1
|
Lin SX, Li XY, Chen QC, Ni Q, Cai WF, Jiang CP, Yi YK, Liu L, Liu Q, Shen CY. Eriodictyol regulates white adipose tissue browning and hepatic lipid metabolism in high fat diet-induced obesity mice via activating AMPK/SIRT1 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118761. [PMID: 39216775 DOI: 10.1016/j.jep.2024.118761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/04/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Blossom of Citrus aurantium L. var. amara Engl. (CAVA) has been popularly consumed as folk medicine and dietary supplement owing to its various beneficial effects and especially anti-obesity potential. Our previous study predicted that eriodictyol was probably one of the key active compounds of the total flavonoids from blossom of CAVA. However, effects of eriodictyol in anti-obesity were still elusive. AIM OF THE STUDY This study was performed to explore the precise role of eriodictyol in white adipose tissue (WAT) browning and hepatic lipid metabolism, and simultaneously, to verify the impact of eriodictyol on the total flavonoids of CAVA in losing weight. MATERIALS AND METHODS The pancreas lipase assay was conducted and oleic acid-induced HepG2 cells were established to preliminarily detect the lipid-lowering potential of eriodictyol. Then, high fat diet-induced obesity (DIO) mouse model was established for in vivo studies. The biochemical indicators of mice were tested by commercial kits. The histopathological changes of WAT and liver in mice were tested by H&E staining, Oil Red O staining and Sirius Red staining. Immunohistochemical, Western blot assay, as well as RT-qPCR analysis were further performed. Additionally, molecular docking assay was used to simulate the binding of eriodictyol with potential target proteins. RESULTS In vitro studies showed that eriodictyol intervention potently inhibited pancreatic lipase activity and reversed hepatic steatosis in oleic acid-induced HepG2 cells. Consistently, long-term medication of eriodictyol also effectively prevented obesity and improved lipid and glucose metabolism in diet-induced obesity mice. Obesity-induced histopathological changes in iWAT, eWAT and BAT, and abnormal expression levels of IL-10, IL-6 and TNF-α in iWAT of DIO mice were also significantly reversed by eriodictyol treatment. Eriodictyol administration significantly and potently promoted browning of iWAT by increasing expression levels of thermogenic marker protein of UCP1, as well as brown adipocyte-specific genes of PGC-1α, SIRT1 and AMPKα1. Further assays revealed that eriodictyol enhanced mitochondrial function, as shown by an increase in compound IV activity and the expression of tricarboxylic acid cycle-related genes. Besides, eriodictyol addition markedly reversed hepatic damages and hepatic inflammation, and enhanced hepatic lipid metabolism in DIO mice, as evidenced by its regulation on p-ACC, CPT1-α, UCP1, PPARα, PGC-1α, SIRT1 and p-AMPKα expression. Molecular docking results further validated that AMPK/SIRT1 pathway was probably the underlying mechanisms by which eriodictyol acted. CONCLUSION Eriodictyol exhibited significant anti-obesity effect, which was comparable to that of the total flavonoids from blossom of CAVA. These findings furnished theoretical basis for the application of eriodictyol in weight loss.
Collapse
Affiliation(s)
- Song-Xia Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Xiao-Yi Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Qi-Cong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Qian Ni
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Wei-Feng Cai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Cui-Ping Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China.
| | - Yan-Kui Yi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China.
| | - Li Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Chun-Yan Shen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Uno K, Uchino T, Suzuki T, Sayama Y, Edo N, Uno-Eder K, Morita K, Ishikawa T, Koizumi M, Honda H, Katagiri H, Tsukamoto K. Rspo3-mediated metabolic liver zonation regulates systemic glucose metabolism and body mass in mice. PLoS Biol 2025; 23:e3002955. [PMID: 39854351 PMCID: PMC11759367 DOI: 10.1371/journal.pbio.3002955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/27/2024] [Indexed: 01/26/2025] Open
Abstract
The unique architecture of the liver consists of hepatic lobules, dividing the hepatic features of metabolism into 2 distinct zones, namely the pericentral and periportal zones, the spatial characteristics of which are broadly defined as metabolic zonation. R-spondin3 (Rspo3), a bioactive protein promoting the Wnt signaling pathway, regulates metabolic features especially around hepatic central veins. However, the functional impact of hepatic metabolic zonation, regulated by the Rspo3/Wnt signaling pathway, on whole-body metabolism homeostasis remains poorly understood. In this study, we analyze the local functions of Rspo3 in the liver and the remote actions of hepatic Rspo3 on other organs of the body by using murine models. Rspo3 expression analysis shows that Rspo3 expression patterns are spatiotemporally controlled in the murine liver such that it locates in the pericentral zones and converges after feeding, and the dynamics of these processes are disturbed in obesity. We find that viral-mediated induction of Rspo3 in hepatic tissue of obesity improves insulin resistance and prevents body weight gain by restoring attenuated organ insulin sensitivities, reducing adipose tissue enlargement and reversing overstimulated adaptive thermogenesis. Denervation of the hepatic vagus suppresses these remote effects, derived from hepatic Rspo3 induction, toward adipose tissues and skeletal muscle, suggesting that signals are transduced via the neuronal communication consisting of afferent vagal and efferent sympathetic nerves. Furthermore, the non-neuronal inter-organ communication up-regulating muscle lipid utilization is partially responsible for the ameliorations of both fatty liver development and reduced skeletal muscle quality in obesity. In contrast, hepatic Rspo3 suppression through Cre-LoxP-mediated recombination system exacerbates diabetes due to glucose intolerance and insulin resistance, promotes fatty liver development and decreases skeletal muscle quality, resulting in obesity. Taken together, our study results reveal that modulation of hepatic Rspo3 contributes to maintaining systemic glucose metabolism and body composition via a newly identified inter-organ communication mechanism.
Collapse
Affiliation(s)
- Kenji Uno
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takuya Uchino
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takashi Suzuki
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yohei Sayama
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Naoki Edo
- Teikyo Academic Research Center, Tokyo, Japan
| | | | - Koji Morita
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Toshio Ishikawa
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Miho Koizumi
- Field of Human Disease Models, Tokyo Women’s Medical University, Tokyo, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Tokyo Women’s Medical University, Tokyo, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhisa Tsukamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
3
|
Gatto L, Di Nunno V, Ghelardini A, Tosoni A, Bartolini S, Asioli S, Ratti S, Di Stefano AL, Franceschi E. Targeting Mitochondria in Glioma: New Hopes for a Cure. Biomedicines 2024; 12:2730. [PMID: 39767637 PMCID: PMC11727304 DOI: 10.3390/biomedicines12122730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Drugs targeting mitochondrial energy metabolism are emerging as promising antitumor therapeutics. Glioma treatment is extremely challenging due to the high complexity of the tumor and the high cellular heterogeneity. From a metabolic perspective, glioma cancer cells can be classified into the oxidative metabolic phenotype (mainly depending on mitochondrial respiration for energy production) and glycolytic phenotype or "Warburg effect" (mainly depending on glycolysis). Herein, we reviewed the function of novel bio-active molecules targeting oxidative phosphorylation (OXPHOS), mitochondrial membrane potential and mitochondrial dynamics. These molecules exhibit intriguing preclinical and clinical results and have been proven to be promising candidates to be further developed for glioma therapy. However, despite these initial encouraging results, it is imperative to rigorously assess the side effects of these metabolic drugs, which have a non-negligible toxicity profile.
Collapse
Affiliation(s)
- Lidia Gatto
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Vincenzo Di Nunno
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Anna Ghelardini
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Sofia Asioli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy;
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Stefano Ratti
- Cellular Signalling Laboratory, Anatomy Center, Department of Biomedical Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy;
| | - Anna Luisa Di Stefano
- Division of Neurosurgery, Azienda USL Toscana Nord Ovest, Spedali Riuniti di Livorno, 56121 Livorno, Italy;
- Department of Neurology, Foch Hospital, 92150 Suresnes, France
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| |
Collapse
|
4
|
Wang H, An Y, Rajput SA, Qi D. Resveratrol and (-)-Epigallocatechin-3-gallate Regulate Lipid Metabolism by Activating the AMPK Pathway in Hepatocytes. BIOLOGY 2024; 13:368. [PMID: 38927248 PMCID: PMC11201192 DOI: 10.3390/biology13060368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
The purpose of this study was to explore the effects of Res and EGCG on cell growth, cellular antioxidant levels, and cellular lipid metabolism in hepatocytes. In this experiment, leghorn male hepatoma (LMH) cells were used as hepatocytes. The results showed that 6.25-25 μM Res and EGCG had no adverse effects on cell viability and growth. Meanwhile, with the increasing dosage of Res and EGCG, the contents of total cholesterol (TC), total glyceride (TG), and malondialdehyde (MDA) in hepatocytes decreased significantly (p < 0.05), while the contents of glutathione peroxidase (GSH-Px), total superoxide dismutase (T-SOD), and catalase (CAT) increased significantly (p < 0.05). In addition, western blot results showed that Res and EGCG could significantly increase the expression of p-AMPK protein and reduce the expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) protein in hepatocytes (p < 0.05). Moreover, q-PCR results showed that with the increase in Res and EGCG, the expression of cholesterol- and fatty acid synthesis-related genes decreased significantly (p < 0.05). In conclusion, Res and EGCG can increase the antioxidant capacity of hepatocytes and reduce the synthesis of TC and TG in hepatocytes by activating AMPK, thereby regulating lipid metabolism in hepatocytes.
Collapse
Affiliation(s)
- Huanbin Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.W.); (Y.A.)
| | - Yu An
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.W.); (Y.A.)
| | - Shahid Ali Rajput
- Department of Animal and Dairy Sciences, Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan 60000, Pakistan;
| | - Desheng Qi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (H.W.); (Y.A.)
| |
Collapse
|
5
|
Onodera K, Hasegawa Y, Yokota N, Tamura S, Kinno H, Takahashi I, Chiba H, Kojima H, Katagiri H, Nata K, Ishigaki Y. A newly identified compound activating UCP1 inhibits obesity and its related metabolic disorders. Obesity (Silver Spring) 2024; 32:324-338. [PMID: 37974549 DOI: 10.1002/oby.23948] [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: 04/08/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE Promoting thermogenesis in adipose tissue has been a promising strategy against obesity and related metabolic complications. We aimed to identify compounds that promote thermogenesis in adipocytes and to elucidate their functions and roles in metabolism. METHODS To identify compounds that directly promote thermogenesis from a structurally diverse set of 4800 compounds, we utilized a cell-based platform for high-throughput screening that induces uncoupling protein 1 (Ucp1) expression in adipocytes. RESULTS We identified one candidate compound that activates UCP1. Additional characterization of this compound revealed that it induced cellular thermogenesis in adipocytes with negligible cytotoxicity. In a subsequent diet-induced obesity model, mice treated with this compound exhibited a slower rate of weight gain, improved insulin sensitivity, and increased energy expenditure. Mechanistic studies have revealed that this compound increases mitochondrial biogenesis by elevating maximal respiration, which is partly mediated by the protein kinase A (PKA)-p38 mitogen-activated protein kinase (MAPK) signaling pathway. A further comprehensive genetic analysis of adipocytes treated with these compounds identified two novel UCP1-dependent thermogenic genes, potassium voltage-gated channel subfamily C member 2 (Kcnc2) and predicted gene 5627 (Gm5627). CONCLUSIONS The identified compound can serve as a potential therapeutic drug for the treatment of obesity and its related metabolic disorders. Furthermore, our newly clarified thermogenic genes play an important role in UCP1-dependent thermogenesis in adipocytes.
Collapse
Affiliation(s)
- Ken Onodera
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Yutaka Hasegawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Nozomi Yokota
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Shukuko Tamura
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Hirofumi Kinno
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Iwao Takahashi
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmacy, Iwate Medical University, Yahaba, Japan
| | - Hiraku Chiba
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Hirotatsu Kojima
- Drug Discovery Initiative, The University of Tokyo, Tokyo, Japan
| | - Hideki Katagiri
- Department of Diabetes and Metabolism, Tohoku University Graduate School of Medicine, Tohoku University Hospital, Sendai, Japan
| | - Koji Nata
- Division of Medical Biochemistry, School of Pharmacy, Iwate Medical University, Yahaba, Japan
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
6
|
Cojocaru KA, Luchian I, Goriuc A, Antoci LM, Ciobanu CG, Popescu R, Vlad CE, Blaj M, Foia LG. Mitochondrial Dysfunction, Oxidative Stress, and Therapeutic Strategies in Diabetes, Obesity, and Cardiovascular Disease. Antioxidants (Basel) 2023; 12:antiox12030658. [PMID: 36978905 PMCID: PMC10045078 DOI: 10.3390/antiox12030658] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Mitochondria are subcellular organelles involved in essential cellular functions, including cytosolic calcium regulation, cell apoptosis, and reactive oxygen species production. They are the site of important biochemical pathways, including the tricarboxylic acid cycle, parts of the ureagenesis cycle, or haem synthesis. Mitochondria are responsible for the majority of cellular ATP production through OXPHOS. Mitochondrial dysfunction has been associated with metabolic pathologies such as diabetes, obesity, hypertension, neurodegenerative diseases, cellular aging, and cancer. In this article, we describe the pathophysiological changes in, and mitochondrial role of, metabolic disorders (diabetes, obesity, and cardiovascular disease) and their correlation with oxidative stress. We highlight the genetic changes identified at the mtDNA level. Additionally, we selected several representative biomarkers involved in oxidative stress and summarize the progress of therapeutic strategies.
Collapse
Affiliation(s)
- Karina-Alexandra Cojocaru
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
| | - Ionut Luchian
- Department of Periodontology, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Ancuta Goriuc
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
- Correspondence: (A.G.); (C.-E.V.)
| | - Lucian-Mihai Antoci
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
| | - Cristian-Gabriel Ciobanu
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
| | - Roxana Popescu
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
- Department of Medical Genetics, “Saint Mary” Emergency Children’s Hospital, Vasile Lupu Street, No. 62, 700309 Iasi, Romania
| | - Cristiana-Elena Vlad
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
- Department of Nephrology-Internal Medicine, “Dr. C. I. Parhon” Clinical Hospital, 700503 Iasi, Romania
- Correspondence: (A.G.); (C.-E.V.)
| | - Mihaela Blaj
- Anaesthesia and Intensive Care Department, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Anaesthesia and Intensive Care Department, Sf. Spiridon University Hospital, 700111 Iasi, Romania
| | - Liliana Georgeta Foia
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
| |
Collapse
|
7
|
Luo Y, Chen Q, Zou J, Fan J, Li Y, Luo Z. Chronic Intermittent Hypoxia Exposure Alternative to Exercise Alleviates High-Fat-Diet-Induced Obesity and Fatty Liver. Int J Mol Sci 2022; 23:ijms23095209. [PMID: 35563600 PMCID: PMC9104027 DOI: 10.3390/ijms23095209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity often concurs with nonalcoholic fatty liver disease (NAFLD), both of which are detrimental to human health. Thus far, exercise appears to be an effective treatment approach. However, its effects cannot last long and, moreover, it is difficult to achieve for many obese people. Thus, it is necessary to look into alternative remedies. The present study explored a noninvasive, easy, tolerable physical alternative. In our experiment, C57BL/6 mice were fed with a high-fat diet (HFD) to induce overweight/obesity and were exposed to 10% oxygen for one hour every day. We found that hypoxia exerted protective effects. First, it offset HFD-induced bodyweight gain and insulin resistance. Secondly, hypoxia reversed the HFD-induced enlargement of white and brown adipocytes and fatty liver, and protected liver function. Thirdly, HFD downregulated the expression of genes required for lipolysis and thermogenesis, such as UCP1, ADR3(beta3-adrenergic receptor), CPT1A, ATGL, PPARα, and PGC1α, M2 macrophage markers arginase and CD206 in the liver, and UCP1 and PPARγ in brown fat, while these molecules were upregulated by hypoxia. Furthermore, hypoxia induced the activation of AMPK, an energy sensing enzyme. Fourthly, our results showed that hypoxia increased serum levels of epinephrine. Indeed, the effects of hypoxia on bodyweight, fatty liver, and associated changes in gene expression ever tested were reproduced by injection of epinephrine and prevented by propranolol at varying degrees. Altogether, our data suggest that hypoxia triggers stress responses where epinephrine plays important roles. Therefore, our study sheds light on the hope to use hypoxia to treat the daunting disorders, obesity and NAFLD.
Collapse
Affiliation(s)
- Yunfei Luo
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Qiongfeng Chen
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Junrong Zou
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Jingjing Fan
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Yuanjun Li
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Zhijun Luo
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
- Queen Mary School, Nanchang University, Nanchang 330031, China
- Correspondence: ; Tel.: +86-158-7917-7010
| |
Collapse
|
8
|
Albracht-Schulte K, Wilson S, Johnson P, Pahlavani M, Ramalingam L, Goonapienuwala B, Kalupahana NS, Festuccia WT, Scoggin S, Kahathuduwa CN, Moustaid-Moussa N. Sex-Dependent Effects of Eicosapentaenoic Acid on Hepatic Steatosis in UCP1 Knockout Mice. Biomedicines 2021; 9:1549. [PMID: 34829779 PMCID: PMC8615653 DOI: 10.3390/biomedicines9111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/26/2022] Open
Abstract
Visceral obesity may be a driving factor in nonalcoholic fatty liver disease (NAFLD) development. Previous studies have shown that the omega-3 polyunsaturated fatty acid, eicosapentaenoic acid (EPA), ameliorates obesity in high-fat (HF) fed male, C57Bl/6 mice at thermoneutral conditions, independent of uncoupling protein 1 (UCP1). Our goals herein were to investigate sex-dependent mechanisms of EPA in the livers of wild type (WT) and UCP1 knockout (KO) male and female mice fed a HF diet (45% kcal fat; WT-HF, KO-HF) with or without supplementation of 36 g/kg EPA (WT-EPA, KO-EPA). KO significantly increased body weight in males, with no significant reductions with EPA in the WT or KO groups. In females, there were no significant differences in body weight among KO groups and no effects of EPA. In males, liver TGs were significantly higher in the KO-HF group and reduced with EPA, which was not observed in females. Accordingly, gene and protein markers of mitochondrial oxidation, peroxisomal biogenesis and oxidation, as well as metabolic futile cycles were sex-dependently impacted by KO and EPA supplementation. These findings suggest a genotypic difference in response to dietary EPA supplementation on the livers of male and female mice with diet-induced obesity and housed at thermoneutrality.
Collapse
Affiliation(s)
- Kembra Albracht-Schulte
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Savanna Wilson
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Paige Johnson
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Mandana Pahlavani
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Latha Ramalingam
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Bimba Goonapienuwala
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Nishan S. Kalupahana
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
- Department of Physiology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - William T. Festuccia
- Department of Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
| | - Shane Scoggin
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Chanaka N. Kahathuduwa
- Texas Tech University Health Sciences Center, Department of Laboratory Sciences and Primary Care, Lubbock, TX 79430, USA;
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| |
Collapse
|
9
|
Milane L, Dolare S, Jahan T, Amiji M. Mitochondrial nanomedicine: Subcellular organelle-specific delivery of molecular medicines. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102422. [PMID: 34175455 DOI: 10.1016/j.nano.2021.102422] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/21/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
As mitochondria network together to act as the master sensors and effectors of apoptosis, ATP production, reactive oxygen species management, mitophagy/autophagy, and homeostasis; this organelle is an ideal target for pharmaceutical manipulation. Mitochondrial dysfunction contributes to many diseases, for example, β-amyloid has been shown to interfere with mitochondrial protein import and induce apoptosis in Alzheimer's Disease while some forms of Parkinson's Disease are associated with dysfunctional mitochondrial PINK1 and Parkin proteins. Mitochondrial medicine has applications in the treatment of an array of pathologies from cancer to cardiovascular disease. A challenge of mitochondrial medicine is directing therapies to a subcellular target. Nanotechnology based approaches combined with mitochondrial targeting strategies can greatly improve the clinical translation and effectiveness of mitochondrial medicine. This review discusses mitochondrial drug delivery approaches and applications of mitochondrial nanomedicines. Nanomedicine approaches have the potential to drive the success of mitochondrial therapies into the clinic.
Collapse
Affiliation(s)
- Lara Milane
- Northeastern University, Department of Pharmaceutical Sciences, Boston, MA.
| | - Saket Dolare
- Northeastern University, Department of Pharmaceutical Sciences, Boston, MA
| | - Tanjheela Jahan
- Northeastern University, Department of Pharmaceutical Sciences, Boston, MA
| | - Mansoor Amiji
- Northeastern University, Department of Pharmaceutical Sciences, Boston, MA
| |
Collapse
|
10
|
Weber M, Mera P, Casas J, Salvador J, Rodríguez A, Alonso S, Sebastián D, Soler-Vázquez MC, Montironi C, Recalde S, Fucho R, Calderón-Domínguez M, Mir JF, Bartrons R, Escola-Gil JC, Sánchez-Infantes D, Zorzano A, Llorente-Cortes V, Casals N, Valentí V, Frühbeck G, Herrero L, Serra D. Liver CPT1A gene therapy reduces diet-induced hepatic steatosis in mice and highlights potential lipid biomarkers for human NAFLD. FASEB J 2020; 34:11816-11837. [PMID: 32666604 DOI: 10.1096/fj.202000678r] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/12/2020] [Accepted: 06/19/2020] [Indexed: 12/25/2022]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has increased drastically due to the global obesity pandemic but at present there are no approved therapies. Here, we aimed to revert high-fat diet (HFD)-induced obesity and NAFLD in mice by enhancing liver fatty acid oxidation (FAO). Moreover, we searched for potential new lipid biomarkers for monitoring liver steatosis in humans. We used adeno-associated virus (AAV) to deliver a permanently active mutant form of human carnitine palmitoyltransferase 1A (hCPT1AM), the key enzyme in FAO, in the liver of a mouse model of HFD-induced obesity and NAFLD. Expression of hCPT1AM enhanced hepatic FAO and autophagy, reduced liver steatosis, and improved glucose homeostasis. Lipidomic analysis in mice and humans before and after therapeutic interventions, such as hepatic AAV9-hCPT1AM administration and RYGB surgery, respectively, led to the identification of specific triacylglyceride (TAG) specie (C50:1) as a potential biomarker to monitor NAFFLD disease. To sum up, here we show for the first time that liver hCPT1AM gene therapy in a mouse model of established obesity, diabetes, and NAFLD can reduce HFD-induced derangements. Moreover, our study highlights TAG (C50:1) as a potential noninvasive biomarker that might be useful to monitor NAFLD in mice and humans.
Collapse
Affiliation(s)
- Minéia Weber
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC)/CSIC, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Salvador
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Sergio Alonso
- Cancer Genetics and Epigenetics Group, Program of Predictive and Personalized Medicine of Cancer, Germans Trias i Pujol Research Institute (IGTP-PMPPC), Campus Can Ruti, Barcelona, Spain
| | - David Sebastián
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - M Carmen Soler-Vázquez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Carla Montironi
- Pathology Department, Hospital Clinic de Barcelona, Barcelona, Spain.,Liver Cancer Translational Research Laboratory, Liver Unit, IDIBAPS-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Sandra Recalde
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Raquel Fucho
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - María Calderón-Domínguez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Joan Francesc Mir
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Ramon Bartrons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Joan Carles Escola-Gil
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.,IIB Sant Pau, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David Sánchez-Infantes
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Germans Trias i Pujol Research Institute (IGTP-PMPPC), Campus Can Ruti, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Vicenta Llorente-Cortes
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.,CIBERCV, Institute of Health Carlos III, Madrid, Spain.,Cardiovascular Research Center, CSIC-ICCC, Barcelona, Spain
| | - Núria Casals
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Víctor Valentí
- Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, Spain.,Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
11
|
Badi RM, Mostafa DG, Khaleel EF, Satti HH. Resveratrol protects against hepatic insulin resistance in a rat's model of non-alcoholic fatty liver disease by down-regulation of GPAT-1 and DGAT2 expression and inhibition of PKC membranous translocation. Clin Exp Pharmacol Physiol 2019; 46:545-555. [PMID: 30773673 DOI: 10.1111/1440-1681.13074] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/16/2019] [Accepted: 02/13/2019] [Indexed: 12/16/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with hepatic insulin resistance (IR). Resveratrol (RES) a potent hypolipidemic dietary polyphenol has been identified for its ability to prevent hepatic steatosis and hepatic IR in high-fat diet (HFD)-fed murine models of NAFLD. In the present study, we have carried an in vivo animal experiment to identify a novel mechanism for RES protective action. Sub-chronic (45 days) RES pretreatment in 3 days HFD-fed adult Wistar rats prevented early hepatic IR through inhibiting PKC/JNK activation; decreasing p-IRS (Ser307 ) and increasing p-IRS(Tyr612 ), p-Akt(Ser473 ) and p-GSK3(Ser9 ). These effects of RES were associated with reduced expression of acyl-CoA:glycerol-sn-3-phosphate acyltransferase (GPAT-1) and diacylglycerol:acyl-CoA acyltransferase (DGAT2), two critical enzymes in the glycerol-3-phosphate pathway for de novo triglycerides synthesis. These data indicate that RES protects against NAFLD, initially, by inhibiting the early development of hepatic IR.
Collapse
Affiliation(s)
- Rehab M Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Physiology, University of Khartoum, Khartoum, Sudan
| | - Dalia G Mostafa
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Medical Physiology, Assiut University, Assiut, Egypt
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Medical Physiology, Cairo University, Cairo, Egypt
| | - Huda H Satti
- Department of Pathology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Pathology, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
12
|
Guo J, Tao H, Alasadi A, Huang Q, Jin S. Niclosamide piperazine prevents high-fat diet-induced obesity and diabetic symptoms in mice. Eat Weight Disord 2019; 24:91-96. [PMID: 28780747 DOI: 10.1007/s40519-017-0424-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 07/21/2017] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Obesity and type 2 diabetes (T2D) have become the major public health challenges globally. Mitochondrial uncoupling, which reduces intracellular lipid loads and corrects the underlying cause of insulin resistance, has emerged as a promising anti-obese and anti-diabetic intervention. Niclosamide is an anthelmintic drug approved by the US FDA with the mechanism of action that uncouples mitochondria of parasitic worms. Recently, niclosamide ethanolamine salt (NEN) was found to be a safe and effective hepatic mitochondrial uncoupler for the prevention and treatment of obesity and T2D in mouse models. The striking features of NEN prompt us to examine the anti-obese and anti-diabetic efficacy of other salt forms of niclosamide, with the ultimate goal to identify a suitable salt formulation for future clinical development. Here, we report the study with niclosamide piperazine salt (NPP), another salt form of niclosamide with documented safety profile. METHODS Mitochondrial uncoupling activity of NEN and NPP were determined by oxygen consumption assay with Seahorse XF24e Analyzer, as well as by mitochondrial membrane potential measurement in cultured cells. The in vivo anti-diabetic and anti-obesity activities were determined in C57BL/6J mice fed high-fat diet (HFD) or HFD containing 2000 ppm. NPP for 11 weeks. RESULTS Niclosamide piperazine salt showed a comparable mitochondrial uncoupling activity to NEN. Oral administration of NPP significantly reduced HFD-induced obesity, hyperglycemia and hepatic steatosis, and sensitized the insulin responses in mice. CONCLUSIONS Niclosamide piperazine salt may hold the promise to become an alternative to NEN as a drug lead for the treatment of obesity and T2D. No level of evidence Animal study.
Collapse
Affiliation(s)
- Jingjing Guo
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Hanlin Tao
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Amer Alasadi
- Department of Physiology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Qingrong Huang
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Shengkan Jin
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| |
Collapse
|
13
|
Bigagli E, Cinci L, Niccolai A, Biondi N, Rodolfi L, D'Ottavio M, D'Ambrosio M, Lodovici M, Tredici MR, Luceri C. Preliminary data on the dietary safety, tolerability and effects on lipid metabolism of the marine microalga Tisochrysis lutea. ALGAL RES 2018. [DOI: 10.1016/j.algal.2018.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
14
|
Zhang Y, Higgins CB, Mayer AL, Mysorekar IU, Razani B, Graham MJ, Hruz PW, DeBosch BJ. TFEB-dependent induction of thermogenesis by the hepatocyte SLC2A inhibitor trehalose. Autophagy 2018; 14:1959-1975. [PMID: 29996716 PMCID: PMC6152536 DOI: 10.1080/15548627.2018.1493044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 02/08/2023] Open
Abstract
The macroautophagy/autophagy-inducing disaccharide, trehalose, has been proposed to be a promising therapeutic agent against neurodegenerative and cardiometabolic diseases. We recently showed that trehalose attenuates hepatic steatosis in part by blocking hepatocyte glucose transport to induce hepatocyte autophagic flux. However, although every major demonstration of trehalose action invokes activating autophagic flux as its primary function, the mechanism of action of trehalose in whole-body energy metabolism remains poorly defined. Here, we demonstrate that trehalose induces hepatocyte TFEB (transcription factor EB)-dependent thermogenesis in vivo, concomitant with upregulation of hepatic and white adipose expression of UCP1 (uncoupling protein 1 [mitochondrial, protein carrier]). Mechanistically, we provide evidence that hepatocyte fasting transcriptional and metabolic responses depend upon PPARGC1A (peroxisome proliferative activated receptor, gamma, coactivator 1 alpha), TFEB, and FGF21 (fibroblast growth factor 21) signaling. Strikingly, hepatocyte-selective TFEB knockdown abrogated trehalose induction of thermogenesis and white adipose tissue UCP1 upregulation in vivo. In contrast, we found that trehalose action on thermogenesis was independent of LEP (leptin) and the autophagy pathway, as there was robust thermogenic induction in trehalose-treated ob/ob, Becn1, Atg16l1, and Epg5 mutant mice. We conclude that trehalose induces metabolically favorable effects on whole-body thermogenesis in part via hepatocyte-centered fasting-like mechanisms that appear to be independent of autophagic flux. Our findings elucidate a novel mechanism by which trehalose acts as a metabolic therapeutic agent by activating hepatic fasting responses. More broadly, the hepatic glucose fasting response may be of clinical utility against overnutrition-driven disease, such as obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yiming Zhang
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Allyson L. Mayer
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Indira U. Mysorekar
- Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Babak Razani
- Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark J. Graham
- IONIS Pharmaceuticals, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul W. Hruz
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian J. DeBosch
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
15
|
Dallak MA. Acylated ghrelin induces but deacylated ghrelin prevents hepatic steatosis and insulin resistance in lean rats: Effects on DAG/ PKC/JNK pathway. Biomed Pharmacother 2018; 105:299-311. [PMID: 29860222 DOI: 10.1016/j.biopha.2018.05.098] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022] Open
Abstract
This study investigated the molecular effects of acylated (AG) and unacylated ghrelin (UAG) or their combination on hepatic lipogenesis pathways and DAG/PKC/JNK signaling in the livers of lean rats fed standard diet. Male rats (n = 10) were classified as control + vehicle (saline, 200 μl), AG, UAG, and AG + UAG-treated groups. All treatments were given at final doses of 200 ng/kg of for 14 days (twice/day, S.C). Administration of AG significantly enhanced circulatory levels of AG and UAG turning the normal ratio of AG/UAG from 1:2.5 to 1:1.2. However, while UAG didn't affect circulatory levels of AG, administration of UAG alone or in combination with AG resulted in AG/UAG ratios of 1:7 and 1:3, respectively. Independent of food intake nor the development of peripheral IR, AG increased hepatic DAG, TGs and CHOL contents and induced hepatic IR. Mechanism of action include 1) upregulation of mRNA and protein levels of DGAT-2 and mtGPAT-1, SREBP-1 and SCD-1, and 2) inhibition of fatty acids (FAs) oxidation mediated by inhibition of AMPK/ PPAR-α/CPT-1 axis. Consequently, AG induced membranous translocation of PKCδ and PKCε leading to activation of JNK and significant inhibition of insulin signaling under basal and insulin stimulation as evident by decreases in the phosphorylation levels of IRS (Tyr612) and Akt (Thr318) and increased phosphorylation of IRS (Ser307). However, while UAG only activated FAs oxidation in control rats, it reversed all alterations in all measured biochemical endpoints seen in the AG-treated group, when administered in combination with AG, leading to significant decreases in hepatic fat accumulation and prevention of hepatic IR. In conclusion, while exogenous administration of AG is at high risk of developing steatohepatitis and hepatic IR, co-administration of a balanced dose of UAG reduces this risk and inhibits hepatic lipid accumulation and enhance hepatic insulin signaling.
Collapse
Affiliation(s)
- Mohammad A Dallak
- Department of Physiology, College of Medicine, King's Khalid University, Abha, 61241, Saudi Arabia.
| |
Collapse
|
16
|
Yamamoto J, Imai J, Izumi T, Takahashi H, Kawana Y, Takahashi K, Kodama S, Kaneko K, Gao J, Uno K, Sawada S, Asano T, Kalinichenko VV, Susaki EA, Kanzaki M, Ueda HR, Ishigaki Y, Yamada T, Katagiri H. Neuronal signals regulate obesity induced β-cell proliferation by FoxM1 dependent mechanism. Nat Commun 2017; 8:1930. [PMID: 29208957 PMCID: PMC5717276 DOI: 10.1038/s41467-017-01869-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 10/23/2017] [Indexed: 12/17/2022] Open
Abstract
Under insulin-resistant conditions such as obesity, pancreatic β-cells proliferate to prevent blood glucose elevations. A liver–brain–pancreas neuronal relay plays an important role in this process. Here, we show the molecular mechanism underlying this compensatory β-cell proliferation. We identify FoxM1 activation in islets from neuronal relay-stimulated mice. Blockade of this relay, including vagotomy, inhibits obesity-induced activation of the β-cell FoxM1 pathway and suppresses β-cell expansion. Inducible β-cell-specific FoxM1 deficiency also blocks compensatory β-cell proliferation. In isolated islets, carbachol and PACAP/VIP synergistically promote β-cell proliferation through a FoxM1-dependent mechanism. These findings indicate that vagal nerves that release several neurotransmitters may allow simultaneous activation of multiple pathways in β-cells selectively, thereby efficiently promoting β-cell proliferation and maintaining glucose homeostasis during obesity development. This neuronal signal-mediated mechanism holds potential for developing novel approaches to regenerating pancreatic β-cells. Neuronal signals, in particular those transmitted via the vagal nerve, regulate both β-cell function and proliferation. Here, Yamamoto et al. show that the forkhead box M1 pathway is required for vagal signal-mediated induction of β-cell proliferation during obesity.
Collapse
Affiliation(s)
- Junpei Yamamoto
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| | - Tomohito Izumi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Hironori Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Yohei Kawana
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Junhong Gao
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Kenji Uno
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Shojiro Sawada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Tomoichiro Asano
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, Hiroshima, 734-8553, Japan
| | - Vladimir V Kalinichenko
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Etsuo A Susaki
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Osaka, 565-0871, Japan.,PRESTO, Japan Science and Technology Agency (JST), Kawaguchi, 332-0012, Japan
| | - Makoto Kanzaki
- Tohoku University Graduate School of Biomedical Engineering, Sendai, 980-8579, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Osaka, 565-0871, Japan
| | - Yasushi Ishigaki
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.,Division of Diabetes and Metabolism, Department of Internal Medicine, Iwate Medical University, Morioka, 020-8505, Japan
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.,Japan Agency for Medical Research and Development, Project for Elucidating and Controlling Mechanisms of Aging and Longevity, Tokyo, 100-0004, Japan.,Japan Agency for Medical Research and Development, CREST, Tokyo, 100-1004, Japan
| |
Collapse
|
17
|
Zingg JM, Hasan ST, Nakagawa K, Canepa E, Ricciarelli R, Villacorta L, Azzi A, Meydani M. Modulation of cAMP levels by high-fat diet and curcumin and regulatory effects on CD36/FAT scavenger receptor/fatty acids transporter gene expression. Biofactors 2017; 43:42-53. [PMID: 27355903 DOI: 10.1002/biof.1307] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/24/2016] [Accepted: 06/03/2016] [Indexed: 02/06/2023]
Abstract
Curcumin, a polyphenol from turmeric (Curcuma longa), reduces inflammation, atherosclerosis, and obesity in several animal studies. In Ldlr-/- mice fed a high-fat diet (HFD), curcumin reduces plasma lipid levels, therefore contributing to a lower accumulation of lipids and to reduced expression of fatty acid transport proteins (CD36/FAT, FABP4/aP2) in peritoneal macrophages. In this study, we analyzed the molecular mechanisms by which curcumin (500, 1000, 1500 mg/kg diet, for 4 months) may influence plasma and tissue lipid levels in Ldlr-/- mice fed an HFD. In liver, HFD significantly suppressed cAMP levels, and curcumin restored almost normal levels. Similar trends were observed in adipose tissues, but not in brain, skeletal muscle, spleen, and kidney. Treatment with curcumin increased phosphorylation of CREB in liver, what may play a role in regulatory effects of curcumin in lipid homeostasis. In cell lines, curcumin increased the level of cAMP, activated the transcription factor CREB and the human CD36 promoter via a sequence containing a consensus CREB response element. Regulatory effects of HFD and Cur on gene expression were observed in liver, less in skeletal muscle and not in brain. Since the cAMP/protein kinase A (PKA)/CREB pathway plays an important role in lipid homeostasis, energy expenditure, and thermogenesis by increasing lipolysis and fatty acid β-oxidation, an increase in cAMP levels induced by curcumin may contribute to its hypolipidemic and anti-atherosclerotic effects. © 2016 BioFactors, 43(1):42-53, 2017.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Vascular Biology Laboratory, JM USDA-Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Syeda T Hasan
- Vascular Biology Laboratory, JM USDA-Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Kiyotaka Nakagawa
- Vascular Biology Laboratory, JM USDA-Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Elisa Canepa
- Department of Experimental Medicine, Section of General Pathology, University of Genoa, Genoa, Italy
| | - Roberta Ricciarelli
- Department of Experimental Medicine, Section of General Pathology, University of Genoa, Genoa, Italy
| | - Luis Villacorta
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Angelo Azzi
- Vascular Biology Laboratory, JM USDA-Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Mohsen Meydani
- Vascular Biology Laboratory, JM USDA-Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| |
Collapse
|
18
|
Winn NC, Vieira-Potter VJ, Gastecki ML, Welly RJ, Scroggins RJ, Zidon TM, Gaines TL, Woodford ML, Karasseva NG, Kanaley JA, Sacks HS, Padilla J. Loss of UCP1 exacerbates Western diet-induced glycemic dysregulation independent of changes in body weight in female mice. Am J Physiol Regul Integr Comp Physiol 2016; 312:R74-R84. [PMID: 27881400 DOI: 10.1152/ajpregu.00425.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/15/2016] [Accepted: 11/17/2016] [Indexed: 01/23/2023]
Abstract
We tested the hypothesis that female mice null for uncoupling protein 1 (UCP1) would have increased susceptibility to Western diet-induced "whitening" of brown adipose tissue (AT) and glucose intolerance. Six-week-old C57BL/6J wild-type (WT) and UCP1 knockout (UCP1-/-) mice, housed at 25°C, were randomized to either a control diet (10% kcal from fat) or Western diet (45% kcal from fat and 1% cholesterol) for 28 wk. Loss of UCP1 had no effect on energy intake, energy expenditure, spontaneous physical activity, weight gain, or visceral white AT mass. Despite similar susceptibility to weight gain compared with WT, UCP1-/- exhibited whitening of brown AT evidenced by a striking ~500% increase in mass and appearance of large unilocular adipocytes, increased expression of genes related to inflammation, immune cell infiltration, and endoplasmic reticulum/oxidative stress (P < 0.05), and decreased mitochondrial subunit protein (COX I, II, III, and IV, P < 0.05), all of which were exacerbated by Western diet (P < 0.05). UCP1-/- mice also developed liver steatosis and glucose intolerance, which was worsened by Western diet. Collectively, these findings demonstrate that loss of UCP1 exacerbates Western diet-induced whitening of brown AT, glucose intolerance, and induces liver steatosis. Notably, the adverse metabolic manifestations of UCP1-/- were independent of changes in body weight, visceral adiposity, and energy expenditure. These novel findings uncover a previously unrecognized metabolic protective role of UCP1 that is independent of its already established role in energy homeostasis.
Collapse
Affiliation(s)
- Nathan C Winn
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Michelle L Gastecki
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Rebecca J Welly
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Rebecca J Scroggins
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Terese M Zidon
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - T'Keaya L Gaines
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Makenzie L Woodford
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Jill A Kanaley
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Harold S Sacks
- Endocrine and Diabetes Division, Veterans Greater Los Angeles Healthcare System and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; .,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and.,Child Health, University of Missouri, Columbia, Missouri
| |
Collapse
|
19
|
Farnesoid X Receptor Signaling Shapes the Gut Microbiota and Controls Hepatic Lipid Metabolism. mSystems 2016; 1:mSystems00070-16. [PMID: 27822554 PMCID: PMC5080402 DOI: 10.1128/msystems.00070-16] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023] Open
Abstract
The farnesoid X receptor (FXR) plays an important role in mediating the dialog between the host and gut microbiota, particularly through modulation of enterohepatic circulation of bile acids. Mounting evidence suggests that genetic ablation of Fxr in the gut or gut-restricted chemical antagonism of the FXR promotes beneficial health effects, including the prevention of nonalcoholic fatty liver disease in rodent models. However, questions remain unanswered, including whether modulation of FXR activity plays a role in shaping the gut microbiota community structure and function and what metabolic pathways of the gut microbiota contribute in an FXR-dependent manner to the host phenotype. In this report, new insights are gained into the metabolic contribution of the gut microbiota to the metabolic phenotypes, including establishing a link between FXR antagonism, bacterial bile salt hydrolase activity, and fermentation. Multiple approaches, including unique mouse models as well as metabolomics and genome-scale metabolic models, were employed to confirm these results. The gut microbiota modulates obesity and associated metabolic phenotypes in part through intestinal farnesoid X receptor (FXR) signaling. Glycine-β-muricholic acid (Gly-MCA), an intestinal FXR antagonist, has been reported to prevent or reverse high-fat diet (HFD)-induced and genetic obesity, insulin resistance, and fatty liver; however, the mechanism by which these phenotypes are improved is not fully understood. The current study investigated the influence of FXR activity on the gut microbiota community structure and function and its impact on hepatic lipid metabolism. Predictions about the metabolic contribution of the gut microbiota to the host were made using 16S rRNA-based PICRUSt (phylogenetic investigation of communities by reconstruction of unobserved states), then validated using 1H nuclear magnetic resonance-based metabolomics, and results were summarized by using genome-scale metabolic models. Oral Gly-MCA administration altered the gut microbial community structure, notably reducing the ratio of Firmicutes to Bacteroidetes and its PICRUSt-predicted metabolic function, including reduced production of short-chain fatty acids (substrates for hepatic gluconeogenesis and de novo lipogenesis) in the ceca of HFD-fed mice. Metabolic improvement was intestinal FXR dependent, as revealed by the lack of changes in HFD-fed intestine-specific Fxr-null (FxrΔIE) mice treated with Gly-MCA. Integrative analyses based on genome-scale metabolic models demonstrated an important link between Lactobacillus and Clostridia bile salt hydrolase activity and bacterial fermentation. Hepatic metabolite levels after Gly-MCA treatment correlated with altered levels of gut bacterial species. In conclusion, modulation of the gut microbiota by inhibition of intestinal FXR signaling alters host liver lipid metabolism and improves obesity-related metabolic dysfunction. IMPORTANCE The farnesoid X receptor (FXR) plays an important role in mediating the dialog between the host and gut microbiota, particularly through modulation of enterohepatic circulation of bile acids. Mounting evidence suggests that genetic ablation of Fxr in the gut or gut-restricted chemical antagonism of the FXR promotes beneficial health effects, including the prevention of nonalcoholic fatty liver disease in rodent models. However, questions remain unanswered, including whether modulation of FXR activity plays a role in shaping the gut microbiota community structure and function and what metabolic pathways of the gut microbiota contribute in an FXR-dependent manner to the host phenotype. In this report, new insights are gained into the metabolic contribution of the gut microbiota to the metabolic phenotypes, including establishing a link between FXR antagonism, bacterial bile salt hydrolase activity, and fermentation. Multiple approaches, including unique mouse models as well as metabolomics and genome-scale metabolic models, were employed to confirm these results.
Collapse
|
20
|
Mitochondrial biology, targets, and drug delivery. J Control Release 2015; 207:40-58. [PMID: 25841699 DOI: 10.1016/j.jconrel.2015.03.036] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 02/07/2023]
Abstract
In recent years, mitochondrial medicine has emerged as a new discipline resting at the intersection of mitochondrial biology, pathology, and pharmaceutics. The central role of mitochondria in critical cellular processes such as metabolism and apoptosis has placed mitochondria at the forefront of cell science. Advances in mitochondrial biology have revealed that these organelles continually undergo fusion and fission while functioning independently and in complex cellular networks, establishing direct membrane contacts with each other and with other organelles. Understanding the diverse cellular functions of mitochondria has contributed to understanding mitochondrial dysfunction in disease states. Polyplasmy and heteroplasmy contribute to mitochondrial phenotypes and associated dysfunction. Residing at the center of cell biology, cellular functions, and disease pathology and being laden with receptors and targets, mitochondria are beacons for pharmaceutical modification. This review presents the current state of mitochondrial medicine with a focus on mitochondrial function, dysfunction, and common disease; mitochondrial receptors, targets, and substrates; and mitochondrial drug design and drug delivery with a focus on the application of nanotechnology to mitochondrial medicine. Mitochondrial medicine is at the precipice of clinical translation; the objective of this review is to aid in the advancement of mitochondrial medicine from infancy to application.
Collapse
|
21
|
Tao H, Zhang Y, Zeng X, Shulman GI, Jin S. Niclosamide ethanolamine-induced mild mitochondrial uncoupling improves diabetic symptoms in mice. Nat Med 2014; 20:1263-9. [PMID: 25282357 PMCID: PMC4299950 DOI: 10.1038/nm.3699] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/21/2014] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes (T2D) has reached an epidemic level globally. Most current treatments ameliorate the hyperglycemic symptom of the disease but are not effective in correcting its underlying cause. One important causal factor of T2D is ectopic accumulation of lipids in metabolically sensitive organs such as liver and muscle. Mitochondrial uncoupling, which reduces cellular energy efficiency and increases lipid oxidation, is an appealing therapeutic strategy. The challenge, however, is to discover safe mitochondrial uncouplers for practical use. Niclosamide is an anthelmintic drug approved by the US Food and Drug Administration that uncouples the mitochondria of parasitic worms. Here we show that niclosamide ethanolamine salt (NEN) uncouples mammalian mitochondria at upper nanomolar concentrations. Oral NEN increases energy expenditure and lipid metabolism in mice. It is also efficacious in preventing and treating hepatic steatosis and insulin resistance induced by a high-fat diet. Moreover, it improves glycemic control and delays disease progression in db/db mice. Given the well-documented safety profile of NEN, our study provides a potentially new and practical pharmacological approach for treating T2D.
Collapse
Affiliation(s)
- Hanlin Tao
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Yong Zhang
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Xiangang Zeng
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
- Zhejiang Key Laboratory of Applied Enzymology, Yangtze Delta Region Research Institute of Tsinghua University, Jiaxing, Zhejiang 314006, China
| | - Gerald I. Shulman
- Howard Hughes Medical Institute, Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shengkan Jin
- Department of Pharmacology, Rutgers University-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| |
Collapse
|
22
|
Kondo K, Ishigaki Y, Gao J, Yamada T, Imai J, Sawada S, Muto A, Oka Y, Igarashi K, Katagiri H. Bach1 deficiency protects pancreatic β-cells from oxidative stress injury. Am J Physiol Endocrinol Metab 2013; 305:E641-8. [PMID: 23880309 DOI: 10.1152/ajpendo.00120.2013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BTB and CNC homology 1 (Bach1) is a transcriptional repressor of antioxidative enzymes, such as heme oxygenase-1 (HO-1). Oxidative stress is reportedly involved in insulin secretion impairment and obesity-associated insulin resistance. However, the role of Bach1 in the development of diabetes is unclear. HO-1 expression in the liver, white adipose tissue, and pancreatic islets was markedly upregulated in Bach1-deficient mice. Unexpectedly, glucose and insulin tolerance tests showed no differences in obese wild-type (WT) and obese Bach1-deficient mice after high-fat diet loading for 6 wk, suggesting minimal roles of Bach1 in the development of insulin resistance. In contrast, Bach1 deficiency significantly suppressed alloxan-induced pancreatic insulin content reduction and the resultant glucose elevation. Furthermore, TUNEL-positive cells in pancreatic islets of Bach1-deficient mice were markedly decreased, by 60%, compared with those in WT mice. HO-1 expression in islets was significantly upregulated in alloxan-injected Bach1-deficient mice, whereas expression of other antioxidative enzymes, e.g., catalase, superoxide dismutase, and glutathione peroxidase, was not changed by either alloxan administration or Bach1 deficiency. Our results suggest that Bach1 deficiency protects pancreatic β-cells from oxidative stress-induced apoptosis and that the enhancement of HO-1 expression plays an important role in this protection.
Collapse
Affiliation(s)
- Keiichi Kondo
- Division of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Tsukita S, Yamada T, Uno K, Takahashi K, Kaneko K, Ishigaki Y, Imai J, Hasegawa Y, Sawada S, Ishihara H, Oka Y, Katagiri H. Hepatic glucokinase modulates obesity predisposition by regulating BAT thermogenesis via neural signals. Cell Metab 2012; 16:825-32. [PMID: 23217261 DOI: 10.1016/j.cmet.2012.11.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 08/01/2012] [Accepted: 11/14/2012] [Indexed: 10/27/2022]
Abstract
Considering the explosive increase in obesity worldwide, there must be an unknown mechanism(s) promoting energy accumulation under conditions of overnutrition. We identified a feed-forward mechanism favoring energy storage, originating in hepatic glucokinase (GK) upregulation. High-fat feeding induced hepatic GK upregulation, and hepatic GK overexpression dose-dependently decreased adaptive thermogenesis by downregulating thermogenesis-related genes in brown adipose tissue (BAT). This intertissue (liver-to-BAT) system consists of the afferent vagus from the liver and sympathetic efferents from the medulla and antagonizes anti-obesity effects of leptin on thermogenesis. Furthermore, upregulation of endogenous GK in the liver by high-fat feeding was more marked in obesity-prone than in obesity-resistant strains and was inversely associated with BAT thermogenesis. Hepatic GK overexpression in obesity-resistant mice promoted weight gain, while hepatic GK knockdown in obesity-prone mice attenuated weight gain with increased adaptive thermogenesis. Thus, this intertissue energy-saving system may contribute to determining obesity predisposition.
Collapse
Affiliation(s)
- Sohei Tsukita
- Department of Metabolic Diseases, Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Saito T, Hasegawa Y, Ishigaki Y, Yamada T, Gao J, Imai J, Uno K, Kaneko K, Ogihara T, Shimosawa T, Asano T, Fujita T, Oka Y, Katagiri H. Importance of endothelial NF-κB signalling in vascular remodelling and aortic aneurysm formation. Cardiovasc Res 2012; 97:106-14. [PMID: 23015640 DOI: 10.1093/cvr/cvs298] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
AIMS Vascular remodelling and aortic aneurysm formation are induced mainly by inflammatory responses in the adventitia and media. However, relatively little is known about the mechanistic significance of endothelium in the pathogenesis of these vascular disorders. The transcription factor nuclear factor-kappa B (NF-κB) regulates the expressions of numerous genes, including those related to pro-inflammatory responses. Therefore, to investigate the roles of endothelial pro-inflammatory responses, we examined the impact of blocking endothelial NF-κB signalling on intimal hyperplasia and aneurysm formation. METHODS AND RESULTS To block endothelial NF-κB signalling, we used transgenic mice expressing dominant-negative IκBα selectively in endothelial cells (E-DNIκB mice). E-DNIκB mice were protected from the development of cuff injury-induced neointimal formation, in association with suppressed arterial expressions of cellular adhesion molecules, a macrophage marker, and inflammatory factors. In addition, the blockade of endothelial NF-κB signalling prevented abdominal aortic aneurysm formation in an experimental model, hypercholesterolaemic apolipoprotein E-deficient mice with angiotensin II infusion. In this aneurysm model as well, aortic expressions of an adhesion molecule, a macrophage marker, and inflammatory factors were suppressed with the inhibited expression and activity of matrix metalloproteinases in the aorta. CONCLUSION Endothelial NF-κB activation up-regulates adhesion molecule expression, which may trigger macrophage infiltration and inflammation in the adventitia and media. Thus, the endothelium plays important roles in vascular remodelling and aneurysm formation through its intracellular NF-κB signalling.
Collapse
Affiliation(s)
- Tokuo Saito
- Department of Metabolic Diseases, Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Moya M, Benet M, Guzmán C, Tolosa L, García-Monzón C, Pareja E, Castell JV, Jover R. Foxa1 reduces lipid accumulation in human hepatocytes and is down-regulated in nonalcoholic fatty liver. PLoS One 2012; 7:e30014. [PMID: 22238690 PMCID: PMC3253125 DOI: 10.1371/journal.pone.0030014] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 12/08/2011] [Indexed: 02/06/2023] Open
Abstract
Triglyceride accumulation in nonalcoholic fatty liver (NAFL) results from unbalanced lipid metabolism which, in the liver, is controlled by several transcription factors. The Foxa subfamily of winged helix/forkhead box (Fox) transcription factors comprises three members which play important roles in controlling both metabolism and homeostasis through the regulation of multiple target genes in the liver, pancreas and adipose tissue. In the mouse liver, Foxa2 is repressed by insulin and mediates fasting responses. Unlike Foxa2 however, the role of Foxa1 in the liver has not yet been investigated in detail. In this study, we evaluate the role of Foxa1 in two human liver cell models, primary cultured hepatocytes and HepG2 cells, by adenoviral infection. Moreover, human and rat livers were analyzed to determine Foxa1 regulation in NAFL. Results demonstrate that Foxa1 is a potent inhibitor of hepatic triglyceride synthesis, accumulation and secretion by repressing the expression of multiple target genes of these pathways (e.g., GPAM, DGAT2, MTP, APOB). Moreover, Foxa1 represses the fatty acid transporter protein FATP2 and lowers fatty acid uptake. Foxa1 also increases the breakdown of fatty acids by inducing peroxisomal fatty acid β-oxidation and ketone body synthesis. Finally, Foxa1 is able to largely up-regulate UCP1, thereby dissipating energy and consistently decreasing the mitochondria membrane potential. We also report that human and rat NAFL have a reduced Foxa1 expression, possibly through a protein kinase C-dependent pathway. We conclude that Foxa1 is an antisteatotic factor that coordinately tunes several lipid metabolic pathways to block triglyceride accumulation in hepatocytes. However, Foxa1 is down-regulated in human and rat NAFL and, therefore, increasing Foxa1 levels could protect from steatosis. Altogether, we suggest that Foxa1 could be a novel therapeutic target for NAFL disease and insulin resistance.
Collapse
Affiliation(s)
- Marta Moya
- Experimental Hepatology Unit, University Hospital La Fe, Valencia, Spain
| | - Marta Benet
- Experimental Hepatology Unit, University Hospital La Fe, Valencia, Spain
- CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain
| | - Carla Guzmán
- Experimental Hepatology Unit, University Hospital La Fe, Valencia, Spain
| | - Laia Tolosa
- Experimental Hepatology Unit, University Hospital La Fe, Valencia, Spain
| | - Carmelo García-Monzón
- CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain
- Liver Research Unit, Instituto de Investigación Sanitaria Princesa, University Hospital Santa Cristina, Madrid, Spain
| | - Eugenia Pareja
- Surgery and Liver Transplantation Unit, University Hospital La Fe, Valencia, Spain
| | - José Vicente Castell
- Experimental Hepatology Unit, University Hospital La Fe, Valencia, Spain
- CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
| | - Ramiro Jover
- Experimental Hepatology Unit, University Hospital La Fe, Valencia, Spain
- CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
26
|
Klaus S, Keipert S, Rossmeisl M, Kopecky J. Augmenting energy expenditure by mitochondrial uncoupling: a role of AMP-activated protein kinase. GENES AND NUTRITION 2011; 7:369-86. [PMID: 22139637 DOI: 10.1007/s12263-011-0260-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 11/18/2011] [Indexed: 11/28/2022]
Abstract
Strategies to prevent and treat obesity aim to decrease energy intake and/or increase energy expenditure. Regarding the increase of energy expenditure, two key intracellular targets may be considered (1) mitochondrial oxidative phosphorylation, the major site of ATP production, and (2) AMP-activated protein kinase (AMPK), the master regulator of cellular energy homeostasis. Experiments performed mainly in transgenic mice revealed a possibility to ameliorate obesity and associated disorders by mitochondrial uncoupling in metabolically relevant tissues, especially in white adipose tissue (WAT), skeletal muscle (SM), and liver. Thus, ectopic expression of brown fat-specific mitochondrial uncoupling protein 1 (UCP1) elicited major metabolic effects both at the cellular/tissue level and at the whole-body level. In addition to expected increases in energy expenditure, surprisingly complex phenotypic effects were detected. The consequences of mitochondrial uncoupling in WAT and SM are not identical, showing robust and stable obesity resistance accompanied by improvement of lipid metabolism in the case of ectopic UCP1 in WAT, while preservation of insulin sensitivity in the context of high-fat feeding represents the major outcome of muscle UCP1 expression. These complex responses could be largely explained by tissue-specific activation of AMPK, triggered by a depression of cellular energy charge. Experimental data support the idea that (1) while being always activated in response to mitochondrial uncoupling and compromised intracellular energy status in general, AMPK could augment energy expenditure and mediate local as well as whole-body effects; and (2) activation of AMPK alone does not lead to induction of energy expenditure and weight reduction.
Collapse
Affiliation(s)
- Susanne Klaus
- German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | | | | | | |
Collapse
|
27
|
Isoleucine or valine deprivation stimulates fat loss via increasing energy expenditure and regulating lipid metabolism in WAT. Amino Acids 2011; 43:725-34. [DOI: 10.1007/s00726-011-1123-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 10/07/2011] [Indexed: 01/14/2023]
|
28
|
Gao J, Ishigaki Y, Yamada T, Kondo K, Yamaguchi S, Imai J, Uno K, Hasegawa Y, Sawada S, Ishihara H, Oyadomari S, Mori M, Oka Y, Katagiri H. Involvement of endoplasmic stress protein C/EBP homologous protein in arteriosclerosis acceleration with augmented biological stress responses. Circulation 2011; 124:830-9. [PMID: 21810656 DOI: 10.1161/circulationaha.110.014050] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND The processes of arteriosclerosis, including atherosclerosis and vascular remodeling, are affected by interactions among numerous biological pathways such as responses to inflammation, oxidative stress, and endoplasmic reticulum stress. C/EBP homologous protein (CHOP), which is well known to induce cellular apoptosis in response to severe endoplasmic reticulum stress, is reportedly upregulated in plaque lesions. METHODS AND RESULTS We examined the effects of CHOP deficiency on 2 types of arteriosclerosis: cuff injury-induced neointimal formation and hypercholesterolemia-induced atherosclerosis. Cuff injury-induced neointimal formation was markedly inhibited in CHOP(-/-) mice with suppressed aortic expression of inflammatory factors and smooth muscle cell proliferation-related proteins. A CHOP deficiency also inhibited aortic plaque formation in hypercholesterolemic apolipoprotein E(-/-) mice with suppressed aortic expression of inflammatory factors and oxidative stress markers. Bone marrow transplantation experiments revealed that recipient CHOP deficiency significantly suppressed both cuff injury-induced neointimal formation and hypercholesterolemia-induced atherosclerotic plaque formation to a greater extent than donor CHOP deficiency, suggesting the importance of CHOP in vascular cells for arteriosclerosis progression. Furthermore, in our in vitro experiments, in not only macrophages but also endothelial and smooth muscle cell lines, endoplasmic reticulum stress inducers upregulated inflammation-, adhesion-, or smooth muscle cell proliferation-related proteins, whereas decreased CHOP expression remarkably suppressed endoplasmic reticulum stress-induced upregulation of these proteins. CONCLUSIONS In addition to the well-known signaling for apoptosis induction, CHOP may play important roles in augmenting potentially pathological biological stress responses. This noncanonical role of CHOP, especially that expressed in vascular cells, may contribute to the progression of vascular remodeling and atherosclerosis.
Collapse
Affiliation(s)
- Junhong Gao
- Department of Metabolic Diseases, Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Nakatani M, Kokubo M, Ohsawa Y, Sunada Y, Tsuchida K. Follistatin-derived peptide expression in muscle decreases adipose tissue mass and prevents hepatic steatosis. Am J Physiol Endocrinol Metab 2011; 300:E543-53. [PMID: 21205933 DOI: 10.1152/ajpendo.00430.2010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myostatin, a member of the transforming growth factor (TGF)-β superfamily, plays a potent inhibitory role in regulating skeletal muscle mass. Inhibition of myostatin by gene disruption, transgenic (Tg) expression of myostatin propeptide, or injection of propeptide or myostatin antibodies causes a widespread increase in skeletal muscle mass. Several peptides, in addition to myostatin propeptide and myostatin antibodies, can bind directly to and neutralize the activity of myostatin. These include follistatin and follistatin-related gene. Overexpression of follistatin or follistatin-related gene in mice increased the muscle mass as in myostatin knockout mice. Follistatin binds to myostatin but also binds to and inhibits other members of the TGF-β superfamily, notably activins. Therefore, follistatin regulates both myostatin and activins in vivo. We previously reported the development and characterization of several follistatin-derived peptides, including FS I-I (Nakatani M, Takehara Y, Sugino H, Matsumoto M, Hashimoto O, Hasegawa Y, Murakami T, Uezumi A, Takeda S, Noji S, Sunada Y, Tsuchida K. FASEB J 22: 477-487, 2008). FS I-I retained myostatin-inhibitory activity without affecting the bioactivity of activins. Here, we found that inhibition of myostatin increases skeletal muscle mass and decreases fat accumulation in FS I-I Tg mice. FS I-I Tg mice also showed decreased fat accumulation even on a control diet. Interestingly, the adipocytes in FS I-I Tg mice were much smaller than those of wild-type mice. Furthermore, FS I-I Tg mice were resistant to high-fat diet-induced obesity and hepatic steatosis and had lower hepatic fatty acid levels and altered fatty acid composition compared with control mice. FS I-I Tg mice have improved glucose tolerance when placed on a high-fat diet. These data indicate that inhibiting myostatin with a follistatin-derived peptide provides a novel therapeutic option to decrease adipocyte size, prevent obesity and hepatic steatosis, and improve glucose tolerance.
Collapse
Affiliation(s)
- Masashi Nakatani
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Aichi, Japan
| | | | | | | | | |
Collapse
|
30
|
Suzuki T, Imai J, Yamada T, Ishigaki Y, Kaneko K, Uno K, Hasegawa Y, Ishihara H, Oka Y, Katagiri H. Interleukin-6 enhances glucose-stimulated insulin secretion from pancreatic beta-cells: potential involvement of the PLC-IP3-dependent pathway. Diabetes 2011; 60:537-47. [PMID: 21270264 PMCID: PMC3028353 DOI: 10.2337/db10-0796] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Interleukin-6 (IL-6) has a significant impact on glucose metabolism. However, the effects of IL-6 on insulin secretion from pancreatic β-cells are controversial. Therefore, we analyzed IL-6 effects on pancreatic β-cell functions both in vivo and in vitro. RESEARCH DESIGN AND METHODS First, to examine the effects of IL-6 on in vivo insulin secretion, we expressed IL-6 in the livers of mice using the adenoviral gene transfer system. In addition, using both MIN-6 cells, a murine β-cell line, and pancreatic islets isolated from mice, we analyzed the in vitro effects of IL-6 pretreatment on insulin secretion. Furthermore, using pharmacological inhibitors and small interfering RNAs, we studied the intracellular signaling pathway through which IL-6 may affect insulin secretion from MIN-6 cells. RESULTS Hepatic IL-6 expression raised circulating IL-6 and improved glucose tolerance due to enhancement of glucose stimulated-insulin secretion (GSIS). In addition, in both isolated pancreatic islets and MIN-6 cells, 24-h pretreatment with IL-6 significantly enhanced GSIS. Furthermore, pretreatment of MIN-6 cells with phospholipase C (PLC) inhibitors with different mechanisms of action, U-73122 and neomycin, and knockdowns of the IL-6 receptor and PLC-β(1), but not with a protein kinase A inhibitor, H-89, inhibited IL-6-induced enhancement of GSIS. An inositol triphosphate (IP(3)) receptor antagonist, Xestospondin C, also abrogated the GSIS enhancement induced by IL-6. CONCLUSIONS The results obtained from both in vivo and in vitro experiments strongly suggest that IL-6 acts directly on pancreatic β-cells and enhances GSIS. The PLC-IP(3)-dependent pathway is likely to be involved in IL-6-mediated enhancements of GSIS.
Collapse
Affiliation(s)
- Toshinobu Suzuki
- Department of Metabolic Diseases, Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Junta Imai
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Tetsuya Yamada
- Department of Metabolic Diseases, Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Yasushi Ishigaki
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Keizo Kaneko
- Department of Metabolic Diseases, Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Kenji Uno
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Yutaka Hasegawa
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Hisamitsu Ishihara
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Yoshitomo Oka
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Hideki Katagiri
- Department of Metabolic Diseases, Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
- Corresponding author: Hideki Katagiri,
| |
Collapse
|
31
|
Susaki E, Kaneko-Oshikawa C, Miyata K, Tabata M, Yamada T, Oike Y, Katagiri H, Nakayama KI. Increased E4 activity in mice leads to ubiquitin-containing aggregates and degeneration of hypothalamic neurons resulting in obesity. J Biol Chem 2010; 285:15538-15547. [PMID: 20190229 DOI: 10.1074/jbc.m110.105841] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Obesity has become a serious worldwide public health problem. Although neural degeneration in specific brain regions has been suggested to contribute to obesity phenotype in humans, a causal relationship between these two conditions has not been demonstrated experimentally. We now show that E4B (also known as UFD2a), a mammalian ubiquitin chain elongation factor (E4), induces the formation of intracellular aggregates positive for ubiquitin and the adaptor protein p62 when overexpressed in cultured cells or the brain. Mice transgenic for E4B manifested neural degeneration in association with aggregate formation, and they exhibited functional impairment specifically in a subset of hypothalamic neurons that regulate food intake and energy expenditure, resulting in development of hyperphagic obesity and related metabolic abnormalities. The neural pathology of E4B transgenic mice was similar to that of human neurodegenerative diseases associated with the formation of intracellular ubiquitin-positive deposits, indicating the existence of a link between such diseases and obesity and related metabolic disorders. Our findings thus provide experimental evidence for a role of hypothalamic neurodegeneration in obesity, and the E4B transgenic mouse should prove to be a useful animal model for studies of the relationship between neurodegenerative diseases and obesity.
Collapse
Affiliation(s)
- Etsuo Susaki
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012
| | - Chie Kaneko-Oshikawa
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556
| | - Mitsuhisa Tabata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556
| | - Tetsuya Yamada
- Division of Advanced Therapeutics for Metabolic Diseases, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556
| | - Hideki Katagiri
- Division of Advanced Therapeutics for Metabolic Diseases, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012.
| |
Collapse
|
32
|
Min L, Yanase T, Tanaka T, Fan W, Nomura M, Kawate H, Okabe T, Takayanagi R, Nawata H. A novel synthetic androgen receptor ligand, S42, works as a selective androgen receptor modulator and possesses metabolic effects with little impact on the prostate. Endocrinology 2009; 150:5606-16. [PMID: 19854864 DOI: 10.1210/en.2009-0405] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We identified a novel synthetic steroid, S42, as a promising candidate of selective androgen receptor (AR) modulator. Results of the whole-cell binding assay using COS-7 cells exogenously expressing various steroid receptors indicated that S42 specifically binds to AR and progesterone receptor. When orchiectomized Sprague Dawley rats were administered with S42 for 3 wk, the muscle weight of the levator ani was increased as markedly as that induced by 5alpha-dihydrotestosterone (DHT), but the weight of the prostate was not elevated at any doses in contrast to DHT. The plasma concentrations of gonadotropin and adiponectin, those down-regulated by DHT, were unaffected by S42. In addition, although the plasma triglyceride level was unaffected by DHT, it was significantly reduced by S42. This effect of S42 was associated with suppression of the SRBP-1c-mediated lipogenic and insulin-desensitizing pathway in the liver and visceral fat. Taken together, S42 works as an AR agonist in muscle and as an AR antagonist in the prostate, pituitary gland, and liver, accompanying beneficial potentials on lipid metabolism.
Collapse
Affiliation(s)
- Liu Min
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Begriche K, Massart J, Fromenty B. Effects of β-aminoisobutyric acid on leptin production and lipid homeostasis: mechanisms and possible relevance for the prevention of obesity. Fundam Clin Pharmacol 2009; 24:269-82. [DOI: 10.1111/j.1472-8206.2009.00765.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
34
|
Li GY, Li CP. Advances in research on the role of phosphatidylinositol-3 kinase in the pathogenesis of nonalcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2009; 17:2277-2282. [DOI: 10.11569/wcjd.v17.i22.2277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Due to improvement in living standards, changes in eating habits, as well as a sharp increase in the incidence of diabetes and metabolic syndrome, the incidence of nonalcoholic fatty liver disease is continuously increasing. Recent studies have shown that the phosphatidylinositol 3-kinase (PI-3K) pathway plays a crucial role in the pathogenesis of nonalcoholic fatty liver disease. In this article, we will review the role of PI-3K in the pathogenesis of nonalcoholic fatty liver disease.
Collapse
|
35
|
Dean JT, Tran L, Beaven S, Tontonoz P, Reue K, Dipple KM, Liao JC. Resistance to diet-induced obesity in mice with synthetic glyoxylate shunt. Cell Metab 2009; 9:525-36. [PMID: 19490907 PMCID: PMC4277884 DOI: 10.1016/j.cmet.2009.04.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 03/17/2009] [Accepted: 04/23/2009] [Indexed: 10/20/2022]
Abstract
Given the success in engineering synthetic phenotypes in microbes and mammalian cells, constructing non-native pathways in mammals has become increasingly attractive for understanding and identifying potential targets for treating metabolic disorders. Here, we introduced the glyoxylate shunt into mouse liver to investigate mammalian fatty acid metabolism. Mice expressing the shunt showed resistance to diet-induced obesity on a high-fat diet despite similar food consumption. This was accompanied by a decrease in total fat mass, circulating leptin levels, plasma triglyceride concentration, and a signaling metabolite in liver, malonyl-CoA, that inhibits fatty acid degradation. Contrary to plants and bacteria, in which the glyoxylate shunt prevents the complete oxidation of fatty acids, this pathway when introduced in mice increases fatty acid oxidation such that resistance to diet-induced obesity develops. This work suggests that using non-native pathways in higher organisms to explore and modulate metabolism may be a useful approach.
Collapse
Affiliation(s)
- Jason T Dean
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Labruna G, Pasanisi F, Nardelli C, Tarantino G, Vitale DF, Bracale R, Finelli C, Genua MP, Contaldo F, Sacchetti L. UCP1 -3826 AG+GG genotypes, adiponectin, and leptin/adiponectin ratio in severe obesity. J Endocrinol Invest 2009; 32:525-529. [PMID: 19474520 DOI: 10.1007/bf03346500] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver disease (NAFLD) and metabolic syndrome (MS) are well-recognized complications of obesity. This study was designed to evaluate the role of the UCP1 -3826 A>G polymorphism, adiponectin levels, leptin/adiponectin ratio (L/A), and main biochemical parameters in 102 unrelated severely obese adults [61 females and 41 males, median body mass index (BMI) = 47.8 kg/m2] with NAFLD, with (MS+) or without MS (MS-) from Southern Italy. SUBJECT AND METHODS The UCP1 polymorphism was tested by the TaqMan method, main biochemical parameters by routinary methods, adiponectin, and leptin serum levels by enzyme-linked immunosorbent assay. MS was diagnosed according to the American Heart Association criteria, liver steatosis was detected by ultrasound. RESULTS MS was present in 53% male and 66% female obese patients. Only total cholesterol (p=0.04 males and p=0.002 females) and L/A ratio (p=0.03 males) differed between MS+ and MS- obese patients. At multivariate analysis, severe liver steatosis was significantly associated with: UCP1 (AG+GG) genotypes [odds ratio-confidence interval (OR-CI): 4.25; 1.12-16.13], MS (OR-CI: 8.47; 1.78-40.25), low adiponectin levels (OR-CI: 0.92; 0.87-0.98), high alanine aminotransferase levels (OR-CI: 1.03; 1.00-1.06), age (ORCI: 1.08; 1.00-1.15), and male gender (OR-CI: 10.78; 1.61- 71.96). CONCLUSION In addition to traditional factors, total cholesterol and L/A ratio appear to contribute to MS characterization in severe obesity. Furthermore, the UCP1 (AG+GG) genotypes and low adiponectin levels could predispose to a more severe liver steatosis independently of MS presence. Based on our data, polymorphic UCP1 (AG+GG) obese patients with low adiponectin levels appear to be high-risk subjects for worsening of liver steatosis, a NAFLD, possibly requiring a second-step evaluation by liver biopsy.
Collapse
Affiliation(s)
- G Labruna
- CEINGE Biotecnologie Avanzate S.C. a R.L., Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Grunnet LG, Brøns C, Jacobsen S, Nilsson E, Astrup A, Hansen T, Pedersen O, Poulsen P, Quistorff B, Vaag A. Increased recovery rates of phosphocreatine and inorganic phosphate after isometric contraction in oxidative muscle fibers and elevated hepatic insulin resistance in homozygous carriers of the A-allele of FTO rs9939609. J Clin Endocrinol Metab 2009; 94:596-602. [PMID: 18984658 DOI: 10.1210/jc.2008-1592] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Recent studies identified the rs9939609 A-allele of the FTO (fat mass and obesity associated) gene as being associated with obesity and type 2 diabetes. We studied the role of the A-allele in the regulation of peripheral organ functions involved in the pathogenesis of obesity and type 2 diabetes. METHODS Forty-six young men underwent a hyperinsulinemic euglycemic clamp with excision of skeletal muscle biopsies, an iv glucose tolerance test, 31phosphorous magnetic resonance spectroscopy, and 24-h whole body metabolism was measured in a respiratory chamber. RESULTS The FTO rs9939609 A-allele was associated with elevated fasting blood glucose and plasma insulin, hepatic insulin resistance, and shorter recovery half-times of phosphocreatine and inorganic phosphate after exercise in a primarily type I muscle. These relationships--except for fasting insulin--remained significant after correction for body fat percentage. The risk allele was not associated with fat distribution, peripheral insulin sensitivity, insulin secretion, 24-h energy expenditure, or glucose and fat oxidation. The FTO genotype did not influence the mRNA expression of FTO or a set of key nuclear or mitochondrially encoded genes in skeletal muscle during rest. CONCLUSION Increased energy efficiency--and potentially increased mitochondrial coupling--as suggested by faster recovery rates of phosphocreatine and inorganic phosphate in oxidative muscle fibers may contribute to the increased risk of obesity and type 2 diabetes in homozygous carriers of the FTO A-risk allele. Hepatic insulin resistance may represent the key metabolic defect responsible for mild elevations of fasting blood glucose associated with the FTO phenotype.
Collapse
Affiliation(s)
- Louise G Grunnet
- Steno Diabetes Center, Niels Steensens vej 2, 2820 Gentofte, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ishigaki Y, Katagiri H, Gao J, Yamada T, Imai J, Uno K, Hasegawa Y, Kaneko K, Ogihara T, Ishihara H, Sato Y, Takikawa K, Nishimichi N, Matsuda H, Sawamura T, Oka Y. Impact of plasma oxidized low-density lipoprotein removal on atherosclerosis. Circulation 2008; 118:75-83. [PMID: 18559699 DOI: 10.1161/circulationaha.107.745174] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Several clinical studies of statin therapy have demonstrated that lowering low-density lipoprotein (LDL) cholesterol prevents atherosclerotic progression and decreases cardiovascular mortality. In addition, oxidized LDL (oxLDL) is suggested to play roles in the formation and progression of atherosclerosis. However, whether lowering oxLDL alone, rather than total LDL, affects atherogenesis remains unclear. METHODS AND RESULTS To clarify the atherogenic impact of oxLDL, lectin-like oxLDL receptor 1 (LOX-1), an oxLDL receptor, was expressed ectopically in the liver with adenovirus administration in apolipoprotein E-deficient mice at 46 weeks of age. Hepatic LOX-1 expression enhanced hepatic oxLDL uptake, indicating functional expression of LOX-1 in the liver. Although plasma total cholesterol, triglyceride, and LDL cholesterol levels were unaffected, plasma oxLDL was markedly and transiently decreased in LOX-1 mice. In controls, atherosclerotic lesions, detected by Oil Red O staining, were markedly increased (by 38%) during the 4-week period after adenoviral administration. In contrast, atherosclerotic progression was almost completely inhibited by hepatic LOX-1 expression. In addition, plasma monocyte chemotactic protein-1 and lipid peroxide levels were decreased, whereas adiponectin was increased, suggesting decreased systemic oxidative stress. Thus, LOX1 expressed in the livers of apolipoprotein E-deficient mice transiently removes oxLDL from circulating blood and possibly decreases systemic oxidative stress, resulting in complete prevention of atherosclerotic progression despite the persistence of severe LDL hypercholesterolemia and hypertriglyceridemia. CONCLUSIONS OxLDL has a major atherogenic impact, and oxLDL removal is a promising therapeutic strategy against atherosclerosis.
Collapse
Affiliation(s)
- Yasushi Ishigaki
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Nonalcoholic fatty liver disease is a spectrum of diseases ranging from simple steatosis to cirrhosis. The hallmark of nonalcoholic fatty liver disease is hepatocyte accumulation of triglycerides. We will review the role of triglyceride synthesis in nonalcoholic fatty liver disease progression and summarize recent findings about triglyceride synthesis inhibition and prevention of progressive disease. RECENT FINDINGS Attempts to inhibit triglyceride synthesis in animal models have resulted in improvement in hepatic steatosis. Studies in animal models of nonalcoholic fatty liver disease demonstrate that inhibition of acyl-coenzyme A:diacylglycerol acyltransferase, the enzyme that catalyzes the final step in triglyceride synthesis, results in improvement in hepatic steatosis and insulin sensitivity. We recently confirmed that hepatic specific inhibition of acyl-coenzyme A:diacylglycerol acyltransferase with antisense oligonucleotides improves hepatic steatosis in obese, diabetic mice but, unexpectedly, exacerbated injury and fibrosis in that model of progressive nonalcoholic fatty liver disease. When hepatocyte triglyceride synthesis was inhibited, free fatty acids accumulated in the liver, leading to induction of fatty acid oxidizing systems that increased hepatic oxidative stress and liver damage. These findings suggest that the ability to synthesize triglycerides may, in fact, be protective in obesity. SUMMARY Nonalcoholic fatty liver disease is strongly associated with obesity and peripheral insulin resistance. Peripheral insulin resistance increases lipolysis in adipose depots, promoting increased free fatty acid delivery to the liver. In states of energy excess, such as obesity, the latter normally triggers hepatic triglyceride synthesis. When hepatic triglyceride synthesis is unable to accommodate increased hepatocyte free fatty acid accumulation, however, lipotoxicity results. Thus, rather than being hepatotoxic, liver triglyceride accumulation is actually hepato-protective in obese, insulin-resistant individuals.
Collapse
Affiliation(s)
- Steve S Choi
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
40
|
Stefanovic-Racic M, Perdomo G, Mantell BS, Sipula IJ, Brown NF, O'Doherty RM. A moderate increase in carnitine palmitoyltransferase 1a activity is sufficient to substantially reduce hepatic triglyceride levels. Am J Physiol Endocrinol Metab 2008; 294:E969-77. [PMID: 18349115 DOI: 10.1152/ajpendo.00497.2007] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), hypertriglyceridemia, and elevated free fatty acids are present in the majority of patients with metabolic syndrome and type 2 diabetes mellitus and are strongly associated with hepatic insulin resistance. In the current study, we tested the hypothesis that an increased rate of fatty acid oxidation in liver would prevent the potentially harmful effects of fatty acid elevation, including hepatic triglyceride (TG) accumulation and elevated TG secretion. Primary rat hepatocytes were transduced with adenovirus encoding carnitine palmitoyltransferase 1a (Adv-CPT-1a) or control adenoviruses encoding either beta-galactosidase (Adv-beta-gal) or carnitine palmitoyltransferase 2 (Adv-CPT-2). Overexpression of CPT-1a increased the rate of beta-oxidation and ketogenesis by approximately 70%, whereas esterification of exogenous fatty acids and de novo lipogenesis were unchanged. Importantly, CPT-1a overexpression was accompanied by a 35% reduction in TG accumulation and a 60% decrease in TG secretion by hepatocytes. There were no changes in secretion of apolipoprotein B (apoB), suggesting the synthesis of smaller, less atherogenic VLDL particles. To evaluate the effect of increasing hepatic CPT-1a activity in vivo, we injected lean or obese male rats with Adv-CPT-1a, Adv-beta-gal, or Adv-CPT-2. Hepatic CPT-1a activity was increased by approximately 46%, and the rate of fatty acid oxidation was increased by approximately 44% in lean and approximately 36% in obese CPT-1a-overexpressing animals compared with Adv-CPT-2- or Adv-beta-gal-treated rats. Similar to observations in vitro, liver TG content was reduced by approximately 37% (lean) and approximately 69% (obese) by this in vivo intervention. We conclude that a moderate stimulation of fatty acid oxidation achieved by an increase in CPT-1a activity is sufficient to substantially reduce hepatic TG accumulation both in vitro and in vivo. Therefore, interventions that increase CPT-1a activity could have potential benefits in the treatment of NAFLD.
Collapse
Affiliation(s)
- Maja Stefanovic-Racic
- Department of Medicine, Division of Endocrinology, University of Pittsburg, Pittsburgh, PA, USA.
| | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Yamada T, Oka Y, Katagiri H. Inter-organ metabolic communication involved in energy homeostasis: potential therapeutic targets for obesity and metabolic syndrome. Pharmacol Ther 2007; 117:188-98. [PMID: 18006064 DOI: 10.1016/j.pharmthera.2007.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 09/05/2007] [Indexed: 01/16/2023]
Abstract
The global rate of obesity is rising alarmingly, exerting a major adverse impact on human health by increasing the prevalences of disorders, such as diabetes, hypertension and heart disease. To maintain systemic energy homeostasis, metabolic information must be communicated among organs/tissues. Obesity-related disorders can be thought of as resulting from dysregulation of this vital inter-tissue communication. Remarkable advances in obesity research during this decade have shown humoral factors manufactured and secreted by adipose tissue (adipocytokines) to be of great importance. In addition to these humoral factors, such as nutrients (glucose, fatty acids and amino acids) and hormones (insulin, adipocytokines and so on), the functional significance of the autonomic nervous system has recently attracted research attention. Autonomic nerves are essential components of the endogenous system for maintaining energy homeostasis, making them potential therapeutic targets for obesity-related disorders. This review focuses on the therapeutic possibilities of targeting inter-organ communication systems.
Collapse
Affiliation(s)
- Tetsuya Yamada
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | |
Collapse
|
43
|
Liu QS, Gao M, Zhu SY, Li SJ, Zhang L, Wang QJ, Du GH. The novel mechanism of recombinant human ciliary neurotrophic factor on the anti-diabetes activity. Basic Clin Pharmacol Toxicol 2007; 101:78-84. [PMID: 17651306 DOI: 10.1111/j.1742-7843.2007.00092.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In a previous study, the ciliary neurotrophic factor (CNTF) were demonstrated to lead to weight-loss partly by up-regulating the energy metabolism and the expression of uncoupling protein-1, mitochondrial transcription factor A and nuclear respiratory factor-1 in adipose tissues or muscle. To investigate the up-stream regulators of the expression, recombinant human CNTF (rhCNTF) (0.1, 0.3 and 0.9 mg/kg/day subcutaneously) were administered to KK-Ay mice for 30 days, resulting in reduction of perirenal fat mass, serum free fatty acids and islet triacylglycerol; furthermore, the values of oral glucose tolerance test were found improved. In brown adipose tissues, the gene expressions of peroxisome proliferator-activated receptor alpha (PPARalpha) and peroxisome proliferator-activated receptor coactivator-1 alpha (PGC-1alpha) were found to be up-regulated by rhCNTF. To the best of our knowledge, the changes of gene expression of PPARalpha and PGC-1alpha represent new insights into the mechanisms of anti-diabetes by rhCNTF. In addition, the activity of mitochondrial complexII was found to be increased by rhCNTF. Stimulation of PPARalpha, PGC-1alpha, uncoupling protein-1 and enhanced activity of mitochondrial complex II may be associated with the effects of anti-diabetes. The present study indicates new mechanisms of the activity and mechanisms on anti-diabetes of rhCNTF, which may be a novel anti-diabetes reagent partly acting by enhancing energy metabolism.
Collapse
|
44
|
Yamada T, Katagiri H. Avenues of communication between the brain and tissues/organs involved in energy homeostasis. Endocr J 2007; 54:497-505. [PMID: 17510499 DOI: 10.1507/endocrj.kr-106] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Obesity is a rapidly increasing public health concern worldwide as a major risk factor for numerous disorders, including diabetes, hypertension and heart disease. Despite remarkable advances in obesity research over the past 10 years, the molecular mechanisms underlying obesity are still not completely understood. To maintain systemic energy homeostasis, it is important that organs/tissues communicate metabolic information among each other. Obesity-related disorders can be thought of as resulting from dysregulation of this inter-tissue communication. This system has both afferent sensing components and efferent effecter limbs. The afferent signals consist of not only humoral factors, such as nutrients (glucose, fatty acids and amino acids) and adipocytokines (leptin, adiponectin and so on), but also autonomic afferent nerve systems. Both converge on brain centers, most importantly within the hypothalamus, where the signals are integrated, and the direction and magnitude of efferent responses are determined. The efferent elements of this physiological system include those regulating energy inputs and outputs, i.e. food intake and metabolic rates. In this review, we will summarize recent advances in research on metabolic information avenues to the brain, which are important for energy homeostasis.
Collapse
Affiliation(s)
- Tetsuya Yamada
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | | |
Collapse
|
45
|
Katagiri H, Yamada T, Oka Y. Adiposity and cardiovascular disorders: disturbance of the regulatory system consisting of humoral and neuronal signals. Circ Res 2007; 101:27-39. [PMID: 17615379 DOI: 10.1161/circresaha.107.151621] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Obesity, a major healthcare issue, is associated with significant cardiovascular morbidities, including hypertension and atherosclerosis. Numerous intensive studies conducted this decade have revealed that adipose tissue is a major endocrine organ that secretes a variety of bioactive substances, termed adipocytokines. Adipocytokine secretion profiles are altered as obesity develops, which may increase the risk of obesity-related cardiovascular disorders. For instance, leptin is upregulated in obese subjects and plays important roles in the pathophysiology of obesity-related atherogenesis through multiple mechanisms, such as its proliferative, proinflammatory, prothrombotic, and prooxidant actions. In contrast, adiponectin, which is downregulated in obese subjects, has protective effects against cardiovascular disorders at various atherogenic stages. In addition to these factors secreted by adipose tissue, neuronal circuits involving autonomic nerves are now being recognized as an important metabolic regulatory system and have thus attracted considerable attentions. Alterations in fat accumulation in intraabdominal organs, such as visceral adipose tissue and the liver, send afferent neuronal signals to the brain, leading to modulation of sympathetic tonus and thereby affecting the vasculature. Moreover, these humoral and neuronal signaling pathways communicate with each other, resulting in cooperative metabolic regulation among tissues/organs throughout the body. Further elucidation of these regulatory systems is anticipated to lead to new approaches to devising therapeutic strategies for the metabolic syndrome.
Collapse
Affiliation(s)
- Hideki Katagiri
- Division of Advanced Therapeutics for Metabolic Diseases, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | |
Collapse
|
46
|
Liu QS, Wang QJ, Du GH, Zhu SY, Gao M, Zhang L, Zhu JM, Cao JF. Recombinant human ciliary neurotrophic factor reduces weight partly by regulating nuclear respiratory factor 1 and mitochondrial transcription factor A. Eur J Pharmacol 2007; 563:77-82. [PMID: 17397829 DOI: 10.1016/j.ejphar.2007.02.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 01/28/2007] [Accepted: 02/01/2007] [Indexed: 11/27/2022]
Abstract
Ciliary neurotrophic factor (CNTF) can lead to weight loss by up-regulating energy metabolism and the expression of UCP-1 in mitochondria. To investigate the up-stream regulators of the expression of UCP-1, recombinant human CNTF (rhCNTF) (0.1, 0.3, 0.9 mg/kg/day s.c.) administered to KK-Ay mice for 30 days resulting in reductions in body weight and perirenal fat mass. In brown adipose tissues, the gene expressions of nuclear respiratory factor (NRF)-1, mitochondrial transcription factor A (TFam) and uncoupling protein (UCP)-1 were found up-regulated by rhCNTF. To the best of our knowledge, these effects represent new insights on the mechanisms of action of weight loss by rhCNTF. In addition, we also found that rhCNTF increased the activity of mitochondrial complex IV. The stimulation of NRF-1, TFam, UCP-1 and the enhanced activity of mitochondrial complex IV may be associated with remedying obesity. The result indicates that rhCNTF can enhance the expressions of NRF-1 and TFam, both of which can up-regulate the expression of UCP-1.
Collapse
Affiliation(s)
- Qing-Shan Liu
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009 PR China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Choi CS, Savage DB, Kulkarni A, Yu XX, Liu ZX, Morino K, Kim S, Distefano A, Samuel VT, Neschen S, Zhang D, Wang A, Zhang XM, Kahn M, Cline GW, Pandey SK, Geisler JG, Bhanot S, Monia BP, Shulman GI. Suppression of diacylglycerol acyltransferase-2 (DGAT2), but not DGAT1, with antisense oligonucleotides reverses diet-induced hepatic steatosis and insulin resistance. J Biol Chem 2007; 282:22678-88. [PMID: 17526931 DOI: 10.1074/jbc.m704213200] [Citation(s) in RCA: 300] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major contributing factor to hepatic insulin resistance in type 2 diabetes. Diacylglycerol acyltransferase (Dgat), of which there are two isoforms (Dgat1 and Dgat2), catalyzes the final step in triglyceride synthesis. We evaluated the metabolic impact of pharmacological reduction of DGAT1 and -2 expression in liver and fat using antisense oligonucleotides (ASOs) in rats with diet-induced NAFLD. Dgat1 and Dgat2 ASO treatment selectively reduced DGAT1 and DGAT2 mRNA levels in liver and fat, but only Dgat2 ASO treatment significantly reduced hepatic lipids (diacylglycerol and triglyceride but not long chain acyl CoAs) and improved hepatic insulin sensitivity. Because Dgat catalyzes triglyceride synthesis from diacylglycerol, and because we have hypothesized that diacylglycerol accumulation triggers fat-induced hepatic insulin resistance through protein kinase C epsilon activation, we next sought to understand the paradoxical reduction in diacylglycerol in Dgat2 ASO-treated rats. Within 3 days of starting Dgat2 ASO therapy in high fat-fed rats, plasma fatty acids increased, whereas hepatic lysophosphatidic acid and diacylglycerol levels were similar to those of control rats. These changes were associated with reduced expression of lipogenic genes (SREBP1c, ACC1, SCD1, and mtGPAT) and increased expression of oxidative/thermogenic genes (CPT1 and UCP2). Taken together, these data suggest that knocking down Dgat2 protects against fat-induced hepatic insulin resistance by paradoxically lowering hepatic diacylglycerol content and protein kinase C epsilon activation through decreased SREBP1c-mediated lipogenesis and increased hepatic fatty acid oxidation.
Collapse
Affiliation(s)
- Cheol Soo Choi
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Buettner R, Schölmerich J, Bollheimer LC. High-fat diets: modeling the metabolic disorders of human obesity in rodents. Obesity (Silver Spring) 2007; 15:798-808. [PMID: 17426312 DOI: 10.1038/oby.2007.608] [Citation(s) in RCA: 784] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
RESEARCH METHODS AND PROCEDURES High-fat (HF) diet feeding can induce obesity and metabolic disorders in rodents that resemble the human metabolic syndrome. However, this dietary intervention is not standardized, and the HF-induced phenotype varies distinctly among different studies. The question which HF diet type is best to model the metabolic deterioration seen in human obesity remains unclear. Therefore, in this review, metabolic data obtained with different HF diet approaches are compiled. Both whole-body and organ-specific diet effects are analyzed. RESULTS On the basis of these results, we conclude that animal fats and omega-6/omega-9-containing plant oils can be used to generate an obese and insulin-resistant phenotype in rodents, whereas fish oil-fed animals do not develop these disorders. DISCUSSION Looking at the present data, it does not seem possible to define an ideal HF diet, and an exact definition of diet composition and a thorough metabolic characterization of the HF diet effects in a researcher's specific laboratory setting remains essential for metabolic studies with this model.
Collapse
Affiliation(s)
- Roland Buettner
- Department of Internal Medicine I, University of Regensburg, 93042 Regensburg, Germany.
| | | | | |
Collapse
|
49
|
Gao J, Katagiri H, Ishigaki Y, Yamada T, Ogihara T, Imai J, Uno K, Hasegawa Y, Kanzaki M, Yamamoto TT, Ishibashi S, Oka Y. Involvement of apolipoprotein E in excess fat accumulation and insulin resistance. Diabetes 2007; 56:24-33. [PMID: 17192461 DOI: 10.2337/db06-0144] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Although apolipoprotein E (apoE) is well known to play a major role in lipid metabolism, its role in glucose and energy homeostasis remains unclear. Herein, we established apoE-deficient genetically obese Ay (apoE(-/-);Ay/+) mice. ApoE deficiency in Ay mice prevented the development of obesity, with decreased fat accumulation in the liver and adipose tissues. ApoE(-/-);Ay/+ mice exhibited better glucose tolerance than apoE(+/+);Ay/+ mice. Insulin tolerance testing and hyperinsulinemic-euglycemic clamp study revealed marked improvement of insulin sensitivity, despite increased plasma free fatty acid levels. These metabolic phenotypes were reversed by adenoviral replenishment of apoE protein, indicating circulating apoE to be involved in increased adiposity and obesity-related metabolic disorders. Uptake of apoE-lacking VLDL into the liver and adipocytes was markedly inhibited, but adipocytes in apoE(-/-);Ay/+ mice exhibited normal differentiation, suggesting that apoE-dependent VLDL transport is involved in the development of obesity, i.e., surplus fat accumulation. Interestingly, apoE(-/-);Ay/+ mice exhibited decreased food intake and increased energy expenditure. Pair-feeding experiments indicate these phenomena to both contribute to the obesity-resistant phenotypes associated with apoE deficiency. Thus, apoE is involved in maintaining energy homeostasis. ApoE-dependent excess fat accumulation is a promising therapeutic target for the metabolic syndrome.
Collapse
Affiliation(s)
- Junhong Gao
- Division of Molecular Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Penza M, Montani C, Romani A, Vignolini P, Pampaloni B, Tanini A, Brandi ML, Alonso-Magdalena P, Nadal A, Ottobrini L, Parolini O, Bignotti E, Calza S, Maggi A, Grigolato PG, Di Lorenzo D. Genistein affects adipose tissue deposition in a dose-dependent and gender-specific manner. Endocrinology 2006; 147:5740-51. [PMID: 16959845 DOI: 10.1210/en.2006-0365] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The soy isoflavone genistein targets adipose tissue and elicits physiological effects that may vary based on dietary intake. We hypothesized that the adipose effects of genistein are dose and gender dependent. Four-week-old C57BL/6 male and female mice received daily oral doses of genistein (50-200,000 microg/kg.d) or 17beta-estradiol (E2) (5 microg/kg.d) for 15 d or a diet containing 800 ppm genistein. Genistein increased epididymal and renal fat pad and adipocyte size at doses up to 50,000 microg/kg.d or at 800 ppm in the diet in males but not in females. The alteration in adipocity correlated with changes in peripheral insulin resistance. These treatments increased genistein serum concentrations from 35+/-6 to 103+/-26 nM 12 h after treatment and lowered plasma triglycerides and cholesterol levels. The 200,000 microg/kg.d genistein dose decreased adipose tissue weight similarly to E2. This genistein dose down-regulated estrogen receptor (beta more than alpha) and progesterone receptor expression and induced estrogen-dependent adipose differentiation factors; it did not change expression of the minimal consensus estrogen-responsive element in ERE-tK-LUC mice, which was positively modulated in other tissues (e.g. the lung). E2 down-regulated almost all examined adipogenic factors. Gene microarray analysis identified factors in fat metabolism and obesity-related phenotypes differentially regulated by low and high doses of genistein, uncovering its adipogenic and antiadipogenic actions. The lower dose induced the phospholipase A2 group 7 and the phospholipid transfer protein genes; the 200,000 microg/kg.d dose inhibited them. The antiadipogenic action of genistein and down-regulation of adipogenic genes required the expression of ERbeta. In conclusion, nutritional doses of genistein are adipogenic in a gender-specific manner, whereas pharmacological doses inhibited adipose deposition.
Collapse
Affiliation(s)
- M Penza
- 3rd Laboratory/Biotechnology, and Department of Diagnostics, Civic Hospital of Brescia, 25123 and Department of Pathology, University of Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|