1
|
Ghesmati Z, Rashid M, Fayezi S, Gieseler F, Alizadeh E, Darabi M. An update on the secretory functions of brown, white, and beige adipose tissue: Towards therapeutic applications. Rev Endocr Metab Disord 2024; 25:279-308. [PMID: 38051471 PMCID: PMC10942928 DOI: 10.1007/s11154-023-09850-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 12/07/2023]
Abstract
Adipose tissue, including white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue, is vital in modulating whole-body energy metabolism. While WAT primarily stores energy, BAT dissipates energy as heat for thermoregulation. Beige adipose tissue is a hybrid form of adipose tissue that shares characteristics with WAT and BAT. Dysregulation of adipose tissue metabolism is linked to various disorders, including obesity, type 2 diabetes, cardiovascular diseases, cancer, and infertility. Both brown and beige adipocytes secrete multiple molecules, such as batokines, packaged in extracellular vesicles or as soluble signaling molecules that play autocrine, paracrine, and endocrine roles. A greater understanding of the adipocyte secretome is essential for identifying novel molecular targets in treating metabolic disorders. Additionally, microRNAs show crucial roles in regulating adipose tissue differentiation and function, highlighting their potential as biomarkers for metabolic disorders. The browning of WAT has emerged as a promising therapeutic approach in treating obesity and associated metabolic disorders. Many browning agents have been identified, and nanotechnology-based drug delivery systems have been developed to enhance their efficacy. This review scrutinizes the characteristics of and differences between white, brown, and beige adipose tissues, the molecular mechanisms involved in the development of the adipocytes, the significant roles of batokines, and regulatory microRNAs active in different adipose tissues. Finally, the potential of WAT browning in treating obesity and atherosclerosis, the relationship of BAT with cancer and fertility disorders, and the crosstalk between adipose tissue with circadian system and circadian disorders are also investigated.
Collapse
Affiliation(s)
- Zeinab Ghesmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Fayezi
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Frank Gieseler
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
2
|
Klein M, Failla AV, Hermey G. Internally tagged Vps10p-domain receptors reveal uptake of the neurotrophin BDNF. J Biol Chem 2023; 299:105216. [PMID: 37660918 PMCID: PMC10540051 DOI: 10.1016/j.jbc.2023.105216] [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: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023] Open
Abstract
The Vps10p-domain (Vps10p-D) receptor family consists of Sortilin, SorLA, SorCS1, SorCS2, and SorCS3. They mediate internalization and intracellular sorting of specific cargo in various cell types, but underlying molecular determinants are incompletely understood. Deciphering the dynamic intracellular itineraries of Vps10p-D receptors is crucial for understanding their role in physiological and cytopathological processes. However, studying their spatial and temporal dynamics by live imaging has been challenging so far, as terminal tagging with fluorophores presumably impedes several of their protein interactions and thus functions. Here, we addressed the lack of appropriate tools and developed functional versions of all family members internally tagged in their ectodomains. We predict folding of the newly designed receptors by bioinformatics and show their exit from the endoplasmic reticulum. We examined their subcellular localization in immortalized cells and primary cultured neurons by immunocytochemistry and live imaging. This was, as far as known, identical to that of wt counterparts. We observed homodimerization of fluorophore-tagged SorCS2 by coimmunoprecipitation and fluorescence lifetime imaging, suggesting functional leucine-rich domains. Through ligand uptake experiments, live imaging and fluorescence lifetime imaging, we show for the first time that all Vps10p-D receptors interact with the neurotrophin brain-derived neurotrophic factor and mediate its uptake, indicating functionality of the Vps10p-Ds. In summary, we developed versions of all Vps10p-D receptors, with internal fluorophore tags that preserve several functions of the cytoplasmic and extracellular domains. These newly developed fluorophore-tagged receptors are likely to serve as powerful functional tools for accurate live studies of the individual cellular functions of Vps10p-D receptors.
Collapse
Affiliation(s)
- Marcel Klein
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | - Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
3
|
Sa M, Yoo ES, Koh W, Park MG, Jang HJ, Yang YR, Bhalla M, Lee JH, Lim J, Won W, Kwon J, Kwon JH, Seong Y, Kim B, An H, Lee SE, Park KD, Suh PG, Sohn JW, Lee CJ. Hypothalamic GABRA5-positive neurons control obesity via astrocytic GABA. Nat Metab 2023; 5:1506-1525. [PMID: 37653043 DOI: 10.1038/s42255-023-00877-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/25/2023] [Indexed: 09/02/2023]
Abstract
The lateral hypothalamic area (LHA) regulates food intake and energy balance. Although LHA neurons innervate adipose tissues, the identity of neurons that regulate fat is undefined. Here we show that GABRA5-positive neurons in LHA (GABRA5LHA) polysynaptically project to brown and white adipose tissues in the periphery. GABRA5LHA are a distinct subpopulation of GABAergic neurons and show decreased pacemaker firing in diet-induced obesity mouse models in males. Chemogenetic inhibition of GABRA5LHA suppresses fat thermogenesis and increases weight gain, whereas gene silencing of GABRA5 in LHA decreases weight gain. In the diet-induced obesity mouse model, GABRA5LHA are tonically inhibited by nearby reactive astrocytes releasing GABA, which is synthesized by monoamine oxidase B (Maob). Gene silencing of astrocytic Maob in LHA facilitates fat thermogenesis and reduces weight gain significantly without affecting food intake, which is recapitulated by administration of a Maob inhibitor, KDS2010. We propose that firing of GABRA5LHA suppresses fat accumulation and selective inhibition of astrocytic GABA is a molecular target for treating obesity.
Collapse
Affiliation(s)
- Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Mingu Gordon Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hyun-Jun Jang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yong Ryoul Yang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Mridula Bhalla
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Woojin Won
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jea Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Joon-Ho Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Yejin Seong
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Byungeun Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Heeyoung An
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Pann-Ghill Suh
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea.
- IBS School, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
4
|
Carobbio S, Vidal-Puig A. Differentiation of Human Pluripotent Stem Cells (hPSCs) into Brown-Like Adipocytes. Methods Mol Biol 2023; 2662:1-9. [PMID: 37076666 DOI: 10.1007/978-1-0716-3167-6_1] [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] [Indexed: 04/21/2023]
Abstract
Increasing brown adipose tissue (BAT) mass and activation is a therapeutic strategy to prevent and treat obesity and associated complications. Obese and diabetic patients possess less BAT; thus, finding an efficient way to expand their mass is necessary. There is limited knowledge about how human BAT develops, differentiates, and is optimally activated. Accessing human BAT is challenging, given its scarcity and anatomical dispersion. These constraints make detailed BAT-related developmental and functional mechanistic studies in human subjects virtually impossible. We have developed a new chemically defined protocol for differentiating human pluripotent stem cells (hPSCs) into bona fide brown adipocytes (BAs) that overcomes current limitations. This protocol recapitulates step by step the physiological developmental path of human BAT.
Collapse
Affiliation(s)
- Stefania Carobbio
- Centro de Investigacion Principe Felipe, Valencia, Spain.
- Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Antonio Vidal-Puig
- Centro de Investigacion Principe Felipe, Valencia, Spain.
- Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| |
Collapse
|
5
|
Salasova A, Monti G, Andersen OM, Nykjaer A. Finding memo: versatile interactions of the VPS10p-Domain receptors in Alzheimer’s disease. Mol Neurodegener 2022; 17:74. [PMID: 36397124 PMCID: PMC9673319 DOI: 10.1186/s13024-022-00576-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
The family of VPS10p-Domain (D) receptors comprises five members named SorLA, Sortilin, SorCS1, SorCS2 and SorCS3. While their physiological roles remain incompletely resolved, they have been recognized for their signaling engagements and trafficking abilities, navigating a number of molecules between endosome, Golgi compartments, and the cell surface. Strikingly, recent studies connected all the VPS10p-D receptors to Alzheimer’s disease (AD) development. In addition, they have been also associated with diseases comorbid with AD such as diabetes mellitus and major depressive disorder. This systematic review elaborates on genetic, functional, and mechanistic insights into how dysfunction in VPS10p-D receptors may contribute to AD etiology, AD onset diversity, and AD comorbidities. Starting with their functions in controlling cellular trafficking of amyloid precursor protein and the metabolism of the amyloid beta peptide, we present and exemplify how these receptors, despite being structurally similar, regulate various and distinct cellular events involved in AD. This includes a plethora of signaling crosstalks that impact on neuronal survival, neuronal wiring, neuronal polarity, and synaptic plasticity. Signaling activities of the VPS10p-D receptors are especially linked, but not limited to, the regulation of neuronal fitness and apoptosis via their physical interaction with pro- and mature neurotrophins and their receptors. By compiling the functional versatility of VPS10p-D receptors and their interactions with AD-related pathways, we aim to further propel the AD research towards VPS10p-D receptor family, knowledge that may lead to new diagnostic markers and therapeutic strategies for AD patients.
Collapse
|
6
|
Yin X, Chen Y, Ruze R, Xu R, Song J, Wang C, Xu Q. The evolving view of thermogenic fat and its implications in cancer and metabolic diseases. Signal Transduct Target Ther 2022; 7:324. [PMID: 36114195 PMCID: PMC9481605 DOI: 10.1038/s41392-022-01178-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 02/07/2023] Open
Abstract
AbstractThe incidence of metabolism-related diseases like obesity and type 2 diabetes mellitus has reached pandemic levels worldwide and increased gradually. Most of them are listed on the table of high-risk factors for malignancy, and metabolic disorders systematically or locally contribute to cancer progression and poor prognosis of patients. Importantly, adipose tissue is fundamental to the occurrence and development of these metabolic disorders. White adipose tissue stores excessive energy, while thermogenic fat including brown and beige adipose tissue dissipates energy to generate heat. In addition to thermogenesis, beige and brown adipocytes also function as dynamic secretory cells and a metabolic sink of nutrients, like glucose, fatty acids, and amino acids. Accordingly, strategies that activate and expand thermogenic adipose tissue offer therapeutic promise to combat overweight, diabetes, and other metabolic disorders through increasing energy expenditure and enhancing glucose tolerance. With a better understanding of its origins and biological functions and the advances in imaging techniques detecting thermogenesis, the roles of thermogenic adipose tissue in tumors have been revealed gradually. On the one hand, enhanced browning of subcutaneous fatty tissue results in weight loss and cancer-associated cachexia. On the other hand, locally activated thermogenic adipocytes in the tumor microenvironment accelerate cancer progression by offering fuel sources and is likely to develop resistance to chemotherapy. Here, we enumerate current knowledge about the significant advances made in the origin and physiological functions of thermogenic fat. In addition, we discuss the multiple roles of thermogenic adipocytes in different tumors. Ultimately, we summarize imaging technologies for identifying thermogenic adipose tissue and pharmacologic agents via modulating thermogenesis in preclinical experiments and clinical trials.
Collapse
|
7
|
The armadillo-repeat containing X-linked protein 3, ARMCX3, is a negative regulator of the browning of adipose tissue associated with obesity. Int J Obes (Lond) 2022; 46:1652-1661. [PMID: 35705702 DOI: 10.1038/s41366-022-01169-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVES To determine the role of armadillo repeat-containing X-linked protein 3 (ARMCX3) in the thermogenic plasticity of adipose tissue. METHODS Adipose tissues were characterized in Armcx3-KO male mice. Armcx3 gene expression was analyzed in adipose tissue from mice exposed to thermogenic inducers (cold, β3-adenergic stimulus) and in differentiating brown and beige cells in culture. Analyses encompassed circulating metabolite and hormonal profiling, tissue characterization, histology, gene expression patterns, and immunoblot assays. Armcx3 gene expression was assessed in subcutaneous adipose tissue from lean individuals and individuals with obesity and was correlated with expression of marker genes of adipose browning. The effects of adenoviral-mediated overexpression of ARMCX3 on differentiating brown adipocyte gene expression and respiratory activity were determined. RESULTS Male mice lacking ARMCX3 showed significant induction of white adipose tissue browning. In humans, ARMCX3 expression in subcutaneous adipose tissue was inversely correlated with the expression of marker genes of thermogenic activity, including CIDEA, mitochondrial transcripts, and creatine kinase-B. Armcx3 expression in adipose tissues was repressed by thermogenic activation (cold or β3-adrenergic stimulation) and was upregulated by obesity in mice and humans. Experimentally-induced increases in Armcx3 caused down-regulation of thermogenesis-related genes and reduced mitochondrial oxidative activity of adipocytes in culture, whereas siRNA-mediated Armcx3 knocking-down enhanced expression of thermogenesis-related genes. CONCLUSION ARMCX3 is a novel player in the control of thermogenic adipose tissue plasticity that acts to repress acquisition of the browning phenotype and shows a direct association with indicators of obesity in mice and humans.
Collapse
|
8
|
Wang Y, Luan M, Xue L, Jin J, Xie A. Evaluation of the relationship between SORL1 gene polymorphism and Parkinson's disease in the Chinese population. Neurosci Lett 2022; 778:136602. [DOI: 10.1016/j.neulet.2022.136602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/26/2022] [Indexed: 11/29/2022]
|
9
|
Joki Y, Konishi H, Ebinuma H, Takasu K, Minamino T. Circulating sLR11 levels predict severity of pulmonary hypertension due to left heart disease. PLoS One 2021; 16:e0261753. [PMID: 34965280 PMCID: PMC8716052 DOI: 10.1371/journal.pone.0261753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/07/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Heart failure is a severe condition often involving pulmonary hypertension (PH). Soluble low-density lipoprotein receptor with 11 ligand-binding repeats (sLR11) has been associated with pulmonary artery hypertension. We examined whether sLR11 correlates with PH in left heart disease and can be used as a predictive marker. METHOD We retrospectively analyzed patients with severe mitral regurgitation who underwent right heart catheterization before surgery for valve replacement or valvuloplasty from November 2005 to October 2012 at Juntendo University. We measured sLR11 levels before right heart catheterization and analyzed correlations with pulmonary hemodynamics. We compared prognoses between a group with normal sLR11 (≤9.4 ng/ml) and a group with high sLR11 (>9.4 ng/ml). Follow-up was continued for 5 years, with end points of hospitalization due to HF and death due to cardiovascular disease. RESULTS Among 34 patients who met the inclusion criteria, sLR11 correlated with mean pulmonary artery pressure (r = 0.54, p<0.001), transpulmonary pressure gradient (r = 0.42, p = 0.012), pulmonary vascular resistance (r = 0.36, p<0.05), and log brain natriuretic peptide (BNP). However, logBNP did not correlate with pulmonary vascular resistance (p = 0.6). Levels of sLR11 were significantly higher in the 10 patients with PH (14.4±4.3 ng/ml) than in patients without PH (9.9±3.9 ng/ml; p = 0.002). At 5 years, the event rate was higher in the high-sLR11 group than in the normal-sLR11 group. The high-sLR11 group showed 5 hospitalizations due to HF (25.0%) and 2 deaths (10.0%), whereas the normal-sLR11 group showed no hospitalizations or deaths. Analyses using receiver operating characteristic curves showed a higher area under the concentration-time curve (AUC) for sLR11 level (AUC = 0.85; 95% confidence interval (CI) = 0.72-0.98) than for BNP (AUC = 0.80, 95%CI = 0.62-0.99) in the diagnosis of PH in left heart disease. CONCLUSIONS Concentration of sLR11 is associated with severity of PH and offers a strong predictor of severe mitral regurgitation in patients after surgery.
Collapse
Affiliation(s)
- Yusuke Joki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo City, Tokyo, Japan
| | - Hakuoh Konishi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo City, Tokyo, Japan
- * E-mail:
| | - Hiroyuki Ebinuma
- Tsukuba Research Institute, Sekisui Medical Co Ltd, Ryugasaki, Japan
| | - Kiyoshi Takasu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo City, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo City, Tokyo, Japan
| |
Collapse
|
10
|
Schmidt V, Horváth C, Dong H, Blüher M, Qvist P, Wolfrum C, Willnow TE. SORLA is required for insulin-induced expansion of the adipocyte precursor pool in visceral fat. J Cell Biol 2021; 220:e202006058. [PMID: 34779857 PMCID: PMC8598079 DOI: 10.1083/jcb.202006058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/19/2021] [Accepted: 09/08/2021] [Indexed: 01/24/2023] Open
Abstract
Visceral adipose tissue shows remarkable plasticity, constantly replacing mature adipocytes from an inherent pool of adipocyte precursors. The number of precursors is set in the juvenile organism and remains constant in adult life. Which signals drive precursor pool expansion in juveniles and why they operate in visceral but not in subcutaneous white adipose tissue (WAT) are unclear. Using mouse models, we identified the insulin-sensitizing receptor SORLA as a molecular factor explaining the distinct proliferative capacity of visceral WAT. High levels of SORLA activity in precursors of juvenile visceral WAT prime these cells for nutritional stimuli provided through insulin, promoting mitotic expansion of the visceral precursor cell pool in overfed juvenile mice. SORLA activity is low in subcutaneous precursors, blunting their response to insulin and preventing diet-induced proliferation of this cell type. Our findings provide a molecular explanation for the unique proliferative properties of juvenile visceral WAT, and for the genetic association of SORLA with visceral obesity in humans.
Collapse
Affiliation(s)
- Vanessa Schmidt
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Carla Horváth
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Hua Dong
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Per Qvist
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, Aarhus University, Aarhus, Denmark
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Thomas E. Willnow
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Brandão BB, Poojari A, Rabiee A. Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential. Int J Mol Sci 2021; 22:5906. [PMID: 34072788 PMCID: PMC8198523 DOI: 10.3390/ijms22115906] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The concerning worldwide increase of obesity and chronic metabolic diseases, such as T2D, dyslipidemia, and cardiovascular disease, motivates further investigations into preventive and alternative therapeutic approaches. Over the past decade, there has been growing evidence that the formation and activation of thermogenic adipocytes (brown and beige) may serve as therapy to treat obesity and its associated diseases owing to its capacity to increase energy expenditure and to modulate circulating lipids and glucose levels. Thus, understanding the molecular mechanism of brown and beige adipocytes formation and activation will facilitate the development of strategies to combat metabolic disorders. Here, we provide a comprehensive overview of pathways and players involved in the development of brown and beige fat, as well as the role of thermogenic adipocytes in energy homeostasis and metabolism. Furthermore, we discuss the alterations in brown and beige adipose tissue function during obesity and explore the therapeutic potential of thermogenic activation to treat metabolic syndrome.
Collapse
Affiliation(s)
- Bruna B. Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Ankita Poojari
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| | - Atefeh Rabiee
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
12
|
Carobbio S, Guenantin AC, Bahri M, Rodriguez-Fdez S, Honig F, Kamzolas I, Samuelson I, Long K, Awad S, Lukovic D, Erceg S, Bassett A, Mendjan S, Vallier L, Rosen BS, Chiarugi D, Vidal-Puig A. Unraveling the Developmental Roadmap toward Human Brown Adipose Tissue. Stem Cell Reports 2021; 16:641-655. [PMID: 33606988 PMCID: PMC7940445 DOI: 10.1016/j.stemcr.2021.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/20/2022] Open
Abstract
Increasing brown adipose tissue (BAT) mass and activation is a therapeutic strategy to treat obesity and complications. Obese and diabetic patients possess low amounts of BAT, so an efficient way to expand their mass is necessary. There is limited knowledge about how human BAT develops, differentiates, and is optimally activated. Accessing human BAT is challenging, given its low volume and anatomical dispersion. These constraints make detailed BAT-related developmental and functional mechanistic studies in humans virtually impossible. We have developed and characterized functionally and molecularly a new chemically defined protocol for the differentiation of human pluripotent stem cells (hPSCs) into brown adipocytes (BAs) that overcomes current limitations. This protocol recapitulates step by step the physiological developmental path of human BAT. The BAs obtained express BA and thermogenic markers, are insulin sensitive, and responsive to β-adrenergic stimuli. This new protocol is scalable, enabling the study of human BAs at early stages of development.
Collapse
Affiliation(s)
- Stefania Carobbio
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Anne-Claire Guenantin
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Myriam Bahri
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | | | - Floris Honig
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Ioannis Kamzolas
- Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Isabella Samuelson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Kathleen Long
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Sherine Awad
- Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Dunja Lukovic
- Retinal Degeneration Lab and National Stem Cell Bank-Valencia Node, Research Center Principe Felipe, Valencia, Spain
| | - Slaven Erceg
- Stem Cell Therapies for Neurodegenerative Diseases Lab and National Stem Cell Bank - Valencia Node, Research Center Principe Felipe, Valencia, Spain
| | - Andrew Bassett
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Sasha Mendjan
- Institute of Molecular Biotechnology, 1030 Vienna, Austria
| | - Ludovic Vallier
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK; Department of Surgery, University of Cambridge, Cambridge, UK
| | - Barry S Rosen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Davide Chiarugi
- Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Jiangbei Area, Nanjing, P.R. China.
| |
Collapse
|
13
|
VPS10P Domain Receptors: Sorting Out Brain Health and Disease. Trends Neurosci 2020; 43:870-885. [DOI: 10.1016/j.tins.2020.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/23/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
|
14
|
Kwok A, Zvetkova I, Virtue S, Luijten I, Huang-Doran I, Tomlinson P, Bulger DA, West J, Murfitt S, Griffin J, Alam R, Hart D, Knox R, Voshol P, Vidal-Puig A, Jensen J, O'Rahilly S, Semple RK. Truncation of Pik3r1 causes severe insulin resistance uncoupled from obesity and dyslipidaemia by increased energy expenditure. Mol Metab 2020; 40:101020. [PMID: 32439336 PMCID: PMC7385515 DOI: 10.1016/j.molmet.2020.101020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Insulin signalling via phosphoinositide 3-kinase (PI3K) requires PIK3R1-encoded regulatory subunits. C-terminal PIK3R1 mutations cause SHORT syndrome, as well as lipodystrophy and insulin resistance (IR), surprisingly without fatty liver or metabolic dyslipidaemia. We sought to investigate this discordance. METHODS The human pathogenic Pik3r1 Y657∗ mutation was knocked into mice by homologous recombination. Growth, body composition, bioenergetic and metabolic profiles were investigated on chow and high-fat diet (HFD). We examined adipose and liver histology, and assessed liver responses to fasting and refeeding transcriptomically. RESULTS Like humans with SHORT syndrome, Pik3r1WT/Y657∗ mice were small with severe IR, and adipose expansion on HFD was markedly reduced. Also as in humans, plasma lipid concentrations were low, and insulin-stimulated hepatic lipogenesis was not increased despite hyperinsulinemia. At odds with lipodystrophy, however, no adipocyte hypertrophy nor adipose inflammation was found. Liver lipogenic gene expression was not significantly altered, and unbiased transcriptomics showed only minor changes, including evidence of reduced endoplasmic reticulum stress in the fed state and diminished Rictor-dependent transcription on fasting. Increased energy expenditure, which was not explained by hyperglycaemia nor intestinal malabsorption, provided an alternative explanation for the uncoupling of IR from dyslipidaemia. CONCLUSIONS Pik3r1 dysfunction in mice phenocopies the IR and reduced adiposity without lipotoxicity of human SHORT syndrome. Decreased adiposity may not reflect bona fide lipodystrophy, but rather, increased energy expenditure, and we suggest that further study of brown adipose tissue in both humans and mice is warranted.
Collapse
Affiliation(s)
- Albert Kwok
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Ilona Zvetkova
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Sam Virtue
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Ineke Luijten
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Isabel Huang-Doran
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Patsy Tomlinson
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - David A Bulger
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - James West
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Steven Murfitt
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Julian Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK; Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Medicine, Imperial College London, The Sir Alexander Fleming Building, London, UK
| | - Rafeah Alam
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge, UK
| | - Daniel Hart
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Rachel Knox
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Peter Voshol
- Louis Bolk Institute, Kosterijland 3-5, NL-3981 AJ, Bunnik, the Netherlands
| | - Antonio Vidal-Puig
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Jørgen Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, P.O. Box 4014, Ulleval Stadion, 0806 Oslo, Norway
| | - Stephen O'Rahilly
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Robert K Semple
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK; The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK; MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK.
| |
Collapse
|
15
|
Soluble SORLA Enhances Neurite Outgrowth and Regeneration through Activation of the EGF Receptor/ERK Signaling Axis. J Neurosci 2020; 40:5908-5921. [PMID: 32601248 DOI: 10.1523/jneurosci.0723-20.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 01/01/2023] Open
Abstract
SORLA is a transmembrane trafficking protein associated with Alzheimer's disease risk. Although SORLA is abundantly expressed in neurons, physiological roles for SORLA remain unclear. Here, we show that cultured transgenic neurons overexpressing SORLA feature longer neurites, and accelerated neurite regeneration with wounding. Enhanced release of a soluble form of SORLA (sSORLA) is observed in transgenic mouse neurons overexpressing human SORLA, while purified sSORLA promotes neurite extension and regeneration. Phosphoproteomic analyses demonstrate enrichment of phosphoproteins related to the epidermal growth factor (EGFR)/ERK pathway in SORLA transgenic mouse hippocampus from both genders. sSORLA coprecipitates with EGFR in vitro, and sSORLA treatment increases EGFR Y1173 phosphorylation, which is involved in ERK activation in cultured neurons. Furthermore, sSORLA triggers ERK activation, whereas pharmacological EGFR or ERK inhibition reverses sSORLA-dependent enhancement of neurite outgrowth. In search for downstream ERK effectors activated by sSORLA, we identified upregulation of Fos expression in hippocampus from male mice overexpressing SORLA by RNAseq analysis. We also found that Fos is upregulated and translocates to the nucleus in an ERK-dependent manner in neurons treated with sSORLA. Together, these results demonstrate that sSORLA is an EGFR-interacting protein that activates EGFR/ERK/Fos signaling to enhance neurite outgrowth and regeneration.SIGNIFICANCE STATEMENT SORLA is a transmembrane trafficking protein previously known to reduce the levels of amyloid-β, which is critical in the pathogenesis of Alzheimer's disease. In addition, SORLA mutations are a risk factor for Alzheimer's disease. Interestingly, the SORLA ectodomain is cleaved into a soluble form, sSORLA, which has been shown to regulate cytoskeletal signaling pathways and cell motility in cells outside the nervous system. We show here that sSORLA binds and activates the EGF receptor to induce downstream signaling through the ERK serine/threonine kinase and the Fos transcription factor, thereby enhancing neurite outgrowth. These findings reveal a novel role for sSORLA in promoting neurite regeneration through the EGF receptor/ERK/Fos pathway, thereby demonstrating a potential neuroprotective mechanism involving SORLA.
Collapse
|
16
|
The kallikrein-kinin pathway as a mechanism for auto-control of brown adipose tissue activity. Nat Commun 2020; 11:2132. [PMID: 32358539 PMCID: PMC7195474 DOI: 10.1038/s41467-020-16009-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Brown adipose tissue (BAT) is known to secrete regulatory factors in response to thermogenic stimuli. Components of the BAT secretome may exert local effects that contribute to BAT recruitment and activation. Here, we found that a thermogenic stimulus leads to enhanced secretion of kininogen (Kng) by BAT, owing to induction of kininogen 2 (Kng2) gene expression. Noradrenergic, cAMP-mediated signals induce KNG2 expression and release in brown adipocytes. Conversely, the expression of kinin receptors, that are activated by the Kng products bradykinin and [Des-Arg9]-bradykinin, are repressed by thermogenic activation of BAT in vivo and of brown adipocytes in vitro. Loss-of-function models for Kng (the circulating-Kng-deficient BN/Ka rat) and bradykinin (pharmacological inhibition of kinin receptors, kinin receptor-null mice) signaling were coincident in showing abnormal overactivation of BAT. Studies in vitro indicated that Kng and bradykinin exert repressive effects on brown adipocyte thermogenic activity by interfering the PKA/p38 MAPK pathway of control of Ucp1 gene transcription, whereas impaired kinin receptor expression enhances it. Our findings identify the kallikrein–kinin system as a relevant component of BAT thermogenic regulation that provides auto-regulatory inhibitory signaling to BAT. Brown adipose tissue, known produce heat by metabolizing fat, is also secretes molecules capable of communicating with other organs. Here the authors show that brown adipose tissue secretes kininogen, a component of heat system regulation, that provides auto-regulatory inhibitory signaling in brown adipose tissue.
Collapse
|
17
|
Lillingston F, Fields P, Waechter R. Auricular Acupuncture Associated with Reduced Waist Circumference in Overweight Women-A Randomized Controlled Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:6471560. [PMID: 31929817 PMCID: PMC6935794 DOI: 10.1155/2019/6471560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/21/2019] [Accepted: 10/01/2019] [Indexed: 11/17/2022]
Abstract
Obesity and subsequent ill health have reached epidemic proportions in developed countries, and many developing countries are on the same trajectory. Weight loss and sustaining a healthy weight have posed a significant challenge for individuals, patients, health-care providers, and public health experts. The literature suggests that dietary advice and lifestyle changes alone have limited sustainable impact for those who are seeking to achieve a healthy weight. Supplementary techniques to control weight, such as acupuncture and auricular acupuncture (AA), have shown mixed results and failed to clearly demonstrate a conclusive impact. This study aimed to provide clarity about the impact of AA on weight loss via a randomized controlled trial. Data were collected from patients to identify measurable girth reduction, weight loss, dietary choices, and mood changes over seven weekly sessions of AA (n = 30) versus sham needle as control (n = 28). Results demonstrated a large and highly significant AA treatment effect for reduced waist circumference over the course of the seven-week intervention. While the treatment effect for weight loss and BMI was not significant, this negative result may have been mediated by the relatively short duration of the study. Results also demonstrated a significant mood improvement across participants in both the AA intervention and control group as the intervention progressed. Further studies are required to determine if the reduction in waist circumference is driven specifically by the AA alone or in conjunction with improved mood. The results also have potentially significant implications for healthcare delivery in the fight against overweight and obesity.
Collapse
Affiliation(s)
| | - Paul Fields
- School of Medicine, St. George's University, True Blue, Grenada
| | | |
Collapse
|
18
|
Ishida H, Jiang M, Ebinuma H, Hiruta N, Schneider WJ, Kinoshita T, Bujo H. Circulating soluble LR11, a differentiation regulator for vascular cells, is increased during pregnancy and exaggerated in patients with pre-eclampsia. Clin Chim Acta 2019; 497:172-177. [PMID: 31299181 DOI: 10.1016/j.cca.2019.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/13/2019] [Accepted: 07/01/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Pre-eclampsia is a pregnancy-specific disease characterized by onset of hypertension and proteinuria, sometimes progressing into damaging other organs. Here, we investigated the pathological significance of the soluble fragment of LR11 (sLR11), a cell differentiation regulator, in comparison to circulating IL-6 and TNF-α, in pre-eclampsia. METHODS The study was conducted in a cross-sectional research design with fourteen pre-eclampsia patients and fifty healthy pregnant subjects. Pre-eclampsia was defined as hypertensive disorders in pregnancy at over 20 weeks of gestation with proteinuria. RESULTS Plasma levels of sLR11 as well as IL-6 in pre-eclampsia were increased compared with those in the healthy pregnant subjects at the first, the second, and the third trimester. Receiver operating characteristic analysis for the detection of pre-eclampsia among third-trimester subjects showed that the areas under the curves of sLR11 and IL-6 were equivalent. sLR11 and IL-6 correlated positively with TNF-α in healthy pregnant subjects. In the pre-eclampsia patients, there was neither a correlation between sLR11 and IL-6 nor between sLR11 and TNF-α. CONCLUSIONS sLR11 increases during pregnancy, with levels further exaggerated in pre-eclampsia, and may be related to the pathology of pre-eclampsia.
Collapse
Affiliation(s)
- Hiroaki Ishida
- Department of Obstetrics and Gynecology, Toho University Sakura Medical Center, Sakura, Japan
| | - Meizi Jiang
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University Sakura Medical Center, Sakura, Japan
| | - Hiroyuki Ebinuma
- Tsukuba Research Institute, Sekisui Medical Co Ltd, Ryugasaki, Japan
| | - Nobuyuki Hiruta
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University Sakura Medical Center, Sakura, Japan; Department of Surgical Pathology, Toho University Sakura Medical Center, Sakura, Japan
| | - Wolfgang J Schneider
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Toshihiko Kinoshita
- Department of Obstetrics and Gynecology, Toho University Sakura Medical Center, Sakura, Japan
| | - Hideaki Bujo
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University Sakura Medical Center, Sakura, Japan.
| |
Collapse
|
19
|
Adipose Tissue-Derived Signatures for Obesity and Type 2 Diabetes: Adipokines, Batokines and MicroRNAs. J Clin Med 2019; 8:jcm8060854. [PMID: 31208019 PMCID: PMC6617388 DOI: 10.3390/jcm8060854] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
: Obesity is one of the main risk factors for type 2 diabetes mellitus (T2DM). It is closely related to metabolic disturbances in the adipose tissue that primarily functions as a fat reservoir. For this reason, adipose tissue is considered as the primary site for initiation and aggravation of obesity and T2DM. As a key endocrine organ, the adipose tissue communicates with other organs, such as the brain, liver, muscle, and pancreas, for the maintenance of energy homeostasis. Two different types of adipose tissues-the white adipose tissue (WAT) and brown adipose tissue (BAT)-secrete bioactive peptides and proteins, known as "adipokines" and "batokines," respectively. Some of them have beneficial anti-inflammatory effects, while others have harmful inflammatory effects. Recently, "exosomal microRNAs (miRNAs)" were identified as novel adipokines, as adipose tissue-derived exosomal miRNAs can affect other organs. In the present review, we discuss the role of adipose-derived secretory factors-adipokines, batokines, and exosomal miRNA-in obesity and T2DM. It will provide new insights into the pathophysiological mechanisms involved in disturbances of adipose-derived factors and will support the development of adipose-derived factors as potential therapeutic targets for obesity and T2DM.
Collapse
|
20
|
Pietilä M, Sahgal P, Peuhu E, Jäntti NZ, Paatero I, Närvä E, Al-Akhrass H, Lilja J, Georgiadou M, Andersen OM, Padzik A, Sihto H, Joensuu H, Blomqvist M, Saarinen I, Boström PJ, Taimen P, Ivaska J. SORLA regulates endosomal trafficking and oncogenic fitness of HER2. Nat Commun 2019; 10:2340. [PMID: 31138794 PMCID: PMC6538630 DOI: 10.1038/s41467-019-10275-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 04/23/2019] [Indexed: 12/31/2022] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is an oncogene targeted by several kinase inhibitors and therapeutic antibodies. While the endosomal trafficking of many other receptor tyrosine kinases is known to regulate their oncogenic signalling, the prevailing view on HER2 is that this receptor is predominantly retained on the cell surface. Here, we find that sortilin-related receptor 1 (SORLA; SORL1) co-precipitates with HER2 in cancer cells and regulates HER2 subcellular distribution by promoting recycling of the endosomal receptor back to the plasma membrane. SORLA protein levels in cancer cell lines and bladder cancers correlates with HER2 levels. Depletion of SORLA triggers HER2 targeting to late endosomal/lysosomal compartments and impairs HER2-driven signalling and in vivo tumour growth. SORLA silencing also disrupts normal lysosome function and sensitizes anti-HER2 therapy sensitive and resistant cancer cells to lysosome-targeting cationic amphiphilic drugs. These findings reveal potentially important SORLA-dependent endosomal trafficking-linked vulnerabilities in HER2-driven cancers. The EGF receptor HER2 is an oncogene protein thought to reside at the plasma membrane, but its endosomal trafficking is currently unclear. Here, the authors report that HER2 is endocytosed and that sortillin-related receptor 1 (SORLA) promotes endosomal HER2 recycling and HER2 oncogenic signalling.
Collapse
Affiliation(s)
- Mika Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
| | - Pranshu Sahgal
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Emilia Peuhu
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Niklas Z Jäntti
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Elisa Närvä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Hussein Al-Akhrass
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Johanna Lilja
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Maria Georgiadou
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience Nordic-EMBL Partnership (DANDRITE), Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000, Aarhus, Denmark
| | - Artur Padzik
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Harri Sihto
- Laboratory of Molecular Oncology, Translational Cancer Biology Program, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, FI-00290, Helsinki, Finland
| | - Heikki Joensuu
- Laboratory of Molecular Oncology, Translational Cancer Biology Program, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, FI-00290, Helsinki, Finland
| | - Matias Blomqvist
- Institute of Biomedicine, University of Turku and Department of Pathology, Turku University Hospital, FI-20520, Turku, Finland
| | - Irena Saarinen
- Institute of Biomedicine, University of Turku and Department of Pathology, Turku University Hospital, FI-20520, Turku, Finland
| | - Peter J Boström
- Department of Urology, University of Turku and Turku University Hospital, FI-20520, Turku, Finland
| | - Pekka Taimen
- Institute of Biomedicine, University of Turku and Department of Pathology, Turku University Hospital, FI-20520, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland. .,Department of Biochemistry, University of Turku, FI-20520, Turku, Finland.
| |
Collapse
|
21
|
Kristóf E, Klusóczki Á, Veress R, Shaw A, Combi ZS, Varga K, Győry F, Balajthy Z, Bai P, Bacso Z, Fésüs L. Interleukin-6 released from differentiating human beige adipocytes improves browning. Exp Cell Res 2019; 377:47-55. [PMID: 30794803 DOI: 10.1016/j.yexcr.2019.02.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/30/2019] [Accepted: 02/18/2019] [Indexed: 01/12/2023]
Abstract
Brown and beige adipocytes contribute significantly to the regulation of whole body energy expenditure and systemic metabolic homeostasis not exclusively by thermogenesis through mitochondrial uncoupling. Several studies have provided evidence in rodents that brown and beige adipocytes produce a set of adipokines ("batokines") which regulate local tissue homeostasis and have beneficial effects on physiological functions of the entire body. We observed elevated secretion of Interleukin (IL)-6, IL-8 and monocyte chemoattractant protein (MCP)-1, but not tumor necrosis factor alpha (TNFα) or IL-1β pro-inflammatory cytokines, by ex vivo differentiating human beige adipocytes (induced by either PPARγ agonist or irisin) compared to white. Higher levels of IL-6, IL-8 and MCP-1 were released from human deep neck adipose tissue biopsies (enriched in browning cells) than from subcutaneous ones. IL-6 was produced in a sustained manner and mostly by the adipocytes and not by the undifferentiated progenitors. Continuous blocking of IL-6 receptor by specific antibody during beige differentiation resulted in downregulation of brown marker genes and increased morphological changes that are characteristic of white adipocytes. The data suggest that beige adipocytes adjust their production of IL-6 to reach an optimal level for differentiation in the medium enhancing browning in an autocrine manner.
Collapse
Affiliation(s)
- Endre Kristóf
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Ágnes Klusóczki
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Roland Veress
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Abhirup Shaw
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Zsolt Sándor Combi
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Klára Varga
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Ferenc Győry
- Department of Surgery, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Zoltán Balajthy
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Péter Bai
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary; Research Center for Molecular Medicine, University of Debrecen, Faculty of Medicine, Debrecen, Hungary; Department of Medical Chemistry, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, University of Debrecen, Faculties of Medicine and Pharmacy, Debrecen, Hungary
| | - László Fésüs
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary; MTA-DE Stem Cells, Apoptosis and Genomics Research Group of the Hungarian Academy of Sciences, Debrecen, Hungary.
| |
Collapse
|
22
|
Talbot H, Saada S, Naves T, Gallet PF, Fauchais AL, Jauberteau MO. Regulatory Roles of Sortilin and SorLA in Immune-Related Processes. Front Pharmacol 2019; 9:1507. [PMID: 30666202 PMCID: PMC6330335 DOI: 10.3389/fphar.2018.01507] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/10/2018] [Indexed: 12/25/2022] Open
Abstract
Sortilin, also known as Neurotensin Receptor-3, and the sorting-related receptor with type-A repeats (SorLA) are both members of the Vps10p domain receptor family. Initially identified in CNS cells, they are expressed in various other cell types where they exert multiple functions. Although mostly studied for its involvement in Alzheimer’s disease, SorLA has recently been shown to be implicated in immune response by regulating IL-6-mediated signaling, as well as driving monocyte migration. Sortilin has been shown to act as a receptor, as a co-receptor and as an intra- and extracellular trafficking regulator. In the last two decades, deregulation of sortilin has been demonstrated to be involved in many human pathophysiologies, including neurodegenerative disorders (Alzheimer and Parkinson diseases), type 2 diabetes and obesity, cancer, and cardiovascular pathologies such as atherosclerosis. Several studies highlighted different functions of sortilin in the immune system, notably in microglia, pro-inflammatory cytokine regulation, phagosome fusion and pathogen clearance. In this review, we will analyze the multiple roles of sortilin and SorLA in the human immune system and how their deregulation may be involved in disease development.
Collapse
Affiliation(s)
- Hugo Talbot
- Faculty of Medicine, University of Limoges, Limoges, France
| | - Sofiane Saada
- Faculty of Medicine, University of Limoges, Limoges, France
| | - Thomas Naves
- Faculty of Medicine, University of Limoges, Limoges, France
| | | | - Anne-Laure Fauchais
- Faculty of Medicine, University of Limoges, Limoges, France.,Department of Internal Medicine, University Hospital Limoges Dupuytren Hospital, Limoges, France
| | - Marie-Odile Jauberteau
- Faculty of Medicine, University of Limoges, Limoges, France.,Department of Immunology, University Hospital Limoges Dupuytren Hospital, Limoges, France
| |
Collapse
|
23
|
Abstract
Brown adipokines are regulatory factors secreted by brown and beige adipocytes that exhibit endocrine, paracrine, and autocrine actions. Peptidic and non-peptidic molecules, including miRNAs and lipids, are constituents of brown adipokines. Brown adipose tissue remodeling to meet thermogenic needs is dependent on the secretory properties of brown/beige adipocytes. The association between brown fat activity and a healthy metabolic profile, in relation to energy balance and glucose and lipid homeostasis, is influenced by the endocrine actions of brown adipokines. A comprehensive knowledge of the brown adipocyte secretome is still lacking. Advancements in the identification and characterization of brown adipokines will facilitate therapeutic interventions for metabolic diseases, as these molecules are obvious candidates to therapeutic agents. Moreover, identification of brown adipokines as circulating biomarkers of brown adipose tissue activity may be particularly useful for noninvasive assessment of brown adipose tissue alterations in human pathologies.
Collapse
Affiliation(s)
- Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Catalonia, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Barcelona, Spain.
| | - Aleix Gavaldà-Navarro
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Barcelona, Spain
| | - Marion Peyrou
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Barcelona, Spain
| | - Joan Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Barcelona, Spain
| |
Collapse
|
24
|
Yao G, Kang L, Li J, Long Y, Wei H, Ferreira CA, Jeffery JJ, Lin Y, Cai W, Wang X. Effective weight control via an implanted self-powered vagus nerve stimulation device. Nat Commun 2018; 9:5349. [PMID: 30559435 PMCID: PMC6297229 DOI: 10.1038/s41467-018-07764-z] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022] Open
Abstract
In vivo vagus nerve stimulation holds great promise in regulating food intake for obesity treatment. Here we present an implanted vagus nerve stimulation system that is battery-free and spontaneously responsive to stomach movement. The vagus nerve stimulation system comprises a flexible and biocompatible nanogenerator that is attached on the surface of stomach. It generates biphasic electric pulses in responsive to the peristalsis of stomach. The electric signals generated by this device can stimulate the vagal afferent fibers to reduce food intake and achieve weight control. This strategy is successfully demonstrated on rat models. Within 100 days, the average body weight is controlled at 350 g, 38% less than the control groups. This work correlates nerve stimulation with targeted organ functionality through a smart, self-responsive system, and demonstrated highly effective weight control. This work also provides a concept in therapeutic technology using artificial nerve signal generated from coordinated body activities. Developing new technologies for the neuromodulation of the vagus nerve can enable therapeutic strategies for body weight control in obese patients. Here, the authors present a battery-free self-powered implantable vagus nerve stimulation system that electrically responds to stomach movement.
Collapse
Affiliation(s)
- Guang Yao
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA.,State Key Laboratory of Electronic Thin films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, People's Republic of China
| | - Lei Kang
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Jun Li
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Yin Long
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA.,State Key Laboratory of Electronic Thin films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, People's Republic of China
| | - Hao Wei
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Carolina A Ferreira
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Justin J Jeffery
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA
| | - Yuan Lin
- State Key Laboratory of Electronic Thin films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, People's Republic of China
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Xudong Wang
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
25
|
Biochemical and cognitive effects of docosahexaenoic acid differ in a developmental and SorLA dependent manner. Behav Brain Res 2018; 348:90-100. [PMID: 29660442 DOI: 10.1016/j.bbr.2018.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/10/2018] [Accepted: 04/12/2018] [Indexed: 11/23/2022]
Abstract
Beneficial effects of omega-3 fatty acid intake on cognition are under debate as some studies show beneficial effects while others show no effects of omega-3 supplementation. These inconsistencies may be a result of inter-individual response variations, potentially caused by gene and diet interactions. SorLA is a multifunctional receptor involved in ligand trafficking including lipoprotein lipase and amyloid precursor protein. Decreased SorLA levels have been correlated to Alzheimer's disease, and omega-3 fatty acid supplementation is known to increase SorLA expression in neuronal cell lines and mouse models. We therefore addressed potential correlations between Sorl1 and dietary omega-3 in SorLA deficient mice (Sorl1-/-) and controls exposed to diets supplemented with or deprived of omega-3 during their entire development and lifespan (lifelong) or solely from the time of weaning (post weaning). Observed diet-induced effects were only evident when exposed to lifelong omega-3 supplementation or deprivation as opposed to post weaning exposure only. Lifelong exposure to omega-3 supplementation resulted in impaired spatial learning in Sorl1-/- mice. The vitamin C antioxidant capacity in the brains of Sorl1-/- mice was reduced, but reduced glutathione and vitamin E levels were increased, leaving the overall antioxidant capacity of the brain inconclusive. No gross morphological differences of hippocampal neurons were found to account for the altered behavior. We found a significant adverse effect in cognitive performance by combining SorLA deficiency with lifelong exposure to omega-3. Our results stress the need for investigations of the underlying molecular mechanisms to clarify the precise circumstances under which omega-3 supplementation may be beneficial.
Collapse
|
26
|
Sponton CH, Kajimura S. Multifaceted Roles of Beige Fat in Energy Homeostasis Beyond UCP1. Endocrinology 2018; 159:2545-2553. [PMID: 29757365 PMCID: PMC6692864 DOI: 10.1210/en.2018-00371] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/05/2018] [Indexed: 02/07/2023]
Abstract
Beige adipocytes are an inducible form of thermogenic adipose cells that emerge within the white adipose tissue in response to a variety of environmental stimuli, such as chronic cold acclimation. Similar to brown adipocytes that reside in brown adipose tissue depots, beige adipocytes are also thermogenic; however, beige adipocytes possess unique, distinguishing characteristics in their developmental regulation and biological function. This review highlights recent advances in our understanding of beige adipocytes, focusing on the diverse roles of beige fat in the regulation of energy homeostasis that are independent of the canonical thermogenic pathway via uncoupling protein 1.
Collapse
Affiliation(s)
- Carlos Henrique Sponton
- Diabetes Center, University of California, San Francisco, San Francisco, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, San Francisco, California
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California
| | - Shingo Kajimura
- Diabetes Center, University of California, San Francisco, San Francisco, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, San Francisco, California
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California
- Correspondence: Shingo Kajimura, PhD, Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, RMB1023, Box 0669, San Francisco, California 94143. E-mail:
| |
Collapse
|
27
|
Transcriptional control of intestinal cholesterol absorption, adipose energy expenditure and lipid handling by Sortilin. Sci Rep 2018; 8:9006. [PMID: 29899496 PMCID: PMC5998044 DOI: 10.1038/s41598-018-27416-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022] Open
Abstract
The sorting receptor Sortilin functions in the regulation of glucose and lipid metabolism. Dysfunctional lipid uptake, storage, and metabolism contribute to several major human diseases including atherosclerosis and obesity. Sortilin associates with cardiovascular disease; however, the role of Sortilin in adipose tissue and lipid metabolism remains unclear. Here we show that in the low-density lipoprotein receptor-deficient (Ldlr−/−) atherosclerosis model, Sortilin deficiency (Sort1−/−) in female mice suppresses Niemann-Pick type C1-Like 1 (Npc1l1) mRNA levels, reduces body and white adipose tissue weight, and improves brown adipose tissue function partially via transcriptional downregulation of Krüppel-like factor 4 and Liver X receptor. Female Ldlr−/−Sort1−/− mice on a high-fat/cholesterol diet had elevated plasma Fibroblast growth factor 21 and Adiponectin, an adipokine that when reduced is associated with obesity and cardiovascular disease-related factors. Additionally, Sort1 deficiency suppressed cholesterol absorption in both female mice ex vivo intestinal tissue and human colon Caco-2 cells in a similar manner to treatment with the NPC1L1 inhibitor ezetimibe. Together our findings support a novel role of Sortilin in energy regulation and lipid homeostasis in female mice, which may be a potential therapeutic target for obesity and cardiovascular disease.
Collapse
|
28
|
Carobbio S, Guénantin AC, Samuelson I, Bahri M, Vidal-Puig A. Brown and beige fat: From molecules to physiology and pathophysiology. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:37-50. [PMID: 29852279 DOI: 10.1016/j.bbalip.2018.05.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/31/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
Abstract
The adipose organ portrays adipocytes of diverse tones: white, brown and beige, each type with distinct functions. Adipocytes orchestrate their adaptation and expansion to provide storage to excess nutrients, the quick mobilisation of fuel to supply peripheral functional demands, insulation, and, in their thermogenic form, heat generation to maintain core body temperature. Thermogenic adipocytes could be targets for anti-obesity and anti-diabetic therapeutic approaches aiming to restore adipose tissue functionality and increase energy dissipation. However, for thermogenic adipose tissue to become therapeutically relevant, a better understanding of its development and origins, its progenitors and their characteristics and the composition of its niche, is essential. Also crucial is the identification of stimuli and molecules promoting its specific differentiation and activation. Here we highlight the structural/cellular differences between human and rodent brown adipose tissue and discuss how obesity and metabolic complication affects brown and beige cells as well as how they could be targeted to improve their activation and improve global metabolic homeostasis. Finally, we describe the limitations of current research models and the advantages of new emerging approaches.
Collapse
Affiliation(s)
- Stefania Carobbio
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Anne-Claire Guénantin
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Isabella Samuelson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Myriam Bahri
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
29
|
Loss of P2X7 receptor function dampens whole body energy expenditure and fatty acid oxidation. Purinergic Signal 2018; 14:299-305. [PMID: 29754194 DOI: 10.1007/s11302-018-9610-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/01/2018] [Indexed: 12/24/2022] Open
Abstract
The established role of ATP-responsive P2X7 receptor in inflammatory, neurodegenerative, and immune diseases is now expanding to include several aspects of metabolic dysregulation. Indeed, P2X7 receptors are involved in β cell function, insulin secretion, and liability to diabetes, and loss of P2X7 function may increase the risk of hepatic steatosis and disrupt adipogenesis. Recently, body weight gain, abnormal lipid accumulation, adipocyte hyperplasia, increased fat mass, and ectopic fat distribution have been found in P2X7 KO mice. Here, we hypothesized that such clinical picture of dysregulated lipid metabolism might be the result of altered in vivo energy metabolism. By indirect calorimetry, we assessed 24 h of energy expenditure (EE) and respiratory exchange ratio (RER) as quotient of carbohydrate to fat oxidation in P2X7 KO mice. Moreover, we assessed the same parameters in aged-matched WT counterparts that underwent a 7-day treatment with the P2X7 antagonist A804598. We found that loss of P2X7 function elicits a severe decrease of EE that was less pronounced in A804598-treated mice. In parallel, P2X7KO mice show a drastic increase of RER, thus indicating the occurrence of a greater ratio of carbohydrate to fat oxidation. Decreased EE and fat oxidation is predictive of body weight gain, which was here confirmed. Taken together, our data provide evidence that P2X7 loss of function produces defective energy homeostasis that, together with disrupted adipogenesis, might help to explain accumulation of adipose tissue and contribute to disclose the potential role of P2X7 in metabolic diseases.
Collapse
|
30
|
Buttenschøn HN, Elfving B, Nielsen M, Skeldal S, Kaas M, Mors O, Glerup S. Exploring the sortilin related receptor, SorLA, in depression. J Affect Disord 2018; 232:260-267. [PMID: 29499509 DOI: 10.1016/j.jad.2018.02.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/29/2018] [Accepted: 02/16/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Studies of individual biomarkers for depression have shown insufficient sensitivity and specificity for clinical use, and most likely combinations of biomarkers may provide a better signature. The sorting-related receptor with A-type repeats (SorLA) is a well-studied pathogenic factor for Alzheimer's. SorLA belongs to the Vps10p domain receptor family, which also encompasses sortilin and SorCS1-3. All family members have been implicated in neurological and mental disorders. Notably, the SORCS3 gene is genome-wide significantly associated with depression and serum protein levels of sortilin are reduced in depressed individuals. SorLA regulates the activity of neurotrophic factors and cytokines and we hence speculated that SorLA might be implicated in depression. METHODS Serum SorLA levels were measured in two well-defined clinical samples using ELISA. Generalized linear models were used in the statistical analyses. RESULTS We identified a multivariate model to discriminate depressed individuals from healthy controls. Interestingly, the model consisted of serum SorLA levels and additional four predictors: previous depressive episode, stressful life events, serum levels of sortilin and VEGF. However, as an isolated factor, we observed no significant difference in SorLA levels between 140 depressed individuals and 140 healthy controls. Nevertheless, we observed a significant increase in SorLA levels following 12 weeks of treatment with nortriptyline, but not escitalopram. LIMITATIONS The number of biomarkers included in the multivariate model for depression and lack of replication limit our study. CONCLUSIONS Our results suggest SorLA as one of five factors that in combination may support the depression diagnosis, but not as an individual biomarker for depression or treatment response.
Collapse
Affiliation(s)
- Henriette N Buttenschøn
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Marit Nielsen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Sune Skeldal
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; The Lundbeck Foundation Research Center, MIND, Aarhus University, Denmark
| |
Collapse
|
31
|
Mikamo H, Jiang M, Noro M, Suzuki Y, Hiruta N, Unoki-Kubota H, Schneider WJ, Bujo H. Susceptibilities of epicardial and subcutaneous fat tissue for browning-gene expression and diet-induced volume reduction are different. Mol Med Rep 2018; 17:6542-6550. [PMID: 29512723 PMCID: PMC5928636 DOI: 10.3892/mmr.2018.8690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/25/2018] [Indexed: 12/13/2022] Open
Abstract
The upregulation of brown or brown-like beige adipocytes is a potential strategy for the prevention or treatment of diabetes and coronary artery diseases in obese patients. Epicardial adipose tissue (EAT) differs significantly from subcutaneous fat tissue (SAT) in metabolic properties. To investigate properties of EAT further, thermogenesis gene expression was investigated in human autopsy and murine samples, and adipocytes differentiated from EAT mesenchymal cells. Subsequently, analyzed EAT volume alterations were observed to be associated with weight reduction in obese patients by imaging. Gene expression analyses of autopsy samples revealed that UCP‑1 mRNA levels in EAT were significantly increased compared with SAT, and β3‑adrenergic receptor (AR) levels tended to be increased; this finding was verified in comparing EAT with SAT in mice. Browning stimulation of human EAT‑derived MCs increased uncoupling protein‑1 and β3‑AR levels by 3.2 fold‑ and 12.6‑fold compared with SAT‑derived MCs, respectively. Subsequent imaging for EAT volume measurement using multi‑detector computed tomography in 10 obese patients revealed that mean EAT volumes did not significantly decrease following weight loss therapy. The EAT volume alterations were not correlated with weight changes, whereas positive correlations were observed in SAT and visceral adipose tissue. Therefore, the studies in man and mouse on EAT properties demonstrated that susceptibilities of EAT and SAT for browning‑gene expression and diet‑induced volume reduction were grossly different. The data suggest a potential association of EAT with local thermogenetic and metabolic homeostasis in cardiac and/or cardiovascular cells, in conjunction with systemic energy metabolism.
Collapse
Affiliation(s)
- Hiroshi Mikamo
- Department of Cardiology, Toho University Sakura Medical Center, Sakura, Chiba 285‑8741, Japan
| | - Meizi Jiang
- Department of Clinical‑Laboratory and Experimental‑Research Medicine, Toho University Sakura Medical Center, Sakura, Chiba 285‑8741, Japan
| | - Mahito Noro
- Department of Cardiology, Toho University Sakura Medical Center, Sakura, Chiba 285‑8741, Japan
| | - Yasuo Suzuki
- Department of Internal Medicine, Toho University Sakura Medical Center, Sakura, Chiba 285‑8741, Japan
| | - Nobuyuki Hiruta
- Department of Pathology, Toho University Sakura Medical Center, Sakura, Chiba 285‑8741, Japan
| | - Hiroyuki Unoki-Kubota
- Department of Diabetic Complications, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Wolfgang J Schneider
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1090, Austria
| | - Hideaki Bujo
- Department of Clinical‑Laboratory and Experimental‑Research Medicine, Toho University Sakura Medical Center, Sakura, Chiba 285‑8741, Japan
| |
Collapse
|
32
|
Chechi K, van Marken Lichtenbelt W, Richard D. Brown and beige adipose tissues: phenotype and metabolic potential in mice and men. J Appl Physiol (1985) 2018; 124:482-496. [PMID: 28302705 PMCID: PMC5867364 DOI: 10.1152/japplphysiol.00021.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
With the recent rediscovery of brown fat in adult humans, our outlook on adipose tissue biology has undergone a paradigm shift. While we attempt to identify, recruit, and activate classic brown fat stores in humans, identification of beige fat has also raised the possibility of browning our white fat stores. Whether such transformation of human white fat depots can be achieved to enhance the whole body oxidative potential remains to be seen. Evidence to date, however, largely points toward a major oxidative role only for classic brown fat depots, at least in rodents. White fat stores seem to provide the main fuel for sustaining thermogenesis via lipolysis. Interestingly, molecular markers consistent with both classic brown and beige fat identity can be observed in human supraclavicular depot, thereby complicating the discussion on beige fat in humans. Here, we review the recent advances made in our understanding of brown and beige fat in humans and mice. We further provide an overview of their plausible physiological relevance to whole body energy metabolism.
Collapse
Affiliation(s)
- Kanta Chechi
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Ville de Québec, Quebec , Canada
| | - Wouter van Marken Lichtenbelt
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Ville de Québec, Quebec , Canada
| |
Collapse
|
33
|
Microfluidic systems for studying dynamic function of adipocytes and adipose tissue. Anal Bioanal Chem 2017; 410:791-800. [PMID: 29214530 DOI: 10.1007/s00216-017-0741-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/12/2017] [Accepted: 11/02/2017] [Indexed: 01/03/2023]
Abstract
Recent breakthroughs in organ-on-a-chip and related technologies have highlighted the extraordinary potential for microfluidics to not only make lasting impacts in the understanding of biological systems but also to create new and important in vitro culture platforms. Adipose tissue (fat), in particular, is one that should be amenable to microfluidic mimics of its microenvironment. While the tissue was traditionally considered important only for energy storage, it is now understood to be an integral part of the endocrine system that secretes hormones and responds to various stimuli. As such, adipocyte function is central to the understanding of pathological conditions such as obesity, diabetes, and metabolic syndrome. Despite the importance of the tissue, only recently have significant strides been made in studying dynamic function of adipocytes or adipose tissues on microfluidic devices. In this critical review, we highlight new developments in the special class of microfluidic systems aimed at culture and interrogation of adipose tissue, a sub-field of microfluidics that we contend is only in its infancy. We close by reflecting on these studies as we forecast a promising future, where microfluidic technologies should be capable of mimicking the adipose tissue microenvironment and provide novel insights into its physiological roles in the normal and diseased states. Graphical abstract This critical review focuses on recent developments and challenges in applying microfluidic systems to the culture and analysis of adipocytes and adipose tissue.
Collapse
|
34
|
Villarroya F, Gavaldà-Navarro A, Peyrou M, Villarroya J, Giralt M. The Lives and Times of Brown Adipokines. Trends Endocrinol Metab 2017; 28:855-867. [PMID: 29113711 DOI: 10.1016/j.tem.2017.10.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/13/2022]
Abstract
Brown adipose tissue (BAT) is responsible for adaptive non-shivering thermogenesis. Moreover, brown fat secretes regulatory factors, so-called brown adipokines, that have autocrine, paracrine, and endocrine actions. Brown adipokines are either polypeptides or nonpeptidic molecules including lipid molecules and microRNAs. The secretory properties of brown fat are essential for tissue remodeling adaptations to thermogenic necessities. The endocrine properties of brown adipokines are thought to contribute to the association between BAT activity and a healthy metabolic profile in relation to glucose and lipid homeostasis. The identification and characterization of brown adipokines may allow the discovery of circulating biomarkers of BAT activity in humans, and will lead to the development of candidate tools for therapeutic interventions in metabolic diseases.
Collapse
Affiliation(s)
- Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain.
| | - Aleix Gavaldà-Navarro
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Marion Peyrou
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Joan Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| |
Collapse
|
35
|
Scheideler M, Herzig S, Georgiadi A. Endocrine and autocrine/paracrine modulators of brown adipose tissue mass and activity as novel therapeutic strategies against obesity and type 2 diabetes. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0043/hmbci-2017-0043.xml. [PMID: 28850545 DOI: 10.1515/hmbci-2017-0043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/28/2017] [Indexed: 12/17/2022]
Abstract
The dramatically increasing world-wide prevalence of obesity is recognized as a risk factor for the development of various diseases. The growing research on the role of adipose tissue in controlling energy homeostasis and insulin sensitivity has revealed that the promotion of brown adipose tissue (BAT) activity and the browning of white adipose tissue (WAT) leads to multiple health benefits and prevents obesity and type 2 diabetes (T2D). Inducible thermogenic adipocytes do exist in adult humans and are linked with increased energy combustion and lower body fat mass. Thus brown adipocytes are currently placed at the center of attention for novel therapeutic strategies against metabolic diseases such as obesity and diabetes. Besides the classical, norepinephrine-mediated sympathetic recruitment and activation of thermogenic adipocytes, a number of novel circulating factors have been recently identified to have a positive or negative impact on thermogenic adipocyte formation and activity. In this review their mechanism of action and the plausible therapeutic applications will be summarized and discussed.
Collapse
|
36
|
Nohara A. sLR11 as a novel predictor of vascular calcification. Atherosclerosis 2017; 265:242-243. [PMID: 28823527 DOI: 10.1016/j.atherosclerosis.2017.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 07/25/2017] [Indexed: 11/27/2022]
Affiliation(s)
- Atsushi Nohara
- Cholesterol Clinic, Department of Cardiology, Kanazawa University Hospital, Kanazawa University Health Service Center, Takara-machi 13-1, Kanazawa 920-8641, Japan.
| |
Collapse
|
37
|
Gusarova V, Banfi S, Alexa-Braun CA, Shihanian LM, Mintah IJ, Lee JS, Xin Y, Su Q, Kamat V, Cohen JC, Hobbs HH, Zambrowicz B, Yancopoulos GD, Murphy AJ, Gromada J. ANGPTL8 Blockade With a Monoclonal Antibody Promotes Triglyceride Clearance, Energy Expenditure, and Weight Loss in Mice. Endocrinology 2017; 158:1252-1259. [PMID: 28204173 PMCID: PMC5460832 DOI: 10.1210/en.2016-1894] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/06/2017] [Indexed: 01/05/2023]
Abstract
Angiopoietin-like protein (ANGPTL)8 is a negative regulator of lipoprotein lipase-mediated plasma triglyceride (TG) clearance. In this study, we describe a fully human monoclonal antibody (REGN3776) that binds monkey and human ANGPTL8 with high affinity. Inhibition of ANGPTL8 with REGN3776 in humanized ANGPTL8 mice decreased plasma TGs and increased lipoprotein lipase activity. Additionally, REGN3776 reduced body weight and fat content. The reduction in body weight was secondary to increased energy expenditure. Finally, single administration of REGN3776 normalized plasma TGs in dyslipidemic cynomolgus monkeys. In conclusion, we show that blockade of ANGPTL8 with monoclonal antibody strongly reduced plasma TGs in mice and monkeys. These data suggest that inhibition of ANGPTL8 may provide a new therapeutic avenue for the treatment of dyslipidemia with beneficial effects on body weight.
Collapse
Affiliation(s)
| | - Serena Banfi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | | | | | | | - Joseph S. Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, 10591
| | - Yurong Xin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, 10591
| | - Qi Su
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, 10591
| | - Vishal Kamat
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, 10591
| | - Jonathan C. Cohen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Helen H. Hobbs
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
- Howard Hughes Medical Institute, Dallas, Texas 75390
| | | | | | | | - Jesper Gromada
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, 10591
| |
Collapse
|
38
|
Modica S, Wolfrum C. The dual role of BMP4 in adipogenesis and metabolism. Adipocyte 2017; 6:141-146. [PMID: 28425843 PMCID: PMC5477726 DOI: 10.1080/21623945.2017.1287637] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/12/2017] [Accepted: 01/22/2017] [Indexed: 12/13/2022] Open
Abstract
BMP4 has a well-established role in triggering commitment of mesenchymal stem cells into the osteogenic and adipogenic linage. We recently described an additional dual function in adipogenesis: after promoting the formation of both white and brown pre-adipocytes, Bmp4 drives terminal differentiation into mature white rather than brown fat cells. Besides this, Bmp4 seems to have a dual role in metabolism either promoting or repressing oxidative metabolism in a cell context dependent manner.
Collapse
Affiliation(s)
- Salvatore Modica
- a Swiss Federal Institute of Technology, Department of Health Science , Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology , Schwerzenbach , Switzerland
| | - Christian Wolfrum
- a Swiss Federal Institute of Technology, Department of Health Science , Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology , Schwerzenbach , Switzerland
| |
Collapse
|
39
|
Kristensen MM, Davidsen PK, Vigelsø A, Hansen CN, Jensen LJ, Jessen N, Bruun JM, Dela F, Helge JW. miRNAs in human subcutaneous adipose tissue: Effects of weight loss induced by hypocaloric diet and exercise. Obesity (Silver Spring) 2017; 25:572-580. [PMID: 28158925 DOI: 10.1002/oby.21765] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/29/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Obesity is central in the development of insulin resistance. However, the underlying mechanisms still need elucidation. Dysregulated microRNAs (miRNAs; post-transcriptional regulators) in adipose tissue may present an important link. METHODS The miRNA expression in subcutaneous adipose tissue from 19 individuals with severe obesity (10 women and 9 men) before and after a 15-week weight loss intervention was studied using genome-wide microarray analysis. The microarray results were validated with RT-qPCR, and pathway enrichment analysis of in silico predicted targets was performed to elucidate the biological consequences of the miRNA dysregulation. Lastly, the messenger RNA (mRNA) and/or protein expression of multiple predicted targets as well as several proteins involved in lipolysis were investigated. RESULTS The intervention led to upregulation of miR-29a-3p and miR-29a-5p and downregulation of miR-20b-5p. The mRNA and protein expression of predicted targets was not significantly affected by the intervention. However, negative correlations between miR-20b-5p and the protein levels of its predicted target, acyl-CoA synthetase long-chain family member 1, were observed. Several other miRNA-target relationships correlated negatively, indicating possible miRNA regulation, including miR-29a-3p and lipoprotein lipase mRNA levels. Proteins involved in lipolysis were not affected by the intervention. CONCLUSIONS Weight loss influenced several miRNAs, some of which were negatively correlated with predicted targets. These dysregulated miRNAs may affect adipocytokine signaling and forkhead box protein O signaling.
Collapse
Affiliation(s)
- Malene M Kristensen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter K Davidsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Vigelsø
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina N Hansen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars J Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jens M Bruun
- Randers Regional Hospital, Medical Department M, Randers, Denmark
- Department of Nutrition, Exercise, and Sports (NEXS), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Dela
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Geriatrics, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Jørn W Helge
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Soluble form of LR11 is highly increased in the vitreous fluids of patients with idiopathic epiretinal membrane. Graefes Arch Clin Exp Ophthalmol 2017; 255:885-891. [DOI: 10.1007/s00417-017-3585-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/24/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022] Open
|
41
|
Abstract
Brown adipose tissue (BAT) is the main site of adaptive thermogenesis and experimental studies have associated BAT activity with protection against obesity and metabolic diseases, such as type 2 diabetes mellitus and dyslipidaemia. Active BAT is present in adult humans and its activity is impaired in patients with obesity. The ability of BAT to protect against chronic metabolic disease has traditionally been attributed to its capacity to utilize glucose and lipids for thermogenesis. However, BAT might also have a secretory role, which could contribute to the systemic consequences of BAT activity. Several BAT-derived molecules that act in a paracrine or autocrine manner have been identified. Most of these factors promote hypertrophy and hyperplasia of BAT, vascularization, innervation and blood flow, processes that are all associated with BAT recruitment when thermogenic activity is enhanced. Additionally, BAT can release regulatory molecules that act on other tissues and organs. This secretory capacity of BAT is thought to be involved in the beneficial effects of BAT transplantation in rodents. Fibroblast growth factor 21, IL-6 and neuregulin 4 are among the first BAT-derived endocrine factors to be identified. In this Review, we discuss the current understanding of the regulatory molecules (the so-called brown adipokines or batokines) that are released by BAT that influence systemic metabolism and convey the beneficial metabolic effects of BAT activation. The identification of such adipokines might also direct drug discovery approaches for managing obesity and its associated chronic metabolic diseases.
Collapse
Affiliation(s)
- Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Rubén Cereijo
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Joan Villarroya
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| |
Collapse
|
42
|
Schmidt V, Subkhangulova A, Willnow TE. Sorting receptor SORLA: cellular mechanisms and implications for disease. Cell Mol Life Sci 2016; 74:1475-1483. [PMID: 27832290 PMCID: PMC5357279 DOI: 10.1007/s00018-016-2410-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 12/21/2022]
Abstract
Sorting-related receptor with A-type repeats (SORLA) is an intracellular sorting receptor that directs cargo proteins, such as kinases, phosphatases, and signaling receptors, to their correct location within the cell. The activity of SORLA assures proper function of cells and tissues, and receptor dysfunction is the underlying cause of common human malignancies, including Alzheimer’s disease, atherosclerosis, and obesity. Here, we discuss the molecular mechanisms that govern sorting of SORLA and its cargo in multiple cell types, and why genetic defects in this receptor results in devastating diseases.
Collapse
Affiliation(s)
- Vanessa Schmidt
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125, Berlin, Germany.
| | - Aygul Subkhangulova
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
43
|
Andersen OM, Rudolph IM, Willnow TE. Risk factor SORL1: from genetic association to functional validation in Alzheimer's disease. Acta Neuropathol 2016; 132:653-665. [PMID: 27638701 PMCID: PMC5073117 DOI: 10.1007/s00401-016-1615-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/12/2016] [Accepted: 09/05/2016] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) represents one of the most dramatic threats to healthy aging and devising effective treatments for this devastating condition remains a major challenge in biomedical research. Much has been learned about the molecular concepts that govern proteolytic processing of the amyloid precursor protein to amyloid-β peptides (Aβ), and how accelerated accumulation of neurotoxic Aβ peptides underlies neuronal cell death in rare familial but also common sporadic forms of this disease. Out of a plethora of proposed modulators of amyloidogenic processing, one protein emerged as a key factor in AD pathology, a neuronal sorting receptor termed SORLA. Independent approaches using human genetics, clinical pathology, or exploratory studies in animal models all converge on this receptor that is now considered a central player in AD-related processes by many. This review will provide a comprehensive overview of the evidence implicating SORLA-mediated protein sorting in neurodegenerative processes, and how receptor gene variants in the human population impair functional receptor expression in sporadic but possibly also in autosomal-dominant forms of AD.
Collapse
Affiliation(s)
- Olav M Andersen
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Ole Worms Alle 3, Aarhus C, 8000, Aarhus, Denmark.
| | - Ina-Maria Rudolph
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Strasse 10, 13125, Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Strasse 10, 13125, Berlin, Germany.
| |
Collapse
|
44
|
Levels of the soluble LDL receptor-relative LR11 decrease in overweight individuals with type 2 diabetes upon diet-induced weight loss. Atherosclerosis 2016; 254:67-72. [DOI: 10.1016/j.atherosclerosis.2016.09.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/15/2016] [Accepted: 09/22/2016] [Indexed: 12/31/2022]
|
45
|
Glastonbury C, Viñuela A, Buil A, Halldorsson G, Thorleifsson G, Helgason H, Thorsteinsdottir U, Stefansson K, Dermitzakis E, Spector T, Small K. Adiposity-Dependent Regulatory Effects on Multi-tissue Transcriptomes. Am J Hum Genet 2016; 99:567-579. [PMID: 27588447 PMCID: PMC5011064 DOI: 10.1016/j.ajhg.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/01/2016] [Indexed: 10/25/2022] Open
Abstract
Obesity is a global epidemic that is causally associated with a range of diseases, including type 2 diabetes and cardiovascular disease, at the population-level. However, there is marked heterogeneity in obesity-related outcomes among individuals. This might reflect genotype-dependent responses to adiposity. Given that adiposity, measured by BMI, is associated with widespread changes in gene expression and regulatory variants mediate the majority of known complex trait loci, we sought to identify gene-by-BMI (G × BMI) interactions on the regulation of gene expression in a multi-tissue RNA-sequencing (RNA-seq) dataset from the TwinsUK cohort (n = 856). At a false discovery rate of 5%, we identified 16 cis G × BMI interactions (top cis interaction: CHURC1, rs7143432, p = 2.0 × 10(-12)) and one variant regulating 53 genes in trans (top trans interaction: ZNF423, rs3851570, p = 8.2 × 10(-13)), all in adipose tissue. The interactions were adipose-specific and enriched for variants overlapping adipocyte enhancers, and regulated genes were enriched for metabolic and inflammatory processes. We replicated a subset of the interactions in an independent adipose RNA-seq dataset (deCODE genetics, n = 754). We also confirmed the interactions with an alternate measure of obesity, dual-energy X-ray absorptiometry (DXA)-derived visceral-fat-volume measurements, in a subset of TwinsUK individuals (n = 682). The identified G × BMI regulatory effects demonstrate the dynamic nature of gene regulation and reveal a functional mechanism underlying the heterogeneous response to obesity. Additionally, we have provided a web browser allowing interactive exploration of the dataset, including of association between expression, BMI, and G × BMI regulatory effects in four tissues.
Collapse
|
46
|
Abstract
Excess and ectopic fat accumulation in obesity is a major risk factor for developing hyperlipidemia, type 2 diabetes and cardiovascular disease. The activation of brown and/or beige adipocytes is a promising target for the treatment of metabolic disorders as the combustion of excess energy by these thermogenic adipocytes may help losing weight and improving plasma parameters including triglyceride, cholesterol and glucose levels. The regulation of heat production by thermogenic adipose tissues is based on a complex crosstalk between the autonomous nervous system, intracellular and secreted factors. This multifaceted alignment regulates thermogenic demands to environmental circumstances in dependence on available energy resources. This review summarizes the current knowledge how thermogenic tissues can be targeted to combat the burden of diseases with a special focus on lipid metabolism and diseases related to lipoprotein metabolism.
Collapse
Affiliation(s)
- Christian Schlein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
47
|
Schmidt V, Schulz N, Yan X, Schürmann A, Kempa S, Kern M, Blüher M, Poy MN, Olivecrona G, Willnow TE. SORLA facilitates insulin receptor signaling in adipocytes and exacerbates obesity. J Clin Invest 2016; 126:2706-20. [PMID: 27322061 DOI: 10.1172/jci84708] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 04/29/2016] [Indexed: 12/22/2022] Open
Abstract
In humans, genetic variation of sortilin-related receptor, L(DLR class) A repeats containing (SORL1), which encodes the intracellular sorting receptor SORLA, is a major genetic risk factor for familial and sporadic forms of Alzheimer's disease. Recent GWAS analysis has also associated SORL1 with obesity in humans and in mouse models, suggesting that this receptor may play a role in regulating metabolism. Here, using mouse models with genetic loss or tissue-specific overexpression of SORLA as well as data from obese human subjects, we observed a gene-dosage effect that links SORLA expression to obesity and glucose tolerance. Overexpression of human SORLA in murine adipose tissue blocked hydrolysis of triacylglycerides and caused excessive adiposity. In contrast, Sorl1 gene inactivation in mice accelerated breakdown of triacylglycerides in adipocytes and protected animals from diet-induced obesity. We then identified the underlying molecular mechanism whereby SORLA promotes insulin-induced suppression of lipolysis in adipocytes. Specifically, we determined that SORLA acts as a sorting factor for the insulin receptor (IR) that redirects internalized receptor molecules from endosomes to the plasma membrane, thereby enhancing IR surface expression and strengthening insulin signal reception in target cells. Our findings provide a molecular mechanism for the association of SORL1 with human obesity and confirm a genetic link between neurodegeneration and metabolism that converges on the receptor SORLA.
Collapse
|
48
|
Circulating soluble form of LR11, a regulator of smooth muscle cell migration, is a novel marker for intima-media thickness of carotid arteries in type 2 diabetes. Clin Chim Acta 2016; 457:137-41. [PMID: 27095609 DOI: 10.1016/j.cca.2016.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/18/2016] [Accepted: 04/14/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Smooth muscle cell (SMC) migration from the media to the intima, a process affecting plaque stability in advanced-stage atherosclerosis, is under the control of LR11. To delineate the clinical significance of the circulating soluble form of LR11 (sLR11) in patients with type 2 diabetes (T2D), we analyzed the correlation of sLR11 levels with intima-media thickness (IMT) of carotid arteries. METHODS Plasma sLR11 levels were measured in 165 patients with T2D (mean age 56.2±10.4 y, 58.2% males, and BMI 24.6±3.6) by ELISA. Averaged IMT levels of common carotid arteries were determined by ultrasonography. RESULTS Circulating sLR11 levels were 9.8±3.5ng/ml, and correlated positively with the classical atherosclerosis risk factors age, sex, systolic blood pressure, low-density lipoprotein-cholesterol (LDL-C), fasting plasma-glucose (FPG), and glycosylated hemoglobin. Multivariate linear regression analysis indicated that only FPG was associated with sLR11; sLR11 correlated positively with IMT, together with age and FPG, but less with LDL-C. Among the serum risk factors for IMT, multivariate linear regression analysis uncovered that sLR11 was independently associated with IMT. Subsequent logistic analysis revealed that FPG correlated best with IMT values at a cut-off of 0.80mm and sLR11 at a cut-off of 0.90mm, respectively, while LDL-C showed lower discriminatory power at any IMT cut-off values. CONCLUSION Increased sLR11 concentrations are highly associated with increased IMT as well as with FPG in middle-aged, non-obese patients with T2D. Circulating sLR11 may be a novel marker representing the pathophysiology of intimal SMCs in patients with T2D.
Collapse
|
49
|
Petrov PD, Palou A, Bonet ML, Ribot J. Cell-Autonomous Brown-Like Adipogenesis of Preadipocytes From Retinoblastoma Haploinsufficient Mice. J Cell Physiol 2016; 231:1941-52. [PMID: 26727985 DOI: 10.1002/jcp.25299] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/04/2016] [Indexed: 12/20/2022]
Abstract
Mechanisms behind the emergence of brown adipocyte-like (brite or beige) adipocytes within white adipose tissue (WAT) are of interest. Retinoblastoma protein gene (Rb) haploinsufficiency associates in mice with improved metabolic regulation linked to a greater capacity for fatty acid oxidation and thermogenesis in WAT. We aimed to explain a feasible mechanism of WAT-to-BAT remodeling in this model. Differentiated primary adipocytes and Sca1-positive preadipocytes derived from adipose depots of Rb(+/-) mice and wild-type siblings were compared. Primary white Rb(+/-) adipocytes displayed under basal conditions increased glucose uptake and an enhanced expression of brown adipocyte-related genes (Pparg, Ppargc1a, Ppargc1b, Prdm16, Cpt1b) but not of purported beige/brite transcriptional markers (Cd137, Tmem26, Tbx1, Slc27a1, Hoxc9, Shox2). Lack of induction of beige markers phenocopied results in WAT of adult Rb(+/-) mice. Flow cytometry analysis evidenced an increased number of preadipocytes in WAT depots of Rb(+/-) mice. Sca1(+) preadipocytes from WAT of Rb(+/-) mice displayed increased gene expression of several transcription factors common to the brown and beige adipogenic programs (Prdm16, Pparg, Ppargc1a) and of receptors of bone morphogenetic proteins (BMPs); however, among the recently proposed beige markers, only Tbx1 was upregulated. Adult Rb(+/-) mice had increased circulating levels of BMP7. These results indicate that preadipose cells resident in WAT depots of Rb(+/-) mice retain an increased capacity for brown-like adipogenesis that appears to be different from beige adipogenesis, and suggest that the contribution of these precursors to the Rb(+/-) adipose phenotype is driven, at least in part, by interaction with BMP7 pathways. J. Cell. Physiol. 231: 1941-1952, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Petar D Petrov
- Laboratory of Molecular Biology, Nutrition and Biotechnology-Nutrigenomics, Universitat de les Illes Balears, Palma de Mallorca, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology-Nutrigenomics, Universitat de les Illes Balears, Palma de Mallorca, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - M Luisa Bonet
- Laboratory of Molecular Biology, Nutrition and Biotechnology-Nutrigenomics, Universitat de les Illes Balears, Palma de Mallorca, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - Joan Ribot
- Laboratory of Molecular Biology, Nutrition and Biotechnology-Nutrigenomics, Universitat de les Illes Balears, Palma de Mallorca, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| |
Collapse
|