Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38948810

ABSTRACT

Objective: Platelet Derived Growth Factor Receptor Beta (Pdgfrß) suppresses the formation of cold temperature-induced beige adipocytes in aged mammals. We aimed to determine if deleting Pdgfrß in aged mice could rejuvenate metabolically active beige adipocytes by activating group 2 innate lymphoid cells (ILC2), and whether this effect could counteract diet-induced obesity-associated beige fat decline. Methods: We employed Pdgfrß gain-of-function and loss-of-function mouse models targeting beige adipocyte progenitor cells (APCs). Our approach included cold exposure, metabolic cage analysis, and age and diet-induced obesity models to examine beige fat development and metabolic function under varied Pdgfrß activity. Results: Acute cold exposure alone enhanced metabolic benefits in aged mice, irrespective of beige fat generation. However, Pdgfrß deletion in aged mice reestablished the formation of metabolically functional beige adipocytes, enhancing metabolism. Conversely, constitutive Pdgfrß activation in young mice stymied beige fat development. Mechanistically, Pdgfrß deletion upregulated IL-33, promoting ILC2 recruitment and activation, whereas Pdgfrß activation reduced IL-33 levels and suppressed ILC2 activity. Notably, diet-induced obesity markedly increased Pdgfrß expression and Stat1 signaling, which inhibited IL-33 induction and ILC2 activation. Genetic deletion of Pdgfrß restored beige fat formation in obese mice, improving whole-body metabolism. Conclusion: This study reveals that cold temperature exposure alone can trigger metabolic activation in aged mammals. However, reversing Pdgfrß signaling in aged and obese mice not only restores beige fat formation but also renews metabolic function and enhances the immunological environment of white adipose tissue (WAT). These findings highlight Pdgfrß as a crucial target for therapeutic strategies aimed at combating age- and obesity-related metabolic decline.

2.
Sci Adv ; 10(26): eadn5229, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38924414

ABSTRACT

There is a regional preference around lymph nodes (LNs) for adipose beiging. Here, we show that local LN removal within inguinal white adipose tissue (iWAT) greatly impairs cold-induced beiging, and this impairment can be restored by injecting M2 macrophages or macrophage-derived C-C motif chemokine (CCL22) into iWAT. CCL22 injection into iWAT effectively promotes iWAT beiging, while blocking CCL22 with antibodies can prevent it. Mechanistically, the CCL22 receptor, C-C motif chemokine receptor 4 (CCR4), within eosinophils and its downstream focal adhesion kinase/p65/interleukin-4 signaling are essential for CCL22-mediated beige adipocyte formation. Moreover, CCL22 levels are inversely correlated with body weight and fat mass in mice and humans. Acute elevation of CCL22 levels effectively prevents diet-induced body weight and fat gain by enhancing adipose beiging. Together, our data identify the CCL22-CCR4 axis as an essential mediator for LN-controlled adaptive thermogenesis and highlight its potential to combat obesity and its associated complications.


Subject(s)
Adipose Tissue, White , Chemokine CCL22 , Energy Metabolism , Lymph Nodes , Macrophages , Thermogenesis , Animals , Female , Humans , Male , Mice , Adipocytes, Beige/metabolism , Adipose Tissue, White/metabolism , Chemokine CCL22/metabolism , Eosinophils/metabolism , Lymph Nodes/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Obesity/metabolism , Receptors, CCR4/metabolism , Signal Transduction
3.
Cell Rep ; 43(5): 114169, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38678562

ABSTRACT

Sympathetic innervation of brown adipose tissue (BAT) controls mammalian adaptative thermogenesis. However, the cellular and molecular underpinnings contributing to BAT innervation remain poorly defined. Here, we show that smooth muscle cells (SMCs) support BAT growth, lipid utilization, and thermogenic plasticity. Moreover, we find that BAT SMCs express and control the bioavailability of Cxcl12. SMC deletion of Cxcl12 fosters brown adipocyte lipid accumulation, reduces energy expenditure, and increases susceptibility to diet-induced metabolic dysfunction. Mechanistically, we find that Cxcl12 stimulates CD301+ macrophage recruitment and supports sympathetic neuronal maintenance. Administering recombinant Cxcl12 to obese mice or leptin-deficient (Ob/Ob) mice is sufficient to boost macrophage presence and drive sympathetic innervation to restore BAT morphology and thermogenic responses. Altogether, our data reveal an SMC chemokine-dependent pathway linking immunological infiltration and sympathetic innervation as a rheostat for BAT maintenance and thermogenesis.


Subject(s)
Adipose Tissue, Brown , Chemokine CXCL12 , Macrophages , Myocytes, Smooth Muscle , Sympathetic Nervous System , Thermogenesis , Animals , Chemokine CXCL12/metabolism , Macrophages/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/innervation , Mice , Myocytes, Smooth Muscle/metabolism , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiology , Mice, Inbred C57BL , Male , Energy Metabolism , Obesity/metabolism , Obesity/pathology
4.
iScience ; 27(1): 108682, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38235323

ABSTRACT

White adipose tissue (WAT) development and adult homeostasis rely on distinct adipocyte progenitor cells (APCs). While adult APCs are defined early during embryogenesis and generate adipocytes after WAT organogenesis, the mechanisms underlying adult adipose lineage determination and preservation remain undefined. Here, we uncover a critical role for platelet-derived growth factor receptor beta (Pdgfrß) in identifying the adult APC lineage. Without Pdgfrß, APCs lose their adipogenic competency to incite fibrotic tissue replacement and inflammation. Through lineage tracing analysis, we reveal that the adult APC lineage is lost and develops into macrophages when Pdgfrß is deleted embryonically. Moreover, to maintain the APC lineage, Pdgfrß activation stimulates p38/MAPK phosphorylation to promote APC proliferation and maintains the APC state by phosphorylating peroxisome proliferator activated receptor gamma (Pparγ) at serine 112. Together, our findings identify a role for Pdgfrß acting as a rheostat for adult adipose lineage confinement to prevent unintended lineage switches.

5.
Front Endocrinol (Lausanne) ; 14: 1150059, 2023.
Article in English | MEDLINE | ID: mdl-37020585

ABSTRACT

The ability to generate thermogenic fat could be a targeted therapy to thwart obesity and improve metabolic health. Brown and beige adipocytes are two types of thermogenic fat cells that regulate energy balance. Both adipocytes share common morphological, biochemical, and thermogenic properties. Yet, recent evidence suggests unique features exist between brown and beige adipocytes, such as their cellular origin and thermogenic regulatory processes. Beige adipocytes also appear highly plastic, responding to environmental stimuli and interconverting between beige and white adipocyte states. Additionally, beige adipocytes appear to be metabolically heterogenic and have substrate specificity. Nevertheless, obese and aged individuals cannot develop beige adipocytes in response to thermogenic fat-inducers, creating a key clinical hurdle to their therapeutic promise. Thus, elucidating the underlying developmental, molecular, and functional mechanisms that govern thermogenic fat cells will improve our understanding of systemic energy regulation and strive for new targeted therapies to generate thermogenic fat. This review will examine the recent advances in thermogenic fat biogenesis, molecular regulation, and the potential mechanisms for their failure.


Subject(s)
Adipocytes, Beige , Adipocytes , Humans , Aged , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipocytes, Beige/metabolism , Energy Metabolism/physiology , Obesity/metabolism
6.
Nat Commun ; 14(1): 1806, 2023 03 31.
Article in English | MEDLINE | ID: mdl-37002214

ABSTRACT

Perivascular adipocyte progenitor cells (APCs) can generate cold temperature-induced thermogenic beige adipocytes within white adipose tissue (WAT), an effect that could counteract excess fat mass and metabolic pathologies. Yet, the ability to generate beige adipocytes declines with age, creating a key challenge for their therapeutic potential. Here we show that ageing beige APCs overexpress platelet derived growth factor receptor beta (Pdgfrß) to prevent beige adipogenesis. We show that genetically deleting Pdgfrß, in adult male mice, restores beige adipocyte generation whereas activating Pdgfrß in juvenile mice blocks beige fat formation. Mechanistically, we find that Stat1 phosphorylation mediates Pdgfrß beige APC signaling to suppress IL-33 induction, which dampens immunological genes such as IL-13 and IL-5. Moreover, pharmacologically targeting Pdgfrß signaling restores beige adipocyte development by rejuvenating the immunological niche. Thus, targeting Pdgfrß signaling could be a strategy to restore WAT immune cell function to stimulate beige fat in adult mammals.


Subject(s)
Adipocytes , Adipogenesis , Male , Mice , Animals , Adipogenesis/genetics , Adipocytes/metabolism , Signal Transduction , Adipose Tissue, White/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Thermogenesis/genetics , Mammals/metabolism
7.
Front Endocrinol (Lausanne) ; 13: 889923, 2022.
Article in English | MEDLINE | ID: mdl-35721736

ABSTRACT

Obesity and its' associated metabolic diseases such as type 2 diabetes and cardiometabolic disorders are significant health problems confronting many countries. A major driver for developing obesity and metabolic dysfunction is the uncontrolled expansion of white adipose tissue (WAT). Specifically, the pathophysiological expansion of visceral WAT is often associated with metabolic dysfunction due to changes in adipokine secretion profiles, reduced vascularization, increased fibrosis, and enrichment of pro-inflammatory immune cells. A critical determinate of body fat distribution and WAT health is the sex steroid estrogen. The bioavailability of estrogen appears to favor metabolically healthy subcutaneous fat over visceral fat growth while protecting against changes in metabolic dysfunction. Our review will focus on the role of estrogen on body fat partitioning, WAT homeostasis, adipogenesis, adipocyte progenitor cell (APC) function, and thermogenesis to control WAT health and systemic metabolism.


Subject(s)
Diabetes Mellitus, Type 2 , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Diabetes Mellitus, Type 2/complications , Estrogens/metabolism , Humans , Obesity/complications
8.
Commun Biol ; 5(1): 584, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35701601

ABSTRACT

Beige adipocytes are induced by cold temperatures or ß3-adrenergic receptor (Adrb3) agonists. They create heat through glucose and fatty acid (FA) oxidation, conferring metabolic benefits. The distinct and shared mechanisms by which these treatments induce beiging are unknown. Here, we perform single-nucleus assay for transposase-accessible chromatin sequencing (snATAC-seq) on adipose tissue from mice exposed to cold or an Adrb3 agonist to identify cellular and chromatin accessibility dynamics during beiging. Both stimuli induce chromatin remodeling that influence vascularization and inflammation in adipose. Beige adipocytes from cold-exposed mice have increased accessibility at genes regulating glycolytic processes, whereas Adrb3 activation increases cAMP responses. While both thermogenic stimuli increase accessibility at genes regulating thermogenesis, lipogenesis, and beige adipocyte development, the kinetics and magnitudes of the changes are distinct for the stimuli. Accessibility changes at lipogenic genes are linked to functional changes in lipid composition of adipose. Both stimuli tend to decrease the proportion of palmitic acids, a saturated FA in adipose. However, Adrb3 activation increases the proportion of monounsaturated FAs, whereas cold increases the proportion of polyunsaturated FAs. These findings reveal common and distinct mechanisms of cold and Adrb3 induced beige adipocyte biogenesis, and identify unique functional consequences of manipulating these pathways in vivo.


Subject(s)
Adipocytes, Beige , Gene Regulatory Networks , Adipocytes, Beige/metabolism , Adipose Tissue , Animals , Chromatin/metabolism , Mice , Thermogenesis/genetics
9.
Dev Cell ; 56(7): 985-999.e4, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33711247

ABSTRACT

Thermogenic beige fat found in white adipose tissue is a potential therapeutic target to curb the global obesity and diabetes epidemic. However, these inducible thermogenic beige adipocytes have been thought to be short-lived and to rapidly convert to "white-like" adipocytes after discontinuing stimuli. In this study, using effective labeling techniques and genetic mouse tools, we demonstrate that a subset of UCP1+ cells that exist within white adipose tissue are able to self-divide and contribute to new beige adipocyte recruitment in response to ß3 stimuli. When these cells are depleted or their adipogenic capability is blocked, ß3-induced beige adipocyte formation is impaired. We also identify a cell-cycle machinery of p21 and CDKN2A as a molecular basis of beige adipocyte regulation. Collectively, our findings provide new insights into the cellular and molecular mechanisms of beige adipocyte regulation and potential therapeutic opportunities to induce the beige phenotype and treat metabolic disease.


Subject(s)
Adipocytes, Beige/physiology , Adipose Tissue, White/cytology , Stem Cells/physiology , Uncoupling Protein 1/analysis , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Deletion , Genes, cdc , Male , Mice , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
10.
Elife ; 92020 06 19.
Article in English | MEDLINE | ID: mdl-32553115

ABSTRACT

Adipocytes arise from distinct progenitor populations during developmental and adult stages but little is known about how developmental progenitors differ from adult progenitors. Here, we investigate the role of platelet-derived growth factor receptor alpha (PDGFRα) in the divergent regulation of the two different adipose progenitor cells (APCs). Using in vivo adipose lineage tracking and deletion mouse models, we found that developmental PDGFRα+ cells are adipogenic and differentiated into mature adipocytes, and the deletion of Pdgfra in developmental adipose lineage disrupted white adipose tissue (WAT) formation. Interestingly, adult PDGFRα+ cells do not significantly contribute to adult adipogenesis, and deleting Pdgfra in adult adipose lineage did not affect WAT homeostasis. Mechanistically, embryonic APCs require PDGFRα for fate maintenance, and without PDGFRα, they underwent fate change from adipogenic to fibrotic lineage. Collectively, our findings indicate that PDGFRα+ cells and Pdgfra gene itself are differentially required for WAT development and adult WAT homeostasis.


Subject(s)
Adipogenesis/genetics , Adipose Tissue/growth & development , Homeostasis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stem Cells/metabolism , Adipose Tissue/metabolism , Adipose Tissue, White/growth & development , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Male , Mice , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/metabolism
11.
Genes Dev ; 33(19-20): 1367-1380, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31488578

ABSTRACT

Fat storage in adult mammals is a highly regulated process that involves the mobilization of adipocyte progenitor cells (APCs) that differentiate to produce new adipocytes. Here we report a role for the broadly conserved miR-26 family of microRNAs (miR-26a-1, miR-26a-2, and miR-26b) as major regulators of APC differentiation and adipose tissue mass. Deletion of all miR-26-encoding loci in mice resulted in a dramatic expansion of adipose tissue in adult animals fed normal chow. Conversely, transgenic overexpression of miR-26a protected mice from high-fat diet-induced obesity. These effects were attributable to a cell-autonomous function of miR-26 as a potent inhibitor of APC differentiation. miR-26 blocks adipogenesis, at least in part, by repressing expression of Fbxl19, a conserved miR-26 target without a previously known role in adipocyte biology that encodes a component of SCF-type E3 ubiquitin ligase complexes. These findings have therefore revealed a novel pathway that plays a critical role in regulating adipose tissue formation in vivo and suggest new potential therapeutic targets for obesity and related disorders.


Subject(s)
Adipogenesis/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , MicroRNAs/metabolism , Obesity/genetics , Stem Cells/cytology , Animals , Diet, High-Fat , Gene Expression , Gene Knockdown Techniques , Mice , MicroRNAs/genetics
12.
Elife ; 62017 10 11.
Article in English | MEDLINE | ID: mdl-29019320

ABSTRACT

Beige/brite adipocytes are induced within white adipose tissues (WAT) and, when activated, consume glucose and fatty acids to produce heat. Classically, two stimuli have been used to trigger a beiging response: cold temperatures and ß3-adrenergic receptor (Adrb3) agonists. These two beiging triggers have been used interchangeably but whether these two stimuli may induce beiging differently at cellular and molecular levels remains unclear. Here, we found that cold-induced beige adipocyte formation requires Adrb1, not Adrb3, activation. Adrb1 activation stimulates WAT resident perivascular (Acta2+) cells to form cold-induced beige adipocytes. In contrast, Adrb3 activation stimulates mature white adipocytes to convert into beige adipocytes. Necessity tests, using mature adipocyte-specific Prdm16 deletion strategies, demonstrated that adipocytes are required and are predominant source to generate Adrb3-induced, but not cold-induced, beige adipocytes. Collectively, we identify that cold temperatures and Adrb3 agonists activate distinct cellular populations that express different ß-adrenergic receptors to induce beige adipogenesis.


Subject(s)
Adipocytes, Beige/physiology , Cell Differentiation , Receptors, Adrenergic, beta-3/metabolism , Animals , Cold Temperature , Mice, Inbred C57BL , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-3/genetics
13.
Nat Commun ; 8: 15926, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28649987

ABSTRACT

Adipose progenitor cells (APCs) reside in a vascular niche, located within the perivascular compartment of adipose tissue blood vessels. Yet, the signals and mechanisms that govern adipose vascular niche formation and APC niche interaction are unknown. Here we show that the assembly and maintenance of the adipose vascular niche is controlled by PPARγ acting within APCs. PPARγ triggers a molecular hierarchy that induces vascular sprouting, APC vessel niche affinity and APC vessel occupancy. Mechanistically, PPARγ transcriptionally activates PDGFRß and VEGF. APC expression and activation of PDGFRß promotes the recruitment and retention of APCs to the niche. Pharmacologically, targeting PDGFRß disrupts APC niche contact thus blocking adipose tissue expansion. Moreover, enhanced APC expression of VEGF stimulates endothelial cell proliferation and expands the adipose niche. Consequently, APC niche communication and retention are boosted by VEGF thereby impairing adipogenesis. Our data indicate that APCs direct adipose tissue niche expansion via a PPARγ-initiated PDGFRß and VEGF transcriptional axis.


Subject(s)
Adipocytes/metabolism , PPAR gamma/metabolism , Stem Cell Niche , Stem Cells/metabolism , Adipocytes/cytology , Adipogenesis , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Proliferation , Female , Male , Mice , PPAR gamma/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stem Cells/cytology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Cell Metab ; 25(1): 166-181, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27889388

ABSTRACT

Cold temperatures induce progenitor cells within white adipose tissue to form beige adipocytes that burn energy and generate heat; this is a potential anti-diabesity therapy. However, the potential to form cold-induced beige adipocytes declines with age. This creates a clinical roadblock to potential therapeutic use in older individuals, who constitute a large percentage of the obesity epidemic. Here we show that aging murine and human beige progenitor cells display a cellular aging, senescence-like phenotype that accounts for their age-dependent failure. Activating the senescence pathway, either genetically or pharmacologically, in young beige progenitors induces premature cellular senescence and blocks their potential to form cold-induced beige adipocytes. Conversely, genetically or pharmacologically reversing cellular aging by targeting the p38/MAPK-p16Ink4a pathway in aged mouse or human beige progenitor cells rejuvenates cold-induced beiging. This in turn increases glucose sensitivity. Collectively, these data indicate that anti-aging or senescence modalities could be a strategy to induce beiging, thereby improving metabolic health in aging humans.


Subject(s)
Adipocytes, Beige/cytology , Adipocytes, Beige/metabolism , Aging/physiology , Cellular Senescence , Cold Temperature , Actins/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Male , Mice, Inbred C57BL , Phenotype , Stem Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Trends Endocrinol Metab ; 27(8): 574-585, 2016 08.
Article in English | MEDLINE | ID: mdl-27262681

ABSTRACT

Stem or progenitor cells are an essential component for the development, homeostasis, expansion, and regeneration of many tissues. Within white adipose tissue (WAT) reside vascular-resident adipose progenitor cells (APCs) that can proliferate and differentiate into either white or beige/brite adipocytes, which may control adiposity. Recent studies have begun to show that APCs can be manipulated to control adiposity and counteract 'diabesity'. However, much remains unknown about the identity of APCs and how they may control adiposity in response to homeostatic and external cues. Here, we discuss recent advances in our understanding of adipose progenitors and cover a range of topics, including the stem cell/progenitor lineage, their niche, their developmental and adult roles, and their role in cold-induced beige/brite adipocyte formation.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/metabolism , Stem Cells/cytology , Animals , Homeostasis/genetics , Homeostasis/physiology , Humans , Thermogenesis/genetics , Thermogenesis/physiology
17.
Nat Commun ; 7: 10184, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26729601

ABSTRACT

Cold temperatures induce formation of beige adipocytes, which convert glucose and fatty acids to heat, and may increase energy expenditure, reduce adiposity and lower blood glucose. This therapeutic potential is unrealized, hindered by a dearth of genetic tools to fate map, track and manipulate beige progenitors and 'beiging'. Here we examined 12 Cre/inducible Cre mouse strains that mark adipocyte, muscle and mural lineages, three proposed beige origins. Among these mouse strains, only those that marked perivascular mural cells tracked the cold-induced beige lineage. Two SMA-based strains, SMA-Cre(ERT2) and SMA-rtTA, fate mapped into the majority of cold-induced beige adipocytes and SMA-marked progenitors appeared essential for beiging. Disruption of the potential of the SMA-tracked progenitors to form beige adipocytes was accompanied by an inability to maintain body temperature and by hyperglycaemia. Thus, SMA-engineered mice may be useful to track and manipulate beige progenitors, beige adipocyte formation and function.


Subject(s)
Adipocytes/classification , Adipocytes/metabolism , Cold Temperature , Animals , Blood Vessels/cytology , Blood Vessels/metabolism , Cells, Cultured , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Inbred Strains , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism
18.
Cell Rep ; 9(3): 1007-22, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25437556

ABSTRACT

Adipose tissues have striking plasticity, highlighted by childhood and adult obesity. Using adipose lineage analyses, smooth muscle actin (SMA)-mural cell-fate mapping, and conditional PPARγ deletion to block adipocyte differentiation, we find two phases of adipocyte generation that emanate from two independent adipose progenitor compartments: developmental and adult. These two compartments are sequentially required for organ formation and maintenance. Although both developmental and adult progenitors are specified during the developmental period and express PPARγ, they have distinct microanatomical, functional, morphogenetic, and molecular profiles. Furthermore, the two compartments derive from different lineages; whereas adult adipose progenitors fate-map from an SMA+ mural lineage, developmental progenitors do not. Remarkably, the adult progenitor compartment appears to be specified earlier than the developmental cells and then enters the already developmentally formed adipose depots. Thus, two distinct cell compartments control adipose organ development and organ homeostasis, which may provide a discrete therapeutic target for childhood and adult obesity.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/growth & development , Cell Lineage , Homeostasis , Organogenesis , Stem Cells/cytology , Adipose Tissue/metabolism , Aging/metabolism , Animals , Mice , PPAR gamma/metabolism , Staining and Labeling , Stem Cells/metabolism , Time Factors
19.
J Biol Chem ; 289(49): 34065-73, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25320093

ABSTRACT

Cellular retinoic acid-binding protein 2 (CRABP2) potently suppresses the growth of various carcinomas, but the mechanism(s) that underlies this activity remains incompletely understood. CRABP2 displays two distinct functions. The classical function of this protein is to directly deliver retinoic acid (RA) to RA receptor (RAR), a nuclear receptor activated by this hormone, in turn inducing the expression of multiple antiproliferative genes. The other function of the protein is exerted in the absence of RA and mediated by the RNA-binding and stabilizing protein HuR. CRABP2 directly binds to HuR, markedly strengthens its interactions with target mRNAs, and thus increases their stability and up-regulates their expression. Here we show that the anticarcinogenic activities of CRABP2 are mediated by both of its functions. Transcriptome analyses revealed that, in the absence of RA, a large cohort of transcripts is regulated in common by CRABP2 and HuR, and many of these are involved in regulation of oncogenic properties. Furthermore, both in cultured cells and in vivo, CRABP2 or a CRABP2 mutant defective in its ability to cooperate with RAR but competent in interactions with HuR suppressed carcinoma growth and did so in the absence of RA. Hence, transcript stabilization by the CRABP2-HuR complex significantly contributes to the ability of CRABP2 to inhibit tumorigenesis. Surprisingly, the observations also revealed that HuR regulates the expression of multiple genes involved in nuclear pore formation and is required for nuclear import of CRABP2 and for transcriptional activation by RAR. The data thus point at a novel function for this important protein.


Subject(s)
ELAV Proteins/genetics , Gene Expression Regulation, Neoplastic , RNA, Messenger/genetics , Receptors, Retinoic Acid/genetics , Transcriptional Activation , Active Transport, Cell Nucleus , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , ELAV Proteins/metabolism , ELAV-Like Protein 1 , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , RNA Stability , RNA, Messenger/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction , Tretinoin/metabolism , Tretinoin/pharmacology
20.
Cancer Res ; 74(21): 6341-51, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25237067

ABSTRACT

Vitamin A, retinol, circulates in blood bound to retinol-binding protein (RBP). At some tissues, RBP is recognized by STRA6, a plasma membrane protein that serves a dual role: it transports retinol from extracellular RBP into cells and it transduces a signaling cascade mediated by the Janus kinase JAK2 and the transcription factors STAT3 and STAT5. We show here that expression of RBP and STRA6 is markedly upregulated in human breast and colon tumors, that holo-RBP/STRA6 signaling promotes oncogenic properties, and that STRA6 expression is critical for tumor formation by colon carcinoma cells in vivo. The holo-RBP/STRA6 pathway also efficiently induces fibroblasts to undergo oncogenic transformation, rendering them highly tumorigenic. These data establish that holo-RBP and its receptor STRA6 are potent oncogenes and suggest that the pathway is a novel target for therapy of some human cancers.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Membrane Proteins/genetics , Vitamin A/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Hep G2 Cells , Humans , Janus Kinase 2/genetics , Membrane Proteins/metabolism , Retinol-Binding Proteins/biosynthesis , Retinol-Binding Proteins/genetics , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...