Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cell Rep ; 43(5): 114240, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38753486

ABSTRACT

Adipose tissue remodeling and dysfunction, characterized by elevated inflammation and insulin resistance, play a central role in obesity-related development of type 2 diabetes (T2D) and cardiovascular diseases. Long intergenic non-coding RNAs (lincRNAs) are important regulators of cellular functions. Here, we describe the functions of linc-ADAIN (adipose anti-inflammatory), an adipose lincRNA that is downregulated in white adipose tissue of obese humans. We demonstrate that linc-ADAIN knockdown (KD) increases KLF5 and interleukin-8 (IL-8) mRNA stability and translation by interacting with IGF2BP2. Upregulation of KLF5 and IL-8, via linc-ADAIN KD, leads to an enhanced adipogenic program and adipose tissue inflammation, mirroring the obese state, in vitro and in vivo. KD of linc-ADAIN in human adipose stromal cell (ASC) hTERT adipocytes implanted into mice increases adipocyte size and macrophage infiltration compared to implanted control adipocytes, mimicking hallmark features of obesity-induced adipose tissue remodeling. linc-ADAIN is an anti-inflammatory lincRNA that limits adipose tissue expansion and lipid storage.

2.
Nat Metab ; 6(5): 793-807, 2024 May.
Article in English | MEDLINE | ID: mdl-38783156

ABSTRACT

Ageing is a conserved biological process, modulated by intrinsic and extrinsic factors, that leads to changes in life expectancy. In humans, ageing is characterized by greatly increased prevalence of cardiometabolic disease, type 2 diabetes and disorders associated with impaired immune surveillance. Adipose tissue displays species-conserved, temporal changes with ageing, including redistribution from peripheral to central depots, loss of thermogenic capacity and expansion within the bone marrow. Adipose tissue is localized to discrete depots, and also diffusely distributed within multiple organs and tissues in direct proximity to specialized cells. Thus, through their potent endocrine properties, adipocytes are capable of modulating tissue and organ function throughout the body. In addition to adipocytes, multipotent progenitor/stem cells in adipose tissue play a crucial role in maintenance and repair of tissues throughout the lifetime. Adipose tissue may therefore be a central driver for organismal ageing and age-associated diseases. Here we review the features of adipose tissue during ageing, and discuss potential mechanisms by which these changes affect whole-body metabolism, immunity and longevity. We also explore the potential of adipose tissue-targeted therapies to ameliorate age-associated disease burdens.


Subject(s)
Adipose Tissue , Aging , Humans , Aging/physiology , Adipose Tissue/metabolism , Animals , Adipocytes/metabolism , Longevity
3.
PNAS Nexus ; 2(12): pgad420, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38130664

ABSTRACT

Adipocyte lipid droplets (LDs) play a crucial role in systemic lipid metabolism by storing and releasing lipids to meet the organism's energy needs. Hormonal signals such as catecholamines and insulin act on adipocyte LDs, and impaired responsiveness to these signals can lead to uncontrolled lipolysis, lipotoxicity, and metabolic disease. To investigate the mechanisms that control LD function in human adipocytes, we applied proximity labeling mediated by enhanced ascorbate peroxidase (APEX2) to identify the interactome of PLIN1 in adipocytes differentiated from human mesenchymal progenitor cells. We identified 70 proteins that interact specifically with PLIN1, including PNPLA2 and LIPE, which are the primary effectors of regulated triglyceride hydrolysis, and 4 members of the 14-3-3 protein family (YWHAB, YWHAE, YWHAZ, and YWHAG), which are known to regulate diverse signaling pathways. Functional studies showed that YWHAB is required for maximum cyclic adenosine monophosphate (cAMP)-stimulated lipolysis, as its CRISPR-Cas9-mediated knockout mitigates lipolysis through a mechanism independent of insulin signaling. These findings reveal a new regulatory mechanism operating in human adipocytes that can impact lipolysis and potentially systemic metabolism.

4.
bioRxiv ; 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37693594

ABSTRACT

Aging and metabolic diseases are accompanied by systemic inflammation, but the mechanisms that induce this state are not known. We developed a human bone-marrow organoid system to explore mechanisms underlying metabolic-disease associated systemic inflammation. We find that a distinct type of hematopoietic stem cell (HSC) develops in the adipose-rich, yellow bone marrow, which is known to gradually replace the hematopoietic red marrow as we age and during metabolic disease. Unlike HSCs derived from the red bone marrow, HSCs derived from the yellow bone marrow have higher proliferation rates, increase myeloid differentiation, skew towards pro-inflammatory M1 macrophage differentiation, and express a distinct transcriptomic profile associated with responsiveness to wounding. Yellow marrow-derived HSCs express higher levels of the leptin receptor, which we find to be further increased in patients with type 2 diabetes. Our work demonstrates that the human long bone yellow marrow is a niche for a distinct class of HSCs which could underlie hematopoietic dysfunction during aging and metabolic disease processes suggesting a shared inflammaging mechanism.

5.
Nat Metab ; 5(6): 1014-1028, 2023 06.
Article in English | MEDLINE | ID: mdl-37337125

ABSTRACT

Mesenchymal stem/progenitor cells are essential for tissue development and repair throughout life, but how they are maintained under chronic differentiation pressure is not known. Using single-cell transcriptomics of human progenitor cells we find that adipose differentiation stimuli elicit two cellular trajectories: one toward mature adipocytes and another toward a pool of non-differentiated cells that maintain progenitor characteristics. These cells are induced by transient Wnt pathway activation and express numerous extracellular matrix genes and are therefore named structural Wnt-regulated adipose tissue cells. We find that the genetic signature of structural Wnt-regulated adipose tissue cells is present in adult human adipose tissue and adipose tissue developed from human progenitor cells in mice. Our results suggest a mechanism whereby adipose differentiation occurs concurrently with the maintenance of a mesenchymal progenitor cell pool, ensuring tissue development, repair and appropriate metabolic control over the lifetime.


Subject(s)
Stem Cells , Wnt Signaling Pathway , Mice , Humans , Animals , Adipogenesis , Adipose Tissue , Adipocytes/metabolism
6.
Elife ; 112022 09 15.
Article in English | MEDLINE | ID: mdl-36107478

ABSTRACT

Mechanisms that control 'beige/brite' thermogenic adipose tissue development may be harnessed to improve human metabolic health. To define these mechanisms, we developed a species-hybrid model in which human mesenchymal progenitor cells were used to develop white or thermogenic/beige adipose tissue in mice. The hybrid adipose tissue developed distinctive features of human adipose tissue, such as larger adipocyte size, despite its neurovascular architecture being entirely of murine origin. Thermogenic adipose tissue recruited a denser, qualitatively distinct vascular network, differing in genes mapping to circadian rhythm pathways, and denser sympathetic innervation. The enhanced thermogenic neurovascular network was associated with human adipocyte expression of THBS4, TNC, NTRK3, and SPARCL1, which enhance neurogenesis, and decreased expression of MAOA and ACHE, which control neurotransmitter tone. Systemic inhibition of MAOA, which is present in human but absent in mouse adipocytes, induced browning of human but not mouse adipose tissue, revealing the physiological relevance of this pathway. Our results reveal species-specific cell type dependencies controlling the development of thermogenic adipose tissue and point to human adipocyte MAOA as a potential target for metabolic disease therapy.


Subject(s)
Monoamine Oxidase , Thermogenesis , Adipogenesis , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Mice , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Neurogenesis , Thermogenesis/genetics
7.
Nat Metab ; 4(8): 963-964, 2022 08.
Article in English | MEDLINE | ID: mdl-35982291
8.
Angiogenesis ; 25(4): 439-453, 2022 11.
Article in English | MEDLINE | ID: mdl-35857195

ABSTRACT

While most tissues exhibit their greatest growth during development, adipose tissue is capable of additional massive expansion in adults. Adipose tissue expandability is advantageous when temporarily storing fuel for use during fasting, but becomes pathological upon continuous food intake, leading to obesity and its many comorbidities. The dense vasculature of adipose tissue provides necessary oxygen and nutrients, and supports delivery of fuel to and from adipocytes under fed or fasting conditions. Moreover, the vasculature of adipose tissue comprises a major niche for multipotent progenitor cells, which give rise to new adipocytes and are necessary for tissue repair. Given the multiple, pivotal roles of the adipose tissue vasculature, impairments in angiogenic capacity may underlie obesity-associated diseases such as diabetes and cardiometabolic disease. Exciting new studies on the single-cell and single-nuclei composition of adipose tissues in mouse and humans are providing new insights into mechanisms of adipose tissue angiogenesis. Moreover, new modes of intercellular communication involving micro vesicle and exosome transfer of proteins, nucleic acids and organelles are also being recognized to play key roles. This review focuses on new insights on the cellular and signaling mechanisms underlying adipose tissue angiogenesis, and on their impact on obesity and its pathophysiological consequences.


Subject(s)
Adipose Tissue , Nucleic Acids , Adipocytes , Adipose Tissue/metabolism , Adult , Animals , Humans , Mice , Neovascularization, Pathologic/pathology , Nucleic Acids/metabolism , Obesity/pathology , Oxygen/metabolism
9.
Endocrinology ; 163(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34223880

ABSTRACT

Adipose tissue distribution in the human body is highly heterogeneous, and the relative mass of different depots is differentially associated with metabolic disease risk. Distinct functions of adipose depots are mediated by their content of specialized adipocyte subtypes, best exemplified by thermogenic adipocytes found in specific depots. Single-cell transcriptome profiling has been used to define the cellular composition of many tissues and organs, but the large size, buoyancy, and fragility of adipocytes have rendered it challenging to apply these techniques to understand the full complexity of adipocyte subtypes in different depots. Discussed here are strategies that have been recently developed for investigating adipocyte heterogeneity, including single-cell RNA-sequencing profiling of the stromal vascular fraction to identify diverse adipocyte progenitors, and single-nuclei profiling to characterize mature adipocytes. These efforts are yielding a more complete characterization of adipocyte subtypes in different depots, insights into the mechanisms of their development, and perturbations associated with different physiological states such as obesity. A better understanding of the adipocyte subtypes that compose different depots will help explain metabolic disease phenotypes associated with adipose tissue distribution and suggest new strategies for improving metabolic health.


Subject(s)
Adipocytes/cytology , Adipose Tissue/metabolism , Adipogenesis , Adipose Tissue, Beige/physiology , Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Animals , Body Height , Body Mass Index , Body Weight , Cell Differentiation , Cell Separation , Humans , Mice , Single-Cell Analysis , Stem Cells/cytology , Stromal Vascular Fraction/metabolism , Thermogenesis
10.
Nat Commun ; 12(1): 6931, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34836963

ABSTRACT

Obesity and type 2 diabetes are associated with disturbances in insulin-regulated glucose and lipid fluxes and severe comorbidities including cardiovascular disease and steatohepatitis. Whole body metabolism is regulated by lipid-storing white adipocytes as well as "brown" and "brite/beige" adipocytes that express thermogenic uncoupling protein 1 (UCP1) and secrete factors favorable to metabolic health. Implantation of brown fat into obese mice improves glucose tolerance, but translation to humans has been stymied by low abundance of primary human beige adipocytes. Here we apply methods to greatly expand human adipocyte progenitors from small samples of human subcutaneous adipose tissue and then disrupt the thermogenic suppressor gene NRIP1 by CRISPR. Ribonucleoprotein consisting of Cas9 and sgRNA delivered ex vivo are fully degraded by the human cells following high efficiency NRIP1 depletion without detectable off-target editing. Implantation of such CRISPR-enhanced human or mouse brown-like adipocytes into high fat diet fed mice decreases adiposity and liver triglycerides while enhancing glucose tolerance compared to implantation with unmodified adipocytes. These findings advance a therapeutic strategy to improve metabolic homeostasis through CRISPR-based genetic enhancement of human adipocytes without exposing the recipient to immunogenic Cas9 or delivery vectors.


Subject(s)
Adipocytes, Brown/transplantation , CRISPR-Cas Systems/genetics , Glucose Intolerance/therapy , Obesity/therapy , Thermogenesis/genetics , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adult Stem Cells/physiology , Animals , Cell Culture Techniques/methods , Cell Differentiation , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Gene Editing/methods , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Humans , Lipid Metabolism/genetics , Male , Mice , Nuclear Receptor Interacting Protein 1/genetics , Nuclear Receptor Interacting Protein 1/metabolism , Obesity/complications , Obesity/metabolism , RNA, Guide, Kinetoplastida/genetics , Subcutaneous Fat/cytology
11.
Cell Metab ; 33(4): 692-699, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33826910

ABSTRACT

Marking insulin's centennial, we share stories of researchers and clinicians whose seminal work has advanced our understanding of insulin, islet biology, insulin resistance, and diabetes. The past century of pursuing the "hormone of hormones" and advancing diabetes therapies is replete with stories of collaboration, perseverance, and triumph.


Subject(s)
Diabetes Mellitus/drug therapy , Insulin/therapeutic use , Biomedical Research/history , Cell- and Tissue-Based Therapy , Drug Delivery Systems , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , History, 20th Century , History, 21st Century , Humans , Insulin/chemistry , Insulin/metabolism , Insulin Resistance , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism
12.
Annu Rev Physiol ; 83: 257-278, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33566675

ABSTRACT

Adipose tissue depots in distinct anatomical locations mediate key aspects of metabolism, including energy storage, nutrient release, and thermogenesis. Although adipocytes make up more than 90% of adipose tissue volume, they represent less than 50% of its cellular content. Here, I review recent advances in genetic lineage tracing and transcriptomics that reveal the identities of the heterogeneous cell populations constituting mouse and human adipose tissues. In addition to mature adipocytes and their progenitors, these include endothelial and various immune cell types that together orchestrate adipose tissue development and functions. One salient finding is the identification of progenitor subtypes that can modulate adipogenic capacity through paracrine mechanisms. Another is the description of fate trajectories of monocyte/macrophages, which can respond maladaptively to nutritional and thermogenic stimuli, leading to metabolic disease. These studies have generated an extraordinary source of publicly available data that can be leveraged to explore commonalities and differences among experimental models, providing new insights into adipose tissues and their role in metabolic disease.


Subject(s)
Adipose Tissue/physiology , Adipocytes/physiology , Adipogenesis/physiology , Animals , Humans , Thermogenesis/physiology
13.
Sci Transl Med ; 12(571)2020 11 25.
Article in English | MEDLINE | ID: mdl-33239385

ABSTRACT

Pregnancy is a physiological state of continuous adaptation to changing maternal and fetal nutritional needs, including a reduction of maternal insulin sensitivity allowing for appropriately enhanced glucose availability to the fetus. However, excessive insulin resistance in conjunction with insufficient insulin secretion results in gestational diabetes mellitus (GDM), greatly increasing the risk for pregnancy complications and predisposing both mothers and offspring to future metabolic disease. Here, we report a signaling pathway connecting pregnancy-associated plasma protein A (PAPPA) with adipose tissue expansion in pregnancy. Adipose tissue plays a central role in the regulation of insulin sensitivity, and we show that, in both mice and humans, pregnancy caused remodeling of adipose tissue evidenced by altered adipocyte size, vascularization, and in vitro expansion capacity. PAPPA is known to be a metalloprotease secreted by human placenta that modulates insulin-like growth factor (IGF) bioavailability through prolteolysis of IGF binding proteins (IGFBPs) 2, 4, and 5. We demonstrate that recombinant PAPPA can stimulate ex vivo human adipose tissue expansion in an IGFBP-5- and IGF-1-dependent manner. Moreover, mice lacking PAPPA displayed impaired adipose tissue remodeling, pregnancy-induced insulin resistance, and hepatic steatosis, recapitulating multiple aspects of human GDM. In a cohort of 6361 pregnant women, concentrations of circulating PAPPA are inversely correlated with glycemia and odds of developing GDM. These data identify PAPPA and the IGF signaling pathway as necessary for the regulation of maternal adipose tissue physiology and systemic glucose homeostasis, with consequences for long-term metabolic risk and potential for therapeutic use.


Subject(s)
Diabetes, Gestational , Insulin Resistance , Pregnancy-Associated Plasma Protein-A/physiology , Adipose Tissue , Animals , Blood Glucose , Female , Humans , Mice , Pregnancy , Pregnancy-Associated Plasma Protein-A/genetics , Pregnancy-Associated Plasma Protein-A/pharmacology
14.
Nat Metab ; 2(5): 397-412, 2020 05.
Article in English | MEDLINE | ID: mdl-32440655

ABSTRACT

Human thermogenic adipose tissue mitigates metabolic disease, raising much interest in understanding its development and function. Here, we show that human thermogenic adipocytes specifically express a primate-specific long non-coding RNA, LINC00473 which is highly correlated with UCP1 expression and decreased in obesity and type-2 diabetes. LINC00473 is detected in progenitor cells, and increases upon differentiation and in response to cAMP. In contrast to other known adipocyte LincRNAs, LINC00473 shuttles out of the nucleus, colocalizes and can be crosslinked to mitochondrial and lipid droplet proteins. Up- or down- regulation of LINC00473 results in reciprocal alterations in lipolysis, respiration and transcription of genes associated with mitochondrial oxidative metabolism. Depletion of PLIN1 results in impaired cAMP-responsive LINC00473 expression and lipolysis, indicating bidirectional interactions between PLIN1, LINC00473 and mitochondrial oxidative functions. Thus, we suggest that LINC00473 is a key regulator of human thermogenic adipocyte function, and reveals a role for a LincRNA in inter-organelle communication and human energy metabolism.


Subject(s)
Adipocytes/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , Thermogenesis/genetics , Thermogenesis/physiology , Adult , Aged , Aged, 80 and over , Cell Communication/genetics , Cell Communication/physiology , Cell Nucleus/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/genetics , Energy Metabolism/physiology , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression Regulation , Humans , Lipid Droplets , Male , Middle Aged , Obesity/genetics , Obesity/metabolism , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Perilipin-1/deficiency , Perilipin-1/genetics , Uncoupling Protein 1/biosynthesis , Uncoupling Protein 1/genetics , Young Adult
15.
Proc Natl Acad Sci U S A ; 116(36): 17970-17979, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31420514

ABSTRACT

Single-cell sequencing technologies have revealed an unexpectedly broad repertoire of cells required to mediate complex functions in multicellular organisms. Despite the multiple roles of adipose tissue in maintaining systemic metabolic homeostasis, adipocytes are thought to be largely homogenous with only 2 major subtypes recognized in humans so far. Here we report the existence and characteristics of 4 distinct human adipocyte subtypes, and of their respective mesenchymal progenitors. The phenotypes of these distinct adipocyte subtypes are differentially associated with key adipose tissue functions, including thermogenesis, lipid storage, and adipokine secretion. The transcriptomic signature of "brite/beige" thermogenic adipocytes reveals mechanisms for iron accumulation and protection from oxidative stress, necessary for mitochondrial biogenesis and respiration upon activation. Importantly, this signature is enriched in human supraclavicular adipose tissue, confirming that these cells comprise thermogenic depots in vivo, and explain previous findings of a rate-limiting role of iron in adipose tissue browning. The mesenchymal progenitors that give rise to beige/brite adipocytes express a unique set of cytokines and transcriptional regulators involved in immune cell modulation of adipose tissue browning. Unexpectedly, we also find adipocyte subtypes specialized for high-level expression of the adipokines adiponectin or leptin, associated with distinct transcription factors previously implicated in adipocyte differentiation. The finding of a broad adipocyte repertoire derived from a distinct set of mesenchymal progenitors, and of the transcriptional regulators that can control their development, provides a framework for understanding human adipose tissue function and role in metabolic disease.


Subject(s)
Adipocytes, Beige/metabolism , Adiponectin/biosynthesis , Leptin/blood , Mesenchymal Stem Cells/metabolism , Thermogenesis , Transcriptome , Adipocytes, Beige/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Female , Gene Expression Profiling , Humans , Male , Mesenchymal Stem Cells/cytology
16.
Int J Mol Sci ; 20(8)2019 Apr 25.
Article in English | MEDLINE | ID: mdl-31027261

ABSTRACT

Exercise mitigates chronic diseases such as diabetes, cardiovascular diseases, and obesity; however, the molecular mechanisms governing protection from these diseases are not completely understood. Here we demonstrate that exercise rescues metabolically compromised high fat diet (HFD) fed mice, and reprograms subcutaneous white adipose tissue (scWAT). Using transcriptomic profiling, scWAT was analyzed for HFD gene expression changes that were rescued by exercise. Gene networks involved in vascularization were identified as prominent targets of exercise, which led us to investigate the vasculature architecture and endothelial phenotype. Vascular density in scWAT was found to be compromised in HFD, and exercise rescued this defect. Similarly, angiogenic capacity as measured by ex vivo capillary sprouting was significantly promoted with exercise. Together, these data demonstrate that exercise enhances scWAT vascularization and functional capacity for angiogenesis, and can prevent the detrimental effects of HFD. The improvement in these indices correlates with improvement of whole-body metabolism, suggesting that scWAT vascularization may be a potential therapeutic target for metabolic disease.


Subject(s)
Neovascularization, Physiologic/genetics , Physical Conditioning, Animal , Signal Transduction/genetics , Subcutaneous Fat/blood supply , Adaptation, Physiological , Animals , Diet, High-Fat , Glucose/metabolism , Homeostasis , Male , Mice, Inbred C57BL , Transcriptome/genetics
17.
Tissue Eng Part A ; 25(11-12): 842-854, 2019 06.
Article in English | MEDLINE | ID: mdl-30306830

ABSTRACT

IMPACT STATEMENT: This research describes the use of human mesenchymal progenitor cells for generating functional adipose tissue in vivo in a nude mouse model. Further preclinical development of the methods and insights described in this article can lead to therapeutic use of these cells in regenerative and reconstructive medicine.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Adipocytes/cytology , Adipose Tissue/cytology , Animals , Heterografts , Humans , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Nude
19.
Mol Metab ; 9: 199-206, 2018 03.
Article in English | MEDLINE | ID: mdl-29396370

ABSTRACT

OBJECTIVE: Perivascular adipose tissue depots around the aorta are regionally distinct and have specific functional properties. Thoracic aorta perivascular adipose tissue (tPVAT) expresses higher levels of thermogenic genes and lower levels of inflammatory genes than abdominal aorta perivascular adipose tissue (aPVAT). It is not known whether this distinction is due to the in-vivo functional environment or to cell-autonomous traits that persist outside the in-vivo setting. In this study, we asked whether the progenitor cells in tPVAT and aPVAT have cell-autonomous traits that lead to formation of regionally distinct PVAT. METHODS: We performed microarray analysis of thoracic and abdominal peri-aortic adipose tissues of C57Bl/6J mice to define gene expression profile of each depot. To derive adipocyte progenitor cells, C57Bl/6J mice were sacrificed and thoracic and abdominal aorta fragments were embedded in Matrigel and cultured under pro-angiogenic conditions. Adipogenesis was induced using the Ppar-γ agonist rosiglitazone, a thiazolidinedione (TZD). TZD-induced adipocyte populations were analyzed using immunofluorescence and qRT-PCR. RESULTS: Microarray analysis showed that tPVAT expressed higher levels of transcription factors related brown adipose tissue development compared to aPVAT. Classic brown adipose tissue (BAT) genes such as Ucp-1, Prdm16, Dio2, Slc27a displayed a concordant trend of higher level expression in tPVAT, while white adipose tissue (WAT) genes such as Hoxc8, Nnat, Sncg, and Mest were expressed at a higher level in aPVAT. The adipokines resistin and retinol binding protein 4 were also higher in aPVAT. Furthermore, adipocyte progenitors from abdominal and thoracic aortic rings responded to TZD with expression of canonical adipocyte genes Acrp30, Plin1, and Glut4. Adipocytes differentiated from thoracic aorta progenitors displayed markedly higher induction of Ucp-1 and Cidea. CONCLUSIONS: Thoracic aorta PVAT expresses higher levels of brown adipocyte transcription factors than aPVAT. Precursor cells from the thoracic aorta give rise to adipocytes that express significantly higher levels of Ucp-1 and Cidea ex vivo, suggesting that progenitor cells in tPVAT and aPVAT have cell-autonomous properties that dictate adipocyte phenotype.


Subject(s)
Adipocytes/cytology , Adipogenesis , Stem Cells/cytology , Adipocytes/metabolism , Animals , Aorta/cytology , Mice , Mice, Inbred C57BL , Phenotype , Stem Cells/metabolism , Transcriptome
20.
J Cell Physiol ; 232(10): 2887-2899, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27987321

ABSTRACT

White adipocytes are plastic cells able to reversibly transdifferentiate into brown adipocytes and into epithelial glandular cells under physiologic stimuli in vivo. These plastic properties could be used in future for regenerative medicine, but are incompletely explored in their details. Here, we focused on plastic properties of human mature adipocytes (MA) combining gene expression profile through microarray analysis with morphologic data obtained by electron and time lapse microscopy. Primary MA showed the classic morphology and gene expression profile of functional mature adipocytes. Notably, despite their committed status, MA expressed high levels of reprogramming genes. MA from ceiling cultures underwent transdifferentiation toward fibroblast-like cells with a well-differentiated morphology and maintaining stem cell gene signatures. The main morphologic aspect of the transdifferentiation process was the secretion of large lipid droplets and the development of organelles necessary for exocrine secretion further supported the liposecretion process. Of note, electron microscope findings suggesting liposecretion phenomena were found also in explants of human fat and rarely in vivo in fat biopsies from obese patients. In conclusion, both MA and post-liposecretion adipocytes show a well-differentiated phenotype with stem cell properties in line with the extraordinary plasticity of adipocytes in vivo. J. Cell. Physiol. 232: 2887-2899, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipogenesis , Cell Plasticity , Lipid Metabolism , Mesenchymal Stem Cells/metabolism , Obesity/metabolism , Adipocytes, Brown/ultrastructure , Adipocytes, White/ultrastructure , Aged , Aged, 80 and over , Cell Lineage , Cell Shape , Cells, Cultured , Cellular Reprogramming , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genetic Markers , Humans , Lipid Droplets/metabolism , Mesenchymal Stem Cells/ultrastructure , Microscopy, Confocal , Microscopy, Electron , Microscopy, Video , Middle Aged , Obesity/pathology , Obesity/physiopathology , Oligonucleotide Array Sequence Analysis , Phenotype , Time Factors , Time-Lapse Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...