barrier: systemic tumor necrosis factor levels, intestinal inflammatory cytokine (duodenum, ileum, sigmoid), and ileal antimicrobial peptide expression; (ii) microbiota composition: 16SrRNA sequencing of duodenal, ileal, and colonic mucosal and fecal microbiota; and (iii) microbial functionality: duodenal fluid and fecal bile acid (BA) profile (conjugation and dehydroxylation status), intestinal BA transporter (ASBT, FXR, FGF-19, SHP) expression, and stool metabolomics using gas chromatography/mass spectrometry. RESULTS: Alc patients demonstrated a significant duodenal, ileal, and colonic mucosal and fecal dysbiosis, compared to NAlc and controls with lower autochthonous bacterial taxa. BA profile skewed toward a potentially toxic profile (higher secondary and glycine-conjugated BAs) in duodenal fluid and stool in Alc patients. Duodenal fluid demonstrated conjugated secondary BAs only in the Alc group. There was a greater expression of all ileal BA transporters in Alc patients. This group also showed higher endotoxemia, systemic and ileal inflammatory expression, and lower amino acid and bioenergetic-associated metabolites, without change in antimicrobial peptide expression. CONCLUSIONS: Despite cirrhosis, continued alcohol misuse predisposes patients to widespread dysbiosis with alterations in microbial functionality such as a toxic BA profile, which can lead to intestinal and systemic inflammation.


Subject(s)
Alcoholism/physiopathology , Dysbiosis/physiopathology , Gastrointestinal Tract/physiopathology , Liver Cirrhosis/physiopathology , Alcoholism/diagnosis , Alcoholism/epidemiology , Dysbiosis/diagnosis , Dysbiosis/epidemiology , Endoscopy, Digestive System/methods , Female , Humans , Liver Cirrhosis/diagnosis , Liver Cirrhosis/epidemiology , Male , Microbiota/physiology , Middle Aged
3.
Hepatol Commun ; 1(1): 61-70, 2017 02.
Article in English | MEDLINE | ID: mdl-29404434

ABSTRACT

We previously reported that alcohol drinkers with and without cirrhosis showed a significant increase in fecal bile acid secretion compared to nondrinkers. We hypothesized this may be due to activation by alcohol of hepatic cyclic adenosine monophosphate responsive element-binding protein 3-like protein 3 (CREBH), which induces cholesterol 7α-hydroxylase (Cyp7a1). Alternatively, the gut microbiota composition in the absence of alcohol might increase bile acid synthesis by up-regulating Cyp7a1. To test this hypothesis, we humanized germ-free (GF) mice with stool from healthy human subjects (Ctrl-Hum), human subjects with cirrhosis (Cirr-Hum), and human subjects with cirrhosis and active alcoholism (Alc-Hum). All animals were fed a normal chow diet, and none demonstrated cirrhosis. Both hepatic Cyp7a1 and sterol 12α-hydroxylase (Cyp8b1) messenger RNA (mRNA) levels were significantly induced in the Alc-Hum and Ctrl-Hum mice but not in the Cirr-Hum mice or GF mice. Liver bile acid concentration was correspondingly increased in the Alc-Hum mice despite fibroblast growth factor 15, fibroblast growth receptor 4, and small heterodimer partner mRNA levels being significantly induced in the large bowel and liver of the Ctrl-Hum mice and Alc-Hum mice but not in the Cirr-Hum mice or GF mice. This suggests that the normal pathways of Cyp7a1 repression were activated in the Alc-Hum mice and Ctrl-Hum mice. CREBH mRNA was significantly induced only in the Ctrl-Hum mice and Alc-Hum mice, possibly indicating that the gut microbiota up-regulate CREBH and induce bile acid synthesis genes. Analysis of stool bile acids showed that the microbiota of the Cirr-Hum and Alc-Hum mice had a greater ability to deconjugate and 7α-dehydroxylate primary bile acids compared to the microbiota of the Cirr-Hum mice. 16S ribosomal RNA gene sequencing of the gut microbiota showed that the relative abundance of taxa that 7-α dehydroxylate primary bile acids was higher in the Ctrl-Hum and Alc-Hum groups. Conclusion: The composition of gut microbiota influences the regulation of the rate-limiting enzymes in bile acid synthesis in the liver. (Hepatology Communications 2017;1:61-70).

4.
Hepatology ; 64(4): 1232-48, 2016 10.
Article in English | MEDLINE | ID: mdl-27339732

ABSTRACT

UNLABELLED: The mechanisms behind the development of hepatic encephalopathy (HE) are unclear, although hyperammonemia and systemic inflammation through gut dysbiosis have been proposed. The aim of this work was to define the individual contribution of hyperammonemia and systemic inflammation on neuroinflammation in cirrhosis using germ-free (GF) and conventional mice. GF and conventional C57BL/6 mice were made cirrhotic using CCl4 gavage. These were compared to their noncirrhotic counterparts. Intestinal microbiota, systemic and neuroinflammation (including microglial and glial activation), serum ammonia, intestinal glutaminase activity, and cecal glutamine content were compared between groups. GF cirrhotic mice developed similar cirrhotic changes to conventional mice after 4 extra weeks (16 vs. 12 weeks) of CCl4 gavage. GF cirrhotic mice exhibited higher ammonia, compared to GF controls, but this was not associated with systemic or neuroinflammation. Ammonia was generated through increased small intestinal glutaminase activity with concomitantly reduced intestinal glutamine levels. However, conventional cirrhotic mice had intestinal dysbiosis as well as systemic inflammation, associated with increased serum ammonia, compared to conventional controls. This was associated with neuroinflammation and glial/microglial activation. Correlation network analysis in conventional mice showed significant linkages between systemic/neuroinflammation, intestinal microbiota, and ammonia. Specifically beneficial, autochthonous taxa were negatively linked with brain and systemic inflammation, ammonia, and with Staphylococcaceae, Lactobacillaceae, and Streptococcaceae. Enterobacteriaceae were positively linked with serum inflammatory cytokines. CONCLUSION: Gut microbiota changes drive development of neuroinflammatory and systemic inflammatory responses in cirrhotic animals. (Hepatology 2016;64:1232-1248).


Subject(s)
Gastrointestinal Microbiome/physiology , Liver Cirrhosis/etiology , Animals , Hyperammonemia/etiology , Inflammation/etiology , Mice , Mice, Inbred C57BL , Neuroglia
5.
Gut Microbes ; 7(1): 22-39, 2016.
Article in English | MEDLINE | ID: mdl-26939849

ABSTRACT

Emerging evidence strongly suggest that the human "microbiome" plays an important role in both health and disease. Bile acids function both as detergents molecules promoting nutrient absorption in the intestines and as hormones regulating nutrient metabolism. Bile acids regulate metabolism via activation of specific nuclear receptors (NR) and G-protein coupled receptors (GPCRs). The circulating bile acid pool composition consists of primary bile acids produced from cholesterol in the liver, and secondary bile acids formed by specific gut bacteria. The various biotransformation of bile acids carried out by gut bacteria appear to regulate the structure of the gut microbiome and host physiology. Increased levels of secondary bile acids are associated with specific diseases of the GI system. Elucidating methods to control the gut microbiome and bile acid pool composition in humans may lead to a reduction in some of the major diseases of the liver, gall bladder and colon.


Subject(s)
Bacteria/metabolism , Bile Acids and Salts/metabolism , Energy Metabolism , Gastrointestinal Microbiome/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acids/metabolism , Diet , Humans
7.
Hepatology ; 63(5): 1736-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26105621
8.
Sci Rep ; 5: 18559, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26692421

ABSTRACT

Diabetes (DM) is prevalent in cirrhosis and may modulate the risk of hospitalization through gut dysbiosis. We aimed to define the role of gut microbiota on 90-day hospitalizations and of concomitant DM on microbiota. Cirrhotic outpatients with/without DM underwent stool and sigmoid mucosal microbial analysis and were followed for 90 days. Microbial composition was compared between those with/without DM, and those who were hospitalized/not. Regression/ROC analyses for hospitalizations were performed using clinical and microbial features. 278 cirrhotics [39% hepatic encephalopathy (HE), 31%DM] underwent stool while 72 underwent mucosal analyses. Ultimately, 94 were hospitalized and they had higher MELD, proton pump inhibitor (PPI) use and HE without difference in DM. Stool/mucosal microbiota were significantly altered in those who were hospitalized (UNIFRAC p < = 1.0e-02). Specifically, lower stool Bacteroidaceae, Clostridiales XIV, Lachnospiraceae, Ruminococcacae and higher Enterococcaceae and Enterobacteriaceae were seen in hospitalized patients. Concomitant DM impacted microbiota UNIFRAC (stool, p = 0.003, mucosa, p = 0.04) with higher stool Bacteroidaceae and lower Ruminococcaeae. Stool Bacteroidaceaeae and Clostridiales XIV predicted 90-day hospitalizations independent of clinical predictors (MELD, HE, PPI). Stool and colonic mucosal microbiome are altered in cirrhotics who get hospitalized with independent prediction using stool Bacteroidaceae and Clostridiales XIV. Concomitant DM distinctly impacts gut microbiota without affecting hospitalizations.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome , Hospitalization , Liver Cirrhosis/complications , Liver Cirrhosis/microbiology , Demography , Feces/microbiology , Female , Humans , Intestinal Mucosa/microbiology , Logistic Models , Male , Middle Aged , Principal Component Analysis , Treatment Outcome
10.
Dig Dis ; 33(3): 338-45, 2015.
Article in English | MEDLINE | ID: mdl-26045267

ABSTRACT

The understanding of the complex role of the bile acid-gut microbiome axis in health and disease processes is evolving rapidly. Our focus revolves around the interaction of the gut microbiota with liver diseases, especially cirrhosis. The bile acid pool size has recently been shown to be a function of microbial metabolism of bile acid, and regulation of the microbiota by bile acids is important in the development and progression of several liver diseases. Humans produce a large, conjugated hydrophilic bile acid pool, maintained through positive-feedback antagonism of farnesoid X receptor (FXR) in the intestine and liver. Microbes use bile acids, and via FXR signaling this results in a smaller, unconjugated hydrophobic bile acid pool. This equilibrium is critical to maintain health. The challenge is to examine the manifold functions of gut bile acids as modulators of antibiotic, probiotic, and disease progression in cirrhosis, metabolic syndrome, and alcohol use. Recent studies have shown potential mechanisms explaining how perturbations in the microbiome affect bile acid pool size and composition. With advancing liver disease and cirrhosis, there is dysbiosis in the fecal, ileal, and colonic mucosa, in addition to a decrease in bile acid concentration in the intestine due to the liver problems. This results in a dramatic shift toward the Firmicutes, particularly Clostridium cluster XIVa, and increasing production of deoxycholic acid. Alcohol intake speeds up these processes in the subjects with and without cirrhosis without significant FXR feedback. Taken together, these pathways can impact intestinal and systemic inflammation while worsening dysbiosis. The interaction between bile acids, alcohol, cirrhosis, and dysbiosis is an important relationship that influences intestinal and systemic inflammation, which in turn determines progression of the overall disease process. These interactions and the impact of commonly used therapies for liver disease can provide insight into the pathogenesis of inflammation in humans.


Subject(s)
Alcohol Drinking/metabolism , Bile Acids and Salts/metabolism , Gastrointestinal Microbiome , Liver Cirrhosis/metabolism , Alcohol Drinking/adverse effects , Bile Acids and Salts/analysis , Dysbiosis/etiology , Dysbiosis/metabolism , Dysbiosis/prevention & control , Ethanol/pharmacology , Feces/chemistry , Gastrointestinal Microbiome/drug effects , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/therapy , Probiotics/administration & dosage , Signal Transduction
11.
Hepatology ; 62(4): 1260-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25820757

ABSTRACT

UNLABELLED: Altered gut microbiome is associated with systemic inflammation and cirrhosis decompensation. However, the correlation of the oral microbiome with inflammation in cirrhosis is unclear. Our aim was to evaluate the oral microbiome in cirrhosis and compare with stool microbiome. Outpatients with cirrhosis (with/without hepatic encephalopathy [HE]) and controls underwent stool/saliva microbiome analysis (for composition and function) and also systemic inflammatory evaluation. Ninety-day liver-related hospitalizations were recorded. Salivary inflammation was studied using T helper 1 cytokines/secretory immunoglobulin A (IgA), histatins and lysozyme in a subsequent group. A total of 102 patients with cirrhosis (43 previous HE) and 32 age-matched controls were included. On principal component analysis (PCA), stool and saliva microbiome clustered far apart, showing differences between sites as a whole. In salivary microbiome, with previous HE, relative abundance of autochthonous families decreased whereas potentially pathogenic ones (Enterobacteriaceae, Enterococcaceae) increased in saliva. Endotoxin-related predicted functions were significantly higher in cirrhotic saliva. In stool microbiome, relative autochthonous taxa abundance reduced in previous HE, along with increased Enterobacteriaceae and Enterococcaceae. Cirrhotic stool microbiota demonstrated a significantly higher correlation with systemic inflammation, compared to saliva microbiota, on correlation networks. Thirty-eight patients were hospitalized within 90 days. Their salivary dysbiosis was significantly worse and predicted this outcome independent of cirrhosis severity. Salivary inflammation was studied in an additional 86 age-matched subjects (43 controls/43 patients with cirrhosis); significantly higher interleukin (IL)-6/IL-1ß, secretory IgA, and lower lysozyme, and histatins 1 and 5 were found in patients with cirrhosis, compared to controls. CONCLUSIONS: Dysbiosis, represented by reduction in autochthonous bacteria, is present in both saliva and stool in patients with cirrhosis, compared to controls. Patients with cirrhosis have impaired salivary defenses and worse inflammation. Salivary dysbiosis was greater in patients with cirrhosis who developed 90-day hospitalizations. These findings could represent a global mucosal-immune interface change in cirrhosis.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome , Hepatic Encephalopathy/microbiology , Intestines/microbiology , Liver Cirrhosis/microbiology , Saliva/microbiology , Dysbiosis , Female , Hepatic Encephalopathy/complications , Humans , Inflammation/microbiology , Liver Cirrhosis/complications , Male , Microbiota , Middle Aged
12.
Infect Immun ; 82(6): 2637-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24711565

ABSTRACT

Prevotella intermedia is an oral bacterium implicated in a variety of oral diseases. Although internalization of this bacterium by nonphagocytic host cells is well established, the molecular players mediating the process are not well known. Here, the properties of a leucine-rich repeat (LRR) domain protein, designated AdpF, are described. This protein contains a leucine-rich region composed of 663 amino acid residues, and molecular modeling shows that it folds into a classical curved solenoid structure. The cell surface localization of recombinant AdpF (rAdpF) was confirmed by electron and confocal microscopy analyses. The recombinant form of this protein bound fibronectin in a dose-dependent manner. Furthermore, the protein was internalized by host cells, with the majority of the process accomplished within 30 min. The internalization of rAdpF was inhibited by nystatin, cytochalasin, latrunculin, nocodazole, and wortmannin, indicating that microtubules, microfilaments, and signal transduction are required for the invasion. It is noteworthy that preincubation of eukaryotic cells with AdpF increased P. intermedia 17 internalization by 5- and 10-fold for HeLa and NIH 3T3 fibroblast cell lines, respectively. The addition of the rAdpF protein was also very effective in inducing bacterial internalization into the oral epithelial cell line HN4, as well as into primary cells, including human oral keratinocytes (HOKs) and human umbilical vein endothelial cells (HUVECs). Finally, cells exposed to P. intermedia 17 internalized the bacteria more readily upon reinfection. Taken together, our data demonstrate that rAdpF plays a role in the internalization of P. intermedia 17 by a variety of host cells.


Subject(s)
Bacterial Proteins/physiology , Eukaryotic Cells/microbiology , Prevotella intermedia/physiology , Proteins/physiology , Analysis of Variance , Fibroblasts/microbiology , Fibronectins/metabolism , Gene Expression Regulation, Bacterial , HeLa Cells/microbiology , Humans , Leucine-Rich Repeat Proteins , Prevotella intermedia/genetics , Prevotella intermedia/pathogenicity
13.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G929-37, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24699327

ABSTRACT

Alcohol abuse with/without cirrhosis is associated with an impaired gut barrier and inflammation. Gut microbiota can transform primary bile acids (BA) to secondary BAs, which can adversely impact the gut barrier. The purpose of this study was to define the effect of active alcohol intake on fecal BA levels and ileal and colonic inflammation in cirrhosis. Five age-matched groups {two noncirrhotic (control and drinkers) and three cirrhotic [nondrinkers/nonalcoholics (NAlc), abstinent alcoholic for >3 mo (AbsAlc), currently drinking (CurrAlc)]} were included. Fecal and serum BA analysis, serum endotoxin, and stool microbiota using pyrosequencing were performed. A subgroup of controls, NAlc, and CurrAlc underwent ileal and sigmoid colonic biopsies on which mRNA expression of TNF-α, IL-1ß, IL-6, and cyclooxygenase-2 (Cox-2) were performed. One hundred three patients (19 healthy, 6 noncirrhotic drinkers, 10 CurrAlc, 38 AbsAlc, and 30 NAlc, age 56 yr, median MELD: 10.5) were included. Five each of healthy, CurrAlc, and NAlc underwent ileal/colonic biopsies. Endotoxin, serum-conjugated DCA and stool total BAs, and secondary-to-primary BA ratios were highest in current drinkers. On biopsies, a significantly higher mRNA expression of TNF-α, IL-1ß, IL-6, and Cox-2 in colon but not ileum was seen in CurrAlc compared with NAlc and controls. Active alcohol use in cirrhosis is associated with a significant increase in the secondary BA formation compared with abstinent alcoholic cirrhotics and nonalcoholic cirrhotics. This increase in secondary BAs is associated with a significant increase in expression of inflammatory cytokines in colonic mucosa but not ileal mucosa, which may contribute to alcohol-induced gut barrier injury.


Subject(s)
Alcoholism/complications , Bile Acids and Salts/metabolism , Colonic Diseases/chemically induced , Inflammation/etiology , Liver Cirrhosis/complications , Bile Acids and Salts/chemistry , Colonic Diseases/pathology , Feces/chemistry , Humans , Inflammation/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Middle Aged
14.
PLoS One ; 7(8): e42694, 2012.
Article in English | MEDLINE | ID: mdl-22880085

ABSTRACT

A disintegrin and metalloprotease 10 (ADAM10) is a key regulator of cellular processes by shedding extracellular domains of transmembrane proteins. We have previously demonstrated that deletion of B cell expressed ADAM10 results in changes in lymphoid tissue architecture and impaired germinal center (GC) formation. In this study, mice were generated in which ADAM10 is deleted in B cells following class switch recombination (ADAM10(Δ/Δ)IgG1-cre(+/-) mice). Despite normal GC formation, antibody responses were impaired in ADAM10(Δ/Δ)IgG1-cre(+/-) mice, implicating ADAM10 in post-GC and extrafollicular B cell terminal differentiation. Surprisingly, plasma cell (PC) numbers were normal in ADAM10(Δ/Δ)IgG1-cre(+/-) mice when compared to controls. However, PCs isolated from ADAM10(Δ/Δ)IgG1-cre(+/-) mice exhibited decreased expression of transcription factors important for PC function: Prdm1, Xbp1 and Irf4. Bcl6 is a GC transcriptional repressor that inhibits the PC transcriptional program and thus must be downregulated for PC differentiation to occur. Bcl6 expression was increased in PCs isolated from ADAM10(Δ/Δ)IgG1-cre(+/-) mice at both the mRNA and protein level. These results demonstrate that ADAM10 is required for proper transcription factor expression in PCs and thus, for normal PC function.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Plasma Cells/metabolism , Transcription Factors/genetics , ADAM10 Protein , Animals , Antibody Formation/immunology , Bacterial Proteins/metabolism , Cell Count , Cell Separation , DNA-Binding Proteins/metabolism , Germinal Center/immunology , Immunoglobulin G/metabolism , Immunologic Memory/immunology , Integrases/metabolism , Luminescent Proteins/metabolism , Mice , Plasma Cells/immunology , Proto-Oncogene Proteins c-bcl-6 , Transcription Factors/metabolism
15.
J Immunol ; 187(10): 5114-22, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21998451

ABSTRACT

A disintegrin and metalloproteinase 10 (ADAM10) is a zinc-dependent proteinase related to matrix metalloproteinases. ADAM10 has emerged as a key regulator of cellular processes by cleaving and shedding extracellular domains of multiple transmembrane receptors and ligands. We have developed B cell-specific ADAM10-deficient mice (ADAM10(B-/-)). In this study, we show that ADAM10 levels are significantly enhanced on germinal center B cells. Moreover, ADAM10(B-/-) mice had severely diminished primary and secondary responses after T-dependent immunization. ADAM10(B-/-) displayed impaired germinal center formation, had fewer follicular Th cells, decreased follicular dendritic cell networks, and altered chemokine expression in draining lymph nodes (LNs). Interestingly, when spleen and LN structures from immunized mice were analyzed for B and T cell localization, tissues structure was aberrant in ADAM10(B-/-) mice. Importantly, when ADAM10-deficient B cells were stimulated in vitro, they produced comparable Ab as wild type B cells. This result demonstrates that the defects in humoral responses in vivo result from inadequate B cell activation, likely because of the decrease in follicular Th cells and the changes in structure. Thus, ADAM10 is essential for the maintenance of lymphoid structure after Ag challenge.


Subject(s)
ADAM Proteins/physiology , Amyloid Precursor Protein Secretases/physiology , Immunity, Humoral , Membrane Proteins/physiology , ADAM Proteins/biosynthesis , ADAM Proteins/deficiency , ADAM10 Protein , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/deficiency , Animals , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , CHO Cells , Cricetinae , Germinal Center/enzymology , Germinal Center/immunology , Germinal Center/pathology , Immunity, Humoral/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Peyer's Patches/enzymology , Peyer's Patches/immunology , Peyer's Patches/pathology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Up-Regulation/genetics , Up-Regulation/immunology
16.
J Immunol ; 186(7): 4244-52, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21368228

ABSTRACT

Although the physiological consequences of Notch signaling in hematopoiesis have been extensively studied, the differential effects of individual notch cleavage products remain to be elucidated. Given that ADAM10 is a critical regulator of Notch and that its deletion is embryonically lethal, we generated mice that overexpress ADAM10 (ADAM10 transgenic [A10Tg]) at early stages of lympho- and myeloid development. Transgene expression resulted in abrogated B cell development, delayed T cell development in the thymus, and unexpected systemic expansion of CD11b(+)Gr-1(+) cells, also known as myeloid-derived suppressor cells. Mixed bone marrow reconstitution assays demonstrated that transgene expression altered hematopoiesis via a cell-intrinsic mechanism. Consistent with previously reported observations, we hypothesized that ADAM10 overexpression dysregulated Notch by uncoupling the highly regulated proteolysis of Notch receptors. This was confirmed using an in vitro model of hematopoiesis via culturing A10Tg hematopoietic Lineage(-)Sca-1(+)c-Kit(+) cells with OP-9 stromal cells in the presence or absence of Delta-like 1, a primary ligand for Notch. Blockade of the site 2 (S2) and site 3 (S3) cleavage of the Notch receptor demonstrated differential effects on hematopoiesis. OP9-DL1 cultures containing the ADAM10 inhibitor (S2 cleavage site) enhanced and rescued B cell development from wild-type and A10Tg Lineage(-)Sca-1(+)c-Kit(+) cells, respectively. In contrast, blockade of γ-secretase at the S3 cleavage site induced accumulation of the S2 product and consequently prevented B cell development and resulted in myeloid cell accumulation. Collectively, these findings indicate that the differential cleavage of Notch into S2 and S3 products regulated by ADAM10 is critical to hematopoietic cell-fate determination.


Subject(s)
ADAM Proteins/genetics , Amyloid Precursor Protein Secretases/genetics , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Membrane Proteins/genetics , Myelopoiesis/genetics , Myelopoiesis/immunology , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , ADAM Proteins/biosynthesis , ADAM Proteins/physiology , ADAM10 Protein , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/physiology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation , Cells, Cultured , Growth Inhibitors/biosynthesis , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hydrolysis , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/metabolism , Receptors, Notch/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology
17.
J Exp Med ; 207(3): 623-35, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20156974

ABSTRACT

The proteolytic activity of a disintegrin and metalloproteinase 10 (ADAM10) regulates cell-fate decisions in Drosophila and mouse embryos. However, in utero lethality of ADAM10(-/-) mice has prevented examination of ADAM10 cleavage events in lymphocytes. To investigate their role in B cell development, we generated B cell-specific ADAM10 knockout mice. Intriguingly, deletion of ADAM10 prevented development of the entire marginal zone B cell (MZB) lineage. Additionally, cleavage of the low affinity IgE receptor, CD23, was profoundly impaired, but subsequent experiments demonstrated that ADAM10 regulates CD23 cleavage and MZB development by independent mechanisms. Development of MZBs is dependent on Notch2 signaling, which requires proteolysis of the Notch2 receptor by a previously unidentified proteinase. Further experiments revealed that Notch2 signaling is severely impaired in ADAM10-null B cells. Thus, ADAM10 critically regulates MZB development by initiating Notch2 signaling. This study identifies ADAM10 as the in vivo CD23 sheddase and an important regulator of B cell development. Moreover, it has important implications for the treatment of numerous CD23- and Notch-mediated pathologies, ranging from allergy to cancer.


Subject(s)
ADAM Proteins/deficiency , Amyloid Precursor Protein Secretases/deficiency , B-Lymphocytes/immunology , Membrane Proteins/deficiency , Receptors, IgE/metabolism , ADAM Proteins/genetics , ADAM Proteins/physiology , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/physiology , Animals , DNA, Complementary/genetics , Exons/genetics , Gene Amplification , Gene Deletion , Macrophages/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Receptor, Notch2/genetics , Receptors, CCR1/genetics , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Spleen/physiology , T-Lymphocytes/immunology
18.
Anaerobe ; 16(2): 137-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19464381

ABSTRACT

Primary bile acids are synthesized from cholesterol in the liver, conjugated to either glycine or taurine and secreted into bile. Bile salts undergo enterohepatic circulation several times each day. During this process, they are biotransformed into a variety of metabolites by gut bacteria. The major biotransformation is the 7alpha-dehydroxylation of cholic acid and chenodeoxycholic acid yielding deoxycholic acid and lithocholic acid, respectively. 7alpha-Dehydroxylation is a multi-step pathway. The genes encoding enzymes in this pathway have been identified in two species of "high" activity strains of clostridia. Here, we report the isolation and characterization of a bile acid inducible (bai) operon in Clostridium hylemonae, a "low" activity 7alpha-dehydroxylating strain. The gene organization and sequence of the baiBCDEFGHI operon was highly conserved between C. hylemonae and "high" activity strains. Surprisingly, the baiA gene was missing from the bai operon of C. hylemonae. The baiA gene was isolated using PCR and degenerate oligonucleotide primers. The mRNA start site for the large bai operon was determined and shown to be only 11bp from the initiation codon of the first gene. It was also discovered that allocholic acid (5alpha) induced the bai operon and stimulated the conversion of [24-(14)C] cholic acid to [24-(14)C] allodeoxycholic acid in cultures of C. scindens and C. hylemonae allodeoxycholic acid. Finally, it was discovered that the addition of testosterone to the growth medium markedly increased 7alpha-dehydroxylation of cholic acid in Clostridium scindens and C. hylemonae. We hypothesize that testosterone may be a gratuitous inducer of genes involved in the reductive arm of the bile acid 7alpha-dehydroxylation pathway.


Subject(s)
Bile Acids and Salts/metabolism , Clostridium/enzymology , Clostridium/genetics , Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , Metabolic Networks and Pathways/genetics , Operon , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Conserved Sequence , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Gene Order , Genes, Bacterial , Molecular Sequence Data , Sequence Analysis, DNA , Testosterone/metabolism , Transcription Initiation Site , Transcriptional Activation
19.
Biochim Biophys Acta ; 1781(1-2): 16-25, 2008.
Article in English | MEDLINE | ID: mdl-18047844

ABSTRACT

Secondary bile acids, formed by intestinal bacteria, are suggested to play a significant role in cancers of the gastrointestinal tract in humans. Bile acid 7alpha/beta-dehydroxylation is carried out by a few species of intestinal clostridia which harbor a multi-gene bile acid inducible (bai) operon. Several genes encoding enzymes in this pathway have been cloned and characterized. However, no gene product(s) has yet been assigned to the production of 3-oxo-Delta4-cholenoic acid intermediates of cholic acid (CA), chenodeoxycholic acid (CDCA) or ursodeoxycholic acid (UDCA). We previously reported that the baiH gene encodes an NADH:flavin oxidoreductase (NADH:FOR); however, the role of this protein in bile acid 7-dehydroxylation is unclear. Homology searches and secondary structural alignments suggest this protein to be similar to flavoproteins which reduce alpha/beta-unsaturated carbonyl compounds. The baiH gene product was expressed in Escherichia coli, purified and discovered to be a stereo-specific NAD(H)-dependent 7beta-hydroxy-3-oxo-Delta4-cholenoic acid oxidoreductase. Additionally, high sequence similarity between the baiH and baiCD gene products suggests the baiCD gene may encode a 3-oxo-Delta4-cholenoic acid oxidoreductase specific for CDCA and CA. We tested this hypothesis using cell extracts prepared from E. coli overexpressing the baiCD gene and discovered that it encodes a stereo-specific NAD(H)-dependent 7alpha-hydroxy-3-oxo-Delta4-cholenoic acid oxidoreductase.


Subject(s)
Carboxylic Acids/chemistry , Carboxylic Acids/metabolism , Clostridium/enzymology , Clostridium/genetics , Oxidoreductases/genetics , Oxidoreductases/metabolism , Amino Acid Sequence , Catalysis , Chromatography, Thin Layer , Conserved Sequence , Molecular Sequence Data , Oxidoreductases/chemistry , Oxidoreductases/isolation & purification , Protein Structure, Secondary , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Stereoisomerism , Substrate Specificity
20.
J Lipid Res ; 47(2): 241-59, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16299351

ABSTRACT

Secondary bile acids, produced solely by intestinal bacteria, can accumulate to high levels in the enterohepatic circulation of some individuals and may contribute to the pathogenesis of colon cancer, gallstones, and other gastrointestinal (GI) diseases. Bile salt hydrolysis and hydroxy group dehydrogenation reactions are carried out by a broad spectrum of intestinal anaerobic bacteria, whereas bile acid 7-dehydroxylation appears restricted to a limited number of intestinal anaerobes representing a small fraction of the total colonic flora. Microbial enzymes modifying bile salts differ between species with respect to pH optima, enzyme kinetics, substrate specificity, cellular location, and possibly physiological function. Crystallization, site-directed mutagenesis, and comparisons of protein secondary structure have provided insight into the mechanisms of several bile acid-biotransforming enzymatic reactions. Molecular cloning of genes encoding bile salt-modifying enzymes has facilitated the understanding of the genetic organization of these pathways and is a means of developing probes for the detection of bile salt-modifying bacteria. The potential exists for altering the bile acid pool by targeting key enzymes in the 7alpha/beta-dehydroxylation pathway through the development of pharmaceuticals or sequestering bile acids biologically in probiotic bacteria, which may result in their effective removal from the host after excretion.


Subject(s)
Bacteria/enzymology , Bile Acids and Salts/metabolism , Intestinal Mucosa/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Amino Acid Sequence , Bacteria/metabolism , Biotransformation , Cholic Acids/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Hydrogen Sulfide/metabolism , Hydroxysteroid Dehydrogenases/chemistry , Hydroxysteroid Dehydrogenases/metabolism , Intestines/microbiology , Models, Molecular , Molecular Sequence Data , Molecular Structure , Sequence Homology, Amino Acid , Taurine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...