Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Cell ; 184(13): 3502-3518.e33, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34048700

ABSTRACT

Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of ß-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3.


Subject(s)
Adipose Tissue, Brown/metabolism , Constitutive Androstane Receptor/metabolism , Lipolysis , Receptors, G-Protein-Coupled/metabolism , Thermogenesis , Adipocytes/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cold Temperature , Dietary Fats/pharmacology , Humans , Mice, Inbred C57BL , Phenotype , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Sympathetic Nervous System/metabolism , Transcription, Genetic
2.
Dev Biol ; 477: 164-176, 2021 09.
Article in English | MEDLINE | ID: mdl-34023333

ABSTRACT

Intraflagellar transport (IFT) is an evolutionarily conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella, including mammalian sperm tails. Depletion of IFT27, a component of the IFT complex, in male germ cells results in infertility associated with disrupted sperm flagella structure and motility. Leucine zipper transcription factor-like 1 (LZTFL1) is an IFT27 associated protein. LZTFL1, also known as BBS17, is a Bardet-Biedl syndrome (BBS) associated protein. Patients carrying biallelic variants of LZTFL1 gene exhibit the common BBS phenotypes. The global Lztfl1 knockout mice showed abnormal growth rate and retinal degeneration, typical of BBS phenotype. However, it is not clear if Lztfl1 has a role in male fertility. The LZTFL1 protein is highly and predominantly expressed in mouse testis. During the first wave of spermatogenesis, the protein is only expressed during spermiogenesis phase from the round spermatid stage and displays a cytoplasmic localization with a vesicular distribution pattern. At the elongated spermatid stage, LZTFL1 is present in the developing flagella and appears also close to the manchette. Fertility of Lztfl1 knockout mice was significantly reduced and associated with low sperm motility and a high level of abnormal sperm (astheno-teratozoospermia). In vitro assessment of fertility revealed reduced fertilization and embryonic development when using sperm from homozygous mutant mice. In addition, we observed a significant decrease of the testicular IFT27 protein level in Lztfl1 mutant mice contrasting with a stable expression levels of other IFT proteins, including IFT20, IFT81, IFT88 and IFT140. Overall, our results support strongly the important role of LZTFL1 in mouse spermatogenesis and male fertility.


Subject(s)
Fertility/physiology , Spermatozoa/physiology , Transcription Factors/physiology , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , Female , Fertility/genetics , HEK293 Cells , Humans , Male , Mice, Knockout , Protein Binding , RNA, Messenger/metabolism , Spermatogenesis/genetics , Spermatogenesis/physiology , Transcription Factors/genetics , rab GTP-Binding Proteins/physiology
3.
J Biol Chem ; 296: 100104, 2021.
Article in English | MEDLINE | ID: mdl-33219129

ABSTRACT

ABHD5 is an essential coactivator of ATGL, the rate-limiting triglyceride (TG) lipase in many cell types. Importantly, ABHD5 also functions as a tumor suppressor, and ABHD5 mRNA expression levels correlate with patient survival for several cancers. Nevertheless, the mechanisms involved in ABHD5-dependent tumor suppression are not known. We found that overexpression of ABHD5 induces cell cycle arrest at the G1 phase and causes growth retardation in a panel of prostate cancer cells. Transcriptomic profiling and biochemical analysis revealed that genetic or pharmacological activation of lipolysis by ABHD5 potently inhibits mTORC1 signaling, leading to a significant downregulation of protein synthesis. Mechanistically, we found that ABHD5 elevates intracellular AMP content, which activates AMPK, leading to inhibition of mTORC1. Interestingly, ABHD5-dependent suppression of mTORC1 was abrogated by pharmacological inhibition of DGAT1 or DGAT2, isoenzymes that re-esterify fatty acids in a process that consumes ATP. Collectively, this study maps out a novel molecular pathway crucial for limiting cancer cell proliferation, in which ABHD5-mediated lipolysis creates an energy-consuming futile cycle between TG hydrolysis and resynthesis, leading to inhibition of mTORC1 and cancer cell growth arrest.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , AMP-Activated Protein Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Humans , Lipolysis/physiology , Metabolism/physiology , RNA-Seq
4.
J Lipid Res ; 62: 100016, 2021.
Article in English | MEDLINE | ID: mdl-33334871

ABSTRACT

Perilipin 5 (PLIN5) is a lipid-droplet-associated protein that coordinates intracellular lipolysis in highly oxidative tissues and is thought to regulate lipid metabolism in response to phosphorylation by protein kinase A (PKA). We sought to identify PKA phosphorylation sites in PLIN5 and assess their functional relevance in cultured cells and the livers of mice. We detected phosphorylation on S155 and identified S155 as a functionally important site for lipid metabolism. Expression of phosphorylation-defective PLIN5 S155A in Plin5 null cells resulted in decreased rates of lipolysis and triglyceride-derived fatty acid oxidation. FLIM-FRET analysis of protein-protein interactions showed that PLIN5 S155 phosphorylation regulates PLIN5 interaction with adipose triglyceride lipase at the lipid droplet, but not with α-ß hydrolase domain-containing 5. Re-expression of PLIN5 S155A in the liver of Plin5 liver-specific null mice reduced lipolysis compared with wild-type PLIN5 re-expression, but was not associated with other changes in hepatic lipid metabolism. Furthermore, glycemic control was impaired in mice with expression of PLIN5 S155A compared with mice expressing PLIN5. Together, these studies demonstrate that PLIN5 S155 is required for PKA-mediated lipolysis and builds on the body of evidence demonstrating a critical role for PLIN5 in coordinating lipid and glucose metabolism.


Subject(s)
Perilipin-5
5.
Biochem J ; 477(3): 583-600, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32026949

ABSTRACT

A central function of adipose tissue is in the management of systemic energy homeostasis that is achieved through the co-ordinated regulation of energy storage and mobilization, adipokine release, and immune functions. With the dramatic increase in the prevalence of obesity and obesity-related metabolic disease over the past 30 years, there has been extensive interest in targeting adipose tissue for therapeutic benefit. However, in order for this goal to be achieved it is essential to establish a comprehensive atlas of adipose tissue cellular composition and define mechanisms of intercellular communication that mediate pathologic and therapeutic responses. While traditional methods, such as fluorescence-activated cell sorting (FACS) and genetic lineage tracing, have greatly advanced the field, these approaches are inherently limited by the choice of markers and the ability to comprehensively identify and characterize dynamic interactions among stromal cells within the tissue microenvironment. Single cell RNA sequencing (scRNAseq) has emerged as a powerful tool for deconvolving cellular heterogeneity and holds promise for understanding the development and plasticity of adipose tissue under normal and pathological conditions. scRNAseq has recently been used to characterize adipose stem cell (ASC) populations and has provided new insights into subpopulations of macrophages that arise during anabolic and catabolic remodeling in white adipose tissue. The current review summarizes recent findings that use this technology to explore adipose tissue heterogeneity and plasticity.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White , Macrophages/metabolism , Stromal Cells/cytology , Adipocytes/immunology , Adipose Tissue/cytology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Cell Communication , Flow Cytometry , Humans , Macrophages/cytology , Obesity/metabolism , Obesity/therapy , Sequence Analysis, RNA , Single-Cell Analysis , Stromal Cells/metabolism , Stromal Cells/pathology
6.
Circulation ; 138(3): 305-315, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30012703

ABSTRACT

Lipid droplets (LDs) are distinct and dynamic organelles that affect the health of cells and organs. Much progress has been made in understanding how these structures are formed, how they interact with other cellular organelles, how they are used for storage of triacylglycerol in adipose tissue, and how they regulate lipolysis. Our understanding of the biology of LDs in the heart and vascular tissue is relatively primitive in comparison with LDs in adipose tissue and liver. The National Heart, Lung, and Blood Institute convened a working group to discuss how LDs affect cardiovascular diseases. The goal of the working group was to examine the current state of knowledge on the cell biology of LDs, including current methods to study them in cells and organs and reflect on how LDs influence the development and progression of cardiovascular diseases. This review summarizes the working group discussion and recommendations on research areas ripe for future investigation that will likely improve our understanding of atherosclerosis and heart function.


Subject(s)
Cardiovascular Diseases/metabolism , Lipid Droplets/metabolism , Myocardium/metabolism , Animals , Cardiovascular Diseases/genetics , Consensus Development Conferences, NIH as Topic , Disease Models, Animal , Gene-Environment Interaction , Humans , Lipid Metabolism , National Heart, Lung, and Blood Institute (U.S.) , United States
7.
J Pharmacol Exp Ther ; 363(3): 367-376, 2017 12.
Article in English | MEDLINE | ID: mdl-28928121

ABSTRACT

Current knowledge regarding acute regulation of adipocyte lipolysis is largely based on receptor-mediated activation or inhibition of pathways that influence intracellular levels of cAMP, thereby affecting protein kinase A (PKA) activity. We recently identified synthetic ligands of α-ß-hydrolase domain containing 5 (ABHD5) that directly activate adipose triglyceride lipase (ATGL) by dissociating ABHD5 from its inhibitory regulator, perilipin-1 (PLIN1). In the current study, we used these novel ligands to determine the direct contribution of ABHD5 to various aspects of lipolysis control in white (3T3-L1) and brown adipocytes. ABHD5 ligands stimulated adipocyte lipolysis without affecting PKA-dependent phosphorylation on consensus sites of PLIN1 or hormone-sensitive lipase (HSL). Cotreatment of adipocytes with synthetic ABHD5 ligands did not alter the potency or maximal lipolysis efficacy of the ß-adrenergic receptor (ADRB) agonist isoproterenol (ISO), indicating that both target a common pool of ABHD5. Reducing ADRB/PKA signaling with insulin or desensitizing ADRB suppressed lipolysis responses to a subsequent challenge with ISO, but not to ABHD5 ligands. Lastly, despite strong treatment differences in PKA-dependent phosphorylation of HSL, we found that ligand-mediated activation of ABHD5 led to complete triglyceride hydrolysis, which predominantly involved ATGL, but also HSL. These results indicate that the overall pattern of lipolysis controlled by ABHD5 ligands is similar to that of isoproterenol, and that ABHD5 plays a central role in the regulation of adipocyte lipolysis. As lipolysis is critical for adaptive thermogenesis and in catabolic tissue remodeling, ABHD5 ligands may provide a means of activating these processes under conditions where receptor signaling is compromised.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Piperazines/pharmacology , Thiazepines/pharmacology , Urea/analogs & derivatives , Urea/pharmacology , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Female , Hydrolysis , Insulin/pharmacology , Ligands , Lipolysis , Mice , Perilipin-1/metabolism , Phosphorylation , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Sterol Esterase/metabolism , Triglycerides/metabolism
8.
Genome Res ; 23(11): 1885-93, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23934932

ABSTRACT

Identifying the pathways that are significantly impacted in a given condition is a crucial step in understanding the underlying biological phenomena. All approaches currently available for this purpose calculate a P-value that aims to quantify the significance of the involvement of each pathway in the given phenotype. These P-values were previously thought to be independent. Here we show that this is not the case, and that many pathways can considerably affect each other's P-values through a "crosstalk" phenomenon. Although it is intuitive that various pathways could influence each other, the presence and extent of this phenomenon have not been rigorously studied and, most importantly, there is no currently available technique able to quantify the amount of such crosstalk. Here, we show that all three major categories of pathway analysis methods (enrichment analysis, functional class scoring, and topology-based methods) are severely influenced by crosstalk phenomena. Using real pathways and data, we show that in some cases pathways with significant P-values are not biologically meaningful, and that some biologically meaningful pathways with nonsignificant P-values become statistically significant when the crosstalk effects of other pathways are removed. We describe a technique able to detect, quantify, and correct crosstalk effects, as well as identify independent functional modules. We assessed this novel approach on data from four experiments involving three phenotypes and two species. This method is expected to allow a better understanding of individual experiment results, as well as a more refined definition of the existing signaling pathways for specific phenotypes.


Subject(s)
Computational Biology/methods , Metabolic Networks and Pathways , Signal Transduction , Adipose Tissue, White/metabolism , Animals , Cervical Ripening , Cervix Uteri/metabolism , Female , Gene Expression , Humans , Mice , Models, Biological , Phenotype , Pregnancy , Species Specificity
9.
Am J Physiol Regul Integr Comp Physiol ; 310(1): R55-65, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26538237

ABSTRACT

De novo brown adipogenesis involves the proliferation and differentiation of progenitors, yet the mechanisms that guide these events in vivo are poorly understood. We previously demonstrated that treatment with a ß3-adrenergic receptor (ADRB3) agonist triggers brown/beige adipogenesis in gonadal white adipose tissue following adipocyte death and clearance by tissue macrophages. The close physical relationship between adipocyte progenitors and tissue macrophages suggested that the macrophages that clear dying adipocytes might generate proadipogenic factors. Flow cytometric analysis of macrophages from mice treated with CL 316,243 identified a subpopulation that contained elevated lipid and expressed CD44. Lipidomic analysis of fluorescence-activated cell sorting-isolated macrophages demonstrated that CD44+ macrophages contained four- to five-fold higher levels of the endogenous peroxisome-proliferator activated receptor gamma (PPARγ) ligands 9-hydroxyoctadecadienoic acid (HODE), and 13-HODE compared with CD44- macrophages. Gene expression profiling and immunohistochemistry demonstrated that ADRB3 agonist treatment upregulated expression of ALOX15, the lipoxygenase responsible for generating 9-HODE and 13-HODE. Using an in vitro model of adipocyte efferocytosis, we found that IL-4-primed tissue macrophages accumulated lipid from dying fat cells and upregulated expression of Alox15. Furthermore, treatment of differentiating adipocytes with 9-HODE and 13-HODE potentiated brown/beige adipogenesis. Collectively, these data indicate that noninflammatory removal of adipocyte remnants and coordinated generation of PPARγ ligands by M2 macrophages provides localized adipogenic signals to support de novo brown/beige adipogenesis.


Subject(s)
Adipocytes, White/drug effects , Adipogenesis/drug effects , Adipose Tissue, White/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Cell Communication/drug effects , Dioxoles/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipocytes, White/pathology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Cell Death , Cell Line , Cell Separation/methods , Coculture Techniques , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Ligands , Macrophages/metabolism , Male , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , Phagocytosis/drug effects , Phenotype , Signal Transduction/drug effects , Time Factors
10.
FASEB J ; 29(1): 286-99, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25392270

ABSTRACT

This work investigated how cold stress induces the appearance of brown adipocytes (BAs) in brown and white adipose tissues (WATs) of adult mice. In interscapular brown adipose tissue (iBAT), cold exposure increased proliferation of endothelial cells and interstitial cells expressing platelet-derived growth factor receptor, α polypeptide (PDGFRα) by 3- to 4-fold. Surprisingly, brown adipogenesis and angiogenesis were largely restricted to the dorsal edge of iBAT. Although cold stress did not increase proliferation in inguinal white adipose tissue (ingWAT), the percentage of BAs, defined as multilocular adipocytes that express uncoupling protein 1, rose from undetectable to 30% of total adipocytes. To trace the origins of cold-induced BAs, we genetically tagged PDGFRα(+) cells and adipocytes prior to cold exposure, using Pdgfra-Cre recombinase estrogen receptor T2 fusion protein (CreER(T2)) and adiponectin-CreER(T2), respectively. In iBAT, cold stress triggered the proliferation and differentiation of PDGFRα(+) cells into BAs. In contrast, all newly observed BAs in ingWAT (5207 out of 5207) were derived from unilocular adipocytes tagged by adiponectin-CreER(T2)-mediated recombination. Surgical denervation of iBAT reduced cold-induced brown adipogenesis by >85%, whereas infusion of norepinephrine (NE) mimicked the effects of cold in warm-adapted mice. NE-induced de novo brown adipogenesis in iBAT was eliminated in mice lacking ß1-adrenergic receptors. These observations identify a novel tissue niche for brown adipogenesis in iBAT and further define depot-specific mechanisms of BA recruitment.


Subject(s)
Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Cold Temperature/adverse effects , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Norepinephrine/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Adrenergic, beta-1/deficiency , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stress, Physiological , Sympathectomy
11.
J Biol Chem ; 289(46): 32178-32185, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25253697

ABSTRACT

Adipocyte lipolysis can increase the production of inflammatory cytokines such as interleukin-6 (IL-6) that promote insulin resistance. However, the mechanisms that link lipolysis with inflammation remain elusive. Acute activation of ß3-adrenergic receptors (ADRB3) triggers lipolysis and up-regulates production of IL-6 in adipocytes, and both of these effects are blocked by pharmacological inhibition of hormone-sensitive lipase. We report that stimulation of ADRB3 induces expression of sphingosine kinase 1 (SphK1) and increases sphingosine 1-phosphate production in adipocytes in a manner that also depends on hormone-sensitive lipase activity. Mechanistically, we found that adipose lipolysis-induced SphK1 up-regulation is mediated by the c-Jun N-terminal kinase (JNK)/activating protein-1 signaling pathway. Inhibition of SphK1 by sphingosine kinase inhibitor 2 diminished the ADRB3-induced IL-6 production both in vitro and in vivo. Induction of IL-6 by ADRB3 activation was suppressed by siRNA knockdown of Sphk1 in cultured adipocytes and was severely attenuated in Sphk1 null mice. Conversely, ectopic expression of SphK1 increased IL-6 expression in adipocytes. Collectively, these data demonstrate that SphK1 is a critical mediator in lipolysis-triggered inflammation in adipocytes.


Subject(s)
Adipocytes/cytology , Inflammation/metabolism , Interleukin-6/metabolism , Lipolysis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Signal Transduction , Sphingolipids/chemistry , Tandem Mass Spectrometry
12.
Biochim Biophys Acta ; 1842(3): 358-69, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23688783

ABSTRACT

Adipose tissue plays an essential role in regulating energy balance through its metabolic, cellular and endocrine functions. Adipose tissue has been historically classified into anabolic white adipose tissue and catabolic brown adipose tissue. An explosion of new data, however, points to the remarkable heterogeneity among the cells types that can become adipocytes, as well as the inherent metabolic plasticity of mature cells. These data indicate that targeting cellular and metabolic plasticity of adipose tissue might provide new avenues for treatment of obesity-related diseases. This review will discuss the developmental origins of adipose tissue, the cellular complexity of adipose tissues, and the identification of progenitors that contribute to adipogenesis throughout development. We will touch upon the pathological remodeling of adipose tissue and discuss how our understanding of adipose tissue remodeling can uncover new therapeutic targets. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.


Subject(s)
Adipogenesis/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Mesenchymal Stem Cells/cytology , Adipocytes/cytology , Adipose Tissue, Brown/growth & development , Adipose Tissue, White/growth & development , Animals , Body Fat Distribution , Cell Differentiation , Humans , Mice
13.
J Lipid Res ; 55(11): 2276-86, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25193997

ABSTRACT

Chronic activation of ß3-adrenergic receptors (ß3-ARs) expands the catabolic activity of both brown and white adipose tissue by engaging uncoupling protein 1 (UCP1)-dependent and UCP1-independent processes. The present work examined de novo lipogenesis (DNL) and TG/glycerol dynamics in classic brown, subcutaneous "beige," and classic white adipose tissues during sustained ß3-AR activation by CL 316,243 (CL) and also addressed the contribution of TG hydrolysis to these dynamics. CL treatment for 7 days dramatically increased DNL and TG turnover similarly in all adipose depots, despite great differences in UCP1 abundance. Increased lipid turnover was accompanied by the simultaneous upregulation of genes involved in FAS, glycerol metabolism, and FA oxidation. Inducible, adipocyte-specific deletion of adipose TG lipase (ATGL), the rate-limiting enzyme for lipolysis, demonstrates that TG hydrolysis is required for CL-induced increases in DNL, TG turnover, and mitochondrial electron transport in all depots. Interestingly, the effect of ATGL deletion on induction of specific genes involved in FA oxidation and synthesis varied among fat depots. Overall, these studies indicate that FAS and FA oxidation are tightly coupled in adipose tissues during chronic adrenergic activation, and this effect critically depends on the activity of adipocyte ATGL.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Lipogenesis/drug effects , Lipolysis/drug effects , Receptors, Adrenergic, beta-3/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Adiposity/drug effects , Animals , Dioxoles/pharmacology , Female , Gene Deletion , Gene Expression Regulation/drug effects , Glycerol/metabolism , Kinetics , Lipase/deficiency , Lipase/genetics , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Time Factors , Triglycerides/metabolism
14.
Am J Physiol Endocrinol Metab ; 307(9): E793-9, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25184993

ABSTRACT

Brown adipocytes (BA) generate heat in response to sympathetic activation and are the main site of nonshivering thermogenesis in mammals. Although most BA are located in classic brown adipose tissue depots, BA are also abundant in the inguinal white adipose tissue (iWAT) before weaning. The number of BA is correlated with the density of sympathetic innervation in iWAT; however, the role of continuous sympathetic tone in the establishment and maintenance of BA in WAT has not been investigated. BA marker expression in iWAT was abundant in weaning mice but was greatly reduced by 8 wk of age. Nonetheless, BA phenotype could be rapidly reinstated by acute ß3-adrenergic stimulation with CL-316,243 (CL). Genetic tagging of adipocytes with adiponectin-CreER(T2) demonstrated that CL reinstates uncoupling protein 1 (UCP1) expression in adipocytes that were present before weaning. Chronic surgical denervation dramatically reduced the ability of CL to induce the expression of UCP1 and other BA markers in the tissue as a whole, and this loss of responsiveness was prevented by concurrent treatment with CL. These results indicate that ongoing sympathetic activity is critical to preserve the ability of iWAT fat cells to express a BA phenotype upon adrenergic stimulation.


Subject(s)
Adipocytes, Brown/cytology , Adipogenesis , Aging , Subcutaneous Fat, Abdominal/cytology , Sympathetic Nervous System/metabolism , Synaptic Transmission , Adipocytes, Brown/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Biomarkers/metabolism , Crosses, Genetic , Denervation/adverse effects , Dioxoles/pharmacology , Gene Expression Regulation, Developmental/drug effects , Groin , Immunohistochemistry , Ion Channels/agonists , Ion Channels/metabolism , Mice, 129 Strain , Mice, Transgenic , Mitochondrial Proteins/agonists , Mitochondrial Proteins/metabolism , Subcutaneous Fat, Abdominal/growth & development , Subcutaneous Fat, Abdominal/innervation , Subcutaneous Fat, Abdominal/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/growth & development , Synaptic Transmission/drug effects , Uncoupling Protein 1 , Weaning
15.
Sci Rep ; 14(1): 19, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167670

ABSTRACT

Long-chain acyl-CoAs (LC-acyl-CoAs) are important intermediary metabolites and are also thought to function as intracellular signaling molecules; however, the direct effects of LC-acyl-CoAs have been difficult to determine in real-time and dissociate from Protein Kinase A (PKA) signaling. Here, we examined the direct role of lipolysis in generating intracellular LC-acyl-CoAs and activating AMPK in white adipocytes by pharmacological activation of ABHD5 (also known as CGI-58), a lipase co-activator. Activation of lipolysis in 3T3-L1 adipocytes independent of PKA with synthetic ABHD5 ligands, resulted in greater activation of AMPK compared to receptor-mediated activation with isoproterenol, a ß-adrenergic receptor agonist. Importantly, the effect of pharmacological activation of ABHD5 on AMPK activation was blocked by inhibiting ATGL, the rate-limiting enzyme for triacylglycerol hydrolysis. Utilizing a novel FRET sensor to detect intracellular LC-acyl-CoAs, we demonstrate that stimulation of lipolysis in 3T3-L1 adipocytes increased the production of LC-acyl-CoAs, an effect which was blocked by inhibition of ATGL. Moreover, ATGL inhibition blocked AMPKß1 S108 phosphorylation, a site required for allosteric regulation. Increasing intracellular LC-acyl-CoAs by removal of BSA in the media and pharmacological inhibition of DGAT1 and 2 resulted in greater activation of AMPK. Finally, inhibiting LC-acyl-CoA generation reduced activation of AMPK; however, did not lower energy charge. Overall, results demonstrate that lipolysis in white adipocytes directly results in allosteric activation of AMPK through the generation of LC-acyl-CoAs.


Subject(s)
Acyl Coenzyme A , Lipolysis , Mice , Animals , Acyl Coenzyme A/metabolism , AMP-Activated Protein Kinases/metabolism , Signal Transduction , Adipocytes, White/metabolism , 3T3-L1 Cells
16.
Cell Rep Methods ; 4(5): 100774, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38749444

ABSTRACT

We present methods for making and testing the membrane biophysics of model lipid droplets (LDs). Methods are described for imaging LDs ranging in size from 0.1 to 40 µm in diameter with high-resolution microscopy and spectroscopy. With known LD compositions, membrane binding, sorting, diffusion, and tension were measured via fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), fluorescence lifetime imaging microscopy (FLIM), atomic force microscopy (AFM), and imaging flow cytometry. Additionally, a custom, small-volume pendant droplet tensiometer is described and used to measure the association of phospholipids to the LD surface. These complementary, cross-validating methods of measuring LD membrane behavior reveal the interplay of biophysical processes on lipid droplet monolayers.


Subject(s)
Lipid Droplets , Lipid Droplets/metabolism , Lipid Droplets/chemistry , Microscopy, Atomic Force/methods , Microscopy, Fluorescence/methods , Fluorescence Recovery After Photobleaching/methods , Humans , Flow Cytometry/methods , Spectrometry, Fluorescence/methods
17.
Nat Commun ; 15(1): 2779, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38555350

ABSTRACT

Adipose tissue (AT) adapts to overnutrition in a complex process, wherein specialized immune cells remove and replace dysfunctional and stressed adipocytes with new fat cells. Among immune cells recruited to AT, lipid-associated macrophages (LAMs) have emerged as key players in obesity and in diseases involving lipid stress and inflammation. Here, we show that LAMs selectively express transmembrane 4 L six family member 19 (TM4SF19), a lysosomal protein that represses acidification through its interaction with Vacuolar-ATPase. Inactivation of TM4SF19 elevates lysosomal acidification and accelerates the clearance of dying/dead adipocytes in vitro and in vivo. TM4SF19 deletion reduces the LAM accumulation and increases the proportion of restorative macrophages in AT of male mice fed a high-fat diet. Importantly, male mice lacking TM4SF19 adapt to high-fat feeding through adipocyte hyperplasia, rather than hypertrophy. This adaptation significantly improves local and systemic insulin sensitivity, and energy expenditure, offering a potential avenue to combat obesity-related metabolic dysfunction.


Subject(s)
Insulin Resistance , Obesity , Male , Mice , Animals , Obesity/complications , Obesity/genetics , Adipose Tissue/metabolism , Inflammation/metabolism , Diet, High-Fat/adverse effects , Lysosomes/metabolism , Lipids , Macrophages/metabolism , Mice, Inbred C57BL
18.
J Biol Chem ; 287(30): 25038-48, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22685301

ABSTRACT

ß-Adrenergic receptors (ß-ARs) promote brown adipose tissue (BAT) thermogenesis by mobilizing fatty acids and inducing the expression of oxidative genes. ß-AR activation increases the expression of oxidative genes by elevating cAMP, but whether lipolytic products can modulate gene expression is not known. This study examined the role that adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) plays in the induction of gene expression. Activation of brown adipocytes by ß-AR agonism or 8-bromo-cyclic AMP increased the expression of PGC1α, PDK4, PPARα, uncoupling protein 1 (UCP1), and neuron-derived orphan receptor-1 (NOR-1), and concurrent inhibition of HSL reduced the induction of PGC1α, PDK4, PPARα, and UCP1 but not NOR-1. Similar results were observed in the BAT of mice following pharmacological or genetic inhibition of HSL and in brown adipocytes with stable knockdown of ATGL. Conversely, treatments that increase endogenous fatty acids elevated the expression of oxidative genes. Pharmacological antagonism and siRNA knockdown indicate that PPARα and PPARδ modulate the induction of oxidative genes by ß-AR agonism. Using a live cell fluorescent reporter assay of PPAR activation, we demonstrated that ligands for PPARα and -δ, but not PPARγ, were rapidly generated at the lipid droplet surface and could transcriptionally activate PPARα and -δ. Knockdown of ATGL reduced cAMP-mediated induction of genes involved in fatty acid oxidation and oxidative phosphorylation. Consequently, ATGL knockdown reduced maximal oxidation of fatty acids, but not pyruvate, in response to cAMP stimulation. Overall, the results indicate that lipolytic products can activate PPARα and PPARδ in brown adipocytes, thereby expanding the oxidative capacity to match enhanced fatty acid supply.


Subject(s)
Adipocytes, Brown/metabolism , Fatty Acids/metabolism , Gene Expression Regulation/physiology , Lipolysis/physiology , PPAR alpha/biosynthesis , PPAR-beta/biosynthesis , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cells, Cultured , Fatty Acids/genetics , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Ion Channels/genetics , Ion Channels/metabolism , Lipase/genetics , Lipase/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidation-Reduction , PPAR alpha/genetics , PPAR-beta/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Uncoupling Protein 1
19.
bioRxiv ; 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36778294

ABSTRACT

The diffusion and reorganization of phospholipids and membrane-associated proteins are fundamental for cellular function. Fluorescence cross-correlation spectroscopy (FCCS) measures the diffusion and molecular interactions at nanomolar concentration in biological systems. We have developed a novel, economical method to simultaneously monitor diffusion and oligomerization with the use of super-continuum laser and spectral deconvolution from a single detector. Customizable excitation wavelengths were chosen from the wide-band source and spectral fitting of the emitted light revealed the interactions for up to four spectrally overlapping fluorophores simultaneously. This method was applied to perform four-color FCCS, as demonstrated with polystyrene nanoparticles, lipid vesicles, and membrane-bound molecules. Up to four individually customizable excitation channels were selected from the broad-spectrum fiber laser to excite the diffusers within a diffraction-limited spot. The fluorescence emission passed through a cleanup filter and a dispersive prism prior to being collected by a sCMOS or EMCCD camera with up to 10 kHz frame rates. The emission intensity versus time of each fluorophore was extracted through a linear least-square fitting of each camera frame and temporally correlated via custom software. Auto- and cross-correlation functions enabled the measurement of the diffusion rates and binding partners. We have measured the induced aggregation of nanobeads and lipid vesicles in solution upon increasing the buffer salinity. Because of the adaptability of investigating four fluorophores simultaneously with a cost-effective method, this technique will have wide application for examining complex homo- and heterooligomerization in model and living systems.

20.
Biomed Opt Express ; 14(7): 3812-3827, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37497523

ABSTRACT

The diffusion and reorganization of phospholipids and membrane-associated proteins are fundamental for cellular function. Fluorescence cross-correlation spectroscopy (FCCS) measures diffusion and molecular interactions at nanomolar concentration in biological systems. We have developed an economical method to simultaneously monitor diffusion and complexation with the use of super-continuum laser and spectral deconvolution from a single detector. Customizable excitation wavelengths were chosen from the wide-band source and spectral fitting of the emitted light revealed the interactions for up to four chromatically overlapping fluorophores simultaneously. This method was applied to perform four-color FCCS that we demonstrated with polystyrene nanoparticles, lipid vesicles, and membrane-bound molecules. Up to four individually customizable excitation channels were selected from the broad-spectrum fiber laser to excite the diffusers within a diffraction-limited spot. The fluorescence emission passed through a cleanup filter and a dispersive prism prior to being collected by a sCMOS or EMCCD camera with up to 1.8 kHz frame rates. The emission intensity versus time of each fluorophore was extracted through a linear least-square fitting of each camera frame and temporally correlated via custom software. Auto- and cross-correlation functions enabled the measurement of the diffusion rates and binding partners. We have measured the induced aggregation of nanobeads and lipid vesicles in solution upon increasing the buffer salinity. Because of the adaptability of investigating four fluorophores simultaneously with a cost-effective method, this technique will have wide application for examining macromolecular complex formation in model and living systems.

SELECTION OF CITATIONS
SEARCH DETAIL