Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Endocrinology ; 162(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33369618

ABSTRACT

Obesity and related metabolic disorders have become epidemic diseases. Intermittent fasting has been shown to promote adipose tissue angiogenesis and have an anti-obesity feature; however, the mechanisms of how intermittent fasting modulates adipose tissues angiogenesis are poorly understood. We investigated the effect of fasting on vascular endothelial growth factor (VEGF) levels in white adipose tissues (WAT) and the function of fibroblast growth factor 21 (FGF21) in 1-time fasting and long-term intermittent fasting-induced VEGF expression. In the current study, fasting induced a selective and drastic elevation of VEGF levels in WAT, which did not occur in interscapular brown adipose tissue and liver. The fasting-induced Vegfa expression occurred predominantly in mature adipocytes, but not in the stromal vascular fraction in epididymal WAT and inguinal WAT (iWAT). Furthermore, a single bolus of recombinant mouse FGF21 injection increased VEGF levels in WAT. Long-term intermittent fasting for 16 weeks increased WAT angiogenesis, iWAT browning, and improved insulin resistance and inflammation, but the effect was blunted in FGF21 liver-specific knockout mice. In summary, these data suggest that FGF21 is a potent regulator of VEGF levels in WAT. The interorgan FGF21 signaling-induced WAT angiogenesis by VEGF could be a potential new therapeutic target in combination with obesity-related metabolic disorders.


Subject(s)
Adipose Tissue, White/blood supply , Eating/physiology , Fibroblast Growth Factors/genetics , Neovascularization, Physiologic/genetics , Adipocytes/metabolism , Animals , Fasting/physiology , Fibroblast Growth Factors/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Angiogenesis ; 23(4): 667-684, 2020 11.
Article in English | MEDLINE | ID: mdl-32699964

ABSTRACT

Platelet-derived growth factor-B (PDGF-B) is a main factor to promote adipose tissue angiogenesis, which is responsible for the tissue expansion in obesity. In this process, PDGF-B induces the dissociation of pericytes from blood vessels; however, its regulatory mechanism remains unclear. In the present study, we found that stromal cell-derived factor 1 (SDF1) plays an essential role in this regulatory mechanism. SDF1 mRNA was increased in epididymal white adipose tissue (eWAT) of obese mice. Ex vivo pharmacological analyses using cultured adipose tissue demonstrated that physiological concentrations (1-100 pg/mL) of SDF1 inhibited the PDGF-B-induced pericyte dissociation from vessels via two cognate SDF1 receptors, CXCR4 and CXCR7. In contrast, higher concentrations (> 1 ng/mL) of SDF1 alone caused the dissociation of pericytes via CXCR4, and this effect disappeared in the cultured tissues from PDGF receptor ß (PDGFRß) knockout mice. To investigate the role of SDF1 in angiogenesis in vivo, the effects of anagliptin, an inhibitor of dipeptidyl peptidase 4 (DPP4) that degrades SDF1, were examined in mice fed a high-fat diet. Anagliptin increased the SDF1 levels in the serum and eWAT. These changes were associated with a reduction of pericyte dissociation and fat accumulation in eWAT. AMD3100, a CXCR4 antagonist, cancelled these anagliptin effects. In flow-cytometry analysis, anagliptin increased and decreased the PDGF-B expression in endothelial cells and macrophages, respectively, whereas anagliptin reduced the PDGFRß expression in pericytes of eWAT. These results suggest that SDF1 negatively regulates the adipose tissue angiogenesis in obesity by altering the reactivity of pericytes to PDGF-B.


Subject(s)
Adipose Tissue, White/pathology , Adipose Tissue, White/physiopathology , Chemokine CXCL12/metabolism , Obesity/pathology , Obesity/physiopathology , Proto-Oncogene Proteins c-sis/metabolism , Vascular Remodeling , Adipose Tissue, White/blood supply , Angiogenesis Inducing Agents/metabolism , Animals , Blood Vessels/pathology , Chemokine CXCL12/blood , Diet, High-Fat , Epididymis/pathology , Feeding Behavior , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Obese , Models, Biological , Neovascularization, Physiologic/drug effects , Pericytes/drug effects , Pericytes/metabolism , Pericytes/pathology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, CXCR/metabolism , Receptors, CXCR4/metabolism , Thinness/pathology
3.
Microvasc Res ; 130: 104003, 2020 07.
Article in English | MEDLINE | ID: mdl-32199946

ABSTRACT

OBJECTIVE: This study aimed to determine the effects of physical exercise on the angio-adaptive response in adipose tissue following weight loss in a mouse model of diet-induced obesity. We hypothesized that physical exercise stimulates angiogenesis through the regulation of Vascular endothelial growth factor-A (VEGF-A) pro-/Thrombospondin-1 (TSP-1) anti-angiogenic signal under the control of the Murine double-minute 2/Forkhead box Os (Mdm2/FoxOs) axis, as reported in skeletal muscle. METHODS: We studied the effects of 7 weeks-voluntary exercise (Ex) in C57Bl/6 control or diet-induced obese (HFS) mice on vascularization of white adipose tissue (AT). RESULTS: Diet-induced obese sedentary (HFSsed) mice presented a powerful angiostatic control in all adipose tissues, under FoxOs protein regulation, leading to capillary rarefaction. Exercise increased expression of Mdm2, repressing the angiostatic control in favor of adipose vascular regrowth in normal chow (NCex) and HFSex mice. This phenomenon was associated with adipocytes microenvironment improvement, such as decreased adipocytes hypertrophy and adipose tissue inflammation. In addition, adipose angiogenesis stimulation by exercise through Mdm2 pro-angiogenic action, improved visceral adipose insulin sensitivity, activated browning process within subcutaneous adipose tissue (ScWAT) and decreased ectopic fat deposition (muscle, heart and liver) in obese HFSex mice. The overall result of this approach of therapy by physical exercise is an improvement of all systemic cardiometabolic parameters. CONCLUSIONS: These data demonstrated the therapeutic efficacy of physical exercise against obesity-associated pathologies, and also offer new prospects for molecular therapies targeting the adipose angio-adaptation in obese humans.


Subject(s)
Adipocytes, White/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Diet, High-Fat , Exercise Therapy , Neovascularization, Physiologic , Obesity/therapy , Proto-Oncogene Proteins c-mdm2/metabolism , Adipocytes, Brown/metabolism , Adipocytes, Brown/pathology , Adipocytes, White/pathology , Adipose Tissue, White/pathology , Animals , Cellular Microenvironment , Disease Models, Animal , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/metabolism , Male , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Signal Transduction , Thrombospondin 1/metabolism , Tissue Culture Techniques , Vascular Endothelial Growth Factor A/metabolism , Weight Loss
4.
Sci Rep ; 10(1): 670, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959796

ABSTRACT

Adipose tissue macrophages (ATMs) play a central role in tissue remodeling and homeostasis. However, whether ATMs promote adipose angiogenesis in obesity remains unclear. We examined the impact of ATMs deletion on adipose angiogenesis and tissue expansion in the epididymal white adipose tissue (eWAT) of high-fat diet (HFD)-fed mice by using liposome-encapsulated clodronate. We further elucidated the induction mechanisms of platelet-derived growth factor (PDGF)-B in macrophages in response to obesity-associated metabolic stresses, since it plays a significant role in the regulation of pericyte behavior for the initiation of neoangiogenesis during tissue expansion. ATM depletion prevented adipose tissue expansion in HFD-fed mice by inhibiting pericyte detachment from vessels, resulting in less vasculature in eWAT. The lipopolysaccharide (LPS) stimulation and high glucose concentration augmented glucose incorporation and glycolytic capacity with the induction of Pdgfb mRNA. This effect was mediated through extracellular signal-regulated kinase (ERK) among mitogen-activated protein kinases coupled with glycolysis in RAW264.7 macrophages. The Pdgfb induction system was distinct from that of inflammatory cytokines mediated by mechanistic target of rapamycin complex 1 (mTORC1) and NFκB signaling. Thus, obesity-associated hyperglycemia and chronic inflammation fuels ERK signaling coupled with glycolysis in pro-inflammatory macrophages, which contribute to the expansion of eWAT through PDGF-B-dependent vascular remodeling.


Subject(s)
Adipose Tissue, White/blood supply , Adipose Tissue, White/cytology , Glycolysis , Macrophages/physiology , Neovascularization, Pathologic , Obesity/pathology , Proto-Oncogene Proteins c-sis/metabolism , Vascular Remodeling/genetics , Animals , Diet, High-Fat/adverse effects , Glycolysis/genetics , Inflammation , Lymphokines/genetics , Lymphokines/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , NF-kappa B/metabolism , Obesity/etiology , Obesity/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , RAW 264.7 Cells , RNA, Messenger/metabolism , Signal Transduction
5.
Mol Biol Rep ; 47(3): 1667-1680, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31933262

ABSTRACT

Obesity is a complex, chronic disease that arises according to the interaction between genetic and environmental factors. The expansion and growth of white adipose tissue (WAT) could be related to angiogenesis. Resveratrol and adrenomedullin (AdM) were used for the inhibition of angiogenesis in metabolically passive WAT for inhibiting the expansion of this tissue, and the activation of angiogenesis in metabolically active brown adipose tissue (BAT) for increasing daily energy consumption as a way of reducing obesity. Rats were divided into eight groups. Four obese groups were fed with a high-fat diet containing 60% fat as energy for three months. After obtaining obesity, 2.5 nmol/kg AdM and 10 mg/kg resveratrol were treated to experiment groups intraperitoneally (i.p.) every other day for four weeks. AdM and vascular endothelial growth factor A (VEGF-A) mRNA levels were detected with semi-quantitative PCR; protein levels were detected with Western Blotting. AdM and resveratrol are multifactorial molecules, thus, this study has revealed a few novel evidence. The results were distinct in the group and treatment levels. The results showed that resveratrol has a role in angiogenesis in obesity and contributed to AdM production. It is observed that AdM has regulated its expression and increased the effect of resveratrol in WAT. AdM and VEGF-A gene expressions could not be detected in BAT; however, it is suggested that resveratrol may have a pro-angiogenic effect in BAT of obese rats according to the protein levels. AdM also has regulated VEGF-A level according to the metabolic situation of the organism.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Adrenomedullin/administration & dosage , Energy Metabolism/drug effects , Obesity/metabolism , Resveratrol/administration & dosage , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Adrenomedullin/genetics , Adrenomedullin/metabolism , Animals , Antioxidants/administration & dosage , Diet, High-Fat/adverse effects , Female , Gene Expression/drug effects , Injections, Intraperitoneal , Obesity/etiology , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vasodilator Agents/administration & dosage
6.
Toxicol Lett ; 318: 12-21, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31622651

ABSTRACT

Maternal smoking during pregnancy and lactation is associated with increased fat mass in the offspring, but the mechanism by which this occurs is not fully understood. Our study focused on the relationships among maternal nicotine exposure, adipose angiogenesis and adipose tissue function in female offspring. Pregnant rats were randomly assigned to nicotine or control groups. Microvascular density, lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins were tested in 4-, 12- and 26-week female offspring. In vitro, nicotine concentration- and time-response experiments were conducted in 3T3-L1. Lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins were tested. The conditioned media of differentiated 3T3-L1 treated with nicotine were used to observe tube formation in human umbilical vein endothelial cells (HUVECs). Nicotine-exposed females presented higher adipose microvascular density. The gene expression of α7nAChR, Egr1 and FGF2 was significantly increased in gonadal white adipose tissue (gWAT) and inguinal subcutaneous WAT (igSWAT) of nicotine-exposed females at 4 weeks of age. The protein expression of α7nAChR, Egr1 and FGF2 was increased in gWAT and igSWAT of nicotine-exposed females at 4 weeks of age, and increased in gWAT at 26 weeks. In vitro, nicotine increased the expression of lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins in a concentration- and time-dependent manner. In the tube formation experiment, adipocytes affected by nicotine promoted HUVEC angiogenesis. Therefore, maternal nicotine exposure promoted the early angiogenesis of adipose tissue via the α7nAChR-Egr1-FGF2 signaling pathway, and this angiogenesis mechanism was associated with increased adipogenesis in adipose tissue of female offspring.


Subject(s)
Adipocytes/drug effects , Adipose Tissue, White/blood supply , Neovascularization, Physiologic/drug effects , Nicotine/toxicity , Nicotinic Agonists/toxicity , Prenatal Exposure Delayed Effects , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipid Metabolism/drug effects , Maternal Exposure , Mice , Pregnancy , Rats, Wistar , Signal Transduction/drug effects , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
7.
J Endocrinol ; 242(2): 79-89, 2019 08.
Article in English | MEDLINE | ID: mdl-31137012

ABSTRACT

Obesity-associated chronic inflammation in adipose tissue is partly attributed to hypoxia with insufficient microcirculation. Previous studies have shown that exenatide, a glucagon-like peptide 1 (GLP-1) receptor agonist, plays an anti-inflammatory role. Here, we investigate its effects on inflammation, hypoxia and microcirculation in white adipose tissue of diet-induced obese (DIO) mice. DIO mice were injected intraperitoneally with exenatide or normal saline for 4 weeks, while mice on chow diet were used as normal controls. The mRNA and protein levels of pro-inflammatory cytokines, hypoxia-induced genes and angiogenic factors were detected. Capillary density was measured by laser confocal microscopy and immunochemistry staining. After 4-week exenatide administration, the dramatically elevated pro-inflammatory cytokines in serum and adipose tissue and macrophage infiltration in adipose tissue of DIO mice were significantly reduced. Exenatide also ameliorated expressions of hypoxia-related genes in obese fat tissue. Protein levels of endothelial markers and pro-angiogenic factors including vascular endothelial growth factor and its receptor 2 were augmented in accordance with increased capillary density by exenatide in DIO mice. Our results indicate that inflammation and hypoxia in adipose tissue can be mitigated by GLP-1 receptor agonist potentially via improved angiogenesis and microcirculation in obesity.


Subject(s)
Adipose Tissue, White/drug effects , Exenatide/pharmacology , Inflammation/prevention & control , Microcirculation/drug effects , Obesity/prevention & control , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat/adverse effects , Exenatide/administration & dosage , Gene Expression/drug effects , Hypoxia/genetics , Hypoxia/metabolism , Inflammation/genetics , Inflammation/metabolism , Injections, Intraperitoneal , Male , Mice, Inbred C57BL , Microcirculation/genetics , Microcirculation/physiology , Obesity/etiology , Obesity/genetics
8.
J Vis Exp ; (144)2019 02 13.
Article in English | MEDLINE | ID: mdl-30829339

ABSTRACT

Recent studies have highlighted the critical role of angiogenesis and sympathetic innervation in adipose tissue remodeling during the development of obesity. Therefore, developing an easy and efficient method to document the dynamic changes in adipose tissue is necessary. Here, we describe a modified immunofluorescent approach that efficiently co-stains blood vessels and nerve fibers in adipose tissues. Compared to traditional and recently developed methods, our approach is relatively easy to follow and more efficient in labeling the blood vessels and nerve fibers with higher densities and less background. Moreover, the higher resolution of the images further allows us to accurately measure the area of the vessels, amount of branching, and length of the fibers by open source software. As a demonstration using our method, we show that brown adipose tissue (BAT) contains higher amounts of blood vessels and nerve fibers compared to white adipose tissue (WAT). We further find that among the WATs, subcutaneous WAT (sWAT) has more blood vessels and nerve fibers compared to epididymal WAT (eWAT). Our method thus provides a useful tool for investigating adipose tissue remodeling.


Subject(s)
Adipose Tissue, Brown/blood supply , Adipose Tissue, White/blood supply , Nerve Fibers/metabolism , Obesity/blood , Subcutaneous Fat/blood supply , Animals , Humans , Male , Mice
9.
Microcirculation ; 26(5): e12540, 2019 07.
Article in English | MEDLINE | ID: mdl-30825241

ABSTRACT

OBJECTIVE: Accumulating evidence suggests the vascular endothelium plays a fundamental role in the pathophysiology of obesity by regulating the functional status of white adipose and systemic metabolism. Robo4 is expressed specifically in endothelial cells and increases vascular stability and inhibits angiogenesis. We sought to determine the role of Robo4 in modulating cardiometabolic function in response to high-fat feeding. METHODS: We examined exercise capacity, glucose tolerance, and white adipose tissue artery gene expression, endothelium-dependent dilation (EDD), and angiogenesis in wild type and Robo4 knockout (KO) mice fed normal chow (NC) or a high-fat diet (HFD). RESULTS: We found Robo4 deletion enhances exercise capacity in NC-fed mice and HFD markedly increased the expression of the Robo4 ligand, Slit2, in white adipose tissue. Deletion of Robo4 increased angiogenesis in white adipose tissue and protected against HFD-induced impairments in white adipose artery vasodilation and glucose intolerance. CONCLUSIONS: We demonstrate a novel functional role for Robo4 in endothelial cell function and metabolic homeostasis in white adipose tissue, with Robo4 deletion protecting against endothelial and metabolic dysfunction associated with a HFD. Our findings suggest that Robo4-dependent signaling pathways may be a novel target in anti-obesity therapy.


Subject(s)
Adipose Tissue, White , Arteries , Dietary Fats/adverse effects , Endothelium, Vascular , Gene Deletion , Gene Expression Regulation/drug effects , Receptors, Cell Surface , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Arteries/metabolism , Arteries/pathology , Dietary Fats/pharmacology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/deficiency , Vasodilation/drug effects , Vasodilation/genetics
10.
Int J Obes (Lond) ; 43(3): 556-566, 2019 03.
Article in English | MEDLINE | ID: mdl-30006585

ABSTRACT

BACKGROUND/OBJECTIVES: The increased prevalence of obesity has prompted great strides in our understanding of specific adipose depots and their involvement in cardio-metabolic health. However, the impact of obesity on dermal white adipose tissue (dWAT) and dermal microvascular functionality remains unclear. This study aimed to investigate the temporal changes that occur in dWAT and dermal microvascular functionality during the development of diet-induced obesity and type 2 diabetes in mice. METHODS: Metabolic phenotyping of a murine model of hypercaloric diet (HCD)-induced obesity and type 2 diabetes was performed at three time points that reflected three distinct stages of disease development; 2 weeks of HCD-overweight-metabolically healthy, 4 weeks of HCD-obese-prediabetic and 12 weeks of HCD-obese-type 2 diabetic mice. Expansion of dWAT was characterized histologically, and changes in dermal microvascular reactivity were assessed in response to pressure and the vasodilators SNP and Ach. RESULTS: HCD resulted in a progressive expansion of dWAT and increased expression of pro-inflammatory markers (IL1ß and COX-2). Impairments in pressure-induced (PIV) and Ach-induced (endothelium-dependent) vasodilation occurred early, in overweight-metabolically healthy mice. Residual vasodilatory responses were NOS-independent but sensitive to COX inhibition. These changes were associated with reductions in NO and adiponectin bioavailability, and rescued by exogenous adiponectin or hyperinsulinemia. Obese-prediabetic mice continued to exhibit impaired Ach-dependent vasodilation but PIV appeared normalized. This normalization coincided with elevated endogenous adiponectin and insulin levels, and was sensitive to NOS, COX and PI3K, inhibition. In obese-type 2 diabetic mice, both Ach-stimulated and pressure-induced vasodilatory responses were increased through enhanced COX-2-dependent prostaglandin response. CONCLUSIONS: We demonstrate that the development of obesity, metabolic dysfunction and type 2 diabetes, in HCD-fed mice, is accompanied by increased dermal adiposity and associated metaflammation in dWAT. Importantly, these temporal changes are also linked to disease stage-specific dermal microvascular reactivity, which may reflect adaptive mechanisms driven by metaflammation.


Subject(s)
Adipose Tissue, White , Diabetes Mellitus, Type 2/physiopathology , Inflammation , Obesity/physiopathology , Skin , Adiponectin/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adipose Tissue, White/physiopathology , Animals , Cytokines/metabolism , Diabetes Mellitus, Experimental , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microvessels/metabolism , Microvessels/pathology , Microvessels/physiopathology , Skin/blood supply , Skin/metabolism , Skin/physiopathology , Vasodilation/physiology
11.
Am J Physiol Heart Circ Physiol ; 316(3): H485-H494, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30550353

ABSTRACT

Blood flow regulation is a critical factor for tissue oxygenation and substrate supply. Increased reactivity of arteries to vasoconstrictors may increase vascular resistance, resulting in reduced blood flow. We aimed to investigate the effect of a high-fat (HF) diet on stiffness and vasoconstrictor reactivity of white adipose tissue (WAT) and brown adipose tissue (BAT) resistance arteries and also investigated the interconversion of both adipose depots in the setting of a HF diet. Vasoconstrictor reactivity and passive morphology and mechanical properties of arteries from B6D2F1 mice (5 mo old) fed normal chow (NC) or a HF diet (8 wk) were measured using pressure myography. Receptor gene expression in WAT and BAT arteries and markers of WAT and BAT were assessed in whole tissue lysates by real-time RT-PCR. Despite greater receptor-independent vasoconstriction (in response to KCl, P < 0.01), vasoconstriction in response to angiotensin II ( P < 0.01) was lower in NC-BAT than NC-WAT arteries and similar in response to endothelin-1 ( P = 0.07) and norepinephrine ( P = 0.11) in NC-BAT and NC-WAT arteries. With the exception of BAT artery reactivity to endothelin-1 and angiotensin II, the HF diet tended to attenuate reactivity in arteries from both adipose depots and increased expression of adipose markers in BAT. No significant differences in morphology or passive mechanical properties were found between adipose types or diet conditions. Alterations in gene expression of adipose markers after the HF diet suggest beiging of BAT. An increase in brown adipocytes in the absence of increased BAT mass may be a compensatory mechanism to dissipate excess energy from a HF diet. NEW & NOTEWORTHY Despite no differences in passive mechanical properties and greater receptor-independent vasoconstriction, receptor-mediated vasoconstriction was either lower in brown than white adipose tissue arteries or similar in brown and white adipose tissue arteries. A high-fat diet has a greater impact on vasoconstrictor responses in white adipose tissue but leads to altered adipose tissue gene expression consistent with beiging of the brown adipose tissue.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Arteries/drug effects , Diet, High-Fat , Dietary Fats/pharmacology , Vasoconstriction , Adipose Tissue, Brown/blood supply , Adipose Tissue, White/blood supply , Angiotensin II/pharmacology , Animals , Arteries/physiology , Endothelin-1/pharmacology , Male , Mice , Norepinephrine/pharmacology , Vasoconstrictor Agents/pharmacology
12.
Mol Metab ; 14: 71-81, 2018 08.
Article in English | MEDLINE | ID: mdl-29914852

ABSTRACT

OBJECTIVE: The vascular system is central to sustaining tissue survival and homeostasis. Blood vessels are densely present in adipose tissues and exert essential roles in their metabolism. However, conventional immunohistochemistry methods have intrinsic limitations in examining the 3D vascular network in adipose tissues as well as other organs in general. METHODS: We established a 3D volume fluorescence-imaging technique to visualize the vasculatures in mouse adipose tissues by combining the optimized steps of whole-mount immunolabeling, tissue optical clearing, and lightsheet volume fluorescence-imaging. To demonstrate the strength of this novel imaging procedure, we comprehensively assessed the intra-adipose vasculatures under obese conditions or in response to a cold challenge. RESULTS: We show the entirety of the vascular network in mouse adipose tissues on the whole-tissue level at a single-capillary resolution for the first time in the field. We accurately quantify the pathological changes of vasculatures in adipose tissues in wild-type or obese mice (ob/ob, db/db, or diet-induced obesity). In addition, we identify significant and reversible changes of the intra-adipose vasculatures in the mice subjected to cold challenge (i.e., 4°). Furthermore, we demonstrate that the cold-induced vascular plasticity depends on the sympathetic-derived catecholamine signal and is involved in the beiging process of white adipose tissues. CONCLUSIONS: We report a 3D volume fluorescence-imaging procedure that is compatible with many areas of vascular research and is poised to serve the field in future investigations of the vascular system in adipose tissues or other research scenarios.


Subject(s)
Adipose Tissue, Beige/cytology , Adipose Tissue, White/cytology , Capillaries/cytology , Imaging, Three-Dimensional/methods , Adipose Tissue, Beige/blood supply , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Animals , Body Temperature Regulation , Capillaries/physiology , Cold-Shock Response , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods
13.
J Physiol Biochem ; 74(3): 441-454, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29797227

ABSTRACT

Physical training (PT) has been considered as a treatment in metabolic syndrome (MS), since it induces thermogenic activity in brown (BAT) and white (WAT) adipose tissues. We evaluated the therapeutic effect of PT on activity of WAT and BAT in rats with MS induced by high-fat diet (30% lard) for 13 weeks and submitted, for the last 6 weeks, to swimming or kept sedentary (SED) rats. MS-SED rats compared to control diet (CT-SED) rats showed low physical fitness and high levels of glucose, insulin, homeostasis evaluation of insulin resistance (HOMA-IR), homeostasis evaluation of the functional capacity of ß-cells (HOMA-ß), and blood pressure. The gastrocnemius muscle decreased in peroxisome proliferator-activated receptor gamma coactivator 1-alpha and beta (PGC-1α, PGC-1ß), and uncoupled protein 2 and 3 (UCP2 and UCP3) expressions. Both WAT and BAT increased in the adipocyte area and decreased in blood vessels and fibroblast numbers. WAT increased in expression of pro-inflammatory adipokines and decreased in anti-inflammatory adipokine and adiponectin. WAT and gastrocnemius showed impairment in the insulin signaling pathway. In response to PT, MS rats showed increased physical fitness and restoration of certain biometric and biochemical parameters and blood pressure. PT also induced thermogenic modulations in skeletal muscle, WAT and BAT, and also improved the insulin signaling pathway. Collectively, PT was effective in treating MS by inducing improvement in physical fitness and interchangeable effects between skeletal muscle, WAT and BAT, suggesting a development of brown-like adipocyte cells.


Subject(s)
Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Adiposity , Insulin Resistance , Metabolic Syndrome/therapy , Physical Conditioning, Animal , Thermogenesis , Adipokines/genetics , Adipokines/metabolism , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Biomarkers/blood , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Gene Expression Regulation , Hyperglycemia/etiology , Hyperglycemia/prevention & control , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Hypertension/etiology , Hypertension/prevention & control , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Random Allocation , Rats, Inbred F344 , Weaning
14.
J Exp Med ; 215(2): 611-626, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29305395

ABSTRACT

Angiogenesis plays an instrumental role in the modulation of adipose tissue mass and metabolism. Targeting adipose vasculature provides an outstanding opportunity for treatment of obesity and metabolic disorders. Here, we report the physiological functions of VEGFR1 in the modulation of adipose angiogenesis, obesity, and global metabolism. Pharmacological inhibition and genetic deletion of endothelial VEGFR1 augmented adipose angiogenesis and browning of subcutaneous white adipose tissue, leading to elevated thermogenesis. In a diet-induced obesity model, endothelial-VEGFR1 deficiency demonstrated a potent anti-obesity effect by improving global metabolism. Along with metabolic changes, fatty liver and insulin sensitivity were also markedly improved in VEGFR1-deficient high fat diet (HFD)-fed mice. Together, our data indicate that targeting of VEGFR1 provides an exciting new opportunity for treatment of obesity and metabolic diseases, such as liver steatosis and type 2 diabetes.


Subject(s)
Adipose Tissue/blood supply , Adipose Tissue/metabolism , Endothelium, Vascular/metabolism , Metabolic Diseases/therapy , Vascular Endothelial Growth Factor Receptor-1/deficiency , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Obesity/etiology , Obesity/metabolism , Obesity/therapy , Thermogenesis , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
15.
Cell Physiol Biochem ; 42(3): 974-986, 2017.
Article in English | MEDLINE | ID: mdl-28662501

ABSTRACT

BACKGROUND/AIMS: To characterize the temporal profile of cold-induced angiogenesis in brown and white adipose tissues of mice in vivo and the temporal changes of angiogenic factors in primary mice brown (BA) and white adipocytes (WA) treated with ß3-adrenoceptor agonist (CL316,243) in vitro. METHODS: 8-week old male C57BL/6J mice were individually housed in conventional cages under cold exposure (4°C) for 1, 2, 3, 4 and 5 days. Interscapular brown adipose tissue (iBAT), inguinal subcutaneous (sWAT) and epididymal white adipose tissues (eWAT) were harvested for immunohistochemical and gene expression analysis. In vitro, primary mice BA and WA treated with or without CL316,243 were harvested for gene expression and protein secretion analysis. RESULTS: A combination of morphological and genetic (Vegfa, Vegfr2, Hif-1α, Pai1 and Pedf) analyses demonstrated depot-specific angiogenesis in response to cold exposure. Upon CL316,243 treatment, angiogenic factors (Vegfa, Vegfr2, Hif-1α, Pai1 and Pedf) and secreted protein VEGFA were transiently increased in both BA and WA. CONCLUSION: Our results show that iBAT is highly responsive to cold-induced angiogenesis that is mainly supported by sWAT with a lesser extent by eWAT. Moreover, the angiogenesis is a transient process with the angiogenic factors may work in an autocrine/paracrine manner.


Subject(s)
Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/physiology , Adipose Tissue, White/blood supply , Adipose Tissue, White/physiology , Cold-Shock Response , Neovascularization, Physiologic , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Cells, Cultured , Cold Temperature , Gene Expression Regulation , Male , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Diabetes ; 66(6): 1479-1490, 2017 06.
Article in English | MEDLINE | ID: mdl-28254844

ABSTRACT

Adipocyte-derived vascular endothelial growth factor-A (VEGF-A) plays a crucial role in angiogenesis and contributes to adipocyte function and systemic metabolism, such as insulin resistance, chronic inflammation, and beiging of subcutaneous adipose tissue. Using a doxycycline-inducible adipocyte-specific VEGF-A-overexpressing mouse model, we investigated the dynamics of local VEGF-A effects on tissue beiging of adipose tissue transplants. VEGF-A overexpression in adipocytes triggers angiogenesis. We also observed a rapid appearance of beige fat cells in subcutaneous white adipose tissue as early as 2 days postinduction of VEGF-A. In contrast to conventional cold-induced beiging, VEGF-A-induced beiging is independent of interleukin-4. We subjected metabolically healthy VEGF-A-overexpressing adipose tissue to autologous transplantation. Transfer of subcutaneous adipose tissues taken from VEGF-A-overexpressing mice into diet-induced obese mice resulted in systemic metabolic benefits, associated with improved survival of adipocytes and a concomitant reduced inflammatory response. These effects of VEGF-A are tissue autonomous, inducing white adipose tissue beiging and angiogenesis within the transplanted tissue. Our findings indicate that manipulation of adipocyte functions with a bona fide angiogenic factor, such as VEGF-A, significantly improves the survival and volume retention of fat grafts and can convey metabolically favorable properties on the recipient on the basis of beiging.


Subject(s)
Adipocytes, Beige/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Cell Differentiation/genetics , Neovascularization, Physiologic/genetics , Obesity/genetics , Vascular Endothelial Growth Factor A/genetics , Adipocytes, Beige/cytology , Adipose Tissue/metabolism , Adipose Tissue/transplantation , Adipose Tissue, Beige/blood supply , Adipose Tissue, Beige/cytology , Adipose Tissue, White/blood supply , Adipose Tissue, White/cytology , Animals , Cell Differentiation/immunology , Fluorescent Antibody Technique , Glucose Tolerance Test , Interleukin-4/immunology , Mice , Polymerase Chain Reaction , Subcutaneous Fat/blood supply , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Vascular Endothelial Growth Factor A/metabolism
17.
Cell Rep ; 18(1): 93-106, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28052263

ABSTRACT

Despite progress in our comprehension of the mechanisms regulating adipose tissue development, the nature of the factors that functionally characterize adipose precursors is still elusive. Defining the early steps regulating adipocyte development is needed for the generation of tools to control adipose tissue size and function. Here, we report the discovery of V-set and transmembrane domain containing 2A (VSTM2A) as a protein expressed and secreted by committed preadipocytes. VSTM2A expression is elevated in the early phases of adipogenesis in vitro and adipose tissue development in vivo. We show that VSTM2A-producing cells associate with the vasculature and express the common surface markers of adipocyte progenitors. Overexpression of VSTM2A induces adipogenesis, whereas its depletion impairs this process. VSTM2A controls preadipocyte determination at least in part by modulating BMP signaling and PPARγ2 activation. We propose a model in which VSTM2A is produced to preserve and amplify the adipogenic capability of adipose precursors.


Subject(s)
Adipogenesis , Cell Lineage , Membrane Proteins/metabolism , Receptors, Immunologic/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/cytology , Animals , Biomarkers/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Gene Knockdown Techniques , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , NIH 3T3 Cells , Neovascularization, Physiologic , PPAR gamma/metabolism , Signal Transduction
18.
Diabetes ; 66(4): 1008-1021, 2017 04.
Article in English | MEDLINE | ID: mdl-28122789

ABSTRACT

Platelet-derived growth factor (PDGF) is a key factor in angiogenesis; however, its role in adult obesity remains unclear. In order to clarify its pathophysiological role, we investigated the significance of PDGF receptor ß (PDGFRß) in adipose tissue expansion and glucose metabolism. Mature vessels in the epididymal white adipose tissue (eWAT) were tightly wrapped with pericytes in normal mice. Pericyte desorption from vessels and the subsequent proliferation of endothelial cells were markedly increased in the eWAT of diet-induced obese mice. Analyses with flow cytometry and adipose tissue cultures indicated that PDGF-B caused the detachment of pericytes from vessels in a concentration-dependent manner. M1-macrophages were a major type of cells expressing PDGF-B in obese adipose tissue. In contrast, pericyte detachment was attenuated and vascularity within eWAT was reduced in tamoxifen-inducible conditional Pdgfrb-knockout mice with decreases in adipocyte size and chronic inflammation. Furthermore, Pdgfrb-knockout mice showed enhanced energy expenditure. Consequently, diet-induced obesity and the associated deterioration of glucose metabolism in wild-type mice were absent in Pdgfrb-knockout mice. Therefore, PDGF-B-PDGFRß signaling plays a significant role in the development of adipose tissue neovascularization and appears to be a fundamental target for the prevention of obesity and type 2 diabetes.


Subject(s)
Adipose Tissue, White/metabolism , Cell Proliferation/genetics , Endothelial Cells/cytology , Glucose/metabolism , Neovascularization, Pathologic/genetics , Obesity/genetics , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Vascular Remodeling/genetics , Adipose Tissue/blood supply , Adipose Tissue/metabolism , Adipose Tissue, White/blood supply , Animals , Blotting, Western , Diet, High-Fat , Flow Cytometry , Glucose Clamp Technique , Macrophages , Male , Mice , Mice, Knockout , Mice, Transgenic , Obesity/metabolism , Pericytes , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 113(20): 5552-7, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27140638

ABSTRACT

The incidence of obesity, which is recognized by the American Medical Association as a disease, has nearly doubled since 1980, and obesity-related comorbidities have become a major threat to human health. Given that adipose tissue expansion and transformation require active growth of new blood vasculature, angiogenesis offers a potential target for the treatment of obesity-associated disorders. Here we construct two peptide-functionalized nanoparticle (NP) platforms to deliver either Peroxisome Proliferator-Activated Receptor gamma (PPARgamma) activator rosiglitazone (Rosi) or prostaglandin E2 analog (16,16-dimethyl PGE2) to adipose tissue vasculature. These NPs were engineered through self-assembly of a biodegradable triblock polymer composed of end-to-end linkages between poly(lactic-coglycolic acid)-b-poly(ethylene glycol) (PLGA-b-PEG) and an endothelial-targeted peptide. In this system, released Rosi promotes both transformation of white adipose tissue (WAT) into brown-like adipose tissue and angiogenesis, which facilitates the homing of targeted NPs to adipose angiogenic vessels, thereby amplifying their delivery. We show that i.v. administration of these NPs can target WAT vasculature, stimulate the angiogenesis that is required for the transformation of adipose tissue, and transform WAT into brown-like adipose tissue, by the up-regulation of angiogenesis and brown adipose tissue markers. In a diet-induced obese mouse model, these angiogenesis-targeted NPs have inhibited body weight gain and modulated several serological markers including cholesterol, triglyceride, and insulin, compared with the control group. These findings suggest that angiogenesis-targeting moieties with angiogenic stimulator-loaded NPs could be incorporated into effective therapeutic regimens for clinical treatment of obesity and other metabolic diseases.


Subject(s)
16,16-Dimethylprostaglandin E2/administration & dosage , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Nanoparticles/administration & dosage , Neovascularization, Physiologic , Obesity/prevention & control , Thiazolidinediones/administration & dosage , Animals , Carbohydrate Metabolism , Diet , Drug Delivery Systems , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Nanomedicine , Rosiglitazone
20.
Curr Obes Rep ; 4(2): 224-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26627217

ABSTRACT

Systemic inflammation resulting from dysfunction of white adipose tissue (WAT) accelerates the pathologies of diabetes and cardiovascular diseases. In contrast to WAT, brown adipose tissue (BAT) is abundant in mitochondria that produce heat by uncoupling respiratory chain process of ATP synthesis. Besides BAT's role in thermogenesis, accumulating evidence has shown that it is involved in regulating systemic metabolism. Studies have analyzed the "browning" processes of WAT as a means to combat obesity, whereas few studies have focused on the impact and molecular mechanisms that contribute to obesity-linked BAT dysfunction--a process that is associated with the "whitening" of this tissue. Compared to WAT, a dense vascular network is required to support the high energy consumption of BAT. Recently, vascular rarefaction was shown to be a significant causal factor in the whitening of BAT in mouse models. Vascular insufficiency leads to mitochondrial dysfunction and loss in BAT and contributes to systemic insulin resistance. These data suggest that BAT "whitening," resulting from vascular dysfunction, can impact obesity and obesity-linked diseases. Conversely, agents that promote BAT function could have utility in the treatment of these conditions.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Blood Vessels/pathology , Cardiovascular Diseases/etiology , Energy Metabolism , Insulin Resistance , Obesity/metabolism , Adipose Tissue, Brown/blood supply , Adipose Tissue, White/blood supply , Animals , Cardiovascular Diseases/metabolism , Humans , Mitochondria/metabolism , Obesity/complications , Obesity/pathology , Thermogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...