1
|
Reguero M, Reglero G, Quintela JC, Ramos-Ruiz R, Ramírez de Molina A, Gómez de Cedrón M. Silymarin-Enriched Extract from Milk Thistle Activates Thermogenesis in a Preclinical Model of High-Fat-Diet-Induced Obesity to Relieve Systemic Meta-Inflammation. Nutrients 2024; 16:4166. [PMID: 39683558 DOI: 10.3390/nu16234166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Obesity and aging are associated with the progressive loss of brown adipose tissue (BAT), an increase in visceral white adipose tissue (vWAT), and a reduction in subcutaneous white adipose tissue (sWAT). The progressive expansion of visceral obesity promotes a low grade of systemic chronic inflammation (meta-inflammation), contributing to the onset of comorbidities such as type 2 diabetes mellitus (T2DM), metabolic syndrome, and even cancer. Thus, preserving the thermogenic activity of adipose tissue and improving the metabolic flexibility of sWAT could be an effective strategy to prevent the development of metabolic chronic diseases and promote healthy aging. Precision nutrition has emerged as a complementary approach to control the metabolic alterations associated with unhealthy obesity and aging. In a previous work, we described that a silymarin-enriched extract from milk thistle (Mthistle) increased markers of browning and thermogenesis in vitro in human differentiated adipocytes (SGBS). OBJECTIVES/METHODS Therefore, this study aims to evaluate the potential of Mthistle to activate thermogenesis in a preclinical model of high-fat diet (HFD)-induced obesity (DIO). RESULTS Our results demonstrate that Mthistle increases systemic energy expenditure (EE), preserves body temperature after cold exposure, improves insulin resistance, and reduces inflammatory markers in WAT. CONCLUSIONS Based on these results, silymarin-enriched extract from Mthistle may be proposed as a nutraceutical for the management of metabolic chronic diseases and/or accelerated aging.
Collapse
Affiliation(s)
- Marina Reguero
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- NATAC BIOTECH, Electronica 7, E28923 Madrid, Spain
| | - Guillermo Reglero
- Production and Characterization of Novel Foods Department, Institute of Food Science Research CIAL, CEI UAM + CSIC, E28049 Madrid, Spain
| | | | - Ricardo Ramos-Ruiz
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Marta Gómez de Cedrón
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- Cell Metabolism Unit, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| |
Collapse
|
2
|
Hu Y, Huang Y, Jiang Y, Weng L, Cai Z, He B. The Different Shades of Thermogenic Adipose Tissue. Curr Obes Rep 2024; 13:440-460. [PMID: 38607478 DOI: 10.1007/s13679-024-00559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF REVIEW By providing a concise overview of adipose tissue types, elucidating the regulation of adipose thermogenic capacity in both physiological contexts and chronic wasting diseases (a protracted hypermetabolic state that precipitates sustained catabolism and consequent progressive corporeal atrophy), and most importantly, delving into the ongoing discourse regarding the role of adipose tissue thermogenic activation in chronic wasting diseases, this review aims to provide researchers with a comprehensive understanding of the field. RECENT FINDINGS Adipose tissue, traditionally classified as white, brown, and beige (brite) based on its thermogenic activity and potential, is intricately regulated by complex mechanisms in response to exercise or cold exposure. This regulation is adipose depot-specific and dependent on the duration of exposure. Excessive thermogenic activation of adipose tissue has been observed in chronic wasting diseases and has been considered a pathological factor that accelerates disease progression. However, this conclusion may be confounded by the detrimental effects of excessive lipolysis. Recent research also suggests that such activation may play a beneficial role in the early stages of chronic wasting disease and provide potential therapeutic effects. A more comprehensive understanding of the changes in adipose tissue thermogenesis under physiological and pathological conditions, as well as the underlying regulatory mechanisms, is essential for the development of novel interventions to improve health and prevent disease.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yijie Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yangjing Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lvkan Weng
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
3
|
Luca T, Pezzino S, Puleo S, Castorina S. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 2024; 22:764. [PMID: 39143643 PMCID: PMC11323604 DOI: 10.1186/s12967-024-05547-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024] Open
Abstract
Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy.
| | | | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
4
|
Karanfil AS, Louis F, Sowa Y, Matsusaki M. Cationic polymer effect on brown adipogenic induction of dedifferentiated fat cells. Mater Today Bio 2024; 27:101157. [PMID: 39113911 PMCID: PMC11304885 DOI: 10.1016/j.mtbio.2024.101157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Obesity and its associated comorbidities place a substantial burden on public health. Given the considerable potential of brown adipose tissue in addressing metabolic disorders that contribute to dysregulation of the body's energy balance, this area is an intriguing avenue for research. This study aimed to assess the impact of various polymers, including collagen type I, fibronectin, laminin, gelatin, gellan gum, and poly-l-lysine (PLL), on the in vitro brown adipogenic differentiation of dedifferentiated fat cells within a fibrin gel matrix. The findings, obtained through RT-qPCR, immunofluorescent imaging, ELISA assay, and mitochondria assessment, revealed that PLL exhibited a significant browning-inducing effect. Compared to fibrin-only brown-like drops after two weeks of incubation in brown adipogenic medium, PLL showed 6 (±3) times higher UCP1 gene expression, 5 (±2) times higher UCP1 concentration by ELISA assay, and 2 (±1) times higher mitochondrial content. This effect can be attributed to PLL's electrostatic properties, which potentially facilitate the cellular uptake of crucial brown adipogenic inducers such as the thyroid hormone, triiodothyronine (T3), and insulin from the induction medium.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Osaka University, Japan
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Osaka University, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Osaka University, Japan
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Osaka University, Japan
| |
Collapse
|
5
|
Rendine M, Cocci P, de Vivo L, Bellesi M, Palermo FA. Effects of Chronic Sleep Restriction on Transcriptional Sirtuin 1 Signaling Regulation in Male Mice White Adipose Tissue. Curr Issues Mol Biol 2024; 46:2144-2154. [PMID: 38534754 DOI: 10.3390/cimb46030138] [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: 02/08/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Chronic sleep restriction (CSR) is a prevalent issue in modern society that is associated with several pathological states, ranging from neuropsychiatric to metabolic diseases. Despite its known impact on metabolism, the specific effects of CSR on the molecular mechanisms involved in maintaining metabolic homeostasis at the level of white adipose tissue (WAT) remain poorly understood. Therefore, this study aimed to investigate the influence of CSR on sirtuin 1 (SIRT1) and the peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway in the WAT of young male mice. Both genes interact with specific targets involved in multiple metabolic processes, including adipocyte differentiation, browning, and lipid metabolism. The quantitative PCR (qPCR) results demonstrated a significant upregulation of SIRT-1 and some of its target genes associated with the transcriptional regulation of lipid homeostasis (i.e., PPARα, PPARγ, PGC-1α, and SREBF) and adipose tissue development (i.e., leptin, adiponectin) in CSR mice. On the contrary, DNA-binding transcription factors (i.e., CEBP-β and C-myc), which play a pivotal function during the adipogenesis process, were found to be down-regulated. Our results also suggest that the induction of SIRT1-dependent molecular pathways prevents weight gain. Overall, these findings offer new, valuable insights into the molecular adaptations of WAT to CSR, in order to support increased energy demand due to sleep loss.
Collapse
Affiliation(s)
- Marco Rendine
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133 Milan, Italy
| | - Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Luisa de Vivo
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy
| | - Michele Bellesi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1QU, UK
| | | |
Collapse
|
6
|
Alemany M. The Metabolic Syndrome, a Human Disease. Int J Mol Sci 2024; 25:2251. [PMID: 38396928 PMCID: PMC10888680 DOI: 10.3390/ijms25042251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review focuses on the question of metabolic syndrome (MS) being a complex, but essentially monophyletic, galaxy of associated diseases/disorders, or just a syndrome of related but rather independent pathologies. The human nature of MS (its exceptionality in Nature and its close interdependence with human action and evolution) is presented and discussed. The text also describes the close interdependence of its components, with special emphasis on the description of their interrelations (including their syndromic development and recruitment), as well as their consequences upon energy handling and partition. The main theories on MS's origin and development are presented in relation to hepatic steatosis, type 2 diabetes, and obesity, but encompass most of the MS components described so far. The differential effects of sex and its biological consequences are considered under the light of human social needs and evolution, which are also directly related to MS epidemiology, severity, and relations with senescence. The triggering and maintenance factors of MS are discussed, with especial emphasis on inflammation, a complex process affecting different levels of organization and which is a critical element for MS development. Inflammation is also related to the operation of connective tissue (including the adipose organ) and the widely studied and acknowledged influence of diet. The role of diet composition, including the transcendence of the anaplerotic maintenance of the Krebs cycle from dietary amino acid supply (and its timing), is developed in the context of testosterone and β-estradiol control of the insulin-glycaemia hepatic core system of carbohydrate-triacylglycerol energy handling. The high probability of MS acting as a unique complex biological control system (essentially monophyletic) is presented, together with additional perspectives/considerations on the treatment of this 'very' human disease.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
7
|
Jalloul W, Moscalu M, Moscalu R, Jalloul D, Grierosu IC, Gutu M, Haba D, Mocanu V, Gutu MM, Stefanescu C. Are MTV and TLG Accurate for Quantifying the Intensity of Brown Adipose Tissue Activation? Biomedicines 2024; 12:151. [PMID: 38255256 PMCID: PMC10813038 DOI: 10.3390/biomedicines12010151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Recent research has suggested that one novel mechanism of action for anti-obesity medications is to stimulate the activation of brown adipose tissue (BAT). 18FDG PET/CT remains the gold standard for defining and quantifying BAT. SUVmax is the most often used quantification tool in clinical practice. However, this parameter does not reflect the entire BAT volume. As a potential method for precisely evaluating BAT, we have utilised metabolic tumour volume (MTV) and total lesion glycolysis (TLG) to answer the question: Are MTV and TLG accurate in quantifying the intensity of BAT activation? After analysing the total number of oncological 18F-FDG PET/CT scans between 2021-2023, we selected patients with active BAT. Based on the BAT SUVmax, the patients were divided into BAT-moderate activation (MA) vs. BAT-high activation (HA). Furthermore, we statistically analysed the accuracy of TLG and MTV in assessing BAT activation intensity. The results showed that both parameters increased their predictive value regarding BAT activation, and presented a significantly high sensitivity and specificity for the correct classification of BAT activation intensity. To conclude, these parameters could be important indicators with increased accuracy for classifying BAT expression, and could bring additional information about the volume of BAT to complement the limitations of the SUVmax.
Collapse
Affiliation(s)
- Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Roxana Moscalu
- Manchester Academic Health Science Centre, Cell Matrix Biology and Regenerative Medicine, The University of Manchester, Manchester M13 9PT, UK;
| | - Despina Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| | - Irena Cristina Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| | - Mihaela Gutu
- County Hospital of Emergency “Saint John the New”, 720224 Suceava, Romania; (M.G.); (M.M.G.)
| | - Danisia Haba
- Department 1 Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Veronica Mocanu
- Department of Morpho-Functional Sciences (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Mihai Marius Gutu
- County Hospital of Emergency “Saint John the New”, 720224 Suceava, Romania; (M.G.); (M.M.G.)
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (W.J.); (D.J.); (I.C.G.); (C.S.)
| |
Collapse
|
8
|
Winarto J, Song DG, Pan CH. The Role of Fucoxanthin in Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:ijms24098203. [PMID: 37175909 PMCID: PMC10179653 DOI: 10.3390/ijms24098203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Chronic liver disease (CLD) has emerged as a leading cause of human deaths. It caused 1.32 million deaths in 2017, which affected men more than women by a two-to-one ratio. There are various causes of CLD, including obesity, excessive alcohol consumption, and viral infection. Among them, non-alcoholic fatty liver disease (NAFLD), one of obesity-induced liver diseases, is the major cause, representing the cause of more than 50% of cases. Fucoxanthin, a carotenoid mainly found in brown seaweed, exhibits various biological activities against NAFLD. Its role in NAFLD appears in several mechanisms, such as inducing thermogenesis in mitochondrial homeostasis, altering lipid metabolism, and promoting anti-inflammatory and anti-oxidant activities. The corresponding altered signaling pathways are the β3-adorenarine receptor (β3Ad), proliferator-activated receptor gamma coactivator (PGC-1), adenosine monophosphate-activated protein kinase (AMPK), peroxisome proliferator-activated receptor (PPAR), sterol regulatory element binding protein (SREBP), nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK), protein kinase B (AKT), SMAD2/3, and P13K/Akt pathways. Fucoxanthin also exhibits anti-fibrogenic activity that prevents non-alcoholic steatohepatitis (NASH) development.
Collapse
Affiliation(s)
- Jessica Winarto
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
| | - Dae-Geun Song
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
| | - Cheol-Ho Pan
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea
- Microalgae Ask US Co., Ltd., Gangneung 25441, Republic of Korea
| |
Collapse
|
9
|
Xue S, Lee D, Berry DC. Thermogenic adipose tissue in energy regulation and metabolic health. Front Endocrinol (Lausanne) 2023; 14:1150059. [PMID: 37020585 PMCID: PMC10067564 DOI: 10.3389/fendo.2023.1150059] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
The ability to generate thermogenic fat could be a targeted therapy to thwart obesity and improve metabolic health. Brown and beige adipocytes are two types of thermogenic fat cells that regulate energy balance. Both adipocytes share common morphological, biochemical, and thermogenic properties. Yet, recent evidence suggests unique features exist between brown and beige adipocytes, such as their cellular origin and thermogenic regulatory processes. Beige adipocytes also appear highly plastic, responding to environmental stimuli and interconverting between beige and white adipocyte states. Additionally, beige adipocytes appear to be metabolically heterogenic and have substrate specificity. Nevertheless, obese and aged individuals cannot develop beige adipocytes in response to thermogenic fat-inducers, creating a key clinical hurdle to their therapeutic promise. Thus, elucidating the underlying developmental, molecular, and functional mechanisms that govern thermogenic fat cells will improve our understanding of systemic energy regulation and strive for new targeted therapies to generate thermogenic fat. This review will examine the recent advances in thermogenic fat biogenesis, molecular regulation, and the potential mechanisms for their failure.
Collapse
Affiliation(s)
| | | | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
10
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
11
|
Generation of mega brown adipose tissue in adults by controlling brown adipocyte differentiation in vivo. Proc Natl Acad Sci U S A 2022; 119:e2203307119. [PMID: 36161914 DOI: 10.1073/pnas.2203307119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Brown adipose tissue (BAT) is a highly specialized adipose tissue in its immobile location and size during the entire adulthood. In response to cold exposure and other β3-adrenoreceptor stimuli, BAT commits energy consumption by nonshivering thermogenesis (NST). However, the molecular machinery in controlling the BAT mass in adults is unknown. Here, we show our surprising findings that the BAT mass and functions can be manipulated in adult animals by controlling BAT adipocyte differentiation in vivo. Platelet-derived growth factor receptor α (PDGFα) expressed in BAT progenitor cells served a signaling function to avert adipose progenitor differentiation. Genetic and pharmacological loss-of-function of PDGFRα eliminated the differentiation barrier and permitted progenitor cell differentiation to mature and functional BAT adipocytes. Consequently, an enlarged BAT mass (megaBAT) was created by PDGFRα inhibition owing to increases of brown adipocyte numbers. Under cold exposure, a microRNA-485 (miR-485) was identified as a master suppressor of the PDGFRα signaling, and delivery of miR-485 also produced megaBAT in adult animals. Noticeably, megaBAT markedly improved global metabolism, insulin sensitivity, high-fat-diet (HFD)-induced obesity, and diabetes by enhancing NST. Together, our findings demonstrate that the adult BAT mass can be increased by blocking the previously unprecedented inhibitory signaling for BAT progenitor cell differentiation. Thus, blocking the PDGFRα for the generation of megaBAT provides an attractive strategy for treating obesity and type 2 diabetes mellitus (T2DM).
Collapse
|
12
|
Rizzetti DA, Corrales P, Uranga-Ocio JA, Medina-Gómez G, Peçanha FM, Vassallo DV, Miguel M, Wiggers GA. Potential benefits of egg white hydrolysate in the prevention of Hg-induced dysfunction in adipose tissue. Food Funct 2022; 13:5996-6007. [PMID: 35575219 DOI: 10.1039/d2fo00561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: To investigate the effects of egg white hydrolysate (EWH) on the lipid and glycemic metabolism disruption in the white adipose tissue (WAT) dysfunction induced by mercury (Hg). Experimental: Wistar rats were treated for 60 days: control (saline, intramuscular - i.m.); hydrolysate (EWH, gavage, 1 g kg-1 day-1); mercury (HgCl2, i.m., 1st dose 4.6 μg kg-1, subsequent doses 0.07 μg kg-1 day-1) and hydrolysate-mercury (EWH-HgCl2). Hg level and histological analyses were performed in epididymal WAT (eWAT), pancreas and liver. GRP78, CHOP, PPARα, PPARγ, leptin, adiponectin, and CD11 mRNA expressions were analyzed in eWAT. The plasma lipid profile, glucose, and insulin levels were measured. Antioxidant status was also evaluated in the plasma and liver. Results: EWH intake prevented the reduced eWAT weight, adipocyte size, insulin levels, and antioxidant defenses and the increased glucose and triglyceride levels induced by Hg exposure; hepatic glutathione levels were higher in rats co-treated with EWH. The increased mRNA expression of CHOP, PPARα, and leptin induced by Hg was reduced in co-treated rats. EWH did not modify the elevated mRNA expression of GRP78, PPARγ and adiponectin in Hg-treated rats. Increased levels of Hg were found in the liver; the co-treatment did not alter this parameter. EWH prevented the morphological and metabolic disorder induced by Hg, by improving antioxidant defenses, inactivating pro-apoptotic pathways and normalizing the mRNA expression of PPARs and adipokines. Its effects enabled an increase in insulin levels and a normal balance between the fat storage and expenditure mechanisms in WAT. Conclusions: EWH may have potential benefits in the prevention and management of Hg-related metabolic disorders.
Collapse
Affiliation(s)
- Danize Aparecida Rizzetti
- Cardiovascular Physiology Laboratory, Universidade Federal do Pampa, BR 472, Km 592, Uruguaiana, Rio Grande do Sul, Brazil.
| | - Patricia Corrales
- Department of Basic Health Sciences, Universidad Rey Juan Carlos, Atenas s/n, Alcorcón, Spain.
| | - José Antonio Uranga-Ocio
- Department of Basic Health Sciences, Universidad Rey Juan Carlos, Atenas s/n, Alcorcón, Spain. .,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), Universidad Rey Juan Carlos, Atenas s/n, Alcorcón, Spain
| | - Gema Medina-Gómez
- Department of Basic Health Sciences, Universidad Rey Juan Carlos, Atenas s/n, Alcorcón, Spain.
| | - Franck Maciel Peçanha
- Cardiovascular Physiology Laboratory, Universidade Federal do Pampa, BR 472, Km 592, Uruguaiana, Rio Grande do Sul, Brazil.
| | - Dalton Valentim Vassallo
- Cardiac Electromechanical and Vascular Reactivity Laboratory, Universidade Federal do Espírito Santo, Marechal Campos, 1468, Vitória, Espírito Santo, Brazil.
| | - Marta Miguel
- Bioactivity and Food Analysis Laboratory, Instituto de Investigación en Ciencias de la Alimentación, Nicolás Cabrera, 9, Campus Universitario de Cantoblanco, Madrid, Spain.
| | - Giulia Alessandra Wiggers
- Cardiovascular Physiology Laboratory, Universidade Federal do Pampa, BR 472, Km 592, Uruguaiana, Rio Grande do Sul, Brazil.
| |
Collapse
|
13
|
Transdermal Delivery of Metformin Using Dissolving Microneedles and Iontophoresis Patches for Browning Subcutaneous Adipose Tissue. Pharmaceutics 2022; 14:pharmaceutics14040879. [PMID: 35456713 PMCID: PMC9029293 DOI: 10.3390/pharmaceutics14040879] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/24/2022] [Accepted: 04/11/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity is a serious public health problem that is strongly associated with increased multiple comorbidities such as diabetes, cardiovascular disease, and some types of cancer. While current anti-obesity treatments have various issues, locally transforming energy-storing white adipose tissue (WAT) into energy-burning brown-like/beige adipose tissue, the so-called browning of WAT, has been suggested to enhance obesity treatment efficiency with minimized side effects. Metformin is a first-line antidiabetes drug and a potent activator of AMP-activated protein kinase. Emerging evidence has suggested that metformin might enhance energy expenditure via the browning of WAT and hence reduce body weight. Subcutaneous WAT is easier to access and has a stronger browning potential than other WAT depots. In this study, we used dissolvable poly (lactic-co-glycolic acid) microneedles (MN) to deliver metformin to the subcutaneous WAT in obese C57BL/6J mice with the assistance of iontophoresis (INT), and then investigated metformin-induced WAT browning and its subsequent thermogenesis effects. Compared with MN alone or INT alone, MN + INT had better anti-obesity activity, as indicated by decreasing body weight and fat gain, increased energy expenditure, decreased fat pad size, and improved energy metabolism through the browning of WAT. Browning subcutaneous WAT by delivering metformin and other browning agents using this MN + INT approach might combat obesity in an effective, easy, and safe regimen.
Collapse
|
14
|
Almeida DL, Moreira VM, Cardoso LE, Junior MDF, Pavanelo A, Ribeiro TA, da Silva Franco CC, Tófolo LP, Peres MNC, Ribeiro MVG, Ferreira ARO, Gomes RM, Miranda RA, Trevenzoli IH, Armitage JA, Palma-Rigo K, de Freitas Mathias PC. Lean in one way, in obesity another: effects of moderate exercise in brown adipose tissue of early overfed male Wistar rats. Int J Obes (Lond) 2022; 46:137-143. [PMID: 34552207 DOI: 10.1038/s41366-021-00969-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Early postnatal overfeeding (PO) induces long-term overweight and reduces brown adipose tissue (BAT) thermogenesis. Exercise has been suggested as a possible intervention to increase BAT function. In this study, we investigated chronical effects of moderate-intensity exercise in BAT function in postnatal overfed male Wistar rats METHODS: Litters' delivery was on postnatal-day 0 - PN0. At PN2, litters were adjusted to nine (normal litter - NL) or three pups (small litter - SL) per dam. Animals were weaned on PN21 and in PN30 randomly divided into sedentary (NL-Sed and SL-Sed) or exercised (NL-Exe and SL-Exe), N of 14 litters per group. Exercise protocol started (PN30) with an effort test; training sessions were performed three times weekly at 60% of the VO2max achieved in effort test, until PN80. On PN81, a temperature transponder was implanted beneath the interscapular BAT, whose temperature was assessed in periods of lights-on and -off from PN87 to PN90. Sympathetic nerve activation of BAT was registered at PN90. Animals were euthanized at PN91 and tissues collected RESULTS: PO impaired BAT thermogenesis in lights-on (pPO < 0.0001) and -off (pPO < 0.01). Exercise increased BAT temperature in lights-on (pExe < 0.0001). In NL-Exe, increased BAT activity was associated with higher sympathetic activity (pExe < 0.05), β3-AR (pExe < 0.001), and UCP1 (pExe < 0.001) content. In SL-Exe, increasing BAT thermogenesis is driven by a combination of tissue morphology remodeling (pExe < 0.0001) with greater effect in increasing UCP1 (pExe < 0.001) and increased β3-AR (pExe < 0.001) content. CONCLUSION Moderate exercise chronically increased BAT thermogenesis in both, NL and SL groups. In NL-Exe by increasing Sympathetic activity, and in SL-Exe by a combination of increased β3-AR and UCP1 content with morphologic remodeling of BAT. Chronically increasing BAT thermogenesis in obese subjects may lead to higher overall energy expenditure, favoring the reduction of obesity and related comorbidities.
Collapse
Affiliation(s)
- Douglas Lopes Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil.
- Department of Physiology, State University of Londrina, Londrina, Paraná, Brazil.
| | - Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
- Department of Physiology, State University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Lucas Eduardo Cardoso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | | | - Audrei Pavanelo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Claudinéia Conationi da Silva Franco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Laize Perón Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maria Natália Chimirri Peres
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Rodrigo Mello Gomes
- Physiological Sciences Department, Federal University of Goiás, Av Esperança, Goiânia/GO, Brazil
| | - Rosiane Aparecida Miranda
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - Isis Hara Trevenzoli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - James Andrew Armitage
- Deakin University, School of Medicine, Optometry, 75 Pigdons Rd, Waurn Ponds, VIC, Australia
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Paulo Cesar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| |
Collapse
|
15
|
Becker-Greene D, Li H, Perez-Cremades D, Wu W, Bestepe F, Ozdemir D, Niosi CE, Aydogan C, Orgill DP, Feinberg MW, Icli B. MiR-409-3p targets a MAP4K3-ZEB1-PLGF signaling axis and controls brown adipose tissue angiogenesis and insulin resistance. Cell Mol Life Sci 2021; 78:7663-7679. [PMID: 34698882 PMCID: PMC8655847 DOI: 10.1007/s00018-021-03960-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/09/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
Endothelial cells (ECs) within the microvasculature of brown adipose tissue (BAT) are important in regulating the plasticity of adipocytes in response to increased metabolic demand by modulating the angiogenic response. However, the mechanism of EC-adipocyte crosstalk during this process is not completely understood. We used RNA sequencing to profile microRNAs derived from BAT ECs of obese mice and identified an anti-angiogenic microRNA, miR-409-3p. MiR-409-3p overexpression inhibited EC angiogenic properties; whereas, its inhibition had the opposite effects. Mechanistic studies revealed that miR-409-3p targets ZEB1 and MAP4K3. Knockdown of ZEB1/MAP4K3 phenocopied the angiogenic effects of miR-409-3p. Adipocytes co-cultured with conditioned media from ECs deficient in miR-409-3p showed increased expression of BAT markers, UCP1 and CIDEA. We identified a pro-angiogenic growth factor, placental growth factor (PLGF), released from ECs in response to miR-409-3p inhibition. Deficiency of ZEB1 or MAP4K3 blocked the release of PLGF from ECs and PLGF stimulation of 3T3-L1 adipocytes increased UCP1 expression in a miR-409-3p dependent manner. MiR-409-3p neutralization improved BAT angiogenesis, glucose and insulin tolerance, and energy expenditure in mice with diet-induced obesity. These findings establish miR-409-3p as a critical regulator of EC-BAT crosstalk by modulating a ZEB1-MAP4K3-PLGF signaling axis, providing new insights for therapeutic intervention in obesity.
Collapse
Affiliation(s)
- Dakota Becker-Greene
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
| | - Hao Li
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
| | - Daniel Perez-Cremades
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
- Department of Physiology, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Winona Wu
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
| | - Furkan Bestepe
- Molecular Cardiology Research Institute, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA
| | - Denizhan Ozdemir
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
- Department of Medical Biology, Hacettepe University, Ankara, Turkey
| | - Carolyn E Niosi
- Molecular Cardiology Research Institute, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA
| | - Ceren Aydogan
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA
- Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Dennis P Orgill
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mark W Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA.
| | - Basak Icli
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Louis Pasteur Avenue 77, Boston, MA, 02115, USA.
- Molecular Cardiology Research Institute, Tufts University School of Medicine, 800 Washington St, Boston, MA, 02111, USA.
| |
Collapse
|
16
|
Weldenegodguad M, Pokharel K, Niiranen L, Soppela P, Ammosov I, Honkatukia M, Lindeberg H, Peippo J, Reilas T, Mazzullo N, Mäkelä KA, Nyman T, Tervahauta A, Herzig KH, Stammler F, Kantanen J. Adipose gene expression profiles reveal insights into the adaptation of northern Eurasian semi-domestic reindeer (Rangifer tarandus). Commun Biol 2021; 4:1170. [PMID: 34620965 PMCID: PMC8497613 DOI: 10.1038/s42003-021-02703-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/21/2021] [Indexed: 02/08/2023] Open
Abstract
Reindeer (Rangifer tarandus) are semi-domesticated animals adapted to the challenging conditions of northern Eurasia. Adipose tissues play a crucial role in northern animals by altering gene expression in their tissues to regulate energy homoeostasis and thermogenic activity. Here, we perform transcriptome profiling by RNA sequencing of adipose tissues from three different anatomical depots: metacarpal (bone marrow), perirenal, and prescapular fat in Finnish and Even reindeer (in Sakha) during spring and winter. A total of 16,212 genes are expressed in our data. Gene expression profiles in metacarpal tissue are distinct from perirenal and prescapular adipose tissues. Notably, metacarpal adipose tissue appears to have a significant role in the regulation of the energy metabolism of reindeer in spring when their nutritional condition is poor after winter. During spring, genes associated with the immune system are upregulated in the perirenal and prescapular adipose tissue. Blood and tissue parameters reflecting general physiological and metabolic status show less seasonal variation in Even reindeer than in Finnish reindeer. This study identifies candidate genes potentially involved in immune response, fat deposition, and energy metabolism and provides new information on the mechanisms by which reindeer adapt to harsh arctic conditions.
Collapse
Affiliation(s)
- Melak Weldenegodguad
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland ,grid.9668.10000 0001 0726 2490Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kisun Pokharel
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Laura Niiranen
- grid.10858.340000 0001 0941 4873Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Päivi Soppela
- grid.37430.330000 0001 0744 995XArctic Centre, University of Lapland, Rovaniemi, Finland
| | - Innokentyi Ammosov
- grid.495192.2Laboratory of Reindeer Husbandry and Traditional Industries, Yakut Scientific Research Institute of Agriculture, Yakutsk, The Sakha Republic (Yakutia) Russia
| | | | - Heli Lindeberg
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Maaninka, Finland
| | - Jaana Peippo
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland ,NordGen—Nordic Genetic Resource Center, Ås, Norway
| | - Tiina Reilas
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Nuccio Mazzullo
- grid.37430.330000 0001 0744 995XArctic Centre, University of Lapland, Rovaniemi, Finland
| | - Kari A. Mäkelä
- grid.10858.340000 0001 0941 4873Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Tommi Nyman
- grid.454322.60000 0004 4910 9859Department of Ecosystems in the Barents Region, Norwegian Institute of Bioeconomy Research, Svanvik, Norway
| | - Arja Tervahauta
- grid.9668.10000 0001 0726 2490Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Karl-Heinz Herzig
- grid.10858.340000 0001 0941 4873Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland ,grid.10858.340000 0001 0941 4873Medical Research Center, Faculty of Medicine, University of Oulu, Oulu, Finland ,grid.412326.00000 0004 4685 4917Oulu University Hospital, Oulu, Finland ,grid.22254.330000 0001 2205 0971Institute of Pediatrics, Poznań University of Medical Sciences, Poznań, Poland
| | - Florian Stammler
- grid.37430.330000 0001 0744 995XArctic Centre, University of Lapland, Rovaniemi, Finland
| | - Juha Kantanen
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland
| |
Collapse
|
17
|
Colleluori G, Perugini J, Barbatelli G, Cinti S. Mammary gland adipocytes in lactation cycle, obesity and breast cancer. Rev Endocr Metab Disord 2021; 22:241-255. [PMID: 33751362 PMCID: PMC8087566 DOI: 10.1007/s11154-021-09633-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
The mammary gland (MG) is an exocrine gland present in female mammals responsible for the production and secretion of milk during the process of lactation. It is mainly composed by epithelial cells and adipocytes. Among the features that make the MG unique there are 1) its highly plastic properties displayed during pregnancy, lactation and involution (all steps belonging to the lactation cycle) and 2) its requirement to grow in close association with adipocytes which are absolutely necessary to ensure MG's proper development at puberty and remodeling during the lactation cycle. Although MG adipocytes play such a critical role for the gland development, most of the studies have focused on its epithelial component only, leaving the role of the neighboring adipocytes largely unexplored. In this review we aim to describe evidences regarding MG's adipocytes role and properties in physiologic conditions (gland development and lactation cycle), obesity and breast cancer, emphasizing the existing gaps in the literature which deserve further investigation.
Collapse
Affiliation(s)
- Georgia Colleluori
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto, 10A 60020, Ancona, Italy.
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto, 10A 60020, Ancona, Italy
| | - Giorgio Barbatelli
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto, 10A 60020, Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto, 10A 60020, Ancona, Italy.
| |
Collapse
|
18
|
Barella LF, Jain S, Kimura T, Pydi SP. Metabolic roles of G protein-coupled receptor signaling in obesity and type 2 diabetes. FEBS J 2021; 288:2622-2644. [PMID: 33682344 DOI: 10.1111/febs.15800] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/31/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022]
Abstract
The incidence of obesity and type 2 diabetes (T2D) has been increasing steadily worldwide. It is estimated that by 2045 more than 800 million people will be suffering from diabetes. Despite the advancements in modern medicine, more effective therapies for treating obesity and T2D are needed. G protein-coupled receptors (GPCRs) have emerged as important drug targets for various chronic diseases, including obesity, T2D, and liver diseases. During the past two decades, many laboratories worldwide focused on understanding the role of GPCR signaling in regulating glucose metabolism and energy homeostasis. The information gained from these studies can guide the development of novel therapeutic agents. In this review, we summarize recent studies providing insights into the role of GPCR signaling in peripheral, metabolically important tissues such as pancreas, liver, skeletal muscle, and adipose tissue, focusing primarily on the use of mutant animal models and human data.
Collapse
Affiliation(s)
- Luiz F Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.,Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Takefumi Kimura
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.,Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| |
Collapse
|
19
|
Wang Z, Hou Y, Ren S, Liu Z, Zuo Z, Huang S, Wang W, Wang H, Chen Y, Xu Y, Yamamoto M, Zhang Q, Fu J, Pi J. CL316243 treatment mitigates the inflammation in white adipose tissues of juvenile adipocyte-specific Nfe2l1 knockout mice. Free Radic Biol Med 2021; 165:289-298. [PMID: 33545311 DOI: 10.1016/j.freeradbiomed.2021.01.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 12/13/2022]
Abstract
Nuclear factor-erythroid 2-related factor 1 (NFE2L1) is a key transcription factor that regulates cellular adaptive responses to various stresses. Our previous studies revealed that adult adipocyte-specific Nfe2l1-knockout [Nfe2l1(f)-KO] mice show adipocyte hypertrophy and severe adipose inflammation, which can be worsened by rosiglitazone, a peroxisome proliferator-activated receptor γ agonist. To further assess the crucial roles of NFE2L1 in adipocytes, we investigated the effect of CL316243, a β3 adrenergic agonist that promotes lipolysis via a post-translational mechanism, on adipose inflammation in juvenile Nfe2l1(f)-KO mice. In contrast to adult mice, 4-week-old juvenile Nfe2l1(f)-KO mice displayed a normal fat distribution but reduced fasting plasma glycerol levels and elevated adipocyte hypertrophy and macrophage infiltration in inguinal and gonadal WAT. In addition, Nfe2l1(f)-KO mice had decreased expression of multiple lipolytic genes and reduced lipolytic activity in WAT. While 7 days of CL316243 treatment showed no significant effect on adipose inflammation in Nfe2l1-Floxed control mice, the same treatment dramatically alleviated macrophage infiltration and mRNA expression of inflammation and pyroptosis-related genes in WAT of Nfe2l1(f)-KO mice. Together with previous findings in adult mice, the current study highlights that NFE2L1 plays a fundamental regulatory role in lipolytic gene expression and thus might be an important target to improve adipose plasticity and lipid homeostasis.
Collapse
Affiliation(s)
- Zhendi Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Yongyong Hou
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Suping Ren
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Zhiyuan Liu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Zhuo Zuo
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Sicui Huang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Wanqi Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Huihui Wang
- Group of Chronic Disease and Environmental Genomics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Yanyan Chen
- The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Road, Heping Area, Shenyang, 110001, PR China
| | - Yuanyuan Xu
- Group of Chronic Disease and Environmental Genomics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, No 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China.
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, No 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China.
| |
Collapse
|
20
|
Smetanina N, Valickas R, Vitkauskiene A, Albertsson-Wikland K, Verkauskienė R. Prevalence of Metabolic Syndrome and Impaired Glucose Metabolism among 10- to 17-Year-Old Overweight and Obese Lithuanian Children and Adolescents. Obes Facts 2021; 14:271-282. [PMID: 33951670 PMCID: PMC8255643 DOI: 10.1159/000514720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 01/19/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Overweight (Ow) and obesity among adults and children increases the risk of metabolic consequences. Metabolic syndrome (MS) and impaired glucose metabolism are well-known risk factors for cardiovascular diseases and type 2 diabetes. The aim of this study was to evaluate the prevalence of MS and impaired glucose metabolism among Ow and obese (Ob) children and adolescents (aged 10-17 years) in Lithuania, and to evaluate the associations between insulin resistance (IR) indices and anthropometric parameters as well as metabolic disturbances. METHODS The study population consisted of 344 OwOb children and adolescents of all pubertal stages. Oral glucose tolerance tests (OGTTs), IR and β cell function indices, lipid profile, and anthropometric parameters of all subjects were analyzed. MS was defined according to the International Diabetes Federation consensus guidelines. RESULTS MS was found in 21.3% of the OwOb children and adolescents, and 12.1% had impaired glucose metabolism (6.9% with impaired fasting glucose, 4.5% with impaired glucose tolerance, and 0.6% with type 2 diabetes). IR was directly related to body mass index and waist circumference, waist-to-height and waist-to-hip ratios, and sum of skin-fold thicknesses. Children with MS were more insulin-resistant, had higher odds ratio for prediabetes and had a more disturbed lipid profile than subjects without MS. Moreover, total cholesterol and low-density lipoprotein cholesterol levels were significantly lower in the more mature OwOb adolescents. CONCLUSION MS and lipid profile disturbances are common in OwOb children and adolescents. MS is directly associated with IR. Therefore, OwOb children and adolescents should be carefully followed up for metabolic abnormalities during late childhood as these can persist into adulthood.
Collapse
Affiliation(s)
- Natalija Smetanina
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
- *Natalija Smetanina,
| | - Raimondas Valickas
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Astra Vitkauskiene
- Department of Laboratory Medicine, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kerstin Albertsson-Wikland
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Rasa Verkauskienė
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
21
|
Anthocyanin Bioactivity in Obesity and Diabetes: The Essential Role of Glucose Transporters in the Gut and Periphery. Cells 2020; 9:cells9112515. [PMID: 33233708 PMCID: PMC7699863 DOI: 10.3390/cells9112515] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Obesity and type-2 diabetes trends continue to worsen in the United States. Dietary anthocyanins (typically provided by berries and other fruits) are reported to have protective effects against both conditions using a variety of experimental research models including animal and human feeding studies. This review highlights studies that explore the biochemical pathways in both tissue and rodent models which could explain clinical improvements noted with anthocyanin consumption. First, the primary mode of intestinal absorption of anthocyanins is through both sGLT1 and GLUT2 glucose transporters. Stronger binding affinities may allow anthocyanins to be more inhibitive to glucose absorption compared to the reverse, where GLUT2 expression may also be affected. Genetic or chemical inhibition of sGLT1 or GLUT2 demonstrate their essential function in anthocyanin absorption across the enterocyte, where the former interacts with a greater variety of anthocyanins but the latter is the major transporter for specific anthocyanin-glycosides. Once absorbed, anthocyanins positively modulate GLUT4 density and function in both skeletal muscle and adipose tissues via the upregulation of AMPK and restoration of insulin sensitivity. Antioxidant properties and phosphodiesterase inhibition by anthocyanins promote both mitochondrial function and density which could be novel targets for dietary management of obesity and its complications.
Collapse
|
22
|
Aging and Immunometabolic Adaptations to Thermogenesis. Ageing Res Rev 2020; 63:101143. [PMID: 32810648 DOI: 10.1016/j.arr.2020.101143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/20/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Abstract
Brown and subcutaneous adipose tissues play a key role in non-shivering thermogenesis both in mice and human, and their activation by adrenergic stimuli promotes energy expenditure, reduces adiposity, and protects against age-related metabolic diseases such as type 2 diabetes (T2D). Low-grade inflammation and insulin resistance characterize T2D. Even though the decline of thermogenic adipose tissues is well-established during ageing, the mechanisms by which this event affects immune system and contributes to the development of T2D is still poorly defined. It is emerging that activation of thermogenic adipose tissues promotes type 2 immunity skewing, limiting type 1 inflammation. Of note, metabolic substrates sustaining type 1 inflammation (e.g. glucose and succinate) are also used by activated adipocytes to promote thermogenesis. Keeping in mind this aspect, a nutrient competition between adipocytes and adipose tissue immune cell infiltrates could be envisaged. Herein, we reviewed the metabolic rewiring of adipocytes during thermogenesis in order to give important insight into the anti-inflammatory role of thermogenic adipose tissues and delineate how their decline during ageing may favor the setting of low-grade inflammatory states that predispose to type 2 diabetes in elderly. A brief description about the contribution of adipokines secreted by thermogenic adipocytes in modulation of immune cell activation is also provided. Finally, we have outlined experimental flow chart procedures and provided technical advices to investigate the physiological processes leading to thermogenic adipose tissue impairment that are behind the immunometabolic decline during aging.
Collapse
|
23
|
Shin S, Pang Y, Park J, Liu L, Lukas BE, Kim SH, Kim KW, Xu P, Berry DC, Jiang Y. Dynamic control of adipose tissue development and adult tissue homeostasis by platelet-derived growth factor receptor alpha. eLife 2020; 9:56189. [PMID: 32553115 PMCID: PMC7338051 DOI: 10.7554/elife.56189] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Adipocytes arise from distinct progenitor populations during developmental and adult stages but little is known about how developmental progenitors differ from adult progenitors. Here, we investigate the role of platelet-derived growth factor receptor alpha (PDGFRα) in the divergent regulation of the two different adipose progenitor cells (APCs). Using in vivo adipose lineage tracking and deletion mouse models, we found that developmental PDGFRα+ cells are adipogenic and differentiated into mature adipocytes, and the deletion of Pdgfra in developmental adipose lineage disrupted white adipose tissue (WAT) formation. Interestingly, adult PDGFRα+ cells do not significantly contribute to adult adipogenesis, and deleting Pdgfra in adult adipose lineage did not affect WAT homeostasis. Mechanistically, embryonic APCs require PDGFRα for fate maintenance, and without PDGFRα, they underwent fate change from adipogenic to fibrotic lineage. Collectively, our findings indicate that PDGFRα+ cells and Pdgfra gene itself are differentially required for WAT development and adult WAT homeostasis.
Collapse
Affiliation(s)
- Sunhye Shin
- Department of Physiology and Biophysics, College of Medicine, The University of Illinois, Chicago, United States
| | - Yiyu Pang
- Department of Physiology and Biophysics, College of Medicine, The University of Illinois, Chicago, United States
| | - Jooman Park
- Department of Physiology and Biophysics, College of Medicine, The University of Illinois, Chicago, United States
| | - Lifeng Liu
- Department of Physiology and Biophysics, College of Medicine, The University of Illinois, Chicago, United States
| | - Brandon E Lukas
- Department of Physiology and Biophysics, College of Medicine, The University of Illinois, Chicago, United States
| | - Seung Hyeon Kim
- Department of Pharmacology, College of Medicine, The University of Illinois, Chicago, United States
| | - Ki-Wook Kim
- Department of Pharmacology, College of Medicine, The University of Illinois, Chicago, United States
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, United States
| | - Daniel C Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, United States
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, The University of Illinois, Chicago, United States
| |
Collapse
|
24
|
Schiffer TA, Lundberg JO, Weitzberg E, Carlström M. Modulation of mitochondria and NADPH oxidase function by the nitrate-nitrite-NO pathway in metabolic disease with focus on type 2 diabetes. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165811. [PMID: 32339643 DOI: 10.1016/j.bbadis.2020.165811] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria play fundamental role in maintaining cellular metabolic homeostasis, and metabolic disorders including type 2 diabetes (T2D) have been associated with mitochondrial dysfunction. Pathophysiological mechanisms are coupled to increased production of reactive oxygen species and oxidative stress, together with reduced bioactivity/signaling of nitric oxide (NO). Novel strategies restoring these abnormalities may have therapeutic potential in order to prevent or even treat T2D and associated cardiovascular and renal co-morbidities. A diet rich in green leafy vegetables, which contains high concentrations of inorganic nitrate, has been shown to reduce the risk of T2D. To this regard research has shown that in addition to the classical NO synthase (NOS) dependent pathway, nitrate from our diet can work as an alternative precursor for NO and other bioactive nitrogen oxide species via serial reductions of nitrate (i.e. nitrate-nitrite-NO pathway). This non-conventional pathway may act as an efficient back-up system during various pathological conditions when the endogenous NOS system is compromised (e.g. acidemia, hypoxia, ischemia, aging, oxidative stress). A number of experimental studies have demonstrated protective effects of nitrate supplementation in models of obesity, metabolic syndrome and T2D. Recently, attention has been directed towards the effects of nitrate/nitrite on mitochondrial functions including beiging/browning of white adipose tissue, PGC-1α and SIRT3 dependent AMPK activation, GLUT4 translocation and mitochondrial fusion-dependent improvements in glucose homeostasis, as well as dampening of NADPH oxidase activity. In this review, we examine recent research related to the effects of bioactive nitrogen oxide species on mitochondrial function with emphasis on T2D.
Collapse
Affiliation(s)
- Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
Rodríguez A, Catalán V, Ramírez B, Unamuno X, Portincasa P, Gómez-Ambrosi J, Frühbeck G, Becerril S. Impact of adipokines and myokines on fat browning. J Physiol Biochem 2020; 76:227-240. [PMID: 32236810 DOI: 10.1007/s13105-020-00736-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Since the discovery of leptin in 1994, the adipose tissue (AT) is not just considered a passive fat storage organ but also an extremely active secretory and endocrine organ that secretes a large variety of hormones, called adipokines, involved in energy metabolism. Adipokines may not only contribute to AT dysfunction and obesity, but also in fat browning, a process that induces a phenotypic switch from energy-storing white adipocytes to thermogenic brown fat-like cells. The fat browning process and, consequently, thermogenesis can also be stimulated by physical exercise. Contracting skeletal muscle is a metabolically active tissue that participates in several endocrine functions through the production of bioactive factors, collectively termed myokines, proposed as the mediators of physical activity-induced health benefits. Myokines affect muscle mass, have profound effects on glucose and lipid metabolism, and promote browning and thermogenesis of white AT in an endocrine and/or paracrine manner. The present review focuses on the role of different myokines and adipokines in the regulation of fat browning, as well as in the potential cross-talk between AT and skeletal muscle, in order to control body weight, energy expenditure and thermogenesis.
Collapse
Affiliation(s)
- A Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - V Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - B Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - X Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain.,Medical Engineering Laboratory, University of Navarra, Pamplona, Spain
| | - P Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, Policlinico Hospital, University of Bari Medical School, 70124, Bari, Italy
| | - J Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - G Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain. .,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
26
|
Taha S, Volkmer E, Haas E, Alberton P, Straub T, David-Rus D, Aszodi A, Giunta R, Saller MM. Differences in the Inflammatory Response of White Adipose Tissue and Adipose-Derived Stem Cells. Int J Mol Sci 2020; 21:ijms21031086. [PMID: 32041245 PMCID: PMC7037886 DOI: 10.3390/ijms21031086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
The application of liposuctioned white adipose tissue (L-WAT) and adipose-derived stem cells (ADSCs) as a novel immunomodulatory treatment option is the currently subject of various clinical trials. Because it is crucial to understand the underlying therapeutic mechanisms, the latest studies focused on the immunomodulatory functions of L-WAT or ADSCs. However, studies that examine the specific transcriptional adaptation of these treatment options to an extrinsic inflammatory stimulus in an unbiased manner are scarce. The aim of this study was to compare the gene expression profile of L-WAT and ADSCs, when subjected to tumor necrosis factor alpha (TNFα), and to identify key factors that might be therapeutically relevant when using L-WAT or ADSCs as an immuno-modulator. Fat tissue was harvested by liposuction from five human donors. ADSCs were isolated from the same donors and shortly subjected to expansion culture. L-WAT and ADSCs were treated with human recombinant TNFα, to trigger a strong inflammatory response. Subsequently, an mRNA deep nextgeneration sequencing was performed to evaluate the different inflammatory responses of L-WAT and ADSCs. We found significant gene expression changes in both experimental groups after TNFα incubation. However, ADSCs showed a more homogenous gene expression profile by predominantly expressing genes involved in immunomodulatory processes such as CCL19, CCL5, TNFSF15 and IL1b when compared to L-WAT, which reacted rather heterogeneously. As RNA sequencing between L-WAT and ADSCS treated with TNFα revealed that L-WAT responded very heterogeneously to TNFα treatment, we therefore conclude that ADSCs are more reliable and predictable when used therapeutically. Our study furthermore yields insight into potential biological processes regarding immune system response, inflammatory response, and cell activation. Our results can help to better understand the different immunomodulatory effects of L-WAT and ADSCs.
Collapse
Affiliation(s)
- Sara Taha
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (S.T.); (E.V.); (E.H.); (P.A.); (A.A.)
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University (LMU), Pettenkoferstraße. 8a, 80336 Munich, Germany
| | - Elias Volkmer
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (S.T.); (E.V.); (E.H.); (P.A.); (A.A.)
- Department of Hand Surgery, Helios Klinikum München West, Steinerweg 5, 81241 Munich, Germany
| | - Elisabeth Haas
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (S.T.); (E.V.); (E.H.); (P.A.); (A.A.)
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University (LMU), Pettenkoferstraße. 8a, 80336 Munich, Germany
| | - Paolo Alberton
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (S.T.); (E.V.); (E.H.); (P.A.); (A.A.)
| | - Tobias Straub
- Bioinformatics Unit, Biomedical Center Munich, Ludwig-Maximilians-University (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany;
| | - Diana David-Rus
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377 Munich, Germany;
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (S.T.); (E.V.); (E.H.); (P.A.); (A.A.)
| | - Riccardo Giunta
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University (LMU), Pettenkoferstraße. 8a, 80336 Munich, Germany
| | - Maximilian Michael Saller
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (S.T.); (E.V.); (E.H.); (P.A.); (A.A.)
- Correspondence: ; Tel.: +49-89-4400-55486
| |
Collapse
|
27
|
Aquaglyceroporins Are Differentially Expressed in Beige and White Adipocytes. Int J Mol Sci 2020; 21:ijms21020610. [PMID: 31963489 PMCID: PMC7014209 DOI: 10.3390/ijms21020610] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/31/2022] Open
Abstract
Browning of white adipocytes has been proposed as a powerful strategy to overcome metabolic complications, since brown adipocytes are more catabolic, expending energy as a heat form. However, the biological pathways involved in the browning process are still unclear. Aquaglyceroporins are a sub-class of aquaporin water channels that also permeate glycerol and are involved in body energy homeostasis. In the adipose tissue, aquaporin-7 (AQP7) is the most representative isoform, being crucial for white adipocyte fully differentiation and glycerol metabolism. The altered expression of AQP7 is involved in the onset of obesity and metabolic disorders. Herein, we investigated if aquaglyceroporins are implicated in beige adipocyte differentiation, similar to white cells. Thus, we optimized a protocol of murine 3T3-L1 preadipocytes browning that displayed increased beige and decreased white adipose tissue features at both gene and protein levels and evaluated aquaporin expression patterns along the differentiation process together with cellular lipid content. Our results revealed that AQP7 and aquaporin-9 (AQP9) expression was downregulated throughout beige adipocyte differentiation compared to white differentiation, which may be related to the beige physiological role of heat production from oxidative metabolism, contrasting with the anabolic/catabolic lipid metabolism requiring glycerol gateways occurring in white adipose cells.
Collapse
|
28
|
Taha S, Saller MM, Haas E, Farkas Z, Aszodi A, Giunta R, Volkmer E. Adipose-derived stem/progenitor cells from lipoaspirates: A comparison between the Lipivage200-5 liposuction system and the Body-Jet liposuction system. J Plast Reconstr Aesthet Surg 2020; 73:166-175. [DOI: 10.1016/j.bjps.2019.06.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/28/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
|
29
|
Riuzzi F, Chiappalupi S, Arcuri C, Giambanco I, Sorci G, Donato R. S100 proteins in obesity: liaisons dangereuses. Cell Mol Life Sci 2020; 77:129-147. [PMID: 31363816 PMCID: PMC11104817 DOI: 10.1007/s00018-019-03257-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
Obesity is an endemic pathophysiological condition and a comorbidity associated with hypercholesterolemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, and cancer. The adipose tissue of obese subjects shows hypertrophic adipocytes, adipocyte hyperplasia, and chronic low-grade inflammation. S100 proteins are Ca2+-binding proteins exclusively expressed in vertebrates in a cell-specific manner. They have been implicated in the regulation of a variety of functions acting as intracellular Ca2+ sensors transducing the Ca2+ signal and extracellular factors affecting cellular activity via ligation of a battery of membrane receptors. Certain S100 proteins, namely S100A4, the S100A8/S100A9 heterodimer and S100B, have been implicated in the pathophysiology of obesity-promoting macrophage-based inflammation via toll-like receptor 4 and/or receptor for advanced glycation end-products ligation. Also, serum levels of S100A4, S100A8/S100A9, S100A12, and S100B correlate with insulin resistance/type 2 diabetes, metabolic risk score, and fat cell size. Yet, secreted S100B appears to exert neurotrophic effects on sympathetic fibers in brown adipose tissue contributing to the larger sympathetic innervation of this latter relative to white adipose tissue. In the present review we first briefly introduce S100 proteins and then critically examine their role(s) in adipose tissue and obesity.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Sara Chiappalupi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, 06132, Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
| |
Collapse
|
30
|
Chung JY, Ain QU, Song Y, Yong SB, Kim YH. Targeted delivery of CRISPR interference system against Fabp4 to white adipocytes ameliorates obesity, inflammation, hepatic steatosis, and insulin resistance. Genome Res 2019; 29:1442-1452. [PMID: 31467027 PMCID: PMC6724665 DOI: 10.1101/gr.246900.118] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 07/23/2019] [Indexed: 12/18/2022]
Abstract
Obesity is an increasing pathophysiological problem in developed societies. Despite all major progress in understanding molecular mechanisms of obesity, currently available anti-obesity drugs have shown limited efficacy with severe side effects. CRISPR interference (CRISPRi) mechanism based on catalytically dead Cas9 (dCas9) and single guide RNA (sgRNA) was combined with a targeted nonviral gene delivery system to treat obesity and obesity-induced type 2 diabetes. A fusion peptide targeting a vascular and cellular marker of adipose tissue, prohibitin, was developed by conjugation of adipocyte targeting sequence (CKGGRAKDC) to 9-mer arginine (ATS-9R). (dCas9/sgFabp4) + ATS-9R oligoplexes showed effective condensation and selective delivery into mature adipocytes. Targeted delivery of the CRISPRi system against Fabp4 to white adipocytes by ATS-9R induced effective silencing of Fabp4, resulting in reduction of body weight and inflammation and restoration of hepatic steatosis in obese mice. This RNA-guided DNA recognition platform provides a simple and safe approach to regress and treat obesity and obesity-induced metabolic syndromes.
Collapse
Affiliation(s)
- Jee Young Chung
- Department of Bioengineering, Institute for Bioengineering and Biopharamceutical Research Hanyang University, 04763 Seoul, South Korea
| | - Qurrat Ul Ain
- Department of Bioengineering, Institute for Bioengineering and Biopharamceutical Research Hanyang University, 04763 Seoul, South Korea
| | - Yoonsung Song
- Department of Bioengineering, Institute for Bioengineering and Biopharamceutical Research Hanyang University, 04763 Seoul, South Korea
- BK21 Plus Future Biopharmaceutical Human Resource Team, Hanyang University, 04763 Seoul, South Korea
| | - Seok-Beom Yong
- Department of Bioengineering, Institute for Bioengineering and Biopharamceutical Research Hanyang University, 04763 Seoul, South Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharamceutical Research Hanyang University, 04763 Seoul, South Korea
- BK21 Plus Future Biopharmaceutical Human Resource Team, Hanyang University, 04763 Seoul, South Korea
| |
Collapse
|
31
|
Di Stefano AB, Massihnia D, Grisafi F, Castiglia M, Toia F, Montesano L, Russo A, Moschella F, Cordova A. Adipose tissue, angiogenesis and angio-MIR under physiological and pathological conditions. Eur J Cell Biol 2019; 98:53-64. [DOI: 10.1016/j.ejcb.2018.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023] Open
|
32
|
Kawarasaki S, Kuwata H, Sawazaki H, Sakamoto T, Nitta T, Kim CS, Jheng HF, Takahashi H, Nomura W, Ara T, Takahashi N, Tomita K, Yu R, Kawada T, Goto T. A new mouse model for noninvasive fluorescence-based monitoring of mitochondrial UCP1 expression. FEBS Lett 2019; 593:1201-1212. [PMID: 31074834 DOI: 10.1002/1873-3468.13430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/09/2019] [Accepted: 05/05/2019] [Indexed: 01/08/2023]
Abstract
Mitochondrial uncoupling protein 1 (UCP1) is well known for its thermogenic function in brown adipose tissue (BAT). Since UCP1 expends energy on thermogenesis, UCP1 activation has been considered an approach to ameliorate obesity. As a tool for uncovering yet unknown mechanisms of UCP1 activation, we generated a transgenic mouse model in which UCP1 expression levels are reflected in fluorescence derived from monomeric red fluorescent protein 1 (mRFP1). In these UCP1-mRFP1 BAC transgenic mice, fluorescence intensity mimics the change in UCP1 expression levels evoked through physiological or pharmacological stimulation. This transgenic mouse model will be useful in the search for bioactive compounds with the ability to induce UCP1 and for revealing undiscovered mechanisms of BAT activation.
Collapse
Affiliation(s)
- Satoko Kawarasaki
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Hidetoshi Kuwata
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Honami Sawazaki
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Tomoya Sakamoto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Takahiro Nitta
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Chuu-Sook Kim
- Department of Food Science and Nutrition, University of Ulsan, South Korea
| | - Huei-Fen Jheng
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Haruya Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Wataru Nomura
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Japan
| | - Takeshi Ara
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | - Nobuyuki Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Japan
| | - Koichi Tomita
- Department of Anatomy and Developmental Neurobiology, Graduate school of Biomedical Sciences, Tokushima University, Japan
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, South Korea
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Japan
| |
Collapse
|
33
|
Choi CJ, Tao W, Doddapaneni R, Acosta-Torres Z, Blessing NW, Lee BW, Pelaez D, Wester ST. The Effect of Prostaglandin Analogue Bimatoprost on Thyroid-Associated Orbitopathy. Invest Ophthalmol Vis Sci 2019; 59:5912-5923. [PMID: 30551199 PMCID: PMC6296211 DOI: 10.1167/iovs.18-25134] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose We characterize the effect of bimatoprost on orbital adipose tissue in thyroid-associated orbitopathy (TAO) with clinicopathologic correlation. Methods Orbital adipose-derived stem cells (OASCs) from types 1 and 2 TAO and control patients with and without exposure to 1 μm bimatoprost were examined via immunohistochemistry, RT-PCR, and Western blot for cell viability, migration capacity, lipid content, adipocyte morphology, mitochondrial content, and levels of adipogenic markers. A retrospective chart review was performed for clinicopathologic correlation. In mice, optical coherence tomography and pattern electroretinography were performed at baseline and at 1 month following a retrobulbar injection of bimatoprost, followed by orbital exenteration for histopathologic examination. Results Types 1 and 2 TAO-derived cells had a significantly higher migration capacity and lipid content than those of healthy controls. With the addition of bimatoprost, types 1 and 2 TAO and control adipocytes exhibited a significant decrease in lipid content with morphologic transformation into smaller and multilocular lipid droplets, and an increase in mitochondrial load and UCP-1 expression consistent with an increase in brown adipose tissue turnover. Retrobulbar injection of bimatoprost in mice did not alter the gross morphology, retinal thickness, or ganglion cell function in vivo. Conclusions Bimatoprost inhibits adipogenesis in OASCs and upregulates pathways involved in the browning of adipocytes. Furthermore, retrobulbar injection of bimatoprost is tolerated without immediate adverse effects in mice. Our results suggest a potential future application of prostaglandin analogues in the treatment of TAO.
Collapse
Affiliation(s)
- Catherine J Choi
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States
| | - Wensi Tao
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States
| | - Ravi Doddapaneni
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States
| | - Zenith Acosta-Torres
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States.,Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Nathan W Blessing
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States
| | - Bradford W Lee
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States
| | - Daniel Pelaez
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States.,Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Sara T Wester
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, Department of Ophthalmology, McKnight Vision Research Center, University of Miami School of Medicine, Miami, Florida, United States
| |
Collapse
|
34
|
Blázquez-Medela AM, Jumabay M, Boström KI. Beyond the bone: Bone morphogenetic protein signaling in adipose tissue. Obes Rev 2019; 20:648-658. [PMID: 30609449 PMCID: PMC6447448 DOI: 10.1111/obr.12822] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/02/2018] [Accepted: 11/25/2018] [Indexed: 02/06/2023]
Abstract
The bone morphogenetic proteins (BMPs) belong to the same superfamily as related to transforming growth factor β (TGFβ), growth and differentiation factors (GDFs), and activins. They were initially described as inducers of bone formation but are now known to be involved in morphogenetic activities and cell differentiation throughout the body, including the development of adipose tissue and adipogenic differentiation. BMP4 and BMP7 are the most studied BMPs in adipose tissue, with major roles in white adipogenesis and brown adipogenesis, respectively, but other BMPs such as BMP2, BMP6, and BMP8b as well as some inhibitors and modulators have been shown to also affect adipogenesis. It has become ever more important to understand adipose regulation, including the BMP pathways, in light of the strong links between obesity and metabolic and cardiovascular disease. In this review, we summarize the available information on BMP signaling in adipose tissue using preferentially articles that have appeared in the last decade, which together demonstrate the importance of BMP signaling in adipose biology.
Collapse
Affiliation(s)
- Ana M Blázquez-Medela
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Medet Jumabay
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States.,Molecular Biology Institute, UCLA, Los Angeles, California, United States
| |
Collapse
|
35
|
Ghosh S, Taylor JL, Mendoza TM, Dang T, Burk DH, Yu Y, Kilroy G, Floyd ZE. Siah2 modulates sex-dependent metabolic and inflammatory responses in adipose tissue to a high-fat diet challenge. Biol Sex Differ 2019; 10:19. [PMID: 30987673 PMCID: PMC6466809 DOI: 10.1186/s13293-019-0233-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/31/2019] [Indexed: 02/08/2023] Open
Abstract
Background The obesity-related risk of developing metabolic syndrome is higher in males than in females of reproductive age, likely due to estrogen-mediated reduced adipose tissue inflammation and fibrosis with hypertrophied adipocytes. Depletion of the ubiquitin ligase Siah2 reduced white adipose tissue inflammation and improved glucose metabolism in obese male mice. Siah2 is a transcriptional target of estrogen, but data is lacking about the effect of Siah2 on adipose tissue of females. We therefore evaluated the impact of Siah2 deficiency on white and brown adipose tissue in females of reproductive age. Methods Body composition, adipose tissue morphology, brown adipose tissue gene, and protein expression and adipocyte sizing were evaluated in wild-type and Siah2KO female and male mice fed a low-fat or high-fat diet. Glucose and insulin tolerance, fasting glucose, insulin, fatty acids and triglycerides, and gene expression of inflammation markers in perigonadal fat were evaluated in wild-type and Siah2KO female mice. Microarray analysis of brown fat gene expression was carried out in both sexes. Statistical analysis was assessed by unpaired two-tailed t test and repeated measures ANOVA. Results Siah2 deficiency improves glucose and insulin tolerance in the presence of hypertrophied white adipocytes in high-fat-fed female mice with percent fat comparable to male mice. While previous studies showed Siah2KO reduces the white adipose tissue inflammatory response in male mice, the response in females is biased toward the upregulation of M2-like markers in white adipose tissue. In contrast, loss of Siah2 leads to increased whitening of brown fat in males, but not in females. This corresponded to increased expression of markers of inflammation (F4/80, Ccl2) and thermogenic genes (Pgc1alpha, Dio2, Ucp-1) and proteins (PGC-1α, UCP-1) in females. Contrary to expectations, increased expression of thermogenic markers in females was coupled with a downregulation of ERalpha and ERRgamma protein levels. Conclusions The most striking sex-related effect of Siah2 deficiency is reduced whitening of brown fat in high-fat-fed females. Protection from accumulating unilocular adipocytes in the brown fat corresponds to increased expression of thermogenic genes and proteins in female, but not in male mice. These results raise the possibility that Siah2 contributes to the estrogen-related effects on brown fat function in males and females. Electronic supplementary material The online version of this article (10.1186/s13293-019-0233-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sujoy Ghosh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.,Cardiovascular and Metabolic Disease Program and Center for Computational Biology, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Jessica L Taylor
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Tamra M Mendoza
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Thanh Dang
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - David H Burk
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Yongmei Yu
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Gail Kilroy
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | |
Collapse
|
36
|
Nerurkar PV, Orias D, Soares N, Kumar M, Nerurkar VR. Momordica charantia (bitter melon) modulates adipose tissue inflammasome gene expression and adipose-gut inflammatory cross talk in high-fat diet (HFD)-fed mice. J Nutr Biochem 2019; 68:16-32. [PMID: 31005847 DOI: 10.1016/j.jnutbio.2019.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 02/19/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
Systemic and tissue-specific inflammation has a profound influence on regulation of metabolism, and therefore, strategies to reduce inflammation are of special interest in prevention and treatment of obesity and type 2 diabetes (T2D). Antiobesity and antidiabetic properties of Momordica charantia (bitter melon, BM) have been linked to its protective effects on inflammation and gut microbial dysbiosis. We investigated the mechanisms by which freeze-dried BM juice reduces adipose inflammation in mice fed a 60% high-fat diet (HFD) for 16 weeks. Although earlier studies indicated that BM inhibited recruitment of macrophages (Mφ) infiltration in adipose tissue of rodents and reduced NF-kB and IL-1β secretions, the mechanisms remain unknown. We demonstrate that freeze-dried BM juice inhibits recruitment of Mφ into adipose tissue and its polarization to inflammatory phenotype possibly due to reduction of sphingokinase 1 (SPK1) mRNA in HFD-fed mice. Furthermore, reduction of IL-1β secretion by freeze-dried BM juice in the adipose tissue of HFD-fed mice is correlated to alleviation of NLRP3 inflammasome components and their downstream signaling targets. We confirm previous observations that BM inhibited inflammation of colon and gut microbial dysbiosis in HFD-fed mice, which in part may be associated with the observed anti-inflammatory effects in adipose tissue if HFD-fed mice. Overall, functional foods such as BM may offer potential dietary interventions that may impact sterile inflammatory diseases such as obesity and T2D.
Collapse
Affiliation(s)
- Pratibha V Nerurkar
- Laboratory of Metabolic Disorders and Alternative Medicine, Department of Molecular Biosciences and Bioengineering (MBBE), College of Tropical Agriculture and Human Resources (CTAHR), University of Hawaii at Manoa, Honolulu, HI 96822, USA.
| | - Daniella Orias
- Laboratory of Metabolic Disorders and Alternative Medicine, Department of Molecular Biosciences and Bioengineering (MBBE), College of Tropical Agriculture and Human Resources (CTAHR), University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Natasha Soares
- Laboratory of Metabolic Disorders and Alternative Medicine, Department of Molecular Biosciences and Bioengineering (MBBE), College of Tropical Agriculture and Human Resources (CTAHR), University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Mukesh Kumar
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Vivek R Nerurkar
- Department of Tropical Medicine, Medical Microbiology and Pharmacology; Pacific Center for Emerging Infectious Diseases Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| |
Collapse
|
37
|
White, brown, beige and pink: A rainbow in the adipose organ. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2019. [DOI: 10.1016/j.coemr.2018.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
Papalou O, Kandaraki EA, Papadakis G, Diamanti-Kandarakis E. Endocrine Disrupting Chemicals: An Occult Mediator of Metabolic Disease. Front Endocrinol (Lausanne) 2019; 10:112. [PMID: 30881345 PMCID: PMC6406073 DOI: 10.3389/fendo.2019.00112] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs), a heterogeneous group of exogenous chemicals that can interfere with any aspect of endogenous hormones, represent an emerging global threat for human metabolism. There is now considerable evidence that the observed upsurge of metabolic disease cannot be fully attributed to increased caloric intake, physical inactivity, sleep deficit, and ageing. Among environmental factors implicated in the global deterioration of metabolic health, EDCs have drawn the biggest attention of scientific community, and not unjustifiably. EDCs unleash a coordinated attack toward multiple components of human metabolism, including crucial, metabolically-active organs such as hypothalamus, adipose tissue, pancreatic beta cells, skeletal muscle, and liver. Specifically, EDCs' impact during critical developmental windows can promote the disruption of individual or multiple systems involved in metabolism, via inducing epigenetic changes that can permanently alter the epigenome in the germline, enabling changes to be transmitted to the subsequent generations. The clear effect of this multifaceted attack is the manifestation of metabolic disease, clinically expressed as obesity, metabolic syndrome, diabetes mellitus, and non-alcoholic fatty liver disease. Although limitations of EDCs research do exist, there is no doubt that EDCs constitute a crucial parameter of the global deterioration of metabolic health we currently encounter.
Collapse
Affiliation(s)
- Olga Papalou
- Department of Endocrinology & Diabetes, Hygeia Hospital, Athens, Greece
| | | | | | - Evanthia Diamanti-Kandarakis
- Department of Endocrinology & Diabetes, Hygeia Hospital, Athens, Greece
- *Correspondence: Evanthia Diamanti-Kandarakis
| |
Collapse
|
39
|
Gao W, Gao Z, Pu S, Dong Y, Xu X, Yang X, Zhang Y, Fang K, Li J, Yu W, Sun N, Hu L, Xu Q, Cheng Z, Gao Y. The Underlying Regulated Mechanisms of Adipose Differentiation and Apoptosis of Breast Cells after Weaning. Curr Protein Pept Sci 2019; 20:696-704. [PMID: 30678617 DOI: 10.2174/1389203720666190124161652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/14/2019] [Indexed: 11/22/2022]
Abstract
Numerous experimental studies have demonstrated that a series of remodeling processes occurred in the adipose tissue during the weaning, such as differentiation. Fibroblasts in the breast at weaning stage could re-differentiate into mature adipocytes. Many transcriptional factors were involved in these processes, especially the PPARγ, C/EBP, and SREBP1. There is cell apoptosis participating in the breast tissue degeneration and secretory epithelial cells loss during weaning. In addition, hormones, especially the estrogen and pituitary hormone, play a vital role in the whole reproductive processes. In this review, we mainly focus on the underlying regulated mechanisms of differentiation of adipose tissue and apoptosis of breast cell to provide a specific insight into the physiological changes during weaning.
Collapse
Affiliation(s)
- Weihang Gao
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhao Gao
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Administration of Sports of Guangdong Province, Guangzhou, Guangdong, 510105, China
| | - Shuqi Pu
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yanbin Dong
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaowen Xu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510405, China
| | - Xingping Yang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Yuan Zhang
- Administration of Sports of Guangdong Province, Guangzhou, Guangdong, 510105, China
| | - Kui Fang
- Administration of Sports of Guangdong Province, Guangzhou, Guangdong, 510105, China
| | - Jie Li
- Administration of Sports of Guangdong Province, Guangzhou, Guangdong, 510105, China
| | - Weijian Yu
- Administration of Sports of Guangdong Province, Guangzhou, Guangdong, 510105, China
| | - Nannan Sun
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510405, China
| | - Ling Hu
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qin Xu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhibin Cheng
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunan, 650201, China
| | - Yong Gao
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| |
Collapse
|
40
|
Zhao C, Sun Q, Tang L, Cao Y, Nourse JL, Pathak MM, Lu X, Yang Q. Mechanosensitive Ion Channel Piezo1 Regulates Diet-Induced Adipose Inflammation and Systemic Insulin Resistance. Front Endocrinol (Lausanne) 2019; 10:373. [PMID: 31263454 PMCID: PMC6584899 DOI: 10.3389/fendo.2019.00373] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Adipocytes function as an energy buffer and undergo significant size and volume changes in response to nutritional cues. This adipocyte plasticity is important for systemic lipid metabolism and insulin sensitivity. Accompanying the adipocyte size and volume changes, the mechanical pressure against cell membrane also changes. However, the role that mechanical pressure plays in lipid metabolism and insulin sensitivity remains to be elucidated. Here we show that Piezo1, a mechanically-activated cation channel stimulated by membrane tension and stretch, was highly expressed in adipocytes. Adipose Piezo1 expression was increased in obese mice. Adipose-specific piezo1 knockout mice (adipose-Piezo1-/-) developed insulin resistance, especially when challenged with a high-fat diet (HFD). Perigonadal white adipose tissue (pgWAT) weight was reduced while pro-inflammatory and lipolysis genes were increased in the pgWAT of HFD-fed adipose-Piezo1-/- mice. The adipose-Piezo1-/- mice also developed hepatic steatosis with elevated expression of fatty acid synthesis genes. In cultured adipocytes, Piezo1 activation decreased, while Piezo1 inhibition elevated pro-inflammatory gene expression. TLR4 antagonist TAK-242 abolished adipocyte inflammation induced by Piezo1 inhibition. Thus, adipose Piezo1 may serve as an adaptive mechanism for adipocyte plasticity restraining pro-inflammatory response in obesity.
Collapse
Affiliation(s)
- Can Zhao
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
- Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, China
| | - Qiushi Sun
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
| | - Lingyi Tang
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
- Department of Cardiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Cao
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
| | - Jamison L. Nourse
- Department of Physiology and Biophysics, Sue and Bill Gross Stem Cell Research Center, Center for Complex Systems Biology, University of California at Irvine, Irvine, CA, United States
| | - Medha M. Pathak
- Department of Physiology and Biophysics, Sue and Bill Gross Stem Cell Research Center, Center for Complex Systems Biology, University of California at Irvine, Irvine, CA, United States
| | - Xiang Lu
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
- Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, China
- *Correspondence: Xiang Lu
| | - Qin Yang
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
- Qin Yang
| |
Collapse
|
41
|
Modulation of aquaporin gene expression by n-3 long-chain PUFA lipid structures in white and brown adipose tissue from hamsters. Br J Nutr 2018; 120:1098-1106. [DOI: 10.1017/s0007114518002519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractEPA (20 : 5n-3) and DHA (22 : 6n-3) fatty acids have weight-reducing properties with physiological activity depending on their molecular structure – that is, as TAG or ethyl esters (EE). Aquaporins (AQP) are membrane protein channels recognised as important players in fat metabolism, but their differential expression in white adipose tissue (WAT) and brown adipose tissue (BAT), as well as their modulation by dietary n-3 long-chain PUFA (LCPUFA) such as EPA and DHA, has never been investigated. In this study, the transcriptional profiles of AQP3, AQP5, AQP7 and selected lipid markers of WAT (subcutaneous and visceral) and BAT (interscapular) from hamsters fed diets containing n-3 LCPUFA in different lipid structures such as fish oil (FO, rich in EPA and DHA in the TAG form) and FO-EE (rich in EPA and DHA in the EE form) were used and compared with linseed oil (LSO) as the reference group. A clear effect of fat depot was observed for AQP3 and leptin (LEP), with the lowest values of mRNA found in BAT relative to WAT. The opposite occurred for PPARα. AQP7 was affected by diet, with FO-fed hamsters having higher mRNA levels compared with LSO-fed hamsters. The relative gene expression of AQP5, adiponectin (ADIPO), GLUT4 and PPARγ was influenced by both fat tissue and diet. Taken together, our results revealed a differential expression profile of AQP and some markers of lipid metabolism in both WAT and BAT in response to feeding n-3 LCPUFA in two different structural formats: TAG v. EE.
Collapse
|
42
|
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
43
|
Moxibustion-Simulating Bipolar Radiofrequency Suppresses Weight Gain and Induces Adipose Tissue Browning via Activation of UCP1 and FGF21 in a Mouse Model of Diet-Induced Obesity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:4737515. [PMID: 30275865 PMCID: PMC6151680 DOI: 10.1155/2018/4737515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/29/2018] [Indexed: 12/13/2022]
Abstract
Background Obesity is a pathological condition associated with various diseases including diabetes, stroke, arthritis, infertility, and heart disease. Moxibustion is widely used to prevent and manage obesity in traditional Asian medicine. We tested our hypothesis that moxibustion-simulating bipolar radiofrequency (M-RF) can suppress total body and white adipose tissue (WAT) weight gain via induction of WAT browning in a mouse model of diet-induced obesity (DIO). Methods We designed an M-RF device that could accurately adjust the depth and temperature at which heat stimulation was administered into the abdomen of DIO mice. High-fat-fed male C57BL/6 mice were treated with the M-RF device every two or three days for three weeks. We then harvested WAT and serum from the mice and measured total body and WAT weight, size of adipocytes, mitochondrial contents, features of the dead adipocyte environment, and levels of uncoupling protein 1 (UCP1) and fibroblast growth factor 21 (FGF21). Results Heat stimulation by M-RF in DIO mice resulted in precise temperature adjustment in the mice abdomen, with variance less than 1°C. Additionally, M-RF stimulation inhibited body and WAT weight gain, resulting in increased formation of beige adipocytes, increased mitochondrial content, and decreased formation of dead adipocytes in WAT. Moreover, treatment of M-RF induced expression of UCP1 and FGF21 in serum and/or epididymal WATs in DIO mice. Conclusion Heat stimulation by M-RF treatment induced upregulation of UCP1 and FGF21 expression in serum and/or WATs, which was correlated with reduced total body and WAT weight gain in DIO mice.
Collapse
|
44
|
Abstract
Adipocytes are lipid-rich parenchymal cells contained in a very plastic organ, whose composition can undergo striking physiologic changes. In standard conditions the organ contains white and brown adipocytes which play opposite roles: lipid storage to meet metabolic requirements and lipid burning for thermogenesis, respectively. During chronic cold exposure, white adipocytes transdifferentiate to brown, to increase thermogenesis, whereas in conditions of chronic positive energy balance brown adipocytes transdifferentiate to white, to increase energy stores. During pregnancy, lactation, and post-lactation, subcutaneous white adipocytes convert to milk-producing glands formed by lipid-rich elements that can be defined as pink adipocytes. Recent fate-mapping data support the conversion of pink to brown adipocytes and the reversible conversion of brown adipocytes to myoepithelial cells of alveoli.
Collapse
Affiliation(s)
- Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, University of Ancona (Politecnica delle Marche), Via Tronto 10a, 60020 Ancona, Italy.
| |
Collapse
|
45
|
Hou Y, Liu Z, Zuo Z, Gao T, Fu J, Wang H, Xu Y, Liu D, Yamamoto M, Zhu B, Zhang Y, Andersen ME, Zhang Q, Pi J. Adipocyte-specific deficiency of Nfe2l1 disrupts plasticity of white adipose tissues and metabolic homeostasis in mice. Biochem Biophys Res Commun 2018; 503:264-270. [DOI: 10.1016/j.bbrc.2018.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/07/2018] [Indexed: 01/10/2023]
|
46
|
Carpentier AC, Blondin DP, Virtanen KA, Richard D, Haman F, Turcotte ÉE. Brown Adipose Tissue Energy Metabolism in Humans. Front Endocrinol (Lausanne) 2018; 9:447. [PMID: 30131768 PMCID: PMC6090055 DOI: 10.3389/fendo.2018.00447] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/20/2018] [Indexed: 12/16/2022] Open
Abstract
The demonstration of metabolically active brown adipose tissue (BAT) in humans primarily using positron emission tomography coupled to computed tomography (PET/CT) with the glucose tracer 18-fluorodeoxyglucose (18FDG) has renewed the interest of the scientific and medical community in the possible role of BAT as a target for the prevention and treatment of obesity and type 2 diabetes (T2D). Here, we offer a comprehensive review of BAT energy metabolism in humans. Considerable advances in methods to measure BAT energy metabolism, including nonesterified fatty acids (NEFA), chylomicron-triglycerides (TG), oxygen, Krebs cycle rate, and intracellular TG have led to very good quantification of energy substrate metabolism per volume of active BAT in vivo. These studies have also shown that intracellular TG are likely the primary energy source of BAT upon activation by cold. Current estimates of BAT's contribution to energy expenditure range at the lower end of what would be potentially clinically relevant if chronically sustained. Yet, 18FDG PET/CT remains the gold-standard defining method to quantify total BAT volume of activity, used to calculate BAT's total energy expenditure. Unfortunately, BAT glucose metabolism better reflects BAT's insulin sensitivity and blood flow. It is now clear that most glucose taken up by BAT does not fuel mitochondrial oxidative metabolism and that BAT glucose uptake can therefore be disconnected from thermogenesis. Furthermore, BAT thermogenesis is efficiently recruited upon repeated cold exposure, doubling to tripling its total oxidative capacity, with reciprocal reduction of muscle thermogenesis. Recent data suggest that total BAT volume may be much larger than the typically observed 50-150 ml with 18FDG PET/CT. Therefore, the current estimates of total BAT thermogenesis, largely relying on total BAT volume using 18FDG PET/CT, may underestimate the true contribution of BAT to total energy expenditure. Quantification of the contribution of BAT to energy expenditure begs for the development of more integrated whole body in vivo methods.
Collapse
Affiliation(s)
- André C. Carpentier
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Kirsi A. Virtanen
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland (UEF), Kuopio, Finland
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, QC, Canada
| | - François Haman
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Éric E. Turcotte
- Department of Nuclear Medicine and Radiobiology, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
47
|
Nielsen KN, Peics J, Ma T, Karavaeva I, Dall M, Chubanava S, Basse AL, Dmytriyeva O, Treebak JT, Gerhart-Hines Z. NAMPT-mediated NAD + biosynthesis is indispensable for adipose tissue plasticity and development of obesity. Mol Metab 2018; 11:178-188. [PMID: 29551635 PMCID: PMC6001355 DOI: 10.1016/j.molmet.2018.02.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/21/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Objective The ability of adipose tissue to expand and contract in response to fluctuations in nutrient availability is essential for the maintenance of whole-body metabolic homeostasis. Given the nutrient scarcity that mammals faced for millions of years, programs involved in this adipose plasticity were likely evolved to be highly efficient in promoting lipid storage. Ironically, this previously advantageous feature may now represent a metabolic liability given the caloric excess of modern society. We speculate that nicotinamide adenine dinucleotide (NAD+) biosynthesis exemplifies this concept. Indeed NAD+/NADH metabolism in fat tissue has been previously linked with obesity, yet whether it plays a causal role in diet-induced adiposity is unknown. Here we investigated how the NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT) supports adipose plasticity and the pathological progression to obesity. Methods We utilized a newly generated Nampt loss-of-function model to investigate the tissue-specific and systemic metabolic consequences of adipose NAD+ deficiency. Energy expenditure, glycemic control, tissue structure, and gene expression were assessed in the contexts of a high dietary fat burden as well as the transition back to normal chow diet. Results Fat-specific Nampt knockout (FANKO) mice were completely resistant to high fat diet (HFD)-induced obesity. This was driven in part by reduced food intake. Furthermore, HFD-fed FANKO mice were unable to undergo healthy expansion of adipose tissue mass, and adipose depots were rendered fibrotic with markedly reduced mitochondrial respiratory capacity. Yet, surprisingly, HFD-fed FANKO mice exhibited improved glucose tolerance compared to control littermates. Removing the HFD burden largely reversed adipose fibrosis and dysfunction in FANKO animals whereas the improved glucose tolerance persisted. Conclusions These findings indicate that adipose NAMPT plays an essential role in handling dietary lipid to modulate fat tissue plasticity, food intake, and systemic glucose homeostasis. Fat-specific Nampt knockout (FANKO) does not alter body composition on chow diet. NAMPT is essential for adipose expansion and weight gain from high dietary fat. Loss of adipose NAD+ decreases food intake and improves glucose tolerance. High fat diet-induced metabolic dysfunction in FANKO mice is reversible.
Collapse
Affiliation(s)
- Karen Nørgaard Nielsen
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Julia Peics
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tao Ma
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Iuliia Karavaeva
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Morten Dall
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sabina Chubanava
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Astrid L Basse
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Laboratory of Neural Plasticity, Institute of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark; Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, Copenhagen University Hospital, 2400 Copenhagen, Denmark
| | - Jonas T Treebak
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Zachary Gerhart-Hines
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
48
|
Kuda O, Rossmeisl M, Kopecky J. Omega-3 fatty acids and adipose tissue biology. Mol Aspects Med 2018; 64:147-160. [PMID: 29329795 DOI: 10.1016/j.mam.2018.01.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 12/16/2022]
Abstract
This review provides evidence for the importance of white and brown adipose tissue (i.e. WAT and BAT) function for the maintenance of healthy metabolic phenotype and its preservation in response to omega-3 polyunsaturated fatty acids (omega-3 PUFA), namely in the context of diseased states linked to aberrant accumulation of body fat, systemic low-grade inflammation, dyslipidemia and insulin resistance. More specifically, the review deals with (i) the concept of immunometabolism, i.e. how adipose-resident immune cells and adipocytes affect each other and define the immune-metabolic interface; and (ii) the characteristic features of "healthy adipocytes" in WAT, which are relatively small fat cells endowed with a high capacity for mitochondrial oxidative phosphorylation, triacylglycerol/fatty acid (TAG/FA) cycling and de novo lipogenesis (DNL). The intrinsic metabolic features of WAT and their flexible regulations, reflecting the presence of "healthy adipocytes", provide beneficial local and systemic effects, including (i) protection against in situ endoplasmic reticulum stress and related inflammatory response during activation of adipocyte lipolysis; (ii) prevention of ectopic fat accumulation and dyslipidemia caused by increased hepatic VLDL synthesis, as well as prevention of lipotoxic damage of insulin signaling in extra-adipose tissues; and also (iii) increased synthesis of anti-inflammatory and insulin-sensitizing lipid mediators with pro-resolving properties, including the branched fatty acid esters of hydroxy fatty acids (FAHFAs), also depending on the activity of DNL in WAT. The "healthy adipocytes" phenotype can be induced in WAT of obese mice in response to various stimuli including dietary omega-3 PUFA, especially when combined with moderate calorie restriction, and possibly also with other life style (e.g. physical activity) or pharmacological (e.g. thiazolidinediones) interventions. While omega-3 PUFA could exert beneficial systemic effects by improving immunometabolism of WAT without a concomitant induction of BAT, it is currently not clear whether the metabolic effects of the combined intervention using omega-3 PUFA and calorie restriction or thiazolidinediones depend also on the activation of BAT function and/or the induction of brite/beige adipocytes in WAT. It remains to be established why omega-3 PUFA intervention in type 2 diabetic subjects does not improve insulin sensitivity and glucose homeostasis despite inducing various anti-inflammatory mediators in WAT, including the recently discovered docosahexaenoyl esters of hydroxy linoleic acid, the lipokines from the FAHFA family, as well as several endocannabinoid-related anti-inflammatory lipids. To answer the question whether and to which extent omega-3 PUFA supplementation could promote the formation of "healthy adipocytes" in WAT of human subjects, namely in the obese insulin-resistant patients, represents a challenging task that is of great importance for the treatment of some serious non-communicable diseases.
Collapse
Affiliation(s)
- Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Martin Rossmeisl
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic.
| |
Collapse
|
49
|
Nam H, Jung H, Kim Y, Kim B, Kim KH, Park SJ, Suh JG. Aged black garlic extract regulates lipid metabolism by inhibiting lipogenesis and promoting lipolysis in mature 3T3-L1 adipocytes. Food Sci Biotechnol 2017; 27:575-579. [PMID: 30263782 DOI: 10.1007/s10068-017-0268-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/30/2017] [Accepted: 11/15/2017] [Indexed: 12/28/2022] Open
Abstract
As the mechanism of aged black garlic (ABG) extract affecting lipid metabolism in adipocytes remains unclear, this study evaluated the effect of ABG extract on lipid metabolism and the expression of related proteins in mature 3T3-L1 adipocytes. ABG extract treatments at 0, 0.625, 1.25, and 2.5, and 5 mg/mL had no effect on cell morphology or viability in adipocytes. ABG extract suppressed lipogenesis and induced lipolysis in a dose-dependent manner compared to control. Furthermore, ABG extract at 2.5 and 5 mg/mL significantly reduced protein expression of proliferator activated receptor γ (PPARγ) and perilipin in mature 3T3-L1 adipocytes. The hormone-sensitive lipase (HSL) and Ser563-pHSL levels were also significantly reduced by treatment with 5 mg/mL of ABG extract. Taken together, these results suggest that ABG extract has anti-lipogenic and lipolytic effects in mature 3T3-L1 adipocytes, indicating a potential in anti-obesity therapies.
Collapse
Affiliation(s)
- Hajin Nam
- 1Department of Medical Genetics, College of Medicine, Hallym University, 1 Hallymdaehakgil, Chuncheon, Gangwon-do 24252 Korea
| | - Harry Jung
- 1Department of Medical Genetics, College of Medicine, Hallym University, 1 Hallymdaehakgil, Chuncheon, Gangwon-do 24252 Korea
| | - Yooyeon Kim
- 1Department of Medical Genetics, College of Medicine, Hallym University, 1 Hallymdaehakgil, Chuncheon, Gangwon-do 24252 Korea
| | - Boyoung Kim
- 1Department of Medical Genetics, College of Medicine, Hallym University, 1 Hallymdaehakgil, Chuncheon, Gangwon-do 24252 Korea
| | - Kyeong Hee Kim
- RND Team, Medience Co. Ltd, Chuncheon, Gangwon-do 19853 Korea
| | - Sang Jae Park
- RND Team, Medience Co. Ltd, Chuncheon, Gangwon-do 19853 Korea
| | - Jun Gyo Suh
- 1Department of Medical Genetics, College of Medicine, Hallym University, 1 Hallymdaehakgil, Chuncheon, Gangwon-do 24252 Korea
| |
Collapse
|
50
|
Schlüter A, Horstmann M, Diaz-Cano S, Plöhn S, Stähr K, Mattheis S, Oeverhaus M, Lang S, Flögel U, Berchner-Pfannschmidt U, Eckstein A, Banga JP. Genetic immunization with mouse thyrotrophin hormone receptor plasmid breaks self-tolerance for a murine model of autoimmune thyroid disease and Graves' orbitopathy. Clin Exp Immunol 2017; 191:255-267. [PMID: 29058307 DOI: 10.1111/cei.13075] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2017] [Indexed: 01/08/2023] Open
Abstract
Experimental models of Graves' hyperthyroid disease accompanied by Graves' orbitopathy (GO) can be induced efficiently in susceptible inbred strains of mice by immunization by electroporation of heterologous human TSH receptor (TSHR) A-subunit plasmid. In this study, we report on the development of a bona fide murine model of autoimmune Graves' disease induced with homologous mouse TSHR A-subunit plasmid. Autoimmune thyroid disease in the self-antigen model was accompanied by GO and characterized by histopathology of hyperplastic glands with large thyroid follicular cells. Examination of orbital tissues showed significant inflammation in extra-ocular muscle with accumulation of T cells and macrophages together with substantial deposition of adipose tissue. Notably, increased levels of brown adipose tissue were present in the orbital tissue of animals undergoing experimental GO. Further analysis of inflammatory loci by 19 F-magnetic resonance imaging showed inflammation to be confined to orbital muscle and optic nerve, but orbital fat showed no difference in inflammatory signs in comparison to control β-Gal-immunized animals. Pathogenic antibodies induced to mouse TSHR were specific for the self-antigen, with minimal cross-reactivity to human TSHR. Moreover, compared to other self-antigen models of murine Graves' disease induced in TSHR knock-out mice, the repertoire of autoantibodies to mouse TSHR generated following the breakdown of thymic self-tolerance is different to those that arise when tolerance is not breached immunologically, as in the knock-out models. Overall, we show that mouse TSHR A-subunit plasmid immunization by electroporation overcomes tolerance to self-antigen to provide a faithful model of Graves' disease and GO.
Collapse
Affiliation(s)
- A Schlüter
- Molecular Ophthalmology, Departments of Ophthalmology University Hospital Essen, Germany.,Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Essen, Essen, Germany
| | - M Horstmann
- Molecular Ophthalmology, Departments of Ophthalmology University Hospital Essen, Germany
| | - S Diaz-Cano
- Department of Histopathology, King's College Hospital NHS, London, UK
| | - S Plöhn
- Molecular Ophthalmology, Departments of Ophthalmology University Hospital Essen, Germany
| | - K Stähr
- Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Essen, Essen, Germany
| | - S Mattheis
- Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Essen, Essen, Germany
| | - M Oeverhaus
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - S Lang
- Oto-Rhino-Laryngology, Head and Neck Surgery, University Hospital Essen, Essen, Germany
| | - U Flögel
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - A Eckstein
- Molecular Ophthalmology, Departments of Ophthalmology University Hospital Essen, Germany.,Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - J P Banga
- Molecular Ophthalmology, Departments of Ophthalmology University Hospital Essen, Germany
| |
Collapse
|