1
|
van Eeghen SA, Pyle L, Narongkiatikhun P, Choi YJ, Obeid W, Parikh CR, Vosters TG, van Valkengoed IG, Krebber MM, Touw DJ, den Heijer M, Bjornstad P, van Raalte DH, Nokoff NJ. Unveiling mechanisms underlying kidney function changes during sex hormone therapy. J Clin Invest 2025; 135:e190850. [PMID: 40193283 PMCID: PMC12043095 DOI: 10.1172/jci190850] [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: 01/02/2025] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUNDMen with chronic kidney disease (CKD) experience faster kidney function decline than women. Studies in individuals undergoing sex hormone therapy suggest a role for sex hormones, as estimated glomerular filtration rate (eGFR) increases with feminizing therapy and decreases with masculinizing therapy. However, effects on measured GFR (mGFR), glomerular and tubular function, and involved molecular mechanisms remain unexplored.METHODSThis prospective, observational study included individuals initiating feminizing (estradiol and antiandrogens; n = 23) or masculinizing (testosterone; n = 21) therapy. Baseline and 3-month assessments included mGFR (iohexol clearance), kidney perfusion (para-aminohippuric acid clearance), tubular injury biomarkers, and plasma proteomics.RESULTSDuring feminizing therapy, mGFR and kidney perfusion increased (+3.6% and +9.1%, respectively; P < 0.05) without increased glomerular pressure. Tubular injury biomarkers, including urine neutrophil gelatinase-associated lipocalin, epidermal growth factor (EGF), monocyte chemoattractant protein-1, and chitinase 3-like protein 1 (YKL-40), decreased significantly (-53%, -42%, -45%, and -58%, respectively). During masculinizing therapy, mGFR and kidney perfusion remained unchanged, but urine YKL-40 and plasma tumor necrosis factor receptor 1 (TNFR-1) increased (+134% and +8%, respectively; P < 0.05). Proteomic analysis revealed differential expression of 49 proteins during feminizing and 356 proteins during masculinizing therapy. Many kidney-protective proteins were positively associated with estradiol and negatively associated with testosterone, including proteins involved in endothelial function (SFRP4, SOD3), inflammation reduction (TSG-6), and maintaining kidney tissue structure (agrin).CONCLUSIONSex hormones influence kidney physiology, with estradiol showing protective effects on glomerular and tubular function, while testosterone predominantly exerts opposing effects. These findings emphasize the role of sex hormones in sexual dimorphism observed in kidney function and physiology and suggest new approaches for sex-specific precision medicine.TRIAL REGISTRATIONDutch Trial Register (ID: NL9517); ClinicalTrials.gov (ID: NCT04482920).
Collapse
Affiliation(s)
- Sarah A. van Eeghen
- Center of Expertise on Gender Dysphoria, Department of Internal Medicine, Amsterdam UMC, Location VU University, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam, Netherlands
- Department of Internal Medicine, Endocrinology and Metabolism, Amsterdam UMC, Location VU University, Amsterdam, Netherlands
| | - Laura Pyle
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Phoom Narongkiatikhun
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, University of Washington School of Medicine, Seattle, Washington, USA
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Ye Ji Choi
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Wassim Obeid
- Division of Nephrology, Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Chirag R. Parikh
- Division of Nephrology, Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Taryn G. Vosters
- Department of Public and Occupational Health, Amsterdam University Medical Centre, Universiteit van Amsterdam, Amsterdam, Netherlands
| | - Irene G.M. van Valkengoed
- Department of Public and Occupational Health, Amsterdam University Medical Centre, Universiteit van Amsterdam, Amsterdam, Netherlands
| | - Merle M. Krebber
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, Netherlands
| | - Daan J. Touw
- Department of Clinical Pharmacy & Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Martin den Heijer
- Center of Expertise on Gender Dysphoria, Department of Internal Medicine, Amsterdam UMC, Location VU University, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam, Netherlands
- Department of Internal Medicine, Endocrinology and Metabolism, Amsterdam UMC, Location VU University, Amsterdam, Netherlands
| | - Petter Bjornstad
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, University of Washington School of Medicine, Seattle, Washington, USA
| | - Daniël H. van Raalte
- Department of Internal Medicine, Endocrinology and Metabolism, Amsterdam UMC, Location VU University, Amsterdam, Netherlands
- Diabetes Center, Department of Internal Medicine, Amsterdam UMC, Location VU University, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences Research Institute, VU University, Amsterdam, Netherlands
| | - Natalie J. Nokoff
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
2
|
Domagalski M, Olszańska J, Pietraszek‐Gremplewicz K, Nowak D. The role of adipogenic niche resident cells in colorectal cancer progression in relation to obesity. Obes Rev 2025; 26:e13873. [PMID: 39763022 PMCID: PMC11884973 DOI: 10.1111/obr.13873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/03/2024] [Accepted: 11/05/2024] [Indexed: 03/08/2025]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and has one of the highest mortality rates. Considering its nonlinear etiology, many risk factors are associated with CRC formation and development, with obesity at the forefront. Obesity is regarded as one of the key environmental risk determinants for the pathogenesis of CRC. Excessive food intake and a sedentary lifestyle, together with genetic predispositions, lead to the overgrowth of adipose tissue along with a disruption in the number and function of its building cells. Adipose tissue-resident cells may constitute part of the CRC microenvironment. Alterations in their physiology and secretory profiles observed in obesity may further contribute to CRC progression, and despite similar localization, their contributions are not equivalent. They can interact with CRC cells, either directly or indirectly, influencing various processes that contribute to tumorigenesis. The main aim of this review is to provide insights into the diversity of adipose tissue resident cells, namely, adipocytes, adipose stromal cells, and immunological cells, regarding the role of particular cell types in co-forming the CRC microenvironment. The scope of this study was also devoted to the abnormalities in adipose tissue physiology observed in obesity states and their impact on CRC development.
Collapse
Affiliation(s)
- Mikołaj Domagalski
- Department of Cell Pathology, Faculty of BiotechnologyUniversity of WroclawWroclawPoland
| | - Joanna Olszańska
- Department of Cell Pathology, Faculty of BiotechnologyUniversity of WroclawWroclawPoland
| | | | - Dorota Nowak
- Department of Cell Pathology, Faculty of BiotechnologyUniversity of WroclawWroclawPoland
| |
Collapse
|
3
|
Ziqubu K, Mazibuko-Mbeje SE, Dludla PV. Regulation of adipokine and batokine secretion by dietary flavonoids, as a prospective therapeutic approach for obesity and its metabolic complications. Biochimie 2025; 230:95-113. [PMID: 39551425 DOI: 10.1016/j.biochi.2024.11.007] [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: 08/17/2024] [Revised: 10/27/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024]
Abstract
Traditionally recognised as the energy reservoir and main site of adaptive thermogenesis, white and brown adipose tissues are complex endocrine organs regulating systemic energy metabolism via the secretion of bioactive molecules, termed "adipokines" and "batokines", respectively. Due to its significant role in regulating whole-body energy metabolism and other physiological processes, adipose tissue has been increasingly explored as a feasible therapeutic target for obesity. Flavonoids are one of the most significant plant polyphenolic compounds holding a great potential as therapeutic agents for combating obesity. However, understanding their mechanisms of action remains largely insufficient to formulate therapeutic theories. This review critically discusses scientific evidence highlighting the role of flavonoids in ameliorating obesity-related metabolic complications, including adipose tissue dysfunction, inflammation, insulin resistance, hepatic steatosis, and cardiovascular comorbidities in part by modulating the release of adipokines and batokines. Further discussion advocates for the use of therapeutics targeting these bioactive molecules as a potential avenue for developing effective treatment for obesity and its adverse metabolic diseases such as type 2 diabetes.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | | | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| |
Collapse
|
4
|
Pomar CA, Trepiana J, Besné-Eseverri I, Castillo P, Palou A, Palou M, Portillo MP, Picó C. Maternal Dietary Improvement or Leptin Supplementation During Suckling Mitigates the Long-Term Impact of Maternal Obesogenic Conditions on Inflammatory and Oxidative Stress Biomarkers in the Offspring of Diet-Induced Obese Rats. Int J Mol Sci 2024; 25:11876. [PMID: 39595945 PMCID: PMC11594198 DOI: 10.3390/ijms252211876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates the impact of maternal nutrition during lactation on inflammation and oxidative stress in the offspring of diet-induced obese rats, along with the potential benefits of leptin supplementation during suckling. Dams were fed either a standard diet (SD), a western diet (WD) before and during gestation and lactation (WD-dams), or a WD switched to an SD during lactation (Rev-dams). Offspring were supplemented with leptin or vehicle during suckling and then fed an SD or WD until four months. Offspring of the Rev-dams exhibited improved metabolic indicators, including lower body weight, reduced plasma levels of TNF-alpha, a higher adiponectin/leptin (A/L) ratio, enhanced liver antioxidant defenses, and decreased inflammation markers in white adipose tissue (WAT) compared to WD-dams, with sex differences. Leptin supplementation further modulated these markers, reducing oxidative stress in liver and inflammation in WAT and liver (e.g., hepatic Tnfa expression decreased by 45% (males) and 41% (females) in the WD group on an SD), and improving the A/L ratio, with effects varying by maternal conditions and sex. In conclusion, this study underscores the importance of maternal nutrition and leptin intake during suckling in shaping long-term metabolic and inflammatory health in offspring, offering strategies to mitigate the adverse effects of maternal obesity on future generations.
Collapse
Affiliation(s)
- Catalina Amadora Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
| | - Jenifer Trepiana
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain
- BIOARABA Institute of Health, 01006 Vitoria-Gasteiz, Spain
| | - Irene Besné-Eseverri
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain
| | - Pedro Castillo
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), 07122 Palma, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
| | - Maria P. Portillo
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain
- BIOARABA Institute of Health, 01006 Vitoria-Gasteiz, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (P.C.); (A.P.); (C.P.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain; (J.T.); (I.B.-E.); (M.P.P.)
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), 07122 Palma, Spain
| |
Collapse
|
5
|
Yao X, Wan R, Li C, Li G, Zhang B, Deng Z, Li H. The hypoglycemic effect of enzymatic modified dietary fiber from bamboo shoot on type 2 diabetes rats. J Food Sci 2024; 89:5900-5911. [PMID: 39150747 DOI: 10.1111/1750-3841.17258] [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: 04/10/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 08/17/2024]
Abstract
Bamboo shoot is a healthy food rich in dietary fiber (DF). However, its highly insoluble DF and fibrous texture limit its application in industrially processed foods. To achieve industrial processing of bamboo shoot, cellulase was used to improve the physical characteristics of bamboo shoot DF in this study. After enzymatic hydrolysis, the content of soluble DF (SDF) of bamboo shoot increased by 99.28% (from 5.53% to 11.02%) significantly (p < 0.01). At the same time, the effect of enzymatic-modified bamboo SDF (EMBSDF) on streptozotocin-induced type 2 diabetes rats was explored. Results demonstrated that the high dose of EMBSDF (312.8 mg/kg) treated rats showed significant improvements in terms of glucose tolerance and insulin sensitivity (p < 0.01) compared with the diabetes rats. Meantime, it was observed that the levels of glucagon-like peptide-1, adiponectin and interleukin-4 of high dose of EMBSDF compared with diabetes rats were increased (p < 0.01) by 57.79%, 159.13%, and 6.17%, respectively. The tumor necrosis factor-α, C-reactive protein, and leptin levels were decreased (p < 0.01) by 62.89%, 31.53%, and 7.84%, respectively. Furthermore, apparent kidney and pancreas histology improvements were found in high-dose and mid-dose EMBSDF-treated diabetes rats. These results indicated that the modified DF significantly improved diabetes.
Collapse
Affiliation(s)
- Xiangjie Yao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, China
| | - Renkou Wan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, China
| | - Chunxiao Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, China
| | - Gongjing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
Madan A, Kelly KP, Bahk P, Sullivan CE, Poling ME, Brent AE, Alassaf M, Dubrulle J, Rajan A. Atg8/LC3 controls systemic nutrient surplus signaling in flies and humans. Curr Biol 2024; 34:3327-3341.e9. [PMID: 38955177 PMCID: PMC11303106 DOI: 10.1016/j.cub.2024.06.005] [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: 05/31/2023] [Revised: 04/26/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Organisms experience constant nutritional flux. Mechanisms at the interface of opposing nutritional states-scarcity and surplus-enable organismal energy homeostasis. Contingent on nutritional stores, adipocytes secrete adipokines, such as the fat hormone leptin, to signal nutrient status to the central brain. Increased leptin secretion underlies metabolic dysregulation during common obesity, but the molecular mechanisms regulating leptin secretion from human adipocytes are poorly understood. Here, we report that Atg8/LC3 family proteins, best known for their role in autophagy during nutrient scarcity, play an evolutionarily conserved role during nutrient surplus by promoting adipokine secretion. We show that in a well-fed state, Atg8/LC3 promotes the secretion of the Drosophila functional leptin ortholog unpaired 2 (Upd2) and leptin from human adipocytes. Proteomic analyses reveal that LC3 directs leptin to a secretory pathway in human cells. We identified LC3-dependent extracellular vesicle (EV) loading and secretion (LDELS) as a required step for leptin release, highlighting a unique secretory route adopted by leptin in human adipocytes. In Drosophila, mutations to Upd2's Atg8 interaction motif (AIM) result in constitutive adipokine retention. Atg8-mediated Upd2 retention alters lipid storage and hunger response and rewires the bulk organismal transcriptome in a manner conducive to starvation survival. Thus, Atg8/LC3's bidirectional role in nutrient sensing-conveying nutrient surplus and responding to nutrient deprivation-enables organisms to manage nutrient flux effectively. We posit that decoding how bidirectional molecular switches-such as Atg8/LC3-operate at the nexus of nutritional scarcity and surplus will inform therapeutic strategies to tackle chronic metabolic disorders.
Collapse
Affiliation(s)
- Aditi Madan
- Basic Sciences Division, Fred Hutch, Seattle, WA 98109, USA
| | - Kevin P Kelly
- Basic Sciences Division, Fred Hutch, Seattle, WA 98109, USA
| | - Patrick Bahk
- Basic Sciences Division, Fred Hutch, Seattle, WA 98109, USA
| | | | | | - Ava E Brent
- Basic Sciences Division, Fred Hutch, Seattle, WA 98109, USA
| | - Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, WA 98109, USA
| | - Julien Dubrulle
- Cellular Imaging Core, Shared Resources, Fred Hutch, Seattle, WA 98109, USA
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, WA 98109, USA.
| |
Collapse
|
7
|
Baird HJM, Shun-Shion AS, Mendes de Oliveira E, Stalder D, Liang L, Eden J, Chambers JE, Farooqi IS, Gershlick DC, Fazakerley DJ. A quantitative pipeline to assess secretion of human leptin coding variants reveals mechanisms underlying leptin deficiencies. J Biol Chem 2024; 300:107562. [PMID: 39002670 PMCID: PMC11366920 DOI: 10.1016/j.jbc.2024.107562] [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: 03/11/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024] Open
Abstract
The hormone leptin, primarily secreted by adipocytes, plays a crucial role in regulating whole-body energy homeostasis. Homozygous loss-of-function mutations in the leptin gene (LEP) cause hyperphagia and severe obesity, primarily through alterations in leptin's affinity for its receptor or changes in serum leptin concentrations. Although serum concentrations are influenced by various factors (e.g., gene expression, protein synthesis, stability in the serum), proper delivery of leptin from its site of synthesis in the endoplasmic reticulum via the secretory pathway to the extracellular serum is a critical step. However, the regulatory mechanisms and specific machinery involved in this trafficking route, particularly in the context of human LEP mutations, remain largely unexplored. We have employed the Retention Using Selective Hooks system to elucidate the secretory pathway of leptin. We have refined this system into a medium-throughput assay for examining the pathophysiology of a range of obesity-associated LEP variants. Our results reveal that leptin follows the default secretory pathway, with no additional regulatory steps identified prior to secretion. Through screening of leptin variants, we identified three mutations that lead to proteasomal degradation of leptin and one variant that significantly decreased leptin secretion, likely through aberrant disulfide bond formation. These observations have identified novel pathogenic effects of leptin variants, which can be informative for therapeutics and diagnostics. Finally, our novel quantitative screening platform can be adapted for other secreted proteins.
Collapse
Affiliation(s)
- Harry J M Baird
- Metabolic Research Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Amber S Shun-Shion
- Metabolic Research Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Edson Mendes de Oliveira
- Metabolic Research Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Danièle Stalder
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Lu Liang
- Metabolic Research Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Jessica Eden
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Joseph E Chambers
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - I Sadaf Farooqi
- Metabolic Research Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom.
| | - David C Gershlick
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom.
| | - Daniel J Fazakerley
- Metabolic Research Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
8
|
López-Alcalá J, Gordon A, Trávez A, Tercero-Alcázar C, Correa-Sáez A, González-Rellán MJ, Rangel-Zúñiga OA, Rodríguez A, Membrives A, Frühbeck G, Nogueiras R, Calzado MA, Guzmán-Ruiz R, Malagón MM. Localization, traffic and function of Rab34 in adipocyte lipid and endocrine functions. J Biomed Sci 2024; 31:2. [PMID: 38183057 PMCID: PMC10770960 DOI: 10.1186/s12929-023-00990-8] [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: 03/21/2023] [Accepted: 12/20/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Excessive lipid accumulation in the adipose tissue in obesity alters the endocrine and energy storage functions of adipocytes. Adipocyte lipid droplets represent key organelles coordinating lipid storage and mobilization in these cells. Recently, we identified the small GTPase, Rab34, in the lipid droplet proteome of adipocytes. Herein, we have characterized the distribution, intracellular transport, and potential contribution of this GTPase to adipocyte physiology and its regulation in obesity. METHODS 3T3-L1 and human primary preadipocytes were differentiated in vitro and Rab34 distribution and trafficking were analyzed using markers of cellular compartments. 3T3-L1 adipocytes were transfected with expression vectors and/or Rab34 siRNA and assessed for secretory activity, lipid accumulation and expression of proteins regulating lipid metabolism. Proteomic and protein interaction analyses were employed for the identification of the Rab34 interactome. These studies were combined with functional analysis to unveil the role played by the GTPase in adipocytes, with a focus on the actions conveyed by Rab34 interacting proteins. Finally, Rab34 regulation in response to obesity was also evaluated. RESULTS Our results show that Rab34 localizes at the Golgi apparatus in preadipocytes. During lipid droplet biogenesis, Rab34 translocates from the Golgi to endoplasmic reticulum-related compartments and then reaches the surface of adipocyte lipid droplets. Rab34 exerts distinct functions related to its intracellular location. Thus, at the Golgi, Rab34 regulates cisternae integrity as well as adiponectin trafficking and oligomerization. At the lipid droplets, this GTPase controls lipid accumulation and lipolysis through its interaction with the E1-ubiquitin ligase, UBA1, which induces the ubiquitination and proteasomal degradation of the fatty acid transporter and member of Rab34 interactome, FABP5. Finally, Rab34 levels in the adipose tissue and adipocytes are regulated in response to obesity and related pathogenic insults (i.e., fibrosis). CONCLUSIONS Rab34 plays relevant roles during adipocyte differentiation, including from the regulation of the oligomerization (i.e., biological activity) and secretion of a major adipokine with insulin-sensitizing actions, adiponectin, to lipid storage and mobilization from lipid droplets. Rab34 dysregulation in obesity may contribute to the altered adipokine secretion and lipid metabolism that characterize adipocyte dysfunction in conditions of excess adiposity.
Collapse
Affiliation(s)
- Jaime López-Alcalá
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Ana Gordon
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain.
| | - Andrés Trávez
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Carmen Tercero-Alcázar
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Alejandro Correa-Sáez
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - María Jesús González-Rellán
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Oriol A Rangel-Zúñiga
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Lipids and Atherosclerosis Unit, IMIBIC/University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Amaia Rodríguez
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Metabolic Research Laboratory, Department of Endocrinology & Nutrition, Clinic, University of Navarra, IdiSNA, Pamplona, Spain
| | - Antonio Membrives
- Department of Medical-Surgical Specialties, University of Córdoba (UCO), Reina Sofia University Hospital (HURS), Córdoba, Spain
| | - Gema Frühbeck
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Metabolic Research Laboratory, Department of Endocrinology & Nutrition, Clinic, University of Navarra, IdiSNA, Pamplona, Spain
| | - Rubén Nogueiras
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Marco A Calzado
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
| | - Rocío Guzmán-Ruiz
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain
| | - María M Malagón
- Department of Cell Biology, Physiology, and Immunology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba (UCO), Reina Sofía University Hospital (HURS), Córdoba, Spain.
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Madrid, Spain.
| |
Collapse
|
9
|
Chiang YT, Wu YY, Lin YC, Huang YY, Lu JC. Cyclodextrin-Mediated Cholesterol Depletion Induces Adiponectin Secretion in 3T3-L1 Adipocytes. Int J Mol Sci 2023; 24:14718. [PMID: 37834165 PMCID: PMC10572842 DOI: 10.3390/ijms241914718] [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: 07/31/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Adipocytes store a significant amount of cholesterol and triglycerides. However, whether cholesterol modulates adipocyte function remains largely unknown. We modulated the cholesterol level in adipocytes to examine its effect on the secretion of adiponectin, an important hormone specifically secreted by adipocytes. Treating differentiated 3T3-L1 adipocytes with 4 mM methyl-β-cyclodextrin (MβCD), a molecule with a high affinity for cholesterol, rapidly depleted cholesterol in adipocytes. Interestingly, MβCD treatment increased adiponectin in the medium without affecting its intracellular level, suggesting a modulation of secretion. By contrast, cholesterol addition did not affect adiponectin secretion, suggesting that cholesterol-depletion-induced intracellular cholesterol trafficking, but not reduced cholesterol level, accounted for MβCD-induced adiponectin secretion. MβCD-induced adiponectin secretion was reduced after 10 μg/mL U18666A treatment that suppressed cholesterol transport out of late endosomes/lysosomes. Depleting Niemann-Pick type C1 (NPC1) or NPC2 proteins, which mediate endosomal/lysosomal cholesterol export, consistently reduced MβCD-induced adiponectin secretion. Furthermore, treatment with 1 μM bafilomycin A1, which neutralized acidic endosomes/lysosomes, also attenuated MβCD-induced adiponectin secretion. Finally, MβCD treatment redistributed cellular adiponectin to lower-density fractions in sucrose gradient fractionation. Our results show that MβCD-mediated cholesterol depletion elevates the secretion of adiponectin, highlighting the involvement of endosomes and lysosomes in adiponectin secretion in adipocytes.
Collapse
Affiliation(s)
- Yu-Ting Chiang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ying-Yu Wu
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Chun Lin
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Yao Huang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Juu-Chin Lu
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| |
Collapse
|
10
|
Krishnan SN, Thanasupawat T, Arreza L, Wong GW, Sfanos K, Trock B, Arock M, Shah GG, Glogowska A, Ghavami S, Hombach-Klonisch S, Klonisch T. Human C1q Tumor Necrosis Factor 8 (CTRP8) defines a novel tryptase+ mast cell subpopulation in the prostate cancer microenvironment. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166681. [PMID: 36921737 DOI: 10.1016/j.bbadis.2023.166681] [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: 09/14/2022] [Revised: 01/26/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
The adipokine C1q Tumor Necrosis Factor 8 (CTRP8) is the least known member of the 15 CTRP proteins and a ligand of the relaxin receptor RXFP1. We previously demonstrated the ability of the CTRP8-RXFP1 interaction to promote motility, matrix invasion, and drug resistance. The lack of specific tools to detect CTRP8 protein severely limits our knowledge on CTRP8 biological functions in normal and tumor tissues. Here, we have generated and characterized the first specific antiserum to human CTRP8 which identified CTRP8 as a novel marker of tryptase+ mast cells (MCT) in normal human tissues and in the prostate cancer (PC) microenvironment. Using human PC tissue microarrays composed of neoplastic and corresponding tumor-adjacent prostate tissues, we have identified a significantly higher number of CTRP8+ MCT in the peritumor versus intratumor compartment of PC tissues of Gleason scores 6 and 7. Higher numbers of CTRP8+ MCT correlated with the clinical parameter of biochemical recurrence. We showed that the human MC line ROSAKIT WT expressed RXFP1 transcripts and responded to CTRP8 treatment with a small but significant increase in cell proliferation. Like the cognate RXFP1 ligand RLN-2 and the small molecule RXFP1 agonist ML-290, CTRP8 reduced degranulation of ROSAKIT WT MC stimulated by the Ca2+-ionophore A14187. In conclusion, this is the first report to identify the RXFP1 agonist CTRP8 as a novel marker of MCT and autocrine/paracrine oncogenic factor within the PC microenvironment.
Collapse
Affiliation(s)
- Sai Nivedita Krishnan
- Dept. of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada
| | - Thatchawan Thanasupawat
- Dept. of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada
| | - Leanne Arreza
- Dept. of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada
| | - G William Wong
- Dept. of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Karen Sfanos
- Dept. of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruce Trock
- Dept. of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michel Arock
- Laboratoire d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, Paris, France
| | - G Girish Shah
- Dept. of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, CHU de Quebec-Laval, Quebec, Canada
| | - Aleksandra Glogowska
- Dept. of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada
| | - Saeid Ghavami
- Dept. of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada; Research Institute of Cancer and Hematology, CancerCare Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Sabine Hombach-Klonisch
- Dept. of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada; Dept. of Pathology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.
| | - Thomas Klonisch
- Dept. of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada; Dept. of Pathology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada; Research Institute of Cancer and Hematology, CancerCare Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada; Dept. of Medical Microbiology & Infectious Diseases, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Canada.
| |
Collapse
|
11
|
Roberts BS, Yang CQ, Neher SB. Characterization of lipoprotein lipase storage vesicles in 3T3-L1 adipocytes. J Cell Sci 2022; 135:jcs258734. [PMID: 34382637 PMCID: PMC8403984 DOI: 10.1242/jcs.258734] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/10/2021] [Indexed: 12/12/2022] Open
Abstract
Lipoprotein lipase (LPL) is a secreted triglyceride lipase involved in the clearance of very-low-density lipoproteins and chylomicrons from circulation. LPL is expressed primarily in adipose and muscle tissues and transported to the capillary lumen. LPL secretion is regulated by insulin in adipose tissue; however, few studies have examined the regulatory and trafficking steps involved in secretion. Here, we describe the intracellular localization and insulin-dependent trafficking of LPL in 3T3-L1 adipocytes. We compared LPL trafficking to the better characterized trafficking pathways taken by leptin and GLUT4 (also known as SLC2A4). We show that the LPL trafficking pathway shares some characteristics of these other pathways, but that LPL subcellular localization and trafficking are distinct from those of GLUT4 and leptin. LPL secretion occurs slowly in response to insulin and rapidly in response to the Ca2+ ionophore ionomycin. This regulated trafficking is dependent on Golgi protein kinase D and the ADP-ribosylation factor GTPase ARF1. Together, these data give support to a new trafficking pathway for soluble cargo that is active in adipocytes.
Collapse
Affiliation(s)
| | | | - Saskia B. Neher
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Adiponectin and Asthma: Knowns, Unknowns and Controversies. Int J Mol Sci 2021; 22:ijms22168971. [PMID: 34445677 PMCID: PMC8396527 DOI: 10.3390/ijms22168971] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Adiponectin is an adipokine associated with the healthy obese phenotype. Adiponectin increases insulin sensitivity and has cardio and vascular protection actions. Studies related to adiponectin, a modulator of the innate and acquired immunity response, have suggested a role of this molecule in asthma. Studies based on various asthma animal models and on the key cells involved in the allergic response have provided important insights about this relation. Some of them indicated protection and others reversed the balance towards negative effects. Many of them described the cellular pathways activated by adiponectin, which are potentially beneficial for asthma prevention or for reduction in the risk of exacerbations. However, conclusive proofs about their efficiency still need to be provided. In this article, we will, briefly, present the general actions of adiponectin and the epidemiological studies supporting the relation with asthma. The main focus of the current review is on the mechanisms of adiponectin and the impact on the pathobiology of asthma. From this perspective, we will provide arguments for and against the positive influence of this molecule in asthma, also indicating the controversies and sketching out the potential directions of research to complete the picture.
Collapse
|
13
|
Gaeini Z, Mirmiran P, Bahadoran Z. Effects of Ramadan intermittent fasting on leptin and adiponectin: a systematic review and meta-analysis. Hormones (Athens) 2021; 20:237-246. [PMID: 33786736 DOI: 10.1007/s42000-021-00285-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 03/17/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Ramadan intermittent fasting may affect whole-body metabolism by affecting appetite-related hormones. This systematic review and meta-analysis aimed to clarify the possible effects of Ramadan intermittent fasting on the main hormones regulating appetite and satiety, including leptin and adiponectin. METHODS All English language papers in the PubMed, Scopus, and Embase databases were searched using the keywords "Ramadan fasting", "adiponectin", and "leptin", up to 2020. Data extraction was conducted based on the main data of the studies; the primary outcomes of the analysis were mean changes of adiponectin and leptin levels during the holy month of Ramadan in fasted subjects. MAIN RESULTS Data of 16 eligible studies, conducted between 2003 and 2020, were included in the systematic review. Of these, 10 studies with complete data on leptin and adiponectin were included in the meta-analysis. A significant decrease in leptin levels was observed after Ramadan fasting (WMD = -2.28 ng/ml, 95% CI = -3.72, -0.84). Ramadan fasting had no significant effect on adiponectin levels (WMD = 2.19 ng/ml, 95% CI = -0.29, 4.67). Sub-group analysis demonstrated a greater decrease in leptin levels among normal-weight subjects compared to those of overweight/obese subjects (WMD = -4.67 ng/ml, 95% CI = -6.03, -3.31 vs. WMD = -3.43 ng/ml, 95% CI = -5.69, -1.17). CONCLUSION Ramadan fasting may decrease leptin levels, especially in normal-weight subjects. There was high heterogeneity, which may be explained by the differences between the wide ranges of study conditions.
Collapse
Affiliation(s)
- Zahra Gaeini
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19395-4763, No. 24, Shahid-Erabi St., Yeman St., Velenjak, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Human Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19395-4763, No. 24, Shahid-Erabi St., Yeman St., Velenjak, Tehran, Iran.
| |
Collapse
|
14
|
Kuramoto K, Kim YJ, Hong JH, He C. The autophagy protein Becn1 improves insulin sensitivity by promoting adiponectin secretion via exocyst binding. Cell Rep 2021; 35:109184. [PMID: 34038729 PMCID: PMC8177967 DOI: 10.1016/j.celrep.2021.109184] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/16/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Autophagy dysregulation is implicated in metabolic diseases, including type 2 diabetes. However, the mechanism by which the autophagy machinery regulates metabolism is largely unknown. Autophagy is generally considered a degradation process via lysosomes. Here, we unveil a metabolically important non-cell-autonomous, non-degradative mechanism regulated by the essential autophagy protein Becn1 in adipose tissue. Upon high-fat diet challenge, autophagy-hyperactive Becn1F121A mice show systemically improved insulin sensitivity and enhanced activation of AMP-activated protein kinase (AMPK), a central regulator of energy homeostasis, via a non-cell-autonomous mechanism mediated by adiponectin, an adipose-derived metabolic hormone. Adipose-specific Becn1F121A expression is sufficient to activate AMPK in non-adipose tissues and improve systemic insulin sensitivity by increasing adiponectin secretion. Further, Becn1 enhances adiponectin secretion by interacting with components of the exocyst complex via the coiled-coil domain. Together, our study demonstrates that Becn1 improves insulin sensitivity by facilitating adiponectin secretion through binding the exocyst in adipose tissue.
Collapse
Affiliation(s)
- Kenta Kuramoto
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yoon-Jin Kim
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jung Hwa Hong
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Congcong He
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
15
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
16
|
Renton MC, McGee SL, Howlett KF. The role of protein kinase D (PKD) in intracellular nutrient sensing and regulation of adaptive responses to the obese environment. Obes Rev 2021; 22:e13145. [PMID: 32929844 DOI: 10.1111/obr.13145] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/19/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022]
Abstract
Obesity is associated with ectopic accumulation of lipids, which is implicated in the development of insulin resistance, type 2 diabetes mellitus and cardiovascular disease. As the global prevalence of obesity continues to rise, it is becoming increasingly important to understand the underlying cellular mechanisms of this disease. Protein kinase D (PKD) is an intracellular signalling kinase with well characterized roles in intracellular vesicle transport and secretion, cancer cell proliferation and cardiac hypertrophy. However, emerging evidence also highlights PKD as a novel nutrient sensor. PKD activation is mediated by the accumulation of the lipid intermediate diacylglycerol, and PKD activity in the liver, heart and adipose tissue increases upon feeding. In obesity, PKD signalling is linked to reduced insulin signalling and dysfunction in adipose tissue, liver and heart, whilst in the pancreas, PKD is essential for the compensatory increase in glucose-stimulated insulin secretion from β-cells during obesity. Collectively, these studies reveal aspects of PKD signalling that are involved in the tissue-specific responses to obesity. This review summarizes the emerging evidence suggesting that PKD plays an important role in regulating the adaptive response to the obese environment.
Collapse
Affiliation(s)
- Mark C Renton
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia
| |
Collapse
|
17
|
Hafiane A, Daskalopoulou SS. Adiponectin's mechanisms in high-density lipoprotein biogenesis and cholesterol efflux. Metabolism 2020; 113:154393. [PMID: 33058851 DOI: 10.1016/j.metabol.2020.154393] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/18/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022]
Abstract
AIM Among adiponectin's beneficial properties is its ability to promote cellular cholesterol efflux, thereby generating high-density lipoprotein (HDL) particles. However, adiponectin's role in the regulation of macrophage lipid metabolism, a crucial process in atherogenesis, remains poorly investigated. The aim of this study was to characterize the adiponectin's role in HDL biogenesis. METHODS AND RESULTS We perform kinetics studies in baby hamster kidney (BHK) and Tamm-Horsfall protein 1 (THP-1) cell lines to elucidate adiponectin's role in HDL biogenesis. In cholesterol-enriched cells, specific molar doses of adiponectin stimulated cholesterol efflux with high efficiency to apoA-I. In the presence of adiponectin, BHK cells expressing ATP binding cassette transporter A1 (ABCA1) or ABCG1 generated lipidated particles having α electrophoretic mobility (α-HDL) and a molecular size of 7.5-20 nm. Interestingly, in THP-1 macrophages, cholesterol efflux was associated with more lipidated preβ1-HDL particles. Direct molecular interaction of adiponectin with apoA-I enhanced the affinity of apoA-I to free cholesterol and resulted in an increase in preβ1-HDL particles from plasma ex vivo. Adiponectin increased ABCA1 and ABCG1 protein expression and activated the formation of ABCA1-linked cholesterol oxidase sensitive plasma membrane domains. CONCLUSION Adiponectin upregulated ABCA1 and ABCG1 protein expression, reduced lipid accumulation, and efficiently promoted nascent HDL formation. These results highlight that these cellular processes are interconnected through adiponectin and ABCA1- and ABCG1-dependent. In this pathway, adiponectin increased the affinity of apoA-I to cholesterol and effectively accelerated cholesterol removal from the plasma membrane to HDL particles. Thus, by accelerating HDL biogenesis, adiponectin may have therapeutic potential for atherosclerotic cardiovascular disease prevention and management.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01.3370H, Montréal, Qc H4A 3J1, Canada.
| | - Stella S Daskalopoulou
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01.3370H, Montréal, Qc H4A 3J1, Canada; Department of Medicine, Division of Internal Medicine, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, EM1.2230, Montreal, Quebec H4A 3J1, Canada.
| |
Collapse
|
18
|
Evaluation of Fat Accumulation and Adipokine Production during the Long-Term Adipogenic Differentiation of Porcine Intramuscular Preadipocytes and Study of the Influence of Immunobiotics. Cells 2020; 9:cells9071715. [PMID: 32708964 PMCID: PMC7408200 DOI: 10.3390/cells9071715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022] Open
Abstract
The degree of fat accumulation and adipokine production are two major indicators of obesity that are correlated with increased adipose tissue mass and chronic inflammatory responses. Adipocytes have been considered effector cells for the inflammatory responses due to their capacity to express Toll-like receptors (TLRs). In this study, we evaluated the degree of fat accumulation and adipokine production in porcine intramuscular preadipocyte (PIP) cells maintained for in vitro differentiation over a long period without or with stimulation of either TNF-α or TLR2-, TLR3-, or TLR4-ligands. The cytosolic fat accumulation was measured by liquid chromatography and the expression of adipokines (CCL2, IL-6, IL-8 and IL-10) were quantified by RT-qPCR and ELISA at several time points (0 to 20 days) of PIP cells differentiation. Long-term adipogenic differentiation (LTAD) induced a progressive fat accumulation in the adipocytes over time. Activation of TLR3 and TLR4 resulted in an increased rate of fat accumulation into the adipocytes over the LTAD. The production of CCL2, IL-8 and IL-6 were significantly increased in unstimulated adipocytes during the LTAD, while IL-10 expression remained stable over the studied period. An increasing trend of adiponectin and leptin production was also observed during the LTAD. On the other hand, the stimulation of adipocytes with TLRs agonists or TNF-α resulted in an increasing trend of CCL2, IL-6 and IL-8 production while IL-10 remained stable in all four treatments during the LTAD. We also examined the influences of several immunoregulatory probiotic strains (immunobiotics) on the modulation of the fat accumulation and adipokine production using supernatants of immunobiotic-treated intestinal immune cells and the LTAD of PIP cells. Immunobiotics have shown a strain-specific ability to modulate the fat accumulation and adipokine production, and differentiation of adipocytes. Here, we expanded the utility and potential application of our in vitro PIP cells model by evaluating an LTAD period (20 days) in order to elucidate further insights of chronic inflammatory pathobiology of adipocytes associated with obesity as well as to explore the prospects of immunomodulatory intervention for obesity such as immunobiotics.
Collapse
|
19
|
Adrenergic stimulation of adiponectin secretion in visceral mouse adipocytes is blunted in high-fat diet induced obesity. Sci Rep 2019; 9:10680. [PMID: 31337827 PMCID: PMC6650418 DOI: 10.1038/s41598-019-47113-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/09/2019] [Indexed: 12/31/2022] Open
Abstract
The hormone adiponectin is secreted by white adipocytes and has been put forward as a key mediator of obesity-linked insulin resistance and the metabolic syndrome. Although adiponectin was discovered two decades ago, the knowledge about the molecular and cellular regulation of its secretion is incomplete. Here we have investigated the adrenergic regulation of adiponectin secretion in primary visceral (gonadal) adipocytes isolated from lean or obese/diabetic mice. We show that visceral adipocyte adiponectin release is triggered by cAMP/catecholamines via signalling pathways involving adrenergic beta-3-receptors (β3ARs) and Exchange Protein directly Activated by cAMP, isoform 1 (Epac1). The adrenergically stimulated adiponectin secretion is blunted in visceral adipocytes isolated from obese and diabetic mice and our results suggest the existence of a secretory defect. We have previously shown that adiponectin secretion in subcutaneous adipocytes is abolished in the obese/diabetic state due to reduced abundance of β3ARs and Epac1. However, here we show that protein levels of β3ARs and Epac1 are maintained in visceral adipocytes from obese/diabetic mice proposing that other molecular defects underlie the blunted adiponectin release. Gene expression analysis indicate diabesity-associated disturbances of the signalling downstream of Epac1 and/or the exocytotic process itself. Our study proposes that visceral adipocytes partake in the regulated secretion of adiponectin and may thus influence circulating levels of the hormone, in health and in metabolic disease.
Collapse
|
20
|
Grewal T, Enrich C, Rentero C, Buechler C. Annexins in Adipose Tissue: Novel Players in Obesity. Int J Mol Sci 2019; 20:ijms20143449. [PMID: 31337068 PMCID: PMC6678658 DOI: 10.3390/ijms20143449] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity and the associated comorbidities are a growing health threat worldwide. Adipose tissue dysfunction, impaired adipokine activity, and inflammation are central to metabolic diseases related to obesity. In particular, the excess storage of lipids in adipose tissues disturbs cellular homeostasis. Amongst others, organelle function and cell signaling, often related to the altered composition of specialized membrane microdomains (lipid rafts), are affected. Within this context, the conserved family of annexins are well known to associate with membranes in a calcium (Ca2+)- and phospholipid-dependent manner in order to regulate membrane-related events, such as trafficking in endo- and exocytosis and membrane microdomain organization. These multiple activities of annexins are facilitated through their diverse interactions with a plethora of lipids and proteins, often in different cellular locations and with consequences for the activity of receptors, transporters, metabolic enzymes, and signaling complexes. While increasing evidence points at the function of annexins in lipid homeostasis and cell metabolism in various cells and organs, their role in adipose tissue, obesity and related metabolic diseases is still not well understood. Annexin A1 (AnxA1) is a potent pro-resolving mediator affecting the regulation of body weight and metabolic health. Relevant for glucose metabolism and fatty acid uptake in adipose tissue, several studies suggest AnxA2 to contribute to coordinate glucose transporter type 4 (GLUT4) translocation and to associate with the fatty acid transporter CD36. On the other hand, AnxA6 has been linked to the control of adipocyte lipolysis and adiponectin release. In addition, several other annexins are expressed in fat tissues, yet their roles in adipocytes are less well examined. The current review article summarizes studies on the expression of annexins in adipocytes and in obesity. Research efforts investigating the potential role of annexins in fat tissue relevant to health and metabolic disease are discussed.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Carlos Enrich
- Department of Biomedicine, Unit of Cell Biology, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carles Rentero
- Department of Biomedicine, Unit of Cell Biology, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany.
| |
Collapse
|
21
|
Schaar A, Sun Y, Sukumaran P, Rosenberger TA, Krout D, Roemmich JN, Brinbaumer L, Claycombe-Larson K, Singh BB. Ca 2+ entry via TRPC1 is essential for cellular differentiation and modulates secretion via the SNARE complex. J Cell Sci 2019; 132:jcs.231878. [PMID: 31182642 PMCID: PMC6633397 DOI: 10.1242/jcs.231878] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/27/2019] [Indexed: 12/17/2022] Open
Abstract
Properties of adipocytes, including differentiation and adipokine secretion, are crucial factors in obesity-associated metabolic syndrome. Here, we provide evidence that Ca2+ influx in primary adipocytes, especially upon Ca2+ store depletion, plays an important role in adipocyte differentiation, functionality and subsequently metabolic regulation. The endogenous Ca2+ entry channel in both subcutaneous and visceral adipocytes was found to be dependent on TRPC1–STIM1, and blocking Ca2+ entry with SKF96365 or using TRPC1−/− knockdown adipocytes inhibited adipocyte differentiation. Additionally, TRPC1−/− mice have decreased organ weight, but increased adipose deposition and reduced serum adiponectin and leptin concentrations, without affecting total adipokine expression. Mechanistically, TRPC1-mediated Ca2+ entry regulated SNARE complex formation, and agonist-mediated secretion of adipokine-loaded vesicles was inhibited in TRPC1−/− adipose. These results suggest an unequivocal role of TRPC1 in adipocyte differentiation and adiponectin secretion, and that loss of TRPC1 disturbs metabolic homeostasis. This article has an associated First Person interview with the first author of the paper. Summary: TRPC1 modulates Ca2+ entry, which is essential in adipocyte differentiation and adiponectin secretion, through facilitating SNARE complex formation, thereby maintaining metabolic homeostasis.
Collapse
Affiliation(s)
- Anne Schaar
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Yuyang Sun
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Pramod Sukumaran
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Thad A Rosenberger
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Danielle Krout
- US Department of Agriculture-Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - James N Roemmich
- US Department of Agriculture-Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Lutz Brinbaumer
- Neurobiology Laboratory, NIHES, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA.,Institute of Biomedical Research, (BIOMED) Catholic University of Argentina, Av. Alicia Moreau de Justo 1300, Edificio San Jose Piso 3, Buenos Aires C1107AAZ, Argentina
| | - Kate Claycombe-Larson
- US Department of Agriculture-Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Brij B Singh
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
22
|
Otelea MR, Streinu-Cercel A, Băicus C, Nitescu M. The Adipokine Profile and the Cardiometabolic Risk in Non-Obese Young Adults. Balkan Med J 2019; 36:155-161. [PMID: 30457109 PMCID: PMC6528526 DOI: 10.4274/balkanmedj.galenos.2018.2018.0789] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022] Open
Abstract
Background Young, non-obese adults are considered at low risk for cardiometabolic diseases, although markers of an unhealthy metabolic state are not uncommon findings in this population. Adipose tissue dysfunction, evaluated by the adipokine profile, significantly influences lipid and glucose metabolism and low-grade systemic inflammation. Aims To determine the relation between adipose tissue dysfunction and the already confirmed cardiometabolic risk indicators, including the atherogenic index of plasma, lipid accumulation product, homeostatic model assessment of insulin resistance, and the low-grade inflammation markers, namely, interleukin 6 and high-sensitivity C-reactive protein. Study Design Cross-sectional study. Methods We recruited 93 non-obese, healthy young adults. Anthropometric, lipid profile, inflammatory markers, and adipokines were measured. An abnormal adipokine profile (high leptin-to-adiponectin ratio) was considered as a marker of a dysfunctional adipose tissue. The correlation between the leptin-to-adiponectin ratio and the anthropometric measurements, atherogenic index of plasma, lipid accumulation product, homeostatic model assessment of insulin resistance, interleukin 6, and high-sensitivity C-reactive protein was determined. Results We found a direct correlation between the abnormal adipokine profile and the cardiometabolic risk indicators mentioned above, except for the low-grade inflammatory markers. In the regression model derived from our data, the leptin-to-adiponectin ratio was best correlated with the unfavorable plasma lipid profile, as estimated by the atherogenic index of plasma (r=0.097, confidence interval=0.015-0.180, p=0.021). A significantly higher leptin-to-adiponectin ratio was found in the insulin-resistant group (p=0.012) and in the highest lipid accumulation product quartile (p=0.032). Conclusion In a non-obese young population, the high rate of leptin-adiponectin may be a good predictor of cardiovascular and metabolic risk assessment.
Collapse
Affiliation(s)
| | - Adrian Streinu-Cercel
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Institute for Infectious Diseases ‘Matei Bals’, Bucharest, Romania
| | - Cristian Băicus
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Clinical Hospital Colentina, Bucharest, Romania
| | - Maria Nitescu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Institute for Infectious Diseases ‘Matei Bals’, Bucharest, Romania
| |
Collapse
|
23
|
Leptin-induced cardiomyocyte hypertrophy is associated with enhanced mitochondrial fission. Mol Cell Biochem 2018; 454:33-44. [PMID: 30251118 DOI: 10.1007/s11010-018-3450-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022]
Abstract
Cardiac pathology including hypertrophy has been associated with an imbalance between mitochondrial fission and fusion. Generally, well-balanced mitochondrial fission and fusion are essential for proper functions of mitochondria. Leptin is a 16-kDa appetite-suppressing protein which has been shown to induce cardiomyocyte hypertrophy. In the present study, we determined whether leptin can influence mitochondrial fission or fusion and whether this can be related to its hypertrophic effect. Cardiomyocytes treated for 24 h with 3.1 nM leptin (50 ng/ml), a concentration representing plasma levels in obese individuals, demonstrated an increase in surface area and a significant 1.6-fold increase in the expression of the β-myosin heavy chain. Mitochondrial staining with MitoTracker Green dye showed elongated structures in control cells with an average length of 4.5 µm. Leptin produced a time-dependent increase in mitochondrial fragmentation with decreasing mitochondrial length. The hypertrophic response to leptin was also associated with increased protein levels of the mitochondrial fission protein dynamin-related protein1 (Drp1) although gene expression of Drp1 was unaffected possibly suggesting post-translational modifications of Drp1. Indeed, leptin treatment was associated with decreased levels of phosphorylated Drp1 and increased translocation of Drp1 to the mitochondria thereby demonstrating a pro-fission effect of leptin. As calcineurin may dephosphorylate Drp1, we determined the effect of a calcineurin inhibitor, FK506, which prevented leptin-induced hypertrophy as well as mitochondrial fission and mitochondrial dysfunction. In conclusion, our data show that leptin-induced cardiomyocyte hypertrophy is associated with enhanced mitochondrial fission via a calcineurin-mediated pathway. The ability of leptin to stimulate mitochondrial fission may be important in understanding the role of this protein in cardiac pathology especially that related to mitochondrial dysfunction.
Collapse
|
24
|
Marques-Oliveira GH, Silva TM, Lima WG, Valadares HMS, Chaves VE. Insulin as a hormone regulator of the synthesis and release of leptin by white adipose tissue. Peptides 2018; 106:49-58. [PMID: 29953915 DOI: 10.1016/j.peptides.2018.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 02/09/2023]
Abstract
Leptin and its receptor are widely distributed in several tissues, mainly in white adipose tissue. The serum leptin is highly correlated with body mass index in rodents and humans, being documented that leptin levels reduces in the fasting state and increase during refeeding, similarly to insulin release by pancreatic islets. Insulin appears to increase leptin mRNA and protein expression and its release by adipocytes. Some studies have suggested that insulin acts through the activation of the transcription factors: sterol regulatory element binding protein 1 (SREBP1), CCAAT enhancer binding protein-α (C/EBP-α) and specificity protein 1 (Sp1). Insulin stimulates the release of preformed and newly synthesized leptin by adipocytes through its signaling cascade. Its effects are blocked by inhibitors of the insulin signaling pathway, as well as by inhibitors of protein synthesis and agents that increase the intracellular cAMP. The literature data suggest that chronic hyperinsulinemia increases serum leptin levels in humans and rodents. In this review, we summarized the most updated knowledge on the effects of insulin on serum leptin levels, presenting the cell mechanisms that control leptin synthesis and release by the white adipose tissue.
Collapse
Affiliation(s)
| | - Thaís Marques Silva
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | - William Gustavo Lima
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | | | - Valéria Ernestânia Chaves
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil.
| |
Collapse
|
25
|
Rödiger M, Werno MW, Wilhelmi I, Baumeier C, Hesse D, Wettschureck N, Offermanns S, Song K, Krauß M, Schürmann A. Adiponectin release and insulin receptor targeting share trans-Golgi-dependent endosomal trafficking routes. Mol Metab 2018; 8:167-179. [PMID: 29203237 PMCID: PMC5985030 DOI: 10.1016/j.molmet.2017.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Intracellular vesicle trafficking maintains cellular structures and functions. The assembly of cargo-laden vesicles at the trans-Golgi network is initiated by the ARF family of small GTPases. Here, we demonstrate the role of the trans-Golgi localized monomeric GTPase ARFRP1 in endosomal-mediated vesicle trafficking of mature adipocytes. METHODS Control (Arfrp1flox/flox) and inducible fat-specific Arfrp1 knockout (Arfrp1iAT-/-) mice were metabolically characterized. In vitro experiments on mature 3T3-L1 cells and primary mouse adipocytes were conducted to validate the impact of ARFRP1 on localization of adiponectin and the insulin receptor. Finally, secretion and transferrin-based uptake and recycling assays were performed with HeLa and HeLa M-C1 cells. RESULTS We identified the ARFRP1-based sorting machinery to be involved in vesicle trafficking relying on the endosomal compartment for cell surface delivery. Secretion of adiponectin from fat depots was selectively reduced in Arfrp1iAT-/- mice, and Arfrp1-depleted 3T3-L1 adipocytes revealed an accumulation of adiponectin in Rab11-positive endosomes. Plasma adiponectin deficiency of Arfrp1iAT-/- mice resulted in deteriorated hepatic insulin sensitivity, increased gluconeogenesis and elevated fasting blood glucose levels. Additionally, the insulin receptor, undergoing endocytic recycling after ligand binding, was less abundant at the plasma membrane of adipocytes lacking Arfrp1. This had detrimental effects on adipose insulin signaling, followed by insufficient suppression of basal lipolytic activity and impaired adipose tissue expansion. CONCLUSIONS Our findings suggest that adiponectin secretion and insulin receptor surface targeting utilize the same post-Golgi trafficking pathways that are essential for an appropriate systemic insulin sensitivity and glucose homeostasis.
Collapse
Affiliation(s)
- Maria Rödiger
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research, München-Neuherberg, 85764 Neuherberg, Germany.
| | - Martin W Werno
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research, München-Neuherberg, 85764 Neuherberg, Germany.
| | - Ilka Wilhelmi
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research, München-Neuherberg, 85764 Neuherberg, Germany.
| | - Christian Baumeier
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research, München-Neuherberg, 85764 Neuherberg, Germany.
| | - Deike Hesse
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research, München-Neuherberg, 85764 Neuherberg, Germany.
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.
| | - Kyungyeun Song
- Department of Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany.
| | - Michael Krauß
- Department of Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany.
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research, München-Neuherberg, 85764 Neuherberg, Germany.
| |
Collapse
|
26
|
Kuten O, Simon M, Hornyák I, De Luna-Preitschopf A, Nehrer S, Lacza Z. The Effects of Hyperacute Serum on Adipogenesis and Cell Proliferation of Mesenchymal Stromal Cells. Tissue Eng Part A 2018; 24:1011-1021. [PMID: 29265000 DOI: 10.1089/ten.tea.2017.0384] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fat tissue, due to its high concentration of stem cells, has a role in aesthetic medicine and reconstructive surgery. However, poor survival of the transplanted cells still limits the usefulness of this material in regenerative medicine. Several studies indicated that platelet-rich plasma (PRP) may improve adipose tissue viability due to its growth factor content. This study aimed at investigating the effects of PRP and hyperacute serum (HAS) on the adipogenic lineage in vitro. PRP was prepared by using two centrifugation steps in the presence of anticoagulants, and HAS was isolated from activated platelet-rich fibrin within 10 min of blood drawing to prevent the propagation of inflammatory cascades. Metabolic activity and proliferation rate of human bone marrow-derived mesenchymal stem cells (hMSCs) cultivated in media supplemented with three types of serum additives (fetal calf serum [FCS], human PRP, or HAS) was determined by using a tetrazolium assay. Adipogenesis was evaluated in standard and pro-adipogenic media and tested by oil red staining, triglyceride content, and expression of specific genes. Adipogenic regulators in the sera were measured by multiplex ELISA assays. We observed that proliferation of hMSCs was supported by both FCS and HAS in a time-dependent manner, but surprisingly, PRP had a much weaker effect (change in proliferation rate after 5 days relative to metabolic activity on day 0-FCS: 5.4-fold change, HAS: 5.8-fold change, serum free 1.9-fold change, PRP: 3.0-fold change, p < 0.05). Lipogenesis was only observed in groups with adipogenic differentiation medium, with HAS showing a significantly stronger effect than PRP. This was confirmed by intensive accumulation of lysochrome dye in lipid droplets, higher triglyceride concentration, and elevated expression of specific adipogenic genes. Measurement of lipogenic proteins in the sera revealed that both PRP and HAS are abundant in them; however, PRP also contains anti-adipogenic factors, which explains its weaker and less reliable effect. The results of this study suggest that HAS provides more robust support than PRP in hMSCs proliferation as well as lipogenic differentiation, indicating that it may be a better adjuvant in fat grafting procedures.
Collapse
Affiliation(s)
- Olga Kuten
- 1 OrthoSera GmbH , Krems an der Donau, Austria
| | - Melinda Simon
- 2 Institute of Clinical Experimental Research, Semmelweis University , Budapest, Hungary
| | - István Hornyák
- 2 Institute of Clinical Experimental Research, Semmelweis University , Budapest, Hungary
| | | | - Stefan Nehrer
- 3 Centre for Regenerative Medicine and Orthopedics, Danube University Krems , Krems, Austria
| | | |
Collapse
|
27
|
Ameen GI, Mora S. Cbl downregulation increases RBP4 expression in adipocytes of female mice. J Endocrinol 2018; 236:29-41. [PMID: 29114012 PMCID: PMC5744582 DOI: 10.1530/joe-17-0359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 11/07/2017] [Indexed: 11/30/2022]
Abstract
Obesity leads to adipose tissue dysfunction, insulin resistance and diabetes. Adipose tissue produces adipokines that contribute to regulate insulin sensitivity. In turn, insulin stimulates the production and release of some adipokines. Casitas-b-lymphoma proteins (c-Cbl, Cbl-b and Cbl3) are intracellular adaptor signalling proteins that are rapidly phosphorylated by activation of tyrosine kinase receptors. c-Cbl is rapidly phosphorylated by insulin in adipocytes. Here, we tested the hypothesis that Cbl signalling regulates adipokine expression in adipose tissue. We determined the adipokine profile of WAT of Cbl-/- and Cbl+/+ mice in the C57BL6 background. Female Cbl-/- mice exhibited altered expression of adiponectin, leptin and RBP4 in visceral adipose tissue, while no significant changes were seen in male mice. TNFα and IL6 levels were unaffected by Cbl depletion. RBP4 expression was unchanged in liver. Adipose tissue of Cbl-/- animals showed increased basal activation of extracellular regulated kinases (ERK1/2) compared to Cbl+/+. c-Cbl knockdown in 3T3L1 adipocytes also increased basal ERK phosphorylation and RBP4 expression. Inhibition of ERK1/2 phosphorylation in Cbl-depleted 3T3L1 adipocytes or in adipose tissue explants of Cbl-/- mice reduced RBP4 mRNA. 17β-Estradiol increased RBP4 mRNA in adipocytes. Cbl depletion did not change ER expression but increased phosphorylation of ERα at S118, a target site for ERK1/2. ERK1/2 inhibition reduced phosphoER and RBP4 levels. These findings suggest that Cbl contributes to regulate RBP4 expression in adipose of female mice through ERK1/2-mediated activation of ERα. Since Cbl signalling is compromised in diabetes, these data highlight a novel mechanism that upregulates RBP4 locally.
Collapse
Affiliation(s)
- Gulizar Issa Ameen
- Department of Cellular and Molecular PhysiologyInstitute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Silvia Mora
- Department of Cellular and Molecular PhysiologyInstitute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
28
|
Rocha N, Payne F, Huang-Doran I, Sleigh A, Fawcett K, Adams C, Stears A, Saudek V, O’Rahilly S, Barroso I, Semple RK. The metabolic syndrome- associated small G protein ARL15 plays a role in adipocyte differentiation and adiponectin secretion. Sci Rep 2017; 7:17593. [PMID: 29242557 PMCID: PMC5730586 DOI: 10.1038/s41598-017-17746-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/30/2017] [Indexed: 02/02/2023] Open
Abstract
Common genetic variants at the ARL15 locus are associated with plasma adiponectin, insulin and HDL cholesterol concentrations, obesity, and coronary atherosclerosis. The ARL15 gene encodes a small GTP-binding protein whose function is currently unknown. In this study adipocyte-autonomous roles for ARL15 were investigated using conditional knockdown of Arl15 in murine 3T3-L1 (pre)adipocytes. Arl15 knockdown in differentiated adipocytes impaired adiponectin secretion but not adipsin secretion or insulin action, while in preadipocytes it impaired adipogenesis. In differentiated adipocytes GFP-tagged ARL15 localized predominantly to the Golgi with lower levels detected at the plasma membrane and intracellular vesicles, suggesting involvement in intracellular trafficking. Sequencing of ARL15 in 375 severely insulin resistant patients identified four rare heterozygous variants, including an early nonsense mutation in a proband with femorogluteal lipodystrophy and non classical congenital adrenal hyperplasia, and an essential splice site mutation in a proband with partial lipodystrophy and a history of childhood yolk sac tumour. No nonsense or essential splice site mutations were found in 2,479 controls, while five such variants were found in the ExAC database. These findings provide evidence that ARL15 plays a role in adipocyte differentiation and adiponectin secretion, and raise the possibility that human ARL15 haploinsufficiency predisposes to lipodystrophy.
Collapse
Affiliation(s)
- Nuno Rocha
- 0000 0004 0369 9638grid.470900.aThe University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK ,grid.454369.9The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Felicity Payne
- 0000 0004 0606 5382grid.10306.34Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Isabel Huang-Doran
- 0000 0004 0369 9638grid.470900.aThe University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK ,grid.454369.9The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Alison Sleigh
- 0000000121885934grid.5335.0Wolfson Brain Imaging Centre, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK ,0000 0004 0383 8386grid.24029.3dNational Institute for Health Research/Wellcome Trust Clinical Research Facility, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Katherine Fawcett
- 0000 0004 0606 5382grid.10306.34Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Claire Adams
- 0000 0004 0369 9638grid.470900.aThe University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK ,grid.454369.9The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Anna Stears
- 0000 0004 0383 8386grid.24029.3dWolfson Diabetes and Endocrine Clinic, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Vladimir Saudek
- 0000 0004 0369 9638grid.470900.aThe University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK ,grid.454369.9The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Stephen O’Rahilly
- 0000 0004 0369 9638grid.470900.aThe University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK ,grid.454369.9The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Inês Barroso
- 0000 0004 0369 9638grid.470900.aThe University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK ,0000 0004 0606 5382grid.10306.34Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Robert K. Semple
- 0000 0004 0369 9638grid.470900.aThe University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK ,grid.454369.9The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK ,0000 0004 1936 7988grid.4305.2Centre for Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
| |
Collapse
|
29
|
Kim S, Lee AY, Kim HJ, Hong SH, Go RE, Choi KC, Kang KS, Cho MH. Exposure to cigarette smoke disturbs adipokines secretion causing intercellular damage and insulin resistance in high fructose diet-induced metabolic disorder mice. Biochem Biophys Res Commun 2017; 494:648-655. [PMID: 29079192 DOI: 10.1016/j.bbrc.2017.10.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
Abstract
A large amount of fructose intake along with smoking is associated with increased incidence of diseases linked to metabolic syndrome. More research is necessary to understand the complex mechanism that ultimately results in metabolic syndrome and the effect, if any, of high fructose dietary intake and smoking on individual health. In this study, we investigated changes in ER-Golgi network and disturbance to secretion of adipokines induced by cigarette smoking (CS) and excess fructose intake and their contribution to the disruption of metabolic homeostasis. We used high fructose-induced metabolic disorder mice model by feeding them with high fructose diet for 8 weeks. For CS exposure experiment, these mice were exposed to CS for 28 days according to OECD guideline 412. Our results clearly showed that the immune system was suppressed and ER stress was induced in mice with exposure to CS and fed with high fructose. Furthermore, their concentrations of adipokines including leptin and adiponectin were aberrant. Such alteration in secretion of adipokines could cause insulin resistance which may lead to the development of type 2 diabetes.
Collapse
Affiliation(s)
- Sanghwa Kim
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Science, Seoul 01812, Republic of Korea; Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Ah Young Lee
- Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeon-Jeong Kim
- Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Seong-Ho Hong
- Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Bio medicine Lab., CKD Research Institute, Yongin 16995, Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Myung-Haing Cho
- Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Graduate Group of Tumor Biology, Seoul National University, Seoul 08826, 151-742, Republic of Korea; Institute of GreenBio Science Technology, Seoul National University, Pyeongchang 25354, Republic of Korea; Graduate School of Convergence Science and Technology, Seoul National University, Suwon 16229, Republic of Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.
| |
Collapse
|
30
|
Brännmark C, Lövfors W, Komai AM, Axelsson T, El Hachmane MF, Musovic S, Paul A, Nyman E, Olofsson CS. Mathematical modeling of white adipocyte exocytosis predicts adiponectin secretion and quantifies the rates of vesicle exo- and endocytosis. J Biol Chem 2017; 292:20032-20043. [PMID: 28972187 DOI: 10.1074/jbc.m117.801225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/22/2017] [Indexed: 11/06/2022] Open
Abstract
Adiponectin is a hormone secreted from white adipocytes and takes part in the regulation of several metabolic processes. Although the pathophysiological importance of adiponectin has been thoroughly investigated, the mechanisms controlling its release are only partly understood. We have recently shown that adiponectin is secreted via regulated exocytosis of adiponectin-containing vesicles, that adiponectin exocytosis is stimulated by cAMP-dependent mechanisms, and that Ca2+ and ATP augment the cAMP-triggered secretion. However, much remains to be discovered regarding the molecular and cellular regulation of adiponectin release. Here, we have used mathematical modeling to extract detailed information contained within our previously obtained high-resolution patch-clamp time-resolved capacitance recordings to produce the first model of adiponectin exocytosis/secretion that combines all mechanistic knowledge deduced from electrophysiological experimental series. This model demonstrates that our previous understanding of the role of intracellular ATP in the control of adiponectin exocytosis needs to be revised to include an additional ATP-dependent step. Validation of the model by introduction of data of secreted adiponectin yielded a very close resemblance between the simulations and experimental results. Moreover, we could show that Ca2+-dependent adiponectin endocytosis contributes to the measured capacitance signal, and we were able to predict the contribution of endocytosis to the measured exocytotic rate under different experimental conditions. In conclusion, using mathematical modeling of published and newly generated data, we have obtained estimates of adiponectin exo- and endocytosis rates, and we have predicted adiponectin secretion. We believe that our model should have multiple applications in the study of metabolic processes and hormonal control thereof.
Collapse
Affiliation(s)
- Cecilia Brännmark
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg
| | - William Lövfors
- Departments of Biomedical Engineering, SE-581 83 Linköping; Mathematics, Linköping University, SE-581 83 Linköping
| | - Ali M Komai
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg
| | - Tom Axelsson
- Departments of Biomedical Engineering, SE-581 83 Linköping
| | - Mickaël F El Hachmane
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg
| | - Saliha Musovic
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg
| | - Alexandra Paul
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10 SE-412 96 Göteborg
| | - Elin Nyman
- Departments of Biomedical Engineering, SE-581 83 Linköping; Cardiovascular and Metabolic Diseases iMed Biotech Unit, AstraZeneca R&D, 431 83 Gothenburg, Sweden.
| | - Charlotta S Olofsson
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg.
| |
Collapse
|
31
|
Bedi D, Dennis JC, Morrison EE, Braden TD, Judd RL. Regulation of intracellular trafficking and secretion of adiponectin by myosin II. Biochem Biophys Res Commun 2017; 490:202-208. [PMID: 28606474 DOI: 10.1016/j.bbrc.2017.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/07/2017] [Indexed: 01/08/2023]
Abstract
Adiponectin is a protein secreted by white adipocytes that plays an important role in insulin action, energy homeostasis and the development of atherosclerosis. The intracellular localization and trafficking of GLUT4 and leptin in adipocytes has been well studied, but little is known regarding the intracellular trafficking of adiponectin. Recent studies have demonstrated that constitutive adiponectin secretion is dependent on PIP2 levels and the integrity of cortical F-actin. Non-muscle myosin II is an actin-based motor that is associated with membrane vesicles and participates in vesicular trafficking in mammalian cells. Therefore, we investigated the role of myosin II in the trafficking and secretion of adiponectin in 3T3-L1 adipocytes. Confocal microscopy revealed that myosin IIA and IIB were dispersed throughout the cytoplasm of the adipocyte. Both myosin isoforms were localized in the Golgi/TGN region as evidenced by colocalization with the cis-Golgi marker, p115 and the trans-Golgi marker, γ-adaptin. Inhibition of myosin II activity by blebbistatin or actin depolymerization by latrunculin B dispersed myosin IIA and IIB towards the periphery while significantly inhibiting adiponectin secretion. Therefore, the constitutive trafficking and secretion of adiponectin in 3T3-L1 adipocytes occurs by an actin-dependent mechanism that involves the actin-based motors, myosin IIA and IIB.
Collapse
Affiliation(s)
- Deepa Bedi
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States.
| | - John C Dennis
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Edward E Morrison
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Tim D Braden
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Robert L Judd
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
32
|
Kobayashi S, Kawasaki Y, Takahashi T, Maeno H, Nomura M. Mechanisms for the anti-obesity actions of bofutsushosan in high-fat diet-fed obese mice. Chin Med 2017; 12:8. [PMID: 28360931 PMCID: PMC5369197 DOI: 10.1186/s13020-017-0129-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 03/04/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The Kampo medicine bofutsushosan (BTS; Pulvis ledebouriellae compositae; Fang Feng Tong Sheng San) has been used as an anti-obesity treatment in overweight patients. In this study, we assessed the underlying physiological changes induced by BTS in obese mice maintained on a high-fat diet. METHODS Male ICR mice were fed a 60% kcal fat diet for 5 weeks starting at 4 weeks of age and then fed the same diet with administration of water (control) or aqueous BTS extract (1.0-2.0 g/kg) for 25 days. Body weight, wet weight of isolated white adipose tissue, and obesity-related serum parameters (glucose, lipids, leptin, adiponectin) were measured after treatment. The mRNA expression levels of leptin, adiponectin, and UCP1 in the adipose tissues were determined by quantitative real-time polymerase chain reaction after the first 5 days of treatment. RESULTS Bofutsushosan (1.5-2.0 g/kg) significantly decreased total body weight and total wet weight of white adipose tissue isolated from subcutaneous (retroperitoneal) and visceral regions (epididymal, mesenteric, and perirenal). At 2.0 g/kg, BTS also decreased total fat mass, visceral fat mass, and ratio of fat mass to body weight as measured by computed tomography, and significantly decreased epididymal adipocyte size after 14 and 25 days' treatment. Twenty-five days' treatment lowered serum glucose, insulin, leptin, and triglycerides, and reduced homeostasis model assessment-insulin resistance. Alternatively, 2.0 g/kg BTS significantly increased mRNA levels of adiponectin, leptin, and UCP1 in interscapular brown adipose tissue but not epididymal white adipose tissue after 5 days' administration. CONCLUSION In the early administration period, BTS increased mRNA expression levels of leptin, adiponectin, and UCP1 in brown adipose tissues. With longer administration, BTS improved insulin resistance, and subsequently reduced serum levels of leptin and triglyceride in parallel with decreased visceral white adipose tissue volume and adipocyte size.
Collapse
Affiliation(s)
- Shinjiro Kobayashi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Yuki Kawasaki
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Tatsuo Takahashi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Hironori Maeno
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Masaaki Nomura
- Center of Clinical Pharmacy Education, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| |
Collapse
|
33
|
Cui J, Pang J, Lin YJ, Jiang P, Gong H, Wang Z, Li J, Cai JP, Huang JD, Zhang TM. Conventional kinesin KIF5B mediates adiponectin secretion in 3T3-L1 adipocytes. Biochem Biophys Res Commun 2016; 476:620-626. [DOI: 10.1016/j.bbrc.2016.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 06/02/2016] [Indexed: 01/15/2023]
|
34
|
Shen KP, Hao CL, Yen HW, Chen CY, Chen JH, Chen FC, Lin HL. Pre-germinated brown rice prevented high fat diet induced hyperlipidemia through ameliorating lipid synthesis and metabolism in C57BL/6J mice. J Clin Biochem Nutr 2016; 59:39-44. [PMID: 27499577 PMCID: PMC4933684 DOI: 10.3164/jcbn.15-117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/24/2016] [Indexed: 12/22/2022] Open
Abstract
Pre-germinated brown rice (PGBR) can ameliorate hyperlipidemia, but the action mechanism is not clear. We focus the mechanisms of PGBR prevented hyperlipidemia. Six-week-old mice were divided into: standard-regular diet (SRD), high-fat diet (HFD) and HFD with PGBR (HFD + PGBR) groups for 16 weeks. The HFD group has higher concentrations of TG, TC, HDL and Non-HDL in the blood, and a higher atherosclerosis index (AI). The TG levels in the liver, and TG, bile acid levels in the feces were enhanced; and the total adipocytokines level in adipose tissue was reduced. The HFD group had higher protein expressions of SREBP-1, SCD-1, FAS, LDLR, and CYP7α1 in the liver. Moreover, the greater expressions of SREBP-1, SCD-1, FAS and the less expressions of PPAR-α and adiponectin were in adipose tissue. In the HFD + PGBR group, the PGBR regulated the levels of TG, TC, HDL, Non-HDL, AI and adipocytokines. PGBR increased more cholesterol and bile acid exhaust in feces. The SREBP-1, SCD-1, FAS, HMGCR, LDLR, CYP7α1 and PPAR-α proteins in the liver; and the SREBP-1, SCD-1, FAS, PPAR-α and adiponectin proteins in adipose tissue were reversed by PGBR. Taken together, PGBR can improve lipid synthesis and metabolism, and we suggest PGBR is a recommendable food for controlling hyperlipidemia.
Collapse
Affiliation(s)
- Kuo-Ping Shen
- Department of Nursing, Meiho University, Pingtung 91202, Taiwan
| | - Chi-Long Hao
- Division of Cardiology, Department of Internal Medicine, Pingtung Christian Hospital, Pingtung 90053, Taiwan
| | - Hsueh-Wei Yen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chun-Yen Chen
- MS program for Applied Health and Biotechnology, Meiho University, Pingtung 91202, Taiwan
| | - Jia-Hao Chen
- MS program for Applied Health and Biotechnology, Meiho University, Pingtung 91202, Taiwan
| | - Fu-Chih Chen
- Department of Chemistry, National Cheng-Kung University, Tainan 70101, Taiwan
| | - Hui-Li Lin
- Department of Food Science and Nutrition, Meiho University, Pingtung 91202, Taiwan
| |
Collapse
|
35
|
Llobet L, Toivonen JM, Montoya J, Ruiz-Pesini E, López-Gallardo E. Xenobiotics that affect oxidative phosphorylation alter differentiation of human adipose-derived stem cells at concentrations that are found in human blood. Dis Model Mech 2015; 8:1441-55. [PMID: 26398948 PMCID: PMC4631789 DOI: 10.1242/dmm.021774] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/03/2015] [Indexed: 12/17/2022] Open
Abstract
Adipogenesis is accompanied by differentiation of adipose tissue-derived stem cells to adipocytes. As part of this differentiation, biogenesis of the oxidative phosphorylation system occurs. Many chemical compounds used in medicine, agriculture or other human activities affect oxidative phosphorylation function. Therefore, these xenobiotics could alter adipogenesis. We have analyzed the effects on adipocyte differentiation of some xenobiotics that act on the oxidative phosphorylation system. The tested concentrations have been previously reported in human blood. Our results show that pharmaceutical drugs that decrease mitochondrial DNA replication, such as nucleoside reverse transcriptase inhibitors, or inhibitors of mitochondrial protein synthesis, such as ribosomal antibiotics, diminish adipocyte differentiation and leptin secretion. By contrast, the environmental chemical pollutant tributyltin chloride, which inhibits the ATP synthase of the oxidative phosphorylation system, can promote adipocyte differentiation and leptin secretion, leading to obesity and metabolic syndrome as postulated by the obesogen hypothesis. Summary: Some medical drugs and environmental chemical pollutants acting on the oxidative phosphorylation system can alter adipocyte differentiation and adipogenesis and, thus, have important consequences for human health.
Collapse
Affiliation(s)
- Laura Llobet
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Janne M Toivonen
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain Fundación ARAID, Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Ester López-Gallardo
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| |
Collapse
|
36
|
DeClercq V, d'Eon B, McLeod RS. Fatty acids increase adiponectin secretion through both classical and exosome pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1123-33. [DOI: 10.1016/j.bbalip.2015.04.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/06/2015] [Accepted: 04/13/2015] [Indexed: 11/26/2022]
|
37
|
Komai AM, Brännmark C, Musovic S, Olofsson CS. PKA-independent cAMP stimulation of white adipocyte exocytosis and adipokine secretion: modulations by Ca2+ and ATP. J Physiol 2014; 592:5169-86. [PMID: 25194045 DOI: 10.1113/jphysiol.2014.280388] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We examined the effects of cAMP, Ca(2+) and ATP on exocytosis and adipokine release in white adipocytes by a combination of membrane capacitance patch-clamp recordings and biochemical measurements of adipokine secretion. 3T3-L1 adipocyte exocytosis proceeded even in the complete absence of intracellular Ca(2+) ([Ca(2+)]i; buffered with BAPTA) provided cAMP (0.1 mm) was included in the intracellular (pipette-filling) solution. Exocytosis typically plateaued within ∼10 min, probably signifying depletion of a releasable vesicle pool. Inclusion of 3 mm ATP in combination with elevation of [Ca(2+)]i to ≥700 nm augmented the rate of cAMP-evoked exocytosis ∼2-fold and exocytosis proceeded for longer periods (≥20 min) than with cAMP alone. Exocytosis was stimulated to a similar extent upon substitution of cAMP by the Epac (exchange proteins activated by cAMP) agonist 8-Br-2'-O-Me-cAMP (1 mm included in the pipette solution). Inhibition of protein kinase A (PKA) by addition of Rp-cAMPS (0.5 mm) to the cAMP-containing pipette solution was without effect. A combination of the adenylate cyclase activator forskolin (10 μm) and the phosphodiesterase inhibitor IBMX (200 μm; forsk-IBMX) augmented adiponectin secretion measured over 30 min 3-fold and 2-fold in 3T3-L1 and human subcutaneous adipocytes, respectively. This effect was unaltered by pre-loading of cells with the Ca(2+) chelator BAPTA-AM and 2-fold amplified upon inclusion of the Ca(2+) ionophore ionomycin (1 μm) in the extracellular solution. Adiponectin release was also stimulated by the membrane-permeable Epac agonist 8-Br-2'-O-Me-cAMP-AM but unaffected by inclusion of the membrane-permeable PKA inhibitor Rp-8-Br-cAMPS (200 μm). The adipokines leptin, resistin and apelin were present in low amounts in the incubation medium (1-6% of measured adiponectin). Adipsin was secreted in substantial quantities (50% of adiponectin concentration) but release of this adipokine was unaffected by forsk-IBMX. We propose that white adipocyte exocytosis is stimulated by cAMP/Epac-dependent but Ca(2+)- and PKA-independent release of vesicles residing in a readily releasable pool and that the release is augmented by a combination of Ca(2+) and ATP. We further suggest that secreted vesicles chiefly contain adiponectin.
Collapse
Affiliation(s)
- Ali M Komai
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30, Göteborg, Sweden
| | - Cecilia Brännmark
- Discovery Sciences, AstraZeneca R&D, Pepparedsleden 1, SE43153, Mölndal, Sweden
| | - Saliha Musovic
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30, Göteborg, Sweden
| | - Charlotta S Olofsson
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30, Göteborg, Sweden
| |
Collapse
|
38
|
Romacho T, Elsen M, Röhrborn D, Eckel J. Adipose tissue and its role in organ crosstalk. Acta Physiol (Oxf) 2014; 210:733-53. [PMID: 24495317 DOI: 10.1111/apha.12246] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/18/2013] [Accepted: 01/28/2014] [Indexed: 12/13/2022]
Abstract
The discovery of adipokines has revealed adipose tissue as a central node in the interorgan crosstalk network, which mediates the regulation of multiple organs and tissues. Adipose tissue is a true endocrine organ that produces and secretes a wide range of mediators regulating adipose tissue function in an auto-/paracrine manner and important distant targets, such as the liver, skeletal muscle, the pancreas and the cardiovascular system. In metabolic disorders such as obesity, enlargement of adipocytes leads to adipose tissue dysfunction and a shift in the secretory profile with an increased release of pro-inflammatory adipokines. Adipose tissue dysfunction has a central role in the development of insulin resistance, type 2 diabetes, and cardiovascular diseases. Besides the well-acknowledged role of adipokines in metabolic diseases, and the increasing number of adipokines being discovered in the last years, the mechanisms underlying the release of many adipokines from adipose tissue remain largely unknown. To combat metabolic diseases, it is crucial to better understand how adipokines can modulate adipose tissue growth and function. Therefore, we will focus on adipokines with a prominent role in auto-/paracrine crosstalk within the adipose tissue such as RBP4, HO-1, WISP2, SFRPs and chemerin. To depict the endocrine crosstalk between adipose tissue with skeletal muscle, the cardiovascular system and the pancreas, we will report the main findings regarding the direct effects of adiponectin, leptin, DPP4 and visfatin on skeletal muscle insulin resistance, cardiovascular function and β-cell growth and function.
Collapse
Affiliation(s)
- T. Romacho
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - M. Elsen
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - D. Röhrborn
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - J. Eckel
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
- German Center for Diabetes Research (DZD e.V.); Düsseldorf Germany
| |
Collapse
|
39
|
Xie L, Fu Q, Ortega TM, Zhou L, Rasmussen D, O’Keefe J, Zhang KK, Chapes SK. Overexpression of IL-10 in C2D macrophages promotes a macrophage phenotypic switch in adipose tissue environments. PLoS One 2014; 9:e86541. [PMID: 24466141 PMCID: PMC3897709 DOI: 10.1371/journal.pone.0086541] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 12/14/2013] [Indexed: 11/19/2022] Open
Abstract
Adipose tissue macrophages are a heterogeneous collection of classically activated (M1) and alternatively activated (M2) macrophages. Interleukin 10 (IL-10) is an anti-inflammatory cytokine, secreted by a variety of cell types including M2 macrophages. We generated a macrophage cell line stably overexpressing IL-10 (C2D-IL10) and analyzed the C2D-IL10 cells for several macrophage markers after exposure to adipocytes compared to C2D cells transfected with an empty vector (C2D-vector). C2D-IL10 macrophage cells expressed more CD206 when co-cultured with adipocytes than C2D-vector cells; while the co-cultured cell mixture also expressed higher levels of Il4, Il10, Il1β and Tnf. Since regular C2D cells traffic to adipose tissue after adoptive transfer, we explored the impact of constitutive IL-10 expression on C2D-IL10 macrophages in adipose tissue in vivo. Adipose tissue-isolated C2D-IL10 cells increased the percentage of CD206(+), CD301(+), CD11c(-)CD206(+) (M2) and CD11c(+)CD206(+) (M1b) on their cell surface, compared to isolated C2D-vector cells. These data suggest that the expression of IL-10 remains stable, alters the C2D-IL10 macrophage cell surface phenotype and may play a role in regulating macrophage interactions with the adipose tissue.
Collapse
Affiliation(s)
- Linglin Xie
- Department of Basic Sciences, School of Medicine and Health Science, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Qiang Fu
- Department of Basic Sciences, School of Medicine and Health Science, University of North Dakota, Grand Forks, North Dakota, United States of America
- Departments of Gerontology and Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Teresa M. Ortega
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Lun Zhou
- Department of Basic Sciences, School of Medicine and Health Science, University of North Dakota, Grand Forks, North Dakota, United States of America
- Departments of Gerontology and Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Dane Rasmussen
- Department of Basic Sciences, School of Medicine and Health Science, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Jacy O’Keefe
- Department of Basic Sciences, School of Medicine and Health Science, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Ke K. Zhang
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
- North Dakota IDeA Network of Biomedical Research Excellence Bioinformatics Core, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Stephen K. Chapes
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| |
Collapse
|
40
|
Schlottmann I, Ehrhart-Bornstein M, Wabitsch M, Bornstein SR, Lamounier-Zepter V. Calcium-dependent release of adipocyte fatty acid binding protein from human adipocytes. Int J Obes (Lond) 2013; 38:1221-7. [PMID: 24352293 DOI: 10.1038/ijo.2013.241] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/18/2013] [Accepted: 12/04/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND Fatty acid binding protein 4 (FABP4) is a predominantly cytosolic protein of the adipocytes, but also abundantly present in human plasma; its plasma concentrations were linked to obesity and metabolic syndrome. Recent studies have suggested a direct extracellular effect of FABP4 in the regulation of glucose metabolism and heart function independently of its effect as a carrier protein. Interestingly, FABP4 has no secretory signal sequence; hence, the mechanisms how FABP4 is released from adipocytes are unclear. METHODS AND RESULTS In this study we investigated the mechanisms for FABP4 secretion from human adipocytes by using isolated primary pre-adipocytes (PAs) and the human adipocyte cell strain Simpson-Golabi-Behmel syndrome. In undifferentiated PAs, FABP4 expression was barely detectable and increased continuously during differentiation. The increase in FABP4 mRNA expression was accompanied by high levels of FABP4 secretion. In differentiated human adipocytes, FABP4 secretion was not abolished by blocking the Golgi-dependent secretory pathway in vitro, supporting a non-classical secretion mechanism for FABP4. However, raising intracellular Ca(2+) levels enhanced FABP4 secretion in a concentration-dependent manner. CONCLUSION This study shows that FABP4 is actively released from human adipocytes in vitro via a non-classical, calcium-dependent mechanism.
Collapse
Affiliation(s)
- I Schlottmann
- Medical Clinic III, Dresden University of Technology, Dresden, Germany
| | | | - M Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, University of Ulm, Germany
| | - S R Bornstein
- Medical Clinic III, Dresden University of Technology, Dresden, Germany
| | | |
Collapse
|
41
|
Kralisch S, Ebert T, Lossner U, Jessnitzer B, Stumvoll M, Fasshauer M. Adipocyte fatty acid-binding protein is released from adipocytes by a non-conventional mechanism. Int J Obes (Lond) 2013; 38:1251-4. [PMID: 24445660 DOI: 10.1038/ijo.2013.232] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/18/2013] [Accepted: 12/08/2013] [Indexed: 01/22/2023]
Abstract
Adipocyte fatty acid-binding protein (AFABP) is an adipokine, which induces insulin resistance. However, AFABP does not possess any secretion-directed signals and the mechanisms for AFABP release have not been thoroughly assessed so far. In the current study, mechanisms for AFABP secretion were elucidated in 3T3-L1 adipocytes in vitro in the presence or absence of hormonal stimulation, calcium ionophore and secretion inhibitors by cell fractionation experiments, immunoblotting and ELISAs. We demonstrate that AFABP secretion is upregulated during adipocyte differentiation. AFABP secretion is not influenced by treatment with protein secretion inhibitors that block vesicular traffic at the endoplasmic reticulum and the Golgi apparatus. AFABP is secreted partially by adipocyte-derived microvesicles (ADMs), an established mechanism for unconventional secretion from adipocytes. Both total and ADM-secreted AFABP are downregulated by insulin and upregulated by the calcium ionophore ionomycin. Furthermore, murine RAW 264.7 macrophages secrete AFABP and AFABP release from these cells is upregulated by lipopolysaccharide treatment. Taken together, these results suggest that AFABP is actively released by unconventional mechanisms and by ADMs from 3T3-L1 adipocytes. Furthermore, AFABP secretion from fat cells is regulated by insulin and intracellular calcium.
Collapse
Affiliation(s)
- S Kralisch
- 1] Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany [2] IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - T Ebert
- 1] Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany [2] IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - U Lossner
- 1] Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany [2] IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - B Jessnitzer
- Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - M Stumvoll
- Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - M Fasshauer
- 1] Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany [2] IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| |
Collapse
|
42
|
Carson BP, Del Bas JM, Moreno-Navarrete JM, Fernandez-Real JM, Mora S. The rab11 effector protein FIP1 regulates adiponectin trafficking and secretion. PLoS One 2013; 8:e74687. [PMID: 24040321 PMCID: PMC3770573 DOI: 10.1371/journal.pone.0074687] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 08/07/2013] [Indexed: 12/28/2022] Open
Abstract
Adiponectin is an adipokine secreted by white adipocytes involved in regulating insulin sensitivity in peripheral tissues. Secretion of adiponectin in adipocytes relies on the endosomal system, however, the intracellular machinery involved in mediating adiponectin release is unknown. We have previously reported that intracellular adiponectin partially compartmentalizes with rab 5 and rab11, markers for the early/sorting and recycling compartments respectively. Here we have examined the role of several rab11 downstream effector proteins (rab11 FIPs) in regulating adiponectin trafficking and secretion. Overexpression of wild type rab11 FIP1, FIP3 and FIP5 decreased the amount of secreted adiponectin expressed in HEK293 cells, whereas overexpression of rab11 FIP2 or FIP4 had no effect. Furthermore shRNA-mediated depletion of FIP1 enhanced adiponectin release whereas knock down of FIP5 decreased adiponectin secretion. Knock down of FIP3 had no effect. In 3T3L1 adipocytes, endogenous FIP1 co-distributed intracellularly with endogenous adiponectin and FIP1 depletion enhanced adiponectin release without altering insulin-mediated trafficking of the glucose transporter Glut4. While adiponectin receptors internalized with transferrin receptors, there were no differences in transferrin receptor recycling between wild type and FIP1 depleted adipocytes. Consistent with its inhibitory role, FIP1 expression was decreased during adipocyte differentiation, by treatment with thiazolidinediones, and with increased BMI in humans. In contrast, FIP1 expression increased upon exposure of adipocytes to TNFα. In all, our findings identify FIP1 as a novel protein involved in the regulation of adiponectin trafficking and release.
Collapse
Affiliation(s)
- Brian P. Carson
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, the University of Liverpool, Liverpool, United Kingdom
| | - Josep Maria Del Bas
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, the University of Liverpool, Liverpool, United Kingdom
| | | | | | - Silvia Mora
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, the University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Finelli C, Tarantino G. Should visceral fat, strictly linked to hepatic steatosis, be depleted to improve survival? Hepatol Int 2013; 7:413-428. [PMID: 26201775 DOI: 10.1007/s12072-012-9406-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 10/01/2012] [Indexed: 02/06/2023]
Abstract
Numerous epidemiologic studies have implicated abdominal obesity as a major risk factor for insulin resistance, type 2 diabetes mellitus, cardiovascular disease, stroke, metabolic syndrome and its further expression, i.e., nonalcoholic fatty liver disease and death. Using novel models of visceral obesity, several studies have demonstrated that the relationship between visceral fat and longevity is causal, while the accrual of subcutaneous fat does not appear to play an important role in the etiology of disease risk. The need of reducing the visceral fat to improve survival, mainly taking into account the strict link between nonalcoholic fatty liver disease and the coronary artery disease is discussed.
Collapse
Affiliation(s)
- Carmine Finelli
- Center of Obesity and Eating Disorder, Stella Maris Mediterraneo Foundation Chiaromonte, Potenza, Italy
| | - Giovanni Tarantino
- Department of Clinical and Experimental Medicine, Federico II University Medical School of Naples, Naples, Italy.
| |
Collapse
|
44
|
D'Angelo CS, Kohl I, Varela MC, de Castro CIE, Kim CA, Bertola DR, Lourenço CM, Perez ABA, Koiffmann CP. Obesity with associated developmental delay and/or learning disability in patients exhibiting additional features: Report of novel pathogenic copy number variants. Am J Med Genet A 2013; 161A:479-86. [DOI: 10.1002/ajmg.a.35761] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 10/13/2012] [Indexed: 01/09/2023]
|
45
|
Than A, Tee WT, Chen P. Apelin secretion and expression of apelin receptors in 3T3-L1 adipocytes are differentially regulated by angiotensin type 1 and type 2 receptors. Mol Cell Endocrinol 2012; 351:296-305. [PMID: 22249006 DOI: 10.1016/j.mce.2012.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/27/2011] [Accepted: 01/04/2012] [Indexed: 12/28/2022]
Abstract
Adipocytes play pivotal roles in regulating metabolism through secretion of a variety of adipokines, which in turn is regulated by other metabolic factors (e.g., insulin). Understanding the regulations of adipokine secretion is important because adipokines are implicated with metabolic disorders, such as, obesity and diabetes mellitus. Here, we investigated the regulatory roles of angiotensin II (AngII) on the secretion of apelin in 3T3-L1 adipocytes, and distinct signaling pathways mediated by AngII receptor type 1 (AT₁) and type 2 (AT₂) were revealed. It was found that activation of AT₁ receptors stimulates apelin secretion in Ca²⁺, protein kinase C, and MAPK kinase dependent ways while activation of AT₂ receptors inhibits apelin secretion through cAMP and cGMP dependent pathways. Furthermore, we demonstrate that the expression of apelin receptor (APJ) is also similarly regulated by AT₁ and AT₂ receptors. Finally, a detailed AngII signaling map is proposed.
Collapse
Affiliation(s)
- Aung Than
- Division of Bioengineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore
| | | | | |
Collapse
|
46
|
Manabe Y, Miyatake S, Takagi M. Myokines: Do they really exist? JOURNAL OF PHYSICAL FITNESS AND SPORTS MEDICINE 2012. [DOI: 10.7600/jpfsm.1.51] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
47
|
Makino-Wakagi Y, Yoshimura Y, Uzawa Y, Zaima N, Moriyama T, Kawamura Y. Ellagic acid in pomegranate suppresses resistin secretion by a novel regulatory mechanism involving the degradation of intracellular resistin protein in adipocytes. Biochem Biophys Res Commun 2012; 417:880-5. [DOI: 10.1016/j.bbrc.2011.12.067] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 12/14/2011] [Indexed: 12/20/2022]
|
48
|
Novel adiponectin variants identified in type 2 diabetic patients reveal multimerization and secretion defects. PLoS One 2011; 6:e26792. [PMID: 22046359 PMCID: PMC3202584 DOI: 10.1371/journal.pone.0026792] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 10/04/2011] [Indexed: 12/14/2022] Open
Abstract
ADIPOQ, encoding adiponectin, is a candidate gene for type 2 diabetes (T2D) identified by genome-wide linkage analyses with supporting evidence showing the protein function in sensitizing insulin actions. In an endeavor to characterize candidate genes causing T2D in Thai patients, we identified 10 novel ADIPOQ variations, several of which were non-synonymous variations observed only in the patients. To examine the impact of these non-synonymous variations on adiponectin structure and biochemical characteristics, we conducted a structural analysis of the wild-type and variant proteins by in silico modeling and further characterized biochemical properties of the variants with predicted structural abnormalities from the modeling by molecular and biochemical studies. The recombinant plasmids containing wild-type and variant ADIPOQ cDNAs derived from the variations identified by our study (R55H, R112H, and R131H) and previous work (G90S and R112C) were constructed and transiently expressed and co-expressed in cultured HEK293T cells to investigate their oligomerization, interaction, and secretion. We found that the novel R55H variant impaired protein multimerization but it did not exert the effect over the co-expressed wild-type protein while novel R131H variant impaired protein secretion and also affected the co-expressed wild-type protein in a dominant negative fashion. The R131H variant could traffic from the endoplasmic reticulum to the Golgi, trans-Golgi network, and early endosome but could not be secreted. The R131H variant was likely to be degraded through the lysosomal system and inhibition of its degradation rescued the variant protein from secretion defect. We have shown the possibility of using in silico modeling for predicting the effect of amino acid substitution on adiponectin oligomerization. This is also the first report that demonstrates a dominant negative effect of the R131H variant on protein secretion and the possibility of using protein degradation inhibitors as therapeutic agents in the patients carrying adiponectin variants with secretion defect.
Collapse
|
49
|
Li X, DiFiglia M. The recycling endosome and its role in neurological disorders. Prog Neurobiol 2011; 97:127-41. [PMID: 22037413 DOI: 10.1016/j.pneurobio.2011.10.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/14/2011] [Accepted: 10/17/2011] [Indexed: 02/08/2023]
Abstract
The recycling endosome (RE) is an organelle in the endocytic pathway where plasma membranes (proteins and lipids) internalized by endocytosis are processed back to the cell surface for reuse. Endocytic recycling is the primary way for the cell to maintain constituents of the plasma membrane (Griffiths et al., 1989), i.e., to maintain the abundance of receptors and transporters on cell surfaces. Membrane traffic through the RE is crucial for several key cellular processes including cytokinesis and cell migration. In polarized cells, including neurons, the RE is vital for the generation and maintenance of the polarity of the plasma membrane. Many RE dependent cargo molecules are known to be important for neuronal function and there is evidence that improper function of key proteins in RE-associated pathways may contribute to the pathogenesis of neurological disorders, including Huntington's disease. The function of the RE in neurons is poorly understood. Therefore, there is need to understand how membrane dynamics in RE-associated pathways are affected or participate in the development or progression of neurological diseases. This review summarizes advances in understanding endocytic recycling associated with the RE, challenges in elucidating molecular mechanisms underlying RE function, and evidence for RE dysfunction in neurological disorders.
Collapse
Affiliation(s)
- Xueyi Li
- Laboratory of Cellular Neurobiology and Department of Neurology, Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | | |
Collapse
|
50
|
Negative regulation of adiponectin secretion by receptor interacting protein 140 (RIP140). Cell Signal 2011; 24:71-6. [PMID: 21872658 DOI: 10.1016/j.cellsig.2011.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 07/24/2011] [Accepted: 07/26/2011] [Indexed: 01/14/2023]
Abstract
RIP140 (receptor-interacting protein 140) is highly expressed in mature adipocytes and functions as a co-repressor for gene expression involved in lipid and glucose metabolism. In adipocytes, activated PKCε (Protein kinase C epsilon) phosphorylates nuclear RIP140 which is then subsequently arginine methylated and exported to the cytoplasm. In the cytoplasm, RI140 can elicit additional activities. Here we report a new functional role for cytoplasmic RIP140 in adipocyte in regulating adiponectin secretion. Targeting cytoplasmic RIP140 by knocking down RIP140 itself or its nuclear export trigger, PKCε, promotes the secretion of adiponectin without affecting the production or oligomerization of adiponectin. Consequentially, conditioned media from either RIP140- or PKCε-silenced adipocytes, which contain higher levels of adiponectin, enhance glucose uptake in C2C12 cells and reduce gluconeogenesis in HepG2 cells. Further, these effects can be inhibited by an adiponectin-neutralizing antibody. The effect of cytoplasmic RIP140 in regulating adiponectin secretion is via interacting with AS160, a known RIP140-interacting protein. This study reveals a new functional role for cytoplasmic RIP140 in modulating adiponectin vesicle secretion, and suggests that targeting cytoplasmic RIP140 may be a potentially effective therapeutic strategy to improve adiponectin secretion and possibly to manage metabolic disorders.
Collapse
|