Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Identification of Hipk2 as an essential regulator of white fat development

Sjölund, Jonas LU ; Pelorosso, Facundo G ; Quigley, David A ; DelRosario, Reyno and Balmain, Allan (2014) In Proceedings of the National Academy of Sciences 111(20). p.8-7373
Abstract

Homeodomain-interacting protein kinase 2 (Hipk2) has previously been implicated in the control of several transcription factors involved in embryonic development, apoptosis, cell proliferation, and tumor development, but very little is understood about the exact mechanisms through which Hipk2 influences these processes. Analysis of gene expression in normal tissues from genetically heterogeneous mouse or human populations can reveal network motifs associated with the structural or functional components of the tissue, and may predict roles for genes of unknown function. Here we have applied this network strategy to uncover a role for the Hipk2 gene in the transcriptional system controlling adipogenesis. Both in vitro and in vivo models... (More)

Homeodomain-interacting protein kinase 2 (Hipk2) has previously been implicated in the control of several transcription factors involved in embryonic development, apoptosis, cell proliferation, and tumor development, but very little is understood about the exact mechanisms through which Hipk2 influences these processes. Analysis of gene expression in normal tissues from genetically heterogeneous mouse or human populations can reveal network motifs associated with the structural or functional components of the tissue, and may predict roles for genes of unknown function. Here we have applied this network strategy to uncover a role for the Hipk2 gene in the transcriptional system controlling adipogenesis. Both in vitro and in vivo models were used to show that knockdown or loss of Hipk2 specifically inhibits white adipose cell differentiation and tissue development. In addition, loss of Hipk2 leads to induction of pockets of multilocular brown fat-like cells in remaining white adipose depots, which express markers of brown and beige fat such as uncoupling protein 1 and transmembrane protein 26. These changes are accompanied by increased insulin sensitivity in Hipk2 knockout mice and reduced high-fat diet-induced weight gain, highlighting a potential role for this kinase in diseases such as diabetes and obesity. Our study underscores the versatility and power of a readily available tissue, such as skin, for network modeling of systemic transcriptional programs involved in multiple pathways, including lipid metabolism and adipogenesis.

(Less)
Please use this url to cite or link to this publication:
author
; ; ; and
publishing date
type
Contribution to journal
publication status
published
keywords
3T3-L1 Cells, Adipocytes, Adipogenesis, Adipose Tissue, Brown, Adipose Tissue, White, Animals, Apoptosis Regulatory Proteins, Carrier Proteins, Cell Differentiation, DNA Fragmentation, Diet, High-Fat, Female, Gene Expression Regulation, Insulin, Male, Mammary Glands, Animal, Mice, Mice, Knockout, Obesity, PPAR gamma, Protein-Serine-Threonine Kinases, Transcription Factors
in
Proceedings of the National Academy of Sciences
volume
111
issue
20
pages
6 pages
publisher
National Academy of Sciences
external identifiers
  • scopus:84901020401
  • pmid:24785298
ISSN
1091-6490
DOI
10.1073/pnas.1322275111
language
English
LU publication?
no
id
2cf51c46-e6f7-44ef-aadc-5053e45d49c7
date added to LUP
2016-09-08 12:41:28
date last changed
2024-03-07 11:42:14
@article{2cf51c46-e6f7-44ef-aadc-5053e45d49c7,
  abstract     = {{<p>Homeodomain-interacting protein kinase 2 (Hipk2) has previously been implicated in the control of several transcription factors involved in embryonic development, apoptosis, cell proliferation, and tumor development, but very little is understood about the exact mechanisms through which Hipk2 influences these processes. Analysis of gene expression in normal tissues from genetically heterogeneous mouse or human populations can reveal network motifs associated with the structural or functional components of the tissue, and may predict roles for genes of unknown function. Here we have applied this network strategy to uncover a role for the Hipk2 gene in the transcriptional system controlling adipogenesis. Both in vitro and in vivo models were used to show that knockdown or loss of Hipk2 specifically inhibits white adipose cell differentiation and tissue development. In addition, loss of Hipk2 leads to induction of pockets of multilocular brown fat-like cells in remaining white adipose depots, which express markers of brown and beige fat such as uncoupling protein 1 and transmembrane protein 26. These changes are accompanied by increased insulin sensitivity in Hipk2 knockout mice and reduced high-fat diet-induced weight gain, highlighting a potential role for this kinase in diseases such as diabetes and obesity. Our study underscores the versatility and power of a readily available tissue, such as skin, for network modeling of systemic transcriptional programs involved in multiple pathways, including lipid metabolism and adipogenesis.</p>}},
  author       = {{Sjölund, Jonas and Pelorosso, Facundo G and Quigley, David A and DelRosario, Reyno and Balmain, Allan}},
  issn         = {{1091-6490}},
  keywords     = {{3T3-L1 Cells; Adipocytes; Adipogenesis; Adipose Tissue, Brown; Adipose Tissue, White; Animals; Apoptosis Regulatory Proteins; Carrier Proteins; Cell Differentiation; DNA Fragmentation; Diet, High-Fat; Female; Gene Expression Regulation; Insulin; Male; Mammary Glands, Animal; Mice; Mice, Knockout; Obesity; PPAR gamma; Protein-Serine-Threonine Kinases; Transcription Factors}},
  language     = {{eng}},
  month        = {{05}},
  number       = {{20}},
  pages        = {{8--7373}},
  publisher    = {{National Academy of Sciences}},
  series       = {{Proceedings of the National Academy of Sciences}},
  title        = {{Identification of Hipk2 as an essential regulator of white fat development}},
  url          = {{http://dx.doi.org/10.1073/pnas.1322275111}},
  doi          = {{10.1073/pnas.1322275111}},
  volume       = {{111}},
  year         = {{2014}},
}