1
|
Seidinger A, Roberts R, Bai Y, Müller M, Pfeil E, Matthey M, Rieck S, Alenfelder J, König GM, Pfeifer A, Kostenis E, Klinke A, Fleischmann BK, Wenzel D. Pharmacological Gq inhibition induces strong pulmonary vasorelaxation and reverses pulmonary hypertension. EMBO Mol Med 2024; 16:1930-1956. [PMID: 38977926 PMCID: PMC11319782 DOI: 10.1038/s44321-024-00096-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 07/10/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease with limited survival. Herein, we propose the pharmacological inhibition of Gq proteins as a novel concept to counteract pulmonary vasoconstriction and proliferation/migration of pulmonary artery smooth muscle cells (PASMCs) in PAH. We demonstrate that the specific pan-Gq inhibitor FR900359 (FR) induced a strong vasorelaxation in large and small pulmonary arteries in mouse, pig, and human subjects ex vivo. Vasorelaxation by FR proved at least as potent as the currently used triple therapy. We also provide in vivo evidence that local pulmonary application of FR prevented right ventricular systolic pressure increase in healthy mice as well as in mice suffering from hypoxia (Hx)-induced pulmonary hypertension (PH). In addition, we demonstrate that chronic application of FR prevented and also reversed Sugen (Su)Hx-induced PH in mice. We also demonstrate that Gq inhibition reduces proliferation and migration of PASMCs in vitro. Thus, our work illustrates a dominant role of Gq proteins for pulmonary vasoconstriction as well as remodeling and proposes direct Gq inhibition as a powerful pharmacological strategy in PH.
Collapse
Affiliation(s)
- Alexander Seidinger
- Institute of Physiology, Department of Systems Physiology, Medical Faculty, Ruhr University of Bochum, Bochum, Germany
| | - Richard Roberts
- Pharmacology Research Group, University Hospital of Nottingham, Nottingham, UK
| | - Yan Bai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Marion Müller
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
| | - Eva Pfeil
- Molecular-, Cellular-, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Institute of Physiology, Department of Systems Physiology, Medical Faculty, Ruhr University of Bochum, Bochum, Germany
| | - Sarah Rieck
- Institute of Physiology I, Life&Brain Center, Medical Faculty, University of Bonn, Bonn, Germany
| | - Judith Alenfelder
- Molecular-, Cellular-, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Gabriele M König
- Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Evi Kostenis
- Molecular-, Cellular-, and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Anna Klinke
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr University of Bochum, Bad Oeynhausen, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life&Brain Center, Medical Faculty, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Institute of Physiology, Department of Systems Physiology, Medical Faculty, Ruhr University of Bochum, Bochum, Germany.
- Institute of Physiology I, Life&Brain Center, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
2
|
Askin L, Abus S, Tanriverdi O. Resistin and Cardiovascular Disease: A Review of the Current Literature Regarding Clinical and Pathological Relationships. Curr Cardiol Rev 2022; 18:e290721195114. [PMID: 34325643 PMCID: PMC9241124 DOI: 10.2174/1573403x17666210729101120] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 11/22/2022] Open
Abstract
Serum resistin, mainly secreted by the bone marrow, monocytes, and macrophages, contributes to many processes, including endothelial dysfunction, Vascular Smooth Muscle Cell (VSMC) proliferation, and atherothrombosis demonstrating effects on the development of hypertension and Coronary Artery Disease (CAD). Previously published clinical studies have shown that plasma resistin levels are significantly associated with cardiovascular disease risk factors and adverse clinical outcomes associated with the condition. Resistin is associated with vascular smooth muscle cell dysfunction in vitro, most plausibly due to its relationship with oxidative stress in advanced atherosclerosis whereas in vivo studies have shown resistin to be associated with intimal hyperplasia. We aimed to summarize the role of resistin on cardiovascular disease (CVD), as we could not find any review focused on the role of resistin on CVD.
Collapse
Affiliation(s)
- Lutfu Askin
- Department of Cardiology, Adiyaman Education and Research Hospital, Adiyaman, Turkey
| | - Sabri Abus
- Department of cardiology, Kahta State Hospital, Kâhta, Turkey
| | - Okan Tanriverdi
- Department of Cardiology, Adiyaman Education and Research Hospital, Adiyaman, Turkey
| |
Collapse
|
3
|
Ceddia RP, Collins S. A compendium of G-protein-coupled receptors and cyclic nucleotide regulation of adipose tissue metabolism and energy expenditure. Clin Sci (Lond) 2020; 134:473-512. [PMID: 32149342 PMCID: PMC9137350 DOI: 10.1042/cs20190579] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022]
Abstract
With the ever-increasing burden of obesity and Type 2 diabetes, it is generally acknowledged that there remains a need for developing new therapeutics. One potential mechanism to combat obesity is to raise energy expenditure via increasing the amount of uncoupled respiration from the mitochondria-rich brown and beige adipocytes. With the recent appreciation of thermogenic adipocytes in humans, much effort is being made to elucidate the signaling pathways that regulate the browning of adipose tissue. In this review, we focus on the ligand-receptor signaling pathways that influence the cyclic nucleotides, cAMP and cGMP, in adipocytes. We chose to focus on G-protein-coupled receptor (GPCR), guanylyl cyclase and phosphodiesterase regulation of adipocytes because they are the targets of a large proportion of all currently available therapeutics. Furthermore, there is a large overlap in their signaling pathways, as signaling events that raise cAMP or cGMP generally increase adipocyte lipolysis and cause changes that are commonly referred to as browning: increasing mitochondrial biogenesis, uncoupling protein 1 (UCP1) expression and respiration.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| |
Collapse
|
4
|
Friebe A, Sandner P, Schmidtko A. cGMP: a unique 2nd messenger molecule - recent developments in cGMP research and development. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:287-302. [PMID: 31853617 PMCID: PMC7260148 DOI: 10.1007/s00210-019-01779-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022]
Abstract
Cyclic guanosine monophosphate (cGMP) is a unique second messenger molecule formed in different cell types and tissues. cGMP targets a variety of downstream effector molecules and, thus, elicits a very broad variety of cellular effects. Its production is triggered by stimulation of either soluble guanylyl cyclase (sGC) or particulate guanylyl cyclase (pGC); both enzymes exist in different isoforms. cGMP-induced effects are regulated by endogenous receptor ligands such as nitric oxide (NO) and natriuretic peptides (NPs). Depending on the distribution of sGC and pGC and the formation of ligands, this pathway regulates not only the cardiovascular system but also the kidney, lung, liver, and brain function; in addition, the cGMP pathway is involved in the pathogenesis of fibrosis, inflammation, or neurodegeneration and may also play a role in infectious diseases such as malaria. Moreover, new pharmacological approaches are being developed which target sGC- and pGC-dependent pathways for the treatment of various diseases. Therefore, it is of key interest to understand this pathway from scratch, beginning with the molecular basis of cGMP generation, the structure and function of both guanylyl cyclases and cGMP downstream targets; research efforts also focus on the subsequent signaling cascades, their potential crosstalk, and also the translational and, ultimately, the clinical implications of cGMP modulation. This review tries to summarize the contributions to the "9th International cGMP Conference on cGMP Generators, Effectors and Therapeutic Implications" held in Mainz in 2019. Presented data will be discussed and extended also in light of recent landmark findings and ongoing activities in the field of preclinical and clinical cGMP research.
Collapse
Affiliation(s)
- Andreas Friebe
- Institute of Physiology, University of Würzburg, Röntgenring 9, D-97070 Würzburg, Germany
| | - Peter Sandner
- Drug Discovery, Bayer AG, Aprather Weg 18a, D-42096 Wuppertal, Germany and Institute of Pharmacology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany
| |
Collapse
|
5
|
Benz PM, Ding Y, Stingl H, Loot AE, Zink J, Wittig I, Popp R, Fleming I. AKAP12 deficiency impairs VEGF-induced endothelial cell migration and sprouting. Acta Physiol (Oxf) 2020; 228:e13325. [PMID: 31162891 PMCID: PMC6916389 DOI: 10.1111/apha.13325] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022]
Abstract
Aim Protein kinase (PK) A anchoring protein (AKAP) 12 is a scaffolding protein that anchors PKA to compartmentalize cyclic AMP signalling. This study assessed the consequences of the downregulation or deletion of AKAP12 on endothelial cell migration and angiogenesis. Methods The consequences of siRNA‐mediated downregulation AKAP12 were studied in primary cultures of human endothelial cells as well as in endothelial cells and retinas from wild‐type versus AKAP12−/− mice. Molecular interactions were investigated using a combination of immunoprecipitation and mass spectrometry. Results AKAP12 was expressed at low levels in confluent endothelial cells but its expression was increased in actively migrating cells, where it localized to lamellipodia. In the postnatal retina, AKAP12 was expressed by actively migrating tip cells at the angiogenic front, and its deletion resulted in defective extension of the vascular plexus. In migrating endothelial cells, AKAP12 was co‐localized with the PKA type II‐α regulatory subunit as well as multiple key regulators of actin dynamics and actin filament‐based movement; including components of the Arp2/3 complex and the vasodilator‐stimulated phosphoprotein (VASP). Fitting with the evidence of a physical VASP/AKAP12/PKA complex, it was possible to demonstrate that the VEGF‐stimulated and PKA‐dependent phosphorylation of VASP was dependent on AKAP12. Indeed, AKAP12 colocalized with phospho‐Ser157 VASP at the leading edge of migrating endothelial cells. Conclusion The results suggest that compartmentalized AKAP12/PKA signalling mediates VASP phosphorylation at the leading edge of migrating endothelial cells to translate angiogenic stimuli into altered actin dynamics and cell movement.
Collapse
Affiliation(s)
- Peter M. Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine Goethe University Frankfurt am Main Germany
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain Frankfurt am Main Germany
| | - Yindi Ding
- Institute for Vascular Signalling, Centre for Molecular Medicine Goethe University Frankfurt am Main Germany
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain Frankfurt am Main Germany
| | - Heike Stingl
- Institute for Vascular Signalling, Centre for Molecular Medicine Goethe University Frankfurt am Main Germany
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain Frankfurt am Main Germany
| | - Annemarieke E. Loot
- Institute for Vascular Signalling, Centre for Molecular Medicine Goethe University Frankfurt am Main Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine Goethe University Frankfurt am Main Germany
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain Frankfurt am Main Germany
| | - Ilka Wittig
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain Frankfurt am Main Germany
- Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine Goethe University Frankfurt am Main Germany
| | - Rüdiger Popp
- Institute for Vascular Signalling, Centre for Molecular Medicine Goethe University Frankfurt am Main Germany
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain Frankfurt am Main Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine Goethe University Frankfurt am Main Germany
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain Frankfurt am Main Germany
| |
Collapse
|
6
|
Klepac K, Yang J, Hildebrand S, Pfeifer A. RGS2: A multifunctional signaling hub that balances brown adipose tissue function and differentiation. Mol Metab 2019; 30:173-183. [PMID: 31767169 PMCID: PMC6807268 DOI: 10.1016/j.molmet.2019.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/28/2019] [Indexed: 12/28/2022] Open
Abstract
Objective Recruitment of brown adipose tissue (BAT) is a potential new strategy for increasing energy expenditure (EE) to treat obesity. G protein–coupled receptors (GPCRs) represent promising targets to activate BAT, as they are the major regulators of BAT biological function. To identify new regulators of GPCR signaling in BAT, we studied the role of Regulator of G protein Signaling 2 (RGS2) in brown adipocytes and BAT. Methods We combined pharmacological and genetic tools to investigate the role of RGS2 in BAT in vitro and in vivo. Adipocyte progenitors were isolated from wild-type (WT) and RGS2 knockout (RGS2−/−) BAT and differentiated to brown adipocytes. This approach was complemented with knockdown of RGS2 using lentiviral shRNAs (shRGS2). Adipogenesis was analyzed by Oil Red O staining and by determining the expression of adipogenic and thermogenic markers. Pharmacological modulators and fluorescence staining of F-acting stress fibers were employed to identify the underlying signaling pathways. In vivo, the activity of BAT was assessed by ex vivo lipolysis and by measuring whole-body EE by indirect calorimetry in metabolic cages. Results RGS2 is highly expressed in BAT, and treatment with cGMP—an important enhancer of brown adipocyte differentiation—further increased RGS2 expression. Loss of RGS2 strongly suppressed adipogenesis and the expression of thermogenic genes in brown adipocytes. Mechanistically, we found increased Gq/Rho/Rho kinase (ROCK) signaling in the absence of RGS2. Surprisingly, in vivo analysis revealed elevated BAT activity in RGS2-deficient mice that was caused by enhanced Gs/cAMP signaling. Conclusion Overall, RGS2 regulates two major signaling pathways in BAT: Gq and Gs. On the one hand, RGS2 promotes brown adipogenesis by counteracting the inhibitory action of Gq/Rho/ROCK signaling. On the other hand, RGS2 decreases the activity of BAT through the inhibition of Gs signaling and cAMP production. Thus, RGS2 might represent a stress modulator that protects BAT from overstimulation. RGS2 regulates brown adipose tissue (BAT) by inhibiting two major G protein-coupled receptor (GPCR) pathways – Gq and Gs. Deletion of RGS2 impairs the differentiation of murine brown adipocytes due to elevated Gq/Rho/ROCK signaling. In vivo, RGS2 knock-out mice show an increase in BAT lipolysis and whole-body energy expenditure.
Collapse
Affiliation(s)
- Katarina Klepac
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany.
| | - JuHee Yang
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany
| | - Staffan Hildebrand
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany; PharmaCenter, University of Bonn, 53127 Bonn, Germany.
| |
Collapse
|
7
|
Reverte-Salisa L, Sanyal A, Pfeifer A. Role of cAMP and cGMP Signaling in Brown Fat. Handb Exp Pharmacol 2019; 251:161-182. [PMID: 29633180 DOI: 10.1007/164_2018_117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cold-induced activation of brown adipose tissue (BAT) is mediated by norepinephrine and adenosine that are released during sympathetic nerve activation. Both signaling molecules induce an increase in intracellular levels of 3',5'-cyclic adenosine monophosphate (cAMP) in murine and human BAT. In brown adipocytes, cAMP plays a central role, because it activates lipolysis, glucose uptake, and thermogenesis. Another well-studied intracellular second messenger is 3',5'-cyclic guanosine monophosphate (cGMP), which closely resembles cAMP. Several studies have shown that intact cGMP signaling is essential for normal adipogenic differentiation and BAT-mediated thermogenesis in mice. This chapter highlights recent observations, demonstrating the physiological significance of cyclic nucleotide signaling in BAT as well as their potential to induce browning of white adipose tissue (WAT) in mice and humans.
Collapse
Affiliation(s)
- Laia Reverte-Salisa
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Abhishek Sanyal
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
8
|
Fan L, Xu H, Yang R, Zang Y, Chen J, Qin H. Combination of Capsaicin and Capsiate Induces Browning in 3T3-L1 White Adipocytes via Activation of the Peroxisome Proliferator-Activated Receptor γ/β 3-Adrenergic Receptor Signaling Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:6232-6240. [PMID: 31075194 DOI: 10.1021/acs.jafc.9b02191] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This study investigated the effects and molecular mechanism of a combination of capsaicin and capsiate on promoting lipid metabolism and inducing browning in 3T3-L1 white adipocytes. The combination significantly suppressed lipid accumulation in adipocytes ( p = 0.019) and robustly improved lipid metabolic profiles, including decreased triacylglycerol (0.6703 ± 0.0385 versus 0.2849 ± 0.0188 mmol/g of protein; p < 0.001), total cholesterol (0.1282 ± 0.0241 versus 0.0651 ± 0.0178 mmol/g of protein; p = 0.003), and low-density lipoprotein cholesterol (0.0021 ± 0.0017 versus 0.0005 ± 0.0002 mmol/g of protein; p = 0.024) and increased high-density lipoprotein cholesterol (0.0162 ± 0.0141 versus 0.1002 ± 0.0167 mmol/g of protein; p = 0.012). Furthermore, this combination markedly upgraded the protein levels of cluster of differentiation 36 ( p = 0.007) and adipose triglyceride lipase ( p = 0.013) and phosphorylation of hormone-sensitive lipase at Ser660, Ser565, and Ser563 ( p < 0.001, p = 0.027, and p = 0.002, respectively), indicating increases of fatty acid transport and lipolysis. The levels of lipid metabolism regulators, phosphorylation of adenosine-monophosphate-activated protein kinases α and β ( p = 0.011, and p < 0.001, respectively), sirtuin 1 ( p = 0.004), and vanilloid transient receptor subtype I ( p = 0.014) were also increased by the combination. Moreover, the combination greatly activated the browning program in adipocytes, as demonstrated by increases in beige-specific gene and protein. Further research found that the protein levels of peroxisome proliferator-activated receptor γ (PPARγ; p = 0.001) and β3-adrenergic receptor (β3-AR; p = 0.026) were elevated by the combination, and most of the beige-specific markers were abolished by pretreatment of antagonists of PPARγ or β3-AR. In conclusion, these results indicated that a combination of capsaicin and capsiate could induce browning in white adipocytes via activation of the PPARγ/β3-AR signaling pathway, and this combination might be worth investigating as a potential cure for obesity.
Collapse
Affiliation(s)
- Li Fan
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health , Central South University , 110 Xiangya Road , Changsha , Hunan 410078 , People's Republic of China
| | - Haiyan Xu
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health , Central South University , 110 Xiangya Road , Changsha , Hunan 410078 , People's Republic of China
| | - Rengui Yang
- Changsha Center for Disease Control and Prevention , Changsha , Hunan 410004 , People's Republic of China
| | - Yufan Zang
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health , Central South University , 110 Xiangya Road , Changsha , Hunan 410078 , People's Republic of China
| | - Jingfang Chen
- Changsha Center for Disease Control and Prevention , Changsha , Hunan 410004 , People's Republic of China
| | - Hong Qin
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health , Central South University , 110 Xiangya Road , Changsha , Hunan 410078 , People's Republic of China
| |
Collapse
|
9
|
Wang J, Jia Y, Wang L, Li D, Wang L, Zhu Y, Liu J, Gong J. Vasodilator-Stimulated Phosphoprotein: Regulators of Adipokines Resistin and Phenotype Conversion of Epicardial Adipocytes. Med Sci Monit 2018; 24:6010-6020. [PMID: 30156215 PMCID: PMC6126413 DOI: 10.12659/msm.908111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background Endothelial dysfunction plays a central part in the pathogenesis of coronary atherosclerosis. The adipokine resistin is one of the key players in endothelial cell dysfunction. In addition, the role of epicardial fat in coronary artery endothelial dysfunction is also emphasized. We investigated whether vasodilator-stimulated phosphoprotein (VASP) is involved in resistin-related endothelial dysfunction and the phenotype conversion of epicardial adipocytes. Material/Methods Cell proliferation and migration were evaluated by MTT and Transwell chamber assay, respectively. Next, we took epicardial fat samples from patients with valvular heart disease and non-coronary artery disease. Gene expression was determined by reverse transcription-quantitative polymerase chain reaction and relative abundance of the protein by Western blotting. Results Resistin induced endothelial proliferation and migration in a dose-dependent manner. Both resistin-induced cell proliferation and migration were effectively blocked by ablation of VASP. The brown adipose tissue-specific genes for uncoupling protein 1 (UCP-1) and PR-domain-missing16 (PRDM16) decreased, but the white adipose tissue-specific genes for resistin and RIP140 increased in VASP-deficient adipocytes compared with the LV-sicntr group. However, disruption of the Ras homolog gene family member A (RhoA) /Rho-associated kinase (ROCK) in VASP-deficient adipocytes with specific inhibitors inverted the adipocyte phenotype existing in VASP-deficient adipocytes. Furthermore, the expressions of proinflammatory cytokines interleukin-6 (IL-6), interleukin-8 (IL-8), and monocyte chemoattractantprotein-1 (MCP-1) in VASP-deficient adipocytes were markedly upregulated compared with the LV-sicntr group. Conclusions These results suggest a physiological role for VASP in coronary atherosclerosis through regulating adipokine resistin and phenotype conversion of epicardial adipose tissue.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Yan Jia
- Department of Cardiology, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Lijun Wang
- Department of Cardiology, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Demin Li
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Lei Wang
- Department of Cardiology, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Ying Zhu
- Department of Cardiology, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Jing Liu
- Department of Cardiology, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Jianbin Gong
- Department of Cardiology, Jinling Hospital, School of Clinical Medicine, Nanjing University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
10
|
Baranski TJ, Kraja AT, Fink JL, Feitosa M, Lenzini PA, Borecki IB, Liu CT, Cupples LA, North KE, Province MA. A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses. PLoS Genet 2018; 14:e1007222. [PMID: 29608557 PMCID: PMC5897035 DOI: 10.1371/journal.pgen.1007222] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 04/12/2018] [Accepted: 01/25/2018] [Indexed: 12/30/2022] Open
Abstract
Human GWAS of obesity have been successful in identifying loci associated with adiposity, but for the most part, these are non-coding SNPs whose function, or even whose gene of action, is unknown. To help identify the genes on which these human BMI loci may be operating, we conducted a high throughput screen in Drosophila melanogaster. Starting with 78 BMI loci from two recently published GWAS meta-analyses, we identified fly orthologs of all nearby genes (± 250KB). We crossed RNAi knockdown lines of each gene with flies containing tissue-specific drivers to knock down (KD) the expression of the genes only in the brain and the fat body. We then raised the flies on a control diet and compared the amount of fat/triglyceride in the tissue-specific KD group compared to the driver-only control flies. 16 of the 78 BMI GWAS loci could not be screened with this approach, as no gene in the 500-kb region had a fly ortholog. Of the remaining 62 GWAS loci testable in the fly, we found a significant fat phenotype in the KD flies for at least one gene for 26 loci (42%) even after correcting for multiple comparisons. By contrast, the rate of significant fat phenotypes in RNAi KD found in a recent genome-wide Drosophila screen (Pospisilik et al. (2010) is ~5%. More interestingly, for 10 of the 26 positive regions, we found that the nearest gene was not the one that showed a significant phenotype in the fly. Specifically, our screen suggests that for the 10 human BMI SNPs rs11057405, rs205262, rs9925964, rs9914578, rs2287019, rs11688816, rs13107325, rs7164727, rs17724992, and rs299412, the functional genes may NOT be the nearest ones (CLIP1, C6orf106, KAT8, SMG6, QPCTL, EHBP1, SLC39A8, ADPGK /ADPGK-AS1, PGPEP1, KCTD15, respectively), but instead, the specific nearby cis genes are the functional target (namely: ZCCHC8, VPS33A, RSRC2; SPDEF, NUDT3; PAGR1; SETD1, VKORC1; SGSM2, SRR; VASP, SIX5; OTX1; BANK1; ARIH1; ELL; CHST8, respectively). The study also suggests further functional experiments to elucidate mechanism of action for genes evolutionarily conserved for fat storage. Human Genome Wide Association Studies have successfully found thousands of novel genetic variants associated with many diseases. While these undoubtedly point to new biology, the field has been slowed in exploiting these new findings to reach a better understanding of exactly how they confer increased risk. Many, if not most, appear to be regulatory not coding variants, so their immediate consequence is not obvious. A real rate limiting step is even identifying which gene these variants might be regulating, and in what tissues they are operating to increase disease risk. In the absence of any other information, a first order assumption is that they may be more likely to be regulating a nearby gene, and such variants are often initially annotated by the “nearest” gene until their function is more definitively validated. Exploiting the idea that many genes may have conserved function across species, we conducted a high-throughput screen of fruit-fly orthologs of human genes nearby 78 well validated GWAS variants for human obesity, in order to more precisely identify the gene(s) of action. We systematically knocked down the function of each of these nearby genes in the brain and fat-body of the flies, raised them on a standard diet, and compared their percent body fat with control flies, in order to validate which genes showed a fat response. 43% of the time when fly orthologs existed in the region, we were able to identify the causal gene. Interestingly, nearly half the time (46%), it was not the nearest gene but another nearby one that regulated fat.
Collapse
Affiliation(s)
- Thomas J. Baranski
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (TJB); (MAP)
| | - Aldi T. Kraja
- Department of Genetics and Center for Genome Sciences and Systems Biology, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jill L. Fink
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mary Feitosa
- Department of Genetics and Center for Genome Sciences and Systems Biology, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Petra A. Lenzini
- Department of Genetics and Center for Genome Sciences and Systems Biology, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ingrid B. Borecki
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Michael A. Province
- Department of Genetics and Center for Genome Sciences and Systems Biology, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (TJB); (MAP)
| |
Collapse
|
11
|
Laban H, Weigert A, Zink J, Elgheznawy A, Schürmann C, Günther L, Abdel Malik R, Bothur S, Wingert S, Bremer R, Rieger MA, Brüne B, Brandes RP, Fleming I, Benz PM. VASP regulates leukocyte infiltration, polarization, and vascular repair after ischemia. J Cell Biol 2018; 217:1503-1519. [PMID: 29507126 PMCID: PMC5881493 DOI: 10.1083/jcb.201702048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/06/2017] [Accepted: 01/26/2018] [Indexed: 01/14/2023] Open
Abstract
In ischemic vascular diseases, leukocyte recruitment and polarization are crucial for revascularization and tissue repair. The study of Laban et al. provides evidence that VASP is a major regulator of leukocyte recruitment and polarization and vascular repair after ischemia. Mechanistically, the study supports a novel role of VASP in chemokine receptor trafficking. In ischemic vascular diseases, leukocyte recruitment and polarization are crucial for revascularization and tissue repair. We investigated the role of vasodilator-stimulated phosphoprotein (VASP) in vascular repair. After hindlimb ischemia induction, blood flow recovery, angiogenesis, arteriogenesis, and leukocyte infiltration into ischemic muscles in VASP−/− mice were accelerated. VASP deficiency also elevated the polarization of the macrophages through increased signal transducer and activator of transcription (STAT) signaling, which augmented the release of chemokines, cytokines, and growth factors to promote leukocyte recruitment and vascular repair. Importantly, VASP deletion in bone marrow–derived cells was sufficient to mimic the increased blood flow recovery of global VASP−/− mice. In chemotaxis experiments, VASP−/− neutrophils/monocytes were significantly more responsive to M1-related chemokines than wild-type controls. Mechanistically, VASP formed complexes with the chemokine receptor CCR2 and β-arrestin-2, and CCR2 receptor internalization was significantly reduced in VASP−/− leukocytes. Our data indicate that VASP is a major regulator of leukocyte recruitment and polarization in postischemic revascularization and support a novel role of VASP in chemokine receptor trafficking.
Collapse
Affiliation(s)
- Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Amro Elgheznawy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Christoph Schürmann
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Lea Günther
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Randa Abdel Malik
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Sabrina Bothur
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Susanne Wingert
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Rolf Bremer
- HBB Datenkommunikation and Abrechnungssysteme, Hannover, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy and Department for Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ralf P Brandes
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Peter M Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany .,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|
12
|
Zhu E, Yang Y, Zhang J, Li Y, Li C, Chen L, Sun B. Liraglutide suppresses obesity and induces brown fat-like phenotype via Soluble Guanylyl Cyclase mediated pathway in vivo and in vitro. Oncotarget 2018; 7:81077-81089. [PMID: 27835589 PMCID: PMC5348377 DOI: 10.18632/oncotarget.13189] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/28/2016] [Indexed: 12/20/2022] Open
Abstract
Strategies for driving white adipose tissue (WAT) to acquire brown-like characteristics are a promising approach to reduce obesity. Liraglutide has been reported to active brown adipose tissue (BAT) thermogenesis and WAT browning by rapid intracerebroventricular injection in mice. In this study, we investigated the effects and possible mechanisms of liraglutide on WAT browning by chronic treatment. Here, we show that liraglutide significantly decreases body weight of mice and reduces the size of white adipocytes. By quantity polymerase chain reaction, immunoblotting analysis, cell immunofluorescence or immunocytochemical staining, we found liraglutide induced WAT browning because it up-regulated lipolytic activity, BAT, as well as mitochondrial marker genes in inguinal and peripheral renal WAT. We also confirmed liraglutide induced browning of 3T3-L1 because it enhanced expression of BAT and mitochondrial specific genes. In further, we observed that, soluble guanylyl cyclase (sGC) and protein kinase G I (PKGI) were up-regulated by liraglutide in vivo and in vitro; stimulation of sGC elevated expression of BAT markers and PKGI, which suggested that liraglutide induced WAT browning via sGC-dependent pathway. Taken together, this study expands our knowledge on the mechanism of liraglutide inducing WAT browning, and provides a theoretical support for clinical usage of liraglutide on obesity treatment.
Collapse
Affiliation(s)
- Endong Zhu
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 300070 Tianjin, China
| | - Yang Yang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 300070 Tianjin, China
| | - Juanjuan Zhang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 300070 Tianjin, China
| | - Yongmei Li
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 300070 Tianjin, China.,Department of Human Anatomy and Histology, Tianjin Medical University, 300070 Tianjin, China
| | - Chunjun Li
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 300070 Tianjin, China
| | - Liming Chen
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 300070 Tianjin, China
| | - Bei Sun
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 300070 Tianjin, China
| |
Collapse
|
13
|
Bae CR, Hino J, Hosoda H, Son C, Makino H, Tokudome T, Tomita T, Hosoda K, Miyazato M, Kangawa K. Adipocyte-specific expression of C-type natriuretic peptide suppresses lipid metabolism and adipocyte hypertrophy in adipose tissues in mice fed high-fat diet. Sci Rep 2018; 8:2093. [PMID: 29391544 PMCID: PMC5794866 DOI: 10.1038/s41598-018-20469-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/19/2018] [Indexed: 02/08/2023] Open
Abstract
C-type natriuretic peptide (CNP) is expressed in diverse tissues, including adipose and endothelium, and exerts its effects by binding to and activating its receptor, guanylyl cyclase B. Natriuretic peptides regulate intracellular cGMP and phosphorylated vasodilator-stimulated phosphoprotein (VASP). We recently revealed that overexpression of CNP in endothelial cells protects against high-fat diet (HFD)-induced obesity in mice. Given that endothelial CNP affects adipose tissue during obesity, CNP in adipocytes might directly regulate adipocyte function during obesity. Therefore, to elucidate the effect of CNP in adipocytes, we assessed 3T3-L1 adipocytes and transgenic (Tg) mice that overexpressed CNP specifically in adipocytes (A-CNP). We found that CNP activates the cGMP–VASP pathway in 3T3-L1 adipocytes. Compared with Wt mice, A-CNP Tg mice showed decreases in fat weight and adipocyte hypertrophy and increases in fatty acid β-oxidation, lipolysis-related gene expression, and energy expenditure during HFD-induced obesity. These effects led to decreased levels of the macrophage marker F4/80 in the mesenteric fat pad and reduced inflammation. Furthermore, A-CNP Tg mice showed improved glucose tolerance and insulin sensitivity, which were associated with enhanced insulin-stimulated Akt phosphorylation. Our results suggest that CNP overexpression in adipocytes protects against adipocyte hypertrophy, excess lipid metabolism, inflammation, and decreased insulin sensitivity during HFD-induced obesity.
Collapse
Affiliation(s)
- Cho-Rong Bae
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | - Hiroshi Hosoda
- Departments of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Cheol Son
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Omics Research Center and National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Hisashi Makino
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Takeshi Tokudome
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Tsutomu Tomita
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Biobank, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Kiminori Hosoda
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| |
Collapse
|
14
|
Li S, Li Y, Xiang L, Dong J, Liu M, Xiang G. Sildenafil induces browning of subcutaneous white adipose tissue in overweight adults. Metabolism 2018; 78:106-117. [PMID: 28986166 DOI: 10.1016/j.metabol.2017.09.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 08/24/2017] [Accepted: 09/19/2017] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To investigate that short-term treatment of sildenafil can induce browning of subcutaneous white adipose tissue (sWAT) in human adults. DESIGN A randomized, double-blinded, placebo-controlled, parallel group trial. METHODS Sixteen eligibility overweight male subjects were recruited, comparing 100mg/day sildenafil versus an identical placebo therapy for 7days. sWAT samples were collected from subjects before and after 7-day sildenafil or placebo interventions. RESULTS The results showed that multilocular UCP1-positive adipocytes existed in sWAT samples from subjects after sildenafil treatment. Compared to before treatment in both group as well as after treatment in placebo, sildenafil significantly decreased adipocyte size, increased the expressions of UCP1 protein and mRNA, mitochondrial density, and leak respiratory capacity in sWAT (p<0.05). Sildenafil also increased plasma cyclic guanosine-3',5'-monophosphate (cGMP) and catecholamine concentrations (p<0.05), and consequently activated the expressions of vasodilator-stimulated phosphoprotein (VASP) and p70 ribosomal S6 kinase 1 (S6K1) (p<0.05). Sildenafil did not activate typical brown fat. CONCLUSIONS The current findings demonstrate that sildenafil can induce browning of sWAT in human, and this action may be through cGMP-dependent protein kinase I and mechanistic/mammalian target of rapamycin (mTOR) signaling pathways. Sldenafil may be a promising treatment for metabolic disease.
Collapse
Affiliation(s)
- Shuguang Li
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Lin Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Jing Dong
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Min Liu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Guangda Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| |
Collapse
|
15
|
Interplay between Obesity-Induced Inflammation and cGMP Signaling in White Adipose Tissue. Cell Rep 2017; 18:225-236. [PMID: 28052251 DOI: 10.1016/j.celrep.2016.12.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/15/2016] [Accepted: 12/08/2016] [Indexed: 11/21/2022] Open
Abstract
Current worldwide figures suggest that obesity is pandemic. Understanding the underlying molecular mechanisms would help develop viable anti-obesity therapies. Here, we assess the influence of obesity-induced inflammation on white adipocyte cyclic guanosine monophosphate (cGMP) signaling, which is beneficial for adipocyte differentiation and thermogenesis. We find that murine gonadal and not inguinal fat is prone to obesity-induced inflammation. Correspondingly, the cGMP cascade is dysregulated in gonadal but not in inguinal fat of obese mice. Analysis of two independent human cohorts reveals a defective cGMP pathway only in visceral fat of obese subjects. Congruently, cGMP signaling is dysregulated in tumor necrosis factor α (TNF-α)-treated primary white adipocytes. TNF-α-mediated suppression of sGCβ1 is mediated via NF-κB, whereas PKG is repressed by JNK signaling. Additionally, TNF-α-activated JNK signaling suppresses PPARγ and aP2. Taken together, the intensity of obesity-induced inflammation dictates the amplitude of cGMP signaling dysregulation in white adipocytes through distinct pathways.
Collapse
|
16
|
Premenopausal Obesity and Breast Cancer Growth Rates in a Rodent Model. Nutrients 2016; 8:214. [PMID: 27077880 PMCID: PMC4848683 DOI: 10.3390/nu8040214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/20/2022] Open
Abstract
Obese premenopausal women with breast cancer have poorer prognosis for long term survival, in part because their tumors are larger at the time of diagnosis than are found in normal weight women. Whether larger tumor mass is due to obesity-related barriers to detection or to effects on tumor biology is not known. This study used polygenic models for obesity and breast cancer to deconstruct this question with the objective of determining whether cell autonomous mechanisms contribute to the link between obesity and breast cancer burden. Assessment of the growth rates of 259 chemically induced mammary carcinomas from rats sensitive to dietary induced obesity (DS) and of 143 carcinomas from rats resistant (DR) to dietary induced obesity revealed that tumors in DS rats grew 1.8 times faster than in DR rats. This difference may be attributed to alterations in cell cycle machinery that permit more rapid tumor cell accumulation. DS tumors displayed protein expression patterns consistent with reduced G1/S checkpoint inhibition and a higher threshold of factors required for execution of the apoptotic cell death pathway. These mechanistic insights identify regulatory targets for life style modifications or pharmacological interventions designed to disrupt the linkage between obesity and tumor burden.
Collapse
|
17
|
The Gq signalling pathway inhibits brown and beige adipose tissue. Nat Commun 2016; 7:10895. [PMID: 26955961 PMCID: PMC4786868 DOI: 10.1038/ncomms10895] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/01/2016] [Indexed: 12/31/2022] Open
Abstract
Brown adipose tissue (BAT) dissipates nutritional energy as heat via the uncoupling protein-1 (UCP1) and BAT activity correlates with leanness in human adults. Here we profile G protein-coupled receptors (GPCRs) in brown adipocytes to identify druggable regulators of BAT. Twenty-one per cent of the GPCRs link to the Gq family, and inhibition of Gq signalling enhances differentiation of human and murine brown adipocytes. In contrast, activation of Gq signalling abrogates brown adipogenesis. We further identify the endothelin/Ednra pathway as an autocrine activator of Gq signalling in brown adipocytes. Expression of a constitutively active Gq protein in mice reduces UCP1 expression in BAT, whole-body energy expenditure and the number of brown-like/beige cells in white adipose tissue (WAT). Furthermore, expression of Gq in human WAT inversely correlates with UCP1 expression. Thus, our data indicate that Gq signalling regulates brown/beige adipocytes and inhibition of Gq signalling may be a novel therapeutic approach to combat obesity.
Collapse
|
18
|
A novel crosstalk between Alk7 and cGMP signaling differentially regulates brown adipocyte function. Mol Metab 2015; 4:576-83. [PMID: 26266090 PMCID: PMC4529496 DOI: 10.1016/j.molmet.2015.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 05/31/2015] [Accepted: 06/05/2015] [Indexed: 12/18/2022] Open
Abstract
Objective Obesity is an enormous burden for patients and health systems world-wide. Brown adipose tissue dissipates energy in response to cold and has been shown to be metabolically active in human adults. The type I transforming growth factor β (TGFβ) receptor Activin receptor-like kinase 7 (Alk7) is highly expressed in adipose tissues and is down-regulated in obese patients. Here, we studied the function of Alk7 in brown adipocytes. Methods Using pharmacological and genetic tools, Alk7 signaling pathway and its effects were studied in murine brown adipocytes. Brown adipocyte differentiation and activation was analyzed. Results Alk7 is highly upregulated during differentiation of brown adipocytes. Interestingly, Alk7 expression is increased by cGMP/protein kinase G (PKG) signaling, which enhances brown adipocyte differentiation. Activin AB effectively activates Alk7 and SMAD3 signaling. Activation of Alk7 in brown preadipocytes suppresses the master adipogenic transcription factor PPARγ and differentiation. Stimulation of Alk7 during late differentiation of brown adipocytes reduces lipid content and adipogenic marker expression but enhances UCP1 expression. Conclusions We found a so far unknown crosstalk between cGMP and Alk7 signaling pathways. Tight regulation of Alk7 is required for efficient differentiation of brown adipocytes. Alk7 has differential effects on adipogenic differentiation and the development of the thermogenic program in brown adipocytes. Alk7 expression in brown adipocytes is modulated by the cGMP/PKGI signaling pathway. Brown adipocyte differentiation requires tight regulation of Alk7 activation. Alk7 signaling via SMAD3 reduces PPARγ and attenuates adipogenic differentiation. Alk7 enhances UCP1 expression in mature brown adipocytes. We found a novel crosstalk between the Alk7 and cGMP signaling pathway.
Collapse
|
19
|
Hoffmann LS, Etzrodt J, Willkomm L, Sanyal A, Scheja L, Fischer AWC, Stasch JP, Bloch W, Friebe A, Heeren J, Pfeifer A. Stimulation of soluble guanylyl cyclase protects against obesity by recruiting brown adipose tissue. Nat Commun 2015; 6:7235. [PMID: 26011238 PMCID: PMC4455111 DOI: 10.1038/ncomms8235] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/21/2015] [Indexed: 12/31/2022] Open
Abstract
Obesity is characterized by a positive energy balance and expansion of white adipose tissue (WAT). In contrast, brown adipose tissue (BAT) combusts energy to produce heat. Here we show that a small molecule stimulator (BAY 41-8543) of soluble guanylyl cyclase (sGC), which produces the second messenger cyclic GMP (cGMP), protects against diet-induced weight gain, induces weight loss in established obesity, and also improves the diabetic phenotype. Mechanistically, the haeme-dependent sGC stimulator BAY 41-8543 enhances lipid uptake into BAT and increases whole-body energy expenditure, whereas ablation of the haeme-containing β1-subunit of sGC severely impairs BAT function. Notably, the sGC stimulator enhances differentiation of human brown adipocytes as well as induces 'browning' of primary white adipocytes. Taken together, our data suggest that sGC is a potential pharmacological target for the treatment of obesity and its comorbidities.
Collapse
Affiliation(s)
- Linda S Hoffmann
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany
| | - Jennifer Etzrodt
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany
| | - Lena Willkomm
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Institute of Cardiovascular Research and Sports Medicine, Cologne D-50735, Germany
| | - Abhishek Sanyal
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany.,Research Training Group 1873, University of Bonn, Bonn D-53127, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Alexander W C Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Johannes-Peter Stasch
- Bayer Pharma AG, Wuppertal D-42113, Germany.,Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle an der Saale D-06120, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Institute of Cardiovascular Research and Sports Medicine, Cologne D-50735, Germany
| | - Andreas Friebe
- Institute of Physiology, Martin Luther University Würzburg, Würzburg D-97070, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany.,Research Training Group 1873, University of Bonn, Bonn D-53127, Germany.,PharmaCenter, University of Bonn, Bonn D-53119, Germany
| |
Collapse
|
20
|
Pan J, Yang Y, Yang B, Yu Y. Artificial polychromatic light affects growth and physiology in chicks. PLoS One 2014; 9:e113595. [PMID: 25469877 PMCID: PMC4254831 DOI: 10.1371/journal.pone.0113595] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/29/2014] [Indexed: 11/18/2022] Open
Abstract
Despite the overwhelming use of artificial light on captive animals, its effect on those animals has rarely been studied experimentally. Housing animals in controlled light conditions is useful for assessing the effects of light. The chicken is one of the best-studied animals in artificial light experiments, and here, we evaluate the effect of polychromatic light with various green and blue components on the growth and physiology in chicks. The results indicate that green-blue dual light has two side-effects on chick body mass, depending on the various green to blue ratios. Green-blue dual light with depleted and medium blue component decreased body mass, whereas enriched blue component promoted body mass in chicks compared with monochromatic green- or blue spectra-treated chicks. Moreover, progressive changes in the green to blue ratios of green-blue dual light could give rise to consistent progressive changes in body mass, as suggested by polychromatic light with higher blue component resulting in higher body mass. Correlation analysis confirmed that food intake was positively correlated with final body mass in chicks (R2 = 0.7664, P = 0.0001), suggesting that increased food intake contributed to the increased body mass in chicks exposed to higher blue component. We also found that chicks exposed to higher blue component exhibited higher blood glucose levels. Furthermore, the glucose level was positively related to the final body mass (R2 = 0.6406, P = 0.0001) and food intake (R2 = 0.784, P = 0.0001). These results demonstrate that spectral composition plays a crucial role in affecting growth and physiology in chicks. Moreover, consistent changes in spectral components might cause the synchronous response of growth and physiology.
Collapse
Affiliation(s)
- Jinming Pan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Yefeng Yang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Yonghua Yu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- * E-mail:
| |
Collapse
|
21
|
Leiss V, Illison J, Domes K, Hofmann F, Lukowski R. Expression of cGMP-dependent protein kinase type I in mature white adipocytes. Biochem Biophys Res Commun 2014; 452:151-6. [DOI: 10.1016/j.bbrc.2014.08.071] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 08/15/2014] [Indexed: 01/01/2023]
|
22
|
Pfeifer A, Hoffmann LS. Brown, beige, and white: the new color code of fat and its pharmacological implications. Annu Rev Pharmacol Toxicol 2014; 55:207-27. [PMID: 25149919 DOI: 10.1146/annurev-pharmtox-010814-124346] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Brown adipose tissue (BAT) was previously regarded as a special type of fat relevant only for defending hibernating animals and newborns against a cold environment. Recently, BAT has received considerable attention following its (re)discovery in humans. Using glucose tracers, multiple laboratories independently found metabolically active BAT in adults. The enormous metabolic powers of BAT in animal models could make it an attractive target for antiobesity therapies in humans. Here, we review the present knowledge on the role of BAT in energy homeostasis and metabolism, focusing on signaling pathways and potential targets for novel therapeutics. We also shine light on ongoing debates, including those about the true color of brown fat in adults, as well as on the requirements for translation of basic research on BAT into clinical medicine.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, 53105 Bonn, Germany;
| | | |
Collapse
|
23
|
André G, Sandoval JE, Retailleau K, Loufrani L, Toumaniantz G, Offermanns S, Rolli-Derkinderen M, Loirand G, Sauzeau V. Smooth muscle specific Rac1 deficiency induces hypertension by preventing p116RIP3-dependent RhoA inhibition. J Am Heart Assoc 2014; 3:e000852. [PMID: 24938713 PMCID: PMC4309079 DOI: 10.1161/jaha.114.000852] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Increasing evidence implicates overactivation of RhoA as a critical component of the pathogenesis of hypertension. Although a substantial body of work has established that Rac1 functions antagonize RhoA in a broad range of physiological processes, the role of Rac1 in the regulation of vascular tone and blood pressure is not fully elucidated. Methods and Results To define the role of Rac1 in vivo in vascular smooth muscle cells (vSMC), we generated smooth muscle (SM)‐specific Rac1 knockout mice (SM‐Rac1‐KO) and performed radiotelemetric blood pressure recordings, contraction measurements in arterial rings, vSMC cultures and biochemical analyses. SM‐Rac1‐KO mice develop high systolic blood pressure sensitive to Rho kinase inhibition by fasudil. Arteries from SM‐Rac1‐KO mice are characterized by a defective NO‐dependent vasodilation and an overactivation of RhoA/Rho kinase signaling. We provide evidence that Rac1 deletion‐induced hypertension is due to an alteration of cGMP signaling resulting from the loss of Rac1‐mediated control of type 5 PDE activity. Consequently, cGMP‐dependent phosphorylation and binding of RhoA with its inhibitory partner, the phosphatase‐RhoA interacting protein (p116RIP3), are decreased. Conclusions Our data reveal that the depletion of Rac1 in SMC decreases cGMP‐dependent p116RIP3/RhoA interaction and the subsequent inhibition of RhoA signaling. Thus, we unveil an in vivo role of Rac1 in arterial blood pressure regulation and a new pathway involving p116RIP3 that contributes to the antagonistic relationship between Rac1 and RhoA in vascular smooth muscle cells and their opposite roles in arterial tone and blood pressure.
Collapse
Affiliation(s)
- Gwennan André
- Inserm UMR_S1087, CNRS UMR_C6291, l'institut du thorax, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) Université de Nantes, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.)
| | - Juan E Sandoval
- Inserm UMR_S1087, CNRS UMR_C6291, l'institut du thorax, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) Université de Nantes, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.)
| | - Kevin Retailleau
- Inserm UMR_S1083, CNRS UMR_C6214, BNMI, Angers, F-49000, France (K.R., L.L.)
| | - Laurent Loufrani
- Inserm UMR_S1083, CNRS UMR_C6214, BNMI, Angers, F-49000, France (K.R., L.L.)
| | - Gilles Toumaniantz
- Inserm UMR_S1087, CNRS UMR_C6291, l'institut du thorax, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) Université de Nantes, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.)
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (S.O.)
| | - Malvyne Rolli-Derkinderen
- Inserm UMR_S1087, CNRS UMR_C6291, l'institut du thorax, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) Université de Nantes, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.)
| | - Gervaise Loirand
- Inserm UMR_S1087, CNRS UMR_C6291, l'institut du thorax, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) Université de Nantes, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) CHU Nantes, l'institut du thorax, Nantes, F-44000, France (G.L., V.S.)
| | - Vincent Sauzeau
- Inserm UMR_S1087, CNRS UMR_C6291, l'institut du thorax, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) Université de Nantes, Nantes, F-44000, France (G.A., J.E.S., G.T., M.R.D., G.L., V.S.) CHU Nantes, l'institut du thorax, Nantes, F-44000, France (G.L., V.S.)
| |
Collapse
|
24
|
Park A, Kim WK, Bae KH. Distinction of white, beige and brown adipocytes derived from mesenchymal stem cells. World J Stem Cells 2014; 6:33-42. [PMID: 24567786 PMCID: PMC3927012 DOI: 10.4252/wjsc.v6.i1.33] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/05/2013] [Accepted: 01/06/2014] [Indexed: 02/06/2023] Open
Abstract
Adipose tissue is a major metabolic organ, and it has been traditionally classified as either white adipose tissue (WAT) or brown adipose tissue (BAT). WAT and BAT are characterized by different anatomical locations, morphological structures, functions, and regulations. WAT and BAT are both involved in energy balance. WAT is mainly involved in the storage and mobilization of energy in the form of triglycerides, whereas BAT specializes in dissipating energy as heat during cold- or diet-induced thermogenesis. Recently, brown-like adipocytes were discovered in WAT. These brown-like adipocytes that appear in WAT are called beige or brite adipocytes. Interestingly, these beige/brite cells resemble white fat cells in the basal state, but they respond to thermogenic stimuli with increased levels of thermogenic genes and increased respiration rates. In addition, beige/brite cells have a gene expression pattern distinct from that of either white or brown fat cells. The current epidemic of obesity has increased the interest in studying adipocyte formation (adipogenesis), especially in beige/brite cells. This review summarizes the developmental process of adipose tissues that originate from the mesenchymal stem cells and the features of these three different types of adipocytes.
Collapse
Affiliation(s)
- Anna Park
- Anna Park, Won Kon Kim, Kwang-Hee Bae, Research Center for Integrated Cellulomics, KRIBB, Daejeon 305-806, South Korea
| | - Won Kon Kim
- Anna Park, Won Kon Kim, Kwang-Hee Bae, Research Center for Integrated Cellulomics, KRIBB, Daejeon 305-806, South Korea
| | - Kwang-Hee Bae
- Anna Park, Won Kon Kim, Kwang-Hee Bae, Research Center for Integrated Cellulomics, KRIBB, Daejeon 305-806, South Korea
| |
Collapse
|
25
|
Colombo G, Colombo MDHP, Schiavon LDL, d'Acampora AJ. Phosphodiesterase 5 as target for adipose tissue disorders. Nitric Oxide 2013; 35:186-192. [PMID: 24177060 DOI: 10.1016/j.niox.2013.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/26/2013] [Accepted: 10/21/2013] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Adipose tissue as an endocrine organ is responsible for the release of multiple cytokines, which have the most diverse metabolic functions. Therefore, it is extremely important to preserve its physiological health in order to avoid local and systemic disorders. Experiments available in literature show the importance of the nitric oxide (NO)/guanosine 3'5' cyclic monophosphate (cGMP)/protein kinase G (PKG) pathway in adipocyte biology. Phosphodiesterase 5 (PDE5) is an enzyme responsible for cGMP inactivation, and the use of its inhibitors can be an alternative in the search of a more balanced adipose tissue. OBJECTIVE This review aims to describe the PDE5 role and the possibility of using PDE5 inhibitors in adipocyte physiology derangements and their consequences. DESIGN AND METHODS Studies published in the last 10years that related PDE5 and its inhibitors to adipose tissue were raised in major databases. RESULTS PDE5 is present in adipocyte, and PDE5 inhibitors can promote adipogenesis, interfere with adipokines secretion, decrease inflammatory markers expression, and increase the thermogenic potential of white adipose tissue. CONCLUSIONS PDE5 plays an important role in adipocyte physiology and the use of its inhibitors may prove a useful tool to combat adipose tissue disorders and its highest expression, metabolic syndrome.
Collapse
|
26
|
miR-155 regulates differentiation of brown and beige adipocytes via a bistable circuit. Nat Commun 2013; 4:1769. [PMID: 23612310 PMCID: PMC3644088 DOI: 10.1038/ncomms2742] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 03/15/2013] [Indexed: 12/11/2022] Open
Abstract
Brown adipocytes are a primary site of energy expenditure and reside not only in classical brown adipose tissue but can also be found in white adipose tissue. Here we show that microRNA 155 is enriched in brown adipose tissue and is highly expressed in proliferating brown preadipocytes but declines after induction of differentiation. Interestingly, microRNA 155 and its target, the adipogenic transcription factor CCAAT/enhancer-binding protein β, form a bistable feedback loop integrating hormonal signals that regulate proliferation or differentiation. Inhibition of microRNA 155 enhances brown adipocyte differentiation and induces a brown adipocyte-like phenotype (‘browning’) in white adipocytes. Consequently, microRNA 155-deficient mice exhibit increased brown adipose tissue function and ‘browning’ of white fat tissue. In contrast, transgenic overexpression of microRNA 155 in mice causes a reduction of brown adipose tissue mass and impairment of brown adipose tissue function. These data demonstrate that the bistable loop involving microRNA 155 and CCAAT/enhancer-binding protein β regulates brown lineage commitment, thereby, controlling the development of brown and beige fat cells. Brown fat can dissipate energy as heat and has an important role in energy homoeostasis of rodents and possibly humans. Chen et al. show that microRNA 155 regulates the differentiation of brown adipocytes as well as the 'browning' of white fat cells in mice.
Collapse
|
27
|
Hildebrand S, Zimmermann K, Wenzel D, Fleischmann BK, Pfeifer A. The role of VASP in cGMP-mediated vascular smooth muscle relaxation. BMC Pharmacol Toxicol 2013. [PMCID: PMC3765648 DOI: 10.1186/2050-6511-14-s1-p28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
28
|
Pfeifer A, Kilić A, Hoffmann LS. Regulation of metabolism by cGMP. Pharmacol Ther 2013; 140:81-91. [PMID: 23756133 DOI: 10.1016/j.pharmthera.2013.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 01/16/2023]
Abstract
The second messenger cyclic guanosine monophosphate (cGMP) mediates the physiological effects of nitric oxide and natriuretic peptides in a broad spectrum of tissues and cells. So far, the major focus of research on cGMP lay on the cardiovascular system. Recent evidence suggests that cGMP also plays a major role in the regulation of cellular and whole-body metabolism. Here, we focus on the role of cGMP in adipose tissue. In addition, other organs important for the regulation of metabolism and their regulation by cGMP are discussed. Targeting the cGMP signaling pathway could be an exciting approach for the regulation of energy expenditure and the treatment of obesity.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, Germany.
| | | | | |
Collapse
|
29
|
Mitschke MM, Hoffmann LS, Gnad T, Scholz D, Kruithoff K, Mayer P, Haas B, Sassmann A, Pfeifer A, Kilic A. Increased cGMP promotes healthy expansion and browning of white adipose tissue. FASEB J 2013; 27:1621-30. [PMID: 23303211 DOI: 10.1096/fj.12-221580] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
With more than half a billion individuals affected worldwide, obesity has reached pandemic proportions. Development of "brown-like" or "brite" adipocytes within white adipose tissue (WAT) has potential antiobesity and insulin-sensitizing effects. We investigated the role of cyclic GMP (cGMP) signaling, focusing on cGMP-dependent protein kinase I (PKGI) in WAT. PKGI is expressed in murine WAT, primary adipocytes, and 3T3-L1. Treatment of adipocytes with cGMP resulted in increased adipogenesis, with a 54% increase in expression of peroxisome proliferator-activated receptor-γ. Lentiviral overexpression of PKGI further increased adipogenesis, whereas loss of PKGI significantly reduced adipogenic differentiation. In addition to adipogenic effects, PKGI had an antihypertrophic and anti-inflammatory effect via RhoA phosphorylation and reduction of proinflammatory adipokine expression. Moreover, PKGI induced a 4.3-fold increase in abundance of UCP-1 and the development of a brown-like thermogenic program in primary adipocytes. Notably, treatment of C57BL/6 mice with phosphodiesterase inhibitor sildenafil (12 mg/kg/d) for 7 d caused 4.6-fold increase in uncoupling protein-1 expression and promoted establishment of a brown fat cell-like phenotype ("browning") of WAT in vivo. Taken together, PKGI is a key regulator of cell size, adipokine secretion and browning of white fat depots and thus could be a valuable target in developing novel treatments for obesity.
Collapse
|
30
|
Klingenspor M, Herzig S, Pfeifer A. Brown fat develops a brite future. Obes Facts 2012; 5:890-6. [PMID: 23296106 DOI: 10.1159/000346337] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 01/02/2023] Open
|