Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters











Publication year range
1.
Genes Dev ; 38(15-16): 772-783, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39266447

ABSTRACT

The distinct anatomic environment in which adipose tissues arise during organogenesis is a principle determinant of their adult expansion capacity. Metabolic disease results from a deficiency in hyperplastic adipose expansion within the dermal/subcutaneous depot; thus, understanding the embryonic origins of dermal adipose is imperative. Using single-cell transcriptomics throughout murine embryogenesis, we characterized cell populations, including Bcl11b + cells, that regulate the development of dermal white adipose tissue (dWAT). We discovered that BCL11b expression modulates the Wnt signaling microenvironment to enable adipogenic differentiation in the dermal compartment. Subcutaneous and visceral adipose arises from a distinct population of Nefl + cells during embryonic organogenesis, whereas Pi16 + /Dpp4 + fibroadipogenic progenitors support obesity-stimulated hypertrophic expansion in the adult. Together, these results highlight the unique regulatory pathways used by anatomically distinct adipose depots, with important implications for human metabolic disease.


Subject(s)
Gene Expression Regulation, Developmental , Repressor Proteins , Animals , Mice , Repressor Proteins/genetics , Repressor Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Adipogenesis/genetics , Adipose Tissue, White/embryology , Adipose Tissue, White/metabolism , Wnt Signaling Pathway/genetics , Adipose Tissue/metabolism , Adipose Tissue/embryology , Cell Differentiation/genetics , Humans
2.
Toxicol Sci ; 164(1): 72-84, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29617909

ABSTRACT

Globally, approximately 10%-25% of women smoke during pregnancy. Since nicotine is highly addictive, women may use nicotine-containing products like nicotine replacement therapies for smoking cessation, but the long-term consequences of early life exposure to nicotine remain poorly defined. Our laboratory has previously demonstrated that maternal nicotine exposed (MNE) rat offspring exhibit hypertriglyceridemia due to increased hepatic de novo lipogenesis. Hypertriglyceridemia may also be attributed to impaired white adipose tissue (WAT) lipid storage; however, the effects of MNE on WAT are not completely understood. We hypothesize that nicotine-induced alterations in adipose function (eg, lipid storage) underlie dyslipidemia in MNE adults. Female 6-month-old rats exposed to nicotine during gestation and lactation exhibited significantly decreased visceral adipocyte cell area by 40%, attributed, in part, to a 3-fold increase in adipose triglyceride lipase (ATGL) protein expression compared with vehicle. Given ATGL has antioxidant properties and in utero nicotine exposure promotes oxidative stress in various tissues, we next investigated if there was evidence of increased oxidative stress in MNE WAT. At both 3 weeks and 6 months, MNE offspring expressed 37%-48% higher protein levels of superoxide dismutase-1 and -2 in WAT. Since oxidative stress can induce inflammation, we examined the inflammatory profile of WAT and found increased expression of cytokines (interleukin-1ß, tumor necrosis factor α, and interleukin-6) by 44%-61% at 6 months. Collectively, this suggests that the expression of WAT ATGL may be induced to counter MNE-induced oxidative stress and inflammation. However, higher levels of ATGL would further promote lipolysis in WAT, culminating in impaired lipid storage and long-term dyslipidemia.


Subject(s)
Adipose Tissue, White/drug effects , Antioxidants/metabolism , Lipase/genetics , Maternal Exposure/adverse effects , Nicotine/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Adipocytes, White/drug effects , Adipocytes, White/enzymology , Adipose Tissue, White/embryology , Adipose Tissue, White/enzymology , Adipose Tissue, White/growth & development , Animals , Escherichia coli Proteins/drug effects , Female , Lipogenesis/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/enzymology , Prenatal Exposure Delayed Effects/genetics , Rats, Wistar
3.
Endocrinology ; 159(5): 2050-2061, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29579167

ABSTRACT

Regulation of adipogenesis is of major interest given that adipose tissue expansion and dysfunction are central to metabolic syndrome. Glucocorticoids (GCs) are important for adipogenesis in vitro. However, establishing a role for GCs in adipogenesis in vivo has been difficult. GC receptor (GR)‒null mice die at birth, a time at which wild-type (WT) mice do not have fully developed white adipose depots. We conducted several studies aimed at defining the role of GC signaling in adipogenesis in vitro and in vivo. First, we showed that GR-null mouse embryonic fibroblasts (MEFs) have compromised ability to form adipocytes in vitro, a phenotype that can be partially rescued with a peroxisome proliferator-activated receptor γ agonist. Next, we demonstrated that MEFs are capable of forming de novo fat pads in mice despite the absence of GR or circulating GCs [by bilateral adrenalectomy (ADX)]. However, ADX and GR-null fat pads and their associated adipocyte areas were smaller than those in controls. Second, using adipocyte-specific luciferase reporter mice, we identified adipocytes in both WT and GR-null embryonic day (E)18 mouse embryos. Lastly, positive perilipin staining in WT and GR-null E18 embryos confirmed the presence of early white inguinal and brown adipocytes. Taken together, these results provide compelling evidence that GCs and GR augment but are not required for the development of functional adipose tissue in vivo.


Subject(s)
Adipogenesis/genetics , Adipose Tissue, Brown/embryology , Adipose Tissue, White/embryology , Fibroblasts/metabolism , Glucocorticoids/metabolism , Receptors, Glucocorticoid/genetics , Adipocytes, Brown , Adipocytes, White , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adrenalectomy , Animals , In Vitro Techniques , Mice , PPAR gamma/agonists , Perilipin-1/metabolism , Signal Transduction
4.
Dev Cell ; 35(5): 568-583, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26625958

ABSTRACT

Progression from brown preadipocytes to adipocytes engages two transcriptional programs: the expression of adipogenic genes common to both brown fat (BAT) and white fat (WAT), and the expression of BAT-selective genes. However, the dynamics of chromatin states and epigenetic enzymes involved remain poorly understood. Here we show that BAT development is selectively marked and guided by repressive H3K27me3 and is executed by its demethylase Jmjd3. We find that a significant subset of BAT-selective genes, but not common fat genes or WAT-selective genes, are demarcated by H3K27me3 in both brown and white preadipocytes. Jmjd3-catalyzed removal of H3K27me3, in part through Rreb1-mediated recruitment, is required for expression of BAT-selective genes and for development of beige adipocytes both in vitro and in vivo. Moreover, gain- and loss-of-function Jmjd3 transgenic mice show age-dependent body weight reduction and cold intolerance, respectively. Together, we identify an epigenetic mechanism governing BAT fate determination and WAT plasticity.


Subject(s)
Adipose Tissue, Brown/embryology , Adipose Tissue, White/embryology , Gene Expression Regulation, Developmental , Jumonji Domain-Containing Histone Demethylases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Body Weight , DNA-Binding Proteins/metabolism , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Point Mutation , Promoter Regions, Genetic , Sequence Analysis, RNA , Thermogenesis/genetics , Transcription Factors/metabolism , Transgenes , Uncoupling Protein 1
5.
Biochim Biophys Acta ; 1851(5): 686-96, 2015 May.
Article in English | MEDLINE | ID: mdl-25668679

ABSTRACT

In this review we discuss the role of developmental transcription factors in adipose tissue biology with a focus on how these developmental genes may contribute to regional variation in adipose tissue distribution and function. Regional, depot-specific, differences in lipid handling and signalling (lipolysis, lipid storage and adipokine/lipokine signalling) are important determinants of metabolic health. At a cellular level, preadipocytes removed from their original depot and cultured in vitro retain depot-specific functional properties, implying that these are intrinsic to the cells and not a function of their environment in situ. High throughput screening has identified a number of developmental transcription factors involved in embryological development, including members of the Homeobox and T-Box gene families, that are strongly differentially expressed between regional white adipose tissue depots and also between brown and white adipose tissue. However, the significance of depot-specific developmental signatures remains unclear. Developmental transcription factors determine body patterning during embryogenesis. The divergent developmental origins of regional adipose tissue depots may explain their differing functional characteristics. There is evidence from human genetics that developmental genes determine adipose tissue distribution: in GWAS studies a number of developmental genes have been identified as being correlated with anthropometric measures of adiposity and fat distribution. Additionally, compelling functional studies have recently implicated developmental genes in both white adipogenesis and the so-called 'browning' of white adipose tissue. Understanding the genetic and developmental pathways in adipose tissue may help uncover novel ways to intervene with the function of adipose tissue in order to promote health.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Transcription Factors/metabolism , Adipokines/metabolism , Adipose Tissue, Brown/embryology , Adipose Tissue, Brown/growth & development , Adipose Tissue, White/embryology , Adipose Tissue, White/growth & development , Adiposity , Animals , Energy Metabolism , Gene Expression Regulation, Developmental , Humans , Lipogenesis , Lipolysis , Morphogenesis , Signal Transduction , Transcription Factors/genetics
6.
Proc Natl Acad Sci U S A ; 111(40): 14466-71, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25197048

ABSTRACT

Brown adipocytes and muscle and dorsal dermis descend from precursor cells in the dermomyotome, but the factors that regulate commitment to the brown adipose lineage are unknown. Here, we prospectively isolated and determined the molecular profile of embryonic brown preadipose cells. Brown adipogenic precursor activity in embryos was confined to platelet-derived growth factor α(+), myogenic factor 5(Cre)-lineage-marked cells. RNA-sequence analysis identified early B-cell factor 2 (Ebf2) as one of the most selectively expressed genes in this cell fraction. Importantly, Ebf2-expressing cells purified from Ebf2(GFP) embryos or brown fat tissue did not express myoblast or dermal cell markers and uniformly differentiated into brown adipocytes. Interestingly, Ebf2-expressing cells from white fat tissue in adult animals differentiated into brown-like (or beige) adipocytes. Loss of Ebf2 in brown preadipose cells reduced the expression levels of brown preadipose-signature genes, whereas ectopic Ebf2 expression in myoblasts activated brown preadipose-specific genes. Altogether, these results indicate that Ebf2 specifically marks and regulates the molecular profile of brown preadipose cells.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Adipocytes/cytology , Adipogenesis/genetics , Adipose Tissue/cytology , Adipose Tissue/embryology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/embryology , Adipose Tissue, White/cytology , Adipose Tissue, White/embryology , Adipose Tissue, White/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Lineage/genetics , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
7.
Cell Rep ; 8(3): 678-87, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25088414

ABSTRACT

Pref-1 is an EGF-repeat-containing protein that inhibits adipocyte differentiation. To better understand the origin and development of white adipose tissue (WAT), we generated transgenic mouse models for transient or permanent fluorescent labeling of cells using the Pref-1 promoter, facilitating inducible ablation. We show that Pref-1-marked cells retain proliferative capacity and are very early adipose precursors, prior to expression of Zfp423 or PPARγ. In addition, the Pref-1-marked cells establish that adipose precursors are mesenchymal, but not endothelial or pericytal, in origin. During embryogenesis, Pref-1-marked cells first appear in the dorsal mesenteric region as early as embryonic day 10.5 (E10.5). These cells become lipid-laden adipocytes at E17.5 in the subcutaneous region, whereas visceral WAT develops after birth. Finally, ablation of Pref-1-marked cells prevents not only embryonic WAT development but also later adult adipose expansion upon high-fat feeding, demonstrating the requirement of Pref-1 cells for adipogenesis.


Subject(s)
Adipogenesis , Adipose Tissue, White/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/cytology , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/embryology , Animals , Calcium-Binding Proteins , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mesoderm/embryology , Mesoderm/metabolism , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Nature ; 510(7503): 76-83, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24899307

ABSTRACT

Our understanding of adipose tissue biology has progressed rapidly since the turn of the century. White adipose tissue has emerged as a key determinant of healthy metabolism and metabolic dysfunction. This realization is paralleled only by the confirmation that adult humans have heat-dissipating brown adipose tissue, an important contributor to energy balance and a possible therapeutic target for the treatment of metabolic disease. We propose that the development of successful strategies to target brown and white adipose tissues will depend on investigations that elucidate their developmental origins and cell-type-specific functional regulators.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes/pathology , Adipogenesis , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/embryology , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/pathology , Adipose Tissue, White/cytology , Adipose Tissue, White/embryology , Adipose Tissue, White/innervation , Adipose Tissue, White/pathology , Animals , Humans , Lipolysis , Obesity/metabolism , Obesity/pathology , Sympathetic Nervous System/metabolism , Thermogenesis
9.
Nat Cell Biol ; 16(4): 367-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24609269

ABSTRACT

Fuelled by the obesity epidemic, there is considerable interest in the developmental origins of white adipose tissue (WAT) and the stem and progenitor cells from which it arises. Whereas increased visceral fat mass is associated with metabolic dysfunction, increased subcutaneous WAT is protective. There are six visceral fat depots: perirenal, gonadal, epicardial, retroperitoneal, omental and mesenteric, and it is a subject of much debate whether these have a common developmental origin and whether this differs from that for subcutaneous WAT. Here we show that all six visceral WAT depots receive a significant contribution from cells expressing Wt1 late in gestation. Conversely, no subcutaneous WAT or brown adipose tissue arises from Wt1-expressing cells. Postnatally, a subset of visceral WAT continues to arise from Wt1-expressing cells, consistent with the finding that Wt1 marks a proportion of cell populations enriched in WAT progenitors. We show that all visceral fat depots have a mesothelial layer like the visceral organs with which they are associated, and provide several lines of evidence that Wt1-expressing mesothelium can produce adipocytes. These results reveal a major ontogenetic difference between visceral and subcutaneous WAT, and pinpoint the lateral plate mesoderm as a major source of visceral WAT. They also support the notion that visceral WAT progenitors are heterogeneous, and suggest that mesothelium is a source of adipocytes.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , WT1 Proteins/metabolism , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/embryology , Adipose Tissue, White/cytology , Adipose Tissue, White/embryology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Lineage/genetics , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Tamoxifen/pharmacology , WT1 Proteins/genetics
10.
Nat Med ; 19(10): 1338-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995282

ABSTRACT

White adipose tissue displays high plasticity. We developed a system for the inducible, permanent labeling of mature adipocytes that we called the AdipoChaser mouse. We monitored adipogenesis during development, high-fat diet (HFD) feeding and cold exposure. During cold-induced 'browning' of subcutaneous fat, most 'beige' adipocytes stem from de novo-differentiated adipocytes. During HFD feeding, epididymal fat initiates adipogenesis after 4 weeks, whereas subcutaneous fat undergoes hypertrophy for a period of up to 12 weeks. Gonadal fat develops postnatally, whereas subcutaneous fat develops between embryonic days 14 and 18. Our results highlight the extensive differences in adipogenic potential in various fat depots.


Subject(s)
Adipogenesis , Adipose Tissue, White/physiology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/embryology , Adipose Tissue, Brown/physiology , Adipose Tissue, White/cytology , Adipose Tissue, White/embryology , Animals , Cell Differentiation , Cold Temperature , Dietary Fats/administration & dosage , Hyperplasia , Mice
SELECTION OF CITATIONS
SEARCH DETAIL