Your browser doesn't support javascript.
loading
Variant-to-Gene-Mapping Analyses Reveal a Role for the Hypothalamus in Genetic Susceptibility to Inflammatory Bowel Disease.
Lasconi, Chiara; Pahl, Matthew C; Cousminer, Diana L; Doege, Claudia A; Chesi, Alessandra; Hodge, Kenyaita M; Leonard, Michelle E; Lu, Sumei; Johnson, Matthew E; Su, Chun; Hammond, Reza K; Pippin, James A; Terry, Natalie A; Ghanem, Louis R; Leibel, Rudolph L; Wells, Andrew D; Grant, Struan F A.
Afiliação
  • Lasconi C; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Pahl MC; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Cousminer DL; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Doege CA; Division of Molecular Genetics (Pediatrics), Naomi Berrie Diabetes Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York.
  • Chesi A; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Hodge KM; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Leonard ME; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Lu S; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Johnson ME; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Su C; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Hammond RK; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Pippin JA; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania.
  • Terry NA; Department of Pediatrics, Philadelphia, Pennsylvania.
  • Ghanem LR; Department of Pediatrics, Philadelphia, Pennsylvania.
  • Leibel RL; Division of Molecular Genetics (Pediatrics), Naomi Berrie Diabetes Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York.
  • Wells AD; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Department of Pathology, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania.
  • Grant SFA; Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania; Division of Diabetes and Endocrinology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Genetics, Perelman School of Medicine, University
Cell Mol Gastroenterol Hepatol ; 11(3): 667-682, 2021.
Article em En | MEDLINE | ID: mdl-33069917
ABSTRACT
BACKGROUND &

AIMS:

Inflammatory bowel disease (IBD) is a polygenic disorder characterized principally by dysregulated inflammation impacting the gastrointestinal tract. However, there also is increasing evidence for a clinical association with stress and depression. Given the role of the hypothalamus in stress responses and in the pathogenesis of depression, useful insights could be gleaned from understanding its genetic role in IBD.

METHODS:

We conducted genetic correlation analyses on publicly available genome-wide association study summary statistics for depression and IBD traits to identify genetic commonalities. We used partitioned linkage disequilibrium score regression, leveraging our ATAC sequencing and promoter-focused Capture C data, to measure enrichment of IBD single-nucleotide polymorphisms within promoter-interacting open chromatin regions of human embryonic stem cell-derived hypothalamic-like neurons (HNs). Using the same data sets, we performed variant-to-gene mapping to implicate putative IBD effector genes in HNs. To contrast these results, we similarly analyzed 3-dimensional genomic data generated in epithelium-derived colonoids from rectal biopsy specimens from donors without pathologic disease noted at the time of colonoscopy. Finally, we conducted enrichment pathway analyses on the implicated genes to identify putative IBD dysfunctional pathways.

RESULTS:

We found significant genetic correlations (rg) of 0.122 with an adjusted P (Padj) = 1.4 × 10-4 for IBD rg = 0.122; Padj = 2.5 × 10-3 for ulcerative colitis and genetic correlation (rg) = 0.094; Padj = 2.5 × 10-3 for Crohn's disease, and significant approximately 4-fold (P = .005) and approximately 7-fold (P = .03) enrichment of IBD single-nucleotide polymorphisms in HNs and colonoids, respectively. We implicated 25 associated genes in HNs, among which CREM, CNTF, and RHOA encode key regulators of stress. Seven genes also additionally were implicated in the colonoids. We observed an overall enrichment for immune and hormonal signaling pathways, and a colonoid-specific enrichment for microbiota-relevant terms.

CONCLUSIONS:

Our results suggest that the hypothalamus warrants further study in the context of IBD pathogenesis.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Estresse Psicológico / Doenças Inflamatórias Intestinais / Predisposição Genética para Doença / Depressão / Hipotálamo Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Estresse Psicológico / Doenças Inflamatórias Intestinais / Predisposição Genética para Doença / Depressão / Hipotálamo Idioma: En Ano de publicação: 2021 Tipo de documento: Article