1
|
Hanscom M, Morales-Soto W, Watts SW, Jackson WF, Gulbransen BD. Innervation of adipocytes is limited in mouse perivascular adipose tissue. Am J Physiol Heart Circ Physiol 2024; 327:H155-H181. [PMID: 38787382 PMCID: PMC11380956 DOI: 10.1152/ajpheart.00041.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
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.
Collapse
Affiliation(s)
- Marie Hanscom
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| | - Wilmarie Morales-Soto
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
2
|
Lorsignol A, Rabiller L, Labit E, Casteilla L, Pénicaud L. The nervous system and adipose tissues: a tale of dialogues. Am J Physiol Endocrinol Metab 2023; 325:E480-E490. [PMID: 37729026 DOI: 10.1152/ajpendo.00115.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/16/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
White, beige, and brown adipose tissues play a crucial role in maintaining energy homeostasis. Due to the heterogeneous and diffuse nature of fat pads, this balance requires a fine and coordinated control of many actors and therefore permanent dialogues between these tissues and the central nervous system. For about two decades, many studies have been devoted to describe the neuro-anatomical and functional complexity involved to ensure this dialogue. Thus, if it is now clearly demonstrated that there is an efferent sympathetic innervation of different fat depots controlling plasticity as well as metabolic functions of the fat pad, the crucial role of sensory innervation capable of detecting local signals informing the central nervous system of the metabolic state of the relevant pads is much more recent. The purpose of this review is to provide the current state of knowledge on this subject.
Collapse
Affiliation(s)
- Anne Lorsignol
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Lise Rabiller
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Elodie Labit
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Louis Casteilla
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Luc Pénicaud
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| |
Collapse
|
3
|
Esaki N, Matsui T, Tsuda T. Lactate induces the development of beige adipocytes via an increase in the level of reactive oxygen species. Food Funct 2023; 14:9725-9733. [PMID: 37817572 DOI: 10.1039/d3fo03287f] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Recent studies have indicated that lactate acts as a signaling molecule in various tissues. We previously demonstrated that intake of an amino acid mixture combined with exercise synergistically induced beige adipocyte formation in inguinal white adipose tissue (iWAT) in mice. Moreover, plasma lactate levels remained significantly elevated in the amino acid mixture + exercise group even 16 h after exercise, indicating that a lactate-mediated pathway may be involved in the induction of beige adipocyte formation. Against this background, we hypothesized that oral intake of lactate would induce beige adipocyte formation via the lactate signaling pathway without exercise. Furthermore, if oral intake of lactate can produce the same effect as exercise, lactate might be used as a food-derived exercise replacement-factor. Oral intake of lactate (100 mM in drinking water) for 4 weeks significantly induced beige adipocyte formation in iWAT in mice as well as a significant elevation of lactate transporter (monocarboxylic acid transporter 1; MCT1) and lactate dehydrogenase B levels. Administration of lactate to adipocytes significantly increased reactive oxygen species (ROS) and superoxide levels and the NADH/NAD+ ratio. The induction of lactate-mediated uncoupling protein 1 (UCP1) expression and ROS production were significantly suppressed by antioxidant treatment or inhibition of MCT1. However, UCP1 induction was not significantly affected by the inhibition of lactate receptor (hydroxycarboxylic acid receptor 1). These findings suggest that lactate-mediated ROS production induces beige adipocyte formation, and thus oral intake of lactate may confer some benefits of exercise without the need to perform exercise.
Collapse
Affiliation(s)
- Nana Esaki
- College of Bioscience and Biotechnology and Graduate School of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan.
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Toshiro Matsui
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takanori Tsuda
- College of Bioscience and Biotechnology and Graduate School of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan.
| |
Collapse
|
4
|
Gunsch G, Paradie E, Townsend KL. Peripheral nervous system glia in support of metabolic tissue functions. Trends Endocrinol Metab 2023; 34:622-639. [PMID: 37591710 DOI: 10.1016/j.tem.2023.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023]
Abstract
The peripheral nervous system (PNS) relays information between organs and tissues and the brain and spine to maintain homeostasis, regulate tissue functions, and respond to interoceptive and exteroceptive signals. Glial cells perform support roles to maintain nerve function, plasticity, and survival. The glia of the central nervous system (CNS) are well characterized, but PNS glia (PNSG) populations, particularly tissue-specific subtypes, are underexplored. PNSG are found in large nerves (such as the sciatic), the ganglia, and the tissues themselves, and can crosstalk with a range of cell types in addition to neurons. PNSG are also subject to phenotypic changes in response to signals from their local tissue environment, including metabolic changes. These topics and the importance of PNSG in metabolically active tissues, such as adipose, muscle, heart, and lymphatic tissues, are outlined in this review.
Collapse
Affiliation(s)
- Gilian Gunsch
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Emma Paradie
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Willows JW, Gunsch G, Paradie E, Blaszkiewicz M, Tonniges JR, Pino MF, Smith SR, Sparks LM, Townsend KL. Schwann cells contribute to demyelinating diabetic neuropathy and nerve terminal structures in white adipose tissue. iScience 2023; 26:106189. [PMID: 36895649 PMCID: PMC9989657 DOI: 10.1016/j.isci.2023.106189] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/09/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
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.
Collapse
Affiliation(s)
- Jake W Willows
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Gilian Gunsch
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Emma Paradie
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | | | - Jeffrey R Tonniges
- Campus Microscopy and Imaging Facility, The Ohio State University, Columbus, OH, USA
| | - Maria F Pino
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
6
|
Maniyadath B, Zhang Q, Gupta RK, Mandrup S. Adipose tissue at single-cell resolution. Cell Metab 2023; 35:386-413. [PMID: 36889280 PMCID: PMC10027403 DOI: 10.1016/j.cmet.2023.02.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 03/09/2023]
Abstract
Adipose tissue exhibits remarkable plasticity with capacity to change in size and cellular composition under physiological and pathophysiological conditions. The emergence of single-cell transcriptomics has rapidly transformed our understanding of the diverse array of cell types and cell states residing in adipose tissues and has provided insight into how transcriptional changes in individual cell types contribute to tissue plasticity. Here, we present a comprehensive overview of the cellular atlas of adipose tissues focusing on the biological insight gained from single-cell and single-nuclei transcriptomics of murine and human adipose tissues. We also offer our perspective on the exciting opportunities for mapping cellular transitions and crosstalk, which have been made possible by single-cell technologies.
Collapse
Affiliation(s)
- Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Qianbin Zhang
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rana K Gupta
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
7
|
Qian X, Meng X, Zhang S, Zeng W. Neuroimmune regulation of white adipose tissues. FEBS J 2022; 289:7830-7853. [PMID: 34564950 DOI: 10.1111/febs.16213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
The white adipose tissues (WAT) are located in distinct depots throughout the body. They serve as an energy reserve, providing fatty acids for other tissues via lipolysis when needed, and function as an endocrine organ to regulate systemic metabolism. Their activities are coordinated through intercellular communications among adipocytes and other cell types such as residential and infiltrating immune cells, which are collectively under neuronal control. The adipocytes and immune subtypes including macrophages/monocytes, eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2s), T and B cells, dendritic cells (DCs), and natural killer (NK) cells display cellular and functional diversity in response to the energy states and contribute to metabolic homeostasis and pathological conditions. Accumulating evidence reveals that neuronal innervations control lipid deposition and mobilization via regulating lipolysis, adipocyte size, and cellularity. Vice versa, the neuronal innervations and activity are influenced by cellular factors in the WAT. Though the literature describing adipose tissue cells is too extensive to cover in detail, we strive to highlight a selected list of neuronal and immune components in this review. The cell-to-cell communications and the perspective of neuroimmune regulation are emphasized to enlighten the potential therapeutic opportunities for treating metabolic disorders.
Collapse
Affiliation(s)
- Xinmin Qian
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xia Meng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Shan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
8
|
Zeng W, Yang F, Shen WL, Zhan C, Zheng P, Hu J. Interactions between central nervous system and peripheral metabolic organs. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1929-1958. [PMID: 35771484 DOI: 10.1007/s11427-021-2103-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/07/2022] [Indexed: 02/08/2023]
Abstract
According to Descartes, minds and bodies are distinct kinds of "substance", and they cannot have causal interactions. However, in neuroscience, the two-way interaction between the brain and peripheral organs is an emerging field of research. Several lines of evidence highlight the importance of such interactions. For example, the peripheral metabolic systems are overwhelmingly regulated by the mind (brain), and anxiety and depression greatly affect the functioning of these systems. Also, psychological stress can cause a variety of physical symptoms, such as bone loss. Moreover, the gut microbiota appears to play a key role in neuropsychiatric and neurodegenerative diseases. Mechanistically, as the command center of the body, the brain can regulate our internal organs and glands through the autonomic nervous system and neuroendocrine system, although it is generally considered to be outside the realm of voluntary control. The autonomic nervous system itself can be further subdivided into the sympathetic and parasympathetic systems. The sympathetic division functions a bit like the accelerator pedal on a car, and the parasympathetic division functions as the brake. The high center of the autonomic nervous system and the neuroendocrine system is the hypothalamus, which contains several subnuclei that control several basic physiological functions, such as the digestion of food and regulation of body temperature. Also, numerous peripheral signals contribute to the regulation of brain functions. Gastrointestinal (GI) hormones, insulin, and leptin are transported into the brain, where they regulate innate behaviors such as feeding, and they are also involved in emotional and cognitive functions. The brain can recognize peripheral inflammatory cytokines and induce a transient syndrome called sick behavior (SB), characterized by fatigue, reduced physical and social activity, and cognitive impairment. In summary, knowledge of the biological basis of the interactions between the central nervous system and peripheral organs will promote the full understanding of how our body works and the rational treatment of disorders. Thus, we summarize current development in our understanding of five types of central-peripheral interactions, including neural control of adipose tissues, energy expenditure, bone metabolism, feeding involving the brain-gut axis and gut microbiota. These interactions are essential for maintaining vital bodily functions, which result in homeostasis, i.e., a natural balance in the body's systems.
Collapse
Affiliation(s)
- Wenwen Zeng
- Institute for Immunology, and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China. .,Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China. .,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, China.
| | - Fan Yang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
| | - Wei L Shen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Cheng Zhan
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China. .,National Institute of Biological Sciences, Beijing, 102206, China. .,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China.
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China. .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China. .,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China.
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| |
Collapse
|
9
|
Choi HE, Jeon EJ, Kim DY, Choi MJ, Yu H, Kim JI, Cheon HG. Sodium salicylate induces browning of white adipocytes via M2 macrophage polarization by HO-1 upregulation. Eur J Pharmacol 2022; 928:175085. [PMID: 35679889 DOI: 10.1016/j.ejphar.2022.175085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/03/2022]
Abstract
Browning, a white to brown-like (beige) adipocyte conversion, offers a promising therapeutic strategy for the treatment of human obesity. In the present study, the effects of sodium salicylate, a nonsteroidal anti-inflammatory drug, on adipocyte browning were investigated. We found sodium salicylate altered the macrophage phenotype to M2 in RAW264.7 cells, mediated by up-regulation of heme oxygenase-1 (HO-1), and sodium salicylate-treated conditioned medium from macrophages (Sal-M2 CM) induced browning of fully differentiated 3T3-L1 adipocytes. Conversely, the conditioned medium obtained from macrophages when treated with sodium salicylate in the presence of either ZnPP (a HO-1 inhibitor) or HO-1 siRNA did not induce browning. In association with macrophage HO-1 induction by sodium salicylate, iron production also increased, and deferoxamine (an iron chelator) blunted the browning effects of Sal-M2 CM, suggesting that iron may play a role in the Sal-M2 CM-induced browning. The in vivo browning effects of sodium salicylate were confirmed in ob/ob mice, whereas in vivo macrophage depletion by clodronate as well as HO-1 blockade by either ZnPP or adeno-associated virus carrying HO-1 shRNA (AAV-HO-1 shRNA) attenuated the browning effects of sodium salicylate. These results reveal sodium salicylate induces browning in vitro and in vivo by up-regulating HO-1 thus promoting M2 polarization.
Collapse
Affiliation(s)
- Hye-Eun Choi
- Department of Pharmacology, Gachon University School of Medicine, Incheon 21999, Republic of Korea
| | - Eun Jeong Jeon
- Department of Pharmacology, Gachon University School of Medicine, Incheon 21999, Republic of Korea
| | - Dong Young Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| | - Mi Jin Choi
- Department of Pharmacology, Gachon University School of Medicine, Incheon 21999, Republic of Korea
| | - Hana Yu
- Department of Pharmacology, Gachon University School of Medicine, Incheon 21999, Republic of Korea
| | - Jea Il Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| | - Hyae Gyeong Cheon
- Department of Pharmacology, Gachon University School of Medicine, Incheon 21999, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea.
| |
Collapse
|
10
|
Nguyen TT, Hulme J, Vo TK, Van Vo G. The Potential Crosstalk Between the Brain and Visceral Adipose Tissue in Alzheimer's Development. Neurochem Res 2022; 47:1503-1512. [PMID: 35298764 DOI: 10.1007/s11064-022-03569-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/25/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
The bidirectional communication between the brain and peripheral organs have been widely documented, but the impact of visceral adipose tissue (VAT) dysfunction and its relation to structural and functional brain changes have yet to be fully elucidated. This review initially examines the clinical evidence supporting associations between the brain and VAT before visiting the roles of the autonomic nervous system, fat and glucose metabolism, neuroinflammation, and metabolites. Finally, the possible effects and potential mechanisms of the brain-VAT axis on the pathogenesis of Alzheimer's disease are discussed, providing new insights regarding future prevention and therapeutic strategies.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, HUTECH University, Ho Chi Minh City, 700000, Vietnam
| | - John Hulme
- Department of BioNano Technology, Gachon University, Seongnam, 461-701, Republic of Korea.
| | - Tuong Kha Vo
- Vietnam Sports Hospital, Ministry of Culture, Sports and Tourism, Hanoi, 100000, Vietnam.,Department of Sports Medicine, University of Medicine and Pharmacy (VNU-UMP), Vietnam National University Hanoi, Hanoi, 100000, Vietnam
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam. .,Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam. .,Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
11
|
Kojima T, Esaki N, Tsuda T. Combination of Exercise and Intake of Amino Acid Mixture Synergistically Induces Beige Adipocyte Formation in Mice. J Nutr Sci Vitaminol (Tokyo) 2021; 67:225-233. [PMID: 34470997 DOI: 10.3177/jnsv.67.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Exercise combined with dietary factors may have significant effects on the suppression of body fat accumulation. Several trials suggest that amino acid mixtures containing alanine, arginine, and phenylalanine (ARF) combined with exercise can significantly reduce body fat accumulation in overweight adults and high-fat diet-induced obesity in mice. We therefore hypothesized that combining ARF and exercise would significantly induce beige adipocyte formation and that this would contribute to reducing body weight, whereas administration of ARF or exercise alone would not. Administration of ARF (1 g/kg body weight, daily) combined with exercise (5 sessions per week) for 4 wk significantly induced formation of beige adipocytes in inguinal white adipose tissue (iWAT) in mice, although ARF or exercise alone did not. Metabolomic analysis showed that plasma lactate concentration was significantly elevated in the exercise+ARF group relative to the exercise group. Furthermore, lactate dehydrogenase B, which increases redox stress by converting lactate to pyruvate in iWAT and triggers induction of uncoupling protein 1 expression was significantly upregulated in iWAT of the exercise+ARF group. These findings demonstrate the unique effect of ARF combined with exercise for inducing beige adipocyte formation, which may be associated with the suggested lactate-mediated pathway. Appropriate mixtures of amino acids could be used as a dietary supplement before exercise and contributed to increasing energy expenditures.
Collapse
Affiliation(s)
- Takuya Kojima
- College of Bioscience and Biotechnology and Graduate School of Bioscience and Biotechnology, Chubu University
| | - Nana Esaki
- College of Bioscience and Biotechnology and Graduate School of Bioscience and Biotechnology, Chubu University
| | - Takanori Tsuda
- College of Bioscience and Biotechnology and Graduate School of Bioscience and Biotechnology, Chubu University
| |
Collapse
|
12
|
The Role of Lipids, Lipid Metabolism and Ectopic Lipid Accumulation in Axon Growth, Regeneration and Repair after CNS Injury and Disease. Cells 2021; 10:cells10051078. [PMID: 34062747 PMCID: PMC8147289 DOI: 10.3390/cells10051078] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/20/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Axons in the adult mammalian nervous system can extend over formidable distances, up to one meter or more in humans. During development, axonal and dendritic growth requires continuous addition of new membrane. Of the three major kinds of membrane lipids, phospholipids are the most abundant in all cell membranes, including neurons. Not only immature axons, but also severed axons in the adult require large amounts of lipids for axon regeneration to occur. Lipids also serve as energy storage, signaling molecules and they contribute to tissue physiology, as demonstrated by a variety of metabolic disorders in which harmful amounts of lipids accumulate in various tissues through the body. Detrimental changes in lipid metabolism and excess accumulation of lipids contribute to a lack of axon regeneration, poor neurological outcome and complications after a variety of central nervous system (CNS) trauma including brain and spinal cord injury. Recent evidence indicates that rewiring lipid metabolism can be manipulated for therapeutic gain, as it favors conditions for axon regeneration and CNS repair. Here, we review the role of lipids, lipid metabolism and ectopic lipid accumulation in axon growth, regeneration and CNS repair. In addition, we outline molecular and pharmacological strategies to fine-tune lipid composition and energy metabolism in neurons and non-neuronal cells that can be exploited to improve neurological recovery after CNS trauma and disease.
Collapse
|
13
|
Abstract
Adipose tissue depots in distinct anatomical locations mediate key aspects of metabolism, including energy storage, nutrient release, and thermogenesis. Although adipocytes make up more than 90% of adipose tissue volume, they represent less than 50% of its cellular content. Here, I review recent advances in genetic lineage tracing and transcriptomics that reveal the identities of the heterogeneous cell populations constituting mouse and human adipose tissues. In addition to mature adipocytes and their progenitors, these include endothelial and various immune cell types that together orchestrate adipose tissue development and functions. One salient finding is the identification of progenitor subtypes that can modulate adipogenic capacity through paracrine mechanisms. Another is the description of fate trajectories of monocyte/macrophages, which can respond maladaptively to nutritional and thermogenic stimuli, leading to metabolic disease. These studies have generated an extraordinary source of publicly available data that can be leveraged to explore commonalities and differences among experimental models, providing new insights into adipose tissues and their role in metabolic disease.
Collapse
Affiliation(s)
- Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA;
| |
Collapse
|
14
|
Abstract
Integrated immunometabolic responses link dietary intake, energy utilization, and storage to immune regulation of tissue function and is therefore essential for the maintenance and restoration of homeostasis. Adipose-resident leukocytes have non-traditional immunological functions that regulate organismal metabolism by controlling insulin action, lipolysis, and mitochondrial respiration to control the usage of substrates for production of heat versus ATP. Energetically expensive vital functions such as immunological responses might have thus evolved to respond accordingly to dietary surplus and deficit of macronutrient intake. Here, we review the interaction of dietary intake of macronutrients and their metabolism with the immune system. We discuss immunometabolic checkpoints that promote healthspan and highlight how dietary fate and regulation of glucose, fat, and protein metabolism might affect immunity.
Collapse
Affiliation(s)
- Aileen H Lee
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Vishwa Deep Dixit
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
15
|
Nishikawa S, Kamiya M, Aoyama H, Yoshimura K, Miyata R, Kumazawa S, Tsuda T. Co-Administration of Curcumin and Artepillin C Induces Development of Brown-Like Adipocytes in Association with Local Norepinephrine Production by Alternatively Activated Macrophages in Mice. J Nutr Sci Vitaminol (Tokyo) 2020; 65:328-334. [PMID: 31474682 DOI: 10.3177/jnsv.65.328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Classical brown adipocytes, characterized by interscapular depots, have multilocular fat depots and are known to release excess energy. Recent studies have shown that induction of brown-like adipocytes, also referred to as beige or brite cells, in white adipose tissue (WAT) results in the release of excess energy through mitochondrial heat production via uncoupling protein 1. This has potential a therapeutic strategy for obesity and related diseases as well as classical brown adipocytes. In our previous studies, we found that artepillin C (ArtC, 10 mg/kg body weight), a characteristic constituent of Brazilian propolis, significantly induced the development of brown-like adipocytes in inguinal WAT (iWAT) of mice. Furthermore, we recently demonstrated that curcumin (Cur, 4.5 mg/kg) also significantly induced the development of brown-like adipocytes in mice. The combined administration of several food-derived factors can enhance their bioactivity and reduce their required functional doses. In this study, we showed that co-administration of Cur and ArtC at lower doses (Cur, 1.5 mg/kg; ArtC, 5 mg/kg) additively induce brown-like adipocyte development in mouse iWAT. Moreover, this induction is associated with the localized production of norepinephrine following accumulation of alternatively activated macrophages in iWAT. These findings suggest that co-administration of Cur and ArtC is significantly effective to reduce the dose and enhance the formation of brown-like adipocyte via a unique molecular mechanism.
Collapse
Affiliation(s)
- Sho Nishikawa
- College of Bioscience and Biotechnology, Chubu University
| | - Misa Kamiya
- College of Bioscience and Biotechnology, Chubu University
| | - Hiroki Aoyama
- College of Bioscience and Biotechnology, Chubu University
| | - Kazuki Yoshimura
- Department of Food and Nutritional Sciences, University of Shizuoka
| | - Ryo Miyata
- Department of Food and Nutritional Sciences, University of Shizuoka
| | | | - Takanori Tsuda
- College of Bioscience and Biotechnology, Chubu University
| |
Collapse
|
16
|
Saxton SN, Clark BJ, Withers SB, Eringa EC, Heagerty AM. Mechanistic Links Between Obesity, Diabetes, and Blood Pressure: Role of Perivascular Adipose Tissue. Physiol Rev 2019; 99:1701-1763. [PMID: 31339053 DOI: 10.1152/physrev.00034.2018] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Obesity is increasingly prevalent and is associated with substantial cardiovascular risk. Adipose tissue distribution and morphology play a key role in determining the degree of adverse effects, and a key factor in the disease process appears to be the inflammatory cell population in adipose tissue. Healthy adipose tissue secretes a number of vasoactive adipokines and anti-inflammatory cytokines, and changes to this secretory profile will contribute to pathogenesis in obesity. In this review, we discuss the links between adipokine dysregulation and the development of hypertension and diabetes and explore the potential for manipulating adipose tissue morphology and its immune cell population to improve cardiovascular health in obesity.
Collapse
Affiliation(s)
- Sophie N Saxton
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Ben J Clark
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Sarah B Withers
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Etto C Eringa
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| | - Anthony M Heagerty
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom; School of Environment and Life Sciences, University of Salford, Salford, United Kingdom; and Department of Physiology, VU University Medical Centre, Amsterdam, Netherlands
| |
Collapse
|
17
|
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.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China
- Institute of Vascular Medicine, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hamidah Abu Bakar
- Health Sciences Department, Universiti Selangor, 40000, Shah Alam, Selangor, Malaysia
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC)-a joint cooperation between the Charité-University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.
- Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Yu Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China.
- Institute of Vascular Medicine, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
18
|
Guilherme A, Henriques F, Bedard AH, Czech MP. Molecular pathways linking adipose innervation to insulin action in obesity and diabetes mellitus. Nat Rev Endocrinol 2019; 15:207-225. [PMID: 30733616 PMCID: PMC7073451 DOI: 10.1038/s41574-019-0165-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adipose tissue comprises adipocytes and many other cell types that engage in dynamic crosstalk in a highly innervated and vascularized tissue matrix. Although adipose tissue has been studied for decades, it has been appreciated only in the past 5 years that extensive arborization of nerve fibres has a dominant role in regulating the function of adipose tissue. This Review summarizes the latest literature, which suggests that adipocytes signal to local sensory nerve fibres in response to perturbations in lipolysis and lipogenesis. Such adipocyte signalling to the central nervous system causes sympathetic output to distant adipose depots and potentially other metabolic tissues to regulate systemic glucose homeostasis. Paracrine factors identified in the past few years that mediate such adipocyte-neuron crosstalk are also reviewed. Similarly, immune cells and endothelial cells within adipose tissue communicate with local nerve fibres to modulate neurotransmitter tone, blood flow, adipocyte differentiation and energy expenditure, including adipose browning to produce heat. This understudied field of neurometabolism related to adipose tissue biology has great potential to reveal new mechanistic insights and potential therapeutic strategies for obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Felipe Henriques
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alexander H Bedard
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
19
|
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.
Collapse
Affiliation(s)
- Sophie N Saxton
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility (3rd floor), 46 Grafton Street, M13 9NT, Manchester, UK.
| | - Sarah B Withers
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility (3rd floor), 46 Grafton Street, M13 9NT, Manchester, UK
- School of Environment and Life Sciences, University of Salford, Manchester, UK
| | - Anthony M Heagerty
- Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility (3rd floor), 46 Grafton Street, M13 9NT, Manchester, UK
| |
Collapse
|
20
|
Nishikawa S, Hyodo T, Nagao T, Nakanishi A, Tandia M, Tsuda T. α-Monoglucosyl Hesperidin but Not Hesperidin Induces Brown-Like Adipocyte Formation and Suppresses White Adipose Tissue Accumulation in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:1948-1954. [PMID: 30691268 DOI: 10.1021/acs.jafc.8b06647] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Hesperidin (HES) is a flavanone glycoside found in citrus peel that contributes to its bitter taste. It has low water solubility and poor oral bioavailability. To improve its solubility and bioavailability, α-monoglucosyl hesperidin (αGH) has been synthesized from HES by transglucosylation using cyclodextrin glucanotransferase. Several reports indicate that αGH significantly decreases body fat, but the underlying molecular mechanism remains unclear. We hypothesized that the antiobesity effects of αGH occur through the induced formation of brown-like adipocytes. The present study verified that dietary αGH induces brown-like adipocytes to form in mouse inguinal white adipose tissue (iWAT), thereby significantly decreasing the weight of white adipose tissue (WAT). Furthermore, dietary αGH significantly induced thermogenesis in iWAT. Dietary αGH also significantly suppressed high-fat-diet-induced WAT accumulation in mice, which may be mediated by brown-like adipocyte formation. These results indicate that dietary αGH induces increased energy expenditure by stimulating the formation of brown-like adipocytes.
Collapse
Affiliation(s)
- Sho Nishikawa
- College of Bioscience and Biotechnology , Chubu University , Matsumoto-cho, Kasugai , Aichi 487-8501 , Japan
| | - Takuma Hyodo
- College of Bioscience and Biotechnology , Chubu University , Matsumoto-cho, Kasugai , Aichi 487-8501 , Japan
| | - Tsubasa Nagao
- College of Bioscience and Biotechnology , Chubu University , Matsumoto-cho, Kasugai , Aichi 487-8501 , Japan
| | - Akihito Nakanishi
- Toyo Sugar Refining Company, Limited , Chuo-ku, Tokyo 103-0016 , Japan
| | - Mahamadou Tandia
- Toyo Sugar Refining Company, Limited , Chuo-ku, Tokyo 103-0016 , Japan
| | - Takanori Tsuda
- College of Bioscience and Biotechnology , Chubu University , Matsumoto-cho, Kasugai , Aichi 487-8501 , Japan
| |
Collapse
|
21
|
Luo Y, Liu B, Yang X, Ma X, Zhang X, Bragin DE, Yang XO, Huang W, Liu M. Myeloid adrenergic signaling via CaMKII forms a feedforward loop of catecholamine biosynthesis. J Mol Cell Biol 2018; 9:422-434. [PMID: 29087480 DOI: 10.1093/jmcb/mjx046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 10/25/2017] [Indexed: 11/13/2022] Open
Abstract
Type 2 immune response has been shown to facilitate cold-induced thermogenesis and browning of white fat. However, whether alternatively activated macrophages produce catecholamine and substantially promote adaptive thermogenesis in adipose tissue remains controversial. Here, we show that tyrosine hydroxylase (TyrH), a rate-limiting enzyme of catecholamine biosynthesis, was expressed and phosphorylated in adipose-resident macrophages. In addition, the plasma level of adrenaline was increased by cold stress in mice, and treatment of macrophages with adrenaline stimulated phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and TyrH. Genetic and pharmacological inhibition of CaMKII or PKA signaling diminished adrenaline-induced phosphorylation of TyrH in primary macrophages. Consistently, overexpression of constitutively active CaMKII upregulated basal TyrH phosphorylation, while suppressing the stimulatory effect of adrenaline on TyrH in macrophages. Myeloid-specific disruption of CaMKIIγ suppressed both the cold-induced production of norepinephrine and adipose UCP1 expression in vivo and the stimulatory effect of adrenaline on macrophage-dependent activation of brown adipocytes in vitro. Lack of CaMKII signaling attenuated catecholamine production mediated by cytokines IL-4 and IL-13, key inducers of type 2 immune response in primary macrophages. Taken together, these results suggest a feedforward mechanism of adrenaline in adipose-resident macrophages, and that myeloid CaMKII signaling plays an important role in catecholamine production and subsequent beige fat activation.
Collapse
Affiliation(s)
- Yan Luo
- Department of Endocrinology and Metabolism, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center School of Medicine, Albuquerque, NM, USA
| | - Bilian Liu
- Department of Endocrinology and Metabolism, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xin Yang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center School of Medicine, Albuquerque, NM, USA
| | - Xiaoxiao Ma
- Department of Diabetes Complications & Metabolism Research, City of Hope, Duarte, CA, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center School of Medicine, Albuquerque, NM, USA.,Department of Microbiology and Molecular Genetics, University of New Mexico Health Sciences Center School of Medicine, Albuquerque, NM, USA
| | - Denis E Bragin
- Department of Neurosurgery, University of New Mexico Health Sciences Center School of Medicine, Albuquerque, NM, USA
| | - Xuexian O Yang
- Department of Microbiology and Molecular Genetics, University of New Mexico Health Sciences Center School of Medicine, Albuquerque, NM, USA
| | - Wendong Huang
- Department of Diabetes Complications & Metabolism Research, City of Hope, Duarte, CA, USA
| | - Meilian Liu
- Department of Endocrinology and Metabolism, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
22
|
Nishikawa S, Kamiya M, Aoyama H, Nomura M, Hyodo T, Ozeki A, Lee H, Takahashi T, Imaizumi A, Tsuda T. Highly Dispersible and Bioavailable Curcumin but not Native Curcumin Induces Brown-Like Adipocyte Formation in Mice. Mol Nutr Food Res 2018; 62. [PMID: 29334590 DOI: 10.1002/mnfr.201700731] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/21/2017] [Indexed: 12/21/2022]
Abstract
SCOPE The induction of brown-like adipocytes in white adipose tissue (WAT) is a potential therapeutic target for the treatment of obesity and metabolic disorders via the ability of these cells to release excess energy as heat in association with uncoupling protein 1. Some experimental trials suggest that curcumin (a yellow pigment from turmeric) has a suppressive effect on the accumulation of body fat. However, there is little evidence to show that curcumin induces the formation of brown-like adipocytes and the molecular mechanisms involved remain elusive. In addition, in most experimental trials, high doses of curcumin are administered. METHODS AND RESULTS Highly dispersible and bioavailable curcumin (HC, i.e., 4.5 mg native curcumin kg-1 ) but not the same dose of native curcumin induces the formation of brown-like adipocytes in mouse inguinal WAT. Moreover, the formation of brown-like adipocytes induced by HC in inguinal WAT may be mediated by the production of local norepinephrine from accumulated alternatively activated macrophages. CONCLUSION These novel findings suggest that curcumin increases energy expenditure by inducing the formation of brown-like adipocytes via a unique molecular mechanism. Importantly, they show that HC has significant bioactive effects in vivo at lower doses of curcumin.
Collapse
Affiliation(s)
- Sho Nishikawa
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Misa Kamiya
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Hiroki Aoyama
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Mami Nomura
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Takuma Hyodo
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Aoi Ozeki
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | | | | | | | - Takanori Tsuda
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| |
Collapse
|
23
|
Jiang H, Ding X, Cao Y, Wang H, Zeng W. Dense Intra-adipose Sympathetic Arborizations Are Essential for Cold-Induced Beiging of Mouse White Adipose Tissue. Cell Metab 2017; 26:686-692.e3. [PMID: 28918935 DOI: 10.1016/j.cmet.2017.08.016] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 12/28/2022]
Abstract
Efferent signals from the central nervous system represent a key layer of regulation of white adipose tissue (WAT). However, the mechanism by which efferent neural signals control WAT metabolism remains to be better understood. Here, we exploit the volume fluorescence-imaging technique to visualize the neural arborizations in mouse inguinal WAT at single-fiber resolution. The imaging reveals a dense network of sympathetic arborizations that had been previously undetected by conventional methods, with sympathetic fibers being in close apposition to > 90% of adipocytes. We demonstrate that these sympathetic fibers originate from the celiac ganglia, which are activated by cold challenge. Sympathetic-specific deletion of TrkA receptor or pharmacologic ablation by 6-hydroxydopamine abolishes these intra-adipose arborizations and, as a result, cold-induced beiging of inguinal WAT. Furthermore, we find that local sympathetic arborizations function through beta-adrenergic receptors in this beiging process. These findings uncover an essential link connecting efferent neural signals with metabolism of individual adipocytes.
Collapse
Affiliation(s)
- Haochen Jiang
- Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaofan Ding
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ying Cao
- Center for Life Sciences, Tsinghua University, Beijing 100084, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Huanhuan Wang
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Wenwen Zeng
- Center for Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
24
|
Kiehn JT, Tsang AH, Heyde I, Leinweber B, Kolbe I, Leliavski A, Oster H. Circadian Rhythms in Adipose Tissue Physiology. Compr Physiol 2017; 7:383-427. [PMID: 28333377 DOI: 10.1002/cphy.c160017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The different types of adipose tissues fulfill a wide range of biological functions-from energy storage to hormone secretion and thermogenesis-many of which show pronounced variations over the course of the day. Such 24-h rhythms in physiology and behavior are coordinated by endogenous circadian clocks found in all tissues and cells, including adipocytes. At the molecular level, these clocks are based on interlocked transcriptional-translational feedback loops comprised of a set of clock genes/proteins. Tissue-specific clock-controlled transcriptional programs translate time-of-day information into physiologically relevant signals. In adipose tissues, clock gene control has been documented for adipocyte proliferation and differentiation, lipid metabolism as well as endocrine function and other adipose oscillations are under control of systemic signals tied to endocrine, neuronal, or behavioral rhythms. Circadian rhythm disruption, for example, by night shift work or through genetic alterations, is associated with changes in adipocyte metabolism and hormone secretion. At the same time, adipose metabolic state feeds back to central and peripheral clocks, adjusting behavioral and physiological rhythms. In this overview article, we summarize our current knowledge about the crosstalk between circadian clocks and energy metabolism with a focus on adipose physiology. © 2017 American Physiological Society. Compr Physiol 7:383-427, 2017.
Collapse
Affiliation(s)
- Jana-Thabea Kiehn
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Anthony H Tsang
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Isabel Heyde
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Brinja Leinweber
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Isa Kolbe
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Alexei Leliavski
- Institute of Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Henrik Oster
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| |
Collapse
|
25
|
Ayala-Lopez N, Watts SW. New actions of an old friend: perivascular adipose tissue's adrenergic mechanisms. Br J Pharmacol 2016; 174:3454-3465. [PMID: 27813085 DOI: 10.1111/bph.13663] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/03/2016] [Accepted: 10/21/2016] [Indexed: 12/17/2022] Open
Abstract
The revolutionary discovery in 1991 by Soltis and Cassis that perivascular adipose tissue (PVAT) has an anti-contractile effect changed how we think about the vasculature. Most experiments on vascular pharmacology begin by removing the fat surrounding vessels. Thus, PVAT was thought to have a minor role in vascular function and its presence was just for structural support. The need to rethink PVAT's role was precipitated by observations that obesity carries a high cardiovascular risk and PVAT dysfunction is associated with obesity. PVAT is a vascular-adipose organ that has intimate connections with the nervous and immune system. A complex world of physiology resides in PVAT, including the presence of an 'adrenergic system' that is able to release, take up and metabolize noradrenaline. Adipocytes, stromal vascular cells and nerves within PVAT contain components that make up this adrenergic system. Some of the great strides in PVAT research came from studying adipose tissue as a whole. Adipose tissue has many roles and participates in regulating energy balance, energy stores, inflammation and thermoregulation. However, PVAT is dissimilar from non-PVAT adipose tissues. PVAT is intimately connected with the vasculature, which is what makes its role in body homeostasis unique. The adrenergic system within PVAT may be an integral link connecting the effects of obesity with the vascular dysfunction observed in obesity-associated hypertension, a condition in which the sympathetic nervous system has a significant role. This review will explore what is known about the adrenergic system in adipose tissue and PVAT, plus the translational importance of these findings. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Nadia Ayala-Lopez
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
26
|
Adipose tissue macrophage in immune regulation of metabolism. SCIENCE CHINA-LIFE SCIENCES 2016; 59:1232-1240. [DOI: 10.1007/s11427-016-0155-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
|
27
|
Nishikawa S, Aoyama H, Kamiya M, Higuchi J, Kato A, Soga M, Kawai T, Yoshimura K, Kumazawa S, Tsuda T. Artepillin C, a Typical Brazilian Propolis-Derived Component, Induces Brown-Like Adipocyte Formation in C3H10T1/2 Cells, Primary Inguinal White Adipose Tissue-Derived Adipocytes, and Mice. PLoS One 2016; 11:e0162512. [PMID: 27598888 PMCID: PMC5012562 DOI: 10.1371/journal.pone.0162512] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/05/2016] [Indexed: 12/15/2022] Open
Abstract
Induction of brown-like adipocytes (beige/brite cells) in white adipose tissue (WAT) suggests a new approach for preventing and treating obesity via induction of thermogenesis associated with uncoupling protein 1 (UCP1). However, whether diet-derived factors can directly induce browning of white adipocytes has not been well established. In addition, the underlying mechanism of induction of brown-like adipocytes by diet-derived factors has been unclear. Here, we demonstrate that artepillin C (ArtC), which is a typical Brazilian propolis-derived component, significantly induces brown-like adipocytes in murine C3H10T1/2 cells and primary inguinal WAT (iWAT)-derived adipocytes. This significant induction is due to activation of peroxisome proliferator-activated receptor γ and stabilization of PRD1-BF-1-RIZ1 homologous domain-containing protein-16 (PRDM16). Furthermore, the oral administration of ArtC (10 mg/kg) for 4 weeks significantly induced brown-like adipocytes accompanied by significant expression of UCP1 and PRDM16 proteins in iWAT of mice, and was independent of the β3-adrenergic signaling pathway via the sympathetic nervous system. These findings may provide insight into browning of white adipocytes including the molecular mechanism mediated by dietary factors and demonstrate that ArtC has a novel biological function with regard to increasing energy expenditure by browning of white adipocytes.
Collapse
MESH Headings
- Adipocytes, Brown/cytology
- Adipocytes, Brown/drug effects
- Adipocytes, Brown/metabolism
- Adipocytes, White/cytology
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Adipose Tissue, Brown/cytology
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Administration, Oral
- Animals
- Anti-Obesity Agents/isolation & purification
- Anti-Obesity Agents/pharmacology
- Cell Line
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Energy Metabolism/drug effects
- Energy Metabolism/genetics
- Gene Expression Regulation
- Male
- Mice
- Mice, Inbred C57BL
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Obesity/prevention & control
- PPAR gamma/agonists
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Phenylpropionates/isolation & purification
- Phenylpropionates/pharmacology
- Primary Cell Culture
- Propolis/chemistry
- Signal Transduction
- Thermogenesis/drug effects
- Thermogenesis/genetics
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Uncoupling Protein 1/agonists
- Uncoupling Protein 1/genetics
- Uncoupling Protein 1/metabolism
Collapse
Affiliation(s)
- Sho Nishikawa
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Hiroki Aoyama
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Misa Kamiya
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Jun Higuchi
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Aiko Kato
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Minoru Soga
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Taeko Kawai
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Kazuki Yoshimura
- Department of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Shigenori Kumazawa
- Department of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | - Takanori Tsuda
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan
| |
Collapse
|
28
|
Garcia E, Becker VGC, McCullough DJ, Stabley JN, Gittemeier EM, Opoku-Acheampong AB, Sieman DW, Behnke BJ. Blood flow responses to mild-intensity exercise in ectopic vs. orthotopic prostate tumors; dependence upon host tissue hemodynamics and vascular reactivity. J Appl Physiol (1985) 2016; 121:15-24. [PMID: 27125846 DOI: 10.1152/japplphysiol.00266.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/27/2016] [Indexed: 12/23/2022] Open
Abstract
Given the critical role of tumor O2 delivery in patient prognosis and the rise in preclinical exercise oncology studies, we investigated tumor and host tissue blood flow at rest and during exercise as well as vascular reactivity using a rat prostate cancer model grown in two transplantation sites. In male COP/CrCrl rats, blood flow (via radiolabeled microspheres) to prostate tumors [R3327-MatLyLu cells injected in the left flank (ectopic) or ventral prostate (orthotopic)] and host tissue was measured at rest and during a bout of mild-intensity exercise. α-Adrenergic vasoconstriction to norepinephrine (NE: 10(-9) to 10(-4) M) was determined in arterioles perforating the tumors and host tissue. To determine host tissue exercise hyperemia in healthy tissue, a sham-operated group was included. Blood flow was lower at rest and during exercise in ectopic tumors and host tissue (subcutaneous adipose) vs. the orthotopic tumor and host tissue (prostate). During exercise, blood flow to the ectopic tumor significantly decreased by 25 ± 5% (SE), whereas flow to the orthotopic tumor increased by 181 ± 30%. Maximal vasoconstriction to NE was not different between arterioles from either tumor location. However, there was a significantly higher peak vasoconstriction to NE in subcutaneous adipose arterioles (92 ± 7%) vs. prostate arterioles (55 ± 7%). Establishment of the tumor did not alter host tissue blood flow from either location at rest or during exercise. These data demonstrate that blood flow in tumors is dependent on host tissue hemodynamics and that the location of the tumor may critically affect how exercise impacts the tumor microenvironment and treatment outcomes.
Collapse
Affiliation(s)
- Emmanuel Garcia
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| | - Veronika G C Becker
- Department of Kinesiology, Kansas State University, Manhattan, Kansas; Department of Sports Science, Leipzig University, Leipzig, Germany
| | - Danielle J McCullough
- Department of Anatomy & Physiology, Edward Via College of Osteopathic Medicine, Auburn Campus, Auburn, Alabama
| | - John N Stabley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | | | | | - Dietmar W Sieman
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, Florida
| | - Bradley J Behnke
- Johnson Cancer Research Center, Kansas State University, Manhattan, Kansas; Department of Kinesiology, Kansas State University, Manhattan, Kansas;
| |
Collapse
|
29
|
Parisi V, Rengo G, Perrone-Filardi P, Pagano G, Femminella GD, Paolillo S, Petraglia L, Gambino G, Caruso A, Grimaldi MG, Baldascino F, Nolano M, Elia A, Cannavo A, De Bellis A, Coscioni E, Pellegrino T, Cuocolo A, Ferrara N, Leosco D. Increased Epicardial Adipose Tissue Volume Correlates With Cardiac Sympathetic Denervation in Patients With Heart Failure. Circ Res 2016; 118:1244-53. [PMID: 26926470 DOI: 10.1161/circresaha.115.307765] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/26/2016] [Indexed: 12/31/2022]
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.
Collapse
Affiliation(s)
- Valentina Parisi
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Giuseppe Rengo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.).
| | - Pasquale Perrone-Filardi
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Gennaro Pagano
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Grazia Daniela Femminella
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Stefania Paolillo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Laura Petraglia
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Giuseppina Gambino
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Aurelio Caruso
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Maria Gabriella Grimaldi
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Francesco Baldascino
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Maria Nolano
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Andrea Elia
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Alessandro Cannavo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Antonio De Bellis
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Enrico Coscioni
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Teresa Pellegrino
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Alberto Cuocolo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Nicola Ferrara
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Dario Leosco
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| |
Collapse
|
30
|
Guigas B, Molofsky AB. A worm of one's own: how helminths modulate host adipose tissue function and metabolism. Trends Parasitol 2015; 31:435-41. [PMID: 25991556 PMCID: PMC4567404 DOI: 10.1016/j.pt.2015.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/15/2015] [Accepted: 04/16/2015] [Indexed: 12/16/2022]
Abstract
Parasitic helminths have coexisted with human beings throughout time. Success in eradicating helminths has limited helminth-induced morbidity and mortality but is also correlated with increasing rates of 'western' diseases, including metabolic syndrome and type 2 diabetes. Recent studies in mice describe how type 2 immune cells, traditionally associated with helminth infection, maintain adipose tissue homeostasis and promote adipose tissue beiging, protecting against obesity and metabolic dysfunction. Here, we review these studies and discuss how helminths and helminth-derived molecules may modulate these physiologic pathways to improve metabolic functions in specific tissues, such as adipose and liver, as well as at the whole-organism level.
Collapse
Affiliation(s)
- Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands; Department of Molecular Cellular Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ari B Molofsky
- Department of Microbiology & Immunology, University of California, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
31
|
Abstract
Adipose tissue resident leukocytes are often cast solely as the effectors of obesity and its attendant pathologies; however, recent observations have demonstrated that these cells support and effect 'healthy' physiologic function as well as pathologic dysfunction. Importantly, these two disparate outcomes are underpinned by similarly disparate immune programs; type 2 responses instruct and promote metabolic normalcy, while type 1 responses drive tissue dysfunction. In this Review, we summarize the literature regarding type 2 immunity's role in adipose tissue physiology and allude to its potential therapeutic implications.
Collapse
|
32
|
Abstract
There are nineteen different receptor proteins for adenosine, adenine and uridine nucleotides, and nucleotide sugars, belonging to three families of G protein-coupled adenosine and P2Y receptors, and ionotropic P2X receptors. The majority are functionally expressed in blood vessels, as purinergic receptors in perivascular nerves, smooth muscle and endothelial cells, and roles in regulation of vascular contractility, immune function and growth have been identified. The endogenous ligands for purine receptors, ATP, ADP, UTP, UDP and adenosine, can be released from different cell types within the vasculature, as well as from circulating blood cells, including erythrocytes and platelets. Many purine receptors can be activated by two or more of the endogenous ligands. Further complexity arises because of interconversion between ligands, notably adenosine formation from the metabolism of ATP, leading to complex integrated responses through activation of different subtypes of purine receptors. The enzymes responsible for this conversion, ectonucleotidases, are present on the surface of smooth muscle and endothelial cells, and may be coreleased with neurotransmitters from nerves. What selectivity there is for the actions of purines/pyrimidines comes from differential expression of their receptors within the vasculature. P2X1 receptors mediate the vasocontractile actions of ATP released as a neurotransmitter with noradrenaline (NA) from sympathetic perivascular nerves, and are located on the vascular smooth muscle adjacent to the nerve varicosities, the sites of neurotransmitter release. The relative contribution of ATP and NA as functional cotransmitters varies with species, type and size of blood vessel, neuronal firing pattern, the tone/pressure of the blood vessel, and in ageing and disease. ATP is also a neurotransmitter in non-adrenergic non-cholinergic perivascular nerves and mediates vasorelaxation via smooth muscle P2Y-like receptors. ATP and adenosine can act as neuromodulators, with the most robust evidence being for prejunctional inhibition of neurotransmission via A1 adenosine receptors, but also prejunctional excitation and inhibition of neurotransmission via P2X and P2Y receptors, respectively. P2Y2, P2Y4 and P2Y6 receptors expressed on the vascular smooth muscle are coupled to vasocontraction, and may have a role in pathophysiological conditions, when purines are released from damaged cells, or when there is damage to the protective barrier that is the endothelium. Adenosine is released during hypoxia to increase blood flow via vasodilator A2A and A2B receptors expressed on the endothelium and smooth muscle. ATP is released from endothelial cells during hypoxia and shear stress and can act at P2Y and P2X4 receptors expressed on the endothelium to increase local blood flow. Activation of endothelial purine receptors leads to the release of nitric oxide, hyperpolarising factors and prostacyclin, which inhibits platelet aggregation and thus ensures patent blood flow. Vascular purine receptors also regulate endothelial and smooth muscle growth, and inflammation, and thus are involved in the underlying processes of a number of cardiovascular diseases.
Collapse
Affiliation(s)
- Vera Ralevic
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.
| | - William R Dunn
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
33
|
Oriowo MA. Perivascular adipose tissue, vascular reactivity and hypertension. Med Princ Pract 2015; 24 Suppl 1:29-37. [PMID: 24503717 PMCID: PMC6489082 DOI: 10.1159/000356380] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 10/09/2013] [Indexed: 12/13/2022] Open
Abstract
Most blood vessels are surrounded by a variable amount of adventitial adipose tissue, perivascular adipose tissue (PVAT), which was originally thought to provide mechanical support for the vessel. It is now known that PVAT secretes a number of bioactive substances including vascular endothelial growth factor, tumor necrosis factor-alpha (TNF-α), leptin, adiponectin, insulin-like growth factor, interleukin-6, plasminogen activator substance, resistin and angiotensinogen. Several studies have shown that PVAT significantly modulated vascular smooth muscle contractions induced by a variety of agonists and electrical stimulation by releasing adipocyte-derived relaxing (ADRF) and contracting factors. The identity of ADRF is not yet known. However, several vasodilators have been suggested including adiponectin, angiotensin 1-7, hydrogen sulfide and methyl palmitate. The anticontractile effect of PVAT is mediated through the activation of potassium channels since it is abrogated by inhibiting potassium channels. Hypertension is characterized by a reduction in the size and amount of PVAT and this is associated with the attenuated anticontractile effect of PVAT in hypertension. However, since a reduction in size and amount of PVAT and the attenuated anticontractile effect of PVAT were already evident in prehypertensive rats with no evidence of impaired release of ADRF, there is the possibility that the anticontractile effect of PVAT was not directly related to an altered function of the adipocytes per se. Hypertension is characterized by low-grade inflammation and infiltration of macrophages. One of the adipokines secreted by macrophages is TNF-α. It has been shown that exogenously administered TNF-α enhanced agonist-induced contraction of a variety of vascular smooth muscle preparations and reduced endothelium-dependent relaxation. Other procontractile factors released by the PVAT include angiotensin II and superoxide. It is therefore possible that the loss could be due to an increased amount of these proinflammatory and procontractile factors. More studies are definitely required to confirm this.
Collapse
Affiliation(s)
- Mabayoje A Oriowo
- Department of Pharmacology and Toxicology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
34
|
Abstract
A detailed appreciation of the control of adipose tissue whether it be white, brown or brite/beige has never been more important to the development of a framework on which to build therapeutic strategies to combat obesity. This is because 1) the rate of fatty acid release into the circulation from lipolysis in white adipose tissue (WAT) is integrally important to the development of obesity, 2) brown adipose tissue (BAT) has now moved back to center stage with the realization that it is present in adult humans and, in its activated form, is inversely proportional to levels of obesity and 3) the identification and characterization of "brown-like" or brite/beige fat is likely to be one of the most exciting developments in adipose tissue biology in the last decade. Central to all of these developments is the role of the CNS in the control of different fat cell functions and central to CNS control is the integrative capacity of the hypothalamus. In this chapter we will attempt to detail key issues relevant to the structure and function of hypothalamic and downstream control of WAT and BAT and highlight the importance of developing an understanding of the neural input to brite/beige fat cells as a precursor to its recruitment as therapeutic target.
Collapse
Affiliation(s)
- A Stefanidis
- Department of Physiology, Monash University, Clayton, 3800, Australia
| | - N M Wiedmann
- Department of Physiology, Monash University, Clayton, 3800, Australia
| | - E S Adler
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - B J Oldfield
- Department of Physiology, Monash University, Clayton, 3800, Australia.
| |
Collapse
|
35
|
Bartness TJ, Liu Y, Shrestha YB, Ryu V. Neural innervation of white adipose tissue and the control of lipolysis. Front Neuroendocrinol 2014; 35:473-93. [PMID: 24736043 PMCID: PMC4175185 DOI: 10.1016/j.yfrne.2014.04.001] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/10/2014] [Accepted: 04/04/2014] [Indexed: 01/22/2023]
Abstract
White adipose tissue (WAT) is innervated by the sympathetic nervous system (SNS) and its activation is necessary for lipolysis. WAT parasympathetic innervation is not supported. Fully-executed SNS-norepinephrine (NE)-mediated WAT lipolysis is dependent on β-adrenoceptor stimulation ultimately hinging on hormone sensitive lipase and perilipin A phosphorylation. WAT sympathetic drive is appropriately measured electrophysiologically and neurochemically (NE turnover) in non-human animals and this drive is fat pad-specific preventing generalizations among WAT depots and non-WAT organs. Leptin-triggered SNS-mediated lipolysis is weakly supported, whereas insulin or adenosine inhibition of SNS/NE-mediated lipolysis is strongly supported. In addition to lipolysis control, increases or decreases in WAT SNS drive/NE inhibit and stimulate white adipocyte proliferation, respectively. WAT sensory nerves are of spinal-origin and sensitive to local leptin and increases in sympathetic drive, the latter implicating lipolysis. Transsynaptic viral tract tracers revealed WAT central sympathetic and sensory circuits including SNS-sensory feedback loops that may control lipolysis.
Collapse
Affiliation(s)
- Timothy J Bartness
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA.
| | - Yang Liu
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA; Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yogendra B Shrestha
- Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vitaly Ryu
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA; Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Eosinophils and type 2 cytokine signaling in macrophages orchestrate development of functional beige fat. Cell 2014; 157:1292-1308. [PMID: 24906148 DOI: 10.1016/j.cell.2014.03.066] [Citation(s) in RCA: 665] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 03/06/2014] [Accepted: 03/31/2014] [Indexed: 12/19/2022]
Abstract
Beige fat, which expresses the thermogenic protein UCP1, provides a defense against cold and obesity. Although a cold environment is the physiologic stimulus for inducing beige fat in mice and humans, the events that lead from the sensing of cold to the development of beige fat remain poorly understood. Here, we identify the efferent beige fat thermogenic circuit, consisting of eosinophils, type 2 cytokines interleukin (IL)-4/13, and alternatively activated macrophages. Genetic loss of eosinophils or IL-4/13 signaling impairs cold-induced biogenesis of beige fat. Mechanistically, macrophages recruited to cold-stressed subcutaneous white adipose tissue (scWAT) undergo alternative activation to induce tyrosine hydroxylase expression and catecholamine production, factors required for browning of scWAT. Conversely, administration of IL-4 to thermoneutral mice increases beige fat mass and thermogenic capacity to ameliorate pre-established obesity. Together, our findings have uncovered the efferent circuit controlling biogenesis of beige fat and provide support for its targeting to treat obesity.
Collapse
|
37
|
Bulloch JM, Daly CJ. Autonomic nerves and perivascular fat: interactive mechanisms. Pharmacol Ther 2014; 143:61-73. [PMID: 24560685 DOI: 10.1016/j.pharmthera.2014.02.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 12/31/2022]
Abstract
The evidence describing the autonomic innervation of body fat is reviewed with a particular focus on the role of the sympathetic neurotransmitters. In compiling the evidence, a strong case emerges for the interaction between autonomic nerves and perivascular adipose tissue (PVAT). Adipocytes have been shown to express receptors for neurotransmitters released from nearby sympathetic varicosities such as adrenoceptors (ARs), purinoceptors and receptors for neuropeptide Y (NPY). Noradrenaline can modulate both lipolysis (via α2- and β3-ARs) and lipogenesis (via α1- and β3-ARs). ATP can inhibit lipolysis (via P1 purinoceptors) or stimulate lipolysis (via P2y purinoceptors). NPY, which can be produced by adipocytes and sympathetic nerves, inhibits lipolysis. Thus the sympathetic triad of transmitters can influence adipocyte free fatty acid (FFA) content. Substance P (SP) released from sensory nerves has also been shown to promote lipolysis. Therefore, we propose a mechanism whereby sympathetic neurotransmission can simultaneously activate smooth muscle cells in the tunica media to cause vasoconstriction and alter FFA content and release from adjacent adipocytes in PVAT. The released FFA can influence endothelial function. Adipocytes also release a range of vasoactive substances, both relaxing and contractile factors, including adiponectin and reactive oxygen species. The action of adipokines (such as adiponectin) and reactive oxygen species (ROS) on cells of the vascular adventitia and nerves has yet to be fully elucidated. We hypothesise a strong link between PVAT and autonomic fibres and suggest that this poorly understood relationship is extremely important for normal vascular function and warrants a detailed study.
Collapse
Affiliation(s)
- Janette M Bulloch
- School of Science, University of the West of Scotland, Hamilton ML3 0JB, Scotland.
| | - Craig J Daly
- School of Life Sciences, University of Glasgow, Glasgow G128QQ, Scotland.
| |
Collapse
|
38
|
Mul JD, O’Duibhir E, Shrestha YB, Koppen A, Vargoviç P, Toonen PW, Zarebidaki E, Kvetnansky R, Kalkhoven E, Cuppen E, Bartness TJ. Pmch-deficiency in rats is associated with normal adipocyte differentiation and lower sympathetic adipose drive. PLoS One 2013; 8:e60214. [PMID: 23555928 PMCID: PMC3608591 DOI: 10.1371/journal.pone.0060214] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 02/22/2013] [Indexed: 02/01/2023] Open
Abstract
The orexigenic neuropeptide melanin-concentrating hormone (MCH), a product of Pmch, is an important mediator of energy homeostasis. Pmch-deficient rodents are lean and smaller, characterized by lower food intake, body-, and fat mass. Pmch is expressed in hypothalamic neurons that ultimately are components in the sympathetic nervous system (SNS) drive to white and interscapular brown adipose tissue (WAT, iBAT, respectively). MCH binds to MCH receptor 1 (MCH1R), which is present on adipocytes. Currently it is unknown if Pmch-ablation changes adipocyte differentiation or sympathetic adipose drive. Using Pmch-deficient and wild-type rats on a standard low-fat diet, we analyzed dorsal subcutaneous and perirenal WAT mass and adipocyte morphology (size and number) throughout development, and indices of sympathetic activation in WAT and iBAT during adulthood. Moreover, using an in vitro approach we investigated the ability of MCH to modulate 3T3-L1 adipocyte differentiation. Pmch-deficiency decreased dorsal subcutaneous and perirenal WAT mass by reducing adipocyte size, but not number. In line with this, in vitro 3T3-L1 adipocyte differentiation was unaffected by MCH. Finally, adult Pmch-deficient rats had lower norepinephrine turnover (an index of sympathetic adipose drive) in WAT and iBAT than wild-type rats. Collectively, our data indicate that MCH/MCH1R-pathway does not modify adipocyte differentiation, whereas Pmch-deficiency in laboratory rats lowers adiposity throughout development and sympathetic adipose drive during adulthood.
Collapse
Affiliation(s)
- Joram D. Mul
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Eoghan O’Duibhir
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yogendra B. Shrestha
- Department of Biology, Neurobiology and Behavior Program, and Exploring and Testing Strategies for Obesity Reversal Center, Georgia State University, Atlanta, Georgia, United States of America
| | - Arjen Koppen
- Department of Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter Vargoviç
- Laboratory for Stress Research, Institute of Experimental Endocrinology, Bratislava, Slovakia
| | - Pim W. Toonen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Eleen Zarebidaki
- Department of Biology, Neurobiology and Behavior Program, and Exploring and Testing Strategies for Obesity Reversal Center, Georgia State University, Atlanta, Georgia, United States of America
| | - Richard Kvetnansky
- Laboratory for Stress Research, Institute of Experimental Endocrinology, Bratislava, Slovakia
| | - Eric Kalkhoven
- Department of Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Edwin Cuppen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Timothy J. Bartness
- Department of Biology, Neurobiology and Behavior Program, and Exploring and Testing Strategies for Obesity Reversal Center, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
39
|
Kvetnansky R, Ukropec J, Laukova M, Manz B, Pacak K, Vargovic P. Stress stimulates production of catecholamines in rat adipocytes. Cell Mol Neurobiol 2012; 32:801-13. [PMID: 22402834 PMCID: PMC3419009 DOI: 10.1007/s10571-012-9822-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 02/17/2012] [Indexed: 11/28/2022]
Abstract
The sympathoadrenal system is the main source of catecholamines (CAs) in adipose tissues and therefore plays the key role in the regulation of adipose tissue metabolism. We recently reported existence of an alternative CA-producing system directly in adipose tissue cells, and here we investigated effect of various stressors-physical (cold) and emotional stress (immobilization) on dynamics of this system. Acute or chronic cold exposure increased intracellular norepinephrine (NE) and epinephrine (EPI) concentration in isolated rat mesenteric adipocytes. Gene expression of CA biosynthetic enzymes did not change in adipocytes but was increased in stromal vascular fraction (SVF) after 28 day cold. Exposure of rats to a single IMO stress caused increases in NE and EPI levels, and also gene expression of CA biosynthetic enzymes in adipocytes. In SVF changes were similar but more pronounced. Animals adapted to a long-term cold exposure (28 days, 4°C) did not show those responses found after a single IMO stress either in adipocytes or SVF. Our data indicate that gene machinery accommodated in adipocytes, which is able to synthesize NE and EPI de novo, is significantly activated by stress. Cold-adapted animals keep their adaptation even after an exposure to a novel stressor. These findings suggest the functionality of CAs produced endogenously in adipocytes. Taken together, the newly discovered CA synthesizing system in adipocytes is activated in stress situations and might significantly contribute to regulation of lipolysis and other metabolic or thermogenetic processes.
Collapse
Affiliation(s)
- R Kvetnansky
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
40
|
van der Spek R, Kreier F, Fliers E, Kalsbeek A. Circadian rhythms in white adipose tissue. PROGRESS IN BRAIN RESEARCH 2012; 199:183-201. [DOI: 10.1016/b978-0-444-59427-3.00011-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
41
|
Cao L, Choi EY, Liu X, Martin A, Wang C, Xu X, During MJ. White to brown fat phenotypic switch induced by genetic and environmental activation of a hypothalamic-adipocyte axis. Cell Metab 2011; 14:324-38. [PMID: 21907139 PMCID: PMC3172615 DOI: 10.1016/j.cmet.2011.06.020] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 05/07/2011] [Accepted: 06/21/2011] [Indexed: 12/12/2022]
Abstract
Living in an enriched environment with complex physical and social stimulation leads to improved cognitive and metabolic health. In white fat, enrichment induced the upregulation of the brown fat cell fate determining gene Prdm16, brown fat-specific markers, and genes involved in thermogenesis and β-adrenergic signaling. Moreover, pockets of cells with prototypical brown fat morphology and high UCP1 levels were observed in the white fat of enriched mice associated with resistance to diet-induced obesity. Hypothalamic overexpression of BDNF reproduced the enrichment-associated activation of the brown fat gene program and lean phenotype. Inhibition of BDNF signaling by genetic knockout or dominant-negative trkB reversed this phenotype. Our genetic and pharmacologic data suggest a mechanism whereby induction of hypothalamic BDNF expression in response to environmental stimuli leads to selective sympathoneural modulation of white fat to induce "browning" and increased energy dissipation.
Collapse
Affiliation(s)
- Lei Cao
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Wu LE, Hocking SL, James DE. Macrophage infiltration and cytokine release in adipose tissue: angiogenesis or inflammation? Diabetol Int 2010. [DOI: 10.1007/s13340-010-0003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
43
|
Pénicaud L. Relationships between adipose tissues and brain: what do we learn from animal studies? DIABETES & METABOLISM 2010; 36 Suppl 3:S39-44. [DOI: 10.1016/s1262-3636(10)70465-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
44
|
Inskip J, Plunet W, Ramer L, Ramsey JB, Yung A, Kozlowski P, Ramer M, Krassioukov A. Cardiometabolic risk factors in experimental spinal cord injury. J Neurotrauma 2010; 27:275-85. [PMID: 19772460 DOI: 10.1089/neu.2009.1064] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cardiometabolic risk factors are sorely underreported after spinal cord injury (SCI), despite the high prevalence of metabolic disorders and cardiovascular mortality in this population. Body-composition analysis and serum-lipid profiling are two assessments that are beginning to be more widely used to document metabolic changes after clinical SCI. Individuals with SCI have been reported to carry increased visceral fat and to exhibit altered serum-lipid levels. However, little is known about the development of these cardiometabolic risk factors in animal models. Using a combination of magnetic resonance imaging (MRI) and adipose tissue dissection, we show that visceral and subcutaneous adipose tissue were both increased at 1 month, but not at 1 week, after complete T3 SCI in rats. Additionally, at 1 month post injury, T3 SCI rats exhibited nonfasting serum hypertriglyceridemia, a result obtained using both standard clinical methods and a home cholesterol monitoring device (CardioChek). Interestingly, at 1 month post injury, rats with complete T10 SCI did not show an increase in either visceral adiposity or serum triglyceride levels. The fact that complete high-thoracic SCI disrupts lipid metabolism and perturbs fat storage in the subacute period, while low-thoracic SCI does not, suggests that differences in descending sympathetic control of adipose tissue might play a role in these changes. These results provide the first evidence of cardiometabolic risk factors in experimental animals with SCI, and are a starting point for investigations of the etiology of obesity and metabolic dysfunctions that often accompany SCI.
Collapse
Affiliation(s)
- Jessica Inskip
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Bartness TJ, Shrestha YB, Vaughan CH, Schwartz GJ, Song CK. Sensory and sympathetic nervous system control of white adipose tissue lipolysis. Mol Cell Endocrinol 2010; 318:34-43. [PMID: 19747957 PMCID: PMC2826518 DOI: 10.1016/j.mce.2009.08.031] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 08/25/2009] [Accepted: 08/26/2009] [Indexed: 12/14/2022]
Abstract
Circulating factors are typically invoked to explain bidirectional communication between the CNS and white adipose tissue (WAT). Thus, initiation of lipolysis has been relegated primarily to adrenal medullary secreted catecholamines and the inhibition of lipolysis primarily to pancreatic insulin, whereas signals of body fat levels to the brain have been ascribed to adipokines such as leptin. By contrast, evidence is given for bidirectional communication between brain and WAT occurring via the sympathetic nervous system (SNS) and sensory innervation of this tissue. Using retrograde transneuronal viral tract tracers, the SNS outflow from brain to WAT has been defined. Functionally, sympathetic denervation of WAT blocks lipolysis to a variety of lipolytic stimuli. Using anterograde transneuronal viral tract tracers, the sensory input from WAT to brain has been defined. Functionally, these WAT sensory nerves respond electrophysiologically to increases in WAT SNS drive suggesting a possible neural negative feedback loop to regulate lipolysis.
Collapse
Affiliation(s)
- Timothy J Bartness
- Department of Biology and Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA.
| | | | | | | | | |
Collapse
|
46
|
Abstract
Circadian clocks time the daily occurrence of multiple aspects of behaviour and physiology. Through studies of chronic misalignment between our internal clocks and the environment (e.g. during shift work), it has long been postulated that disruption of circadian rhythms is detrimental to human health. Recent advances in understanding of the cellular and molecular basis of mammalian circadian timing mechanisms have identified many key genes involved in circadian rhythm generation and demonstrated the presence of clocks throughout the body. Furthermore, clear links between sleep, circadian rhythms and metabolic function have been revealed, and much current research is studying these links in more detail. Here, we review the evidence linking circadian rhythms, clock genes and adipose biology. We also highlight gaps in our understanding and finally suggest avenues for future research.
Collapse
Affiliation(s)
- J D Johnston
- Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK.
| | | | | |
Collapse
|
47
|
Pettersson US, Henriksnäs J, Jansson L. Reversal of high pancreatic islet and white adipose tissue blood flow in type 2 diabetic GK rats by administration of the beta3-adrenoceptor inhibitor SR-59230A. Am J Physiol Endocrinol Metab 2009; 297:E490-4. [PMID: 19491297 DOI: 10.1152/ajpendo.00140.2009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies have shown that the Goto-Kakizaki (GK) rat, a nonobese type 2 diabetes model, has an increased white adipose tissue (WAT) and islet blood flow when compared with control rats. The aim of the study was to examine if these increased blood flow values in GK rats could be affected by the beta(3)-adrenoceptor antagonist SR-59230A. We measured organ blood flow with a microsphere technique 10 min after administration of SR-59230A (1 mg/kg body wt), or the corresponding volume of 0.9% NaCl solution (1 ml/kg body wt) in rats anaesthetized with thiobutabarbital. The GK rat had an increased blood flow in all intra-abdominal adipose tissue depots except for the sternal fat pad compared with Wistar-Furth (WF) rats. However, no differences were seen in the blood perfusion of subcutaneous white or brown adipose tissue. The blood flow was also increased in both the pancreas and in the islets in the GK rat compared with WF rats. SR-59230A treatment affected neither WAT nor pancreatic blood flow in WF rats. In GK rats, on the other hand, SR-59230A decreased both WAT and islet blood flow values to values similar to those seen in control WF rats. The whole pancreatic blood flow was not affected by SR-59230A administration in GK rats. Interestingly, the brown adipose tissue blood flow in GK rats increased after SR-59230A administration. These results suggest that beta(3)-adrenoceptors are involved in regulation of blood flow both in islet and in adipose tissue.
Collapse
MESH Headings
- Adipose Tissue, Brown/blood supply
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, White/blood supply
- Adipose Tissue, White/drug effects
- Adrenergic beta-3 Receptor Antagonists
- Angiogenesis Inhibitors/therapeutic use
- Animal Structures/blood supply
- Animal Structures/drug effects
- Animals
- Diabetes Mellitus, Type 2/drug therapy
- Drug Evaluation, Preclinical
- Gene Expression/drug effects
- Islets of Langerhans/blood supply
- Islets of Langerhans/drug effects
- Male
- Propanolamines/therapeutic use
- Rats
- Rats, Inbred Strains
- Rats, Inbred WF
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
- Regional Blood Flow/drug effects
Collapse
Affiliation(s)
- U S Pettersson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| | | | | |
Collapse
|
48
|
Tchivileva IE, Tan KS, Gambarian M, Nackley AG, Medvedev AV, Romanov S, Flood PM, Maixner W, Makarov SS, Diatchenko L. Signaling pathways mediating beta3-adrenergic receptor-induced production of interleukin-6 in adipocytes. Mol Immunol 2009; 46:2256-66. [PMID: 19477016 DOI: 10.1016/j.molimm.2009.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 04/03/2009] [Accepted: 04/07/2009] [Indexed: 12/25/2022]
Abstract
The beta(3)-adrenergic receptor (beta(3)AR) is an essential regulator of metabolic and endocrine functions. A major cellular and clinically significant consequence of beta(3)AR activation is the substantial elevation in interleukin-6 (IL-6) levels. Although the beta(3)AR-dependent regulation of IL-6 expression is well established, the cellular pathways underlying this regulation have not been characterized. Using a novel method of homogenous reporters, we assessed the pattern of activation of 43 transcription factors in response to the specific beta(3)AR agonist CL316243 in adipocytes, cells that exhibit the highest expression of beta(3)ARs. We observed a unique and robust activation of the CRE-response element, suggesting that IL-6 transcription is regulated via the G(s)-protein/cAMP/protein kinase A (PKA) but not nuclear factor kappa B (NF-kappaB) pathway. However, pretreatment of adipocytes with pharmacologic inhibitors of PKA pathway failed to block beta(3)AR-mediated IL-6 up-regulation. Additionally, stimulation of adipocytes with the exchange protein directly activated by cAMP (Epac) agonist did not induce IL-6 expression. Instead, the beta(3)AR-mediated transcription of IL-6 required activation of both the p38 and PKC pathways. Western blot analysis further showed that transcription factors CREB and ATF-2 but not ATF-1 were activated in a p38- and PKC-dependent manner. Collectively, our results suggest that while stimulation of the beta(3)AR leads to a specific activation of CRE-dependent transcription, there are several independent cellular pathways that converge at the level of CRE-response element activation, and in the case of IL-6 this activation is mediated by p38 and PKC but not PKA pathways.
Collapse
Affiliation(s)
- Inna E Tchivileva
- The Center for Neurosensory Disorders, School of Dentistry, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Brown and white adipose tissue have recently gained prominence as key players in obesity and related health problems, such as type-2 diabetes and cardiovascular disease. Brown adipose tissue-dependent nonshivering thermogenesis significantly affects the body's energy balance. Originally considered as a passive store of lipids, white adipose tissue has recently been found to secrete a number of hormones and cytokines and to be thus involved in the control of body metabolism and energy balance at multiple sites. These findings have renewed the interest in adipose organ biology, including its innervation by the autonomic nervous system and sensory nerves. Here, we describe our protocols for detecting different types of adipose tissue nerves by light microscopy using peroxidase immunostaining and by laser scanning confocal microscopy using immunofluorescence. With these techniques, the presence, distribution, and colocalization of autonomic and sensory nerves can be effectively investigated in subcutaneous and visceral adipose depots of normal and obese animals.
Collapse
Affiliation(s)
- Antonio Giordano
- Institute of Normal Human Morphology, School of Medicine, Marche Polytechnic University, Via Tronto, 10/A, 60020 Ancona, Italy
| | | | | |
Collapse
|
50
|
Ramsey MW, Behnke BJ, Prisby RD, Delp MD. Effects of aging on adipose resistance artery vasoconstriction: possible implications for orthostatic blood pressure regulation. J Appl Physiol (1985) 2007; 103:1636-43. [PMID: 17885023 DOI: 10.1152/japplphysiol.00637.2007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The purpose of this investigation was to determine mean arterial pressure (MAP) and regional vascular conductance responses in young and aged Fisher-344 rats during orthostatic stress, i.e., 70 degrees head-up tilt (HUT). Both groups demonstrated directionally different changes in MAP during HUT (young, 7% increase; aged, 7% decrease). Vascular conductance during HUT in young rats decreased in most tissues but largely remained unchanged in the aged animals. Based on the higher vascular conductance of white adipose tissue from aged rats during HUT, resistance arteries from white visceral fat were isolated and studied in vitro. There was diminished maximal vasoconstriction to phenylephrine and norepinephrine (NE: young, 42 +/- 5%; old, 18 +/- 6%) in adipose resistance arteries from aged rats. These results demonstrate that aging reduces the ability to maintain MAP during orthostatic stress, and this is associated with a diminished vasoconstriction of adipose resistance arteries.
Collapse
Affiliation(s)
- Michael W Ramsey
- Department of Kinesiology, Leisure, and Sport Sciences, East Tennessee State University, Johnson City, Tennessee, USA
| | | | | | | |
Collapse
|