1
|
Mercurio G, Giacco A, Scopigno N, Vigliotti M, Goglia F, Cioffi F, Silvestri E. Mitochondria at the Crossroads: Linking the Mediterranean Diet to Metabolic Health and Non-Pharmacological Approaches to NAFLD. Nutrients 2025; 17:1214. [PMID: 40218971 PMCID: PMC11990101 DOI: 10.3390/nu17071214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing global health concern that is closely linked to metabolic syndrome, yet no approved pharmacological treatment exists. The Mediterranean diet (MD) emerged as a first-line dietary intervention for NAFLD, offering metabolic and hepatoprotective benefits. Now conceptualized as a complex chemical matrix rich in bioactive compounds, the MD exerts antioxidant and anti-inflammatory effects, improving insulin sensitivity and lipid metabolism. Mitochondria play a central role in NAFLD pathophysiology, influencing energy metabolism, oxidative stress, and lipid homeostasis. Emerging evidence suggests that the MD's bioactive compounds enhance mitochondrial function by modulating oxidative phosphorylation, biogenesis, and mitophagy. However, most research has focused on individual compounds rather than the MD as a whole, leaving gaps in understanding its collective impact as a complex dietary pattern. This narrative review explores how the MD and its bioactive compounds influence mitochondrial health in NAFLD, highlighting key pathways such as mitochondrial substrate control, dynamics, and energy efficiency. A literature search was conducted to identify relevant studies on the MD, mitochondria, and NAFLD. While the search was promising, our understanding remains incomplete, particularly when current knowledge is limited by the lack of mechanistic and comprehensive studies on the MD's holistic impact. Future research integrating cutting-edge experimental approaches is needed to elucidate the intricate diet-mitochondria interactions. A deeper understanding of how the MD influences mitochondrial health in NAFLD is essential for developing precision-targeted nutritional strategies that can effectively prevent and manage the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Elena Silvestri
- Department of Science and Technology, University of Sannio, Via De Sanctis, 82100 Benevento, Italy; (G.M.); (A.G.); (N.S.); (M.V.); (F.G.); (F.C.)
| |
Collapse
|
2
|
Heinitz S, Traurig M, Krakoff J, Rabe P, Stäubert C, Kobes S, Hanson RL, Stumvoll M, Blüher M, Bogardus C, Baier L, Piaggi P. An E115A Missense Variant in CERS2 Is Associated With Increased Sleeping Energy Expenditure and Hepatic Insulin Resistance in American Indians. Diabetes 2024; 73:1361-1371. [PMID: 38776413 PMCID: PMC11262042 DOI: 10.2337/db23-0690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Genetic determinants of interindividual differences in energy expenditure (EE) are largely unknown. Sphingolipids, such as ceramides, have been implicated in the regulation of human EE via mitochondrial uncoupling. In this study, we investigated whether genetic variants within enzymes involved in sphingolipid synthesis and degradation affect EE and insulin-related traits in a cohort of American Indians informative for 24-h EE and glucose disposal rates during a hyperinsulinemic-euglycemic clamp. Association analysis of 10,084 genetic variants within 28 genes involved in sphingolipid pathways identified a missense variant (rs267738, A>C, E115A) in exon 4 of CERS2 that was associated with higher sleeping EE (116 kcal/day) and increased rates of endogenous glucose production during basal (5%) and insulin-stimulated (43%) conditions, both indicators of hepatic insulin resistance. The rs267738 variant did not affect ceramide synthesis in HepG2 cells but resulted in a 30% decrease in basal mitochondrial respiration. In conclusion, we provide evidence that the CERS2 rs267738 missense variant may influence hepatic glucose production and postabsorptive sleeping metabolic rate. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sascha Heinitz
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
- Department of Internal Medicine, Clinic for Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Philipp-Rosenthal-Strasse 27, Leipzig, Germany
| | - Michael Traurig
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Philipp Rabe
- Faculty of Medicine, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Claudia Stäubert
- Faculty of Medicine, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Robert L. Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Michael Stumvoll
- Department of Internal Medicine, Clinic for Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Philipp-Rosenthal-Strasse 27, Leipzig, Germany
| | - Matthias Blüher
- Department of Internal Medicine, Clinic for Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Philipp-Rosenthal-Strasse 27, Leipzig, Germany
| | - Clifton Bogardus
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Leslie Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
- Department of Information Engineering, University of Pisa, Pisa, Italy
| |
Collapse
|
3
|
Tian J, Fan J, Zhang T. Mitochondria as a target for exercise-mitigated type 2 diabetes. J Mol Histol 2023; 54:543-557. [PMID: 37874501 DOI: 10.1007/s10735-023-10158-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 09/17/2023] [Indexed: 10/25/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is one of most common metabolic diseases and continues to be a leading cause of death worldwide. Although great efforts have been made to elucidate the pathogenesis of diabetes, the underlying mechanism still remains unclear. Notably, overwhelming evidence has demonstrated that mitochondria are tightly correlated with the development of T2DM, and the defects of mitochondrial function in peripheral insulin-responsive tissues, such as skeletal muscle, liver and adipose tissue, are crucial drivers of T2DM. Furthermore, exercise training is considered as an effective stimulus for improving insulin sensitivity and hence is regarded as the best strategy to prevent and treat T2DM. Although the precise mechanisms by which exercise alleviates T2DM are not fully understood, mitochondria may be critical for the beneficial effects of exercise.
Collapse
Affiliation(s)
- Jingjing Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Jingcheng Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Tan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China.
| |
Collapse
|
4
|
Liu K, Liu S, Wu C, Wang Y, Zhang Y, Yu J, Liu S, Li X, Qi X, Su S, Qi X, Zhou L, Li Y. Rhynchophylline relieves nonalcoholic fatty liver disease by activating lipase and increasing energy metabolism. Int Immunopharmacol 2023; 117:109948. [PMID: 37012893 DOI: 10.1016/j.intimp.2023.109948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 03/13/2023]
Abstract
Hepatic fat metabolism may be altered in the context of overnutrition and obesity, often resulting in the accumulation of triglycerides in hepatocytes and leading to nonalcoholic fatty liver disease (NAFLD). Natural plant alkaloids have demonstrated great potential for the prevention and treatment of NAFLD. However, the role of rhynchophylline (RHY) in lipid metabolism is not clear. We explored the role of RHY in lipid metabolism in cells treated with oleic and palmitic acids to mimic high-fat diet (HFD) conditions. RHY attenuated oleic and palmitic acid-induced increases in triglyceride accumulation in HepG2, AML12, and LMH cells. RHY also increased energy metabolism and reduced oxidative stress. We further investigated the effect of RHY on hepatic lipid metabolism in mice fed an HFD including 40 mg/kg RHY. RHY alleviated hepatic steatosis, reduced fat deposition, promoted energy metabolism, and improved glucose metabolism. We investigated the mechanism responsible for this activity by docking with key proteins of lipid metabolism disorders using Discovery Studio software, which showed that RHY interacted well with lipases. Finally, we found that adding RHY promoted lipase activity and lipolysis. In conclusion, RHY ameliorated HFD-induced NAFLD and its complications by increasing lipase activity.
Collapse
Affiliation(s)
- Kang Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Songsong Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Chou Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Yuwei Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Yurou Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Jingsu Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Siqi Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Xiangling Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Xinyu Qi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Songtao Su
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Xinyi Qi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Lei Zhou
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Yixing Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, PR China.
| |
Collapse
|
5
|
Recent Overview of Potent Antioxidant Activity of Coordination Compounds. Antioxidants (Basel) 2023; 12:antiox12020213. [PMID: 36829772 PMCID: PMC9952845 DOI: 10.3390/antiox12020213] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
During recent decades, the complexation of organic ligands toward several metal ions of s-p and d-block has been applied as a plan to enhance its antioxidant performance. Due to their wide range of beneficial impacts, coordination compounds are widely used in industries, specifically in the medicinal and pharmaceutical fields. The activity is generally improved by chelation consequently knowing that the characteristics of both ligands and metals can lead to the development of greatly active compounds. Chelation compounds are a substitute for using the traditional synthetic antioxidants, because metal chelates present benefits, including a variety in geometry, oxidation states, and coordination number, that assist and favor the redox methods associated with antioxidant action. As well as understanding the best studied anti-oxidative assets of these compounds, coordination compounds are involved in the free radical scavenging process and protecting human organisms from the opposing effects of these radicals. The antioxidant ability can be assessed by various interrelated systems. The methodological modification offers the most knowledge on the antioxidant property of metal chelates. Colorimetric techniques are the most used, though electron paramagnetic resonance (EPR) is an alternative for metallic compounds, since color does not affect the results. Information about systems, with their benefits, and restrictions, permits a dependable valuation of the antioxidant performance of coordination compounds, as well as assisting application in various states wherever antioxidant drugs are required, such as in food protection, appropriate good-packaged foods, dietary supplements, and others. Because of the new exhaustive analysis of organic ligands, it has become a separate field of research in chemistry. The present investigation will be respected for providing a foundation for the antioxidant properties of organic ligands, future tests on organic ligands, and building high-quality antioxidative compounds.
Collapse
|
6
|
Berberine mitigates hepatic insulin resistance by enhancing mitochondrial architecture via the SIRT1/Opa1 signalling pathway. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1464-1475. [PMID: 36269134 PMCID: PMC9827808 DOI: 10.3724/abbs.2022146] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The aberrant changes of fussion/fission-related proteins can trigger mitochondrial dynamics imbalance, which cause mitochondrial dysfunctions and result insulin resistance (IR). However, the relationship between the inner mitochondrial membrane fusion protein optic atrophy 1 (Opa1) and hepatic IR as well as the specific molecular mechanisms of signal transduction has not been fully elucidated. In this study, we explore whether abnormalities in the Opa1 cause hepatic IR and whether berberine (BBR) can prevent hepatic IR through the SIRT1/Opa1 signalling pathway. High-fat diet (HFD)-fed mice and db/db mice are used as animal models to study hepatic IR in vivo. IR, morphological changes, and mitochondrial injury of the liver are examined to explore the effects of BBR. SIRT1/Opa1 protein expression is determined to confirm whether the signalling pathway is damaged in the model animals and is involved in BBR treatment-mediated mitigation of hepatic IR. A palmitate (PA)-induced hepatocyte IR model is established in HepG2 cells in vitro. Opa1 silencing and SIRT1 overexpression are induced to verify whether Opa1 deficiency causes hepatocyte IR and whether SIRT1 improves this dysfunction. BBR treatment and SIRT1 silencing are employed to confirm that BBR can prevent hepatic IR by activating the SIRT1/Opa1 signalling pathway. Western blot analysis and JC-1 fluorescent staining results show that Opa1 deficiency causes an imbalance in mitochondrial fusion/fission and impairs insulin signalling in HepG2 cells. SIRT1 and BBR overexpression ameliorates PA-induced IR, increases Opa1, and improves mitochondrial function. SIRT1 silencing partly reverses the effects of BBR on HepG2 cells. SIRT1 and Opa1 expressions are downregulated in the animal models. BBR attenuates hepatic IR and enhances SIRT1/Opa1 signalling in db/db mice. In summary, Opa1 silencing-mediated mitochondrial fusion/fission imbalance could lead to hepatocyte IR. BBR may improve hepatic IR by regulating the SIRT1/Opa1 signalling pathway, and thus, it may be used to treat type-2 diabetes.
Collapse
|
7
|
Time-of-Day Circadian Modulation of Grape-Seed Procyanidin Extract (GSPE) in Hepatic Mitochondrial Dynamics in Cafeteria-Diet-Induced Obese Rats. Nutrients 2022; 14:nu14040774. [PMID: 35215423 PMCID: PMC8876123 DOI: 10.3390/nu14040774] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Major susceptibility to alterations in liver function (e.g., hepatic steatosis) in a prone environment due to circadian misalignments represents a common consequence of recent sociobiological behavior (i.e., food excess and sleep deprivation). Natural compounds and, more concisely, polyphenols have been shown as an interesting tool for fighting against metabolic syndrome and related consequences. Furthermore, mitochondria have been identified as an important target for mediation of the health effects of these compounds. Additionally, mitochondrial function and dynamics are strongly regulated in a circadian way. Thus, we wondered whether some of the beneficial effects of grape-seed procyanidin extract (GSPE) on metabolic syndrome could be mediated by a circadian modulation of mitochondrial homeostasis. For this purpose, rats were subjected to “standard”, “cafeteria” and “cafeteria diet + GSPE” treatments (n = 4/group) for 9 weeks (the last 4 weeks, GSPE/vehicle) of treatment, administering the extract/vehicle at diurnal or nocturnal times (ZT0 or ZT12). For circadian assessment, one hour after turning the light on (ZT1), animals were sacrificed every 6 h (ZT1, ZT7, ZT13 and ZT19). Interestingly, GSPE was able to restore the rhythm on clock hepatic genes (Bmal1, Per2, Cry1, Rorα), as this correction was more evident in nocturnal treatment. Additionally, during nocturnal treatment, an increase in hepatic fusion genes and a decrease in fission genes were observed. Regarding mitochondrial complex activity, there was a strong effect of cafeteria diet at nearly all ZTs, and GSPE was able to restore activity at discrete ZTs, mainly in the diurnal treatment (ZT0). Furthermore, a differential behavior was observed in tricarboxylic acid (TCA) metabolites between GSPE diurnal and nocturnal administration times. Therefore, GSPE may serve as a nutritional preventive strategy in the recovery of hepatic-related metabolic disease by modulating mitochondrial dynamics, which is concomitant to the restoration of the hepatic circadian machinery.
Collapse
|
8
|
Lee H, Ha TY, Jung CH, Nirmala FS, Park SY, Huh YH, Ahn J. Mitochondrial dysfunction in skeletal muscle contributes to the development of acute insulin resistance in mice. J Cachexia Sarcopenia Muscle 2021; 12:1925-1939. [PMID: 34605225 PMCID: PMC8718067 DOI: 10.1002/jcsm.12794] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/12/2021] [Accepted: 08/23/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Although mounting evidence indicates that insulin resistance (IR) co-occurs with mitochondrial dysfunction in skeletal muscle, there is no clear causal link between mitochondrial dysfunction and IR pathogenesis. In this study, the exact role of mitochondria in IR development was determined. METHODS Six-week-old C57BL/6 mice were fed a high-fat diet for 2 weeks to induce acute IR or for 24 weeks to induce chronic IR (n = 8 per group). To characterize mitochondrial function, we measured citrate synthase activity, ATP content, mitochondrial DNA (mtDNA) content, and oxygen consumption rate in gastrocnemius and liver tissues. We intraperitoneally administered mitochondrial division inhibitor 1 (mdivi-1) to mice with acute IR and measured mitochondrial adaptive responses such as mitophagy, mitochondrial unfolded protein response (UPRmt), and oxidative stress (n = 6 per group). RESULTS Acute IR occurred coincidently with impaired mitochondrial function, including reduced citrate synthase activity (-37.8%, P < 0.01), ATP production (-88.0%, P < 0.01), mtDNA (-53.1%, P < 0.01), and mitochondrial respiration (-52.2% for maximal respiration, P < 0.05) in skeletal muscle but not in liver. Administration of mdivi-1 attenuated IR development by increasing mitochondrial function (+58.5% for mtDNA content, P < 0.01; 4.06 ± 0.69 to 5.84 ± 0.95 pmol/min/mg for citrate synthase activity, P < 0.05; 13.06 ± 0.70 to 34.87 ± 0.70 pmol/min/g for maximal respiration, P < 0.001). Western blot analysis showed acute IR resulted in increased autophagy (mitophagy) and UPRmt induction in muscle tissue. This adaptive response was inhibited by mdivi-1, which reduced the mitochondrial oxidative stress of skeletal muscle during acute IR. CONCLUSIONS Acute IR induced mitochondrial oxidative stress that impaired mitochondrial function in skeletal muscle. Improving mitochondrial function has important potential for treating acute IR.
Collapse
Affiliation(s)
- Hyunjung Lee
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea
| | - Tae Youl Ha
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Chang Hwa Jung
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Farida Sukma Nirmala
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Yang Hoon Huh
- Center for Electron Microscopy Research, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Jiyun Ahn
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Nunes S, Viana SD, Preguiça I, Alves A, Fernandes R, Teodoro JS, Matos P, Figueirinha A, Salgueiro L, André A, Silva S, Jarak I, Carvalho RA, Cavadas C, Rolo AP, Palmeira CM, Pintado MM, Reis F. Blueberry Counteracts Prediabetes in a Hypercaloric Diet-Induced Rat Model and Rescues Hepatic Mitochondrial Bioenergetics. Nutrients 2021; 13:4192. [PMID: 34959746 PMCID: PMC8706913 DOI: 10.3390/nu13124192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/11/2022] Open
Abstract
The paramount importance of a healthy diet in the prevention of type 2 diabetes is now well recognized. Blueberries (BBs) have been described as attractive functional fruits for this purpose. This study aimed to elucidate the cellular and molecular mechanisms pertaining to the protective impact of blueberry juice (BJ) on prediabetes. Using a hypercaloric diet-induced prediabetic rat model, we evaluated the effects of BJ on glucose, insulin, and lipid profiles; gut microbiota composition; intestinal barrier integrity; and metabolic endotoxemia, as well as on hepatic metabolic surrogates, including several related to mitochondria bioenergetics. BJ supplementation for 14 weeks counteracted diet-evoked metabolic deregulation, improving glucose tolerance, insulin sensitivity, and hypertriglyceridemia, along with systemic and hepatic antioxidant properties, without a significant impact on the gut microbiota composition and related mechanisms. In addition, BJ treatment effectively alleviated hepatic steatosis and mitochondrial dysfunction observed in the prediabetic animals, as suggested by the amelioration of bioenergetics parameters and key targets of inflammation, insulin signaling, ketogenesis, and fatty acids oxidation. In conclusion, the beneficial metabolic impact of BJ in prediabetes may be mainly explained by the rescue of hepatic mitochondrial bioenergetics. These findings pave the way to support the use of BJ in prediabetes to prevent diabetes and its complications.
Collapse
Affiliation(s)
- Sara Nunes
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Sofia D. Viana
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Polytechnic Institute of Coimbra, ESTESC-Coimbra Health School, Pharmacy/Biomedical Laboratory Sciences, 3046-854 Coimbra, Portugal;
| | - Inês Preguiça
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - André Alves
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Rosa Fernandes
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - João S. Teodoro
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Patrícia Matos
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (A.F.); (L.S.)
- LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, 3000-456 Coimbra, Portugal
- CIEPQPF, Chemical Process Engineering and Forest Products Research Centre Research Center, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (A.F.); (L.S.)
- LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (A.F.); (L.S.)
- CIEPQPF, Chemical Process Engineering and Forest Products Research Centre Research Center, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Alexandra André
- Polytechnic Institute of Coimbra, ESTESC-Coimbra Health School, Pharmacy/Biomedical Laboratory Sciences, 3046-854 Coimbra, Portugal;
| | - Sara Silva
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (S.S.); (M.M.P.)
| | - Ivana Jarak
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal;
| | - Rui A. Carvalho
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Associated Laboratory for Green Chemistry-Clean Technologies and Processes, REQUIMTE, Faculty of Sciences and Technology, University of Porto, 4050-313 Porto, Portugal
| | - Cláudia Cavadas
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (A.F.); (L.S.)
| | - Anabela P. Rolo
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carlos M. Palmeira
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Maria M. Pintado
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (S.S.); (M.M.P.)
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| |
Collapse
|
10
|
Experimental models of lipid overload and their relevance in understanding skeletal muscle insulin resistance and pathological changes in mitochondrial oxidative capacity. Biochimie 2021; 196:182-193. [PMID: 34563603 DOI: 10.1016/j.biochi.2021.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
It remains essential to decipher some of the pathological mechanisms that link obesity with deteriorating human health. Insulin resistance, due to enhanced free fatty acid substrate delivery, results in disrupted glucose homeostasis and altered mitochondrial oxidative capacity, which is a characteristic feature of an obese state. In fact, as a major site for regulating glucose homeostasis and energy production in response to insulin, the skeletal muscle has become an interesting target tissue to understand the impact of lipid overload on the development of insulin resistance and impaired mitochondrial respiratory function. In addition to systematically retrieving the discussed data, the current review brings an essential perspective in understanding the relevance of experimental models of lipid overload such as high fat diets in understanding the pathological link between insulin resistance and pathological changes in mitochondrial oxidative capacity. Importantly, inclusion of evidence from transgenic model highlights some of the unique molecular targets that are implicated in the development of insulin resistance and inefficient mitochondrial respiration processes within an obese state. Importantly, saturation with lipid products such as ceramides and diacylglycerols, especially within the skeletal muscle, appears to be instrumental in paving the path leading to worsening of metabolic complications. These metabolic consequences mostly interfere with the efficiency of the mitochondrial electron transport chain, leading to overproduction of toxic reactive oxygen species. Therefore, therapeutic agents that reverse the effects of lipid overload by improving insulin sensitivity and mitochondrial oxidative capacity are crucial for the management or even treatment of metabolic diseases.
Collapse
|
11
|
Mucha P, Skoczyńska A, Małecka M, Hikisz P, Budzisz E. Overview of the Antioxidant and Anti-Inflammatory Activities of Selected Plant Compounds and Their Metal Ions Complexes. Molecules 2021; 26:4886. [PMID: 34443474 PMCID: PMC8398118 DOI: 10.3390/molecules26164886] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous plant compounds and their metal-ion complexes exert antioxidative, anti-inflammatory, anticancer, and other beneficial effects. This review highlights the different bioactivities of flavonoids, chromones, and coumarins and their metal-ions complexes due to different structural characteristics. In addition to insight into the most studied antioxidative properties of these compounds, the first part of the review provides a comprehensive overview of exogenous and endogenous sources of reactive oxygen and nitrogen species, oxidative stress-mediated damages of lipids and proteins, and on protective roles of antioxidant defense systems, including plant-derived antioxidants. Additionally, the review covers the anti-inflammatory and antimicrobial activities of flavonoids, chromones, coumarins and their metal-ion complexes which support its application in medicine, pharmacy, and cosmetology.
Collapse
Affiliation(s)
- Paulina Mucha
- Department of the Chemistry of Cosmetic Raw Materials, Faculty of Pharmacy, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland
| | - Anna Skoczyńska
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Poniatowskiego 15, 41-200 Sosnowiec, Poland;
| | - Magdalena Małecka
- Department of Physical Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163/165, 90-236 Łódź, Poland;
| | - Paweł Hikisz
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Łódź, Poland;
| | - Elzbieta Budzisz
- Department of the Chemistry of Cosmetic Raw Materials, Faculty of Pharmacy, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland
| |
Collapse
|
12
|
Mazzoli A, Gatto C, Crescenzo R, Cigliano L, Iossa S. Prolonged Changes in Hepatic Mitochondrial Activity and Insulin Sensitivity by High Fructose Intake in Adolescent Rats. Nutrients 2021; 13:nu13041370. [PMID: 33921866 PMCID: PMC8073121 DOI: 10.3390/nu13041370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/16/2022] Open
Abstract
Persistence of damage induced by unhealthy diets during youth has been little addressed. Therefore, we investigated the impact of a short-term fructose-rich diet on liver metabolic activity in adolescent rats and the putative persistence of alterations after removing fructose from the diet. Adolescent rats were fed a fructose-rich diet for three weeks and then switched to a control diet for further three weeks. Body composition and energy balance were not affected by fructose-rich diet, while increased body lipids and lipid gain were found after the rescue period. Switching to a control diet reversed the upregulation of plasma fructose, uric acid, lipocalin, and haptoglobin, while plasma triglycerides, alanine aminotransferase, lipopolysaccharide, and tumor necrosis factor alpha remained higher. Hepatic steatosis and ceramide were increased by fructose-rich diet, but reversed by returning to a control diet, while altered hepatic response to insulin persisted. Liver fatty acid synthase and stearoyl-CoA desaturase (SCD) activities were upregulated by fructose-rich diet, and SCD activity remained higher after returning to the control diet. Fructose-induced upregulation of complex II-driven mitochondrial respiration, peroxisome proliferator-activated receptor-gamma coactivator 1 alpha, and peroxisome proliferator activated receptor α also persisted after switching to control diet. In conclusion, our results show prolonged fructose-induced dysregulation of liver metabolic activity.
Collapse
|
13
|
Rodrigues RR, Gurung M, Li Z, García-Jaramillo M, Greer R, Gaulke C, Bauchinger F, You H, Pederson JW, Vasquez-Perez S, White KD, Frink B, Philmus B, Jump DB, Trinchieri G, Berry D, Sharpton TJ, Dzutsev A, Morgun A, Shulzhenko N. Transkingdom interactions between Lactobacilli and hepatic mitochondria attenuate western diet-induced diabetes. Nat Commun 2021; 12:101. [PMID: 33397942 PMCID: PMC7782853 DOI: 10.1038/s41467-020-20313-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Western diet (WD) is one of the major culprits of metabolic disease including type 2 diabetes (T2D) with gut microbiota playing an important role in modulating effects of the diet. Herein, we use a data-driven approach (Transkingdom Network analysis) to model host-microbiome interactions under WD to infer which members of microbiota contribute to the altered host metabolism. Interrogation of this network pointed to taxa with potential beneficial or harmful effects on host's metabolism. We then validate the functional role of the predicted bacteria in regulating metabolism and show that they act via different host pathways. Our gene expression and electron microscopy studies show that two species from Lactobacillus genus act upon mitochondria in the liver leading to the improvement of lipid metabolism. Metabolomics analyses revealed that reduced glutathione may mediate these effects. Our study identifies potential probiotic strains for T2D and provides important insights into mechanisms of their action.
Collapse
Affiliation(s)
| | - Manoj Gurung
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Zhipeng Li
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | | | - Renee Greer
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | | | - Franziska Bauchinger
- Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
| | - Hyekyoung You
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Jacob W Pederson
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | | | - Kimberly D White
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Briana Frink
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Benjamin Philmus
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Donald B Jump
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David Berry
- Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
| | | | - Amiran Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, USA.
| | | |
Collapse
|
14
|
Park S, Zhang T, Wu X, Qiu JY. A mixture of mulberry and silk amino acids protected against D-galactosamine induced acute liver damage by attenuating oxidative stress and inflammation in HepG2 cells and rats. Exp Ther Med 2020; 19:3611-3619. [PMID: 32346425 PMCID: PMC7185172 DOI: 10.3892/etm.2020.8636] [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: 01/31/2019] [Accepted: 10/23/2019] [Indexed: 12/23/2022] Open
Abstract
The liver is an important organ for the removal of toxins and utilization of nutrients. The present study then investigated whether a mixture of mulberry water extracts and silk amino acids protected against acute liver damage in rats induced by intraperitoneal injection of D-galactosamine and the action mechanism. D-galactosamine injection is widely used to develop experimental animal models of acute hepatic disease. In the present study, male Sprague-Dawley rats received intraperitoneal injection of D-galactosamine followed by 200 and 600 mg/kg body weight (BW) of mulberry extracts and silk amino acids (1:3, w/w; MS1:3-L and MS1:3-H), the same amounts of MS with different ratios (1:5, w/w; MS1:5-L and MS1:5-H), and 600 mg/kg bw cellulose (control) for 1 week. The normal-control group received an injection of saline instead of D-galactosamine with the same diet as the control group. D-galactosamine injection (control rats) increased serum ALT, AST and γ-GPT levels, indicating the induction of acute liver damage. The control rats also exhibited reduced glycogen depositions, which contributed to increasing fat synthesis from glucose and elevated serum triglyceride levels. Oxidative stress and inflammation in the liver of the control increased in response to the decreasing antioxidant activity and mRNA expression and increasing TNF-α expression, respectively. Both MS1:3 and MS1:5 reduced serum ALT, AST and γ-GPT levels to ameliorate liver damage. MS1:3 reduced oxidative stress by increasing the activity and expression of antioxidant enzymes, whereas MS1:5 decreased the expression TNF-α in the liver. MS1:3 and MS1:5 improved the necrosis of hepatocytes in H&E staining, which was associated with increased glycogen deposition in PAS staining. MS1:5 had better effects on glycogen accumulation. In conclusion, MS1:3 and MS1:5 can be used as therapeutic agents for acute liver damage.
Collapse
Affiliation(s)
- Sunmin Park
- Department of Food and Nutrition, Obesity/Diabetes Center, Hoseo University, Asan, Chungcheong 336-795, Republic of Korea
| | - Ting Zhang
- Department of Food and Nutrition, Obesity/Diabetes Center, Hoseo University, Asan, Chungcheong 336-795, Republic of Korea
| | - Xuangao Wu
- Department of Food and Nutrition, Obesity/Diabetes Center, Hoseo University, Asan, Chungcheong 336-795, Republic of Korea
| | - Jing Yi Qiu
- Department of Food and Nutrition, Obesity/Diabetes Center, Hoseo University, Asan, Chungcheong 336-795, Republic of Korea
| |
Collapse
|
15
|
Mazzoli A, Crescenzo R, Cigliano L, Spagnuolo MS, Cancelliere R, Gatto C, Iossa S. Early Hepatic Oxidative Stress and Mitochondrial Changes Following Western Diet in Middle Aged Rats. Nutrients 2019; 11:nu11112670. [PMID: 31694213 PMCID: PMC6893784 DOI: 10.3390/nu11112670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 01/06/2023] Open
Abstract
To assess the effect of 4 weeks of high fat-high fructose feeding on whole body composition, energy balance, specific markers of oxidative stress and inflammation, and insulin sensitivity in the liver of middle-aged rats, rats (1 year) were fed a diet rich in saturated fatty acids and fructose (HFF rats), mimicking the “Western diet”, and compared with rats of the same age that were fed a low fat diet (LF rats). HFF rats exhibited a significant increase in the gain of body weight, energy, and lipids compared to LF rats. HFF rats also showed hepatic insulin resistance, together with an increase in plasma triglycerides, cholesterol, and tumor necrosis factor alpha. Hepatic lipids, triglycerides and cholesterol were higher in HFF rats, while a significant decrease in Stearoyl-CoA desaturase activity was found in this tissue. A marked increase in the protein amount of complex I, concomitant to a decrease in its contribution to mitochondrial respiration, was found in HFF rats. Lipid peroxidation and Nitro-Tyrosine content, taken as markers of oxidative stress, as well as NADPH oxidase activity, were significantly higher in HFF rats, while the antioxidant enzyme catalase decreased in these rats. Myeloperoxidase activity and lipocalin content increased, while peroxisome proliferator activated receptor gamma decreased in HFF rats. The present results provide evidence that middle-aged rats show susceptibility to a short-term “Western diet”, exhibiting altered redox homeostasis, insulin resistance, and early mitochondrial alterations in the liver. Therefore, this type of dietary habits should be drastically limited to pursue a “healthy aging”.
Collapse
Affiliation(s)
- Arianna Mazzoli
- Department of Biology, Federico II University, Via Cintia,80126 Naples, Italy; (A.M.); (R.C.); (L.C.); (R.C.); (C.G.)
| | - Raffaella Crescenzo
- Department of Biology, Federico II University, Via Cintia,80126 Naples, Italy; (A.M.); (R.C.); (L.C.); (R.C.); (C.G.)
| | - Luisa Cigliano
- Department of Biology, Federico II University, Via Cintia,80126 Naples, Italy; (A.M.); (R.C.); (L.C.); (R.C.); (C.G.)
| | - Maria Stefania Spagnuolo
- Department of Bio-Agrofood Science, Institute for the Animal Production System in Mediterranean Environment, National Research Council Naples (CNR-ISPAAM), 80147 Naples, Italy;
| | - Rosa Cancelliere
- Department of Biology, Federico II University, Via Cintia,80126 Naples, Italy; (A.M.); (R.C.); (L.C.); (R.C.); (C.G.)
| | - Cristina Gatto
- Department of Biology, Federico II University, Via Cintia,80126 Naples, Italy; (A.M.); (R.C.); (L.C.); (R.C.); (C.G.)
| | - Susanna Iossa
- Department of Biology, Federico II University, Via Cintia,80126 Naples, Italy; (A.M.); (R.C.); (L.C.); (R.C.); (C.G.)
- Correspondence: ; Tel.: +39-081-2538111
| |
Collapse
|
16
|
Guo Q, Li F, Duan Y, Wen C, Wang W, Zhang L, Huang R, Yin Y. Oxidative stress, nutritional antioxidants and beyond. SCIENCE CHINA-LIFE SCIENCES 2019; 63:866-874. [PMID: 31705360 DOI: 10.1007/s11427-019-9591-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022]
Abstract
Free radical-induced oxidative stress contributes to the development of metabolic syndromes (Mets), including overweight, hyperglycemia, insulin resistance and pro-inflammatory state. Most free radicals are generated from the mitochondrial electron transport chain; under physiological conditions, their levels are maintained by efficient antioxidant systems. A variety of transcription factors have been identified and characterized that control gene expression in response to oxidative stress status. Natural antioxidant compounds have been largely studied for their strong antioxidant capacities. This review discusses the recent progress in oxidative stress and mitochondrial dysfunction in Mets and highlights the anti-Mets, anti-oxidative, and anti-inflammatory effect of polyphenols as potential nutritional therapy.
Collapse
Affiliation(s)
- Qiuping Guo
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Changsha, 410125, China.,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, 410125, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Fengna Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China. .,Key Laboratory of Agro-ecological Processes in Subtropical Region, Changsha, 410125, China. .,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China. .,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, 410125, China. .,Hunan Co-Innovation Center of Animal Production Safety, Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, 410128, China.
| | - Yehui Duan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Chaoyue Wen
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, 410018, China
| | - Wenlong Wang
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, 410018, China
| | - Lingyu Zhang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Changsha, 410125, China.,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, 410125, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Ruilin Huang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Changsha, 410125, China.,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, 410125, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China. .,Key Laboratory of Agro-ecological Processes in Subtropical Region, Changsha, 410125, China. .,Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China. .,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, 410125, China. .,Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, 410018, China.
| |
Collapse
|
17
|
Su Z, Nie Y, Huang X, Zhu Y, Feng B, Tang L, Zheng G. Mitophagy in Hepatic Insulin Resistance: Therapeutic Potential and Concerns. Front Pharmacol 2019; 10:1193. [PMID: 31649547 PMCID: PMC6795753 DOI: 10.3389/fphar.2019.01193] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/17/2019] [Indexed: 12/23/2022] Open
Abstract
Metabolic syndrome, characterized by central obesity, hypertension, and hyperlipidemia, increases the morbidity and mortality of cardiovascular disease, type 2 diabetes, nonalcoholic fatty liver disease, and other metabolic diseases. It is well known that insulin resistance, especially hepatic insulin resistance, is a risk factor for metabolic syndrome. Current research has shown that hepatic fatty acid accumulation can cause hepatic insulin resistance through increased gluconeogenesis, lipogenesis, chronic inflammation, oxidative stress and endoplasmic reticulum stress, and impaired insulin signal pathway. Mitochondria are the major sites of fatty acid β-oxidation, which is the major degradation mechanism of fatty acids. Mitochondrial dysfunction has been shown to be involved in the development of hepatic fatty acid–induced hepatic insulin resistance. Mitochondrial autophagy (mitophagy), a catabolic process, selectively degrades damaged mitochondria to reverse mitochondrial dysfunction and preserve mitochondrial dynamics and function. Therefore, mitophagy can promote mitochondrial fatty acid oxidation to inhibit hepatic fatty acid accumulation and improve hepatic insulin resistance. Here, we review advances in our understanding of the relationship between mitophagy and hepatic insulin resistance. Additionally, we also highlight the potential value of mitophagy in the treatment of hepatic insulin resistance and metabolic syndrome.
Collapse
Affiliation(s)
- Zuqing Su
- Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yutong Nie
- Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiufang Huang
- Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Zhu
- Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bing Feng
- Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lipeng Tang
- Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangjuan Zheng
- Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
Adami GF, Carbone F, Montecucco F, Camerini G, Cordera R. Adipose Tissue Composition in Obesity and After Bariatric Surgery. Obes Surg 2019; 29:3030-3038. [PMID: 31190263 DOI: 10.1007/s11695-019-04030-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The adipose tissue is a complex organ that regulates food intake and energy expenditure as well as induces low-grade inflammation. This review deals with changes in the composition and activity of the adipose organ after bariatric surgery, focusing on epicardial and ectopic fat and on relationships between white and brown adipose tissues. Postoperative improvements of ectopic fat and epicardial fat size and composition account for the metabolic recovery and the decreased cardiovascular risk. Following Roux-en-Y gastric bypass or biliopancreatic diversion, a proportional increase in the size and activity of the metabolically active brown adipose tissue was observed, most likely related to the postoperative rearrangement of the entero-hormonal pattern with an increase of GLP-1 production: this aspect would promote the postoperative weight loss and maintenance of post-surgery benefits.
Collapse
Affiliation(s)
- Gian Franco Adami
- Department of Internal Medicine, University of Genova, 8, viale Benedetto XV, 16132, Genova, Italy.
- IRCCS, Azienda Ospedale-Universitaria San Martino, Genoa, Italy.
| | - Federico Carbone
- Department of Internal Medicine, University of Genova, 8, viale Benedetto XV, 16132, Genova, Italy
- IRCCS, Azienda Ospedale-Universitaria San Martino, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genova, 8, viale Benedetto XV, 16132, Genova, Italy
- IRCCS, Azienda Ospedale-Universitaria San Martino, Genoa, Italy
| | - Giovanni Camerini
- Department of Internal Medicine, University of Genova, 8, viale Benedetto XV, 16132, Genova, Italy
- IRCCS, Azienda Ospedale-Universitaria San Martino, Genoa, Italy
- Department of Surgery, University of Genova, Genoa, Italy
| | - Renzo Cordera
- Department of Internal Medicine, University of Genova, 8, viale Benedetto XV, 16132, Genova, Italy
- IRCCS, Azienda Ospedale-Universitaria San Martino, Genoa, Italy
| |
Collapse
|
19
|
Abstract
The health-promoting effects of physical activity to prevent and treat metabolic disorders are numerous. However, the underlying molecular mechanisms are not yet completely deciphered. In recent years, studies have referred to the liver as an endocrine organ, since it releases specific proteins called hepatokines. Some of these hepatokines are involved in whole body metabolic homeostasis and are theorized to participate in the development of metabolic disease. In this regard, the present review describes the role of Fibroblast Growth Factor 21, Fetuin-A, Angiopoietin-like protein 4, and Follistatin in metabolic disease and their production in response to acute exercise. Also, we discuss the potential role of hepatokines in mediating the beneficial effects of regular exercise and the future challenges to the discovery of new exercise-induced hepatokines.
Collapse
Affiliation(s)
- Gaël Ennequin
- PEPITE EA4267, EPSI, Université de Bourgogne Franche-Comté , Besançon , France
| | - Pascal Sirvent
- Université Clermont Auvergne, Laboratoire des Adaptations Métaboliques à l'Exercice en conditions Physiologiques et Pathologiques (AME2P), CRNH Auvergne, Clermont-Ferrand , France
| | - Martin Whitham
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham , Birmingham , United Kingdom
| |
Collapse
|
20
|
Neeland IJ, Boone SC, Mook‐Kanamori DO, Ayers C, Smit RAJ, Tzoulaki I, Karaman I, Boulange C, Vaidya D, Punjabi N, Allison M, Herrington DM, Jukema JW, Rosendaal FR, Lamb HJ, van Dijk KW, Greenland P, de Mutsert R. Metabolomics Profiling of Visceral Adipose Tissue: Results From MESA and the NEO Study. J Am Heart Assoc 2019; 8:e010810. [PMID: 31017036 PMCID: PMC6512086 DOI: 10.1161/jaha.118.010810] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/19/2019] [Indexed: 12/22/2022]
Abstract
Background Identifying associations between serum metabolites and visceral adipose tissue ( VAT ) could provide novel biomarkers of VAT and insights into the pathogenesis of obesity-related diseases. We aimed to discover and replicate metabolites reflecting pathways related to VAT . Methods and Results Associations between fasting serum metabolites and VAT area (by computed tomography or magnetic resonance imaging) were assessed with cross-sectional linear regression of individual-level data from participants in MESA (Multi-Ethnic Study of Atherosclerosis; discovery, N=1103) and the NEO (Netherlands Epidemiology of Obesity) study (replication, N=2537). Untargeted 1H nuclear magnetic resonance metabolomics profiling of serum was performed in MESA, and metabolites were replicated in the NEO study using targeted 1H nuclear magnetic resonance spectroscopy. A total of 30 590 metabolomic spectral variables were evaluated. After adjustment for age, sex, race/ethnicity, socioeconomic status, smoking, physical activity, glucose/lipid-lowering medication, and body mass index, 2104 variables representing 24 nonlipid and 49 lipid/lipoprotein subclass metabolites remained significantly associated with VAT ( P=4.88×10-20-1.16×10-3). These included conventional metabolites, amino acids, acetylglycoproteins, intermediates of glucose and hepatic metabolism, organic acids, and subclasses of apolipoproteins, cholesterol, phospholipids, and triglycerides. Metabolites mapped to 31 biochemical pathways, including amino acid substrate use/metabolism and glycolysis/gluconeogenesis. In the replication cohort, acetylglycoproteins, branched-chain amino acids, lactate, glutamine (inversely), and atherogenic lipids remained associated with VAT ( P=1.90×10-35-8.46×10-7), with most associations remaining after additional adjustment for surrogates of VAT (glucose level, waist circumference, and serum triglycerides), reflecting novel independent associations. Conclusions We identified and replicated a metabolite panel associated with VAT in 2 community-based cohorts. These findings persisted after adjustment for body mass index and appear to define a metabolic signature of visceral adiposity.
Collapse
Affiliation(s)
- Ian J. Neeland
- Division of CardiologyDepartment of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTX
| | - Sebastiaan C. Boone
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Dennis O. Mook‐Kanamori
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
- Department of Public Health and Primary CareLeiden University Medical CenterLeidenthe Netherlands
| | - Colby Ayers
- Department of Clinical SciencesUniversity of Texas Southwestern Medical CenterDallasTX
| | - Roelof A. J. Smit
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Ioanna Tzoulaki
- Department of Epidemiology and BiostatisticsImperial College LondonLondonUnited Kingdom
| | - Ibrahim Karaman
- Department of Epidemiology and BiostatisticsImperial College LondonLondonUnited Kingdom
| | | | | | - Naresh Punjabi
- Department of MedicineJohns Hopkins UniversityBaltimoreMD
| | - Matthew Allison
- Department of Family Medicine and Public HealthUniversity of California San DiegoLa JollaCA
| | | | - J. Wouter Jukema
- Department of CardiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Frits R. Rosendaal
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Hildo J. Lamb
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Ko Willems van Dijk
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of Internal MedicineLeiden University Medical CenterLeidenthe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Philip Greenland
- Department of Preventive MedicineNorthwestern UniversityChicagoIL
| | - Renée de Mutsert
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
21
|
A mix of dietary fermentable fibers improves lipids handling by the liver of overfed minipigs. J Nutr Biochem 2018; 65:72-82. [PMID: 30654277 DOI: 10.1016/j.jnutbio.2018.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/17/2018] [Accepted: 12/05/2018] [Indexed: 02/06/2023]
Abstract
Obesity induced by overfeeding ultimately can lead to nonalcoholic fatty liver disease, whereas dietary fiber consumption is known to have a beneficial effect. We aimed to determine if a supplementation of a mix of fibers (inulin, resistant starch and pectin) could limit or alleviate overfeeding-induced metabolic perturbations. Twenty female minipigs were fed with a control diet (C) or an enriched fat/sucrose diet supplemented (O + F) or not (O) with fibers. Between 0 and 56 days of overfeeding, insulin (+88%), HOMA (+102%), cholesterol (+45%) and lactate (+63%) were increased, without any beneficial effect of fibers supplementation. However, fibers supplementation limited body weight gain (vs. O, -15% at D56) and the accumulation of hepatic lipids droplets induced by overfeeding. This could be explained by a decreased lipids transport potential (-50% FABP1 mRNA, O + F vs. O) inducing a down-regulation of regulatory elements of lipids metabolism / lipogenesis (-36% SREBP1c mRNA, O + F vs. O) but not to an increased oxidation (O + F not different from O and C for proteins and mRNA measured). Glucose metabolism was also differentially regulated by fibers supplementation, with an increased net hepatic release of glucose in the fasted state (diet × time effect, P<.05 at D56) that can be explained partially by a possible increased glycogen synthesis in the fed state (+82% GYS2 protein, O + F vs. O, P=.09). The direct role of short chain fatty acids on gluconeogenesis stimulation is questioned, with probably a short-term impact (D14) but no effect on a long-term (D56) basis.
Collapse
|
22
|
Zhang C, Seo J, Murakami K, Salem ESB, Bernhard E, Borra VJ, Choi K, Yuan CL, Chan CC, Chen X, Huang T, Weirauch MT, Divanovic S, Qi NR, Thomas HE, Mercer CA, Siomi H, Nakamura T. Hepatic Ago2-mediated RNA silencing controls energy metabolism linked to AMPK activation and obesity-associated pathophysiology. Nat Commun 2018; 9:3658. [PMID: 30201950 PMCID: PMC6131149 DOI: 10.1038/s41467-018-05870-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/26/2018] [Indexed: 12/21/2022] Open
Abstract
RNA silencing inhibits mRNA translation. While mRNA translation accounts for the majority of cellular energy expenditure, it is unclear if RNA silencing regulates energy homeostasis. Here, we report that hepatic Argonaute 2 (Ago2)-mediated RNA silencing regulates both intrinsic energy production and consumption and disturbs energy metabolism in the pathogenesis of obesity. Ago2 regulates expression of specific miRNAs including miR-802, miR-103/107, and miR-148a/152, causing metabolic disruption, while simultaneously suppressing the expression of genes regulating glucose and lipid metabolism, including Hnf1β, Cav1, and Ampka1. Liver-specific Ago2-deletion enhances mitochondrial oxidation and ATP consumption associated with mRNA translation, which results in AMPK activation, and improves obesity-associated pathophysiology. Notably, hepatic Ago2-deficiency improves glucose metabolism in conditions of insulin receptor antagonist treatment, high-fat diet challenge, and hepatic AMPKα1-deletion. The regulation of energy metabolism by Ago2 provides a novel paradigm in which RNA silencing plays an integral role in determining basal metabolic activity in obesity-associated sequelae.
Collapse
Affiliation(s)
- Cai Zhang
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Joonbae Seo
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kazutoshi Murakami
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Esam S B Salem
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elise Bernhard
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vishnupriya J Borra
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Celvie L Yuan
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Calvin C Chan
- Medical Scientist Training Program, Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Xiaoting Chen
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Taosheng Huang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nathan R Qi
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hala Einakat Thomas
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Carol A Mercer
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Haruhiko Siomi
- Department of Molecular Biology, Keio University School of Medicine, Tokyo, Japan
| | - Takahisa Nakamura
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
23
|
Kim Y, Natarajan SK, Chung S. Gamma-Tocotrienol Attenuates the Hepatic Inflammation and Fibrosis by Suppressing Endoplasmic Reticulum Stress in Mice. Mol Nutr Food Res 2018; 62:e1800519. [DOI: 10.1002/mnfr.201800519] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/26/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Yongeun Kim
- Department of Nutrition and Health Sciences; University of Nebraska-Lincoln; Lincoln NE 68583 USA
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences; University of Nebraska-Lincoln; Lincoln NE 68583 USA
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences; University of Nebraska-Lincoln; Lincoln NE 68583 USA
| |
Collapse
|
24
|
Small L, Brandon AE, Turner N, Cooney GJ. Modeling insulin resistance in rodents by alterations in diet: what have high-fat and high-calorie diets revealed? Am J Physiol Endocrinol Metab 2018; 314:E251-E265. [PMID: 29118016 DOI: 10.1152/ajpendo.00337.2017] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
For over half a century, researchers have been feeding different diets to rodents to examine the effects of macronutrients on whole body and tissue insulin action. During this period, the number of different diets and the source of macronutrients employed have grown dramatically. Because of the large heterogeneity in both the source and percentage of different macronutrients used for studies, it is not surprising that different high-calorie diets do not produce the same changes in insulin action. Despite this, diverse high-calorie diets continue to be employed in an attempt to generate a "generic" insulin resistance. The high-fat diet in particular varies greatly between studies with regard to the source, complexity, and ratio of dietary fat, carbohydrate, and protein. This review examines the range of rodent dietary models and methods for assessing insulin action. In almost all studies reviewed, rodents fed diets that had more than 45% of dietary energy as fat or simple carbohydrates had reduced whole body insulin action compared with chow. However, different high-calorie diets produced significantly different effects in liver, muscle, and whole body insulin action when insulin action was measured by the hyperinsulinemic-euglycemic clamp method. Rodent dietary models remain an important tool for exploring potential mechanisms of insulin resistance, but more attention needs to be given to the total macronutrient content and composition when interpreting dietary effects on insulin action.
Collapse
Affiliation(s)
- Lewin Small
- Diabetes and Metabolism Division, Garvan Institute , Sydney, New South Wales , Australia
| | - Amanda E Brandon
- Diabetes and Metabolism Division, Garvan Institute , Sydney, New South Wales , Australia
- Sydney Medical School, Charles Perkins Centre, The University of Sydney , New South Wales , Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Science, University of New South Wales , Sydney, New South Wales , Australia
| | - Gregory J Cooney
- Diabetes and Metabolism Division, Garvan Institute , Sydney, New South Wales , Australia
- Sydney Medical School, Charles Perkins Centre, The University of Sydney , New South Wales , Australia
| |
Collapse
|
25
|
Proinflammatory cytokine MIF plays a role in the pathogenesis of type-2 diabetes mellitus, but does not affect hepatic mitochondrial function. Cytokine 2017; 99:214-224. [PMID: 28780379 DOI: 10.1016/j.cyto.2017.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/08/2017] [Accepted: 07/13/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that plays an important role in the pathogenesis of type 2 diabetes mellitus (T2DM). Although the effect of high glucose on liver function has been described, the role of MIF in hepatic mitochondrial function during T2DM has not been studied. OBJECTIVE We examine the influence of MIF to hepatic mitochondrial function in T2DM mouse model. METHODS WT and Mif-/- BALB/c mice were treated with a single dose of streptozotocin (STZ). After an 8-week follow-up, serum glucose, proinflammatory cytokines, C-reactive protein (CRP), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzyme quantification, and liver histological analyses were performed. Liver mitochondria were extracted, and mitochondrial function was evaluated by oximetry, swelling and peroxide production. RESULTS Following treatment with STZ, WT mice (WT/STZ) developed significant hyperglycemia and high serum levels of MIF, tumor necrosis factor (TNF)-α, interleukin-β (IL-β), and CRP. Liver damage enzymes ALT and AST were found at high levels. In contrast, Mif-/-STZ lacked serum MIF levels and showed smaller increases in blood glucose, less TNF-α, IL-1β, CPR, ALT and AST, and failure to develop clinical signs of disease compared to the WT/STZ group. Mitochondria extracted from the Mif-/-STZ liver showed similar respiratory control (RC) to WT/STZ or healthy mice with glutamate/malate or succinate as substrates. The four respiratory chain complexes also had comparable activities. WT/STZ-isolated mitochondria showed low swelling with calcium compared to mitochondria from Mif-/-STZ or healthy mice. Peroxide production was comparable in all groups. CONCLUSION These results show although high systemic levels of MIF contribute to the development of T2DM pathology, the liver mitochondria remain unaltered. Importantly, the absence of MIF reduced the pathology of T2DM, also without altering liver mitochondrial function. These support MIF as a therapeutic target for the treatment of this disease in humans.
Collapse
|
26
|
García-Arroyo FE, Tapia E, Blas-Marron MG, Gonzaga G, Silverio O, Cristóbal M, Osorio H, Arellano-Buendía AS, Zazueta C, Aparicio-Trejo OE, Reyes-García JG, Pedraza-Chaverri J, Soto V, Roncal-Jiménez C, Johnson RJ, Sánchez-Lozada LG. Vasopressin Mediates the Renal Damage Induced by Limited Fructose Rehydration in Recurrently Dehydrated Rats. Int J Biol Sci 2017; 13:961-975. [PMID: 28924378 PMCID: PMC5599902 DOI: 10.7150/ijbs.20074] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/26/2017] [Indexed: 12/26/2022] Open
Abstract
Recurrent dehydration and heat stress cause chronic kidney damage in experimental animals. The injury is exacerbated by rehydration with fructose-containing beverages. Fructose may amplify dehydration-induced injury by directly stimulating vasopressin release and also by acting as a substrate for the aldose reductase-fructokinase pathway, as both of these systems are active during dehydration. The role of vasopressin in heat stress associated injury has not to date been explored. Here we show that the amplification of renal damage mediated by fructose in thermal dehydration is mediated by vasopressin. Fructose rehydration markedly enhanced vasopressin (copeptin) levels and activation of the aldose reductase-fructokinase pathway in the kidney. Moreover, the amplification of the renal functional changes (decreased creatinine clearance and tubular injury with systemic inflammation, renal oxidative stress, and mitochondrial dysfunction) were prevented by the blockade of V1a and V2 vasopressin receptors with conivaptan. On the other hand, there are also other operative mechanisms when water is used as rehydration fluid that produce milder renal damage that is not fully corrected by vasopressin blockade. Therefore, we clearly showed evidence of the cross-talk between fructose, even at small doses, and vasopressin that interact to amplify the renal damage induced by dehydration. These data may be relevant for heat stress nephropathy as well as for other renal pathologies due to the current generalized consumption of fructose and deficient hydration habits.
Collapse
Affiliation(s)
| | - Edilia Tapia
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | | | - Guillermo Gonzaga
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico
| | - Octaviano Silverio
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico
| | - Magdalena Cristóbal
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | - Horacio Osorio
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | - Abraham S Arellano-Buendía
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| | - Cecilia Zazueta
- Dept. of Cardiovascular Biomedicine. INC Ignacio Chávez. Mexico City. Mexico
| | | | - Juan G Reyes-García
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, IPN Mexico City. Mexico
| | | | - Virgilia Soto
- Dept. of Pathology. INC Ignacio Chávez. Mexico City. Mexico
| | | | | | - Laura G Sánchez-Lozada
- Laboratory of Renal Physiopathology. INC Ignacio Chávez. Mexico City. Mexico.,Dept. of Nephrology. INC Ignacio Chávez. Mexico City. Mexico
| |
Collapse
|
27
|
Franko A, Kunze A, Böse M, von Kleist-Retzow JC, Paulsson M, Hartmann U, Wiesner RJ. Impaired Insulin Signaling is Associated with Hepatic Mitochondrial Dysfunction in IR +/--IRS-1 +/- Double Heterozygous (IR-IRS1dh) Mice. Int J Mol Sci 2017; 18:ijms18061156. [PMID: 28556799 PMCID: PMC5485980 DOI: 10.3390/ijms18061156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/21/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Mitochondria play a pivotal role in energy metabolism, but whether insulin signaling per se could regulate mitochondrial function has not been identified yet. To investigate whether mitochondrial function is regulated by insulin signaling, we analyzed muscle and liver of insulin receptor (IR)+/−-insulin receptor substrate-1 (IRS-1)+/− double heterozygous (IR-IRS1dh) mice, a well described model for insulin resistance. IR-IRS1dh mice were studied at the age of 6 and 12 months and glucose metabolism was determined by glucose and insulin tolerance tests. Mitochondrial enzyme activities, oxygen consumption, and membrane potential were assessed using spectrophotometric, respirometric, and proton motive force analysis, respectively. IR-IRS1dh mice showed elevated serum insulin levels. Hepatic mitochondrial oxygen consumption was reduced in IR-IRS1dh animals at 12 months of age. Furthermore, 6-month-old IR-IRS1dh mice demonstrated enhanced mitochondrial respiration in skeletal muscle, but a tendency of impaired glucose tolerance. On the other hand, 12-month-old IR-IRS1dh mice showed improved glucose tolerance, but normal muscle mitochondrial function. Our data revealed that deficiency in IR/IRS-1 resulted in normal or even elevated skeletal muscle, but impaired hepatic mitochondrial function, suggesting a direct cross-talk between insulin signaling and mitochondria in the liver.
Collapse
Affiliation(s)
- Andras Franko
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str. 10, D-72076 Tübingen, Germany.
| | - Alexander Kunze
- Center for Biochemistry, Medical Faculty, University of Köln, Joseph-Stelzmann-Str. 52, D-50931 Cologne, Germany.
| | - Marlen Böse
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
| | - Jürgen-Christoph von Kleist-Retzow
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
- Department of Pediatrics, University Hospital Cologne, Kerpener Str. 62, D-50937 Cologne, Germany.
| | - Mats Paulsson
- Center for Biochemistry, Medical Faculty, University of Köln, Joseph-Stelzmann-Str. 52, D-50931 Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Köln, D-50931 Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Köln, D-50931 Cologne, Germany.
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Köln, Joseph-Stelzmann-Str. 52, D-50931 Cologne, Germany.
| | - Rudolf J Wiesner
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Köln, D-50931 Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Köln, D-50931 Cologne, Germany.
| |
Collapse
|
28
|
Spahis S, Borys JM, Levy E. Metabolic Syndrome as a Multifaceted Risk Factor for Oxidative Stress. Antioxid Redox Signal 2017; 26:445-461. [PMID: 27302002 DOI: 10.1089/ars.2016.6756] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Metabolic syndrome (MetS) is associated with a greater risk of diabetes and cardiovascular diseases. It is estimated that this multifactorial condition affects 20%-30% of the world's population. A detailed understanding of MetS mechanisms is crucial for the development of effective prevention strategies and adequate intervention tools that could curb its increasing prevalence and limit its comorbidities, particularly in younger age groups. With advances in basic redox biology, oxidative stress (OxS) involvement in the complex pathophysiology of MetS has become widely accepted. Nevertheless, its clear association with and causative effects on MetS require further elucidation. Recent Advances: Although a better understanding of the causes, risks, and effects of MetS is essential, studies suggest that oxidant/antioxidant imbalance is a key contributor to this condition. OxS is now understood to be a major underlying mechanism for mitochondrial dysfunction, ectopic lipid accumulation, and gut microbiota impairment. CRITICAL ISSUES Further studies, particularly in the field of translational research, are clearly required to understand and control the production of reactive oxygen species (ROS) levels, especially in the mitochondria, since the various therapeutic trials conducted to date have not targeted this major ROS-generating system, aimed to delay MetS onset, or prevent its progression. FUTURE DIRECTIONS Multiple relevant markers need to be identified to clarify the role of ROS in the etiology of MetS. Future clinical trials should provide important proof of concept for the effectiveness of antioxidants as useful therapeutic approaches to simultaneously counteract mitochondrial OxS, alleviate MetS symptoms, and prevent complications. Antioxid. Redox Signal. 26, 445-461.
Collapse
Affiliation(s)
- Schohraya Spahis
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada
| | | | - Emile Levy
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada .,3 EPODE International Network , Paris, France
| |
Collapse
|
29
|
Neeland IJ, Hughes C, Ayers CR, Malloy CR, Jin ES. Effects of visceral adiposity on glycerol pathways in gluconeogenesis. Metabolism 2017; 67:80-89. [PMID: 28081781 PMCID: PMC5244471 DOI: 10.1016/j.metabol.2016.11.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/27/2016] [Accepted: 11/22/2016] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To determine the feasibility of using oral 13C labeled glycerol to assess effects of visceral adiposity on gluconeogenic pathways in obese humans. RESEARCH DESIGN AND METHODS Obese (BMI ≥30kg/m2) participants without type 2 diabetes underwent visceral adipose tissue (VAT) assessment and stratification by median VAT into high VAT-fasting (n=3), low VAT-fasting (n=4), and high VAT-refed (n=2) groups. Participants ingested [U-13C3] glycerol and blood samples were subsequently analyzed at multiple time points over 3h by NMR spectroscopy. The fractions of plasma glucose (enrichment) derived from [U-13C3] glycerol via hepatic gluconeogenesis, pentose phosphate pathway (PPP), and tricarboxylic acid (TCA) cycle were assessed using 13C NMR analysis of glucose. Mixed linear models were used to compare 13C enrichment in glucose between groups. RESULTS Mean age, BMI, and baseline glucose were 49years, 40.1kg/m2, and 98mg/dl, respectively. Up to 20% of glycerol was metabolized in the TCA cycle prior to gluconeogenesis and PPP activity was minor (<1% of total glucose) in all participants. There was a 21% decrease in 13C enrichment in plasma glucose in the high VAT-fasting compared with low VAT-fasting group (p=0.03), suggesting dilution by endogenous glycerol. High VAT-refed participants had 37% less 13C enrichment in glucose compared with high VAT-fasting (p=0.02). There was a trend toward lower [1,2-13C2] (via PPP) and [5,6-13C2]/[4,5,6-13C3] (via TCA cycle) glucose in high VAT versus low VAT groups. CONCLUSIONS We applied a simple method to detect gluconeogenesis from glycerol in obese humans. Our findings provide preliminary evidence that excess visceral fat disrupts multiple pathways in hepatic gluconeogenesis from glycerol.
Collapse
Affiliation(s)
- Ian J Neeland
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Connor Hughes
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Colby R Ayers
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Craig R Malloy
- Advanced Imaging Research Center and Departments of Internal Medicine and Radiology, University of Texas Southwestern Medical Center and VA North Texas Healthcare System, Dallas, TX, USA
| | - Eunsook S Jin
- Advanced Imaging Research Center and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
30
|
Abstract
Recent human and animal studies investigating the roles of the genome, epigenome, and environmental cues have identified associations between offspring predisposition to life-long obesity/metabolic disease and epigenetic modifications such as DNA methylation. This review explores the mechanisms by which maternal exposures impair the health of not only the next generation but also potentially future generations of offspring.
Collapse
Affiliation(s)
- Kathleen Jaeger
- Washington University School of Medicine, St. Louis, Missouri
| | - Jessica L Saben
- Washington University School of Medicine, St. Louis, Missouri
| | - Kelle H Moley
- Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
31
|
Johns M, Esmaeili Mohsen Abadi S, Malik N, Lee J, Neumann WL, Rausaria S, Imani-Nejad M, McPherson T, Schober J, Kwon G. Oral administration of SR-110, a peroxynitrite decomposing catalyst, enhances glucose homeostasis, insulin signaling, and islet architecture in B6D2F1 mice fed a high fat diet. Arch Biochem Biophys 2016; 596:126-37. [PMID: 26970045 DOI: 10.1016/j.abb.2016.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/23/2016] [Accepted: 03/02/2016] [Indexed: 01/03/2023]
Abstract
Peroxynitrite has been implicated in type 2 diabetes and diabetic complications. As a follow-up study to our previous work on SR-135 (Arch Biochem Biophys 577-578: 49-59, 2015), we provide evidence that this series of compounds are effective when administered orally, and their mechanisms of actions extend to the peripheral tissues. A more soluble analogue of SR-135, SR-110 (from a new class of Mn(III) bis(hydroxyphenyl)-dipyrromethene complexes) was orally administered for 2 weeks to B6D2F1 mice fed a high fat-diet (HFD). Mice fed a HFD for 4 months gained significantly higher body weights compared to lean diet-fed mice (52 ± 1.5 g vs 34 ± 1.3 g). SR-110 (10 mg/kg daily) treatment significantly reduced fasting blood glucose and insulin levels, and enhanced glucose tolerance as compared to HFD control or vehicle (peanut butter) group. SR-110 treatment enhanced insulin signaling in the peripheral organs, liver, heart, and skeletal muscle, and reduced lipid accumulation in the liver. Furthermore, SR-110 increased insulin content, restored islet architecture, decreased islet size, and reduced tyrosine nitration. These results suggest that a peroxynitrite decomposing catalyst is effective in improving glucose homeostasis and restoring islet morphology and β-cell insulin content under nutrient overload.
Collapse
Affiliation(s)
- Michael Johns
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | | | - Nehal Malik
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - Joshua Lee
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - William L Neumann
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - Smita Rausaria
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - Maryam Imani-Nejad
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - Timothy McPherson
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - Joseph Schober
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA
| | - Guim Kwon
- School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL, 62026, USA.
| |
Collapse
|