Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Cell Host Microbe ; 29(6): 988-1001.e6, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34010595

ABSTRACT

Intestinal Paneth cells modulate innate immunity and infection. In Crohn's disease, genetic mutations together with environmental triggers can disable Paneth cell function. Here, we find that a western diet (WD) similarly leads to Paneth cell dysfunction through mechanisms dependent on the microbiome and farnesoid X receptor (FXR) and type I interferon (IFN) signaling. Analysis of multiple human cohorts suggests that obesity is associated with Paneth cell dysfunction. In mouse models, consumption of a WD for as little as 4 weeks led to Paneth cell dysfunction. WD consumption in conjunction with Clostridium spp. increased the secondary bile acid deoxycholic acid levels in the ileum, which in turn inhibited Paneth cell function. The process required excess signaling of both FXR and IFN within intestinal epithelial cells. Our findings provide a mechanistic link between poor diet and inhibition of gut innate immunity and uncover an effect of FXR activation in gut inflammation.


Subject(s)
Diet, Western/adverse effects , Gastrointestinal Microbiome/drug effects , Interferon Type I/metabolism , Obesity/metabolism , Paneth Cells/drug effects , Paneth Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/metabolism , Cells, Cultured , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Expression Profiling , Humans , Immunity, Innate/drug effects , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
2.
Science ; 371(6534): 1154-1159, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33707263

ABSTRACT

Alterations of the mycobiota composition associated with Crohn's disease (CD) are challenging to link to defining elements of pathophysiology, such as poor injury repair. Using culture-dependent and -independent methods, we discovered that Debaryomyces hansenii preferentially localized to and was abundant within incompletely healed intestinal wounds of mice and inflamed mucosal tissues of CD human subjects. D. hansenii cultures from injured mice and inflamed CD tissues impaired colonic healing when introduced into injured conventionally raised or gnotobiotic mice. We reisolated D. hansenii from injured areas of these mice, fulfilling Koch's postulates. Mechanistically, D. hansenii impaired mucosal healing through the myeloid cell-specific type 1 interferon-CCL5 axis. Taken together, we have identified a fungus that inhabits inflamed CD tissue and can lead to dysregulated mucosal healing.


Subject(s)
Crohn Disease/microbiology , Crohn Disease/pathology , Debaryomyces/isolation & purification , Debaryomyces/physiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Amphotericin B/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Chemokine CCL5/metabolism , Colon/microbiology , Colon/pathology , Crohn Disease/immunology , Debaryomyces/growth & development , Female , Gastrointestinal Microbiome , Germ-Free Life , Humans , Ileum/microbiology , Ileum/pathology , Inflammation , Interferon Type I/metabolism , Intestinal Mucosa/immunology , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL
3.
Am J Physiol Lung Cell Mol Physiol ; 317(2): L259-L270, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31116580

ABSTRACT

Epithelial cells that line lung airways produce and secrete proteins with important roles in barrier function and host defense. Secretion of airway goblet cells is controlled by autophagy proteins during inflammatory conditions, resulting in accumulation of mucin proteins. We hypothesized that autophagy proteins would also be important in the function of club cells, dominant secretory airway epithelial cells that are dysregulated in chronic lung disease. We found that in the absence of an inflammatory stimulus, mice with club cells deficient for the autophagy protein Atg5 had a markedly diminished expression of secreted host defense proteins secretoglobulin family 1A, member 1 (Scgb1a1) and surfactant proteins A1 and D (Sftpa1 and Sftpd), as well as abnormal club cell morphology. Adult mice with targeted loss of Atg5 also showed diminished levels of host defense proteins in regenerating cells following ablation with naphthalene. A mouse strain with global deficiency of Atg16-like 1 (Atg16l1), an Atg5 binding partner, had a similar loss of host defense proteins and abnormal club cell morphology. Cigarette smoke exposure reduced levels of Scgb1a1 in wild-type mice as expected. Smoke exposure was not required to trigger club cell abnormalities in mice bearing the human ATG16 variant Atg16l1T300A/T300A, which had low Scgb1a1 levels independent of this environmental stress. Evaluation of lung tissues from former smokers with severe chronic obstructive pulmonary disease showed evidence of reduced autophagy and SCGB1A1 expression in club cells. Thus, autophagy proteins are required for the function of club cells, independent of the cellular stress of cigarette smoke, with roles that appear to be distinct from those of other secretory cell types.


Subject(s)
Autophagy/physiology , Epithelial Cells/metabolism , Goblet Cells/metabolism , Lung/metabolism , Animals , Bronchioles/metabolism , Female , Humans , Male , Mice, Transgenic , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Respiratory Mucosa/metabolism
4.
J Clin Invest ; 128(11): 5110-5122, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30137026

ABSTRACT

It is suggested that subtyping of complex inflammatory diseases can be based on genetic susceptibility and relevant environmental exposure (G+E). We propose that using matched cellular phenotypes in human subjects and corresponding preclinical models with the same G+E combinations is useful to this end. As an example, defective Paneth cells can subtype Crohn's disease (CD) subjects; Paneth cell defects have been linked to multiple CD susceptibility genes and are associated with poor outcome. We hypothesized that CD susceptibility genes interact with cigarette smoking, a major CD environmental risk factor, to trigger Paneth cell defects. We found that both CD subjects and mice with ATG16L1T300A (T300A; a prevalent CD susceptibility allele) developed Paneth cell defects triggered by tobacco smoke. Transcriptional analysis of full-thickness ileum and Paneth cell-enriched crypt base cells showed the T300A-smoking combination altered distinct pathways, including proapoptosis, metabolic dysregulation, and selective downregulation of the PPARγ pathway. Pharmacologic intervention by either apoptosis inhibitor or PPARγ agonist rosiglitazone prevented smoking-induced crypt apoptosis and Paneth cell defects in T300A mice and mice with conditional Paneth cell-specific knockout of Atg16l1. This study demonstrates how explicit G+E can drive disease-relevant phenotype and provides rational strategies for identifying actionable targets.


Subject(s)
Autophagy-Related Proteins/metabolism , Carrier Proteins/metabolism , Crohn Disease/metabolism , Genetic Predisposition to Disease , Mutation, Missense , Paneth Cells/metabolism , Smoking/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy-Related Proteins/genetics , Carrier Proteins/genetics , Crohn Disease/genetics , Crohn Disease/pathology , Female , Humans , Male , Mice , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , Paneth Cells/pathology , Rosiglitazone/pharmacology , Smoking/genetics
SELECTION OF CITATIONS
SEARCH DETAIL