Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 609(7927): 569-574, 2022 09.
Article in English | MEDLINE | ID: mdl-36045288

ABSTRACT

Adipose tissues communicate with the central nervous system to maintain whole-body energy homeostasis. The mainstream view is that circulating hormones secreted by the fat convey the metabolic state to the brain, which integrates peripheral information and regulates adipocyte function through noradrenergic sympathetic output1. Moreover, somatosensory neurons of the dorsal root ganglia innervate adipose tissue2. However, the lack of genetic tools to selectively target these neurons has limited understanding of their physiological importance. Here we developed viral, genetic and imaging strategies to manipulate sensory nerves in an organ-specific manner in mice. This enabled us to visualize the entire axonal projection of dorsal root ganglia from the soma to subcutaneous adipocytes, establishing the anatomical underpinnings of adipose sensory innervation. Functionally, selective sensory ablation in adipose tissue enhanced the lipogenic and thermogenetic transcriptional programs, resulting in an enlarged fat pad, enrichment of beige adipocytes and elevated body temperature under thermoneutral conditions. The sensory-ablation-induced phenotypes required intact sympathetic function. We postulate that beige-fat-innervating sensory neurons modulate adipocyte function by acting as a brake on the sympathetic system. These results reveal an important role of the innervation by dorsal root ganglia of adipose tissues, and could enable future studies to examine the role of sensory innervation of disparate interoceptive systems.


Subject(s)
Adipose Tissue , Sensory Receptor Cells , Adipose Tissue/innervation , Adipose Tissue/metabolism , Adipose Tissue, Beige/innervation , Adipose Tissue, Beige/metabolism , Animals , Axons , Energy Metabolism , Ganglia, Spinal/physiology , Homeostasis , Hormones/metabolism , Mice , Organ Specificity , Sensory Receptor Cells/physiology , Subcutaneous Fat/innervation , Subcutaneous Fat/metabolism , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Thermogenesis/genetics
2.
Nature ; 597(7876): 410-414, 2021 09.
Article in English | MEDLINE | ID: mdl-34408322

ABSTRACT

Signals from sympathetic neurons and immune cells regulate adipocytes and thereby contribute to fat tissue biology. Interactions between the nervous and immune systems have recently emerged as important regulators of host defence and inflammation1-4. Nevertheless, it is unclear whether neuronal and immune cells co-operate in brain-body axes to orchestrate metabolism and obesity. Here we describe a neuro-mesenchymal unit that controls group 2 innate lymphoid cells (ILC2s), adipose tissue physiology, metabolism and obesity via a brain-adipose circuit. We found that sympathetic nerve terminals act on neighbouring adipose mesenchymal cells via the ß2-adrenergic receptor to control the expression of glial-derived neurotrophic factor (GDNF) and the activity of ILC2s in gonadal fat. Accordingly, ILC2-autonomous manipulation of the GDNF receptor machinery led to alterations in ILC2 function, energy expenditure, insulin resistance and propensity to obesity. Retrograde tracing and chemical, surgical and chemogenetic manipulations identified a sympathetic aorticorenal circuit that modulates ILC2s in gonadal fat and connects to higher-order brain areas, including the paraventricular nucleus of the hypothalamus. Our results identify a neuro-mesenchymal unit that translates cues from long-range neuronal circuitry into adipose-resident ILC2 function, thereby shaping host metabolism and obesity.


Subject(s)
Adipose Tissue/innervation , Adipose Tissue/metabolism , Brain/metabolism , Immunity, Innate/immunology , Mesoderm/cytology , Neural Pathways , Neurons/cytology , Obesity/metabolism , Adipose Tissue/cytology , Animals , Brain/cytology , Cues , Cytokines/metabolism , Energy Metabolism , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Gonads/metabolism , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Receptors, Adrenergic, beta-2/metabolism , Sympathetic Nervous System/cytology , Sympathetic Nervous System/metabolism
3.
Nature ; 578(7796): 610-614, 2020 02.
Article in English | MEDLINE | ID: mdl-32076265

ABSTRACT

The sympathetic nervous system innervates peripheral organs to regulate their function and maintain homeostasis, whereas target cells also produce neurotrophic factors to promote sympathetic innervation1,2. The molecular basis of this bi-directional communication remains to be fully determined. Here we use thermogenic adipose tissue from mice as a model system to show that T cells, specifically γδ T cells, have a crucial role in promoting sympathetic innervation, at least in part by driving the expression of TGFß1 in parenchymal cells via the IL-17 receptor C (IL-17RC). Ablation of IL-17RC specifically in adipose tissue reduces expression of TGFß1 in adipocytes, impairs local sympathetic innervation and causes obesity and other metabolic phenotypes that are consistent with defective thermogenesis; innervation can be fully rescued by restoring TGFß1 expression. Ablating γδ Τ cells and the IL-17RC signalling pathway also impairs sympathetic innervation in other tissues such as salivary glands. These findings demonstrate coordination between T cells and parenchymal cells to regulate sympathetic innervation.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/innervation , Adipose Tissue/metabolism , Interleukin-17/metabolism , Sympathetic Nervous System/physiology , T-Lymphocytes/metabolism , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Interleukin-17/deficiency , Interleukin-17/genetics , Male , Mice , Mice, Knockout , Organ Specificity , Parenchymal Tissue/cytology , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
4.
Nature ; 583(7818): 839-844, 2020 07.
Article in English | MEDLINE | ID: mdl-32699414

ABSTRACT

Mutations in the leptin gene (ob) result in a metabolic disorder that includes severe obesity1, and defects in thermogenesis2 and lipolysis3, both of which are adipose tissue functions regulated by the sympathetic nervous system. However, the basis of these sympathetic-associated abnormalities remains unclear. Furthermore, chronic leptin administration reverses these abnormalities in adipose tissue, but the underlying mechanism remains to be discovered. Here we report that ob/ob mice, as well as leptin-resistant diet-induced obese mice, show significant reductions of sympathetic innervation of subcutaneous white and brown adipose tissue. Chronic leptin treatment of ob/ob mice restores adipose tissue sympathetic innervation, which in turn is necessary to correct the associated functional defects. The effects of leptin on innervation are mediated via agouti-related peptide and pro-opiomelanocortin neurons in the hypothalamic arcuate nucleus. Deletion of the gene encoding the leptin receptor in either population leads to reduced innervation in fat. These agouti-related peptide and pro-opiomelanocortin neurons act via brain-derived neurotropic factor-expressing neurons in the paraventricular nucleus of the hypothalamus (BDNFPVH). Deletion of BDNFPVH blunts the effects of leptin on innervation. These data show that leptin signalling regulates the plasticity of sympathetic architecture of adipose tissue via a top-down neural pathway that is crucial for energy homeostasis.


Subject(s)
Adipose Tissue/innervation , Adipose Tissue/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Leptin/metabolism , Sympathetic Nervous System/physiology , Agouti-Related Protein/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Leptin/deficiency , Lipolysis , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Signal Transduction , Subcutaneous Fat/innervation , Subcutaneous Fat/metabolism , Thermogenesis
5.
Am J Physiol Heart Circ Physiol ; 327(1): H155-H181, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38787382

ABSTRACT

Perivascular adipose tissue (PVAT) regulates vascular tone by releasing anticontractile factors. These anticontractile factors are driven by processes downstream of adipocyte stimulation by norepinephrine; however, whether norepinephrine originates from neural innervation or other sources is unknown. The goal of this study was to test the hypothesis that neurons innervating PVAT provide the adrenergic drive to stimulate adipocytes in aortic and mesenteric perivascular adipose tissue (aPVAT and mPVAT), and white adipose tissue (WAT). Healthy male and female mice (8-13 wk) were used in all experiments. Expression of genes associated with synaptic transmission were quantified by qPCR and adipocyte activity in response to neurotransmitters and neuron depolarization was assessed in AdipoqCre+;GCaMP5g-tdTf/WT mice. Immunostaining, tissue clearing, and transgenic reporter lines were used to assess anatomical relationships between nerves and adipocytes. Although synaptic transmission component genes are expressed in adipose tissues (aPVAT, mPVAT, and WAT), strong nerve stimulation with electrical field stimulation does not significantly trigger calcium responses in adipocytes. However, norepinephrine consistently elicits strong calcium responses in adipocytes from all adipose tissues studied. Bethanechol induces minimal adipocyte responses. Imaging neural innervation using various techniques reveals that nerve fibers primarily run alongside blood vessels and rarely branch into the adipose tissue. Although nerve fibers are associated with blood vessels in adipose tissue, they demonstrate limited anatomical and functional interactions with adjacent adipocytes, challenging the concept of classical innervation. These findings dispute the significant involvement of neural input in regulating PVAT adipocyte function and emphasize alternative mechanisms governing adrenergic-driven anticontractile functions of PVAT.NEW & NOTEWORTHY This study challenges prevailing views on neural innervation in perivascular adipose tissue (PVAT) and its role in adrenergic-driven anticontractile effects on vasculature. Contrary to existing paradigms, limited anatomical and functional connections were found between PVAT nerve fibers and adipocytes, underscoring the importance of exploring alternative mechanistic pathways. Understanding the mechanisms involved in PVAT's anticontractile effects is critical for developing potential therapeutic interventions against dysregulated vascular tone, hypertension, and cardiovascular disease.


Subject(s)
Adipocytes , Norepinephrine , Animals , Male , Female , Adipocytes/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Mice , Adipose Tissue/innervation , Adipose Tissue/metabolism , Mice, Inbred C57BL , Synaptic Transmission , Adipose Tissue, White/innervation , Adipose Tissue, White/metabolism , Mice, Transgenic , Calcium Signaling
6.
Nat Rev Neurosci ; 19(3): 153-165, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29449715

ABSTRACT

Interactions between the brain and distinct adipose depots have a key role in maintaining energy balance, thereby promoting survival in response to metabolic challenges such as cold exposure and starvation. Recently, there has been renewed interest in the specific central neuronal circuits that regulate adipose depots. Here, we review anatomical, genetic and pharmacological studies on the neural regulation of adipose function, including lipolysis, non-shivering thermogenesis, browning and leptin secretion. In particular, we emphasize the role of leptin-sensitive neurons and the sympathetic nervous system in modulating the activity of brown, white and beige adipose tissues. We provide an overview of advances in the understanding of the heterogeneity of the brain regulation of adipose tissues and offer a perspective on the challenges and paradoxes that the community is facing regarding the actions of leptin on this system.


Subject(s)
Adipose Tissue/physiology , Brain/physiology , Leptin/physiology , Adipose Tissue/innervation , Animals , Energy Metabolism , Humans , Lipolysis , Neurons/physiology , Sympathetic Nervous System/physiology , Thermogenesis
7.
Am J Physiol Endocrinol Metab ; 318(4): E453-E461, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31961706

ABSTRACT

Beige adipocytes have become a promising therapeutic target to combat obesity. Our senior author Dr. B. Xue previously discovered a transient but significant induction of beige adipocytes in mice during early postnatal development, which peaked at postnatal day (P) 20 and then disappeared thereafter. However, the physiological mechanism underlying the transient induction of the developmental beige cells remains mystery. Interestingly, there exists a postnatal surge of leptin in mice at P10 before the appearance of the developmental beige adipocytes. Given the neurotropic effect of leptin during neuronal development and its role in activating the sympathetic nervous system (SNS), we tested the hypothesis that postnatal leptin surge is required for the transient induction of developmental beige adipocytes through sympathetic innervation. Unlike wild-type (WT) mice that were able to acquire the developmentally induced beige adipocytes at P20, ob/ob mice had much less uncoupling protein 1 (UCP1)-positive multilocular cells in inguinal white adipose tissue at the same age. This was consistent with reduced expression of UCP1 mRNA and protein levels in white fat of ob/ob mice. In contrast, daily injection of ob/ob mice with leptin between P8 and P16, mimicking the postnatal leptin surge, largely rescued the ability of these mice to acquire the developmentally induced beige adipocytes at P20, which was associated with enhanced sympathetic nerve innervation assessed by whole mount adipose tissue immunostaining of tyrosine hydroxylase. Our data demonstrate that the postnatal leptin surge is essential for the developmentally induced beige adipocyte formation in mice, possibly through increasing sympathetic nerve innervation.


Subject(s)
Adipocytes, Beige/metabolism , Adipose Tissue/growth & development , Leptin/metabolism , Adipocytes, Beige/drug effects , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Adipose Tissue/drug effects , Adipose Tissue/innervation , Aging , Animals , Dose-Response Relationship, Drug , Female , Leptin/pharmacology , Male , Mice , Mice, Obese , Sympathetic Nervous System , Tyrosine 3-Monooxygenase/metabolism , Uncoupling Protein 1/metabolism
8.
Mol Med ; 26(1): 126, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33297933

ABSTRACT

BACKGROUND: Innervation of adipose tissue is essential for the proper function of this critical metabolic organ. Numerous surgical and chemical denervation studies have demonstrated how maintenance of brain-adipose communication through both sympathetic efferent and sensory afferent nerves helps regulate adipocyte size, cell number, lipolysis, and 'browning' of white adipose tissue. Neurotrophic factors are growth factors that promote neuron survival, regeneration, and plasticity, including neurite outgrowth and synapse formation. Peripheral immune cells have been shown to be a source of neurotrophic factors in humans and mice. Although a number of immune cells reside in the adipose stromal vascular fraction (SVF), it has remained unclear what roles they play in adipose innervation. We previously demonstrated that adipose SVF secretes brain derived neurotrophic factor (BDNF). METHODS: We now show that deletion of this neurotrophic factor from the myeloid lineage of immune cells led to a 'genetic denervation' of inguinal subcutaneous white adipose tissue (scWAT), thereby causing decreased energy expenditure, increased adipose mass, and a blunted UCP1 response to cold stimulation. RESULTS: We and others have previously shown that noradrenergic stimulation via cold exposure increases adipose innervation in the inguinal depot. Here we have identified a subset of myeloid cells that home to scWAT upon cold exposure and are Ly6C+ CCR2+ Cx3CR1+ monocytes/macrophages that express noradrenergic receptors and BDNF. This subset of myeloid lineage cells also clearly interacted with peripheral nerves in the scWAT and were therefore considered neuroimmune cells. CONCLUSIONS: We propose that these myeloid lineage, cold induced neuroimmune cells (CINCs) are key players in maintaining adipose innervation as well as promoting adipose nerve remodeling under noradrenergic stimulation, such as cold exposure.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/innervation , Adipose Tissue/metabolism , Neuroimmunomodulation , Adipose Tissue, White/immunology , Adipose Tissue, White/innervation , Adipose Tissue, White/metabolism , Animals , Biomarkers , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cold Temperature , Diet , Energy Metabolism , Female , Gene Expression , Male , Mice , Mice, Knockout , Neuroimmunomodulation/genetics , Phenotype , Stress, Physiological
9.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R379-R389, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31868518

ABSTRACT

Blood pressure regulation in health and disease involves a balance between afferent and efferent signals from multiple organs and tissues. Although there are numerous reviews focused on the role of sympathetic nerves in different models of hypertension, few have revised the contribution of afferent nerves innervating adipose tissue and their role in the development of obesity-induced hypertension. Both clinical and basic research support the beneficial effects of bilateral renal denervation in lowering blood pressure. However, recent studies revealed that afferent signals from adipose tissue, in an adipose-brain-peripheral pathway, could contribute to the increased sympathetic activation and blood pressure during obesity. This review focuses on the role of adipose tissue afferent reflexes and briefly describes a number of other afferent reflexes modulating blood pressure. A comprehensive understanding of how multiple afferent reflexes contribute to the pathophysiology of essential and/or obesity-induced hypertension may provide significant insights into improving antihypertensive therapeutic approaches.


Subject(s)
Adipose Tissue/innervation , Blood Pressure , Cardiovascular System/innervation , Hypertension/physiopathology , Obesity/physiopathology , Reflex , Sensory Receptor Cells/metabolism , Sympathetic Nervous System/physiopathology , Humans , Hypertension/etiology , Hypertension/metabolism , Male , Obesity/complications , Obesity/metabolism , Prognosis , Risk Factors
10.
Am J Physiol Gastrointest Liver Physiol ; 317(5): G694-G706, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31509431

ABSTRACT

The pancreas consists of both the exocrine (acini and ducts) and endocrine (islets) compartments to participate in and regulate the body's digestive and metabolic activities. These activities are subjected to neural modulation, but characterization of the human pancreatic afferent and efferent nerves remains difficult because of the lack of three-dimensional (3-D) image data. Here we prepare transparent human donor pancreases for 3-D histology to reveal the pancreatic microstructure, vasculature, and innervation in a global and integrated fashion. The pancreatic neural network consists of the substance P (SP)-positive sensory (afferent) nerves, the vesicular acetylcholine transporter (VAChT)-positive parasympathetic (efferent) nerves, and the tyrosine hydroxylase (TH)-positive sympathetic (efferent) nerves. The SP+ afferent nerves were found residing along the basal domain of the interlobular ducts. The VAChT+ and TH+ efferent nerves were identified at the peri-acinar and perivascular spaces, which follow the blood vessels to the islets. In the intrapancreatic ganglia, the SP+ (scattered minority, ~7%) and VAChT+ neurons co-localize, suggesting a local afferent-efferent interaction. Compared with the mouse pancreas, the human pancreas differs in 1) the lack of SP+ afferent nerves in the islet, 2) the lower ganglionic density, and 3) the obvious presence of VAChT+ and TH+ nerves around the intralobular adipocytes. The latter implicates the neural influence on the pancreatic steatosis. Overall, our 3-D image data reveal the human pancreatic afferent and efferent innervation patterns and provide the anatomical foundation for future high-definition analyses of neural remodeling in human pancreatic diseases.NEW & NOTEWORTHY Modern three-dimensional (3-D) histology with multiplex optical signals identifies the afferent and efferent innervation patterns of human pancreas, which otherwise cannot be defined with standard histology. Our 3-D image data reveal the unexpected association of sensory and parasympathetic nerves/neurons in the intrapancreatic ganglia and identify the sympathetic and parasympathetic nerve contacts with the infiltrated adipocytes. The multiplex approach offers a new way to characterize the human pancreas in remodeling (e.g., fatty infiltration and duct lesion progression).


Subject(s)
Islets of Langerhans/cytology , Neurons, Afferent/cytology , Neurons, Efferent/cytology , Pancreas, Exocrine/cytology , Acinar Cells/cytology , Adipose Tissue/cytology , Adipose Tissue/innervation , Adult , Animals , Female , Humans , Imaging, Three-Dimensional , Islets of Langerhans/innervation , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neuroanatomical Tract-Tracing Techniques , Neurons, Afferent/metabolism , Neurons, Efferent/metabolism , Pancreas, Exocrine/innervation , Substance P/genetics , Substance P/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism
12.
Arterioscler Thromb Vasc Biol ; 38(4): 880-891, 2018 04.
Article in English | MEDLINE | ID: mdl-29496660

ABSTRACT

OBJECTIVE: Healthy perivascular adipose tissue (PVAT) exerts an anticontractile effect on resistance arteries which is vital in regulating arterial tone. Activation of ß3-adrenoceptors by sympathetic nerve-derived NA (noradrenaline) may be implicated in this effect and may stimulate the release of the vasodilator adiponectin from adipocytes. Understanding the mechanisms responsible is vital for determining how PVAT may modify vascular resistance in vivo. APPROACH AND RESULTS: Electrical field stimulation profiles of healthy C57BL/6J mouse mesenteric resistance arteries were characterized using wire myography. During electrical field stimulation, PVAT elicits a reproducible anticontractile effect, which is endothelium independent. To demonstrate the release of an anticontractile factor, the solution surrounding stimulated exogenous PVAT was transferred to a PVAT-denuded vessel. Post-transfer contractility was significantly reduced confirming that stimulated PVAT releases a transferable anticontractile factor. Sympathetic denervation of PVAT using tetrodotoxin or 6-hydroxydopamine completely abolished the anticontractile effect. ß3-adrenoceptor antagonist SR59203A reduced the anticontractile effect, although the PVAT remained overall anticontractile. When the antagonist was used in combination with an OCT3 (organic cation transporter 3) inhibitor, corticosterone, the anticontractile effect was completely abolished. Application of an adiponectin receptor-1 blocking peptide significantly reduced the anticontractile effect in +PVAT arteries. When used in combination with the ß3-adrenoceptor antagonist, there was no further reduction. In adiponectin knockout mice, the anticontractile effect is absent. CONCLUSIONS: The roles of PVAT are 2-fold. First, sympathetic stimulation in PVAT triggers the release of adiponectin via ß3-adrenoceptor activation. Second, PVAT acts as a reservoir for NA, preventing it from reaching the vessel and causing contraction.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/innervation , Adipose Tissue/metabolism , Mesenteric Arteries/metabolism , Norepinephrine/metabolism , Paracrine Communication , Sympathetic Nervous System/metabolism , Vasodilation , Adiponectin/genetics , Adiponectin/metabolism , Animals , In Vitro Techniques , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , Vasoconstriction
13.
Cardiovasc Drugs Ther ; 33(2): 245-259, 2019 04.
Article in English | MEDLINE | ID: mdl-30747398

ABSTRACT

Perivascular adipose tissue (PVAT) is no longer recognised as simply a structural support for the vasculature, and we now know that PVAT releases vasoactive factors which modulate vascular function. Since the discovery of this function in 1991, PVAT research is rapidly growing and the importance of PVAT function in disease is becoming increasingly clear. Obesity is associated with a plethora of vascular conditions; therefore, the study of adipocytes and their effects on the vasculature is vital. PVAT contains an adrenergic system including nerves, adrenoceptors and transporters. In obesity, the autonomic nervous system is dysfunctional; therefore, sympathetic innervation of PVAT may be the key mechanistic link between increased adiposity and vascular disease. In addition, not all obese people develop vascular disease, but a common feature amongst those that do appears to be the inflammatory cell population in PVAT. This review will discuss what is known about sympathetic innervation of PVAT, and the links between nerve activation and inflammation in obesity. In addition, we will examine the therapeutic potential of exercise in sympathetic stimulation of adipose tissue.


Subject(s)
Adipose Tissue/innervation , Cardiovascular Diseases/physiopathology , Inflammation/physiopathology , Obesity/physiopathology , Sympathetic Nervous System/physiopathology , Adipocytes/metabolism , Adipokines/metabolism , Adrenergic Fibers/metabolism , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/prevention & control , Energy Metabolism , Exercise Therapy , Humans , Inflammation/metabolism , Inflammation/prevention & control , Inflammation Mediators/metabolism , Obesity/metabolism , Obesity/therapy , Sympathetic Nervous System/metabolism
14.
Int J Mol Sci ; 20(11)2019 Jun 01.
Article in English | MEDLINE | ID: mdl-31159462

ABSTRACT

Despite tremendous research efforts to identify regulatory factors that control energy metabolism, the prevalence of obesity has been continuously rising, with nearly 40% of US adults being obese. Interactions between secretory factors from adipose tissues and the nervous system innervating adipose tissues play key roles in maintaining energy metabolism and promoting survival in response to metabolic challenges. It is currently accepted that there are three types of adipose tissues, white (WAT), brown (BAT), and beige (BeAT), all of which play essential roles in maintaining energy homeostasis. WAT mainly stores energy under positive energy balance, while it releases fuels under negative energy balance. Thermogenic BAT and BeAT dissipate energy as heat under cold exposure to maintain body temperature. Adipose tissues require neural and endocrine communication with the brain. A number of WAT adipokines and BAT batokines interact with the neural circuits extending from the brain to cooperatively regulate whole-body lipid metabolism and energy homeostasis. We review neuroanatomical, histological, genetic, and pharmacological studies in neuroendocrine regulation of adipose function, including lipid storage and mobilization of WAT, non-shivering thermogenesis of BAT, and browning of BeAT. Recent whole-tissue imaging and transcriptome analysis of differential gene expression in WAT and BAT yield promising findings to better understand the interaction between secretory factors and neural circuits, which represents a novel opportunity to tackle obesity.


Subject(s)
Adipose Tissue/metabolism , Energy Metabolism , Neurosecretory Systems/metabolism , Adipose Tissue/innervation , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Biomarkers , Fatty Acids/metabolism , Homeostasis , Humans , Oxidation-Reduction , Sympathetic Nervous System , Thermogenesis
15.
Cell Immunol ; 330: 183-187, 2018 08.
Article in English | MEDLINE | ID: mdl-29807623

ABSTRACT

Obesity is a worldwide public health concern yet no safe therapies are currently available. The activity of sympathetic neurons is necessary and sufficient for fat mass reduction, via norepinephrine (NE) signaling. Macrophage accumulation in the adipose tissue is thought to play the central role in the onset of obesity, yet their relation to NE has been controversial. We have identified a population of sympathetic neuron-associated macrophages (SAMs) that control obesity via the uptake and clearing of NE. Here we focus on the neuro-immune regulation of obesity by discussing the genetic, cellular and functional signatures of SAMs vis-a-vis adipose tissue macrophages (ATMs).


Subject(s)
Adipose Tissue/immunology , Macrophages/immunology , Neurons/immunology , Obesity/immunology , Sympathetic Nervous System/immunology , Adipocytes/immunology , Adipose Tissue/innervation , Animals , Homeostasis/immunology , Humans , Norepinephrine/immunology , Norepinephrine/metabolism , Obesity/physiopathology , Sympathetic Nervous System/physiopathology
16.
Circ Res ; 118(8): 1244-53, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26926470

ABSTRACT

RATIONALE: It has been reported that epicardial adipose tissue (EAT) may affect myocardial autonomic function. OBJECTIVE: The aim of this study was to explore the relationship between EAT and cardiac sympathetic nerve activity in patients with heart failure. METHODS AND RESULTS: In 110 patients with systolic heart failure, we evaluated the correlation between echocardiographic EAT thickness and cardiac adrenergic nerve activity assessed by (123)I-metaiodobenzylguanidine ((123)I-MIBG). The predictive value of EAT thickness on cardiac sympathetic denervation ((123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score) was tested in a multivariate analysis. Furthermore, catecholamine levels, catecholamine biosynthetic enzymes, and sympathetic nerve fibers were measured in EAT and subcutaneous adipose tissue biopsies obtained from patients with heart failure who underwent cardiac surgery. EAT thickness correlated with (123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score, but not with left ventricular ejection fraction. Moreover, EAT resulted as an independent predictor of (123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score and showed a significant additive predictive value on (123)I-MIBG planar and single-photon emission computed tomography results over demographic and clinical data. Although no differences were found in sympathetic innervation between EAT and subcutaneous adipose tissue, EAT showed an enhanced adrenergic activity demonstrated by the increased catecholamine levels and expression of catecholamine biosynthetic enzymes. CONCLUSIONS: This study provides the first evidence of a direct correlation between increased EAT thickness and cardiac sympathetic denervation in heart failure.


Subject(s)
Adipose Tissue/innervation , Adrenergic Fibers/diagnostic imaging , Heart Failure/diagnostic imaging , Pericardium/innervation , Adipose Tissue/diagnostic imaging , Aged , Cardiac-Gated Single-Photon Emission Computer-Assisted Tomography/methods , Echocardiography/methods , Female , Humans , Male , Middle Aged , Organ Size , Pericardium/diagnostic imaging
17.
Cardiovasc Drugs Ther ; 32(5): 481-502, 2018 10.
Article in English | MEDLINE | ID: mdl-30171461

ABSTRACT

Perivascular adipose tissue (PVAT) refers to the local aggregate of adipose tissue surrounding the vascular tree, exhibiting phenotypes from white to brown and beige adipocytes. Although PVAT has long been regarded as simply a structural unit providing mechanical support to vasculature, it is now gaining reputation as an integral endocrine/paracrine component, in addition to the well-established modulator endothelium, in regulating vascular tone. Since the discovery of anti-contractile effect of PVAT in 1991, the use of multiple rodent models of reduced amounts of PVAT has revealed its regulatory role in vascular remodeling and cardiovascular implications, including atherosclerosis. PVAT does not only release PVAT-derived relaxing factors (PVRFs) to activate multiple subsets of endothelial and vascular smooth muscle potassium channels and anti-inflammatory signals in the vasculature, but it does also provide an interface for neuron-adipocyte interactions in the vascular wall to regulate arterial vascular tone. In this review, we outline our current understanding towards PVAT and attempt to provide hints about future studies that can sharpen the therapeutic potential of PVAT against cardiovascular diseases and their complications.


Subject(s)
Adipocytes/metabolism , Adipokines/metabolism , Adipose Tissue/metabolism , Blood Vessels/metabolism , Vascular Diseases/metabolism , Adipocytes/pathology , Adipose Tissue/innervation , Adipose Tissue/physiopathology , Adiposity , Animals , Blood Vessels/innervation , Blood Vessels/physiopathology , Cellular Microenvironment , Humans , MicroRNAs/metabolism , Paracrine Communication , Phenotype , Signal Transduction , Vascular Diseases/diagnosis , Vascular Diseases/physiopathology , Vasodilation
18.
Lasers Med Sci ; 33(3): 619-625, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29396730

ABSTRACT

Precise nerve localization is of major importance in both surgery and regional anesthesia. Optically based techniques can identify tissue through differences in optical properties, like absorption and scattering. The aim of this study was to evaluate the potential of optical spectroscopy (diffuse reflectance spectroscopy) for clinical nerve identification in vivo. Eighteen patients (8 male, 10 female, age 53 ± 13 years) undergoing inguinal lymph node resection or resection or a soft tissue tumor in the groin were included to measure the femoral or sciatic nerve and the surrounding tissues. In vivo optical measurements were performed using Diffuse Reflectance Spectroscopy (400-1600 nm) on nerve, near nerve adipose tissue, muscle, and subcutaneous fat using a needle-shaped probe. Model-based analyses were used to derive verified quantitative parameters as concentrations of optical absorbers and several parameters describing scattering. A total of 628 optical spectra were recorded. Measured spectra reveal noticeable tissue specific characteristics. Optical absorption of water, fat, and oxy- and deoxyhemoglobin was manifested in the measured spectra. The parameters water and fat content showed significant differences (P < 0.005) between nerve and all surrounding tissues. Classification using k-Nearest Neighbor based on the derived parameters revealed a sensitivity of 85% and a specificity of 79%, for identifying nerve from surrounding tissues. Diffuse Reflectance Spectroscopy identifies peripheral nerve bundles. The differences found between tissue groups are assignable to the tissue composition and structure.


Subject(s)
Optical Imaging/methods , Peripheral Nerves/surgery , Spectrum Analysis/methods , Adipose Tissue/innervation , Female , Hemoglobins , Humans , Male , Middle Aged , Subcutaneous Fat/innervation
19.
Biochem Biophys Res Commun ; 493(1): 40-45, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28928093

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors have both anti-diabetic and anti-obesity effects. However, the precise mechanism of the anti-obesity effect remains unclear. We previously demonstrated that the glycogen depletion signal triggers lipolysis in adipose tissue via liver-brain-adipose neurocircuitry. In this study, therefore, we investigated whether the anti-obesity mechanism of SGLT2 inhibitor is mediated by this mechanism. Diet-induced obese mice were subjected to hepatic vagotomy (HVx) or sham operation and loaded with high fat diet containing 0.015% tofogliflozin (TOFO), a highly selective SGLT2 inhibitor, for 3 weeks. TOFO-treated mice showed a decrease in fat mass and the effect of TOFO was attenuated in HVx group. Although both HVx and sham mice showed a similar level of reduction in hepatic glycogen by TOFO treatment, HVx mice exhibited an attenuated response in protein phosphorylation by protein kinase A (PKA) in white adipose tissue compared with the sham group. As PKA pathway is known to act as an effector of the liver-brain-adipose axis and activate triglyceride lipases in adipocytes, these results indicated that SGLT2 inhibition triggered glycogen depletion signal and actuated liver-brain-adipose axis, resulting in PKA activation in adipocytes. Taken together, it was concluded that the effect of SGLT2 inhibition on weight loss is in part mediated via the liver-brain-adipose neurocircuitry.


Subject(s)
Adipose Tissue/physiology , Benzhydryl Compounds/administration & dosage , Brain/physiology , Glucosides/administration & dosage , Liver/physiology , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2/metabolism , Weight Loss/physiology , Adipose Tissue/drug effects , Adipose Tissue/innervation , Animals , Anti-Obesity Agents/administration & dosage , Brain/drug effects , Liver/drug effects , Liver/innervation , Male , Mice , Mice, Inbred C57BL , Vagotomy , Vagus Nerve/drug effects , Vagus Nerve/physiology , Vagus Nerve/surgery
20.
J Musculoskelet Neuronal Interact ; 17(2): 97-103, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28574416

ABSTRACT

BACKGROUND: Tennis elbow is difficult to treat. The results of surgical treatments are not convincing. Treatment studies on Achilles and patellar tendinopathy targeting the richly innervated and vascularized soft tissues outside the tendon have shown promising outcomes. The innervation patterns in the fibrous/fatty tissues superficially to the elbow extensor origin have not been clarified. METHODS: Nine tissue specimens from the fibrous/fatty tissue covering the extensor origin was taken from seven patients (mean age: 45 years) undergoing surgical treatment for chronic painful tennis elbow. The specimens were stained for morphology (haematoxylin and eosin, H and E) and immunohistochemically for general nerve marker protein gene product 9.5 (PGP 9.5) and markers for sympathetic (tyrosine hydroxylase, TH) and sensory nerve fibres (calcitonin gene-related peptide, CGRP). RESULTS: All specimens contained multiple blood vessels and nerve structures indicated by morphology and immunoreactions. There was a frequent occurrence of TH reactions, especially peri-vascularly, but also in nerve fascicles. Immunoreactions for CGRP were seen in nerve fascicles and isolated nerve fibres. CONCLUSION: The results provide new information on the innervation patterns of the superficial tissues of the extensor origin and their potential as source of tennis elbow pain. LEVEL OF EVIDENCE: IV.


Subject(s)
Elbow Joint/innervation , Tennis Elbow/pathology , Adipose Tissue/diagnostic imaging , Adipose Tissue/innervation , Adult , Connective Tissue/diagnostic imaging , Connective Tissue/innervation , Elbow Joint/diagnostic imaging , Female , Humans , Male , Middle Aged , Tennis Elbow/diagnostic imaging , Ultrasonography, Doppler, Color
SELECTION OF CITATIONS
SEARCH DETAIL