101
|
Cheng F, Yun SJ, Cao JL, Chang MC, Meng JL, Liu JY, Cheng YF, Feng CP. Differential Gene Expression and Biological Analyses of Primary Hepatocytes Following D-Chiro-Inositol Supplement. Front Endocrinol (Lausanne) 2021; 12:700049. [PMID: 34335474 PMCID: PMC8320774 DOI: 10.3389/fendo.2021.700049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Dietary supplements have improved the prevention of insulin resistance and metabolic diseases, which became a research hotspot in food science and nutrition. Obesity and insulin resistance, caused by a high-fat diet, eventually result in severe metabolic diseases, can be prevented with the dietary supplement D-chiro-inositol (DCI). In this work, we isolated mice primary hepatocytes with palmitic acid stimulation and DCI was applied to compare and contrast its effects of in primary hepatocyte biology. Before and after intervention with DCI, we used RNA-Seq technology to establish a primary hepatocyte transcriptome gene profile. We found that both PA and DCI cause a wide variation in gene expression. Particularly, we found that DCI plays critical role in this model by acting on glycolysis and gluconeogenesis. Overall, we generated extensive transcripts from primary hepatocytes and uncovered new functions and gene targets for DCI.
Collapse
Affiliation(s)
- Feier Cheng
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
| | - Shao-jun Yun
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
| | - Jin-ling Cao
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
| | - Ming-chang Chang
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
- Shanxi Research Station for Engineering Technology of Edible Fungi, Shanxi Agricultural University, Taigu, China
| | - Jun-long Meng
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
- Shanxi Research Station for Engineering Technology of Edible Fungi, Shanxi Agricultural University, Taigu, China
| | - Jing-yu Liu
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
| | - Yan-fen Cheng
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
| | - Cui-ping Feng
- College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
- *Correspondence: Cui-ping Feng,
| |
Collapse
|
102
|
Han MS, Perry RJ, Camporez JP, Scherer PE, Shulman GI, Gao G, Davis RJ. A feed-forward regulatory loop in adipose tissue promotes signaling by the hepatokine FGF21. Genes Dev 2020; 35:133-146. [PMID: 33334822 PMCID: PMC7778269 DOI: 10.1101/gad.344556.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
In this study, Han et al. demonstrate that JNK signaling in adipocytes causes an increased circulating concentration of the hepatokine fibroblast growth factor 21 (FGF21) that regulates systemic metabolism. This regulatory loop represents a novel signaling paradigm that connects autocrine and endocrine signaling modes of the same hormone in different tissues. The cJun NH2-terminal kinase (JNK) signaling pathway is activated by metabolic stress and promotes the development of metabolic syndrome, including hyperglycemia, hyperlipidemia, and insulin resistance. This integrated physiological response involves cross-talk between different organs. Here we demonstrate that JNK signaling in adipocytes causes an increased circulating concentration of the hepatokine fibroblast growth factor 21 (FGF21) that regulates systemic metabolism. The mechanism of organ crosstalk is mediated by a feed-forward regulatory loop caused by JNK-regulated FGF21 autocrine signaling in adipocytes that promotes increased expression of the adipokine adiponectin and subsequent hepatic expression of the hormone FGF21. The mechanism of organ cross-talk places circulating adiponectin downstream of autocrine FGF21 expressed by adipocytes and upstream of endocrine FGF21 expressed by hepatocytes. This regulatory loop represents a novel signaling paradigm that connects autocrine and endocrine signaling modes of the same hormone in different tissues.
Collapse
Affiliation(s)
- Myoung Sook Han
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - João-Paulo Camporez
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
103
|
Abstract
The current paradigm of type 2 diabetes (T2D) is gluco-centric, being exclusively categorized by glycemic characteristics. The gluco-centric paradigm views hyperglycemia as the primary target, being driven by resistance to insulin combined with progressive beta cells failure, and considers glycemic control its ultimate treatment goal. Most importantly, the gluco-centric paradigm considers the non-glycemic diseases associated with T2D, e.g., obesity, dyslipidemia, hypertension, macrovascular disease, microvascular disease and fatty liver as 'risk factors' and/or 'outcomes' and/or 'comorbidities', rather than primary inherent disease aspects of T2D. That is in spite of their high prevalence (60-90%) and major role in profiling T2D morbidity and mortality. Moreover, the gluco-centric paradigm fails to realize that the non-glycemic diseases of T2D are driven by insulin and, except for glycemic control, response to insulin in T2D is essentially the rule rather than the exception. Failure of the gluco-centric paradigm to offer an exhaustive unifying view of the glycemic and non-glycemic diseases of T2D may have contributed to T2D being still an unmet need. An mTORC1-centric paradigm maintains that hyperactive mTORC1 drives the glycemic and non-glycemic disease aspects of T2D. Hyperactive mTORC1 is proposed to act as double-edged agent, namely, to interfere with glycemic control by disrupting the insulin receptor-Akt transduction pathway, while concomitantly driving the non-glycemic diseases of T2D. The mTORC1-centric paradigm may offer a novel perspective for T2D in terms of pathogenesis, clinical focus and treatment strategy.
Collapse
Affiliation(s)
- Jacob Bar-Tana
- Hebrew University Medical School, 91120, Jerusalem, Israel.
| |
Collapse
|
104
|
Castorani V, Polidori N, Giannini C, Blasetti A, Chiarelli F. Insulin resistance and type 2 diabetes in children. Ann Pediatr Endocrinol Metab 2020; 25:217-226. [PMID: 33401880 PMCID: PMC7788344 DOI: 10.6065/apem.2040090.045] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/05/2020] [Indexed: 12/27/2022] Open
Abstract
Type 2 diabetes (T2D) is an emerging health risk in obese children and adolescents. Both environmental (lack of physical activity, excess nutritional intake, sedentary lifestyle) and genetic factors contribute to this global epidemic. The growing prevalence of T2D in youth is also associated with a consistently increased incidence of metabolic and cardiovascular complications. Insulin resistance (IR), i.e., whole-body decreased glucose uptake in response to physiological insulin levels, determines impaired glucose homeostasis and it is recognized as cardinal trigger of T2D and cardiovascular disease in both adults and children. In particular, IR and beta-cell dysfunction lead to the persistent hyperglycemia which characterizes T2D. Indeed, both pathological states influence each other and presumably play a crucial, synergistic role in the pathogenesis of T2D, although the precise mechanisms are not completely understood. However, beta-cell dysfunction and IR induce impaired glucose metabolism, thus leading to the progression to T2D. Therefore, understanding the mechanisms correlated with the decline of beta-cell function and IR is crucial in order to control, prevent, and treat T2D in youth. This review focuses on the current knowledge regarding IR and T2D in children and adolescents and showcases interesting opportunities and stimulating challenges for the development of new preventative approaches and therapeutic strategies for young patients with T2D.
Collapse
Affiliation(s)
| | - Nella Polidori
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | - Cosimo Giannini
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | | | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti, Chieti, Italy,Address for correspondence: Francesco Chiarelli, MD, PhD Department of Pediatrics, University of Chieti, Via dei Vestini, 5, I-66100 Chieti, Italy Tel: +39-0871-358015 Fax: +39-0871-574538 E-mail:
| |
Collapse
|
105
|
Benchoula K, Arya A, Parhar IS, Hwa WE. FoxO1 signaling as a therapeutic target for type 2 diabetes and obesity. Eur J Pharmacol 2020; 891:173758. [PMID: 33249079 DOI: 10.1016/j.ejphar.2020.173758] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/12/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
Glucose production and the consumption of high levels of carbohydrate increase the chance of insulin resistance, especially in cases of obesity. Therefore, maintaining a balanced glucose homeostasis might form a strategy to prevent or cure diabetes and obesity. The activation and inhibition of glucose production is complicated due to the presence of many interfering pathways. These pathways can be viewed at the downstream level because they activate certain transcription factors, which include the Forkhead-O1 (FoxO1). This has been identified as a significant agent in the pancreas, liver, and adipose tissue, which is significant in the regulation of lipids and glucose. The objective of this review is to discuss the intersecting portrayal of FoxO1 and its parallel cross-talk which highlights obesity-induced insulin susceptibility in the discovery of a targeted remedy. The review also analyses current progress and provides a blueprint on therapeutics, small molecules, and extracts/phytochemicals which are explored at the pre-clinical level.
Collapse
Affiliation(s)
- Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia; Department of Pharmacology and Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, 3010, Australia; Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), Bukit Gambir, Gelugor, Pulau Pinang, Malaysia
| | - Ishwar S Parhar
- Monash University (Malaysia) BRIMS, Jeffrey Cheah School of Medicine & Health Sciences, Malaysia
| | - Wong Eng Hwa
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia.
| |
Collapse
|
106
|
Rojas-Rodriguez R, Ziegler R, DeSouza T, Majid S, Madore AS, Amir N, Pace VA, Nachreiner D, Alfego D, Mathew J, Leung K, Moore Simas TA, Corvera S. PAPPA-mediated adipose tissue remodeling mitigates insulin resistance and protects against gestational diabetes in mice and humans. Sci Transl Med 2020; 12:eaay4145. [PMID: 33239385 PMCID: PMC8375243 DOI: 10.1126/scitranslmed.aay4145] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 04/25/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022]
Abstract
Pregnancy is a physiological state of continuous adaptation to changing maternal and fetal nutritional needs, including a reduction of maternal insulin sensitivity allowing for appropriately enhanced glucose availability to the fetus. However, excessive insulin resistance in conjunction with insufficient insulin secretion results in gestational diabetes mellitus (GDM), greatly increasing the risk for pregnancy complications and predisposing both mothers and offspring to future metabolic disease. Here, we report a signaling pathway connecting pregnancy-associated plasma protein A (PAPPA) with adipose tissue expansion in pregnancy. Adipose tissue plays a central role in the regulation of insulin sensitivity, and we show that, in both mice and humans, pregnancy caused remodeling of adipose tissue evidenced by altered adipocyte size, vascularization, and in vitro expansion capacity. PAPPA is known to be a metalloprotease secreted by human placenta that modulates insulin-like growth factor (IGF) bioavailability through prolteolysis of IGF binding proteins (IGFBPs) 2, 4, and 5. We demonstrate that recombinant PAPPA can stimulate ex vivo human adipose tissue expansion in an IGFBP-5- and IGF-1-dependent manner. Moreover, mice lacking PAPPA displayed impaired adipose tissue remodeling, pregnancy-induced insulin resistance, and hepatic steatosis, recapitulating multiple aspects of human GDM. In a cohort of 6361 pregnant women, concentrations of circulating PAPPA are inversely correlated with glycemia and odds of developing GDM. These data identify PAPPA and the IGF signaling pathway as necessary for the regulation of maternal adipose tissue physiology and systemic glucose homeostasis, with consequences for long-term metabolic risk and potential for therapeutic use.
Collapse
Affiliation(s)
- Raziel Rojas-Rodriguez
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Rachel Ziegler
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sana Majid
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Aylin S Madore
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Nili Amir
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Veronica A Pace
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Daniel Nachreiner
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David Alfego
- Division of Data Sciences and Technology, IT, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jomol Mathew
- Division of Data Sciences and Technology, IT, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Katherine Leung
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Tiffany A Moore Simas
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
107
|
Vav2 catalysis-dependent pathways contribute to skeletal muscle growth and metabolic homeostasis. Nat Commun 2020; 11:5808. [PMID: 33199701 PMCID: PMC7669868 DOI: 10.1038/s41467-020-19489-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle promotes metabolic balance by regulating glucose uptake and the stimulation of multiple interorgan crosstalk. We show here that the catalytic activity of Vav2, a Rho GTPase activator, modulates the signaling output of the IGF1- and insulin-stimulated phosphatidylinositol 3-kinase pathway in that tissue. Consistent with this, mice bearing a Vav2 protein with decreased catalytic activity exhibit reduced muscle mass, lack of proper insulin responsiveness and, at much later times, a metabolic syndrome-like condition. Conversely, mice expressing a catalytically hyperactive Vav2 develop muscle hypertrophy and increased insulin responsiveness. Of note, while hypoactive Vav2 predisposes to, hyperactive Vav2 protects against high fat diet-induced metabolic imbalance. These data unveil a regulatory layer affecting the signaling output of insulin family factors in muscle. Skeletal muscle plays a key role in regulating systemic glucose and metabolic homeostasis. Here, the authors show that the catalytic activity of Vav2, an activator of Rho GTPases, modulates those processes by favoring the responsiveness of this tissue to insulin and related factors.
Collapse
|
108
|
Miyamoto JÉ, Reginato A, Portovedo M, Dos Santos RM, Stahl MA, Le Stunff H, Latorraca MQ, de Barros Reis MA, Arantes VC, Doneda DL, Ignacio-Souza LM, Torsoni AS, Grimaldi R, Ribeiro APB, Torsoni MA, Milanski M. Interesterified palm oil impairs glucose homeostasis and induces deleterious effects in liver of Swiss mice. Metabolism 2020; 112:154350. [PMID: 32910938 DOI: 10.1016/j.metabol.2020.154350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/30/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Interesterified fats have largely replaced the partially hydrogenated oils which are the main dietary source of trans fat in industrialized food. This process promotes a random rearrangement of the native fatty acids and the results are different triacylglycerol (TAG) molecules without generating trans isomers. The role of interesterified fats in metabolism remains unclear. We evaluated metabolic parameters, glucose homeostasis and inflammatory markers in mice fed with normocaloric and normolipidic diets or hypercaloric and high-fat diet enriched with interesterified palm oil. METHODS Male Swiss mice were randomly divided into four experimental groups and submitted to either normolipidic palm oil diet (PO), normolipidic interesterified palm oil diet (IPO), palm oil high-fat diet (POHF) or interesterified palm oil high-fat diet (IPOHF) during an 8 weeks period. RESULTS When compared to the PO group, IPO group presented higher body mass, hyperglycemia, impaired glucose tolerance, evidence of insulin resistance and greater production of glucose in basal state during pyruvate in situ assay. We also observed higher protein content of hepatic PEPCK and increased cytokine mRNA expression in the IPO group when compared to PO. Interestingly, IPO group showed similar parameters to POHF and IPOHF groups. CONCLUSION The results indicate that substitution of palm oil for interesterified palm oil even on normocaloric and normolipidic diet could negatively modulate metabolic parameters and glucose homeostasis as well as cytokine gene expression in the liver and white adipose tissue. This data support concerns about the effects of interesterified fats on health and could promote further discussions about the safety of the utilization of this unnatural fat by food industry.
Collapse
Affiliation(s)
- Josiane Érica Miyamoto
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Andressa Reginato
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Mariana Portovedo
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Raísa Magno Dos Santos
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Hervé Le Stunff
- Paris-Saclay Institute of Neuroscience, CNRS UMR 9197, Université Paris-Sud, University Paris Saclay, Orsay, France
| | | | | | | | - Diego Luiz Doneda
- Physiology Laboratory, Department of Basic Health Sciences, Federal University of Mato Grosso, Cuiabá, Brazil
| | - Leticia Martins Ignacio-Souza
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Adriana Souza Torsoni
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Renato Grimaldi
- School of Food Engineering, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Marcio Alberto Torsoni
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Marciane Milanski
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil.
| |
Collapse
|
109
|
Raje V, Ahern KW, Martinez BA, Howell NL, Oenarto V, Granade ME, Kim JW, Tundup S, Bottermann K, Gödecke A, Keller SR, Kadl A, Bland ML, Harris TE. Adipocyte lipolysis drives acute stress-induced insulin resistance. Sci Rep 2020; 10:18166. [PMID: 33097799 PMCID: PMC7584576 DOI: 10.1038/s41598-020-75321-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Stress hyperglycemia and insulin resistance are evolutionarily conserved metabolic adaptations to severe injury including major trauma, burns, or hemorrhagic shock (HS). In response to injury, the neuroendocrine system increases secretion of counterregulatory hormones that promote rapid mobilization of nutrient stores, impair insulin action, and ultimately cause hyperglycemia, a condition known to impair recovery from injury in the clinical setting. We investigated the contributions of adipocyte lipolysis to the metabolic response to acute stress. Both surgical injury with HS and counterregulatory hormone (epinephrine) infusion profoundly stimulated adipocyte lipolysis and simultaneously triggered insulin resistance and hyperglycemia. When lipolysis was inhibited, the stress-induced insulin resistance and hyperglycemia were largely abolished demonstrating an essential requirement for adipocyte lipolysis in promoting stress-induced insulin resistance. Interestingly, circulating non-esterified fatty acid levels did not increase with lipolysis or correlate with insulin resistance during acute stress. Instead, we show that impaired insulin sensitivity correlated with circulating levels of the adipokine resistin in a lipolysis-dependent manner. Our findings demonstrate the central importance of adipocyte lipolysis in the metabolic response to injury. This insight suggests new approaches to prevent insulin resistance and stress hyperglycemia in trauma and surgery patients and thereby improve outcomes.
Collapse
Affiliation(s)
- Vidisha Raje
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Katelyn W Ahern
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Brittany A Martinez
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Nancy L Howell
- Department of Medicine, Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Vici Oenarto
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.,Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mitchell E Granade
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Jae Woo Kim
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Smanla Tundup
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | | | - Axel Gödecke
- Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Susanna R Keller
- Department of Medicine, Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Alexandra Kadl
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.,Department of Medicine, Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | - Michelle L Bland
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
110
|
Adipocyte lipolysis: from molecular mechanisms of regulation to disease and therapeutics. Biochem J 2020; 477:985-1008. [PMID: 32168372 DOI: 10.1042/bcj20190468] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/19/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
Fatty acids (FAs) are stored safely in the form of triacylglycerol (TAG) in lipid droplet (LD) organelles by professional storage cells called adipocytes. These lipids are mobilized during adipocyte lipolysis, the fundamental process of hydrolyzing TAG to FAs for internal or systemic energy use. Our understanding of adipocyte lipolysis has greatly increased over the past 50 years from a basic enzymatic process to a dynamic regulatory one, involving the assembly and disassembly of protein complexes on the surface of LDs. These dynamic interactions are regulated by hormonal signals such as catecholamines and insulin which have opposing effects on lipolysis. Upon stimulation, patatin-like phospholipase domain containing 2 (PNPLA2)/adipocyte triglyceride lipase (ATGL), the rate limiting enzyme for TAG hydrolysis, is activated by the interaction with its co-activator, alpha/beta hydrolase domain-containing protein 5 (ABHD5), which is normally bound to perilipin 1 (PLIN1). Recently identified negative regulators of lipolysis include G0/G1 switch gene 2 (G0S2) and PNPLA3 which interact with PNPLA2 and ABHD5, respectively. This review focuses on the dynamic protein-protein interactions involved in lipolysis and discusses some of the emerging concepts in the control of lipolysis that include allosteric regulation and protein turnover. Furthermore, recent research demonstrates that many of the proteins involved in adipocyte lipolysis are multifunctional enzymes and that lipolysis can mediate homeostatic metabolic signals at both the cellular and whole-body level to promote inter-organ communication. Finally, adipocyte lipolysis is involved in various diseases such as cancer, type 2 diabetes and fatty liver disease, and targeting adipocyte lipolysis is of therapeutic interest.
Collapse
|
111
|
Guo CY, Liao WT, Qiu RJ, Zhou DS, Ni WJ, Yu CP, Zeng Y. Aurantio-obtusin improves obesity and insulin resistance induced by high-fat diet in obese mice. Phytother Res 2020; 35:346-360. [PMID: 32749748 DOI: 10.1002/ptr.6805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 02/02/2023]
Abstract
Aurantio-obtusin (AUR) is the main bioactive compound among the anthraquinones, from Cassia seed extract. This study was conducted to identify whether AUR could improve obesity and insulin resistance, induced by a high-fat diet in obese mice. Mice were fed a high-fat diet for 6 weeks and were then assigned to the high-fat diet (HFD) control group, the AUR 5 mg/kg group, or the AUR 10 mg/kg group. AUR improves glucose by activating the expression of PI3K, Akt and GLUT4, GLUT2. AUR altered the expression levels of several lipid metabolism-related and adipokine genes. AUR decreased the mRNA expression of PPAR-γ, FAS and increased the mRNA expression of PPAR-α in liver. AUR lowered SREBP-1c, FAS, SCD-1, inflammatory cytokines, and increased the expression of PPAR-γ, PPAR-α, CPT-1, and adiponectin in white adipose tissue (WAT). AUR docking with the insulin receptor showed that the residues of the insulin receptor, ectodomain, were the same as those around the emodin. The effect of AUR may be elicited by regulating the activity of the insulin signaling pathway, expression of lipid metabolism-related genes, and expression of inflammatory cytokine markers to improve adiposity, insulin resistance, and dyslipidemia.
Collapse
Affiliation(s)
- Cong-Ying Guo
- Guangdong Pharmaceutical University, Guangzhou, China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangzhou, China
| | - Wei-Tao Liao
- Guangdong Pharmaceutical University, Guangzhou, China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangzhou, China
| | - Rui-Jin Qiu
- Wuzhou Institute of Agricultural Sciences, Wuzhou, China
| | - Dan-Shui Zhou
- Guangdong Pharmaceutical University, Guangzhou, China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangzhou, China
| | - Wei-Ju Ni
- Guangdong Pharmaceutical University, Guangzhou, China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangzhou, China
| | - Cui-Ping Yu
- Wuzhou Institute of Agricultural Sciences, Wuzhou, China
| | - Yu Zeng
- Guangdong Pharmaceutical University, Guangzhou, China.,Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangzhou, China
| |
Collapse
|
112
|
Jiang S, Young JL, Wang K, Qian Y, Cai L. Diabetic‑induced alterations in hepatic glucose and lipid metabolism: The role of type 1 and type 2 diabetes mellitus (Review). Mol Med Rep 2020; 22:603-611. [PMID: 32468027 PMCID: PMC7339764 DOI: 10.3892/mmr.2020.11175] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 03/06/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a growing health concern in society. Type 1 and type 2 DM are the two main types of diabetes; both types are chronic diseases that affect glucose metabolism in the body and the impaired regulation of glucose and lipid metabolism promotes the development and progression of DM. During the physiological metabolism process, the liver serves a unique role in glucose and lipid metabolism. The present article aimed to review the association between DM and glucose metabolism in the liver and discuss the changes of the following hepatic glucose fluxes: Gluconeogenesis, glucose/glucose 6‑phosphate cycling, glycogenolysis, glycogenesis and the pentose phosphate pathway. Moreover, the incidence of fatty liver in DM was also investigated.
Collapse
Affiliation(s)
- Saizhi Jiang
- Department of Paediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
- Department of Paediatrics, Paediatric Research Institute, University of Louisville, Louisville, KY 40202, USA
| | - Jamie L. Young
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Kai Wang
- Department of Paediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
- Department of Paediatrics, Paediatric Research Institute, University of Louisville, Louisville, KY 40202, USA
| | - Yan Qian
- Department of Paediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lu Cai
- Department of Paediatrics, Paediatric Research Institute, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
- Radiation Oncology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
113
|
Banerjee S, Ghoshal S, Stevens JR, McCommis KS, Gao S, Castro-Sepulveda M, Mizgier ML, Girardet C, Kumar KG, Galgani JE, Niehoff ML, Farr SA, Zhang J, Butler AA. Hepatocyte expression of the micropeptide adropin regulates the liver fasting response and is enhanced by caloric restriction. J Biol Chem 2020; 295:13753-13768. [PMID: 32727846 DOI: 10.1074/jbc.ra120.014381] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
The micropeptide adropin encoded by the clock-controlled energy homeostasis-associated gene is implicated in the regulation of glucose metabolism. However, its links to rhythms of nutrient intake, energy balance, and metabolic control remain poorly defined. Using surveys of Gene Expression Omnibus data sets, we confirm that fasting suppresses liver adropin expression in lean C57BL/6J (B6) mice. However, circadian rhythm data are inconsistent. In lean mice, caloric restriction (CR) induces bouts of compulsive binge feeding separated by prolonged fasting intervals, increasing NAD-dependent deacetylase sirtuin-1 signaling important for glucose and lipid metabolism regulation. CR up-regulates adropin expression and induces rhythms correlating with cellular stress-response pathways. Furthermore, adropin expression correlates positively with phosphoenolpyruvate carboxokinase-1 (Pck1) expression, suggesting a link with gluconeogenesis. Our previous data suggest that adropin suppresses gluconeogenesis in hepatocytes. Liver-specific adropin knockout (LAdrKO) mice exhibit increased glucose excursions following pyruvate injections, indicating increased gluconeogenesis. Gluconeogenesis is also increased in primary cultured hepatocytes derived from LAdrKO mice. Analysis of circulating insulin levels and liver expression of fasting-responsive cAMP-dependent protein kinase A (PKA) signaling pathways also suggests enhanced responses in LAdrKO mice during a glucagon tolerance test (250 µg/kg intraperitoneally). Fasting-associated changes in PKA signaling are attenuated in transgenic mice constitutively expressing adropin and in fasting mice treated acutely with adropin peptide. In summary, hepatic adropin expression is regulated by nutrient- and clock-dependent extrahepatic signals. CR induces pronounced postprandial peaks in hepatic adropin expression. Rhythms of hepatic adropin expression appear to link energy balance and cellular stress to the intracellular signal transduction pathways that drive the liver fasting response.
Collapse
Affiliation(s)
- Subhashis Banerjee
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Sarbani Ghoshal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Joseph R Stevens
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Saint Louis University Liver Center, Saint Louis University School of Medicine, St. Louis, Missouri USA
| | - Su Gao
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA
| | - Mauricio Castro-Sepulveda
- Laboratorio de Ciencias del Ejercicio. Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Maria L Mizgier
- Departamento de Ciencias de la SaludCarrera de Nutrición y Dietética and Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Clemence Girardet
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - K Ganesh Kumar
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA
| | - Jose E Galgani
- Departamento de Ciencias de la SaludCarrera de Nutrición y Dietética and Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Michael L Niehoff
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Saint Louis University School of Medicine; Research Service, John Cochran Division, Saint Louis Veterans Affairs Medical Center, Missouri, USA
| | - Susan A Farr
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Saint Louis University School of Medicine; Research Service, John Cochran Division, Saint Louis Veterans Affairs Medical Center, Missouri, USA
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
114
|
Zheng T, Wang Q, Dong Y, Ma W, Zhang Y, Zhao Y, Bian F, Chen L. High Glucose-Aggravated Hepatic Insulin Resistance: Role of the NLRP3 Inflammasome in Kupffer Cells. Obesity (Silver Spring) 2020; 28:1270-1282. [PMID: 32538511 DOI: 10.1002/oby.22821] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/17/2020] [Accepted: 03/31/2020] [Indexed: 01/09/2023]
Abstract
OBJECTIVE This study aimed to investigate whether the NLRP3 inflammasome in Kupffer cells (KCs) can be activated in response to high glucose (HG) and to evaluate its influence on hepatic insulin sensitivity. METHODS Primary KCs and hepatocytes were isolated from mice, and lipid accumulation, glucose output, and insulin sensitivity of hepatocytes were investigated after culturing either alone or with KCs exposed to HG. The influence of HG-induced NLRP3 inflammasome activation in KCs on insulin sensitivity of hepatocytes was examined. Treatment with gadolinium trichloride caused KC depletion, and, subsequently, a streptozotocin-induced hyperglycemic mouse model was used to confirm the influence of KCs on hepatic insulin sensitivity. RESULTS Hepatocytes cocultured with KCs showed enhanced lipid accumulation, glucose output, and impaired insulin sensitivity when exposed to HG. Enhanced NLRP3 inflammasome activation was also evident in both hepatocytes and KCs. Moreover, KCs that were pretreated with caspase-1 inhibitor, NLRP3 inhibitor, and NLRP3 small interfering RNA corrected coculture-induced aberrances in insulin action and NLRP3 inflammasome activation in hepatocytes. KC coculture also increased interleukin-1β (IL-1β)-mediated nuclear factor-κB (NF-κB) activation in hepatocytes. In hyperglycemic mice, KC depletion inhibited NLRP3 inflammasome activation and improved hepatic insulin sensitivity. CONCLUSIONS NLRP3 inflammasome activation impaired insulin sensitivity through KC-derived IL-1β-mediated NF-κB activation in hepatocytes exposed to HG.
Collapse
Affiliation(s)
- Tao Zheng
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Qibin Wang
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yongcheng Dong
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Weidong Ma
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yonghong Zhang
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yan Zhao
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Fang Bian
- Department of Pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Li Chen
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
115
|
Li W, Wan H, Yan S, Yan Z, Chen Y, Guo P, Ramesh T, Cui Y, Ning L. Gold nanoparticles synthesized with Poria cocos modulates the anti-obesity parameters in high-fat diet and streptozotocin induced obese diabetes rat model. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2020.04.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
116
|
Yang YM, Kuen DS, Chung Y, Kurose H, Kim SG. Gα 12/13 signaling in metabolic diseases. Exp Mol Med 2020; 52:896-910. [PMID: 32576930 PMCID: PMC7338450 DOI: 10.1038/s12276-020-0454-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
As the key governors of diverse physiological processes, G protein-coupled receptors (GPCRs) have drawn attention as primary targets for several diseases, including diabetes and cardiovascular disease. Heterotrimeric G proteins converge signals from ~800 members of the GPCR family. Among the members of the G protein α family, the Gα12 family members comprising Gα12 and Gα13 have been referred to as gep oncogenes. Gα12/13 levels are altered in metabolic organs, including the liver and muscles, in metabolic diseases. The roles of Gα12/13 in metabolic diseases have been investigated. In this review, we highlight findings demonstrating Gα12/13 amplifying or dampening regulators of phenotype changes. We discuss the molecular basis of G protein biology in the context of posttranslational modifications to heterotrimeric G proteins and the cell signaling axis. We also highlight findings providing insights into the organ-specific, metabolic and pathological roles of G proteins in changes associated with specific cells, energy homeostasis, glucose metabolism, liver fibrosis and the immune and cardiovascular systems. This review summarizes the currently available knowledge on the importance of Gα12/13 in the physiology and pathogenesis of metabolic diseases, which is presented according to the basic understanding of their metabolic actions and underlying cellular and molecular bases. Understanding the activities of two members of a vital category of proteins called G proteins, which initiate metabolic changes when signaling molecules bind to cells, could lead to new therapies for many diseases. Researchers in South Korea and Japan, led by Sang Geon Kim at Seoul National University, review the significance of the Gα12 and Gα13 proteins in diseases characterised by significant changes in metabolism, including liver conditions and disorders of the cardiovascular and immune systems. Specific roles for the proteins have been identified by a variety of methods, including studying the effect of disabling the genes that code for them in mice. Recent insights suggest that drugs interfering with the activity of these Gα proteins might help treat many conditions in which the molecular signalling networks involving the proteins are disrupted.
Collapse
Affiliation(s)
- Yoon Mee Yang
- College of Pharmacy, Kangwon National University, Chuncheon, 24341, South Korea
| | - Da-Sol Kuen
- College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Yeonseok Chung
- College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Sang Geon Kim
- College of Pharmacy, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
117
|
Giudetti AM, Micioni Di Bonaventura MV, Ferramosca A, Longo S, Micioni Di Bonaventura E, Friuli M, Romano A, Gaetani S, Cifani C. Brief daily access to cafeteria-style diet impairs hepatic metabolism even in the absence of excessive body weight gain in rats. FASEB J 2020; 34:9358-9371. [PMID: 32463138 DOI: 10.1096/fj.201902757r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Numerous nutritional approaches aimed at reducing body weight have been developed as a strategy to reduce obesity. Most of these interventions rely on reducing caloric intake or limiting calories access to a few hours per day. In this work, we analyzed the effects of the extended (24 hours/day) or restricted (1 hour/day) access to a cafeteria-style (CAF) diet, on rat body weight and hepatic lipid metabolism, with respect to control rats (CTR) fed with a standard chow diet. The body weight gain of restricted-fed rats was not different from CTR, despite the slightly higher total caloric intake, but resulted significantly lower than extended-fed rats, which showed a CAF diet-induced obesity and a dramatically higher total caloric intake. However, both CAF-fed groups of rats showed, compared to CTR, unhealthy serum and hepatic parameters such as higher serum glucose level, lower HDL values, and increased hepatic triacylglycerol and cholesterol amount. The hepatic expression and activity of key enzymes of fatty acid synthesis, acetyl-CoA carboxylase (ACC), and fatty acid synthase (FAS), was similarly reduced in both CAF-fed groups of rats with respect to CTR. Anyway, while in extended-fed rats this reduction was associated to a long-term mechanism involving sterol regulatory element-binding protein-1 (SREBP-1), in restricted-fed animals a short-term mechanism based on PKA and AMPK activation occurred in the liver. Furthermore, hepatic fatty acid oxidation (FAO) and oxidative stress resulted significantly increased in extended, but not in restricted-fed rats, as compared to CTR. Overall, these results demonstrate that although limiting the total caloric intake might successfully fight obesity development, the nutritional content of the diet is the major determinant for the health status.
Collapse
Affiliation(s)
- Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | | | - Alessandra Ferramosca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Serena Longo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | | | - Marzia Friuli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
118
|
Bai Y, Liu J, Yang L, Zhong L. New insights into serum/extracellular thioredoxin in regulating hepatic insulin receptor activation. Biochim Biophys Acta Gen Subj 2020; 1864:129630. [PMID: 32376199 DOI: 10.1016/j.bbagen.2020.129630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/15/2020] [Accepted: 04/28/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Serum thioredoxin of type-2 diabetic patients is significantly higher than that of healthy people. Pathophysiological significance is unclear. METHODS Effects of serum/extracellular thioredoxin on phosphorylation (activation) of hepatic insulin receptor (IR) were investigated by using methods in biochemistry, cell/molecular biology and mass spectrometry. RESULTS In human serum, thioredoxin and insulin may interact. Their mixture contains a mixed disulfide between insulin B-chain and thioredoxin-Cys73, which limits their activities. In contrast, free form of serum/extracellular thioredoxin is active, and can regulate phosphorylation of insulin receptor β-subunits (IRβ) via direct/indirect mechanisms. The direct mechanism associates with positive regulation. Serum/extracellular thioredoxin increases insulin binding to IR, facilitating insulin-induced phosphorylation of IRβ and downstream AKT. The indirect mechanism is involved in negative regulation. Entry of extracellular thioredoxin into hepatic cells via IR enhances the expression and activity of cellular protein-tyrosine phosphatase 1B (PTP1B), which negatively regulates IRβ phosphorylation. After coordination between these two mechanisms, the positive impact of serum/extracellular thioredoxin overwhelms its negative impact on IRβ phosphorylation, which subsequently accelerates hepatic glucose uptake. In hepatic cells with thioredoxin deficiency, insulin-induced IRβ phosphorylation is decreased, which could be restored by extracellular thioredoxin entry. Moreover, the results from assaying 475 serum samples demonstrate a discriminating value of serum thioredoxin activity in diagnosing type-2 diabetes. CONCLUSION Serum/extracellular thioredoxin plays a critical role in regulating hepatic IRβ phosphorylation. GENERAL SIGNIFICANCE In case of insulin resistance/type-2 diabetes, hepatic IRβ is at low phosphorylation level, thereby the improvement effect of serum/extracellular thioredoxin on insulin-induced IRβ phosphorylation seems particularly important.
Collapse
Affiliation(s)
- Yun Bai
- Medical School, University of Chinese Academy of Sciences, the Campus of Yanqi, Huai Rou, Beijing 101407, China
| | - Jia Liu
- Medical School, University of Chinese Academy of Sciences, the Campus of Yanqi, Huai Rou, Beijing 101407, China
| | - Lijuan Yang
- Department of Endocrinology, Chinese PLA General Hospital, Beijing 100853, China.
| | - Liangwei Zhong
- Medical School, University of Chinese Academy of Sciences, the Campus of Yanqi, Huai Rou, Beijing 101407, China.
| |
Collapse
|
119
|
Thota RN, Moughan PJ, Singh H, Garg ML. GlucoTRIG: a novel tool to determine the nutritional quality of foods and meals in general population. Lipids Health Dis 2020; 19:83. [PMID: 32366255 PMCID: PMC7199359 DOI: 10.1186/s12944-020-01268-w] [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: 02/17/2020] [Accepted: 04/24/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND This study aimed to develop a novel criterion, GlucoTRIG, to rank meals for healthiness, that considers both glycaemic (serum insulin) and lipaemic (serum triglycerides) responses. METHODS Healthy volunteers (n = 10) were recruited with the aim of deriving a standard GlucoTRIG value for a reference meal. Volunteers consumed the reference meal (2 regular slices of wholemeal bread; 250 mL chocolate flavoured milk; 7 g butter and 11 g peanut butter) comprising of carbohydrate, fat and protein (41, 40 and 16% energy respectively) on three different occasions with a minimum washout period of 3 days. The GlucoTRIG value was determined as the difference between the product of insulin and triglyceride obtained from venous blood samples at baseline and the product of insulin and triglyceride at 180 min. RESULTS There were no significant differences in the participants' dietary intakes and their metabolic parameters between three visits (P > 0.005). The GlucoTRIG value obtained from three mean values of the reference meal was found to be 19 ± 3.5. There were no significant (P = 0.2303) differences observed between the GlucoTRIG values for the three visits. CONCLUSION GlucoTRIG, consisting of both glycaemic and lipaemic responses, may be a physiologically relevant tool to rank foods and meals for reducing the risk of metabolic diseases. TRIAL REGISTRATION ACTRN12619000973112.
Collapse
Affiliation(s)
- Rohith N Thota
- Nutraceuticals Research Program, School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Riddet Institute, Massey University, Palmerston North, New Zealand.,Priority Research Centre in Physical Activity & Nutrition, University of Newcastle, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Paul J Moughan
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Harjinder Singh
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Manohar L Garg
- Nutraceuticals Research Program, School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, NSW, Australia. .,Riddet Institute, Massey University, Palmerston North, New Zealand. .,Priority Research Centre in Physical Activity & Nutrition, University of Newcastle, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
120
|
Schmitz-Peiffer C. Deconstructing the Role of PKC Epsilon in Glucose Homeostasis. Trends Endocrinol Metab 2020; 31:344-356. [PMID: 32305097 DOI: 10.1016/j.tem.2020.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
The failure of insulin to suppress glucose production by the liver is a key aspect of the insulin resistance seen in type 2 diabetes. Lipid-activated protein kinase C epsilon has long been identified as an important mediator of diet-induced glucose intolerance and hepatic insulin resistance and the current view emphasizes a mechanism involving phosphorylation of the insulin receptor by the kinase to inhibit downstream insulin action. However, the significance of this direct effect in the liver has now been challenged by tissue-specific deletion of PKCε, which demonstrated a more prominent role for the kinase in adipose tissue to promote glucose intolerance. New insights regarding the role of PKCε therefore contribute to the understanding of indirect effects on hepatic glucose metabolism.
Collapse
Affiliation(s)
- Carsten Schmitz-Peiffer
- Garvan Institute of Medical Research, Darlinghurst Sydney, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia.
| |
Collapse
|
121
|
Merry TL, Hedges CP, Masson SW, Laube B, Pöhlmann D, Wueest S, Walsh ME, Arnold M, Langhans W, Konrad D, Zarse K, Ristow M. Partial impairment of insulin receptor expression mimics fasting to prevent diet-induced fatty liver disease. Nat Commun 2020; 11:2080. [PMID: 32350271 PMCID: PMC7190665 DOI: 10.1038/s41467-020-15623-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 03/19/2020] [Indexed: 12/28/2022] Open
Abstract
Excessive insulin signaling through the insulin receptor (IR) may play a role in the pathogenesis of diet-induced metabolic disease, including obesity and type 2 diabetes. Here we investigate whether heterozygous impairment of insulin receptor (IR) expression limited to peripheral, i.e. non-CNS, tissues of adult mice impacts the development of high-fat diet-induced metabolic deterioration. While exhibiting some features of insulin resistance, PerIRKO+/− mice display a hepatic energy deficit accompanied by induction of energy-sensing AMPK, mitochondrial biogenesis, PPARα, unexpectedly leading to protection from, and reversal of hepatic lipid accumulation (steatosis hepatis, NAFLD). Consistently, and unlike in control mice, the PPARα activator fenofibrate fails to further affect hepatic lipid accumulation in PerIRKO+/− mice. Taken together, and opposing previously established diabetogenic features of insulin resistance, incomplete impairment of insulin signaling may mimic central aspects of calorie restriction to limit hepatic lipid accumulation during conditions of metabolic stress. Hyper-insulinemia associated with excess calorie intake may cause metabolic dysfunction. Here the authors report that mice with partially reduced insulin receptor expression in peripheral tissues are protected from and experience reversal of fatty liver disease.
Collapse
Affiliation(s)
- Troy L Merry
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland. .,Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand.
| | - Chris P Hedges
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Stewart W Masson
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Beate Laube
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Doris Pöhlmann
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Michael E Walsh
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, Institute of Food and Nutrition, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Institute of Food and Nutrition, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Kim Zarse
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland.
| |
Collapse
|
122
|
Czech MP. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol Metab 2020; 34:27-42. [PMID: 32180558 PMCID: PMC6997501 DOI: 10.1016/j.molmet.2019.12.014] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/21/2019] [Accepted: 12/23/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The diminished glucose lowering effect of insulin in obesity, called "insulin resistance," is associated with glucose intolerance, type 2 diabetes, and other serious maladies. Many publications on this topic have suggested numerous hypotheses on the molecular and cellular disruptions that contribute to the syndrome. However, significant uncertainty remains on the mechanisms of its initiation and long-term maintenance. SCOPE OF REVIEW To simplify insulin resistance analysis, this review focuses on the unifying concept that adipose tissue is a central regulator of systemic glucose homeostasis by controlling liver and skeletal muscle metabolism. Key aspects of adipose function related to insulin resistance reviewed are: 1) the modes by which specific adipose tissues control hepatic glucose output and systemic glucose disposal, 2) recently acquired understanding of the underlying mechanisms of these modes of regulation, and 3) the steps in these pathways adversely affected by obesity that cause insulin resistance. MAJOR CONCLUSIONS Adipocyte heterogeneity is required to mediate the multiple pathways that control systemic glucose tolerance. White adipocytes specialize in sequestering triglycerides away from the liver, muscle, and other tissues to limit toxicity. In contrast, brown/beige adipocytes are very active in directly taking up glucose in response to β adrenergic signaling and insulin and enhancing energy expenditure. Nonetheless, white, beige, and brown adipocytes all share the common feature of secreting factors and possibly exosomes that act on distant tissues to control glucose homeostasis. Obesity exerts deleterious effects on each of these adipocyte functions to cause insulin resistance.
Collapse
Affiliation(s)
- Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
123
|
López-Soldado I, Bertini A, Adrover A, Duran J, Guinovart JJ. Maintenance of liver glycogen during long-term fasting preserves energy state in mice. FEBS Lett 2020; 594:1698-1710. [PMID: 32159852 DOI: 10.1002/1873-3468.13770] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/20/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Abstract
Glycogen shortage during fasting coincides with dramatic changes in hepatic adenine nucleotide levels. The aim of this work was to study the relevance of liver glycogen in the regulation of the hepatic energy state during food deprivation. To this end, we examined the response of mice with sustained increased liver glycogen content to prolonged fasting. In order to increase hepatic glycogen content, we generated mice that overexpress protein targeting to glycogen (PTG) in the liver (PTGOE mice). Control and PTGOE mice were fed ad libitum or fasted for 36 h. Upon fasting, PTGOE mice retained significant hepatic glycogen stores and maintained hepatic energy status. Furthermore, we show that liver glycogen controls insulin sensitivity, gluconeogenesis, lipid metabolism, and ketogenesis upon nutrient deprivation.
Collapse
Affiliation(s)
- Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Angelo Bertini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain
| | - Anna Adrover
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.,Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| |
Collapse
|
124
|
Uncarboxylated osteocalcin ameliorates hepatic glucose and lipid metabolism in KKAy mice via activating insulin signaling pathway. Acta Pharmacol Sin 2020; 41:383-393. [PMID: 31659239 PMCID: PMC7470804 DOI: 10.1038/s41401-019-0311-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 09/19/2019] [Indexed: 12/21/2022]
Abstract
Osteocalcin, expressed in osteoblasts of the bone marrow, undergoes post-translational carboxylation and deposits in mineralized bone matrix. A portion of osteocalcin remains uncarboxylated (uncarboxylated osteocalcin, GluOC) that is released into blood where it functions as a hormone to regulate insulin secretion and insulin sensitivity. As insulin resistance is closely associated with metabolic syndrome, this study is aimed to elucidate how GluOC regulates glucose and lipid metabolism in KKAy mice, an animal model displaying obese, hyperglycemia, hyperinsulinemia, insulin resistance, and hepatic steatosis. GluOC (3, 30 ng/g per day, ig) was orally administered to female KKAy mice for 4 weeks. Whole-body insulin sensitivity, glucose metabolism, hepatic steatosis, dyslipidemia were examined using routine laboratory assays. We found that GluOC administration significantly enhanced insulin sensitivity in KKAy mice by activating hepatic IRβ/PI3K/Akt pathway and elevated the whole-body insulin sensitivity with decreased FPI and HOMA-IR index. Furthermore, GluOC administration alleviated hyperglycemia through suppressing gluconeogenesis and promoting glycogen synthesis in KKAy mice and in cultured hepatocytes in vitro. Moreover, GluOC administration dose-dependently ameliorated dyslipidemia and attenuated hepatic steatosis in KKAy mice by inhibiting hepatic de novo lipogenesis and promoting fatty-acid β-oxidation. These results demonstrate that GluOC effectively enhances hepatic insulin sensitivity, improves hyperglycemia and ameliorates hepatic steatosis in KKAy mice, suggesting that GluOC could be a promising drug candidate for treating metabolic syndrome.
Collapse
|
125
|
Bredemolic Acid Ameliorates Selected Liver Function Biomarkers in a Diet-Induced Prediabetic Rat Model. Can J Gastroenterol Hepatol 2020; 2020:2475301. [PMID: 32149046 PMCID: PMC7053450 DOI: 10.1155/2020/2475301] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/21/2019] [Accepted: 01/06/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Prediabetes is an intermediary hyperglycaemic state that precedes type 2 diabetes mellitus (T2DM) in which abnormal metabolism of glucose and lipids occurs in organs such as the liver. Evidence has shown that, about 70% of T2DM patients develop hepatic dysfunction which is found to begin during the prediabetic stage. Bredemolic acid, a pentacyclic triterpene, has been found to improve insulin sensitivity in diet-induced prediabetic rats. The effects of this compound on liver function, however, are unknown. This study was therefore designed to investigate the effects of BA on liver function in high fat-high carbohydrate (HFHC) diet-induced prediabetic rats. METHODS Thirty-six (36) male rats that weigh 150 g-180 g were divided into two groups, the non-prediabetic (n = 6) and the prediabetic groups (n = 6) and the prediabetic groups (n = 6) and the prediabetic groups (. RESULTS The induction of prediabetes resulted in increased release of liver enzymes (AST and ALT), increased liver glycogen and triglyceride, lipid peroxidation, and decreased sterol regulatory element-binding protein (SREBP1c) and antioxidant enzymes. However, the administration of BA decreased liver enzyme concentrations, decreased hepatic oxidative stress, and improved antioxidant enzymes such as SOD and GPx. CONCLUSION BA administration improved liver function in diet-induced prediabetic rats in the presence or absence of dietary intervention.
Collapse
|
126
|
Teofilović A, Brkljačić J, Djordjevic A, VojnovićMilutinović D, Tappy L, Matić G, Veličković N. Impact of insulin and glucocorticoid signalling on hepatic glucose homeostasis in the rat exposed to high-fructose diet and chronic stress. Int J Food Sci Nutr 2020; 71:815-825. [PMID: 32070154 DOI: 10.1080/09637486.2020.1728236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Overconsumption of fructose-enriched beverages and everyday stress are involved in the pathogenesis of metabolic disorders through modulation of hepatic glucose metabolism. The aim of the study was to investigate whether interaction of high-fructose diet and chronic stress alter insulin and glucocorticoid signalling thus affecting hepatic glucose homeostasis. High-fructose diet led to hyperinsulinemia, increased glucose transporter 2 level, elevated protein kinase B (Akt) phosphorylation, increased glucokinase mRNA and phospho-to-total glycogen synthase kinase 3 ratio and decreased expression of gluconeogenic genes. Fructose diet also led to stimulated glucocorticoid prereceptor metabolism, but downstream signalling remained unchanged due to increased glucocorticoid clearance. Stress did not affect hepatic insulin and glucocorticoid signalling nor glucose metabolism, while the interaction of the factors was observed only for glucokinase expression. The results suggest that, under conditions of fructose-induced hyperinsulinemia, suppression of gluconeogenesis and glycogen synthase activation contribute to the maintenance of glucose homeostasis. The increased glucocorticoid inactivation may represent an adaptive mechanism to prevent hyperglycaemia.
Collapse
Affiliation(s)
- Ana Teofilović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Brkljačić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela VojnovićMilutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Luc Tappy
- Department of Physiology, University of Lausanne, UNIL-CHUV, Lausanne, Switzerland
| | - Gordana Matić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nataša Veličković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
127
|
Enhanced insulin signaling and its downstream effects in iron-overloaded primary hepatocytes from hepcidin knock-out mice. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118621. [DOI: 10.1016/j.bbamcr.2019.118621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/08/2019] [Accepted: 12/03/2019] [Indexed: 12/22/2022]
|
128
|
Virtue S, Petkevicius K, Moreno-Navarrete JM, Jenkins B, Hart D, Dale M, Koulman A, Fernández-Real JM, Vidal-Puig A. Peroxisome Proliferator-Activated Receptor γ2 Controls the Rate of Adipose Tissue Lipid Storage and Determines Metabolic Flexibility. Cell Rep 2020; 24:2005-2012.e7. [PMID: 30134163 PMCID: PMC6113930 DOI: 10.1016/j.celrep.2018.07.063] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/15/2018] [Accepted: 07/18/2018] [Indexed: 01/06/2023] Open
Abstract
One understudied function of white adipose tissue (AT) is its role in postprandial lipid buffering. In this study, we demonstrate that mice lacking the adipose tissue-specific transcription factor peroxisome proliferator-activated receptor γ2 (PPARγ2) exhibit a defect in their rate of adipose tissue lipid storage. Impaired adipose tissue storage rate reduces metabolic flexibility, without compromising fasted glucose tolerance or insulin sensitivity, even following prolonged high-fat feeding. However, acutely overfeeding PPARγ2-KO mice caused a 10-fold increase in insulin levels compared with controls. Although impaired adipose tissue storage rate did not result in insulin resistance in young mice, 1-year-old PPARγ2-KO mice developed skeletal muscle insulin resistance. Our data indicate that failed adipose tissue storage may occur prior to defects in glucose handling and that overfeeding protocols may uncover genes controlling adipose tissue storage rate, as opposed to capacity, and act as a diagnostic test for early-stage human metabolic disease. Mice lacking PPARγ2 have impaired adipose tissue lipid storage rate Low adipose tissue storage rate leads to metabolic inflexibility Acute hypercaloric challenges can detect impaired adipose tissue lipid storage rate Chronic adipose tissue metabolic inflexibility leads to insulin resistance with age
Collapse
Affiliation(s)
- Sam Virtue
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK.
| | - Kasparas Petkevicius
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - José Maria Moreno-Navarrete
- Biomedical Research Institute of Girona (IDIBGI), CIBERobn Pathophysiology of Obesity and Nutrition, Hospital of Girona "Dr. Josep Trueta," Avinguda de França s/n, and Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Benjamin Jenkins
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Daniel Hart
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Martin Dale
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Albert Koulman
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - José Manuel Fernández-Real
- Biomedical Research Institute of Girona (IDIBGI), CIBERobn Pathophysiology of Obesity and Nutrition, Hospital of Girona "Dr. Josep Trueta," Avinguda de França s/n, and Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Antonio Vidal-Puig
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK; Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK.
| |
Collapse
|
129
|
Loss of Transcriptional Repression by BCL6 Confers Insulin Sensitivity in the Setting of Obesity. Cell Rep 2019; 25:3283-3298.e6. [PMID: 30566857 PMCID: PMC6377366 DOI: 10.1016/j.celrep.2018.11.074] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/24/2018] [Accepted: 11/16/2018] [Indexed: 12/27/2022] Open
Abstract
Accumulation of visceral adiposity is directly linked to the morbidity of obesity, while subcutaneous body fat is considered more benign. We have identified an unexpected role for B cell lymphoma 6 (BCL6), a critical regulator of immunity, in the developmental expansion of subcutaneous adipose tissue. In adipocyte-specific knockout mice (Bcl6AKO), we found that Bcl6 deletion results in strikingly increased inguinal, but not perigonadal, adipocyte size and tissue mass in addition to marked insulin sensitivity. Genome-wide RNA expression and DNA binding analyses revealed that BCL6 controls gene networks involved in cell growth and fatty acid biosynthesis. Using deuterium label incorporation and comprehensive adipokine and lipid profiling, we discovered that ablation of adipocyte Bcl6 enhances subcutaneous adipocyte lipogenesis, increases levels of adiponectin and fatty acid esters of hydroxy fatty acids (FAHFAs), and prevents steatosis. Thus, our studies identify BCL6 as a negative regulator of subcutaneous adipose tissue expansion and metabolic health. Senagolage et al. identify BCL6 as a key regulator of body fat distribution. BCL6 directly represses fatty acid biosynthetic and growth genes in adipocytes. Mice constitutively lacking adipocyte Bcl6 exhibit expansion of their subcutaneous adipose tissue, enhanced insulin sensitivity, and protection from hepatic steatosis.
Collapse
|
130
|
Patel BM, Goyal RK. Liver and insulin resistance: New wine in old bottle!!! Eur J Pharmacol 2019; 862:172657. [DOI: 10.1016/j.ejphar.2019.172657] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022]
|
131
|
Bergman RN, Piccinini F, Kabir M, Ader M. Novel aspects of the role of the liver in carbohydrate metabolism. Metabolism 2019; 99:119-125. [PMID: 31158368 PMCID: PMC7216693 DOI: 10.1016/j.metabol.2019.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 01/21/2023]
Abstract
Malfunction of the liver is a central factor in metabolic disease. Glucose production by liver is complex and controlled via indirect mechanisms; insulin regulates adipose tissue lipolysis, and free fatty acids in turn regulate liver glucose output. This latter concept is confirmed by studies in L-Akt-Foxo1 knockout mice. The adipocyte is a likely locus of hepatic insulin resistance. Also, kidneys play a role in regulating glucose production; denervated kidneys abrogate the effect of fat feeding to cause insulin resistance. Glucose itself is an important regulator of liver metabolism ("glucose effectiveness"); after entering liver, glucose is phosphorylated and can be exported as lactate. Using the dynamic glucose/lactate relationship, we have been able to estimate glucose effectiveness in intact animals and human subjects. Families have been identified with a glucokinase regulatory protein defect; modeling demonstrates elevated glucokinase activity. Insulin clearance by liver is highly variable among normal individuals, and is under environmental control: high fat diet reduces clearance by 30%. Liver insulin clearance is significantly lower in African American (AA) adults and children compared to European American participants, accounting for fasting hyperinsulinemia in AA. We hypothesize that reduced hepatic insulin clearance causes peripheral insulin resistance and increased Type 2 diabetes in AA.
Collapse
Affiliation(s)
- Richard N Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America.
| | - Francesca Piccinini
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Morvarid Kabir
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Marilyn Ader
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| |
Collapse
|
132
|
Hörbelt T, Knebel B, Fahlbusch P, Barbosa D, de Wiza DH, Van de Velde F, Van Nieuwenhove Y, Lapauw B, Thoresen GH, Al-Hasani H, Müller-Wieland D, Ouwens DM, Kotzka J. The adipokine sFRP4 induces insulin resistance and lipogenesis in the liver. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2671-2684. [DOI: 10.1016/j.bbadis.2019.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/19/2019] [Accepted: 07/18/2019] [Indexed: 01/04/2023]
|
133
|
Zhou P, Santoro A, Peroni OD, Nelson AT, Saghatelian A, Siegel D, Kahn BB. PAHSAs enhance hepatic and systemic insulin sensitivity through direct and indirect mechanisms. J Clin Invest 2019; 129:4138-4150. [PMID: 31449056 PMCID: PMC6763232 DOI: 10.1172/jci127092] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/24/2019] [Indexed: 12/30/2022] Open
Abstract
Palmitic acid esters of hydroxy stearic acids (PAHSAs) are bioactive lipids with antiinflammatory and antidiabetic effects. PAHSAs reduce ambient glycemia and improve glucose tolerance and insulin sensitivity in insulin-resistant aged chow- and high-fat diet-fed (HFD-fed) mice. Here, we aimed to determine the mechanisms by which PAHSAs improve insulin sensitivity. Both acute and chronic PAHSA treatment enhanced the action of insulin to suppress endogenous glucose production (EGP) in chow- and HFD-fed mice. Moreover, chronic PAHSA treatment augmented insulin-stimulated glucose uptake in glycolytic muscle and heart in HFD-fed mice. The mechanisms by which PAHSAs enhanced hepatic insulin sensitivity included direct and indirect actions involving intertissue communication between adipose tissue and liver. PAHSAs inhibited lipolysis directly in WAT explants and enhanced the action of insulin to suppress lipolysis during the clamp in vivo. Preventing the reduction of free fatty acids during the clamp with Intralipid infusion reduced PAHSAs' effects on EGP in HFD-fed mice but not in chow-fed mice. Direct hepatic actions of PAHSAs may also be important, as PAHSAs inhibited basal and glucagon-stimulated EGP directly in isolated hepatocytes through a cAMP-dependent pathway involving Gαi protein-coupled receptors. Thus, this study advances our understanding of PAHSA biology and the physiologic mechanisms by which PAHSAs exert beneficial metabolic effects.
Collapse
Affiliation(s)
- Peng Zhou
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Anna Santoro
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Odile D. Peroni
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew T. Nelson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Barbara B. Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
134
|
Zong J, Li S, Wang Y, Mo W, Sun R, Yu M. Bromodomain-containing protein 2 promotes lipolysis via ERK/HSL signalling pathway in white adipose tissue of mice. Gen Comp Endocrinol 2019; 281:105-116. [PMID: 31121164 DOI: 10.1016/j.ygcen.2019.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/17/2019] [Accepted: 05/14/2019] [Indexed: 01/10/2023]
Abstract
White adipose tissue (WAT) dysfunction is prevalent among patients with type 2 diabetes mellitus (T2DM). Uncontrolled free fatty acid (FFA) release from WAT stores has detrimental effects on lipid metabolism, leading to insulin resistance. Bromodomain-containing protein 2 (Brd2) has emerged as a central transcriptional regulator of adipocyte differentiation and pancreatic β-cell bioactivity. A recent study shows that Brd2 overexpression leads to insulin resistance in mice. However, the mechanisms underlying these effects have not been fully elucidated. This study provides the first evidence that adenoviral-mediated Brd2 overexpression in the WAT of mice increases lipolysis-related gene expression in addition to significantly reducing WAT size and promoting plasma FFA release. Brd2 overexpression in adipocytes also inhibits fat synthesis-related gene expression, while activating hormone-sensitive lipase (HSL) expression and ERK-dependent perilipin 1 inhibition as well as promoting glycerol release, which are all involved in lipolysis. Collectively, these results indicate that Brd2 triggers insulin resistance via lipolysis-mediated FFA release.
Collapse
Affiliation(s)
- Jiuyu Zong
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Shuting Li
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Yuxiong Wang
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Wei Mo
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Ruixin Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China.
| | - Min Yu
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
135
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide a brief summary of recent advances in our understanding of liver metabolism. The critical role of the liver in controlling whole-body energy homeostasis makes such understanding crucial to efficiently design new treatments for metabolic syndrome diseases, including type 2 diabetes (T2D). RECENT FINDINGS Significant advances have been made regarding our understanding of the direct and indirect effects of insulin on hepatic metabolism and the communication between the liver and other tissues. Moreover, the catabolic functions of glucagon, as well as the importance of hepatic redox status for the regulation of glucose production, are emerging as potential targets to reduce hyperglycemia. A resolution to the long-standing question "insulin suppression of hepatic glucose production, direct or indirect effect?" is starting to emerge. New advances in our understanding of important fasting-induced hepatic metabolic fluxes may help design better therapies for T2D.
Collapse
Affiliation(s)
- Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, 450 Brookline Av. LC-6219H, Boston, MA, 02215, USA.
| | - Clint D J Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, 450 Brookline Av. LC-6219H, Boston, MA, 02215, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, 450 Brookline Av. LC-6219H, Boston, MA, 02215, USA.
- Dana-Farber Cancer Institute, 450 Brookline Av. LC-6213, Boston, MA, 02215, USA.
| |
Collapse
|
136
|
Bergman RN, Piccinini F, Kabir M, Kolka CM, Ader M. Hypothesis: Role of Reduced Hepatic Insulin Clearance in the Pathogenesis of Type 2 Diabetes. Diabetes 2019; 68:1709-1716. [PMID: 31431441 PMCID: PMC6702636 DOI: 10.2337/db19-0098] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/02/2019] [Indexed: 12/19/2022]
Abstract
There is wide variance among individuals in the fraction of insulin cleared by the liver (20% to 80%). Hepatic insulin clearance is 67% lower in African Americans than European Americans. Clearance is also lower in African American children 7-13 years of age. Lower hepatic insulin clearance will result in peripheral hyperinsulinemia: this exacerbates insulin resistance, which stresses the β-cells, possibly resulting in their ultimate failure and onset of type 2 diabetes. We hypothesize that lower insulin clearance can be a primary cause of type 2 diabetes in at-risk individuals.
Collapse
Affiliation(s)
- Richard N Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Francesca Piccinini
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Morvarid Kabir
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Cathryn M Kolka
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Marilyn Ader
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
137
|
Jaiswal N, Gavin MG, Quinn WJ, Luongo TS, Gelfer RG, Baur JA, Titchenell PM. The role of skeletal muscle Akt in the regulation of muscle mass and glucose homeostasis. Mol Metab 2019; 28:1-13. [PMID: 31444134 PMCID: PMC6822261 DOI: 10.1016/j.molmet.2019.08.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Skeletal muscle insulin signaling is a major determinant of muscle growth and glucose homeostasis. Protein kinase B/Akt plays a prominent role in mediating many of the metabolic effects of insulin. Mice and humans harboring systemic loss-of-function mutations in Akt2, the most abundant Akt isoform in metabolic tissues, are glucose intolerant and insulin resistant. Since the skeletal muscle accounts for a significant amount of postprandial glucose disposal, a popular hypothesis in the diabetes field suggests that a reduction in Akt, specifically in skeletal muscle, leads to systemic glucose intolerance and insulin resistance. Despite this common belief, the specific role of skeletal muscle Akt in muscle growth and insulin sensitivity remains undefined. METHODS We generated multiple mouse models of skeletal muscle Akt deficiency to evaluate the role of muscle Akt signaling in vivo. The effects of these genetic perturbations on muscle mass, glucose homeostasis and insulin sensitivity were assessed using both in vivo and ex vivo assays. RESULTS Surprisingly, mice lacking Akt2 alone in skeletal muscle displayed normal skeletal muscle insulin signaling, glucose tolerance, and insulin sensitivity despite a dramatic reduction in phosphorylated Akt. In contrast, deletion of both Akt isoforms (M-AktDKO) prevented downstream signaling and resulted in muscle atrophy. Despite the absence of Akt signaling, in vivo and ex vivo insulin-stimulated glucose uptake were normal in M-AktDKO mice. Similar effects on insulin sensitivity were observed in mice with prolonged deletion (4 weeks) of both skeletal muscle Akt isoforms selectively in adulthood. Conversely, short term deletion (2 weeks) of skeletal muscle specific Akt in adult muscles impaired insulin tolerance paralleling the effect observed by acute pharmacological inhibition of Akt in vitro. Mechanistically, chronic ablation of Akt induced mitochondrial dysfunction and activation of AMPK, which was required for insulin-stimulated glucose uptake in the absence of Akt. CONCLUSIONS Together, these data indicate that chronic reduction in Akt activity alone in skeletal muscle is not sufficient to induce insulin resistance or prevent glucose uptake in all conditions. Therefore, since insulin-stimulated glucose disposal in skeletal muscle is markedly impaired in insulin-resistant states, we hypothesize that alterations in signaling molecules in addition to skeletal muscle Akt are necessary to perturb glucose tolerance and insulin sensitivity in vivo.
Collapse
Affiliation(s)
- N Jaiswal
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - M G Gavin
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - W J Quinn
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - T S Luongo
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - R G Gelfer
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - J A Baur
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - P M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
138
|
Dittmann A, Kennedy NJ, Soltero NL, Morshed N, Mana MD, Yilmaz ÖH, Davis RJ, White FM. High-fat diet in a mouse insulin-resistant model induces widespread rewiring of the phosphotyrosine signaling network. Mol Syst Biol 2019; 15:e8849. [PMID: 31464373 PMCID: PMC6674232 DOI: 10.15252/msb.20198849] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022] Open
Abstract
Obesity-associated type 2 diabetes and accompanying diseases have developed into a leading human health risk across industrialized and developing countries. The complex molecular underpinnings of how lipid overload and lipid metabolites lead to the deregulation of metabolic processes are incompletely understood. We assessed hepatic post-translational alterations in response to treatment of cells with saturated and unsaturated free fatty acids and the consumption of a high-fat diet by mice. These data revealed widespread tyrosine phosphorylation changes affecting a large number of enzymes involved in metabolic processes as well as canonical receptor-mediated signal transduction networks. Targeting two of the most prominently affected molecular features in our data, SRC-family kinase activity and elevated reactive oxygen species, significantly abrogated the effects of saturated fat exposure in vitro and high-fat diet in vivo. In summary, we present a comprehensive view of diet-induced alterations of tyrosine signaling networks, including proteins involved in fundamental metabolic pathways.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Fatty Acids/pharmacology
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Obesity/etiology
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Phosphorylation/drug effects
- Phosphotyrosine/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Processing, Post-Translational
- Proteomics/methods
- Rats
- Reactive Oxygen Species/agonists
- Reactive Oxygen Species/metabolism
- Signal Transduction
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Antje Dittmann
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Norman J Kennedy
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Nina L Soltero
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Nader Morshed
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Miyeko D Mana
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Broad Institute of Harvard and MITCambridgeMAUSA
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Broad Institute of Harvard and MITCambridgeMAUSA
- Department of PathologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Roger J Davis
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Howard Hughes Medical InstituteWorcesterMAUSA
| | - Forest M White
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| |
Collapse
|
139
|
Kim Y, Rouse M, González-Mariscal I, Egan JM, O'Connell JF. Dietary curcumin enhances insulin clearance in diet-induced obese mice via regulation of hepatic PI3K-AKT axis and IDE, and preservation of islet integrity. Nutr Metab (Lond) 2019; 16:48. [PMID: 31372175 PMCID: PMC6659288 DOI: 10.1186/s12986-019-0377-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/19/2019] [Indexed: 12/23/2022] Open
Abstract
Background Although type 2 diabetes mellitus (T2DM) is primarily characterized by sustained high levels of circulating glucose, other factors, such as obesity, chronic inflammation, fatty liver, and islet dysfunction significantly contribute to the development of this disease. To date, curcumin (CUR), a natural polyphenol and primary component of turmeric, shows putative therapeutic properties such as reducing the incidence of obesity-related diseases in mice. However, the mechanism by which CUR regulates insulin levels remains unclear. Methods This study investigates how dietary CUR improves insulin clearance and maintains a proper range of circulating insulin level in the diet-induced obesity (DIO) mouse model. Male C57BL/6 J mice were fed a control, a high fat/high sugar (HFS) or a HFS diet containing 0.4% (w/w) curcumin (HFS + CUR) (N = 16 per group) for 16 weeks. Results Mice given HFS + CUR had reduced body weight and fat accumulation in the liver and had lower blood insulin levels under fasting conditions compared to mice on HFS alone, resulting from significantly improved insulin clearance via upregulation of hepatic insulin-degrading enzyme (IDE). We also observed restoration of phosphoinositide 3-kinase (PI3K), especially class Ia catalytic subunits, p110α and p110β, and class Ib regulatory subunit, p101, and phosphorylated protein kinase B (AKT) expression levels in liver on HFS + CUR diet. Additionally, HFS + CUR fed mice had significantly smaller islets of Langerhans and increased glucagon contents compared to HFS fed mice, indicating less secretion of insulin in pancreas. The expression of thioredoxin interacting protein (TXNIP), a pro-oxidant and pro-apoptotic protein, was significantly elevated in mouse and human islets cultured under HFS mimicking conditions, which was mitigated by CUR treatment. Conclusions CUR supplementation in obese subjects may alleviate the burden imposed by HFS diets. Our data indicate administration of dietary CUR reinstates PI3K, AKT and IDE levels in obese mice. Additionally, CUR treatment preserves islet integrity by downregulation of TXNIP transcription levels. Therefore, dietary CUR may have the potential to serve as a novel therapeutic agent to address the underlying links of obesity and T2DM.
Collapse
Affiliation(s)
- Yoo Kim
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224 USA
| | - Michael Rouse
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224 USA
| | - Isabel González-Mariscal
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224 USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224 USA
| | - Jennifer F O'Connell
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224 USA
| |
Collapse
|
140
|
Carcinoembryonic Cell Adhesion-Related Molecule 2 Regulates Insulin Secretion and Energy Balance. Int J Mol Sci 2019; 20:ijms20133231. [PMID: 31266142 PMCID: PMC6651791 DOI: 10.3390/ijms20133231] [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: 05/17/2019] [Revised: 06/12/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
The Carcinoembryonic Antigen-Related Cell Adhesion Molecule (CEACAM) family of proteins plays a significant role in regulating peripheral insulin action by participating in the regulation of insulin metabolism and energy balance. In light of their differential expression, CEACAM1 regulates chiefly insulin extraction, whereas CEACAM2 appears to play a more important role in regulating insulin secretion and overall energy balance, including food intake, energy expenditure and spontaneous physical activity. We will focus this review on the role of CEACAM2 in regulating insulin metabolism and energy balance with an overarching goal to emphasize the importance of the coordinated regulatory effect of these related plasma membrane glycoproteins on insulin metabolism and action.
Collapse
|
141
|
Najjar SM, Perdomo G. Hepatic Insulin Clearance: Mechanism and Physiology. Physiology (Bethesda) 2019; 34:198-215. [PMID: 30968756 DOI: 10.1152/physiol.00048.2018] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Upon its secretion from pancreatic β-cells, insulin reaches the liver through the portal circulation to exert its action and eventually undergo clearance in the hepatocytes. In addition to insulin secretion, hepatic insulin clearance regulates the homeostatic level of insulin that is required to reach peripheral insulin target tissues to elicit proper insulin action. Receptor-mediated insulin uptake followed by its degradation constitutes the basic mechanism of insulin clearance. Upon its phosphorylation by the insulin receptor tyrosine kinase, carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) takes part in the insulin-insulin receptor complex to increase the rate of its endocytosis and targeting to the degradation pathways. This review summarizes how this process is regulated and how it is associated with insulin-degrading enzyme in the liver. It also discusses the physiological implications of impaired hepatic insulin clearance: Whereas reduced insulin clearance cooperates with increased insulin secretion to compensate for insulin resistance, it can also cause hepatic insulin resistance. Because chronic hyperinsulinemia stimulates hepatic de novo lipogenesis, impaired insulin clearance also causes hepatic steatosis. Thus impaired insulin clearance can underlie the link between hepatic insulin resistance and hepatic steatosis. Delineating these regulatory pathways should lead to building more effective therapeutic strategies against metabolic syndrome.
Collapse
Affiliation(s)
- Sonia M Najjar
- Department of Biomedical Sciences, Ohio University , Athens, Ohio.,Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio
| | - Germán Perdomo
- Departamento de Ciencias de la Salud, Universidad de Burgos , Burgos , Spain
| |
Collapse
|
142
|
Molinaro A, Becattini B, Mazzoli A, Bleve A, Radici L, Maxvall I, Sopasakis VR, Molinaro A, Bäckhed F, Solinas G. Insulin-Driven PI3K-AKT Signaling in the Hepatocyte Is Mediated by Redundant PI3Kα and PI3Kβ Activities and Is Promoted by RAS. Cell Metab 2019; 29:1400-1409.e5. [PMID: 30982732 DOI: 10.1016/j.cmet.2019.03.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 02/04/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Phosphatidylinositol-3-kinase (PI3K) activity is aberrant in tumors, and PI3K inhibitors are investigated as cancer therapeutics. PI3K signaling mediates insulin action in metabolism, but the role of PI3K isoforms in insulin signaling remains unresolved. Defining the role of PI3K isoforms in insulin signaling is necessary for a mechanistic understanding of insulin action and to develop PI3K inhibitors with optimal therapeutic index. We show that insulin-driven PI3K-AKT signaling depends on redundant PI3Kα and PI3Kβ activities, whereas PI3Kδ and PI3Kγ are largely dispensable. We have also found that RAS activity promotes AKT phosphorylation in insulin-stimulated hepatocytes and that promotion of insulin-driven AKT phosphorylation by RAS depends on PI3Kα. These findings reveal the detailed mechanism by which insulin activates AKT, providing an improved mechanistic understanding of insulin signaling. This improved model for insulin signaling predicts that isoform-selective PI3K inhibitors discriminating between PI3Kα and PI3Kβ should be dosed below their hyperglycemic threshold to achieve isoform selectivity.
Collapse
Affiliation(s)
- Angela Molinaro
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Barbara Becattini
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Arianna Mazzoli
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Augusto Bleve
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Lucia Radici
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ingela Maxvall
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Victoria Rotter Sopasakis
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Antonio Molinaro
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Giovanni Solinas
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
143
|
Charidemou E, Ashmore T, Li X, McNally BD, West JA, Liggi S, Harvey M, Orford E, Griffin JL. A randomized 3-way crossover study indicates that high-protein feeding induces de novo lipogenesis in healthy humans. JCI Insight 2019; 4:124819. [PMID: 31145699 PMCID: PMC6629161 DOI: 10.1172/jci.insight.124819] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 05/08/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Dietary changes have led to the growing prevalence of type 2 diabetes and nonalcoholic fatty liver disease. A hallmark of both disorders is hepatic lipid accumulation, derived in part from increased de novo lipogenesis. Despite the popularity of high-protein diets for weight loss, the effect of dietary protein on de novo lipogenesis is poorly studied. We aimed to characterize the effect of dietary protein on de novo lipid synthesis. METHODS We use a 3-way crossover interventional study in healthy males to determine the effect of high-protein feeding on de novo lipogenesis, combined with in vitro models to determine the lipogenic effects of specific amino acids. The primary outcome was a change in de novo lipogenesis–associated triglycerides in response to protein feeding. RESULTS We demonstrate that high-protein feeding, rich in glutamate, increases de novo lipogenesis–associated triglycerides in plasma (1.5-fold compared with control; P < 0.0001) and liver-derived very low-density lipoprotein particles (1.8-fold; P < 0.0001) in samples from human subjects (n = 9 per group). In hepatocytes, we show that glutamate-derived carbon is incorporated into triglycerides via palmitate. In addition, supplementation with glutamate, glutamine, and leucine, but not lysine, increased triglyceride synthesis and decreased glucose uptake. Glutamate, glutamine, and leucine increased activation of protein kinase B, suggesting that induction of de novo lipogenesis occurs via the insulin signaling cascade. CONCLUSION These findings provide mechanistic insight into how select amino acids induce de novo lipogenesis and insulin resistance, suggesting that high-protein feeding to tackle diabetes and obesity requires greater consideration. FUNDING The research was supported by UK Medical Research Council grants MR/P011705/1, MC_UP_A090_1006 and MR/P01836X/1. JLG is supported by the Imperial Biomedical Research Centre, National Institute for Health Research (NIHR). A subset of amino acids may induce de novo lipogenesis in humans, suggesting that use of high-protein diets to tackle diabetes requires greater consideration.
Collapse
Affiliation(s)
- Evelina Charidemou
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Tom Ashmore
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Xuefei Li
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ben D McNally
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - James A West
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Sonia Liggi
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Matthew Harvey
- Medical Research Council - Elsie Widdowson Laboratory, Cambridge, United Kingdom
| | - Elise Orford
- Medical Research Council - Elsie Widdowson Laboratory, Cambridge, United Kingdom
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom.,Computational and Systems Medicine, Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
144
|
Stöckli J, Zadoorian A, Cooke KC, Deshpande V, Yau B, Herrmann G, Kebede MA, Humphrey SJ, James DE. ABHD15 regulates adipose tissue lipolysis and hepatic lipid accumulation. Mol Metab 2019; 25:83-94. [PMID: 31105056 PMCID: PMC6601125 DOI: 10.1016/j.molmet.2019.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/26/2019] [Accepted: 05/01/2019] [Indexed: 01/01/2023] Open
Abstract
Objective Insulin suppresses adipose tissue lipolysis after a meal, playing a key role in metabolic homeostasis. This is mediated via the kinase Akt and its substrate phosphodiesterase 3B (PDE3B). Once phosphorylated and activated, PDE3B hydrolyses cAMP leading to the inactivation of cAMP-dependent protein kinase (PKA) and suppression of lipolysis. However, several gaps have emerged in this model. Here we investigated the role of the PDE3B-interacting protein, α/β-hydrolase ABHD15 in this process. Methods Lipolysis, glucose uptake, and signaling were assessed in ABHD15 knock down and knock out adipocytes and fat explants in response to insulin and/or β-adrenergic receptor agonist. Glucose and fatty acid metabolism were determined in wild type and ABHD15−/− littermate mice. Results Deletion of ABHD15 in adipocytes resulted in a significant defect in insulin-mediated suppression of lipolysis with no effect on insulin-mediated glucose uptake. ABHD15 played a role in suppressing PKA signaling as phosphorylation of the PKA substrate Perilipin-1 remained elevated in response to insulin upon ABHD15 deletion. ABHD15−/− mice had normal glucose metabolism but defective fatty acid metabolism: plasma fatty acids were elevated upon fasting and in response to insulin, and this was accompanied by elevated liver triglycerides upon β-adrenergic receptor activation. This is likely due to hyperactive lipolysis as evident by the larger triglyceride depletion in brown adipose tissue in these mice. Finally, ABHD15 protein levels were reduced in adipocytes from mice fed a Western diet, further implicating this protein in metabolic homeostasis. Conclusions Collectively, ABHD15 regulates adipocyte lipolysis and liver lipid accumulation, providing novel therapeutic opportunities for modulating lipid homeostasis in disease. Insulin was unable to suppress lipolysis in the absence of ABHD15 in adipocytes in vitro, ex vivo and in mice in vivo. The lipolysis defect was associated with defective signalling via protein kinase A and its substrate Perilipin-1. The defect was specific for lipolysis with no impairment in insulin signalling or insulin-stimulated glucose uptake. Deletion of ABHD15 caused a significant increase in fatty acid deposition in liver in response to stress.
Collapse
Affiliation(s)
- Jacqueline Stöckli
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Armella Zadoorian
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Kristen C Cooke
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Vinita Deshpande
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Belinda Yau
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Gaia Herrmann
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Melkam A Kebede
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
145
|
Mazibuko-Mbeje SE, Dludla PV, Johnson R, Joubert E, Louw J, Ziqubu K, Tiano L, Silvestri S, Orlando P, Opoku AR, Muller CJF. Aspalathin, a natural product with the potential to reverse hepatic insulin resistance by improving energy metabolism and mitochondrial respiration. PLoS One 2019; 14:e0216172. [PMID: 31048842 PMCID: PMC6497260 DOI: 10.1371/journal.pone.0216172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/14/2019] [Indexed: 02/07/2023] Open
Abstract
Aspalathin is a rooibos flavonoid with established blood glucose lowering properties, however, its efficacy to moderate complications associated with hepatic insulin resistance is unknown. To study such effects, C3A liver cells exposed to palmitate were used as a model of hepatic insulin resistance. These hepatocytes displayed impaired substrate metabolism, including reduced glucose transport and free fatty acid uptake. These defects included impaired insulin signaling, evident through reduced phosphatidylinositol-4,5-bisphosphate 3-kinase/ protein kinase B (PI3K/AKT) protein expression, and mitochondrial dysfunction, depicted by a lower mitochondrial respiration rate. Aspalathin was able to ameliorate these defects by correcting altered substrate metabolism, improving insulin signaling and mitochondrial bioenergetics. Activation of 5'-adenosine monophosphate-activated protein kinase (AMPK) may be a plausible mechanism by which aspalathin increases hepatic energy expenditure. Overall, these results encourage further studies assessing the potential use of aspalathin as a nutraceutical to improve hepatocellular energy expenditure, and reverse metabolic disease-associated complications.
Collapse
Affiliation(s)
- Sithandiwe E. Mazibuko-Mbeje
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Phiwayinkosi V. Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Elizabeth Joubert
- Plant Bioactives Group, Post-Harvest and Agro-Processing Technologies, Agricultural Research Council, Infruitec-Nietvoorbij, Stellenbosch, South Africa
- Department of Food Science, Stellenbosch University, Stellenbosch, South Africa
| | - Johan Louw
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Khanyisani Ziqubu
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Andy R. Opoku
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| |
Collapse
|
146
|
Zhang K, Guo X, Yan H, Wu Y, Pan Q, Shen JZ, Li X, Chen Y, Li L, Qi Y, Xu Z, Xie W, Zhang W, Threadgill D, He L, Villarreal D, Sun Y, White MF, Zheng H, Guo S. Phosphorylation of Forkhead Protein FoxO1 at S253 Regulates Glucose Homeostasis in Mice. Endocrinology 2019; 160:1333-1347. [PMID: 30951171 PMCID: PMC6482038 DOI: 10.1210/en.2018-00853] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/29/2019] [Indexed: 01/04/2023]
Abstract
The transcription factor forkhead box O1 (FoxO1) is a key mediator in the insulin signaling pathway and controls multiple physiological functions, including hepatic glucose production (HGP) and pancreatic β-cell function. We previously demonstrated that S256 in human FOXO1 (FOXO1-S256), equivalent to S253 in mouse FoxO1 (FoxO1-S253), is a key phosphorylation site mediating the effect of insulin as a target of protein kinase B on suppression of FOXO1 activity and expression of target genes responsible for gluconeogenesis. Here, we investigated the role of FoxO1-S253 phosphorylation in control of glucose homeostasis in vivo by generating global FoxO1-S253A/A knockin mice, in which FoxO1-S253 alleles were replaced with alanine (A substitution) blocking FoxO1-S253 phosphorylation. FoxO1-S253A/A mice displayed mild increases in feeding blood glucose and insulin levels but decreases in fasting blood glucose and glucagon concentrations, as well as a reduction in the ratio of pancreatic α-cells/β-cells per islet. FoxO1-S253A/A mice exhibited a slight increase in energy expenditure but barely altered food intake and glucose uptake among tissues. Further analyses revealed that FoxO1-S253A/A enhances FoxO1 nuclear localization and promotes the effect of glucagon on HGP. We conclude that dephosphorylation of S253 in FoxO1 may reflect a molecular basis of pancreatic plasticity during the development of insulin resistance.
Collapse
Affiliation(s)
- Kebin Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Xiaoqin Guo
- Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hui Yan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yuxin Wu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
- Queens University Belfast School of Biological Sciences, Belfast, United Kingdom
| | - Quan Pan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - James Zheng Shen
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Xiaopeng Li
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yunmei Chen
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Ling Li
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yajuan Qi
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Zihui Xu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Wei Xie
- Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Weiping Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - David Threadgill
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Ling He
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Daniel Villarreal
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yuxiang Sun
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Morris F White
- Division of Endocrinology, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Hongting Zheng
- Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shaodong Guo
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
- Correspondence: Shaodong Guo, PhD, Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, 123A Cater-Mattil Hall, College Station, Texas 77843. E-mail:
| |
Collapse
|
147
|
Guilherme A, Henriques F, Bedard AH, Czech MP. Molecular pathways linking adipose innervation to insulin action in obesity and diabetes mellitus. Nat Rev Endocrinol 2019; 15:207-225. [PMID: 30733616 PMCID: PMC7073451 DOI: 10.1038/s41574-019-0165-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adipose tissue comprises adipocytes and many other cell types that engage in dynamic crosstalk in a highly innervated and vascularized tissue matrix. Although adipose tissue has been studied for decades, it has been appreciated only in the past 5 years that extensive arborization of nerve fibres has a dominant role in regulating the function of adipose tissue. This Review summarizes the latest literature, which suggests that adipocytes signal to local sensory nerve fibres in response to perturbations in lipolysis and lipogenesis. Such adipocyte signalling to the central nervous system causes sympathetic output to distant adipose depots and potentially other metabolic tissues to regulate systemic glucose homeostasis. Paracrine factors identified in the past few years that mediate such adipocyte-neuron crosstalk are also reviewed. Similarly, immune cells and endothelial cells within adipose tissue communicate with local nerve fibres to modulate neurotransmitter tone, blood flow, adipocyte differentiation and energy expenditure, including adipose browning to produce heat. This understudied field of neurometabolism related to adipose tissue biology has great potential to reveal new mechanistic insights and potential therapeutic strategies for obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Felipe Henriques
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alexander H Bedard
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
148
|
Nagarajan SR, Paul-Heng M, Krycer JR, Fazakerley DJ, Sharland AF, Hoy AJ. Lipid and glucose metabolism in hepatocyte cell lines and primary mouse hepatocytes: a comprehensive resource for in vitro studies of hepatic metabolism. Am J Physiol Endocrinol Metab 2019; 316:E578-E589. [PMID: 30694691 DOI: 10.1152/ajpendo.00365.2018] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The liver is a critical tissue for maintaining glucose, fatty acid, and cholesterol homeostasis. Primary hepatocytes represent the gold standard for studying the mechanisms controlling hepatic glucose, lipid, and cholesterol metabolism in vitro. However, access to primary hepatocytes can be limiting, and therefore, other immortalized hepatocyte models are commonly used. Here, we describe substrate metabolism of cultured AML12, IHH, and PH5CH8 cells, hepatocellular carcinoma-derived HepG2s, and primary mouse hepatocytes (PMH) to identify which of these cell lines most accurately phenocopy PMH basal and insulin-stimulated metabolism. Insulin-stimulated glucose metabolism in PH5CH8 cells, and to a lesser extent AML12 cells, responded most similarly to PMH. Notably, glucose incorporation in HepG2 cells were 14-fold greater than PMH. The differences in glucose metabolic activity were not explained by differential protein expression of key regulators of these pathways, for example glycogen synthase and glycogen content. In contrast, fatty acid metabolism in IHH cells was the closest to PMHs, yet insulin-responsive fatty acid metabolism in AML12 and HepG2 cells was most similar to PMH. Finally, incorporation of acetate into intracellular-free cholesterol was comparable for all cells to PMH; however, insulin-stimulated glucose conversion into lipids and the incorporation of acetate into intracellular cholesterol esters were strikingly different between PMHs and all tested cell lines. In general, AML12 cells most closely phenocopied PMH in vitro energy metabolism. However, the cell line most representative of PMHs differed depending on the mode of metabolism being investigated, and so careful consideration is needed in model selection.
Collapse
Affiliation(s)
- Shilpa R Nagarajan
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney , New South Wales , Australia
| | - Moumita Paul-Heng
- Discipline of Surgery, Central Clinical School & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney , New South Wales , Australia
| | - James R Krycer
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney , New South Wales , Australia
| | - Daniel J Fazakerley
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney , New South Wales , Australia
| | - Alexandra F Sharland
- Discipline of Surgery, Central Clinical School & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney , New South Wales , Australia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney , New South Wales , Australia
| |
Collapse
|
149
|
Prins GH, Luangmonkong T, Oosterhuis D, Mutsaers HAM, Dekker FJ, Olinga P. A Pathophysiological Model of Non-Alcoholic Fatty Liver Disease Using Precision-Cut Liver Slices. Nutrients 2019; 11:E507. [PMID: 30818824 PMCID: PMC6470479 DOI: 10.3390/nu11030507] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/13/2019] [Accepted: 02/20/2019] [Indexed: 01/02/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common liver disorder closely related to metabolic syndrome. NAFLD can progress to an inflammatory state called non-alcoholic steatohepatitis (NASH), which may result in the development of fibrosis and hepatocellular carcinoma. To develop therapeutic strategies against NAFLD, a better understanding of the molecular mechanism is needed. Current in vitro NAFLD models fail to capture the essential interactions between liver cell types and often do not reflect the pathophysiological status of patients. To overcome limitations of commonly used in vitro and in vivo models, precision-cut liver slices (PCLSs) were used in this study. PCLSs, prepared from liver tissue obtained from male Wistar rats, were cultured in supraphysiological concentrations of glucose, fructose, insulin, and palmitic acid to mimic metabolic syndrome. Accumulation of lipid droplets was visible and measurable after 24 h in PCLSs incubated with glucose, fructose, and insulin, both in the presence and absence of palmitic acid. Upregulation of acetyl-CoA carboxylase 1 and 2, and of sterol responsive element binding protein 1c, suggests increased de novo lipogenesis in PCLSs cultured under these conditions. Additionally, carnitine palmitoyltransferase 1 expression was reduced, which indicates impaired fatty acid transport and disrupted mitochondrial β-oxidation. Thus, steatosis was successfully induced in PCLSs with modified culture medium. This novel ex vivo NAFLD model could be used to investigate the multicellular and molecular mechanisms that drive NAFLD development and progression, and to study potential anti-steatotic drugs.
Collapse
Affiliation(s)
- Grietje H Prins
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9712VM Groningen, The Netherlands.
| | - Theerut Luangmonkong
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9712VM Groningen, The Netherlands.
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, 10400 Bangkok, Thailand.
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9712VM Groningen, The Netherlands.
| | - Henricus A M Mutsaers
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9712VM Groningen, The Netherlands.
| | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, University of Groningen, 9712VM Groningen, The Netherlands.
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9712VM Groningen, The Netherlands.
| |
Collapse
|
150
|
Edgerton DS, Scott M, Farmer B, Williams PE, Madsen P, Kjeldsen T, Brand CL, Fledelius C, Nishimura E, Cherrington AD. Targeting insulin to the liver corrects defects in glucose metabolism caused by peripheral insulin delivery. JCI Insight 2019; 5:126974. [PMID: 30830873 DOI: 10.1172/jci.insight.126974] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Peripheral hyperinsulinemia resulting from subcutaneous insulin injection is associated with metabolic defects which include abnormal glucose metabolism. The first aim of this study was to quantify the impairments in liver and muscle glucose metabolism that occur when insulin is delivered via a peripheral vein compared to when it is given through its endogenous secretory route (the hepatic portal vein) in overnight fasted conscious dogs. The second aim was to determine if peripheral delivery of a hepato-preferential insulin analog could restore the physiologic response to insulin that occurs under meal feeding conditions. This study is the first to show that hepatic glucose uptake correlates with insulin's direct effects on the liver under hyperinsulinemic-hyperglycemic conditions. In addition, glucose uptake was equally divided between the liver and muscle when insulin was infused into the portal vein, but when it was delivered into a peripheral vein the percentage of glucose taken up by muscle was 4-times greater than that going to the liver, with liver glucose uptake being less than half of normal. These defects could not be corrected by adjusting the dose of peripheral insulin. On the other hand, hepatic and non-hepatic glucose metabolism could be fully normalized by a hepato-preferential insulin analog.
Collapse
Affiliation(s)
- Dale S Edgerton
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Melanie Scott
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Ben Farmer
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Phillip E Williams
- Vanderbilt University Medical Center, Division of Surgical Research, Nashville, Tennessee, USA
| | - Peter Madsen
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Thomas Kjeldsen
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Christian L Brand
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Christian Fledelius
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Erica Nishimura
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Alan D Cherrington
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| |
Collapse
|