Mesenteric Adipose Tissue Derived Stromal Cells from Crohn's Disease Patients Induce Lactoferrin-Dependent Protective Responses in Colonic Epithelial Cells and Mice with Colitis

2017 ◽  
Vol 152 (5) ◽  
pp. S86-S87
Author(s):  
Jill M. Hoffman ◽  
Aristea Sideri ◽  
Jonathan J. Ruiz ◽  
Jerrold R. Turner ◽  
Charalabos Pothoulakis ◽  
...  
2018 ◽  
Author(s):  
Raquel Franco Leal ◽  
Lívia Bitencourt Pascoal ◽  
Francesca Aparecida Ramos da Silva ◽  
Bruno Lima Rodrigues

PLoS ONE ◽  
2019 ◽  
Vol 14 (2) ◽  
pp. e0212850 ◽  
Author(s):  
Stephen M. Matthews ◽  
Melanie A. Eshelman ◽  
Arthur S. Berg ◽  
Walter A. Koltun ◽  
Gregory S. Yochum

PLoS ONE ◽  
2019 ◽  
Vol 14 (9) ◽  
pp. e0223105 ◽  
Author(s):  
Andressa Coope ◽  
Lívia Bitencourt Pascoal ◽  
José Diego Botezelli ◽  
Francesca Aparecida Ramos da Silva ◽  
Maria de Lourdes Setsuko Ayrizono ◽  
...  

2001 ◽  
Vol 60 (3) ◽  
pp. 365-374 ◽  
Author(s):  
Caroline M. Pond

Redistribution of white adipose tissue is a long-term symptom of several chronic diseases. Although the roles of adipocytes in acute illness have been thoroughly studied, how or why short-term responses of adipose tissue to disease sometimes produce long-term redistribution, and the causal relationship between the anatomical changes and the associated metabolic syndromes are poorly understood. The present paper reviews explanations for the redistribution of adipose tissue after infection with HIV, and in Crohn's disease; both conditions that share the peculiarity of selective expansion of certain adipose depots while others are depleted. HIV adipose tissue redistribution syndrome (HARS) develops gradually after several months of infection with the HIV both in untreated patients and in those taking protease inhibitors and nucleoside reverse transcriptase inhibitors. Some current theories about the causes of HARS are critically assessed, and reasons presented for implicating local interactions between the immune system and perinodal adipocytes. Some evolutionary aspects of conspicuous long-term changes in the distribution of human adipose tissue are discussed. Adipose tissue acts as a social signal, indicating dietary history and previous exposure to pathogens. A distinctive symptom of Crohn's disease is selective enlargement of the mesenteric adipose tissue near the diseased lymph nodes and intestine. Perinodal adipocytes have site-specific properties not found in adipocytes from nodeless depots, such as perirenal and epididymal, that may equip them to interact locally with lymph-node lymphoid cells, making polyunsaturated fatty acids selectively and rapidly available to activated immune cells. Studies of the time course of activation of perinodal adipocytes via the lymph nodes they enclose indicate that prolonged or frequent stimulation recruits more adipocytes to control by immune cells, which may lead to selective enlargement of node-containing depots. These concepts suggest hypotheses about HARS and the anomalous development of mesenteric adipose tissue in Crohn's disease that could form the basis for further investigations.


Microbiome ◽  
2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Zhen He ◽  
Jinjie Wu ◽  
Junli Gong ◽  
Jia Ke ◽  
Tao Ding ◽  
...  

Abstract Background Mesenteric adipose tissue (mAT) hyperplasia, known as creeping fat is a pathologic characteristic of Crohn’s disease (CD). The reserve of creeping fat in surgery is associated with poor prognosis of CD patients, but the mechanism remains unknown. Methods Mesenteric microbiome, metabolome, and host transcriptome were characterized using a cohort of 48 patients with CD and 16 non-CD controls. Multidimensional data including 16S ribosomal RNA gene sequencing (16S rRNA), host RNA sequencing, and metabolome were integrated to reveal network interaction. Mesenteric resident bacteria were isolated from mAT and functionally investigated both in the dextran sulfate sodium (DSS) model and in the Il10 gene-deficient (Il10−/−) mouse colitis model to validate their pro-inflammatory roles. Results Mesenteric microbiota contributed to aberrant metabolites production and transcripts in mATs from patients with CD. The presence of mAT resident microbiota was associated with the development of CD. Achromobacter pulmonis (A. pulmonis) isolated from CD mAT could translocate to mAT and exacerbate both DSS-induced and Il10 gene-deficient (Il10−/−) spontaneous colitis in mice. The levels of A. pulmonis in both mAT and mucous layer from CD patients were higher compared to those from the non-CD group. Conclusions This study suggests that the mesenteric microbiota from patients with CD sculpt a detrimental microenvironment and promote intestinal inflammation.


2021 ◽  
Author(s):  
Zhen He ◽  
Jinjie Wu ◽  
Junli Gong ◽  
Jia Ke ◽  
Tao Ding ◽  
...  

Abstract Background: Mesenteric adipose tissue (mAT) hyperplasia, known as creeping fat is a pathologic characteristic of Crohn’s disease (CD). The reserve of creeping fat in surgery is associated with poor prognosis of CD patients, but the mechanism remains unknown.Methods: Mesenteric microbiome, metabolome and host transcriptome were characterized using a cohort of 48 patients with CD and 16 non-CD controls. Multidimensional data including 16S ribosomal RNA gene sequencing (16S rRNA), host RNA sequencing and metabolome were integrated to reveal network interaction. Mesenteric resident bacteria were isolated from mAT and functionally investigated both in dextran sulfate sodium (DSS) model and Il10 gene deficient (Il10-/-) mouse colitis model to validate their pro-inflammatory roles. Results: Mesenteric microbiota contributed to aberrant metabolites production and transcripts in mATs from patients with CD. Presence of mAT resident microbiota was associated with the development of CD. Achromobacter pulmonis (A. pulmonis) isolated from CD mAT could translocate to mAT and exacerbate both DSS-induced and Il10 gene deficient (Il10-/-) spontaneous colitis in mice. The levels of A. pulmonis both in mAT and mucous layer from CD patients were higher compared to those from non-CD group. Conclusions: This study suggests that the mesenteric microbiota from patients with CD sculpt a detrimental microenvironment and promote intestinal inflammation.


Sign in / Sign up

Export Citation Format

Share Document