4.6 Article

Adipose stem cells from patients with Crohn's disease show a distinctive DNA methylation pattern

Journal

CLINICAL EPIGENETICS
Volume 12, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s13148-020-00843-3

Keywords

Methylome; Inflammatory bowel disease; Adipose tissue; Epigenetics; Gene expression

Funding

  1. Spanish Ministry of Economy and Competitiveness [PI18/00037, PI15/00143, PI14/00228, PI17/01503, SAF2015-65019-R, RTI2018-093919]
  2. European Regional Development Fund (ERDF)
  3. European Crohn's and Colitis Organization (ECCO) grant
  4. Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM) of the Instituto de Salud Carlos III [CB07708/0012]
  5. Ramon y Cajal program from the Ministerio de Educacion y Ciencia [RYC2013-13186]
  6. ERDF
  7. Miguel Servet tenure track program from the Fondo de Investigacion Sanitaria [CP10/00438, CPII16/0008]
  8. Generalitat de Catalunya - ERDF [SLT002/16/00120]
  9. Rio Hortega IISCIII fellowship - ERDF [CM18/00029]

Ask authors/readers for more resources

Background Crohn's disease (CD) is characterized by persistent inflammation and ulceration of the small or large bowel, and expansion of mesenteric adipose tissue, termed creeping fat (CF). We previously demonstrated that human adipose-derived stem cells (hASCs) from CF of patients with CD exhibit dysfunctional phenotypes, including a pro-inflammatory profile, high phagocytic capacity, and weak immunosuppressive properties. Importantly, these phenotypes persist in patients in remission and are found in all adipose depots explored including subcutaneous fat. We hypothesized that changes in hASCs are a consequence of epigenetic modifications. Methods We applied epigenome-wide profiling with a methylation array (Illumina EPIC/850k array) and gene expression analysis to explore the impact of CD on the methylation signature of hASCs isolated from the subcutaneous fat of patients with CD and healthy controls (n = 7 and 5, respectively; cohort I). Differentially methylated positions (p value cutoff < 1 x 10(-4) and ten or more DMPs per gene) and regions (inclusion threshold 0.2, p value cutoff < 1 x 10(-2) and more than 2 DMRs per gene) were identified using dmpfinder and Bumphunter (minfi), respectively. Changes in the expression of differentially methylated genes in hASCs were validated in a second cohort (n = 10/10 inactive and active CD and 10 controls; including patients from cohort I) and also in peripheral blood mononuclear cells (PBMCs) of patients with active/inactive CD and of healthy controls (cohort III; n = 30 independent subjects). Results We found a distinct DNA methylation landscape in hASCs from patients with CD, leading to changes in the expression of differentially methylated genes involved in immune response, metabolic, cell differentiation, and development processes. Notably, the expression of several of these genes in hASCs and PBMCs such as tumor necrosis factor alpha (TNFA) and PR domain zinc finger protein 16 (PRDM16) were not restored to normal (healthy) levels after disease remission. Conclusions hASCs of patients with CD exhibit a unique DNA methylation and gene expression profile, but the expression of several genes are only partially restored in patients with inactive CD, both in hASCs and PBMCs. Understanding how CD shapes the functionality of hASCs is critical for investigating the complex pathophysiology of this disease, as well as for the success of cell-based therapies.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available