Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Ther ; 32(5): 1407-1424, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38429927

ABSTRACT

Maintaining functional adipose innervation is critical for metabolic health. We found that subcutaneous white adipose tissue (scWAT) undergoes peripheral neuropathy (PN) with obesity, diabetes, and aging (reduced small-fiber innervation and nerve/synaptic/growth-cone/vesicle markers, altered nerve activity). Unlike with nerve injuries, peripheral nerves do not regenerate with PN, and therefore new therapies are needed for treatment of this condition affecting 20-30 million Americans. Here, we validated a gene therapy approach using an adipocyte-tropic adeno-associated virus (AAV; serotype Rec2) to deliver neurotrophic factors (brain-derived neurotrophic factor [BDNF] and nerve growth factor [NGF]) directly to scWAT to improve tissue-specific PN as a proof-of-concept approach. AAVRec2-BDNF intra-adipose delivery improved tissue innervation in obese/diabetic mice with PN, but after longer periods of dietary obesity there was reduced efficacy, revealing a key time window for therapies. AAVRec2-NGF also increased scWAT innervation in obese mice and was more effective than BDNF, likely because Rec2 targeted adipocytes, the tissue's endogenous NGF source. AAVRec2-NGF also worked well even after 25 weeks of dietary obesity, unlike BDNF, which likely needs a vector that targets its physiological cellular source (stromal vascular fraction cells). Given the differing effects of AAVs carrying NGF versus BDNF, a combined therapy may be ideal for PN.


Subject(s)
Adipocytes , Brain-Derived Neurotrophic Factor , Dependovirus , Genetic Therapy , Genetic Vectors , Obesity , Subcutaneous Fat , Animals , Dependovirus/genetics , Obesity/therapy , Obesity/metabolism , Mice , Genetic Therapy/methods , Adipocytes/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Subcutaneous Fat/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Nerve Growth Factor/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Gene Transfer Techniques , Humans , Male , Peripheral Nervous System Diseases/therapy , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/genetics , Transduction, Genetic
2.
Compr Physiol ; 13(3): 4985-5021, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37358505

ABSTRACT

The sympathetic nervous system (SNS) is a crucial arm of the peripheral nervous system (PNS) and includes catecholaminergic neurons that release norepinephrine (NE) onto numerous effector tissues and organs in the body. SNS innervation of both white (WAT) and brown adipose tissue (BAT) is clearly essential for proper tissue function and metabolic control, as decades of surgical, chemical, and genetic denervation studies have demonstrated. Despite our vast knowledge about adipose sympathetic innervation, especially in the context of cold-stimulated browning and thermogenesis that are under SNS control, newer data now provide a nuanced view of the SNS supply to adipose, including its regulation by local neuroimmune cells and neurotrophic factors, the co-release of modulatory neuropeptides along with NE, the importance of local SNS drive to adipose versus systemic increases in circulating catecholamines, and the long-overlooked interplay between adipose sympathetic and sensory nerves. This article brings a modern view to the regulation of sympathetic innervation patterns in WAT and BAT, how to image and quantify the nerve supply, contributions of adipose SNS to tissue functions, and how adipose tissue nerves respond to tissue remodeling and plasticity with changing energy demands. © 2023 American Physiological Society. Compr Physiol 13:4985-5021, 2023.


Subject(s)
Adipose Tissue, Brown , Adipose Tissue, White , Humans , Adipose Tissue, White/metabolism , Adipose Tissue, Brown/metabolism , Norepinephrine/metabolism , Thermogenesis , Obesity/metabolism , Sympathetic Nervous System/physiology
3.
iScience ; 26(3): 106189, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36895649

ABSTRACT

Peripheral neuropathy, which can include axonal degeneration and/or demyelination, impacts adipose tissues with obesity, diabetes, and aging. However, the presence of demyelinating neuropathy had not yet been explored in adipose. Both demyelinating neuropathies and axonopathies implicate Schwann cells (SCs), a glial support cell that myelinates axons and contributes to nerve regeneration after injury. We performed a comprehensive assessment of SCs and myelination patterns of subcutaneous white adipose tissue (scWAT) nerves, and changes across altered energy balance states. We found that mouse scWAT contains both myelinated and unmyelinated nerves and is populated by SCs, including SCs that were associated with synaptic vesicle-containing nerve terminals. BTBR ob/ob mice, a model of diabetic peripheral neuropathy, exhibited small fiber demyelinating neuropathy and alterations in SC marker gene expression in adipose that were similar to obese human adipose. These data indicate that adipose SCs regulate the plasticity of tissue nerves and become dysregulated in diabetes.

4.
Aging Cell ; 22(4): e13784, 2023 04.
Article in English | MEDLINE | ID: mdl-36798047

ABSTRACT

Neural communication between the brain and adipose tissues regulates energy expenditure and metabolism through modulation of adipose tissue functions. We have recently demonstrated that under pathophysiological conditions (obesity, diabetes, and aging), total subcutaneous white adipose tissue (scWAT) innervation is decreased ('adipose neuropathy'). With advanced age in the C57BL/6J mouse, small fiber peripheral nerve endings in adipose tissue die back, resulting in reduced contact with adipose-resident blood vessels and other cells. This vascular neuropathy and parenchymal neuropathy together likely pose a physiological challenge for tissue function. In the current work, we used the genetically diverse HET3 mouse model to investigate the incidence of peripheral neuropathy and adipose tissue dysregulation across several ages in both male and female mice. We also investigated the anti-aging treatment rapamycin, an mTOR inhibitor, as a means to prevent or reduce adipose neuropathy. We found that HET3 mice displayed a reduced neuropathy phenotype compared to inbred C56BL/6 J mice, indicating genetic contributions to this aging phenotype. Compared to female HET3 mice, male HET3 mice had worse neuropathic phenotypes by 62 weeks of age. Female HET3 mice appeared to have increased protection from neuropathy until advanced age (126 weeks), after reproductive senescence. We found that rapamycin overall had little impact on neuropathy measures, and actually worsened adipose tissue inflammation and fibrosis. Despite its success as a longevity treatment in mice, higher doses and longer delivery paradigms for rapamycin may lead to a disconnect between life span and beneficial health outcomes.


Subject(s)
Peripheral Nervous System Diseases , Sirolimus , Male , Female , Animals , Mice , Sirolimus/pharmacology , Longevity/genetics , Mice, Inbred C57BL , Adipose Tissue/metabolism , Obesity/metabolism , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/genetics
5.
Front Endocrinol (Lausanne) ; 13: 864925, 2022.
Article in English | MEDLINE | ID: mdl-35795142

ABSTRACT

Peripheral nerves allow a bidirectional communication between brain and adipose tissues, and many studies have clearly demonstrated that a loss of the adipose nerve supply results in tissue dysfunction and metabolic dysregulation. Neuroimmune cells closely associate with nerves in many tissues, including subcutaneous white adipose tissue (scWAT). However, in scWAT, their functions beyond degrading norepinephrine in an obese state remain largely unexplored. We previously reported that a myeloid-lineage knockout (KO) of brain-derived neurotrophic factor (BDNF) resulted in decreased innervation of scWAT, accompanied by an inability to brown scWAT after cold stimulation, and increased adiposity after a high-fat diet. These data underscored that adipose tissue neuroimmune cells support the peripheral nerve supply to adipose and impact the tissue's metabolic functions. We also reported that a subset of myeloid-lineage monocyte/macrophages (Ly6c+CCR2+Cx3cr1+) is recruited to scWAT in response to cold, a process known to increase neurite density in adipose and promote metabolically healthy processes. These cold-induced neuroimmune cells (CINCs) also expressed BDNF. Here we performed RNAseq on CINCs from cold-exposed and room temperature-housed mice, which revealed a striking and coordinated differential expression of numerous genes involved in neuronal function, including neurotrophin signaling and axonal guidance, further supporting that CINCs fulfill a nerve-supporting role in adipose. The increased expression of leukocyte transendothelial migration genes in cold-stimulated CINCs also confirms prior evidence that they are recruited to scWAT and are not tissue resident. We now provide whole-depot imaging of scWAT from LysM-BDNF KO mice, revealing a striking reduction of innervation across the depot fitting with their reduced energy expenditure phenotype. By contrast, Cx3cr1-BDNF KO mice (a macrophage subset of LysM+ cells) exhibited increased thermogenesis and energy expenditure, with compensatory increased food intake and no change in adiposity or body weight. While these KO mice also exhibit a significantly reduced innervation of scWAT, especially around the subiliac lymph node, they displayed an increase in small fiber sympathetic neurite branching, which may underlie their increased thermogenesis. We propose a homeostatic role of scWAT myeloid-lineage neuroimmune cells together in nerve maintenance and neuro-adipose regulation of energy expenditure.


Subject(s)
Adipose Tissue, White , Brain-Derived Neurotrophic Factor , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Mice , Mice, Knockout , Neuronal Plasticity , Obesity/metabolism , Thermogenesis/genetics
6.
Front Bioeng Biotechnol ; 10: 884601, 2022.
Article in English | MEDLINE | ID: mdl-35646839

ABSTRACT

Objective: Increasing the mass and/or activity of brown adipose tissue (BAT) is one promising avenue for treating obesity and related metabolic conditions, given that BAT has a high potential for energy expenditure and is capable of improving glucose and lipid homeostasis. BAT occurs either in discrete "classical" depots, or interspersed in white adipose tissue (WAT), termed "inducible/recruitable" BAT, or 'beige/brite' adipocytes. We and others have demonstrated that bone morphogenetic protein 7 (BMP7) induces brown adipogenesis in committed and uncommitted progenitor cells, resulting in increased energy expenditure and reduced weight gain in mice. BMP7 is therefore a reliable growth factor to induce browning of WAT. Methods: In this study, we sought to deliver BMP7 specifically to subcutaneous (sc)WAT in order to induce tissue-resident progenitor cells to differentiate into energy-expending recruitable brown adipocytes, without off-target effects like bone formation, which can occur when BMPs are in the presence of bone progenitor cells (outside of WAT). BMP7 delivery directly to WAT may also promote tissue innervation, or directly activate mitochondrial activity in brown adipocytes, as we have demonstrated previously. We utilized silk protein in the form of an injectable hydrogel carrying BMP7. Silk scaffolds are useful for in vivo delivery of substances due to favorable material properties, including controlled release of therapeutic proteins in an active form, biocompatibility with minimal immunogenic response, and prior FDA approval for some medical materials. For this study, the silk was engineered to meet desirable release kinetics for BMP7 in order to mimic our prior in vitro brown adipocyte differentiation studies. Fluorescently-labeled silk hydrogel loaded with BMP7 was directly injected into WAT through the skin and monitored by non-invasive in vivo whole body imaging, including in UCP1-luciferase reporter mice, thereby enabling an approach that is translatable to humans. Results: Injection of the BMP7-loaded silk hydrogels into the subcutaneous WAT of mice resulted in "browning", including the development of multilocular, uncoupling protein 1 (UCP1)-positive brown adipocytes, and an increase in whole-body energy expenditure and skin temperature. In diet-induced obese mice, BMP7-loaded silk delivery to subcutaneous WAT resulted in less weight gain, reduced circulating glucose and lower respiratory exchange ratio (RER). Conclusions: In summary, BMP7 delivery via silk scaffolds directly into scWAT is a novel translational approach to increase browning and energy expenditure, and represents a potential therapeutic avenue for delivering substances directly to adipose depots in pursuit of metabolic treatments.

7.
STAR Protoc ; 3(1): 101109, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35106499

ABSTRACT

Here we provide a clearing-free protocol for processing intact, whole mount subcutaneous white adipose tissue (scWAT) for immunofluorescence as an alternative to current clearing-based approaches. We use a combination of Z-depth reduction and autofluorescence quenching techniques to fluorescently label, image, and quantify adipose tissue innervation effectively throughout intact mouse tissues without the need for optical clearing or light sheet microscopy. This protocol has been optimized and validated for adipose neurovascular labeling. For complete details on the use and execution of this protocol, please refer to Willows et al. (2021).


Subject(s)
Adipose Tissue , Imaging, Three-Dimensional , Adipose Tissue/diagnostic imaging , Animals , Fluorescent Antibody Technique , Imaging, Three-Dimensional/methods , Mice , Microscopy , Staining and Labeling
8.
iScience ; 24(10): 103127, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34622172

ABSTRACT

Little is known about the diversity and function of adipose tissue nerves, due in part to the inability to effectively visualize the tissue's diverse nerve subtypes and the patterns of innervation across an intact depot. The tools to image and quantify adipose tissue innervation are currently limited. Here, we present a method of tissue processing that decreases tissue thickness in the z-axis while leaving cells intact for subsequent immunostaining. This was combined with autofluorescence quenching techniques to permit intact whole tissues to be mounted on slides and imaged by confocal microscopy, with a complementary means to perform whole tissue neurite density quantification after capture of tiled z-stack images. Additionally, we demonstrate how to visualize nerve terminals (the neuro-adipose nexus) in intact blocks of adipose tissue without z-depth reduction. We have included examples of data demonstrating nerve subtypes, neurovascular interactions, label-free imaging of collagen, and nerve bundle digital cross-sections.

9.
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
10.
PLoS One ; 14(9): e0221766, 2019.
Article in English | MEDLINE | ID: mdl-31509546

ABSTRACT

The difficulty in obtaining as well as maintaining weight loss, together with the impairment of metabolic control in conditions like diabetes and cardiovascular disease, may represent pathological situations of inadequate neural communication between the brain and peripheral organs and tissues. Innervation of adipose tissues by peripheral nerves provides a means of communication between the master metabolic regulator in the brain (chiefly the hypothalamus), and energy-expending and energy-storing cells in the body (primarily adipocytes). Although chemical and surgical denervation studies have clearly demonstrated how crucial adipose tissue neural innervation is for maintaining proper metabolic health, we have uncovered that adipose tissue becomes neuropathic (ie: reduction in neurites) in various conditions of metabolic dysregulation. Here, utilizing both human and mouse adipose tissues, we present evidence of adipose tissue neuropathy, or loss of proper innervation, under pathophysiological conditions such as obesity, diabetes, and aging, all of which are concomitant with insult to the adipose organ as well as metabolic dysfunction. Neuropathy is indicated by loss of nerve fiber protein expression, reduction in synaptic markers, and lower neurotrophic factor expression in adipose tissue. Aging-related adipose neuropathy particularly results in loss of innervation around the tissue vasculature, which cannot be reversed by exercise. Together with indications of neuropathy in muscle and bone, these findings underscore that peripheral neuropathy is not restricted to classic tissues like the skin of distal extremities, and that loss of innervation to adipose may trigger or exacerbate metabolic diseases. In addition, we have demonstrated stimulation of adipose tissue neural plasticity with cold exposure, which may ameliorate adipose neuropathy and be a potential therapeutic option to re-innervate adipose and restore metabolic health.


Subject(s)
Adipose Tissue, White/innervation , Aging/metabolism , Diabetes Mellitus/metabolism , Obesity/metabolism , Peripheral Nervous System Diseases/metabolism , Subcutaneous Fat/innervation , Adipose Tissue, White/metabolism , Animals , Body Mass Index , Cold Temperature , Disease Models, Animal , Energy Metabolism , Humans , Male , Mice , Nerve Growth Factors/metabolism , Neuronal Plasticity , Obesity/complications
11.
Biology (Basel) ; 8(1)2019 Feb 12.
Article in English | MEDLINE | ID: mdl-30759876

ABSTRACT

Brown and white adipose tissues are essential for maintenance of proper energy balance and metabolic health. In order to function efficiently, these tissues require both endocrine and neural communication with the brain. Brown adipose tissue (BAT), as well as the inducible brown adipocytes that appear in white adipose tissue (WAT) after simulation, are thermogenic and energy expending. This uncoupling protein 1 (UCP1)-mediated process requires input from sympathetic nerves releasing norepinephrine. In addition to sympathetic noradrenergic signaling, adipose tissue contains sensory nerves that may be important for relaying fuel status to the brain. Chemical and surgical denervation studies of both WAT and BAT have clearly demonstrated the role of peripheral nerves in browning, thermogenesis, lipolysis, and adipogenesis. However, much is still unknown about which subtypes of nerves are present in BAT versus WAT, what nerve products are released from adipose nerves and how they act to mediate metabolic homeostasis, as well as which cell types in adipose are receiving synaptic input. Recent advances in whole-depot imaging and quantification of adipose nerve fibers, as well as other new research findings, have reinvigorated this field of research. This review summarizes the history of research into adipose innervation and brain⁻adipose communication, and also covers landmark and recent research on this topic to outline what we currently know and do not know about adipose tissue nerve supply and communication with the brain.

12.
J Nutr Biochem ; 64: 50-60, 2019 02.
Article in English | MEDLINE | ID: mdl-30439568

ABSTRACT

Consumption of diets that differ in fat type and amount, and sequestration of various fatty acids to tissues and organs likely have effects on overall physiology and metabolic health. However, the contributions of dietary lipids to brain-adipose communication and adipose tissue function are poorly understood. We designed six custom diets that differed only in amount and type of dietary fat, with high or low levels of saturated fatty acids (SFA), omega-6 polyunsaturated fatty acids (n-6 PUFA) or omega-3 (n-3) PUFA. Mice fed the n-3 PUFA diet for 16 weeks displayed a striking reduction in weight gain accompanied by smaller adipose depots and improved glucose sensitivity. Reduced body weight occurred despite lowered energy expenditure and no difference in food intake. Despite the apparent beneficial effects to whole body physiology, we have demonstrated for the first time that a peroxidized n-3-enriched diet led to lipotoxicity of white adipose tissue, as evidenced by increased fibrosis, lipofuscin, reduced anti-inflammatory markers and loss of proper nerve supply. While healthful, n-3 fats are prone to peroxidation, and we observed peroxidated lipid metabolites in the adipose tissue of mice on these diets. Furthermore, using a lipidomics approach, we have observed that brain, white adipose tissue and brown adipose tissue accumulate lipid metabolites differently. The brain remained mostly shielded from changes in dietary fat type and amount, but differences in adipose lipid metabolites across these six diets may have affected metabolic function and brain-adipose communication, as observed in this study.


Subject(s)
Adipose Tissue, White/drug effects , Brain/drug effects , Fatty Acids, Omega-3/adverse effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Brain/metabolism , Dietary Fats, Unsaturated/adverse effects , Energy Metabolism/drug effects , Fatty Acids/analysis , Fatty Acids/metabolism , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/pharmacology , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Peroxides/chemistry , Tissue Distribution , Weight Gain/drug effects
13.
J Diabetes Complications ; 32(3): 249-257, 2018 03.
Article in English | MEDLINE | ID: mdl-29306589

ABSTRACT

BACKGROUND: A complication of diabetes is neuropathy, a condition of sensory axon degeneration that originates in the epidermis. The mechanisms remain unknown but reactive oxygen species (ROS) have been implicated in this condition. In this study, we assessed the role of ROS and a candidate downstream target, MMP-13 in glucose-induced sensory axon degeneration in zebrafish and mice. METHODS: The effects of glucose on metabolism and sensory axon degeneration were assessed using qPCR and live imaging. ROS were analyzed using pentafluorobenzene-sulfonyl fluorescein and activation of the NF-κB stress response was determined using Tg(NF-κB:GFP) zebrafish. The role of MMP-13 and ROS in glucose-dependent axon degeneration was determined in zebrafish following treatment with the antioxidant, N-acetylcysteine and the MMP-13 inhibitor, DB04760. Neuropathic mice fed on a high-fat/high-sugar diet were treated with the MMP-13 inhibitor, CL-82198 to assess sensory recovery. RESULTS: Glucose treatment of zebrafish induced metabolic changes that resemble diabetes. Sensory axon degeneration was mediated by ROS-induced MMP-13 and prevented upon antioxidant treatment or MMP-13 inhibition. MMP-13 inhibition also reversed neuropathy in diabetic mice. CONCLUSION: We demonstrate that zebrafish are suitable to study glucose-induced neurotoxicity. Given the effects in zebrafish and mice, MMP-13 inhibition may be beneficial in the treatment of human diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/etiology , Matrix Metalloproteinase 13/physiology , Neurotoxicity Syndromes/etiology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Glucose , Male , Mice, Inbred C57BL , Zebrafish
14.
Endocrinology ; 158(12): 4233-4245, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29040444

ABSTRACT

The regulation of energy balance involves complex processes in the brain, including coordination by hypothalamic neurons that contain pro-opiomelanocortin (POMC). We previously demonstrated that central bone morphogenetic protein (BMP) 7 reduced appetite. Now we show that a type 1 BMP receptor, BMPR1A, is colocalized with POMC neurons and that POMC-BMPR1A-knockout (KO) mice are hyperphagic, revealing physiological involvement of BMP signaling in anorectic POMC neurons in the regulation of appetite. Surprisingly, the hyperphagic POMC-BMPR1A-KO mice exhibited a lack of obesity, even on a 45% high-fat diet. This is because the brown adipose tissue (BAT) of KO animals exhibited increased sympathetic activation and greater thermogenic capacity owing to a reestablishment of energy balance, most likely stemming from a compensatory increase of BMPR1A in the whole hypothalamus of KO mice. Indeed, control animals given central BMP7 displayed increased energy expenditure and a specific increase in sympathetic nerve activity (SNA) in BAT. In these animals, pharmacological blockade of BMPR1A-SMAD signaling blunted the ability of BMP7 to increase energy expenditure or BAT SNA. Together, we demonstrated an important role for hypothalamic BMP signaling in the regulation of energy balance, including BMPR1A-mediated appetite regulation in POMC neurons as well as hypothalamic BMP-SMAD regulation of the sympathetic drive to BAT for thermogenesis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Energy Metabolism , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Adipose Tissue, Brown/metabolism , Animals , Appetite Regulation/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Diet, High-Fat , Hypothalamus/metabolism , Male , Mice, Knockout , Mice, Transgenic , Obesity/genetics , Obesity/metabolism , Pro-Opiomelanocortin/genetics , Thermogenesis/genetics
15.
Curr Obes Rep ; 5(2): 241-50, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27055864

ABSTRACT

Increasing energy expenditure is an appealing therapeutic target for the prevention and reversal of metabolic conditions such as obesity or type 2 diabetes. However, not enough research has investigated how to exploit pre-existing neural pathways, both in the central nervous system (CNS) and peripheral nervous system (PNS), in order to meet these needs. Here, we review several research areas in this field, including centrally acting pathways known to drive the activation of sympathetic nerves that can increase lipolysis and browning in white adipose tissue (WAT) or increase thermogenesis in brown adipose tissue (BAT), as well as other central and peripheral pathways able to increase energy expenditure of these tissues. In addition, we describe new work investigating the family of transient receptor potential (TRP) channels on metabolically important sensory nerves, as well as the role of the vagus nerve in regulating energy balance.


Subject(s)
Adipose Tissue, Brown/metabolism , Central Nervous System/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Obesity/physiopathology , Sympathetic Nervous System/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/prevention & control , Energy Intake , Energy Metabolism , Humans , Neural Pathways , Obesity/metabolism , Obesity/prevention & control , Thermogenesis , Transient Receptor Potential Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...