1
|
Feng J, Xu R, Dou Z, Hao Y, Xu R, Khoso MA, Shi Y, Liu L, Sun H, Chen C, Li X, Liu H, Han W, Cheng M, Tang P, Li J, Zhang Y, Liu X. Tetrahydroberberrubine improves hyperlipidemia by activating the AMPK/SREBP2/PCSK9/LDL receptor signaling pathway. Eur J Pharmacol 2025; 989:177228. [PMID: 39755242 DOI: 10.1016/j.ejphar.2024.177228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/07/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
Hyperlipidemia is a major risk factor for hypertension, coronary heart disease, diabetes and stroke, triggering an intensified research efforts into its prevention and treatment. Tetrahydroberberrubine (THBru) is a derivative of berberine (BBR) that has been shown to have higher bioavailability and lower toxicity compared to its parent compound. However, its impact on hyperlipidemia has not been fully explored. This study was aimed to investigate the effects and potential mechanisms of THBru on hyperlipidemia. Herein, we constructed the hyperlipidemia animal model in C57BL/6J mice through the administration of a 20-week high-fat diet (HFD). The liver damage and lipid metabolism disorders in hyperlipidemic mice were effectively alleviated by THBru (25 or 50 mg/kg) administration. Molecular docking and cellular thermal shift assay (CETSA) have revealed a direct interaction between THBru and the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK). THBru was found to downregulate the expression of sterol regulatory element-binding protein 2 (SREBP2) and proprotein convertase subtilisin/kexin type 9 (PCSK9), while upregulate the expression of low-density lipoprotein cholesterol (LDL-C) in the liver of hyperlipidemic mice and lipid metabolism abnormalities cells. The application of AMPK inhibitor in HepG2 cells was able to effectively reverse the regulatory effect of THBru on the AMPK/SREBP2/PCSK9/LDL receptor signaling pathway. In summary, this study for the first time found that THBru is a potential agonist of AMPK, regulate the SREBP2/PCSK9/LDL receptor pathway to improve hyperlipidemia, providing new insights into the prevention and treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Jing Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Run Xu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Zijia Dou
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Yutong Hao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Ranchen Xu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Muneer Ahmed Khoso
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Yang Shi
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Ling Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Heyang Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Chen Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Xiaohan Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Heng Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Weina Han
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Ming Cheng
- Cardiovascular Surgery Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150006, China
| | - Pengcheng Tang
- Cardiovascular Surgery Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150006, China
| | - Junquan Li
- Cardiovascular Surgery Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150006, China.
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China.
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China.
| |
Collapse
|
2
|
Lee Y, Choi D, Park J, Kim JG, Choi T, Youn D. The Effects of Warm Acupuncture on the Expression of AMPK in High-Fat Diet-Induced MAFLD Rats. Curr Issues Mol Biol 2024; 46:11580-11592. [PMID: 39451567 PMCID: PMC11506734 DOI: 10.3390/cimb46100687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
This study investigated the effects of acupuncture and warm acupuncture on the expression and mechanism of the AMP-activated protein kinase (AMPK) signalling pathway associated with lipid accumulation in the liver tissue of rats with metabolic dysfunction-associated fatty liver disease (MAFLD) induced by a high-fat diet. Sprague-Dawley rats were categorised into four groups: control (CON), untreated MAFLD (MAFLD), and two MAFLD groups treated with acupuncture (ACU) and warm acupuncture (WA). The treatment groups underwent 16 application sessions over 8 weeks at the SP9 and BL18 acupoints. We measured the expression levels of AMPK, sterol regulatory element-binding protein1 (SREBP1), acetyl-coenzyme A carboxylase (ACC), peroxisome proliferator-activated receptorα (PPARα), carnitine palmitoyltransferase1 (CPT1), and CPT2. AMPK was activated in both ACU and WA groups. WA downregulated both SREBP1 and ACC expression at the protein level, whereas the acupuncture treatment downregulated SREBP1 expression. Additionally, WA selectively induced the activation of signalling pathways related to AMPK, PPARα, CPT1, and CPT2 at the mRNA level. Histological observations confirmed that fat accumulation was reduced in both the ACU and the WA groups compared to the MAFLD group. The WA treatment-promoted amelioration of HFD-induced MAFLD may be related to the activation of the AMPK/SREBP1/ACC pathway in the liver.
Collapse
Affiliation(s)
- Yumi Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (Y.L.); (J.P.); (J.G.K.)
| | - Donghee Choi
- Department of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea;
| | - Junghye Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (Y.L.); (J.P.); (J.G.K.)
| | - Jae Gwan Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (Y.L.); (J.P.); (J.G.K.)
| | - Taejin Choi
- DongHaeng Convalescent Hospital, Gwangju 61251, Republic of Korea;
| | - Daehwan Youn
- Department of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea;
| |
Collapse
|
3
|
González-Blázquez R, Gil-Ortega M, Alcalá M, González-Moreno D, Viana M, Chowen JA, Sanz-Gómez M, Fernández-Alfonso MS, Somoza B. Short-term dietary intervention improves endothelial dysfunction induced by high-fat feeding in mice through upregulation of the AMPK-CREB signaling pathway. Acta Physiol (Oxf) 2023; 239:e14023. [PMID: 37553856 DOI: 10.1111/apha.14023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023]
Abstract
AIM In addition to functioning as an energy sensor switch, AMPK plays a key role in the maintenance of cardiovascular homeostasis. However, obesity disrupts AMPK signaling, contributing to endothelial dysfunction and cardiovascular disease. This study aimed to elucidate if a short-term dietary intervention consisting in replacing the high-fat diet with a standard diet for 2 weeks could reverse obesity-induced endothelial dysfunction via AMPK-CREB activation. METHODS For this, 5-week-old male C57BL6J mice were fed a standard (Chow) or a high-fat (HF) diet for 8 weeks. The HF diet was replaced by the chow diet for the last 2 weeks in half of HF mice, generating 3 groups: Chow, HF and HF-Chow. Vascular reactivity and western-blot assays were performed in the thoracic aorta. RESULTS Returning to a chow diet significantly reduced body weight and glucose intolerance. Relaxant responses to acetylcholine and the AMPK activator (AICAR) were significantly impaired in HF mice but improved in HF-Chow mice. The protein levels of AMPKα, p-CREB and antioxidant systems (heme oxygenase-1 (HO-1) and catalase) were significantly reduced in HF but normalized in HF-Chow mice. CONCLUSION Improving dietary intake by replacing a HF diet with a standard diet improves AMPK-mediated responses due to the upregulation of the AMPK/CREB/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Raquel González-Blázquez
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Martín Alcalá
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Daniel González-Moreno
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Marta Viana
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Julie A Chowen
- Department of Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Marta Sanz-Gómez
- Instituto Pluridisciplinar, Unidad de Cartografía Cerebral, Universidad Complutense de Madrid, Madrid, Spain
| | - María S Fernández-Alfonso
- Instituto Pluridisciplinar, Unidad de Cartografía Cerebral, Universidad Complutense de Madrid, Madrid, Spain
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
4
|
Wang X, Yang S, Li Y, Jin X, Lu J, Wu M. Role of emodin in atherosclerosis and other cardiovascular diseases: Pharmacological effects, mechanisms, and potential therapeutic target as a phytochemical. Biomed Pharmacother 2023; 161:114539. [PMID: 36933375 DOI: 10.1016/j.biopha.2023.114539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/20/2023] Open
Abstract
The morbidity and mortality of cardiovascular diseases (CVDs) are increasing in recent years, and atherosclerosis (AS), a major CVD, becomes a disorder that afflicts human beings severely, especially the elders. AS is recognized as the primary cause and pathological basis of some other CVDs. The active constituents of Chinese herbal medicines have garnered increasing interest in recent researches owing to their influence on AS and other CVDs. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is a naturally occurring anthraquinone derivative found in some Chinese herbal medicines such as Rhei radix et rhizome, Polygoni cuspidati rhizoma et radix and Polygoni multiflori root. In this paper, we first review the latest researches about emodin's pharmacology, metabolism and toxicity. Meanwhile, it has been shown to be effective in treating CVDs caused by AS in dozens of previous studies. Therefore, we systematically reviewed the mechanisms by which emodin treats AS. In summary, these mechanisms include anti-inflammatory activity, lipid metabolism regulation, anti-oxidative stress, anti-apoptosis and vascular protection. The mechanisms of emodin in other CVDs are also discussed, such as vasodilation, inhibition of myocardial fibrosis, inhibition of cardiac valve calcification and antiviral properties. We have further summarized the potential clinical applications of emodin. Through this review, we hope to provide guidance for clinical and preclinical drug development.
Collapse
Affiliation(s)
- Xinyue Wang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujuan Li
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Jin
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Beijing University of Chinese Medicine, Beijing, China
| | - Min Wu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Cubillos-Angulo JM, Nogueira BMF, Arriaga MB, Barreto-Duarte B, Araújo-Pereira M, Fernandes CD, Vinhaes CL, Villalva-Serra K, Nunes VM, Miguez-Pinto JP, Amaral EP, Andrade BB. Host-directed therapies in pulmonary tuberculosis: Updates on anti-inflammatory drugs. Front Med (Lausanne) 2022; 9:970408. [PMID: 36213651 PMCID: PMC9537567 DOI: 10.3389/fmed.2022.970408] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/22/2022] [Indexed: 11/26/2022] Open
Abstract
Tuberculosis (TB) is a lethal disease and remains one of the top ten causes of mortality by an infectious disease worldwide. It can also result in significant morbidity related to persistent inflammation and tissue damage. Pulmonary TB treatment depends on the prolonged use of multiple drugs ranging from 6 months for drug-susceptible TB to 6-20 months in cases of multi-drug resistant disease, with limited patient tolerance resulting from side effects. Treatment success rates remain low and thus represent a barrier to TB control. Adjunct host-directed therapy (HDT) is an emerging strategy in TB treatment that aims to target the host immune response to Mycobacterium tuberculosis in addition to antimycobacterial drugs. Combined multi-drug treatment with HDT could potentially result in more effective therapies by shortening treatment duration, improving cure success rates and reducing residual tissue damage. This review explores the rationale and challenges to the development and implementation of HDTs through a succinct report of the medications that have completed or are currently being evaluated in ongoing clinical trials.
Collapse
Affiliation(s)
- Juan M. Cubillos-Angulo
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
| | - Betânia M. F. Nogueira
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
| | - María B. Arriaga
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
| | - Beatriz Barreto-Duarte
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
- Curso de Medicina, Universidade Salvador, Salvador, Brazil
- Programa de Pós-Graduação em Clínica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Araújo-Pereira
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
| | - Catarina D. Fernandes
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
| | - Caian L. Vinhaes
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
- Bahiana School of Medicine and Public Health, Bahia Foundation for the Development of Sciences, Salvador, Brazil
| | - Klauss Villalva-Serra
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
- Curso de Medicina, Universidade Salvador, Salvador, Brazil
| | | | | | - Eduardo P. Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Bruno B. Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Brazil
- Programa de Pós-Graduação em Clínica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Bahiana School of Medicine and Public Health, Bahia Foundation for the Development of Sciences, Salvador, Brazil
| |
Collapse
|
6
|
Kothari V, Babu JR, Mathews ST. AMP activated kinase negatively regulates hepatic Fetuin-A via p38 MAPK-C/EBPβ/E3 Ubiquitin Ligase Signaling pathway. PLoS One 2022; 17:e0266472. [PMID: 35522655 PMCID: PMC9075660 DOI: 10.1371/journal.pone.0266472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
Fetuin-A (Fet-A) is a liver-secreted phosphorylated protein, known to impair insulin signaling, which has been shown to be associated with obesity, insulin resistance, and incident diabetes. Fet-A interacts with the insulin-stimulated insulin receptor (IR) and inhibits IR tyrosine kinase activity and glucose uptake. It has been shown that high glucose increases Fet-A expression through the ERK1/2 signaling pathway. However, factors that downregulate Fet-A expression and their potential mechanisms are unclear. We examined the effect of AMP-activated protein kinase (AMPK) on high-glucose induced Fet-A expression in HepG2 cells, Hep3B cells and primary rat hepatocytes. High glucose increased Fet-A and phosphorylated (Ser312) fetuin-A (pFet-A) expression, which are known to impair insulin signaling. AICAR-induced AMPK activation significantly down-regulated high glucose-induced Fet-A expression and secretion of pFet-A while treatment with Compound C (AMPK inhibitor), SB202190 (p38 MAPK inhibitor) or p38 MAPK siRNA transfection prevented AICAR-induced downregulation of Fet-A expression. In addition, activation of p38 MAPK, by anisomycin, decreased the hepatic expression of Fet-A. Further, we our studies have shown that short-term effect of AICAR-treatment on Fet-A expression was mediated by proteosomal degradation, and long-term treatment of AICAR was associated with decrease in hepatic expression of C/EBP beta, an important transcription factor involved in the regulation of Fet-A. Taken together, our studies implicate a critical role for AMPK-p38 MAPK-C/EBPb-ubiquitin-proteosomal axis in the regulation of the expression of hepatic Fet-A.
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Nutrition and Dietetics, Boshell Diabetes and Metabolic Diseases Research Program, Auburn University, Auburn, AL, United States of America
| | - Jeganathan Ramesh Babu
- Department of Nutrition and Dietetics, Boshell Diabetes and Metabolic Diseases Research Program, Auburn University, Auburn, AL, United States of America
| | - Suresh T. Mathews
- Department of Nutrition and Dietetics, Boshell Diabetes and Metabolic Diseases Research Program, Auburn University, Auburn, AL, United States of America
- Department of Nutrition and Dietetics, Samford University, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
7
|
Hao Q, Huang Z, Li Q, Liu D, Wang P, Wang K, Li J, Cao W, Deng W, Wu K, Su R, Liu Z, Vadgama J, Wu Y. A Novel Metabolic Reprogramming Strategy for the Treatment of Diabetes-Associated Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102303. [PMID: 35023320 PMCID: PMC8867195 DOI: 10.1002/advs.202102303] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/08/2021] [Indexed: 05/11/2023]
Abstract
Diabetes is directly related to the risk of breast cancer (BC) occurrence and worsened BC prognosis. Currently, there are no specific treatments for diabetes-associated BC. This paper aims to understand the fundamental mechanisms of diabetes-induced BC progression and to develop personalized treatments. It reports a metabolic reprogramming strategy (MRS) that pharmaceutical induction of glucose import and glycolysis with metformin and NF-κB inhibitor (NF-κBi) while blocking the export of excessive lactate via inhibiting monocarboxylate transporter 4 (MCT4) leads to a metabolic crisis within the cancer cells. It demonstrates that the MRS shifts the metabolism of BC cells toward higher production of lactate, blocks lactate secretion, prompts intracellular acidification and induces significant cytotoxicity. Moreover, a novel MCT4 inhibitor CB-2 has been identified by structure-based virtual screening. A triple combination of metformin, CB-2, and trabectedin, a drug that impedes NF-κB signaling, strongly inhibits BC cells. Compared to normal glucose condition, MRS elicits more potent cancer cell-killing effects under high glucose condition. Animal model studies show that diabetic conditions promote the proliferation and progression of BC xenografts in nude mice and that MRS treatment significantly inhibits HG-induced BC progression. Therefore, inhibition of MCT4 combined with metformin/NF-κBi is a promising cancer therapy, especially for diabetes-associated BC.
Collapse
Affiliation(s)
- Qiongyu Hao
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
| | - Zhimin Huang
- Key Laboratory of Cell Differentiation and ApoptosisMinistry of EducationDepartment of PathophysiologyShanghai Jiao‐Tong University School of MedicineShanghai200025China
- Department of BioengineeringRice UniversityHoustonTX77005USA
| | - Qun Li
- Department of OncologyShanghai East HospitalSchool of MedicineTongji UniversityShanghai200123China
| | - Dingxie Liu
- Bluewater Biotech LLCNew ProvidenceNJ07974USA
| | - Piwen Wang
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
| | - Kun Wang
- Department of Breast CancerCancer CenterGuangdong Provincial People's Hospital & Guangdong Academy of Medical SciencesGuangzhou510080China
| | - Jieqing Li
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
- Department of Breast CancerCancer CenterGuangdong Provincial People's Hospital & Guangdong Academy of Medical SciencesGuangzhou510080China
| | - Wei Cao
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
| | - Wenhong Deng
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
- Department of General SurgeryRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Ke Wu
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
| | - Rui Su
- College of EngineeringUniversity of CaliforniaBerkeleyCA94720USA
| | - Zhongmin Liu
- The Institute for Biomedical Engineering & Nano ScienceShanghai East HospitalTongji University School of MedicineShanghai200120China
| | - Jay Vadgama
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
| | - Yong Wu
- Division of Cancer Research and TrainingDepartment of Internal MedicineCharles Drew University of Medicine and ScienceDavid Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer CenterLos AngelesCA90095USA
| |
Collapse
|
8
|
He LF, Wang C, Zhang YF, Guo CC, Wan Y, Li YX. Effect of Emodin on Hyperlipidemia and Hepatic Lipid Metabolism in Zebrafish Larvae Fed a High-Cholesterol Diet. Chem Biodivers 2021; 19:e202100675. [PMID: 34866324 DOI: 10.1002/cbdv.202100675] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022]
Abstract
Hyperlipidemia (HLP) is a complex pathological condition results from lipid metabolism disorder, which is closely related to obesity, atherosclerosis and steatohepatitis. Emodin (EM), a natural anthraquinone, exhibits prominent hypolipidemic effects. However, its exact mechanism is still unclear. In this study, we successfully established hyperlipidemic zebrafish model induced by 4 % high-cholesterol diet (HCD) for 10 days and explored the anti-hyperlipidemic roles and underlying mechanisms of EM. The results indicated that EM attenuated the mortality and body mass index (BMI) of zebrafish with HLP, and ameliorated abnormal lipid levels involved in TC, TG, LDL-C and HDL-C levels. Besides, EM effectively reduced lipid accumulation in blood vessels and liver, alleviated hepatic histological damage, and inhibited vascular neutrophil inflammation. Finally, the mRNA expression of molecules related to lipid metabolism were studied by using real-time quantitative polymerase chain reaction (RT-qPCR) to investigated the underlying mechanism. Further results found that treatment with EM up-regulated AMPKα, LDLR, ABCA1 and ABCG1, and down-regulated SREBP-2, PCSK9 and HMGCR expression. In conclusion, EM showed a prominent mitigative effect on lipid metabolism disorder in zebrafish larvae with HCD-stimulated HLP, which was associated with the enhancement of LDL-C uptake and reverse cholesterol transport, and inhibition of cholesterol synthesis.
Collapse
Affiliation(s)
- Lin-Feng He
- National Key Laboratory of Southwestern Chinese Medicine Resources & Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education & School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, China
| | - Cheng Wang
- National Key Laboratory of Southwestern Chinese Medicine Resources & Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education & School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, China
| | - Ya-Fang Zhang
- National Key Laboratory of Southwestern Chinese Medicine Resources & Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education & School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, China
| | - Chao-Cheng Guo
- National Key Laboratory of Southwestern Chinese Medicine Resources & Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education & School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, China
| | - Yan Wan
- National Key Laboratory of Southwestern Chinese Medicine Resources & Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education & School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, China
| | - Yun-Xia Li
- National Key Laboratory of Southwestern Chinese Medicine Resources & Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education & School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, China
| |
Collapse
|
9
|
Barone E, Di Domenico F, Perluigi M, Butterfield DA. The interplay among oxidative stress, brain insulin resistance and AMPK dysfunction contribute to neurodegeneration in type 2 diabetes and Alzheimer disease. Free Radic Biol Med 2021; 176:16-33. [PMID: 34530075 PMCID: PMC8595768 DOI: 10.1016/j.freeradbiomed.2021.09.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly followed by vascular dementia. In addition to clinically diagnosed dementia, cognitive dysfunction has been reported in diabetic patients. Recent studies are now beginning to recognize type 2 diabetes mellitus (T2DM), characterized by chronic hyperglycemia and insulin resistance, as a risk factor for AD and other cognitive disorders. While studies on insulin action have remained traditionally in the domain of peripheral tissues, the detrimental effects of insulin resistance in the central nervous system on cognitive dysfunction are increasingly being reported in recent clinical and preclinical studies. Brain functions require continuous supply of glucose and oxygen and a tight regulation of metabolic processes. Loss of this metabolic regulation has been proposed to be a contributor to memory dysfunction associated with neurodegeneration. Within the above scenario, this review will focus on the interplay among oxidative stress (OS), insulin resistance and AMPK dysfunctions in the brain by highlighting how these neurotoxic events contribute to neurodegeneration. We provide an overview on the detrimental effects of OS on proteins regulating insulin signaling and how these alterations impact cell metabolic dysfunctions through AMPK dysregulation. Such processes, we assert, are critically involved in the molecular pathways that underlie AD.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA.
| |
Collapse
|
10
|
Rogacka D. Insulin resistance in glomerular podocytes: Potential mechanisms of induction. Arch Biochem Biophys 2021; 710:109005. [PMID: 34371008 DOI: 10.1016/j.abb.2021.109005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/19/2021] [Accepted: 08/05/2021] [Indexed: 01/15/2023]
Abstract
Glomerular podocytes are a target for the actions of insulin. Accumulating evidence indicates that exposure to nutrient overload induces insulin resistance in these cells, manifested by abolition of the stimulatory effect of insulin on glucose uptake. Numerous recent studies have investigated potential mechanisms of the induction of insulin resistance in podocytes. High glucose concentrations stimulated reactive oxygen species production through NADPH oxidase activation, decreased adenosine monophosphate-activated protein kinase (AMPK) phosphorylation, and reduced deacetylase sirtuin 1 (SIRT1) protein levels and activity. Calcium signaling involving transient receptor potential cation channel C, member 6 (TRPC6) also was demonstrated to play an essential role in the regulation of insulin-dependent signaling and glucose uptake in podocytes. Furthermore, podocytes exposed to diabetic environment, with elevated insulin levels become insulin resistant as a result of degradation of insulin receptor (IR), resulting in attenuation of insulin signaling responsiveness. Also elevated levels of palmitic acid appear to be an important factor and contributor to podocytes insulin resistance. This review summarizes cellular and molecular alterations that contribute to the development of insulin resistance in glomerular podocytes.
Collapse
Affiliation(s)
- Dorota Rogacka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| |
Collapse
|
11
|
Weiss M, Fellmann L, Regnard P, Bousquet P, Monassier L, Niederhoffer N. Protective effects of the imidazoline-like drug lnp599 in a marmoset model of obesity-induced metabolic disorders. Int J Obes (Lond) 2021; 45:1229-1239. [PMID: 33654274 DOI: 10.1038/s41366-021-00786-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/27/2021] [Accepted: 02/05/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND/OBJECTIVES Overweight and obesity are undoubtable risk factors for type 2 diabetes and cardiovascular diseases and significantly contribute to the global morbi-mortality. We previoulsy reported that LNP599, a pharmacological imidazoline-like activator of hepatic AMPK/adiponectin signaling, protects against the development of adiposity and obesity and the associated cardio-metabolic disorders, suggesting that it may be a suitable drug candidate for a therapeutic approach targeting the development of obesity at very early stages. The objective of the present study was to evaluate the metabolic effects of LNP599 in a model of diet-induced overweight and metabolic disorders in a nonhuman primate, the common marmoset (Callithrix jacchus), and more particularly to establish the impact of the compound on cholesterol homeostasis, i.e., HDL and LDL/VLDL lipoproteins. METHODS Marmosets were fed normal (NC) or hypercaloric (HC) chow during 16 weeks. Diet-induced changes in body weight and metabolism were assessed. Effects of LNP599 were evaluated in a subset of HC animals (HC-LNP) receiving the compound at a daily dose of 10 mg/kg over the 16 weeks. RESULTS HC-feeding induced significant overweight associated with a marked dyslipidemia (hypertriglyceridemia, hypercholesterolemia, and reduced HDL over LDL/VLDL cholesterol ratio). LNP599 blunted the diet-induced body weight gain and largely protected against the development of hypertriglyceridemia. Total cholesterol was unchanged but the ratio of HDL over LDL/VLDL cholesterol was more than doubled. CONCLUSIONS The profile of metabolic troubles obtained upon enriched diet mimicked the disorders associated with spontaneous obesity in marmosets. HC marmosets represent an experimental model of high clinical relevance to study the pathophysiology of obesity and related dyslipidemia and to evaluate the effects of emerging therapies targeting these disorders. Our data confirm the preventing effects of LNP599 in a nonhuman primate model and demonstrate for the first time the high potency of this drug in promoting HDL-cholesterol.
Collapse
Affiliation(s)
- Maud Weiss
- Laboratoire de Pharmacologie et Toxicologie NeuroCardiovasculaire - UR7296, Fédération de Médecine Translationnelle de Strasbourg, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - Lyne Fellmann
- SILABE, Université de Strasbourg, Fort Foch, Niederhausbergen, Strasbourg, France
| | - Pierrick Regnard
- SILABE, Université de Strasbourg, Fort Foch, Niederhausbergen, Strasbourg, France
| | - Pascal Bousquet
- Laboratoire de Pharmacologie et Toxicologie NeuroCardiovasculaire - UR7296, Fédération de Médecine Translationnelle de Strasbourg, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - Laurent Monassier
- Laboratoire de Pharmacologie et Toxicologie NeuroCardiovasculaire - UR7296, Fédération de Médecine Translationnelle de Strasbourg, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - Nathalie Niederhoffer
- Laboratoire de Pharmacologie et Toxicologie NeuroCardiovasculaire - UR7296, Fédération de Médecine Translationnelle de Strasbourg, Faculté de Médecine, Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
12
|
Li X, Hu X, Pan T, Dong L, Ding L, Wang Z, Song R, Wang X, Wang N, Zhang Y, Wang J, Yang B. Kanglexin, a new anthraquinone compound, attenuates lipid accumulation by activating the AMPK/SREBP-2/PCSK9/LDLR signalling pathway. Biomed Pharmacother 2021; 133:110802. [PMID: 33202286 DOI: 10.1016/j.biopha.2020.110802] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
Hyperlipidaemia is one of the major risk factors for atherosclerosis, coronary heart disease, stroke and diabetes. In the present study, we synthesized a new anthraquinone compound, 1,8-dihydroxy-3-succinic acid monoethyl ester-6-methylanthraquinone, and named it Kanglexin (KLX). The aim of this study was to evaluate whether KLX has a lipid-lowering effect and to explore the potential molecular mechanism. In this study, Sprague-Dawley rats were fed a high fat diet (HFD) for 5 weeks to establish a hyperlipidaemia model; then, the rats were orally administered KLX (20, 40, and 80 mg kg-1·d-1) or atorvastatin calcium (AT, 10 mg kg-1·d-1) once a day for 2 weeks. KLX had prominent effects on reducing blood lipids, hepatic lipid accumulation, body weight and the ratio of liver weight/body weight. Furthermore, KLXdramatically reduced the total cholesterol (TC) and triglyceride (TG) levels and lipid accumulation in a HepG2 cell model of dyslipidaemia induced by 1 mmol/L oleic acid (OA). KLX may decrease lipid levels by phosphorylating adenosine monophosphate-activated protein kinase (AMPK) and the downstream sterol regulatory element binding protein 2 (SREBP-2)/proprotein convertase subtilisin/kexin type 9 (PCSK9)/low-density lipoprotein receptor (LDLR) signalling pathway in the HFD rats and OA-treated HepG2 cells. The effects of KLX on the AMPK/SREBP-2/PCSK9/LDLR signalling pathway were abolished when AMPK was inhibited by compound C (a specific AMPK inhibitor) in HepG2 cells. In summary, KLX has an efficient lipid-lowering effect mediated by activation of the AMPK/SREBP-2/PCSK9/LDLR signalling pathway. Our findings may provide new insight into and evidence for the discovery of a new lipid-lowering drug for the prevention and treatment of hyperlipidaemia, fatty liver, and cardiovascular disease in the clinic.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Xueling Hu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Tengfei Pan
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Lei Dong
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Lili Ding
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical CO. LTD, Jiangsu, Lianyungang 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu, Lianyungang 222001, China.
| | - Rui Song
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Xiuzhu Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Ning Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jinhui Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
13
|
Peroxisome Proliferator-Activated Receptors as Molecular Links between Caloric Restriction and Circadian Rhythm. Nutrients 2020; 12:nu12113476. [PMID: 33198317 PMCID: PMC7696073 DOI: 10.3390/nu12113476] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The circadian rhythm plays a chief role in the adaptation of all bodily processes to internal and environmental changes on the daily basis. Next to light/dark phases, feeding patterns constitute the most essential element entraining daily oscillations, and therefore, timely and appropriate restrictive diets have a great capacity to restore the circadian rhythm. One of the restrictive nutritional approaches, caloric restriction (CR) achieves stunning results in extending health span and life span via coordinated changes in multiple biological functions from the molecular, cellular, to the whole-body levels. The main molecular pathways affected by CR include mTOR, insulin signaling, AMPK, and sirtuins. Members of the family of nuclear receptors, the three peroxisome proliferator-activated receptors (PPARs), PPARα, PPARβ/δ, and PPARγ take part in the modulation of these pathways. In this non-systematic review, we describe the molecular interconnection between circadian rhythm, CR-associated pathways, and PPARs. Further, we identify a link between circadian rhythm and the outcomes of CR on the whole-body level including oxidative stress, inflammation, and aging. Since PPARs contribute to many changes triggered by CR, we discuss the potential involvement of PPARs in bridging CR and circadian rhythm.
Collapse
|
14
|
Lee HA, Cho JH, Afinanisa Q, An GH, Han JG, Kang HJ, Choi SH, Seong HA. Ganoderma lucidum Extract Reduces Insulin Resistance by Enhancing AMPK Activation in High-Fat Diet-Induced Obese Mice. Nutrients 2020; 12:nu12113338. [PMID: 33142995 PMCID: PMC7693844 DOI: 10.3390/nu12113338] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Ganoderma lucidum is used widely in oriental medicine to treat obesity and metabolic diseases. Bioactive substances extracted from G. lucidum have been shown to ameliorate dyslipidemia, insulin resistance, and type 2 diabetes in mice via multiple 5' AMP-activated protein kinase (AMPK)-mediated mechanisms; however, further studies are required to elucidate the anti-obesity effects of G. lucidum in vivo. In this study, we demonstrated that 3% G. lucidum extract powder (GEP) can be used to prevent obesity and insulin resistance in a mouse model. C57BL/6 mice were provided with a normal diet (ND) or a high-fat diet (HFD) supplemented with 1, 3, or 5% GEP for 12 weeks and the effect of GEP on body weight, liver, adipose tissue, adipokines, insulin and glucose tolerance (ITT and GTT), glucose uptake, glucose-metabolism related proteins, and lipogenesis related genes was examined. GEP administration was found to reduce weight gain in the liver and fat tissues of the mice. In addition, serum parameters were significantly lower in the 3% and 5% GEP mice groups than in those fed a HFD alone, whereas adiponectin levels were significantly higher. We also observed that GEP improved glucose metabolism, reduced lipid accumulation in the liver, and reduced adipocyte size. These effects may have been mediated by enhanced AMPK activation, which attenuated the transcription and translation of lipogenic genes such as fatty acid synthase (FAS), stearoyl-CoA desaturase 1 (SCD1), and sterol regulatory element-binding protein-1c (SREBP1c). Moreover, AMP-activated protein kinase (AMPK) activation increased acetyl-CoA carboxylase (ACC), insulin receptor (IR), IR substrate 1 (IRS1), and Akt protein expression and activation, as well as glucose transporter type 1/4 (GLUT1/4) protein production, thereby improving insulin sensitivity and glucose metabolism. Together, these findings demonstrate that G. lucidum may effectively prevent obesity and suppress obesity-induced insulin resistance via AMPK activation.
Collapse
Affiliation(s)
- Hyeon A Lee
- Department of Biochemistry, School of Biological Sciences, Chungbuk National University, Cheongju 28644, Korea; (H.A.L.); (Q.A.)
| | - Jae-Han Cho
- Mushroom Research Division, National Institute of Horticultural and Herbal Science, RDA Eumseong, Chungbuk 27709, Korea; (J.-H.C.); (G.-H.A.); (J.-G.H.)
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Qonita Afinanisa
- Department of Biochemistry, School of Biological Sciences, Chungbuk National University, Cheongju 28644, Korea; (H.A.L.); (Q.A.)
| | - Gi-Hong An
- Mushroom Research Division, National Institute of Horticultural and Herbal Science, RDA Eumseong, Chungbuk 27709, Korea; (J.-H.C.); (G.-H.A.); (J.-G.H.)
| | - Jae-Gu Han
- Mushroom Research Division, National Institute of Horticultural and Herbal Science, RDA Eumseong, Chungbuk 27709, Korea; (J.-H.C.); (G.-H.A.); (J.-G.H.)
| | - Hyo Jeung Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (H.J.K.); (S.H.C.); (H.-A.S.); Tel.: +82-43-261-2308 (H.-A.S.); Fax: +82-43-261-2306 (H.-A.S.)
| | - Seong Ho Choi
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Korea
- Correspondence: (H.J.K.); (S.H.C.); (H.-A.S.); Tel.: +82-43-261-2308 (H.-A.S.); Fax: +82-43-261-2306 (H.-A.S.)
| | - Hyun-A Seong
- Department of Biochemistry, School of Biological Sciences, Chungbuk National University, Cheongju 28644, Korea; (H.A.L.); (Q.A.)
- Correspondence: (H.J.K.); (S.H.C.); (H.-A.S.); Tel.: +82-43-261-2308 (H.-A.S.); Fax: +82-43-261-2306 (H.-A.S.)
| |
Collapse
|
15
|
Hashimoto T, Okada Y, Yamanaka A, Ono N, Uryu K, Maru I. The effect of eleutherococcus senticosus on metabolism-associated protein expression in 3T3-L1 and C2C12 cells. Phys Act Nutr 2020; 24:13-18. [PMID: 33108713 PMCID: PMC7669464 DOI: 10.20463/pan.2020.0016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 09/16/2020] [Indexed: 12/06/2022] Open
Abstract
[Purpose] In vivo studies have demonstrated the ergogenic benefits of eleutherococcus senticosus (ES) supplementation. ES has been observed to enhance endurance capacity, improve cardiovascular function, and alter metabolic functions (e.g., increased fat utilization); however, the exact mechanisms involved remain unknown. We aimed to determine whether ES could effectively induce fat loss and improve muscle metabolic profiles through increases in lipolysis- and lipid metabolism-associated protein expression in 3T3-L1 adipocytes and C2C12 skeletal muscle cells, respectively, to uncover the direct effects of ES on adipocytes and skeletal muscle cells. [Methods] Different doses of ES extracts (0.2, 0.5, and 1.0 mg/mL) were added to cells (0.2 ES, 0.5 ES, and 1.0 ES, respectively) for 72 h and compared to the vehicle control (control). [Results] The intracellular triacylglycerol (TG) content significantly decreased (p < 0.05 for 0.2 ES, p < 0.01 for 0.5 ES and 1.0 ES) in 3T3-L1 cells. Adipose triglyceride lipase, which is involved in active lipolysis, was significantly higher in the 1.0 ES group than in the control group (p < 0.01) of 3T3-L1 adipocytes. In C2C12 cells, the mitochondrial protein voltage-dependent anion channel (VDAC) was significantly increased in the 1.0 ES group (p < 0.01). Furthermore, we found that 1.0 ES activated both 5' AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) in skeletal muscle cells (p < 0.01). [Conclusion] These findings suggest that ES extracts decreased TG content, presumably by increasing lipase in adipocytes and metabolism-associated protein expression as well as mitochondrial biogenesis in muscle cells. These effects may corroborate previous in vivo findings regarding the ergogenic effects of ES supplementation.
Collapse
Affiliation(s)
- Takeshi Hashimoto
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Yoko Okada
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | | | | | - Keisuke Uryu
- Bizen Chemical Co., Ltd., Akaiwa, Okayama, Japan
| | - Isafumi Maru
- Bizen Chemical Co., Ltd., Akaiwa, Okayama, Japan
| |
Collapse
|
16
|
Jeju Magma-Seawater Inhibits α-MSH-Induced Melanogenesis via CaMKKβ-AMPK Signaling Pathways in B16F10 Melanoma Cells. Mar Drugs 2020; 18:md18090473. [PMID: 32962063 PMCID: PMC7551477 DOI: 10.3390/md18090473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022] Open
Abstract
Melanin protects skin from ultraviolet radiation, toxic drugs, and chemicals. Its synthesis is sophisticatedly regulated by multiple mechanisms, including transcriptional and enzymatic controls. However, uncontrolled excessive production of melanin can cause serious dermatological disorders, such as freckles, melasma, solar lentigo, and cancer. Moreover, melanogenesis disorders are also linked to neurodegenerative diseases. Therefore, there is a huge demand for safer and more potent inhibitors of melanogenesis. In the present study, we report novel inhibitory effects of Jeju magma-seawater (JMS) on melanogenesis induced by α-melanocyte stimulating hormone (α-MSH) in B16F10 melanoma cells. JMS is the abundant underground seawater found in Jeju Island, a volcanic island of Korea. Research into the physiological effects of JMS is rapidly increasing due to its high contents of various minerals that are essential to human health. However, little is known about the effects of JMS on melanogenesis. Here, we demonstrate that JMS safely and effectively inhibits α-MSH-induced melanogenesis via the CaMKKβ (calcium/calmodulin-dependent protein kinase β)-AMPK (5′ adenosine monophosphate-activated protein kinase) signaling pathway. We further demonstrate that AMPK inhibits the signaling pathways of protein kinase A and MAPKs (mitogen-activated protein kinase), which are critical for melanogenesis-related gene expression. Our results highlight the potential of JMS as a novel therapeutic agent for ameliorating skin pigmentation-related disorders.
Collapse
|
17
|
Mario L, Roberto M, Marta L, Teresa CM, Laura M. Hypothesis of COVID-19 Therapy with Sildenafil. Int J Prev Med 2020. [PMID: 32939238 DOI: 10.4103/ijpvm.ijpvm_258_20.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Backgound Bacterial or viral infections often cause acute and severe systemic inflammation, which affects the lungs lipopolysaccharide (LPS), a pathogenic component of the membrane of gram-negative bacteria, stimulates active innate immune cells, monocytes, macrophages to produce inducible nitric oxide synthase (iNOS). Excess production of this compound occurs in COVID-19 resulting in inflammatory cascade and thromboembolism. We intend to propose the use of sildenafil to reduce this production. Method The analysis of biochemical pathways shows that viral infection produces a high amount of nitric oxide (NO), with an acute inflammatory process. Results In the case of COVID-19 infection we verified that numerous biochemical processes activate a cascade of inflammatory processes through the activation of iNOS with uncontrolled generation of (NO). Conclusions iNOS is the cause of damage to host cells with a consequent pulmonary thromboembolic lung phenomenon in a contest of interstitial pneumonia. This study proposes the use of sildenafil to counter the inflammatory cascade and thromboembolic episodes.
Collapse
Affiliation(s)
- Limodio Mario
- Researcher, UOC, Infectious and Tropical Diseases, Spaziani Hospital, Frosinone, Italy
| | | | | | | | - Menicagli Laura
- Department of Radiodiagnostic, Policliclinico San Donato, Milan University, Italy
| |
Collapse
|
18
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
19
|
Mario L, Roberto M, Marta L, Teresa CM, Laura M. Hypothesis of COVID-19 Therapy with Sildenafil. Int J Prev Med 2020; 11:76. [PMID: 32939238 PMCID: PMC7478270 DOI: 10.4103/ijpvm.ijpvm_258_20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/10/2020] [Indexed: 11/04/2022] Open
Abstract
Backgound Bacterial or viral infections often cause acute and severe systemic inflammation, which affects the lungs lipopolysaccharide (LPS), a pathogenic component of the membrane of gram-negative bacteria, stimulates active innate immune cells, monocytes, macrophages to produce inducible nitric oxide synthase (iNOS). Excess production of this compound occurs in COVID-19 resulting in inflammatory cascade and thromboembolism. We intend to propose the use of sildenafil to reduce this production. Method The analysis of biochemical pathways shows that viral infection produces a high amount of nitric oxide (NO), with an acute inflammatory process. Results In the case of COVID-19 infection we verified that numerous biochemical processes activate a cascade of inflammatory processes through the activation of iNOS with uncontrolled generation of (NO). Conclusions iNOS is the cause of damage to host cells with a consequent pulmonary thromboembolic lung phenomenon in a contest of interstitial pneumonia. This study proposes the use of sildenafil to counter the inflammatory cascade and thromboembolic episodes.
Collapse
Affiliation(s)
- Limodio Mario
- Researcher, UOC, Infectious and Tropical Diseases, Spaziani Hospital, Frosinone, Italy
| | | | | | | | - Menicagli Laura
- Department of Radiodiagnostic, Policliclinico San Donato, Milan University, Italy
| |
Collapse
|
20
|
Park M, Han J, Lee HJ. Anti-Adipogenic Effect of Neferine in 3T3-L1 Cells and Primary White Adipocytes. Nutrients 2020; 12:nu12061858. [PMID: 32580414 PMCID: PMC7353287 DOI: 10.3390/nu12061858] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Neferine, an alkaloid component extracted from lotus seed embryos, is known for its anti-inflammatory, anticancer, and antioxidant properties. However, the anti-adipogenic activity of neferine has not been thoroughly investigated. In this study, neferine was found to inhibit lipid accumulation in a dose-dependent manner during the differentiation of 3T3-L1 cells without inducing cytotoxicity. Real-time polymerase chain reaction and immunoblot analysis revealed the downregulation in the expression of peroxisome proliferator activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein alpha (C/EBPα), sterol regulatory element-binding protein-1c (SREBP-1c), and fatty acid synthase (FAS) and the upregulation in carnitine palmitoyltransferase-1 (CPT-1) and sirtuin 1 (SIRT1) levels following neferine treatment. Furthermore, neferine increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), which is an important regulator of fatty acid oxidation. Our result indicates that neferine attenuates adipogenesis and promotes lipid metabolism by activating AMPK-mediated signaling. Therefore, neferine may serve as a therapeutic candidate for obesity treatment.
Collapse
Affiliation(s)
- Miey Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Gyeonggi-do 13120, Korea; (M.P.); (J.H.)
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
| | - Jinyoung Han
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Gyeonggi-do 13120, Korea; (M.P.); (J.H.)
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Gyeonggi-do 13120, Korea; (M.P.); (J.H.)
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
- Correspondence: ; Tel.: +82-31-750-5968; Fax: +82-31-724-4411
| |
Collapse
|
21
|
Yoshikawa M, Hosokawa M, Miyashita K, Fujita T, Nishino H, Hashimoto T. Fucoxanthinol attenuates oxidative stress-induced atrophy and loss in myotubes and reduces the triacylglycerol content in mature adipocytes. Mol Biol Rep 2020; 47:2703-2711. [PMID: 32180086 DOI: 10.1007/s11033-020-05369-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
The combination of sarcopenia and obesity (i.e., sarcopenic obesity) is more strongly associated with disability and metabolic/cardiovascular diseases than obesity or sarcopenia alone. Therefore, countermeasures that simultaneously suppress fat gain and muscle atrophy to prevent an increase in sarcopenic obesity are warranted. The aim of this study was to investigate the simultaneous effects of fucoxanthinol (FXOH) on fat loss in mature adipocytes and the inhibition of atrophy and loss in myotubes induced by oxidative stress. C2C12 myotubes were treated with FXOH for 24 h and further incubated with hydrogen peroxide (H2O2) for 24 h. The area of myosin heavy chain-positive myotubes and the ROS concentration were measured. Mature 3T3-L1 adipocytes were treated with FXOH for 72 h. The triacylglycerol (TG) content and glycerol and fatty acid (FA) release were biochemically measured. The myotube area was smaller in H2O2-treated cells than that in control cells. However, FXOH protected against the H2O2-induced decreases in myotube area. Further, the ROS concentration was significantly higher in the FXOH-treated cells compared with that in the control cells, although it was significantly lower than that in the H2O2-treated cells. On the other hand, in the mature adipocytes, the TG content was significantly decreased by FXOH treatment compared to that in the control. Moreover, FXOH treatment significantly increased glycerol and FA release compared with that of the control. These results suggest that FXOH inhibits H2O2-induced atrophy and loss in myotubes and activates lipolysis and decreases the TG content in mature adipocytes. Accordingly, FXOH has the potential to exert anti-sarcopenic obesity effects.
Collapse
Affiliation(s)
- Maki Yoshikawa
- Faculty of Sport and Health Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Masashi Hosokawa
- Faculty of Fisheries Sciences, Hokkaido University, Hokkaido, Japan
| | - Kazuo Miyashita
- Faculty of Fisheries Sciences, Hokkaido University, Hokkaido, Japan
| | - Takashi Fujita
- Faculty of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | | | - Takeshi Hashimoto
- Faculty of Sport and Health Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
22
|
Kunjiappan S, Theivendren P, Pavadai P, Govindaraj S, Sankaranarayanan M, Somasundaram B, Arunachalam S, Ram Kumar Pandian S, Ammunje DN. Design and in silico modeling of Indoloquinoxaline incorporated keratin nanoparticles for modulation of glucose metabolism in 3T3-L1 adipocytes. Biotechnol Prog 2019; 36:e2904. [PMID: 31496124 DOI: 10.1002/btpr.2904] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/25/2022]
Abstract
The following study was done to assess the glucose utilizing efficiency of Indoloquinoxaline derivative incorporated keratin nanoparticles (NPs) in 3T3-L1 adipocytes. Indoloquinoxaline derivative had wide range of biological activities including antidiabetic activity. In this view, Indoloquinoxaline moiety containing N, N-dimethyl (3-fluoro-6H-indolo [3,2-b] quinoxalin-6-yl) methanamine compound was designed and synthesized, and further it is incorporated into keratin nanoparticles. The formulated NPs, drug entrapment efficiency, releasing capacity, stability, and physicochemical properties were characterized by various spectral analyzer and obtained results of characterizations were confirmed the properties of NPs. The analysis of mechanism underlying the glucose utilization of NPs was examined through molecular docking with identified target, and observed in silico study reports shown strong interaction of NPs in the binding pockets of AMPK and PTP1B. Based on the in silico screening, the formulated NPs was performed for in vitro cellular viability and glucose uptake studies on 3T3-L1 adipocytes. Interestingly, 40 μg of NPs displayed 78.2 ± 2.76% cellular viability, and no cell death was observed at lower concentrations. Further, the concentration dependent glucose utilization was observed at different concentrations of NPs in 3T3-L1 adipocytes. The results of NPs (40 μg) on glucose utilization have revealed eminent result 58.56 ± 4.54% compared to that of Metformin (10 μM) and Insulin (10 μM). The identified results clearly indicated that Indoloquinoxaline derivative incorporated keratin NPs significantly increased glucose utilization efficiency and protect the cells against the insulin resistance.
Collapse
Affiliation(s)
- Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu, India
| | | | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M S Ramaiah University of Applied Sciences, Bengaluru, Karnataka, India
| | - Saravanan Govindaraj
- Department of Pharmaceutical Chemistry, MNR College of Pharmacy, Sangareddy, Telangana, India
| | | | - Balasubramanian Somasundaram
- Sir CV Raman-KS Krishnan International Research Center, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu, India
| | - Sankarganesh Arunachalam
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu, India
| | | | - Damodar Nayak Ammunje
- Department of Pharmacology, Faculty of Pharmacy, M S Ramaiah University of Applied Sciences, Bengaluru, Karnataka, India
| |
Collapse
|
23
|
Bian Y, Li X, Li X, Ju J, Liang H, Hu X, Dong L, Wang N, Li J, Zhang Y, Yang B. Daming capsule, a hypolipidaemic drug, lowers blood lipids by activating the AMPK signalling pathway. Biomed Pharmacother 2019; 117:109176. [DOI: 10.1016/j.biopha.2019.109176] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/18/2019] [Accepted: 06/25/2019] [Indexed: 02/01/2023] Open
|
24
|
Lima RDCL, Berg RS, Rønning SB, Afseth NK, Knutsen SH, Staerk D, Wubshet SG. Peptides from chicken processing by-product inhibit DPP-IV and promote cellular glucose uptake: potential ingredients for T2D management. Food Funct 2019; 10:1619-1628. [PMID: 30821796 DOI: 10.1039/c8fo02450b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inhibition of dipeptidyl peptidase IV (DPP-IV) and stimulation of muscle glucose uptake are two of the key strategies for management of type-2-diabetes (T2D). In the present study, four protein hydrolysates generated by enzymatic hydrolysis of chicken by-product, i.e., mechanical chicken deboning residue, were evaluated for their DPP-IV inhibitory activity as well as their effect on glucose uptake by skeletal muscle cells. The DPP-IV inhibitory assay was performed at two concentrations (1000 μg mL-1 and 10 μg mL-1) for the crude chicken protein hydrolysates. The hydrolysate with the highest DPP-IV inhibition was selected for preparative-scale fractionation using size-exclusion chromatography (SEC). The SEC fractions were tested for DPP-IV inhibitory activity as well as their effect on glucose uptake and metabolic activity of skeletal muscle cells. The muscle cells were treated with the SEC fractions and glucose uptake was measured based on luminescence detection of 2-deoxyglucose-6-phosphate (2DG6P). A fraction with peptides in the lower molecular weight range was shown to promote glucose uptake and to inhibit DPP-IV. Further chromatographic fractionation followed by inhibition assaying of the most potent SEC fraction led to isolation of five refined peptide fractions with more than 80% DPP-IV inhibition, which were subsequently analyzed with LC-HRMS/MS. This led to identification of 14 peptides as potential DPP-IV inhibitors from protein hydrolysates of mechanical chicken deboning residue.
Collapse
|
25
|
MicroRNA-451 Inhibits Migration of Glioblastoma while Making It More Susceptible to Conventional Therapy. Noncoding RNA 2019; 5:ncrna5010025. [PMID: 30875963 PMCID: PMC6468936 DOI: 10.3390/ncrna5010025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/18/2022] Open
Abstract
Malignant glioblastoma (GBM, glioma) is the most common and aggressive primary adult brain tumor. The prognosis of GBM patients remains poor, despite surgery, radiation and chemotherapy. The major obstacles for successful remedy are invasiveness and therapy resistance of GBM cells. Invasive glioma cells leave primary tumor core and infiltrate surrounding normal brain leading to inevitable recurrence, even after surgical resection, radiation and chemotherapy. Therapy resistance allowing for selection of more aggressive and resistant sub-populations including GBM stem-like cells (GSCs) upon treatment is another serious impediment to successful treatment. Through their regulation of multiple genes, microRNAs can orchestrate complex programs of gene expression and act as master regulators of cellular processes. MicroRNA-based therapeutics could thus impact broad cellular programs, leading to inhibition of invasion and sensitization to radio/chemotherapy. Our data show that miR-451 attenuates glioma cell migration in vitro and invasion in vivo. In addition, we have found that miR-451 sensitizes glioma cells to conventional chemo- and radio-therapy. Our data also show that miR-451 is regulated in vivo by AMPK pathway and that AMPK/miR-451 loop has the ability to switch between proliferative and migratory pattern of glioma cells behavior. We therefore postulate that AMPK/miR-451 negative reciprocal feedback loop allows GBM cells/GSCs to adapt to tumor “ecosystem” by metabolic and behavioral flexibility, and that disruption of such a loop reduces invasiveness and diminishes therapy resistance.
Collapse
|
26
|
Zha D, Yao T, Bao L, Gao P, Wu X. Telmisartan attenuates diabetic nephropathy progression by inhibiting the dimerization of angiotensin type-1 receptor and adiponectin receptor-1. Life Sci 2019; 221:109-120. [DOI: 10.1016/j.lfs.2019.01.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/25/2019] [Accepted: 01/26/2019] [Indexed: 02/07/2023]
|
27
|
Cam ME, Hazar-Yavuz AN, Yildiz S, Ertas B, Ayaz Adakul B, Taskin T, Alan S, Kabasakal L. The methanolic extract of Thymus praecox subsp. skorpilii var. skorpilii restores glucose homeostasis, ameliorates insulin resistance and improves pancreatic β-cell function on streptozotocin/nicotinamide-induced type 2 diabetic rats. JOURNAL OF ETHNOPHARMACOLOGY 2019; 231:29-38. [PMID: 30399410 DOI: 10.1016/j.jep.2018.10.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 10/15/2018] [Accepted: 10/21/2018] [Indexed: 05/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Thymus praecox subsp. skorpilii var. skorpilii (syn. Thymus praecox subsp. jankae (Celak.) Jalas) is consumed as a Turkish folk medicine for the treatment of spasm, sore throat and shortness of breath, also having strong antioxidant activity and the leaves of the plant have been utilized for the treatment of diabetes as the decoction in Turkey. AIM OF THE STUDY In the present study, we aimed to investigate the potential mechanism of antidiabetic action of Thymus praecox subsp. skorpilii var. skorpilii methanolic extract (TPSE) on streptozotocin (STZ)/nicotinamide (NA)-induced type 2 diabetic rats. MATERIALS AND METHODS Sprague Dawley rats were randomly divided into four groups; control, diabetes, TPSE (100 mg/kg b.w, p.o.) and metformin group (400 mg/kg b.w, p.o.). Diabetes was established in all groups except control group by 55 mg/kg STZ (i.p.) for once 15 min after 100 mg/kg NA injection. 3 days after STZ/NA injection, treatments were administered for three weeks and then rats were decapitated; tissue and blood samples were obtained for measuring the level of glucose transporters (both GLUTs and sodium glucose co-transporters (SGLTs)), enzymes related to glucose (Hexokinase (HK), phosphoenolpyruvate carboxykinase (PEPCK), α-glucosidase) and lipid metabolism (Acetyl-coenzyme carboxylase (ACC)), AST, ALT, creatinine, insulin, anti-inflammatory (IL-10) and inflammatory (TNF-α, IL-1β, IL-6) cytokines, AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptor gamma (PPAR-γ) and glucagon like peptide-1 (GLP-1). Histopathological alterations of the pancreas were examined. RESULTS After three weeks of treatment, TPSE has exhibited a significant reduction of plasma levels of the proinflammatory cytokines. Besides, TPSE treatment elevated plasma insulin levels and normalized blood glucose levels. Moreover, it improved the values of AMPK in liver and GLP-1 in pancreas. Increased α-glucosidase, PEPCK, GLUT-2 and SGLTs levels with the induction of diabetes considerably lowered with TPSE treatment. Especially on SGLT-2, TPSE achieved a more prominent decrease. After the atrophy in Langerhans islets due to diabetes induction, treatment was found to prevent the damage of islets. CONCLUSIONS Based on the findings presented here, it has been concluded that TPSE has marked antidiabetic effects through various pathways on STZ/NA-induced diabetic rats and it may potentially be used as an effective treatment for type 2 diabetes mellitus (T2DM). Further research on isolation of the bioactive components is underway.
Collapse
Affiliation(s)
- Muhammet Emin Cam
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Haydarpasa, 34668 Istanbul, Turkey; Department of Mechanical Engineering, University College London, Torrington Place, WC1E 7JE London, UK; Advanced Nanomaterials Research Laboratory, Faculty of Technology, Marmara University, Goztepe, 34722 Istanbul, Turkey.
| | - Ayse Nur Hazar-Yavuz
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Haydarpasa, 34668 Istanbul, Turkey.
| | - Sila Yildiz
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Haydarpasa, 34668 Istanbul, Turkey.
| | - Busra Ertas
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Haydarpasa, 34668 Istanbul, Turkey.
| | - Betul Ayaz Adakul
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Haydarpasa, 34668 Istanbul, Turkey.
| | - Turgut Taskin
- Department of Pharmacognosy, Faculty of Pharmacy, Marmara University, Haydarpasa, 34668 Istanbul, Turkey.
| | - Saadet Alan
- Department of Pathology, Faculty of Medicine, Inonu University, Malatya, Turkey.
| | - Levent Kabasakal
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Haydarpasa, 34668 Istanbul, Turkey.
| |
Collapse
|
28
|
Global phosphoproteomic analysis reveals ARMC10 as an AMPK substrate that regulates mitochondrial dynamics. Nat Commun 2019; 10:104. [PMID: 30631047 PMCID: PMC6328551 DOI: 10.1038/s41467-018-08004-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 12/03/2018] [Indexed: 01/06/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is a key regulator of cellular energy homeostasis. Although AMPK has been studied extensively in cellular processes, understanding of its substrates and downstream functional network, and their contributions to cell fate and disease development, remains incomplete. To elucidate the AMPK-dependent signaling pathways, we performed global quantitative phosphoproteomic analysis using wild-type and AMPKα1/α2-double knockout cells and discovered 160 AMPK-dependent phosphorylation sites. Further analysis using an AMPK consensus phosphorylation motif indicated that 32 of these sites are likely direct AMPK phosphorylation sites. We validated one uncharacterized protein, ARMC10, and demonstrated that the S45 site of ARMC10 can be phosphorylated by AMPK both in vitro and in vivo. Moreover, ARMC10 overexpression was sufficient to promote mitochondrial fission, whereas ARMC10 knockout prevented AMPK-mediated mitochondrial fission. These results demonstrate that ARMC10 is an effector of AMPK that participates in dynamic regulation of mitochondrial fission and fusion. AMPK is regulates cellular energy and has been extensively studied, although our knowledge of downstream substrates is incomplete. Here, Chen et al. perform global quantitative analysis for AMPK-dependent sites and validate one hit, ARMC10, as a direct AMPK effector of mitochondrial dynamics.
Collapse
|
29
|
Zhang JY, Li CJ, Zhang QM, Yu P, Shi JY, Tang GJ, Ma LL, Yu DM. Protective Effects of Reduced Beta 2 Glycoprotein I on Liver Injury in Streptozotocin (STZ)-Diabetic Rats by Activation of AMP-Activated Protein Kinase. Med Sci Monit 2018; 24:7577-7584. [PMID: 30352988 PMCID: PMC6210935 DOI: 10.12659/msm.909598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Protective effects of reduced beta 2 glycoprotein I (Rβ2GPI) against vascular injury of diabetes mellitus have been extensively investigated. However, the effects of Rβ2GPI on liver injury in diabetic animals have not been reported. Material/Methods A diabetic rat model of was produced by systemic injection of streptozotocin (STZ). Rats were divided into a normal control group, a model group, and an Rβ2GPI treatment group (N=6 in each group). After treatments, blood serum and liver tissue were collected to test the protection of Rβ2GPI. AMP-activated protein kinase (AMPK) was detected by immunohistochemistry and Western blotting. Results Our results revealed that Rβ2GPI reduced blood glucose, serum creatinine, and urea nitrogen levels, as well as serum inflammation cytokines, including interleukin (IL)-6, tumor necrosis factor (TNF)-a and C-reactive protein in the diabetic rats. Importantly, Rβ2GPI prevented liver injury in the diabetic rats as confirmed by hematoxylin-eosin (H&E) staining, alanine transaminase, aspartate transaminase, and gamma-glutamyl transferase. Reactive oxygen species (ROS) were promoted by diabetic modeling and were attenuated by Rβ2GPI administration. Moreover, Rβ2GPI significantly reduced liver catalase, malondialdehyde, and superoxide dismutase levels in the diabetic rats. Rβ2GPI reduced liver glycolipid storage in STZ diabetic rats. Both immunohistochemistry and Western blotting demonstrated that Rβ2GPI promoted AMPK phosphorylation in the diabetic rats. Conclusions Our data proved that Rβ2GPI prevented liver injury in diabetic rats, likely through activating the AMPK signaling pathway.
Collapse
Affiliation(s)
- Jing-Yun Zhang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| | - Chun-Jun Li
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| | - Qiu-Mei Zhang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| | - Pei Yu
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| | - Jian-Ying Shi
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| | - Guang-Jie Tang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| | - Lin-Lin Ma
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| | - De-Min Yu
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China (mainland)
| |
Collapse
|
30
|
de Bem GF, Costa CA, Santos IB, Cristino Cordeiro VDS, de Carvalho LCRM, de Souza MAV, Soares RDA, Sousa PJDC, Ognibene DT, Resende AC, de Moura RS. Antidiabetic effect of Euterpe oleracea Mart. (açaí) extract and exercise training on high-fat diet and streptozotocin-induced diabetic rats: A positive interaction. PLoS One 2018; 13:e0199207. [PMID: 29920546 PMCID: PMC6007924 DOI: 10.1371/journal.pone.0199207] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022] Open
Abstract
A growing body of evidence suggests a protective role of polyphenols and exercise training on the disorders of type 2 diabetes mellitus (T2DM). We aimed to assess the effect of the açaí seed extract (ASE) associated with exercise training on diabetic complications induced by high-fat (HF) diet plus streptozotocin (STZ) in rats. Type 2 diabetes was induced by feeding rats with HF diet (55% fat) for 5 weeks and a single dose of STZ (35 mg/kg i.p.). Control (C) and Diabetic (D) animals were subdivided into four groups each: Sedentary, Training, ASE Sedentary, and ASE Training. ASE (200 mg/kg/day) was administered by gavage and the exercise training was performed on a treadmill (30min/day; 5 days/week) for 4 weeks after the diabetes induction. In type 2 diabetic rats, the treatment with ASE reduced blood glucose, insulin resistance, leptin and IL-6 levels, lipid profile, and vascular dysfunction. ASE increased the expression of insulin signaling proteins in skeletal muscle and adipose tissue and plasma GLP-1 levels. ASE associated with exercise training potentiated the reduction of glycemia by decreasing TNF-α levels, increasing pAKT and adiponectin expressions in adipose tissue, and IR and pAMPK expressions in skeletal muscle of type 2 diabetic rats. In conclusion, ASE treatment has an antidiabetic effect in type 2 diabetic rats by activating the insulin-signaling pathway in muscle and adipose tissue, increasing GLP-1 levels, and an anti-inflammatory action. Exercise training potentiates the glucose-lowering effect of ASE by activating adiponectin-AMPK pathway and increasing IR expression.
Collapse
Affiliation(s)
- Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Cristiane Aguiar Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Izabelle Barcellos Santos
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | | | | | | | - Ricardo de Andrade Soares
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | | | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Angela Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
- * E-mail: ,
| | - Roberto Soares de Moura
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
31
|
Bao X, Zhao L, Guan H, Li F. Inhibition of LCMR1 and ATG12 by demethylation-activated miR-570-3p is involved in the anti-metastasis effects of metformin on human osteosarcoma. Cell Death Dis 2018; 9:611. [PMID: 29795113 PMCID: PMC5966512 DOI: 10.1038/s41419-018-0620-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/06/2018] [Accepted: 04/20/2018] [Indexed: 01/02/2023]
Abstract
Epidemiological studies have demonstrated that metformin could mitigate the progression of several tumors. Although it has been proved that metformin could cause demethylation of DNA and lead to up-regulation of some encoding genes and non-coding RNAs, there is little data about the effects of metformin on metastasis, and the interaction between metastasis and autophagy in human osteosarcoma cells. Here, we found miR-570-3p was significantly down-regulated in human metastatic osteosarcoma tissues but not in non-metastatic osteosarcoma tissues. Metformin attenuates the metastasis and autophagy in osteosarcoma. Interestingly, this autophagy favors osteosarcoma cells invasion. Moreover, reduction of metformin-induced inhibition of autophagy could reverse the invasion suppression in osteosarcoma. Mechanistically, metformin increases miR-570-3p by the demethylation of DNA, and the upregulation of miR-570-3p repressed the translation of its target, LCMR1 and ATG12. Our results, for the first time, presents evidence that the miR-570-3p-mediated suppression of LCMR1 and ATG12 is involved in the metformin-induced inhibition of metastasis in osteosarcoma cells.
Collapse
Affiliation(s)
- Xing Bao
- Department of Orthopedics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jiefang Ave, Wuhan, 430030, People's Republic of China
| | - Libo Zhao
- Department of Orthopedics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jiefang Ave, Wuhan, 430030, People's Republic of China
| | - Hanfeng Guan
- Department of Orthopedics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jiefang Ave, Wuhan, 430030, People's Republic of China
| | - Feng Li
- Department of Orthopedics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jiefang Ave, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
32
|
Li W, Ji M, Lin Y, Miao Y, Chen S, Li H. DEPP/DEPP1/C10ORF10 regulates hepatic glucose and fat metabolism partly via ROS-induced FGF21. FASEB J 2018; 32:5459-5469. [PMID: 29702025 DOI: 10.1096/fj.201800357r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Decidual protein induced by progesterone (DEPP/DEPP1/C10ORF10) is induced by denying access to food and reduced by refeeding in insulin-sensitive organs in vivo. The negative regulation of DEPP by insulin is also proven in several cell lines. However, the functions of DEPP in insulin-sensitive organs remain unknown. In the present study, we investigated the impact of DEPP on hepatic energy metabolism and addressed the underlying mechanisms. The metabolic effects of DEPP were investigated in mice with adenovirus-mediated hepatic overexpression. Liver triglyceride (TG), glycogen, and serum metabolites were detected by biochemical assays. Energy homeostasis was measured by indirect calorimetry. Quantitative PCR was used to examine expression of genes involved in fatty acid oxidation, ketogenesis, lipogenesis, and gluconeogenesis. To evaluate the role of fibroblast growth factor 21 (FGF21) mediating the metabolic effects of DEPP, FGF21 antibody was administrated intraperitoneally to mice at 24 h after the delivery of adenovirus, and the metabolic alterations were examined. Reactive oxygen species (ROS) levels were measured by catalase activity assay, live cell fluorescence, or quantitative PCR. Effects of DEPP on the phenotype of db/db mice were also assessed. Acute hepatic overexpression of DEPP significantly reduced serum glucose and TG levels, dramatically elevated β-hydroxybutyrate levels, and improved glucose clearance. Compared with controls, DEPP overexpression reduced food intake, the energy expenditure rate, and the respiratory quotient. DEPP overexpression significantly increased fatty acid oxidation and ketogenesis but suppressed lipid synthesis and gluconeogenesis. Investigations of the underlying mechanisms revealed that DEPP regulates energy metabolism by inducing oxidative stress. With the impairment of the ROS scavenging system and promotion of ROS formation, DEPP overexpression leads to ROS accumulation. FGF21 is upregulated in response to oxidative stress and mediates the effects of DEPP on fatty acid oxidation, ketogenesis, and lipid synthesis but not gluconeogenesis, as evidenced by the fact that the FGF21 antibody dramatically suppressed a DEPP-induced increase of fatty acid oxidation and ketogenesis, reversed the reduction of lipid synthesis, but did not change the suppression of gluconeogenesis. Moreover, overexpression of DEPP in db/ db mice led to a marked reduction in body weight and serum glucose levels and significantly improved insulin sensitivity. Hepatic overexpression of DEPP in mice promotes fatty acid oxidation and ketogenesis and suppresses lipogenesis and gluconeogenesis, which is partly mediated by FGF21 induced by elevated cellular ROS levels.-Li, W., Ji, M., Lin, Y., Miao, Y., Chen, S., Li, H. DEPP/DEPP1/C10ORF10 regulates hepatic glucose and fat metabolism partly via ROS-induced FGF21.
Collapse
Affiliation(s)
- Wenli Li
- Department of Pathophysiology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
| | - Meiling Ji
- Department of Pathophysiology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
| | - Yandie Lin
- Department of Pathophysiology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
| | - Yi Miao
- Department of Pathophysiology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
| | - Simin Chen
- Department of Pathophysiology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
| | - Hao Li
- Department of Pathophysiology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Naceiri Mrabti N, Elhallaoui M. QSAR study and molecular docking of benzimidazole derivatives as potent activators of AMP-activated protein kinase. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2018. [DOI: 10.1016/j.jtusci.2016.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Nidal Naceiri Mrabti
- Engineering Materials, Modeling and Environmental Laboratory, Faculty of Science, University Sidi Mohammed Ben Abdellah Dhar Mehraz, B.P. 1796, Atlas Fes Morocco
| | - Menana Elhallaoui
- Department of Chemistry, Faculty of Science, University Sidi Mohammed Ben Abdellah Dhar Mehraz, B.P. 1796, AtlasFes Morocco
| |
Collapse
|
34
|
Kang MC, Ding Y, Kim J, Kim EA, Fernando IPS, Heo SJ, Lee SH. 3-Chloro-4,5-dihydroxybenzaldehyde inhibits adipogenesis in 3T3-L1 adipocytes by regulating expression of adipogenic transcription factors and AMPK activation. Chem Biol Interact 2018; 287:27-31. [PMID: 29630878 DOI: 10.1016/j.cbi.2018.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/26/2018] [Accepted: 04/05/2018] [Indexed: 02/06/2023]
Abstract
Obesity is a serious health issue in many industrialized countries. It is a medical condition with excessive levels of fat accumulated in adipocytes. The objective of the present study was to determine the inhibitory effect of 3-chloro-4,5-dihydroxybenzaldehyde (CDB) on adipogenesis in 3T3-L1 adipocyte cells. CDB suppressed the differentiation and decreased lipid accumulation and triglycerides contents in 3T3-L1 adipocytes. Its suppression effect on fat accumulation was mediated via expression of adipogenesis factors (C/EBPα, SREBP-1c, PPARγ, and adiponectin) during adipocyte differentiation in white adipocyte cells. CDB's ability to suppress fat accumulation was increased in a concentration-dependent manner. It inhibited fatty acid synthesis related proteins including FAS, FABP4, leptin, and perilipin. It also increased expression of phosphorylated AMPK in adipocytes cells. These observations suggest that CDB has potential anti-obesity effect with ability to improve metabolic diseases.
Collapse
Affiliation(s)
- Min-Cheol Kang
- Department of Marine Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yuling Ding
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Junseong Kim
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science &Technology (KIOST), Jeju, 63349, Republic of Korea
| | - Eun-A Kim
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science &Technology (KIOST), Jeju, 63349, Republic of Korea
| | - I P Shanura Fernando
- Department of Marine Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Soo-Jin Heo
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science &Technology (KIOST), Jeju, 63349, Republic of Korea.
| | - Seung-Hong Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea.
| |
Collapse
|
35
|
Sutjarit N, Sueajai J, Boonmuen N, Sornkaew N, Suksamrarn A, Tuchinda P, Zhu W, Weerachayaphorn J, Piyachaturawat P. Curcuma comosa reduces visceral adipose tissue and improves dyslipidemia in ovariectomized rats. JOURNAL OF ETHNOPHARMACOLOGY 2018; 215:167-175. [PMID: 29273438 DOI: 10.1016/j.jep.2017.12.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcuma comosa Roxb. (C. comosa) or Wan chak motluk Zingiberaceae family, is widely used in Thai traditional medicine for treatment of gynecological problems as well as relief of postmenopausal symptoms. Since C. comosa contains phytoestrogen and causes lipid lowering effect by an unknown mechanism, we investigated its effect on adiposity and lipid metabolism in estrogen-deprived rats. MATERIALS AND METHODS Adult female rats were ovariectomized (OVX) and received daily doses of either a phytoestrogen from C. comosa [(3R)-1,7-diphenyl-(4E,6E)-4,6-heptadien-3-ol; DPHD], C. comosa extract, or estrogen (17β-estradiol; E2) for 12 weeks. Adipose tissue mass, serum levels of lipids and adipokines were determined. In addition, genes and proteins involved in lipid synthesis and fatty acid oxidation in visceral adipose tissue were analyzed. RESULTS Ovariectomy for 12 weeks elevated level of serum lipids and increased visceral fat mass and adipocyte size. These alterations were accompanied with the up-regulation of lipogenic mRNA and protein expressions including LXR-α, SREBP1c and their downstream targets. OVX rats showed decrease in proteins involved in fatty acid oxidation including AMPK-α and PPAR-α in adipose tissue, as well as alteration of adipokines; leptin and adiponectin. Treatments with E2, DPHD or C. comosa extract in OVX rats prevented an increase in adiposity, down-regulated lipogenic genes and proteins with marked increases in the protein levels of AMPK-α and PPAR-α. These findings indicated that their lipid lowering effects were mediated via the suppression of lipid synthesis in concert with an increase in fatty acid oxidation. CONCLUSIONS C. comosa exerts a lipid lowering effect in the estrogen deficient rats through the modulations of lipid synthesis and AMPK-α activity in adipose tissues, supporting the use of this plant for health promotion in the post-menopausal women.
Collapse
Affiliation(s)
- Nareerat Sutjarit
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Jetjamnong Sueajai
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Nittaya Boonmuen
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Nilubon Sornkaew
- Department of Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Patoomratana Tuchinda
- Department of Chemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Weiming Zhu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | | | - Pawinee Piyachaturawat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
36
|
Haczeyni F, Bell-Anderson KS, Farrell GC. Causes and mechanisms of adipocyte enlargement and adipose expansion. Obes Rev 2018; 19:406-420. [PMID: 29243339 DOI: 10.1111/obr.12646] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/28/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
Abstract
Adipose tissue plays a significant role in whole body energy homeostasis. Obesity-associated diabetes, fatty liver and metabolic syndrome are closely linked to adipose stress and dysfunction. Genetic predisposition, overeating and physical inactivity influence the expansion of adipose tissues. Under conditions of constant energy surplus, adipocytes become hypertrophic and adipose tissues undergo hyperplasia so as to increase their lipid storage capacity, thereby keeping circulating blood glucose and fatty acids below toxic levels. Nonetheless, adipocytes have a saturation point where they lose capacity to store more lipids. At this stage, when adipocytes are fully lipid-engorged, they express stress signals. Adipose depots (particularly visceral compartments) from obese individuals with a severe metabolic phenotype are characterized by the high proportion of hypertrophic adipocytes. This review focuses on the mechanisms of adipocyte enlargement in relation to adipose fatty acid and cholesterol metabolism, and considers how this may be related to adipose dysfunction.
Collapse
Affiliation(s)
- F Haczeyni
- Liver Research Group, Australian National University Medical School at The Canberra Hospital, Canberra, ACT, Australia
| | - K S Bell-Anderson
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - G C Farrell
- Liver Research Group, Australian National University Medical School at The Canberra Hospital, Canberra, ACT, Australia
| |
Collapse
|
37
|
Pulito C, Mori F, Sacconi A, Goeman F, Ferraiuolo M, Pasanisi P, Campagnoli C, Berrino F, Fanciulli M, Ford RJ, Levrero M, Pediconi N, Ciuffreda L, Milella M, Steinberg GR, Cioce M, Muti P, Strano S, Blandino G. Metformin-induced ablation of microRNA 21-5p releases Sestrin-1 and CAB39L antitumoral activities. Cell Discov 2017; 3:17022. [PMID: 28698800 PMCID: PMC5501975 DOI: 10.1038/celldisc.2017.22] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/05/2017] [Accepted: 06/01/2017] [Indexed: 12/16/2022] Open
Abstract
Metformin is a commonly prescribed type II diabetes medication that exhibits promising anticancer effects. Recently, these effects were found to be associated, at least in part, with a modulation of microRNA expression. However, the mechanisms by which single modulated microRNAs mediate the anticancer effects of metformin are not entirely clear and knowledge of such a process could be vital to maximize the potential therapeutic benefits of this safe and well-tolerated therapy. Our analysis here revealed that the expression of miR-21-5p was downregulated in multiple breast cancer cell lines treated with pharmacologically relevant doses of metformin. Interestingly, the inhibition of miR-21-5p following metformin treatment was also observed in mouse breast cancer xenografts and in sera from 96 breast cancer patients. This modulation occurred at the levels of both pri-miR-21 and pre-miR-21, suggesting transcriptional modulation. Antagomir-mediated ablation of miR-21-5p phenocopied the effects of metformin on both the clonogenicity and migration of the treated cells, while ectopic expression of miR-21-5p had the opposite effect. Mechanistically, this reduction in miR-21-5p enhanced the expression of critical upstream activators of the AMP-activated protein kinase, calcium-binding protein 39-like and Sestrin-1, leading to AMP-activated protein kinase activation and inhibition of mammalian target of rapamycin signaling. Importantly, these effects of metformin were synergistic with those of everolimus, a clinically relevant mammalian target of rapamycin inhibitor, and were independent of the phosphatase and tensin homolog status. This highlights the potential relevance of metformin in combinatorial settings for the treatment of breast cancer.
Collapse
Affiliation(s)
- Claudio Pulito
- Molecular Chemoprevention Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Federica Mori
- Molecular Chemoprevention Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Andrea Sacconi
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Frauke Goeman
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Maria Ferraiuolo
- Molecular Chemoprevention Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Patrizia Pasanisi
- Department of Preventive & Predictive Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Carlo Campagnoli
- Unit of Endocrinological Gynecology, Ospedale Sant’Anna di Torino, Turin, Italy
| | - Franco Berrino
- Department of Preventive & Predictive Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | | | - Rebecca J Ford
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Massimo Levrero
- Epigénétique et Épigénomique des Carcinomes Hépathocellulaires Viro-Induits du Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Natalia Pediconi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Ludovica Ciuffreda
- Division of Medical Oncology A, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Michele Milella
- Division of Medical Oncology A, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mario Cioce
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
| | - Paola Muti
- Department of Oncology, Juravinski Cancer Center, McMaster University, Hamilton, Ontario, Canada
| | - Sabrina Strano
- Molecular Chemoprevention Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
- Department of Oncology, Juravinski Cancer Center, McMaster University, Hamilton, Ontario, Canada
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute ‘Regina Elena’, Rome, Italy
- Department of Oncology, Juravinski Cancer Center, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
38
|
Liu J, Chen D, Liu X, Liu Z. Cyclosporine A attenuates cardiac dysfunction induced by sepsis via inhibiting calcineurin and activating AMPK signaling. Mol Med Rep 2017; 15:3739-3746. [PMID: 28393192 DOI: 10.3892/mmr.2017.6421] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 01/26/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate whether cyclosporine A (CSA) improved cardiac dysfunction at an early stage of sepsis. Male Wistar rats were randomly divided into the following three groups: the sham‑operated control group, the cecal ligation puncture (CLP) procedure‑induced sepsis group and the CSA intervention group. Cecal ligation was performed to generate a sepsis model. At different time points (2, 6, 12, 24 and 72 h) following sepsis induction, blood pressure, cardiac function, and non‑esterified free fatty acid (NEFA) levels in the plasma and myocardia were measured, and the expression levels of components associated with the AMP‑activated protein kinase (AMPK)‑acetyl CoA carboxylase (ACC)‑carnitine palmitoyl transferase 1 (CPT1) signaling pathway were compared among the three groups. Sepsis induced a decrease in blood pressure and cardiac function at 24 h following sepsis induction in the CLP group, and CSA treatment ameliorated these pathophysiological alterations. In addition, rats in the CLP group exhibited significant increases in calcineurin activity and NEFA accumulation in the heart when compared with those in the sham group. These effects were attenuated by CSA treatment. Mechanistically, the activity of the AMPK‑ACC‑CPT1 pathway was enhanced by CSA treatment. The present study revealed that CSA treatment increases cardiac function at an early stage of sepsis in rats. This treatment partially suppresses calcineurin activity while activating the AMPK‑TCC‑CPT1 pathway.
Collapse
Affiliation(s)
- Jingmiao Liu
- Department of Emergency Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Da Chen
- Department of Emergency Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaowei Liu
- Department of Emergency Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhi Liu
- Department of Emergency Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
39
|
Cheng CY, Gutierrez NM, Marzuki MB, Lu X, Foreman TW, Paleja B, Lee B, Balachander A, Chen J, Tsenova L, Kurepina N, Teng KWW, West K, Mehra S, Zolezzi F, Poidinger M, Kreiswirth B, Kaushal D, Kornfeld H, Newell EW, Singhal A. Host sirtuin 1 regulates mycobacterial immunopathogenesis and represents a therapeutic target against tuberculosis. Sci Immunol 2017; 2. [PMID: 28707004 DOI: 10.1126/sciimmunol.aaj1789] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mycobacterium tuberculosis (Mtb) executes a plethora of immune-evasive mechanisms, which contribute to its pathogenesis, limited efficacy of current therapy, and the emergence of drug-resistant strains. This has led to resurgence in attempts to develop new therapeutic strategies/targets against tuberculosis (TB). We show that Mtb down-regulates sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, in monocytes/macrophages, TB animal models, and TB patients with active disease. Activation of SIRT1 reduced intracellular growth of drug-susceptible and drug-resistant strains of Mtb and induced phagosome-lysosome fusion and autophagy in a SIRT1-dependent manner. SIRT1 activation dampened Mtb-mediated persistent inflammatory responses via deacetylation of RelA/p65, leading to impaired binding of RelA/p65 on the promoter of inflammatory genes. In Mtb-infected mice, the use of SIRT1 activators ameliorated lung pathology, reduced chronic inflammation, and enhanced efficacy of anti-TB drug. Mass cytometry-based high-dimensional analysis revealed that SIRT1 activation mediated modulation of lung myeloid cells in Mtb-infected mice. Myeloid cell-specific SIRT1 knockout mice display increased inflammatory responses and susceptibility to Mtb infection. Collectively, these results provide a link between SIRT1 activation and TB pathogenesis and indicate a potential of SIRT1 activators in designing an effective and clinically relevant host-directed therapies for TB.
Collapse
Affiliation(s)
- Catherine Y Cheng
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Nuria M Gutierrez
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Mardiana B Marzuki
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Xiaohua Lu
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Taylor W Foreman
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Bhairav Paleja
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Bernett Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Akhila Balachander
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Liana Tsenova
- Public Health Research Institute of Rutgers Biomedical and Health Sciences, Rutgers University, Newark, NJ 07103, USA.,New York City College of Technology, Brooklyn, NY 11201, USA
| | - Natalia Kurepina
- Public Health Research Institute of Rutgers Biomedical and Health Sciences, Rutgers University, Newark, NJ 07103, USA
| | - Karen W W Teng
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Kim West
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Smriti Mehra
- Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Francesca Zolezzi
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Michael Poidinger
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Barry Kreiswirth
- Public Health Research Institute of Rutgers Biomedical and Health Sciences, Rutgers University, Newark, NJ 07103, USA
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Hardy Kornfeld
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Evan W Newell
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore
| | - Amit Singhal
- Singapore Immunology Network, Agency for Science, Technology and Research (A STAR), Singapore 138648, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
40
|
Xiao C, Wu Q, Zhang J, Xie Y, Cai W, Tan J. Antidiabetic activity of Ganoderma lucidum polysaccharides F31 down-regulated hepatic glucose regulatory enzymes in diabetic mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 196:47-57. [PMID: 27902927 DOI: 10.1016/j.jep.2016.11.044] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 11/23/2016] [Accepted: 11/26/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ganoderma lucidum (Lin Zhi) has been used to treat diabetes in Chinese folk for centuries. Our laboratory previously demonstrated that Ganoderma lucidum polysaccharides (GLPs) had hypoglycemic effects in diabetic mice. Our aim was to identify the main bioactives in GLPs and corresponding mechanism of action. MATERIALS AND METHODS Four polysaccharide-enriched fraction were isolated from GLPs and the antidiabetic activities were evaluated by type 2 diabetic mice. Fasting serum glucose (FSG), fasting serum insulin (FSI) and epididymal fat/BW ratio were measured at the end of the experiment. In liver, the mRNA levels of hepatic glucose regulatory enzymes were determined by quantitative polymerase chain reaction (qPCR) and the protein levels of phospho-AMP-activated protein kinase (p-AMPK)/AMPK were determined by western blotting test. In epididymal fat tissue, the mRNA and protein levels GLUT4, resistin, fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC1) were determined by qPCR and immuno-histochemistry. The structure of polysaccharide F31 was obtained from GPC, FTIR NMR and GC-MS spectroscopy, RESULTS: F31 significantly decreased FSG (P<0.05), FSI and epididymal fat/BW ratio (P<0.01). In liver, F31 decreased the mRNA levels of hepatic glucose regulatory enzymes, and up-regulated the ratio of phospho-AMP-activated protein kinase (p-AMPK)/AMPK. In epididymal fat tissue, F31 increased the mRNA levels of GLUT4 but decreased fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC1) and resistin. Immuno-histochemistry results revealed F31 increased the protein levels of GLUT4 and decreased resistin. CONCLUSION Data suggested that the main bioactives in GLPs was F31, which was determined to be a β-heteropolysaccharide with the weight-average molecular weight of 15.9kDa. The possible action mechanism of F31 may be associated with down-regulation of the hepatic glucose regulated enzyme mRNA levels via AMPK activation, improvement of insulin resistance and decrease of epididymal fat/BW ratio. These results strongly suggest that F31 has antidiabetic potential.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Acetyl-CoA Carboxylase/genetics
- Acetyl-CoA Carboxylase/metabolism
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Animals
- Blood Glucose/analysis
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Down-Regulation
- Fasting/blood
- Fatty Acid Synthase, Type I/genetics
- Fatty Acid Synthase, Type I/metabolism
- Fruiting Bodies, Fungal
- Fungal Polysaccharides/pharmacology
- Fungal Polysaccharides/therapeutic use
- Ganoderma
- Glucose Transporter Type 4/genetics
- Glucose Transporter Type 4/metabolism
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/blood
- Liver/drug effects
- Liver/metabolism
- Male
- Mice, Inbred C57BL
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Chun Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Qingping Wu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Jumei Zhang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Yizhen Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Wen Cai
- Department of Toxicology, Center for Disease Control and Prevention of Guangdong Province, Guangzhou 510020, China.
| | - Jianbin Tan
- Department of Toxicology, Center for Disease Control and Prevention of Guangdong Province, Guangzhou 510020, China.
| |
Collapse
|
41
|
Metformin suppresses hypoxia-induced stabilization of HIF-1α through reprogramming of oxygen metabolism in hepatocellular carcinoma. Oncotarget 2016; 7:873-84. [PMID: 26621849 PMCID: PMC4808039 DOI: 10.18632/oncotarget.6418] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 11/16/2015] [Indexed: 12/16/2022] Open
Abstract
Overexpression of hypoxia-induced factor 1α (HIF-1α) has been shown to be involved in the development and progression of hepatocellular carcinoma (HCC). HIF-1α should therefore be a promising molecular target for the development of anti-HCC agents. Metformin, an established antidiabetic drug, has proved to also be effective in treating cancer although the precise underlying mechanisms of this activity are not fully elucidated. The aim of this study was to investigate the effects of metformin on the expression of HIF-1α and oxygen metabolism in HCC. The results showed that metformin inhibited hypoxia-induced HIF-1α accumulation and activation independent of AMP-activated protein kinase (AMPK). Moreover, this decrease in HIF-1α accumulation was accompanied by promotion of HIF-1α protein degradation. In addition, metformin significantly decreased oxygen consumption, ultimately leading to increased intracellular oxygen tension and decreased staining with the hypoxia marker pimonidazole. In vivo studies demonstrated that metformin delayed tumor growth and attenuated the expression of HIF-1α in HCC tumor xenografts. Together, these findings suggest that metformin decreases hypoxia-induced HIF-1α accumulation by actively suppressing mitochondrial oxygen consumption and enhancing cellular oxygenation ability, providing a fundamental mechanism of metformin activity against HCC.
Collapse
|
42
|
Gutiérrez-Salmeán G, Meaney E, Lanaspa MA, Cicerchi C, Johnson RJ, Dugar S, Taub P, Ramírez-Sánchez I, Villarreal F, Schreiner G, Ceballos G. A randomized, placebo-controlled, double-blind study on the effects of (-)-epicatechin on the triglyceride/HDLc ratio and cardiometabolic profile of subjects with hypertriglyceridemia: Unique in vitro effects. Int J Cardiol 2016; 223:500-506. [PMID: 27552564 DOI: 10.1016/j.ijcard.2016.08.158] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/07/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cardiometabolic disruptions such as insulin resistance, obesity, high blood pressure, hyperglycemia, and dyslipidemias, are known to increase the risk for cardiovascular and metabolic diseases such as type 2 diabetes mellitus and atherosclerosis. Several screening tools for assessing cardiometabolic risk have been developed including the TG/HDLc ratio, which has been, demonstrated to possess a strong association with insulin resistance and coronary disease. Dietary modifications, together with regular moderate exercise have proven to be effective in attenuating cardiometabolic disruptions. However, they often exhibit poor long-term patient compliance. Nutraceutics, including (-)-epicatechin (EPI), have gained increasing interest as coadjuvant effective and safe therapies that are able to attenuate hypertension, hyperglycemia, hyperinsulinemia, hypertriglyceridemia and hypoalphalipoproteinemia. METHODS The aims of this study were: 1) to compare the in vitro effect of EPI vs. (+)-catechin on fructose induced triglyceride accumulation and mitochondrial function in Hep2 cells in culture, 2) to evaluate the efficacy of EPI treatment in reducing fasting blood triglycerides and improving the TG/HDLc ratio in hypertriglyceridemic patients with a total daily dose of 100mg of EPI. Secondary clinical variables included total cholesterol, LDLc, fructosamine, glucose, insulin, and high sensitivity C-reactive protein blood levels. RESULTS AND CONCLUSION Our results provide preliminary evidence as to favorable effects of EPI on glycemia homeostasis, lipid profile and systemic inflammation such bioactive actions are not class-effects (i.e. limited to their antioxidant potential) but instead, may result from the specific activation of associated downstream signaling pathways since catechin has no effects.
Collapse
Affiliation(s)
- Gabriela Gutiérrez-Salmeán
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Eduardo Meaney
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | | | | | | | | | - Pam Taub
- Department of Medicine, University of California San Diego, United States
| | - Israel Ramírez-Sánchez
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico; Department of Medicine, University of California San Diego, United States
| | | | | | - Guillermo Ceballos
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico.
| |
Collapse
|
43
|
Jung DW, Lee OH, Kang IJ. Sanguisorba officinalis L. Extracts Exert Antiobesity Effects in 3T3-L1 Adipocytes and C57BL/6J Mice Fed High-Fat Diets. J Med Food 2016; 19:768-79. [DOI: 10.1089/jmf.2016.3704] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Da-Woon Jung
- Department of Food Science and Nutrition, Hallym University, Gangwon, Korea
| | - Ok-Hwan Lee
- Department of Food Science and Biotechnology, Kangwon National University, Gangwon, Korea
| | - Il-Jun Kang
- Department of Food Science and Nutrition, Hallym University, Gangwon, Korea
| |
Collapse
|
44
|
Abstract
Activation of the adenosine monophosphate (AMP)-activated kinase (AMPK) contributes to beneficial effects such as improvement of the hyperglycemic state in diabetes as well as reduction of obesity and inflammatory processes. Furthermore, stimulation of AMPK activity has been associated with increased exercise capacity. A study published in 2008, directly before the Olympic Games in Beijing, showed that the AMPK activator AICAR (5-amino-1-β-D-ribofuranosyl-imidazole-4-carboxamide) increased the running capacity of mice without any training and thus, prompted the World Anti-Doping Agency (WADA) to include certain AMPK activators in the list of forbidden drugs. This raises the question as to whether all AMPK activators should be considered for registration or whether the increase in exercise performance is only associated with specific AMPK-activating substances. In this review, we intend to shed light on currently published AMPK-activating drugs, their working mechanisms, and their impact on body fitness.
Collapse
|
45
|
Bullón P, Alcocer-Gómez E, Carrión AM, Marín-Aguilar F, Garrido-Maraver J, Román-Malo L, Ruiz-Cabello J, Culic O, Ryffel B, Apetoh L, Ghiringhelli F, Battino M, Sánchez-Alcazar JA, Cordero MD. AMPK Phosphorylation Modulates Pain by Activation of NLRP3 Inflammasome. Antioxid Redox Signal 2016; 24:157-70. [PMID: 26132721 PMCID: PMC4742979 DOI: 10.1089/ars.2014.6120] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
AIMS Impairment in adenosine monophosphate-activated protein kinase (AMPK) activity and NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation are associated with several metabolic and inflammatory diseases. In this study, we investigated the role of AMPK/NLRP3 inflammasome axis in the molecular mechanism underlying pain perception. RESULTS Impairment in AMPK activation induced by compound C or sunitinib, two AMPK inhibitors, provoked hyperalgesia in mice (p<0.001) associated with marked NLRP3 inflammasome protein activation and increased serum levels of interleukin-1β (IL-1β) (24.56±0.82 pg/ml) and IL-18 (23.83±1.882 pg/ml) compared with vehicle groups (IL-1β: 8.15±0.44; IL-18: 4.92±0.4). This effect was rescued by increasing AMPK phosphorylation via metformin treatment (p<0.001), caloric restriction diet (p<0.001), or NLRP3 inflammasome genetic inactivation using NLRP3 knockout (nlrp3(-/-)) mice (p<0.001). Deficient AMPK activation and overactivation of NLRP3 inflammasome axis were also observed in blood cells from patients with fibromyalgia (FM), a prevalent human chronic pain disease. In addition, metformin treatment (200 mg/daily), which increased AMPK activation, restored all biochemical alterations examined by us in blood cells and significantly improved clinical symptoms, such as, pain, fatigue, depression, disturbed sleep, and tender points, in patients with FM. INNOVATION AND CONCLUSIONS These data suggest that AMPK/NLRP3 inflammasome axis participates in chronic pain and that NLRP3 inflammasome inhibition by AMPK modulation may be a novel therapeutic target to fight against chronic pain and inflammatory diseases as FM.
Collapse
Affiliation(s)
- Pedro Bullón
- Institute of Biomedicine of Seville (IBIS)/Research Laboratory, Oral Medicine Department, Universidad de Sevilla, Sevilla, Spain
| | - Elísabet Alcocer-Gómez
- Institute of Biomedicine of Seville (IBIS)/Research Laboratory, Oral Medicine Department, Universidad de Sevilla, Sevilla, Spain
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-CSIC-Junta de Andalucía and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Sevilla, Spain
| | - Angel M. Carrión
- División de Neurociencias, Universidad Pablo de Olavide de Sevilla, Sevilla, Spain
| | - Fabiola Marín-Aguilar
- Institute of Biomedicine of Seville (IBIS)/Research Laboratory, Oral Medicine Department, Universidad de Sevilla, Sevilla, Spain
| | - Juan Garrido-Maraver
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-CSIC-Junta de Andalucía and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Sevilla, Spain
| | - Lourdes Román-Malo
- Institute of Biomedicine of Seville (IBIS)/Research Laboratory, Oral Medicine Department, Universidad de Sevilla, Sevilla, Spain
| | - Jesus Ruiz-Cabello
- CIBER de Enfermedades Respiratorias, Madrid, Spain; Advanced Imaging Unit, Centro Nacional de Investigaciones Cardiovasculares, and Universidad Complutense Madrid, Madrid, Spain
| | - Ognjen Culic
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | | | - Lionel Apetoh
- INSERM, UMR866, Dijon, France
- Centre Georges François Leclerc, Dijon, France
- Université de Bourgogne, Dijon, France
| | - François Ghiringhelli
- INSERM, UMR866, Dijon, France
- Centre Georges François Leclerc, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche–Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy
| | - José Antonio Sánchez-Alcazar
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-CSIC-Junta de Andalucía and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Sevilla, Spain
| | - Mario D. Cordero
- Institute of Biomedicine of Seville (IBIS)/Research Laboratory, Oral Medicine Department, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
46
|
AMPK Regulation of Cell Growth, Apoptosis, Autophagy, and Bioenergetics. EXPERIENTIA SUPPLEMENTUM (2012) 2016; 107:45-71. [PMID: 27812976 DOI: 10.1007/978-3-319-43589-3_3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In eukaryotic cells, AMP-activated protein kinase (AMPK) generally promotes catabolic pathways that produce ATP and at the same time inhibits anabolic pathways involved in different processes that consume ATP. As an energy sensor, AMPK is involved in the main cellular functions implicated in cell fate, such as cell growth and autophagy.Recently, AMPK has been connected with apoptosis regulation, although the molecular mechanism by which AMPK induces and/or inhibits cell death is not clear.This chapter reviews the essential role of AMPK in signaling pathways that respond to cellular stress and damage, highlighting the complex and reciprocal regulation between AMPK and their targets and effectors. The therapeutic implications of the role of AMPK in different pathologies such as diabetes, cancer, or mitochondrial dysfunctions are still controversial, and it is necessary to further investigate the molecular mechanisms underlying AMPK activation.
Collapse
|
47
|
Kim HL, Park J, Park H, Jung Y, Youn DH, Kang J, Jeong MY, Um JY. Platycodon grandiflorum A. De Candolle Ethanolic Extract Inhibits Adipogenic Regulators in 3T3-L1 Cells and Induces Mitochondrial Biogenesis in Primary Brown Preadipocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7721-7730. [PMID: 26244589 DOI: 10.1021/acs.jafc.5b01908] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This study was designed to evaluate the effects of Platycodon grandiflorum A. DC. ethanolic extract (PG) on obesity in brown/white preadipocytes. The effect of PG on the differentiation and mitochondrial biogenesis of brown adipocytes is still not examined. An in vivo study showed that PG induced weight loss in mice with high-fat-diet-induced obesity. PG successfully suppressed the differentiation of 3T3-L1 cells by down-regulating cellular induction of the peroxisome proliferators activated receptor γ (PPARγ), CCAAT enhancer binding protein α (C/EBPα), lipin-1, and adiponectin but increasing expression of silent mating type information regulation 2 homologue 1 (SIRT1) and the phosphorylation of AMP-activated protein kinase α (AMPKα). The effect of PG on the adipogenic factors was compared with that of its bioactive compound platycodin D. In addition, PG increased expressions of mitochondria-related genes, including uncoupling protein 1 (UCP1), peroxisome proliferator activated receptor-coactivator 1 α (PGC1α), PR domain containing 16 (PRDM16), SIRT3, nuclear respiratory factor (NRF), and cytochrome C (CytC) in primary brown adipocytes. These results indicate that PG stimulates the differentiation of brown adipocytes through modulation of mitochondria-related genes and could offer clinical benefits as a supplement to treat obesity.
Collapse
Affiliation(s)
- Hye-Lin Kim
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Hyewon Park
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Yunu Jung
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Dong-Hyun Youn
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - JongWook Kang
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Mi-Young Jeong
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Jae-Young Um
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| |
Collapse
|
48
|
Smith KJ, Germain M. Polycystic ovary syndrome (PCOS) with melanocytic mucosal macules: the role of STK11 gene polymorphisms in PCOS and Peutz-Jeghers syndrome. Int J Dermatol 2015; 55:177-80. [PMID: 26147831 DOI: 10.1111/ijd.12787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 06/22/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex genetic disorder that is the most common endocrinopathy that affects women. OBSERVATIONS We report two individuals with PCOS with a genetic polymorphism in serine threonine kinase 11 (STK11). Both these individuals developed mucosal pigmentation suggesting Peutz-Jeghers syndrome (PJS), which is associated with mutations in STK11. Both individuals showed some improvement in their metabolic and endocrine dysregulation with therapies commonly used for PCOS. However, they continued to show progression of mucosa pigmentation. CONCLUSIONS This is the first report of clinical overlap in individuals with PCOS and PJS, even though some individuals with PCOS show a polymorphism in STK11, which is the gene mutated in PJS. The importance of this clinical association is not clear but may be significant because of the association of STK11 dysregulation and the development of internal tumors.
Collapse
Affiliation(s)
- Kathleen J Smith
- DermPath and Dermatology Consultants, Atlanta, GA, USA.,Charleston, Mt. Pleasant, SC, USA
| | | |
Collapse
|
49
|
Glade MJ, Smith K. A glance at … exercise and glucose uptake. Nutrition 2015; 31:893-7. [PMID: 25933500 DOI: 10.1016/j.nut.2014.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Affiliation(s)
| | - Kyl Smith
- Progressive Laboratories Inc., Irving, Texas
| |
Collapse
|
50
|
García-Prieto CF, Gil-Ortega M, Aránguez I, Ortiz-Besoain M, Somoza B, Fernández-Alfonso MS. Vascular AMPK as an attractive target in the treatment of vascular complications of obesity. Vascul Pharmacol 2015; 67-69:10-20. [PMID: 25869500 DOI: 10.1016/j.vph.2015.02.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/23/2014] [Accepted: 02/02/2015] [Indexed: 02/06/2023]
Abstract
The key for the survival of all organisms is the regulation and control of energy metabolism. Thus, several strategies have evolved in each tissue in order to balance nutrient supply with energy demand. Adenosine monophosphate-activated protein kinase (AMPK) is now recognized as a key participant in energy metabolism. It ensures an appropriate energetic supply by promoting energy conserving pathways in detriment of anabolic processes not essential for cell survival. Vascular AMPK plays a critical role in the regulation of blood flow and vascular tone through several mechanisms, including vasodilation by stimulating nitric oxide release in endothelial cells. Since obesity leads to endothelial damage and AMPK dysregulation, AMPK activation might be an important strategy to restore vascular function in cardiometabolic alterations. In the present review we focus on the role of vascular AMPK in both endothelial and smooth muscle cells, paying special attention to its dysregulation in obesity- and high-fat diet-related complications, as well as to the mechanisms and benefits of vascular AMPK activation.
Collapse
Affiliation(s)
- C F García-Prieto
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28668 Madrid, Spain
| | - M Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28668 Madrid, Spain
| | - I Aránguez
- Instituto Pluridisciplinar and Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; Departamento de Bioquímica, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - M Ortiz-Besoain
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias, Edificio N3, Universidad Católica del Norte de Chile, Angamos, 0610 Antofagasta, Chile
| | - B Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28668 Madrid, Spain
| | - M S Fernández-Alfonso
- Instituto Pluridisciplinar and Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain.
| |
Collapse
|