Transcriptional Pathways in cPGI2-Induced Adipocyte Progenitor Activation for Browning

De novo formation of beige/brite adipocytes from progenitor cells contributes to the thermogenic adaptation of adipose tissue and holds great potential for the therapeutic remodeling of fat as a treatment for obesity. Despite the recent identification of several factors regulating browning of white fat, there is a lack of physiological cell models for the mechanistic investigation of progenitor-mediated beige/brite differentiation. We have previously revealed prostacyclin (PGI2) as one of the few known endogenous extracellular mediators promoting de novo beige/brite formation by relaying β-adrenergic stimulation to the progenitor level. Here, we present a cell model based on murine primary progenitor cells defined by markers previously shown to be relevant for in vivo browning, including a simplified isolation procedure. We demonstrate the specific and broad induction of thermogenic gene expression by PGI2 signaling in the absence of lineage conversion, and reveal the previously unidentified nuclear relocalization of the Ucp1 gene locus in association with transcriptional activation. By profiling the time course of the progenitor response, we show that PGI2 signaling promoted progenitor cell activation through cell cycle and adhesion pathways prior to metabolic maturation toward an oxidative cell phenotype. Our results highlight the importance of core progenitor activation pathways for the recruitment of thermogenic cells and provide a resource for further mechanistic investigation.

[1]  L. Sidossis,et al.  Brown and beige fat in humans: thermogenic adipocytes that control energy and glucose homeostasis. , 2015, The Journal of clinical investigation.

[2]  S. Farmer,et al.  Myocardin-Related Transcription Factor A Regulates Conversion of Progenitors to Beige Adipocytes , 2015, Cell.

[3]  J. Granneman,et al.  Cellular origins of cold‐induced brown adipocytes in adult mice , 2015, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[4]  Robert S. Illingworth,et al.  Chromatin decondensation is sufficient to alter nuclear organization in embryonic stem cells , 2014, Science.

[5]  S. Herzig,et al.  Thermogenic adipocytes: from cells to physiology and medicine. , 2014, Metabolism: clinical and experimental.

[6]  C. Kahn,et al.  ASC-1, PAT2, and P2RX5 are cell surface markers for white, beige, and brown adipocytes , 2014, Science Translational Medicine.

[7]  Caroline Tao,et al.  Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. , 2014, Cell metabolism.

[8]  R. Palmiter,et al.  Eosinophils and Type 2 Cytokine Signaling in Macrophages Orchestrate Development of Functional Beige Fat , 2014, Cell.

[9]  P. Seale,et al.  Brown and beige fat: development, function and therapeutic potential , 2013, Nature Medicine.

[10]  J. Granneman,et al.  Identification of an adipogenic niche for adipose tissue remodeling and restoration. , 2013, Cell metabolism.

[11]  P. Scherer,et al.  Tracking adipogenesis during white adipose tissue development, expansion and regeneration , 2013, Nature Medicine.

[12]  T. Rülicke,et al.  Bi-directional interconversion of brite and white adipocytes , 2013, Nature Cell Biology.

[13]  P. Meister,et al.  Building silent compartments at the nuclear periphery: a recurrent theme. , 2013, Current opinion in genetics & development.

[14]  Ryan Berry,et al.  Characterization of the adipocyte cellular lineage in vivo , 2013, Nature Cell Biology.

[15]  B. Spiegelman,et al.  Beige Adipocytes Are a Distinct Type of Thermogenic Fat Cell in Mouse and Human , 2012, Cell.

[16]  Yun-Hee Lee,et al.  In vivo identification of bipotential adipocyte progenitors recruited by β3-adrenoceptor activation and high-fat feeding. , 2012, Cell metabolism.

[17]  A. Wagers,et al.  Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat , 2010, Proceedings of the National Academy of Sciences.

[18]  K. Kristiansen,et al.  UCP1 Induction during Recruitment of Brown Adipocytes in White Adipose Tissue Is Dependent on Cyclooxygenase Activity , 2010, PloS one.

[19]  K. Kristiansen,et al.  The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. , 2010, American journal of physiology. Endocrinology and metabolism.

[20]  W. Wahli,et al.  Cyclooxygenase-2 Controls Energy Homeostasis in Mice by de Novo Recruitment of Brown Adipocytes , 2010, Science.

[21]  Elad Katz,et al.  Activation of Estrogen-Responsive Genes Does Not Require Their Nuclear Co-Localization , 2010, PLoS genetics.

[22]  Yvette Lahbib-Mansais,et al.  NEMO: a tool for analyzing gene and chromosome territory distributions from 3D-FISH experiments , 2010, Bioinform..

[23]  Jan Nedergaard,et al.  Chronic Peroxisome Proliferator-activated Receptor γ (PPARγ) Activation of Epididymally Derived White Adipocyte Cultures Reveals a Population of Thermogenically Competent, UCP1-containing Adipocytes Molecularly Distinct from Classic Brown Adipocytes* , 2009, The Journal of Biological Chemistry.

[24]  Arpad M. Danos,et al.  Altering PPARγ Ligand Selectivity Impairs Adipogenesis by Thiazolidinediones But Not Hormonal Inducers , 2009, Obesity.

[25]  J. Friedman,et al.  Identification of White Adipocyte Progenitor Cells In Vivo , 2008, Cell.

[26]  Jan Nedergaard,et al.  Myogenic gene expression signature establishes that brown and white adipocytes originate from distinct cell lineages , 2007, Proceedings of the National Academy of Sciences.

[27]  Mary-Ellen Harper,et al.  Rb and p107 regulate preadipocyte differentiation into white versus brown fat through repression of PGC-1alpha. , 2005, Cell metabolism.

[28]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[29]  K. Kristiansen,et al.  Retinoblastoma protein functions as a molecular switch determining white versus brown adipocyte differentiation , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[30]  J. Giacobino,et al.  Beta 3-adrenoceptor knockout in C57BL/6J mice depresses the occurrence of brown adipocytes in white fat. , 2003, European journal of biochemistry.

[31]  A I Saeed,et al.  TM4: a free, open-source system for microarray data management and analysis. , 2003, BioTechniques.

[32]  M. Lane,et al.  Mitotic clonal expansion: A synchronous process required for adipogenesis , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[33]  J. Ragoussis,et al.  Large-scale chromatin organization of the major histocompatibility complex and other regions of human chromosome 6 and its response to interferon in interphase nuclei. , 2000, Journal of cell science.

[34]  S. Narumiya,et al.  Prostanoid receptors: structures, properties, and functions. , 1999, Physiological reviews.

[35]  K. Helin,et al.  Regulation of cell proliferation by the E2F transcription factors. , 1998, Current opinion in genetics & development.

[36]  B. Spiegelman,et al.  Differential activation of adipogenesis by multiple PPAR isoforms. , 1996, Genes & development.

[37]  J. Lehmann,et al.  An Antidiabetic Thiazolidinedione Is a High Affinity Ligand for Peroxisome Proliferator-activated Receptor γ (PPARγ) (*) , 1995, The Journal of Biological Chemistry.

[38]  G. Ailhaud,et al.  Prostacyclin as a potent effector of adipose-cell differentiation. , 1989, The Biochemical journal.

[39]  J. Timmons,et al.  Recruited vs. nonrecruited molecular signatures of brown, "brite," and white adipose tissues. , 2012, American journal of physiology. Endocrinology and metabolism.

[40]  J. Lehmann,et al.  An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma). , 1995, The Journal of biological chemistry.