1
|
Mason T, Alesi S, Fernando M, Vanky E, Teede HJ, Mousa A. Metformin in gestational diabetes: physiological actions and clinical applications. Nat Rev Endocrinol 2025; 21:77-91. [PMID: 39455749 DOI: 10.1038/s41574-024-01049-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2024] [Indexed: 10/28/2024]
Abstract
Metformin is an effective oral hypoglycaemic agent used in the treatment of type 2 diabetes mellitus; however, its use in pregnancy for the treatment of gestational diabetes mellitus (GDM) remains controversial owing to concerns around safety and efficacy. This comprehensive review outlines the physiological metabolic functions of metformin and synthesizes existing literature and key knowledge gaps pertaining to the use of metformin in pregnancy across various end points in women with GDM. On the basis of current evidence, metformin reduces gestational weight gain, neonatal hypoglycaemia and macrosomia and increases insulin sensitivity. However, considerable heterogeneity between existing studies and the grouping of aggregate and often inharmonious data within meta-analyses has led to disparate findings regarding the efficacy of metformin in treating hyperglycaemia in GDM. Innovative analytical approaches with stratification by individual-level characteristics (for example, obesity, ethnicity, GDM severity and so on) and treatment regimens (diagnostic criteria, treatment timing and follow-up duration) are needed to establish efficacy across a range of end points and to identify which, if any, subgroups might benefit from metformin treatment during pregnancy.
Collapse
Affiliation(s)
- Taitum Mason
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
| | - Simon Alesi
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
| | - Melinda Fernando
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Helena J Teede
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia
- Department of Endocrinology and Diabetes, Monash Health, Clayton, Victoria, Melbourne, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Melbourne, Australia.
| |
Collapse
|
2
|
Yoshimura Y, Matsui T, Kaneko N, Kobayashi I. Digestion and absorption of triacetin, a short-chain triacylglycerol. Lipids 2025. [PMID: 39891375 DOI: 10.1002/lipd.12433] [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: 08/17/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Triacylglycerol (TG) is categorized into long-, medium-, and short-chain TG (SCTG). While the digestion of long- and medium-chain TG is well established, the process for SCTG remains unclear. This study investigated SCTG digestion by administering 2 mmol of triacetin to rats and analyzing acetin, acetic acid, and glycerol levels in the portal blood and small intestine. Triacetin was fully degraded in the upper gastrointestinal tract and absorbed as acetic acid and glycerol. Glycerol influx into the liver promoted gluconeogenesis, while acetate activated AMPK, resulting in the suppression of fatty acid synthesis-related genes and the upregulation of fatty acid β-oxidation-related genes. These findings demonstrate that triacetin not only serves as a substrate for energy metabolism but also regulates hepatic gene expression, highlighting its dual role as both a metabolic substrate and signaling molecule. Triacetin thus shows potential as a dietary modulator for improving metabolic health.
Collapse
Affiliation(s)
| | - Tomoka Matsui
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| | - Nagisa Kaneko
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| | - Ikuha Kobayashi
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| |
Collapse
|
3
|
Yang W, Zeng S, Shao R, Jin R, Huang J, Wang X, Liu E, Zhou T, Li F, Chen Y, Chen D. Sulforaphane regulation autophagy-mediated pyroptosis in autoimmune hepatitis via AMPK/mTOR pathway. Int Immunopharmacol 2025; 146:113826. [PMID: 39673998 DOI: 10.1016/j.intimp.2024.113826] [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: 10/15/2024] [Revised: 11/28/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Autoimmune hepatitis (AIH) is a liver disease marked by inflammation of unknown origin. If untreated, it can progress to cirrhosis or liver failure, posing a significant health risk. Currently, effective drug therapies are lacking in clinical practice. Sulforaphane (SFN), a natural anti-inflammatory and antioxidant compound found in various cruciferous vegetables, alleviate pyroptosis and improve impaired autophagic flux, both of which contribute to AIH progression. However, whether SFN modulates autophagic flux and pyroptosis in S100-induced EAH through the AMPK/mTOR pathway remains unclear. Therefore, this study aims to investigate whether SFN can regulate AIH and elucidate its potential mechanisms of action. In this study, experimental AIH (EAH) was induced in male C57BL/6 J mice through intraperitoneal (i.p.) injection of S100. SFN was administered intraperitoneally every other day. After 28 days, the mice were euthanized, and their livers and serum were collected for histological and biochemical analyses. AML12 cells were used for the in vitro studies. The results showed that SFN mitigated pyroptosis by inhibiting the NLRP3 inflammasome and improving autophagic flux, which alleviates S100-induced EAH. Conversely, the autophagy inhibitor 3-MA negated the protective effects of SFN against inflammasome-mediated pyroptosis. Furthermore, SFN activated the AMPK/mTOR signaling pathway, offering protection against S100-induced EAH. Selective inhibition of AMPK suppressed the improvement in autophagic flux and protected against SFN-induced pyroptosis. Overall, SFN significantly ameliorates S100-induced EAH by enhancing autophagic flux and mitigating pyroptosis through activation of the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Weijian Yang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Shiyi Zeng
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Rongrong Shao
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Ru Jin
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Jiayin Huang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Xinyu Wang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Enqian Liu
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Tenghui Zhou
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Fengfan Li
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Yongping Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China.
| | - Dazhi Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China; School of Clinical Medicine, The First People's Hospital of Lin'an District, Hangzhou, Lin'an People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou 311300, China.
| |
Collapse
|
4
|
Yang B, Yang R, Zhang X, Wang W, Kan J. Hovenia dulcis (Guaizao) polysaccharide ameliorates hyperglycemia through multiple signaling pathways in rats with type 2 diabetes mellitus. Int J Biol Macromol 2024; 285:138338. [PMID: 39638196 DOI: 10.1016/j.ijbiomac.2024.138338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/07/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Type 2 diabetes mellitus (T2DM) poses a significant threat to human health, with its incidence and mortality rates increasing annually. This study investigated the hypoglycemic effects and underlying mechanisms of pure Hovenia dulcis (Guaizao) polysaccharide (HDPs-2A) in rats subjected to a high-fat and high-sugar diet combined with streptozotocin-induced T2DM. Oral administration of HDPs-2A resulted in significant increases in body weight and liver glycogen levels compared to untreated controls. Moreover, a reduction in fasting blood glucose levels, alleviation of hyperinsulinemia, enhanced glucose tolerance, and improved insulin resistance were observed in the HDPs-2A-treated group. HDPs-2A also effectively reversed diabetes-induced dyslipidemia, as evidenced by decreased total cholesterol and triglyceride levels, alongside increased high-density lipoprotein cholesterol levels. Histopathological analyses confirmed that HDPs-2A partially repaired liver tissue damage by mitigating oxidative stress responses in the liver. Additionally, treatment with HDPs-2A significantly elevated short-chain fatty acid levels in T2DM rats. Real-time quantitative PCR and Western blot analyses indicated that HDPs-2A significantly enhanced the expression of InsR, IRS2, PI3K, Akt, and GLUT4, suggesting that HDPs-2A regulates insulin resistance and glycometabolism through the activation of the PI3K/Akt signaling pathway. Furthermore, HDPs-2A appeared to modulate the expression of GS, GSK-3β, and FoxO1 to improve glucose metabolism and reduce insulin resistance. It also improved glucose metabolism by activating the AMPK pathway and modulating G6Pase and PEPCK expression. This study provides novel insights into the antidiabetic effects of HDPs, positioning them as promising nutritional agents for the management of T2DM.
Collapse
Affiliation(s)
- Bing Yang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China.
| | - Ruyan Yang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Xinyu Zhang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Wanjia Wang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Jianquan Kan
- College of Food Science, Southwest University, 2 Tiansheng Road, Beibei, Chongqing 400715, PR China.
| |
Collapse
|
5
|
Tran J, Parekh S, Rockcole J, Wilson D, Parmar MS. Repurposing antidiabetic drugs for Alzheimer's disease: A review of preclinical and clinical evidence and overcoming challenges. Life Sci 2024; 355:123001. [PMID: 39173996 DOI: 10.1016/j.lfs.2024.123001] [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: 07/05/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Repurposing antidiabetic drugs for the treatment of Alzheimer's disease (AD) has emerged as a promising therapeutic strategy. This review examines the potential of repurposing antidiabetic drugs for AD treatment, focusing on preclinical evidence, clinical trials, and observational studies. In addition, the review aims to explore challenges and opportunities in repurposing antidiabetic drugs for AD, emphasizing the importance of well-designed clinical trials that consider patient selection criteria, refined outcome measures, adverse effects, and combination therapies to enhance therapeutic efficacy. Preclinical evidence suggests that glucagon-like peptide-1 (GLP-1) analogs, dipeptidyl peptidase-4 (DPP4) inhibitors, metformin, thiazolidinediones, and sodium-glucose co-transporter-2 (SGLT2) inhibitors exhibit neuroprotective effects in AD preclinical models. In preclinical studies, antidiabetic drugs have demonstrated neuroprotective effects by reducing amyloid beta (Aβ) plaques, tau hyperphosphorylation, neuroinflammation, and cognitive impairment. Antidiabetic drug classes, notably GLP-1 analogs and SGLT2 inhibitors, and a reduced risk of dementia in patients with diabetes mellitus. While the evidence for DPP4 inhibitors is mixed, some studies suggest a potential protective effect. On the other hand, alpha-glucosidase inhibitors (AGIs) and sulfonylureas may potentially increase the risk, especially in those experiencing recurrent hypoglycemic events. Repurposing antidiabetic drugs for AD is a promising therapeutic strategy, but challenges such as disease heterogeneity, limited biomarkers, and benefits versus risk evaluation need to be addressed. Ongoing clinical trials in mild cognitive impairment (MCI) and early AD patients without diabetes will be crucial in determining the clinical efficacy and safety of the antidiabetic drugs, paving the way for potential treatments for AD.
Collapse
Affiliation(s)
- Jacky Tran
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Sneh Parekh
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Julia Rockcole
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Danielle Wilson
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Mayur S Parmar
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
| |
Collapse
|
6
|
Wei F, Zhang W, Kang S, Liu P, Yao Y, Liu W, Aikemu A, Pang K, Yang X. Phenolic Constituents with Glucose Uptake and GLUT4 Translocation Bioactivities from the Fruits of Cordia dichotoma. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16298-16311. [PMID: 38982710 DOI: 10.1021/acs.jafc.4c02672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
From the fruits of Cordia dichotoma, 11 new phenolic compounds, dichotomins A-K, were isolated, together with 19 known compounds. Through the analysis of detailed NMR data and HRESIMS data, the planar structures of all compounds were confirmed. Using NMR calculations, the absolute configuration of dichotomins A-K was elucidated by comparing their observed and computed electronic circular dichroism (ECD) spectra. Dichotomin H (8) and dichotomin I (9) were determined as two pairs of enantiomers. The enantiomers of compounds 8 and 9 were separated using chiral-phase high-performance liquid chromatography (HPLC), and the stereostructure of each enantiomer was determined by similarly calculating the ECD. Compounds 3, 5, 7, 17, 18, 23-25, and 27-30 increased glucose uptake by 1.04- to 2.85-folds at concentrations of 30 μg/mL. Further studies revealed that compounds 3 and 5 had a moderate effect on glucose transporter 4 (GLUT4) translocation activity in L6 cells. At 30 μg/mL, compound 3 significantly enhanced AMPK phosphorylation and GLUT4 expression. As a whole, compound 3 has the potential to be a drug candidate for the treatment of type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Feng Wei
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Wenting Zhang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiwen Kang
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Pengxin Liu
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Yudi Yao
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Wenqi Liu
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Ainiwaer Aikemu
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan 848000, China
| | - Kejian Pang
- College of Biological Sciences and Technology, Yili Normal University, Yili 835000, China
| | - Xinzhou Yang
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| |
Collapse
|
7
|
Kakoti BB, Alom S, Deka K, Halder RK. AMPK pathway: an emerging target to control diabetes mellitus and its related complications. J Diabetes Metab Disord 2024; 23:441-459. [PMID: 38932895 PMCID: PMC11196491 DOI: 10.1007/s40200-024-01420-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/07/2024] [Indexed: 06/28/2024]
Abstract
Purpose In this extensive review work, the important role of AMP-activated protein kinase (AMPK) in causing of diabetes mellitus has been highlighted. Structural feature of AMPK as well its regulations and roles are described nicely, and the association of AMPK with the diabetic complications like nephropathy, neuropathy and retinopathy are also explained along with the connection between AMPK and β-cell function, insulin resistivity, mTOR, protein metabolism, autophagy and mitophagy and effect on protein and lipid metabolism. Methods Published journals were searched on the database like PubMed, Medline, Scopus and Web of Science by using keywords such as AMPK, diabetes mellitus, regulation of AMPK, complications of diabetes mellitus, autophagy, apoptosis etc. Result After extensive review, it has been found that, kinase enzyme like AMPK is having vital role in management of type II diabetes mellitus. AMPK involve in enhance the concentration of glucose transporter like GLUT 1 and GLUT 4 which result in lowering of blood glucose level in influx of blood glucose into the cells; AMPK increases the insulin sensitivity and decreases the insulin resistance and further AMPK decreases the apoptosis of β-cells which result into secretion of insulin and AMPK is also involve in declining of oxidative stress, lipotoxicity and inflammation, owing to which organ damage due to diabetes mellitus can be lowered by activation of AMPK. Conclusion As AMPK activation leads to overall control of diabetes mellitus, designing and developing of small molecules or peptide that can act as AMPK agonist will be highly beneficial for control or manage diabetes mellitus.
Collapse
Affiliation(s)
- Bibhuti B. Kakoti
- Department of Pharmaceutical Sciences, Dibrugarh University, 786004 Dibrugarh, Assam India
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Dibrugarh University, 786004 Dibrugarh, Assam India
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Sciences, Girijananda Chowdhury University- Tezpur campus, 784501 Sonitpur, Assam India
| | - Kangkan Deka
- Department of Pharmaceutical Sciences, Dibrugarh University, 786004 Dibrugarh, Assam India
- Department of Pharmacognosy, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, 781125 Mirza, Kamrup, Assam India
| | - Raj Kumar Halder
- Ruhvenile Biomedical, Plot -8 OCF Pocket Institution, Sarita Vihar, 110076 Delhi, India
| |
Collapse
|
8
|
Min SH, Song DK, Lee CH, Roh E, Kim MS. Hypothalamic AMP-Activated Protein Kinase as a Whole-Body Energy Sensor and Regulator. Endocrinol Metab (Seoul) 2024; 39:1-11. [PMID: 38356211 PMCID: PMC10901667 DOI: 10.3803/enm.2024.1922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
5´-Adenosine monophosphate (AMP)-activated protein kinase (AMPK), a cellular energy sensor, is an essential enzyme that helps cells maintain stable energy levels during metabolic stress. The hypothalamus is pivotal in regulating energy balance within the body. Certain neurons in the hypothalamus are sensitive to fluctuations in food availability and energy stores, triggering adaptive responses to preserve systemic energy equilibrium. AMPK, expressed in these hypothalamic neurons, is instrumental in these regulatory processes. Hypothalamic AMPK activity is modulated by key metabolic hormones. Anorexigenic hormones, including leptin, insulin, and glucagon-like peptide 1, suppress hypothalamic AMPK activity, whereas the hunger hormone ghrelin activates it. These hormonal influences on hypothalamic AMPK activity are central to their roles in controlling food consumption and energy expenditure. Additionally, hypothalamic AMPK activity responds to variations in glucose concentrations. It becomes active during hypoglycemia but is deactivated when glucose is introduced directly into the hypothalamus. These shifts in AMPK activity within hypothalamic neurons are critical for maintaining glucose balance. Considering the vital function of hypothalamic AMPK in the regulation of overall energy and glucose balance, developing chemical agents that target the hypothalamus to modulate AMPK activity presents a promising therapeutic approach for metabolic conditions such as obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Se Hee Min
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Do Kyeong Song
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Chan Hee Lee
- Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Korea
| | - Eun Roh
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Tyagi S, Mani S. Combined Administration of Metformin and Vitamin D: A Futuristic Approach for Management of Hyperglycemia. Cardiovasc Hematol Agents Med Chem 2024; 22:258-275. [PMID: 37929731 DOI: 10.2174/0118715257261643231018102928] [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: 06/09/2023] [Revised: 08/28/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
Diabetes is a series of metabolic disorders that can be categorized into three types depending on different aspects associated with age at onset, intensity of insulin resistance, and beta- cell dysfunction: Type 1 and 2 Diabetes, and Gestational Diabetes Mellitus. Type 2 Diabetes Mellitus (T2DM) has recently been found to account for more than 85% of diabetic cases. The current review intends to raise awareness among clinicians/researchers that combining vitamin D3 with metformin may pave the way for better T2DM treatment and management. An extensive literature survey was performed to analyze vitamin D's role in regulating insulin secretion, their action on the target cells and thus maintaining the normal glucose level. On the other side, the anti-hyperglycemic effect of metformin as well as its detailed mechanism of action was also studied. Interestingly both compounds are known to exhibit the antioxidant effect too. Literature supporting the correlation between diabetic phenotypes and deficiency of vitamin D was also explored further. To thoroughly understand the common/overlapping pathways responsible for the antidiabetic as well as antioxidant nature of metformin and vitamin D3, we compared their antihyperglycemic and antioxidant activities. With this background, we are proposing the hypothesis that it would be of great interest if these two compounds could work in synergy to better manage the condition of T2DM and associated disorders.
Collapse
Affiliation(s)
- Sakshi Tyagi
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
10
|
Chan MP, Takenaka N, Satoh T. Impaired Insulin Signaling Mediated by the Small GTPase Rac1 in Skeletal Muscle of the Leptin-Deficient Obese Mouse. Int J Mol Sci 2023; 24:11531. [PMID: 37511290 PMCID: PMC10380855 DOI: 10.3390/ijms241411531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Insulin-stimulated glucose uptake in skeletal muscle is mediated by the glucose transporter GLUT4. The small GTPase Rac1 acts as a switch of signal transduction that regulates GLUT4 translocation to the plasma membrane following insulin stimulation. However, it remains obscure whether signaling cascades upstream and downstream of Rac1 in skeletal muscle are impaired by obesity that causes insulin resistance and type 2 diabetes. In an attempt to clarify this point, we investigated Rac1 signaling in the leptin-deficient (Lepob/ob) mouse model. Here, we show that insulin-stimulated GLUT4 translocation and Rac1 activation are almost completely abolished in Lepob/ob mouse skeletal muscle. Phosphorylation of the protein kinase Akt2 and plasma membrane translocation of the guanine nucleotide exchange factor FLJ00068 following insulin stimulation were also diminished in Lepob/ob mice. On the other hand, the activation of another small GTPase RalA, which acts downstream of Rac1, by the constitutively activated form of Akt2, FLJ00068, or Rac1, was partially abrogated in Lepob/ob mice. Taken together, we conclude that insulin-stimulated glucose uptake is impaired by two mechanisms in Lepob/ob mouse skeletal muscle: one is the complete inhibition of Akt2-mediated activation of Rac1, and the other is the partial inhibition of RalA activation downstream of Rac1.
Collapse
Affiliation(s)
| | | | - Takaya Satoh
- Laboratory of Cell Biology, Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Sakai 599-8531, Japan; (M.P.C.); (N.T.)
| |
Collapse
|
11
|
Gorący A, Rosik J, Szostak J, Szostak B, Retfiński S, Machaj F, Pawlik A. Improving mitochondrial function in preclinical models of heart failure: therapeutic targets for future clinical therapies? Expert Opin Ther Targets 2023; 27:593-608. [PMID: 37477241 DOI: 10.1080/14728222.2023.2240021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/19/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION Heart failure is a complex clinical syndrome resulting from the unsuccessful compensation of symptoms of myocardial damage. Mitochondrial dysfunction is a process that occurs because of an attempt to adapt to the disruption of metabolic and energetic pathways occurring in the myocardium. This, in turn, leads to further dysfunction in cardiomyocyte processes. Currently, many therapeutic strategies have been implemented to improve mitochondrial function, but their effectiveness varies widely. AREAS COVERED This review focuses on new models of therapeutic strategies targeting mitochondrial function in the treatment of heart failure. EXPERT OPINION Therapeutic strategies targeting mitochondria appear to be a valuable option for treating heart failure. Currently, the greatest challenge is to develop new research models that could restore the disrupted metabolic processes in mitochondria as comprehensively as possible. Only the development of therapies that focus on improving as many dysregulated mitochondrial processes as possible in patients with heart failure will be able to bring the expected clinical improvement, along with inhibition of disease progression. Combined strategies involving the reduction of the effects of oxidative stress and mitochondrial dysfunction, appear to be a promising possibility for developing new therapies for a complex and multifactorial disease such as heart failure.
Collapse
Affiliation(s)
- Anna Gorący
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Joanna Szostak
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Szymon Retfiński
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Filip Machaj
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
- Department of Medical Biology, Medical University of Warsaw, Warsaw, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
12
|
Chen J, Xu L, Zhang XQ, Liu X, Zhang ZX, Zhu QM, Liu JY, Iqbal MO, Ding N, Shao CL, Wei MY, Gu YC. Discovery of a natural small-molecule AMP-activated kinase activator that alleviates nonalcoholic steatohepatitis. MARINE LIFE SCIENCE & TECHNOLOGY 2023; 5:196-210. [PMID: 37275542 PMCID: PMC10232707 DOI: 10.1007/s42995-023-00168-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/08/2023] [Indexed: 06/07/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a primary cause of cirrhosis and hepatocellular carcinoma. Unfortunately, there is no approved drug treatment for NASH. AMP-activated kinase (AMPK) is an important metabolic sensor and whole-body regulator. It has been proposed that AMPK activators could be used for treating metabolic diseases such as obesity, type 2 diabetes and NASH. In this study, we screened a marine natural compound library by monitoring AMPK activity and found a potent AMPK activator, candidusin A (CHNQD-0803). Further studies showed that CHNQD-0803 directly binds recombinant AMPK with a KD value of 4.728 × 10-8 M and activates AMPK at both molecular and intracellular levels. We then investigated the roles and mechanisms of CHNQD-0803 in PA-induced fat deposition, LPS-stimulated inflammation, TGF-β-induced fibrosis cell models and the MCD-induced mouse model of NASH. The results showed that CHNQD-0803 inhibited the expression of adipogenesis genes and reduced fat deposition, negatively regulated the NF-κB-TNFα inflammatory axis to suppress inflammation, and ameliorated liver injury and fibrosis. These data indicate that CHNQD-0803 as an AMPK activator is a novel potential therapeutic candidate for NASH treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-023-00168-z.
Collapse
Affiliation(s)
- Jin Chen
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
- Key Laboratory of Glycoscience and Glycotechnology of Shandong Province, Qingdao, 266003 China
| | - Li Xu
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
| | - Xue-Qing Zhang
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
| | - Xue Liu
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
- Key Laboratory of Glycoscience and Glycotechnology of Shandong Province, Qingdao, 266003 China
| | - Zi-Xuan Zhang
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
| | - Qiu-Mei Zhu
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
- Key Laboratory of Glycoscience and Glycotechnology of Shandong Province, Qingdao, 266003 China
| | - Jian-Yu Liu
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
| | - Muhammad Omer Iqbal
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
- Key Laboratory of Glycoscience and Glycotechnology of Shandong Province, Qingdao, 266003 China
| | - Ning Ding
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114 USA
| | - Chang-Lun Shao
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 China
| | - Yu-Chao Gu
- Key Laboratory of Marine Drugs, the Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao, 266237 China
- Key Laboratory of Glycoscience and Glycotechnology of Shandong Province, Qingdao, 266003 China
| |
Collapse
|
13
|
Wang J, Qi Z, Wu Y, Wang A, Liu Q, Zou F, Wang B, Qi S, Cao J, Hu C, Shi C, Liang Q, Wang L, Liu J, Wang W, Liu Q. Discovery of IHMT-MST1-39 as a novel MST1 kinase inhibitor and AMPK activator for the treatment of diabetes mellitus. Signal Transduct Target Ther 2023; 8:143. [PMID: 37015918 PMCID: PMC10073293 DOI: 10.1038/s41392-023-01352-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 01/03/2023] [Accepted: 02/01/2023] [Indexed: 04/06/2023] Open
Abstract
Insulin-producing pancreatic β cell death is the fundamental cause of type 1 diabetes (T1D) and a contributing factor to type 2 diabetes (T2D). Moreover, metabolic disorder is another hallmark of T2D. Mammalian sterile 20-like kinase 1 (MST1) contributes to the progression of diabetes mellitus through apoptosis induction and acceleration of pancreatic β cell dysfunction. AMP-activated protein kinase (AMPK) is an energy sensing kinase and its activation has been suggested as a treatment option for metabolic diseases. Thus, pharmacological inhibition of MST1 and activation of AMPK simultaneously represents a promising approach for diabetes therapy. Here, we discovered a novel selective MST1 kinase inhibitor IHMT-MST1-39, which exhibits anti-apoptosis efficacy and improves the survival of pancreatic β cells under diabetogenic conditions, as well as primary pancreatic islets in an ex vivo disease model. Mechanistically, IHMT-MST1-39 activated AMPK signaling pathway in hepatocytes in vitro, combination of IHMT-MST1-39 and metformin synergistically prevented hyperglycemia and significantly ameliorated glucose tolerance and insulin resistance in diabetic mice. Taken together, IHMT-MST1-39 is a promising anti-diabetic candidate as a single agent or in combination therapy for both T1D and T2D.
Collapse
Affiliation(s)
- Junjie Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Ziping Qi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Yun Wu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Aoli Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Qingwang Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Fengming Zou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Shuang Qi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Jiangyan Cao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Chen Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Chenliang Shi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Qianmao Liang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Li Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
| | - Wenchao Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Precision Medicine Research Laboratory of Anhui Province, Hefei, Anhui, 230088, P. R. China.
| |
Collapse
|
14
|
Phair IR, Nisr RB, Howden AJM, Sovakova M, Alqurashi N, Foretz M, Lamont D, Viollet B, Rena G. AMPK integrates metabolite and kinase-based immunometabolic control in macrophages. Mol Metab 2023; 68:101661. [PMID: 36586434 PMCID: PMC9842865 DOI: 10.1016/j.molmet.2022.101661] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/25/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Previous mechanistic studies on immunometabolism have focused on metabolite-based paradigms of regulation, such as itaconate. Here, we, demonstrate integration of metabolite and kinase-based immunometabolic control by AMP kinase. METHODS We combined whole cell quantitative proteomics with gene knockout of AMPKα1. RESULTS Comparing macrophages with AMPKα1 catalytic subunit deletion with wild-type, inflammatory markers are largely unchanged in unstimulated cells, but with an LPS stimulus, AMPKα1 knockout leads to a striking M1 hyperpolarisation. Deletion of AMPKα1 also resulted in increased expression of rate-limiting enzymes involved in itaconate synthesis, metabolism of glucose, arginine, prostaglandins and cholesterol. Consistent with this, we observed functional changes in prostaglandin synthesis and arginine metabolism. Selective AMPKα1 activation also unlocks additional regulation of IL-6 and IL-12 in M1 macrophages. CONCLUSIONS Together, our results validate AMPK as a pivotal immunometabolic regulator in macrophages.
Collapse
Affiliation(s)
- Iain R Phair
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Raid B Nisr
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Andrew J M Howden
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Magdalena Sovakova
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Noor Alqurashi
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| | - Marc Foretz
- Université Paris Cité, Institut Cochin, CNRS, INSERM, F-75014 Paris, France.
| | - Douglas Lamont
- Centre for Advanced Scientific Technologies, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Benoit Viollet
- Université Paris Cité, Institut Cochin, CNRS, INSERM, F-75014 Paris, France.
| | - Graham Rena
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
15
|
Gandhi GR, Hillary VE, Antony PJ, Zhong LLD, Yogesh D, Krishnakumar NM, Ceasar SA, Gan RY. A systematic review on anti-diabetic plant essential oil compounds: Dietary sources, effects, molecular mechanisms, and safety. Crit Rev Food Sci Nutr 2023; 64:6526-6545. [PMID: 36708221 DOI: 10.1080/10408398.2023.2170320] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifaceted metabolic syndrome defined through the dysfunction of pancreatic β-cells driven by a confluence of genetic and environmental elements. Insulin resistance, mediated by interleukins and other inflammatory elements, is one of the key factors contributing to the progression of T2DM. Many essential oils derived from dietary plants are beneficial against various chronic diseases. We reviewed the anti-diabetic properties of dietary plant-derived essential oil compounds, with a focus on their molecular mechanisms by modulating specific signaling pathways and other critical inflammatory mediators involved in insulin resistance. High-quality literature published in the last 12 years, from 2010 to 2022, was collected from the Scopus, Web of Science, PubMed, and Embase databases using the search terms "dietary plants," "essential oils," "anti-diabetic," "insulin resistance," "antihyperglycemic," "T2DM," "anti-diabetic essential oils," and anti-diabetic mechanism." According to the results, the essential oil compounds, including cinnamaldehyde, carvacrol, zingerone, sclareol, zerumbone, myrtenol, thujone, geraniol, citral, eugenol, thymoquinone, thymol, citronellol, α-terpineol, and linalool have been demonstrated to contain strong anti-diabetic effects via modulating various signal transduction pathways linked to glucose metabolism. Additionally, in diabetes-related animal models, they can also considerably reduce the expression of TNF-α, IL-1β, IL-4, IL-6, iNOS, and COX-2. The main signaling molecules regulated by these compounds include AMPK, GLUT4, Caspase-3, PPARγ, PPARα, NF-κB, p-IκBα, MyD88, MCP-1, SREBP-1c, AGEs, RAGE, VEGF, Nrf2/HO-1, and SIRT-1. They can also significantly inhibit the generation of TBARS and MDA, reduce oxidative stress, increase insulin levels, adiponectin, and glycoprotein enzymes, boost antioxidant enzymes like SOD, CAT, and GPx, as well as reduce glutathione and vital glycolytic enzymes. Besides, they can significantly lower the levels of liver enzymes and lipid profile markers. Moreover, most essential oil compounds are generally safe based on animal studies. In conclusion, dietary plant-derived essential oil compounds have potential anti-diabetic effects by influencing different signaling pathways and molecular targets linked to glucose metabolism, and should be safe and beneficial against diabetes and related complications.
Collapse
Affiliation(s)
- Gopalsamy Rajiv Gandhi
- Division of Phytochemistry and Drug-Design, Department of Biosciences, Rajagiri College of Social Sciences (Autonomous), Kochi, India
| | - Varghese Edwin Hillary
- Division of Phytochemistry and Drug-Design, Department of Biosciences, Rajagiri College of Social Sciences (Autonomous), Kochi, India
| | | | - Linda L D Zhong
- Biomedical Sciences and Chinese Medicine, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Devarajan Yogesh
- Department of Biochemistry, University of Madras, Chennai, India
| | | | - Stanislaus Antony Ceasar
- Division of Plant Molecular Biology and Biotechnology, Department of Biosciences, Rajagiri College of Social Sciences, Kochi, India
| | - Ren-You Gan
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
16
|
Wu SY, Chen WM, Chen YC, Chiang MF, Lee MC, Soong RS. Effects of H1-Antihistamines on hepatocellular carcinoma risk in patients with type 2 diabetes mellitus. DIABETES & METABOLISM 2023; 49:101393. [PMID: 36170945 DOI: 10.1016/j.diabet.2022.101393] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 01/28/2023]
Abstract
PURPOSE H1-antihistamines (AHs) may exert protective effects against cancer. We investigated the association of AH use with hepatocellular carcinoma (HCC) risk in type 2 diabetes mellitus (T2DM) patients without hepatitis B virus (HBV) or hepatitis C virus (HCV) infection. METHODS The data of patients with T2DM enrolled from Taiwan's National Health Insurance Research Database were examined for the period of January 1, 2008, to December 31, 2018. We used the Kaplan-Meier method and Cox proportional hazards regression to evaluate the AH use-HCC risk association. RESULTS After 1:1 propensity score matching was performed, the two cohorts were each divided into AH users (n = 47,990) and nonusers (n = 47,990). The risk of HCC was significantly lower in AH users than in AH nonusers (adjusted hazard ratio [aHR]: 0.55 95% confidence interval [95% CI], 0.46 to 0.67; IRR: 0.70; 95% CI, 0.60 to 0.84), respectively. The dose-response relationship between AH use and HCC risk was also observed (aHRs: 0.58, 0.56, 0.50, and 0.41 for 28-35, 36-49, 50-77, and >77 cumulative defined daily doses of AH, respectively). CONCLUSION AH use can reduce HCC risk in T2DM patients without HBV or HCV infection in a dose-dependent manner.
Collapse
Affiliation(s)
- Szu-Yuan Wu
- 1Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan; Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan; Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wan-Ming Chen
- 1Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yi-Chan Chen
- Department of General Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Ming-Feng Chiang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 265, Taiwan
| | - Ming-Che Lee
- Department of Surgery, Taipei Municipal Wanfang Hospital, Taipei, Taiwan; College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ruey-Shyang Soong
- Department of Surgery, Taipei Municipal Wanfang Hospital, Taipei, Taiwan; College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
17
|
Tarasiuk O, Miceli M, Di Domizio A, Nicolini G. AMPK and Diseases: State of the Art Regulation by AMPK-Targeting Molecules. BIOLOGY 2022; 11:biology11071041. [PMID: 36101419 PMCID: PMC9312068 DOI: 10.3390/biology11071041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022]
Abstract
5′-adenosine monophosphate (AMP)-activated protein kinase (AMPK) is an enzyme that regulates cellular energy homeostasis, glucose, fatty acid uptake, and oxidation at low cellular ATP levels. AMPK plays an important role in several molecular mechanisms and physiological conditions. It has been shown that AMPK can be dysregulated in different chronic diseases, such as inflammation, diabetes, obesity, and cancer. Due to its fundamental role in physiological and pathological cellular processes, AMPK is considered one of the most important targets for treating different diseases. Over decades, different AMPK targeting compounds have been discovered, starting from those that activate AMPK indirectly by altering intracellular AMP:ATP ratio to compounds that activate AMPK directly by binding to its activation sites. However, indirect altering of intracellular AMP:ATP ratio influences different cellular processes and induces side effects. Direct AMPK activators showed more promising results in eliminating side effects as well as the possibility to engineer drugs for specific AMPK isoforms activation. In this review, we discuss AMPK targeting drugs, especially concentrating on those compounds that activate AMPK by mimicking AMP. These compounds are poorly described in the literature and still, a lot of questions remain unanswered about the exact mechanism of AMP regulation. Future investigation of the mechanism of AMP binding will make it possible to develop new compounds that, in combination with others, can activate AMPK in a synergistic manner.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
- Correspondence:
| | - Matteo Miceli
- SPILLOproject—Innovative In Silico Solutions for Drug R&D and Pharmacology, 20037 Paderno Dugnano, Italy; (M.M.); (A.D.D.)
| | - Alessandro Di Domizio
- SPILLOproject—Innovative In Silico Solutions for Drug R&D and Pharmacology, 20037 Paderno Dugnano, Italy; (M.M.); (A.D.D.)
| | - Gabriella Nicolini
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
18
|
Park JW, Park JE, Kim SR, Sim MK, Kang CM, Kim KS. Metformin alleviates ionizing radiation-induced senescence by restoring BARD1-mediated DNA repair in human aortic endothelial cells. Exp Gerontol 2022; 160:111706. [DOI: 10.1016/j.exger.2022.111706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/27/2021] [Accepted: 01/13/2022] [Indexed: 12/20/2022]
|
19
|
H. Alhowai A, Almogbel Y, A.H. Abdel A, A. Aldubay M, Alfheeaid HA, Felemban SG, Chigurupat S, F. Alharbi I, S. Alharbi H. Metformin Induced Cognitive Impairment and Neuroinflammation in CMF-Treated Rats. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.228.235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
20
|
The early reduction of left ventricular mass after sleeve gastrectomy depends on the fall of branched-chain amino acid circulating levels. EBioMedicine 2022; 76:103864. [PMID: 35131692 PMCID: PMC8829082 DOI: 10.1016/j.ebiom.2022.103864] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/24/2021] [Accepted: 01/19/2022] [Indexed: 01/03/2023] Open
Abstract
Introduction Body-mass index is a major determinant of left-ventricular-mass (LVM). Bariatric-metabolic surgery (BMS) reduces cardiovascular mortality. Its mechanism of action, however, often encompasses a weight-dependent effect. In this translational study, we aimed at investigating the mechanisms by which BMS leads to LVM reduction and functional improvement. Methods Twenty patients (45.2 ± 8.5years) were studied with echocardiography at baseline and at 1,6,12 and 48 months after sleeve-gastrectomy (SG). Ten Wistar rats aged 10-weeks received high-fat diet ad libitum for 10 weeks before and 4 weeks after SG or sham-operation. An oral-glucose-tolerance-test was performed to measure whole-body insulin-sensitivity. Plasma metabolomics was analysed in both human and rodent samples. RNA quantitative Real-Time PCR and western blots were performed in rodent heart biopsies. The best-fitted partial-least-square discriminant-analysis model was used to explore the variable importance in the projection score of all metabolites. Findings Echocardiographic LVM (-12%,-23%,-28% and -43% at 1,6,12 and 48 months, respectively) and epicardial fat decreased overtime after SG in humans while insulin-sensitivity improved. In rats, SG significantly reduced LVM and epicardial fat, enhanced ejection-fraction and improved insulin-sensitivity compared to sham-operation. Metabolomics showed a progressive decline of plasma branched-chain amino-acids (BCAA), alanine, lactate, 3-OH-butyrate, acetoacetate, creatine and creatinine levels in both humans and rodents. Hearts of SG rats had a more efficient BCAA, glucose and fatty-acid metabolism and insulin signaling than sham-operation. BCAAs in cardiomyocyte culture-medium stimulated lipogenic gene transcription and reduced mRNA levels of key mitochondrial β-oxidation enzymes promoting lipid droplet accumulation and glycolysis. Interpretation After SG a prompt and sustained decrease of the LVM, epicardial fat and insulin resistance was found. Animal and in vitro studies showed that SG improves cardiac BCAA metabolism with consequent amelioration of fat oxidation and insulin signaling translating into decreased intra-myocytic fat accumulation and reduced lipotoxicity.
Collapse
|
21
|
Clinical Study on the Relationship between the SNP rs8192675 (C/C) Site of SLC2A2 Gene and the Hypoglycemic Effect of Metformin in Type 2 Diabetes. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3645336. [PMID: 35140900 PMCID: PMC8820847 DOI: 10.1155/2022/3645336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 11/29/2022]
Abstract
This study investigates the correlation between the gene polymorphism of rs8192675 (C/C) locus of SLC2A2 in patients with type 2 diabetes (T2DM) and the efficacy of metformin. For this purpose, we have selected 110 T2DM patients (T2DM group) and 110 healthy people (control group) who were treated in our hospital from January 2019 to January 2020 as the research subjects. PCR-restriction fragment length polymorphism (PCR-RFLP) method detects the distribution frequency of gene polymorphism. The patients in the T2DM group were treated with metformin and followed up for 90 days to analyze the relationship between the efficacy of metformin and the SLC2A2 gene polymorphism. The genotypes of SLC2A2 rs8192675 in the control group and in the T2DM group conformed to the Hardy–Weinberg equilibrium law. Compared with the control group, the CT type and the CC type at rs8192675 in the T2DM group were significantly higher (P < 0.05). For rs8192675, there was no significant difference in TT, CT, CC FPG, 2hPBG, and HbA1c levels before treatment (P > 0.05); after metformin treatment, the reduction in FPG, 2hPBG, and HbA1c in CC patients was lower than that of TT and CT patients (P < 0.05). SLC2A2 gene polymorphism site rs8192675 CC type T2DM patients are sensitive to metformin and have a better hypoglycemic effect.
Collapse
|
22
|
Mukherjee S, Skrede S, Milbank E, Andriantsitohaina R, López M, Fernø J. Understanding the Effects of Antipsychotics on Appetite Control. Front Nutr 2022; 8:815456. [PMID: 35047549 PMCID: PMC8762106 DOI: 10.3389/fnut.2021.815456] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022] Open
Abstract
Antipsychotic drugs (APDs) represent a cornerstone in the treatment of schizophrenia and other psychoses. The effectiveness of the first generation (typical) APDs are hampered by so-called extrapyramidal side effects, and they have gradually been replaced by second (atypical) and third-generation APDs, with less extrapyramidal side effects and, in some cases, improved efficacy. However, the use of many of the current APDs has been limited due to their propensity to stimulate appetite, weight gain, and increased risk for developing type 2 diabetes and cardiovascular disease in this patient group. The mechanisms behind the appetite-stimulating effects of the various APDs are not fully elucidated, partly because their diverse receptor binding profiles may affect different downstream pathways. It is critical to identify the molecular mechanisms underlying drug-induced hyperphagia, both because this may lead to the development of new APDs, with lower appetite-stimulating effects but also because such insight may provide new knowledge about appetite regulation in general. Hence, in this review, we discuss the receptor binding profile of various APDs in relation to the potential mechanisms by which they affect appetite.
Collapse
Affiliation(s)
- Sayani Mukherjee
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Silje Skrede
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Edward Milbank
- NeurObesity Group, Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Madrid, Spain.,SOPAM, U1063, INSERM, University of Angers, SFR ICAT, Bat IRIS-IBS, Angers, France
| | | | - Miguel López
- NeurObesity Group, Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
23
|
Abdallah BM, Alzahrani AM. A-769662 stimulates the differentiation of bone marrow-derived mesenchymal stem cells into osteoblasts via AMP-activated protein kinase-dependent mechanism. J Appl Biomed 2021; 19:159-169. [PMID: 34907759 DOI: 10.32725/jab.2021.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/21/2021] [Indexed: 12/25/2022] Open
Abstract
AMP-activated protein kinase (AMPK) signaling shows an important role in energy metabolism and has recently been involved in osteogenic and adipogenic differentiation. In this study we aimed to investigate the role of AMPK activator, A-769662, in regulating the differentiation of mesenchymal stem cells derived from bone marrow (BMSCs) into osteoblastic and adipocytic cell lineage. The effect of A-769662 on osteogenesis was assessed by quantitative alkaline phosphatase (ALP) activity, matrix mineralization stained with Alizarin red, and gene expression analysis by quantitative polymerase chain reaction (qPCR). Adipogenesis was determined by Oil Red O staining for fat droplets and qPCR analysis of adipogenic markers. A-769662 activated the phosphorylation of AMPKα1 during the osteogenesis of mBMSCs as revealed by western blot analysis. A-769662 promoted the early stage of the commitment of mouse (m) BMSCs differentiation into osteoblasts, while inhibiting their differentiation into adipocytes in a dose-dependent manner. The effects of A-769662 on stimulating osteogenesis and inhibiting adipogenesis of mBMSCs were significantly eliminated in the presence of either AMPKα1 siRNA or Compound C, an inhibitor of AMPK pathway. In conclusion, we identified A-769662 as a new compound that promotes the commitment of BMSCs into osteoblasts versus adipocytes via AMPK-dependent mechanism. Thus our data show A-769662 as a potential osteo-anabolic drug for treatment of osteoporosis.
Collapse
Affiliation(s)
| | - Abdullah M Alzahrani
- King Faisal University, College of Science, Biological Sciences Department, Al-Ahsa, Saudi Arabia
| |
Collapse
|
24
|
Li J, Sun Q, Qiu X, Zhang J, Zheng Y, Luo L, Tan X. Downregulation of AMPK dependent FOXO3 and TFEB involves in the inhibition of autophagy in diabetic cataract. Curr Eye Res 2021; 47:555-564. [PMID: 34872443 DOI: 10.1080/02713683.2021.2009516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE Autophagy plays a crucial role in intracellular quality control of crystalline lens and AMPK has regulatory effect on autophagy. However, whether AMPK regulated autophagy is involved in diabetic cataract (DC) progression remains unknown. This study aims to investigate the AMPK-FOXO3 and AMPK-TFEB induced autophagy activity in DC patients. MATERIALS AND METHODS First, anterior capsule specimens from DC and age-related cataract (ARC) patients were obtained to compare the expression difference of autophagy-related genes. The phosphorylation levels of AMPK, AKT, and mTOR and the expression of FOXO3 and TFEB were measured. Then, human lens epithelial cells (LECs, SRA 01/04) were cultured with 30 mM or 5.5 mM glucose, and AMPK activator (AICAR) and inhibitor (Compound C) were applied to further investigate the regulatory role of AMPK on autophagy. RESULTS Compared with ARC patients, the expression of autophagy-related genes ATG5, FYCO1, ATG8, ATG12, Beclin1, and ULK1 in anterior capsules LECs of DC patients were significantly down-regulated. Meanwhile, AMPK and AMPK-dependent transcription factors, FOXO3 and TFEB were also inhibited. Similar results were found in high glucose (HG) treated SRA 01/04 model. Notably, this down-regulation of autophagy activity was rescued by AICAR in vitro, which was manifested by inhibition of AKT and mTOR phosphorylation and up-regulation of FOXO3, TFEB, Beclin1 and LC3B-II expression. CONCLUSIONS Down-regulation of AMPK-FOXO3 and AMPK-TFEB induced autophagy activity was found in both LECs of anterior capsule from DC patients and SRA 01/04 cells under HG condition, which may be the underlying mechanism of DC formation. Thus, targeting AMPK-induced autophagy may be a potential therapeutic approach for diabetic cataract.
Collapse
Affiliation(s)
- Jiani Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qihang Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaozhang Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jiaqing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxing Zheng
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xuhua Tan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
25
|
|
26
|
Abtahi SH, Mohammadi MH, Allahbakhshian Farsani M, Aghelan Z, Salari S. Evaluation of Sestrin 2, Adiponectin, AMPK, and mTOR Genes Expression in Acute Myeloid Leukemia Patients. IRANIAN JOURNAL OF BIOTECHNOLOGY 2021; 19:e2860. [PMID: 34435062 PMCID: PMC8358177 DOI: 10.30498/ijb.2021.2860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Effective treatment of acute myeloid leukemia (AML) is still controversial, therefore; a comprehensive understanding regarding the impaired cellular signaling pathways in AML can be useful
in designing new therapeutic approaches. Among signaling pathways involved in AML, the mammalian target of rapamycin (mTOR) signaling pathway is of particular importance.
While dysregulation of mTOR signaling has been reported in a wide range of patients with AML, but most studies have focused on mTOR downstream targets, and mTOR upstream targets have been overlooked. Objective: In this study, expression of mTOR genes and three upstream targets (5' adenosine monophosphate-activated protein kinase (AMPK, adiponectin, and sestrin 2)
involved in mTOR signaling was investigated. Materials and Methods: In this study, expression of mTOR, AMPK, sestrin 2, and adiponectin genes in 60 patients with AML were evaluated compared to those of 30 healthy individuals as controls
using the Real-Time polymerase chain reaction (Real-Time RT-PCR) method. Results: According to the results, there was a significant difference in the expression of all the studied genes in patients in comparison to the normal control group (P <0.05).
Expression of the mTOR gene was increased, while expression of AMPK, sestrin 2, and adiponectin genes was decreased in the patients with AML. Mean expression of the genes (2-ΔCt)
(AMPK, sestrin 2, adiponectin, and mTOR) was equal to 7.9, 3.2, 3.74, and 1.49 for controls and 6, 2.1, 2.83, and 2.64 for patients with AML, respectively. Conclusions: Given the decreased expression levels of sestrin 2, adiponectin, and AMPK genes as tumor inhibitors and the increased expression level of the mTOR gene as an oncogene in the
patients with AML in our study, it is thought that disruption of this pathway may be involved in leukemogenesis and can be considered as an effective factor in the progression of cancer.
Collapse
Affiliation(s)
- Seyed Hossein Abtahi
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Mohammadi
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,HSCT Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mehdi Allahbakhshian Farsani
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,HSCT Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Zahra Aghelan
- Department of Clinical Biochemistry, School of Medical Siences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sina Salari
- Department of Medical Oncology, Hematology and Bone Marrow Transplantation, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Park HE, Lee W, Shin MK, Shin SJ. Understanding the Reciprocal Interplay Between Antibiotics and Host Immune System: How Can We Improve the Anti-Mycobacterial Activity of Current Drugs to Better Control Tuberculosis? Front Immunol 2021; 12:703060. [PMID: 34262571 PMCID: PMC8273550 DOI: 10.3389/fimmu.2021.703060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, remains a global health threat despite recent advances and insights into host-pathogen interactions and the identification of diverse pathways that may be novel therapeutic targets for TB treatment. In addition, the emergence and spread of multidrug-resistant Mtb strains led to a low success rate of TB treatments. Thus, novel strategies involving the host immune system that boost the effectiveness of existing antibiotics have been recently suggested to better control TB. However, the lack of comprehensive understanding of the immunomodulatory effects of anti-TB drugs, including first-line drugs and newly introduced antibiotics, on bystander and effector immune cells curtailed the development of effective therapeutic strategies to combat Mtb infection. In this review, we focus on the influence of host immune-mediated stresses, such as lysosomal activation, metabolic changes, oxidative stress, mitochondrial damage, and immune mediators, on the activities of anti-TB drugs. In addition, we discuss how anti-TB drugs facilitate the generation of Mtb populations that are resistant to host immune response or disrupt host immunity. Thus, further understanding the interplay between anti-TB drugs and host immune responses may enhance effective host antimicrobial activities and prevent Mtb tolerance to antibiotic and immune attacks. Finally, this review highlights novel adjunctive therapeutic approaches against Mtb infection for better disease outcomes, shorter treatment duration, and improved treatment efficacy based on reciprocal interactions between current TB antibiotics and host immune cells.
Collapse
Affiliation(s)
- Hyun-Eui Park
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
28
|
Maldonado M, Chen J, Lujun Y, Duan H, Raja MA, Qu T, Huang T, Gu J, Zhong Y. The consequences of a high-calorie diet background before calorie restriction on skeletal muscles in a mouse model. Aging (Albany NY) 2021; 13:16834-16858. [PMID: 34166224 PMCID: PMC8266348 DOI: 10.18632/aging.203237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/31/2021] [Indexed: 02/05/2023]
Abstract
The beneficial effects of calorie restriction (CR) are numerous. However, there is no scientific evidence about how a high-calorie diet (HCD) background influences the mechanisms underlying CR on skeletal muscles in an experimental mouse model. Herein we present empirical evidence showing significant interactions between HCD (4 months) and CR (3 months). Pectoralis major and quadriceps femoris vastus medialis, in the experimental and control groups, displayed metabolic and physiologic heterogeneity and remarkable plasticity, according to the dietary interventions. HCD-CR not only altered genetic activation patterns of satellite SC markers but also boosted the expression of myogenic regulatory factors and key activators of mitochondrial biogenesis, which in turn were also associated with metabolic fiber transition. Our data prompt us to theorize that the effects of CR may vary according to the physiologic, metabolic, and genetic peculiarities of the skeletal muscle described here and that INTM/IM lipid infiltration and tissue-specific fuel-energy status (demand/supply) both hold dependent-interacting roles with other key anti-aging mechanisms triggered by CR. Systematic integration of an HCD with CR appears to bring potential benefits for skeletal muscle function and energy metabolism. However, at this stage of our research, an optimal balance between the two dietary conditions, where anti-aging effects can be accomplished, is under intensive investigation in combination with other tissues and organs at different levels of organization within the organ system.
Collapse
Affiliation(s)
- Martin Maldonado
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| | - Jianying Chen
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| | - Yang Lujun
- Translational Medical Center, Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, P.R. China
| | - Huiqin Duan
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| | - Mazhar Ali Raja
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| | - Ting Qu
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| | - Tianhua Huang
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| | - Jiang Gu
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| | - Ying Zhong
- Chengdu Jinxin Institute of Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu 610066, China
| |
Collapse
|
29
|
Aledavood E, Forte A, Estarellas C, Javier Luque F. Structural basis of the selective activation of enzyme isoforms: Allosteric response to activators of β1- and β2-containing AMPK complexes. Comput Struct Biotechnol J 2021; 19:3394-3406. [PMID: 34194666 PMCID: PMC8217686 DOI: 10.1016/j.csbj.2021.05.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/30/2021] [Accepted: 05/30/2021] [Indexed: 12/21/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a key energy sensor regulating the cell metabolism in response to energy supply and demand. The evolutionary adaptation of AMPK to different tissues is accomplished through the expression of distinct isoforms that can form up to 12 complexes, which exhibit notable differences in the sensitivity to allosteric activators. To shed light into the molecular determinants of the allosteric regulation of this energy sensor, we have examined the structural and dynamical properties of β1- and β2-containing AMPK complexes formed with small molecule activators A-769662 and SC4, and dissected the mechanical response leading to active-like enzyme conformations through the analysis of interaction networks between structural domains. The results reveal the mechanical sensitivity of the α2β1 complex, in contrast with a larger resilience of the α2β2 species, especially regarding modulation by A-769662. Furthermore, binding of activators to α2β1 consistently promotes the pre-organization of the ATP-binding site, favoring the adoption of activated states of the enzyme. These findings are discussed in light of the changes in the residue content of β-subunit isoforms, particularly regarding the β1Asn111 → β2Asp111 substitution as a key factor in modulating the mechanical sensitivity of β1- and β2-containing AMPK complexes. Our studies pave the way for the design of activators tailored for improving the therapeutic treatment of tissue-specific metabolic disorders.
Collapse
Affiliation(s)
| | - Alessia Forte
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB) and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Av. Prat de la Riba 171, Santa Coloma de Gramenet 08921, Spain
| | | | | |
Collapse
|
30
|
Regulation of AMPK Activity by CRBN Is Independent of the Thalidomide-CRL4 CRBN Protein Degradation Axis. Pharmaceuticals (Basel) 2021; 14:ph14060512. [PMID: 34073624 PMCID: PMC8228114 DOI: 10.3390/ph14060512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022] Open
Abstract
Cereblon (CRBN), a primary target of immune-modulatory imide drugs (IMiDs), functions as a substrate receptor in the CUL4-RBX1-DDB1-CRBN (known as CRL4CRBN) E3 ubiquitin ligase complex. Binding of IMiDs to CRBN redirects the CRL4CRBN E3 ubiquitin ligase to recruit or displace its substrates. Interaction between CRBN and the AMPK α subunit leads to CRL4CRBN-dependent degradation of the γ subunit and inhibits AMPK activity. However, the effect of thalidomide on the function of CRBN as a negative regulator of AMPK through interaction with the α subunit remains unclear. Here, we show that thalidomide does not affect AMPK activation or the binding affinity between CRBN and the AMPK α subunit. Thalidomide had no effect on AMPK activity independent of CRBN expression. The N-terminal region and C-terminal tail of CRBN, which is distinct from the IMiD binding site, were critical for interaction with the AMPK α subunit. The present results suggest that CRL4CRBN negatively regulates AMPK through a pathway independent from the CRBN-IMiD binding region.
Collapse
|
31
|
Lim YJ, Kim KM, Jang WG. Chrysophanol increases osteoblast differentiation via AMPK/Smad1/5/9 phosphorylation in vitro and in vivo. Clin Exp Pharmacol Physiol 2021; 48:515-523. [PMID: 33300218 DOI: 10.1111/1440-1681.13443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/05/2020] [Indexed: 01/20/2023]
Abstract
Chrysophanol (Chrysophanic acid; CA) is a natural anthraquinone found in Senna tora and rhubarb that has various characteristic features, including the ability to suppress adipogenesis. However, its effects on osteoblast differentiation have not been investigated. Herein, this study aimed to demonstrate the mechanism by which CA induces the osteoblast differentiation. CA increased the expression of osteogenic genes. The staining levels Alkaline phosphatase (ALP) and Alizarin Red S (ARS) were increased by chrysophanol. CA induced osteoblast differentiation through AMP-activated protein kinase (AMPK)/Small mothers against decapentaplegic (Smad1/5/9) activation in MC3T3-E1 cells. In addition, compound C, AMPK inhibitor (Comp. C)-induced cells suppressed osteogenic genes expression and AMPK/Smad1/5/9 activation. Interestingly, AMPK in the CA-induced AMPK/Smad1/5/9 signalling pathway was an upstream regulator of Smad1/5/9. In order to further dissect in bone development, we used a zebrafish model to investigate the effect of CA on bone development. These results suggest that CA stimulated bone development via AMPK/Smad1/5/9. Overall, our results demonstrate that CA promotes osteoblast differentiation via AMPK/Smad1/5/9 expression in vitro and in vivo.
Collapse
Affiliation(s)
- Young-Ju Lim
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongbuk, Korea
- Research Institute of Anti-Aging, Daegu University, Gyeongbuk, Korea
| | - Kyeong-Min Kim
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongbuk, Korea
- Research Institute of Anti-Aging, Daegu University, Gyeongbuk, Korea
| | - Won-Gu Jang
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongbuk, Korea
- Research Institute of Anti-Aging, Daegu University, Gyeongbuk, Korea
| |
Collapse
|
32
|
Podhorecka M. Metformin - its anti-cancer effects in hematologic malignancies. Oncol Rev 2021; 15:514. [PMID: 33747367 PMCID: PMC7967492 DOI: 10.4081/oncol.2021.514] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/26/2021] [Indexed: 02/08/2023] Open
Abstract
The main anti-diabetic effect of metformin mediated through stimulation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) is the inhibition of hepatic gluconeogenesis and triggering glucose uptake in skeletal muscles. Additionally, some new pathways, besides the AMPK activation, were discovered, that can explain wide-range properties of metformin. All these properties are now attracting the attention of researchers in the fields other than diabetes and the drug has been reported to have anti-cancer, immunoregulatory and anti-aging effects. Among others, the beneficial effects of metformin in hematological disorders like leukemias, lymphomas, and multiple myeloma were reported. Despite a great progress in therapy, these diseases are still incurable in most cases. Thus, there is an urgent need to discover novel, less toxic and more effective drugs especially for older or chemotherapy-resistant patients. In this review article, the current findings on the anti-cancer effect of metformin together with underlying possible mechanisms in blood cancers are discussed. However. to evaluate precisely these promising effects of metformin, more studies are required, because many of the published results are preclinical.
Collapse
Affiliation(s)
- Monika Podhorecka
- Department of Hematooncology and Bone Marrow Transplantation Medical University of Lublin, Poland
| |
Collapse
|
33
|
Ren C, Han R, Hu J, Li H, Li S, Liu Y, Cheng Z, Ji X, Ding Y. Hypoxia post-conditioning promoted glycolysis in mice cerebral ischemic model. Brain Res 2020; 1748:147044. [DOI: 10.1016/j.brainres.2020.147044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 01/10/2023]
|
34
|
Irungbam K, Roderfeld M, Glimm H, Hempel F, Schneider F, Hehr L, Glebe D, Churin Y, Morlock G, Yüce I, Roeb E. Cholestasis impairs hepatic lipid storage via AMPK and CREB signaling in hepatitis B virus surface protein transgenic mice. J Transl Med 2020; 100:1411-1424. [PMID: 32612285 PMCID: PMC7572243 DOI: 10.1038/s41374-020-0457-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Clinical studies demonstrated that nonalcoholic steatohepatitis is associated with liver-related outcomes in chronic hepatitis B. Furthermore, primary biliary fibrosis and biliary atresia occurred in patients with HBV infection. Interestingly, hepatitis B virus surface protein (HBs) transgenic mice spontaneously develop hepatic steatosis. Our aim is to investigate the effect of Abcb4 knockout-induced cholestasis on liver steatosis in HBs transgenic mice. Hybrids of HBs transgenic and Abcb4-/- mice were bred on the BALB/c genetic background. Lipid synthesis, storage, and catabolism as well as proteins and genes that control lipid metabolism were analyzed using HPTLC, qPCR, western blot, electrophoretic mobility shift assay (EMSA), lipid staining, and immunohistochemistry. Hepatic neutral lipid depots were increased in HBs transgenic mice and remarkably reduced in Abcb4-/- and HBs/Abcb4-/- mice. Similarly, HPTLC-based quantification analyses of total hepatic lipid extracts revealed a significant reduction in the amount of triacylglycerols (TAG), while the amount of free fatty acids (FFA) was increased in Abcb4-/- and HBs/Abcb4-/- in comparison to wild-type and HBs mice. PLIN2, a lipid droplet-associated protein, was less expressed in Abcb4-/- and HBs/Abcb4-/-. The expression of genes-encoding proteins involved in TAG synthesis and de novo lipogenesis (Agpat1, Gpat1, Mgat1, Dgat1, Dgat2, Fasn, Hmgcs1, Acc1, Srebp1-c, and Pparγ) was suppressed, and AMPK and CREB were activated in Abcb4-/- and HBs/Abcb4-/- compared to wild-type and HBs mice. Simulating cholestatic conditions in cell culture resulted in AMPK and CREB activation while FASN and PLIN2 were reduced. A concurrent inhibition of AMPK signaling revealed normal expression level of FASN and PLIN2, suggesting that activation of AMPK-CREB signaling regulates hepatic lipid metabolism, i.e. synthesis and storage, under cholestatic condition. In conclusions, in vivo and mechanistic in vitro data suggest that cholestasis reduces hepatic lipid storage via AMPK and CREB signaling. The results of the current study could be the basis for novel therapeutic strategies as NASH is a crucial factor that can aggravate chronic liver diseases.
Collapse
Affiliation(s)
- Karuna Irungbam
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Hannah Glimm
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Felix Hempel
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Franziska Schneider
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Laura Hehr
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Dieter Glebe
- Institute of Medical Virology, National Reference Centre for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Yuri Churin
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Gertrud Morlock
- Institute of Nutritional Science, Chair of Food Science, and TransMIT Center for Effect-Directed Analysis, Justus Liebig University Giessen, Giessen, Germany
| | - Imanuel Yüce
- Institute of Nutritional Science, Chair of Food Science, and TransMIT Center for Effect-Directed Analysis, Justus Liebig University Giessen, Giessen, Germany
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
35
|
Choi EH, Chun YS, Kim J, Ku SK, Jeon S, Park TS, Shim SM. Modulating lipid and glucose metabolism by glycosylated kaempferol rich roasted leaves of Lycium chinense via upregulating adiponectin and AMPK activation in obese mice-induced type 2 diabetes. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
36
|
Sachar M, Kumar V, Gormsen LC, Munk OL, Unadkat JD. Successful Prediction of Positron Emission Tomography-Imaged Metformin Hepatic Uptake Clearance in Humans Using the Quantitative Proteomics-Informed Relative Expression Factor Approach. Drug Metab Dispos 2020; 48:1210-1216. [PMID: 32843330 DOI: 10.1124/dmd.120.000156] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
Predicting transporter-mediated in vivo hepatic drug clearance (CL) from in vitro data (IVIVE) is important in drug development to estimate first-in-human dose and the impact of drug interactions and pharmacogenetics on hepatic drug CL. For IVIVE, one can use human hepatocytes and the traditional milligrams of protein content per gram of liver tissue (MGPGL) approach. However, this approach has been found to consistently underpredict the observed in vivo hepatic drug CL. Therefore, we hypothesized that using transporter-expressing cells and the relative expression factor (REF), determined using targeted quantitative proteomics, will accurately predict in vivo hepatic CL of drugs. We have successfully tested this hypothesis in rats with rosuvastatin, which is transported by hepatic Organic anion transporting polypeptides (OATPs). Here, we tested this hypothesis for another drug and another transporter; namely, organic cation transporter (OCT)1-mediated hepatic distributional CL of metformin. First, we estimated the in vivo metformin hepatic sinusoidal uptake CL (CLh,s,in) of metformin by reanalysis of previously published human positron emission tomography imaging data. Next, using the REF approach, we predicted the in vivo metformin CLh,s,in using OCT1-transporter-expressing HEK293 cells or plated human hepatocytes. Finally, we compared this REF-based prediction with that using the MGPGL approach. The REF approach accurately predicted the in vivo metformin hepatic CLh,s,in, whereas the MGPGL approach considerably underpredicted the in vivo metformin CLh,s,in Based on these and previously published data, the REF approach appears to be superior to the MGPGL approach for a diverse set of drugs transported by different transporters. SIGNIFICANCE STATEMENT: This study is the first to use organic cation transporter 1-expressing cells and plated hepatocytes to compare proteomics-informed REF approach with the traditional MGPGL approach to predict hepatic uptake CL of metformin in humans. The proteomics-informed REF approach, which corrected for plasma membrane abundance, accurately predicted the positron emission tomography-imaged metformin hepatic uptake CL, whereas the MGPGL approach consistently underpredicted this CL.
Collapse
Affiliation(s)
- Madhav Sachar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Vineet Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Lars C Gormsen
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Ole Lajord Munk
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| |
Collapse
|
37
|
Two isoprenylated flavonoids from Dorstenia psilurus activate AMPK, stimulate glucose uptake, inhibit glucose production and lower glycemia. Biochem J 2020; 476:3687-3704. [PMID: 31782497 DOI: 10.1042/bcj20190326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022]
Abstract
Root extracts of a Cameroon medicinal plant, Dorstenia psilurus, were purified by screening for AMP-activated protein kinase (AMPK) activation in incubated mouse embryo fibroblasts (MEFs). Two isoprenylated flavones that activated AMPK were isolated. Compound 1 was identified as artelasticin by high-resolution electrospray ionization mass spectrometry and 2D-NMR while its structural isomer, compound 2, was isolated for the first time and differed only by the position of one double bond on one isoprenyl substituent. Treatment of MEFs with purified compound 1 or compound 2 led to rapid and robust AMPK activation at low micromolar concentrations and increased the intracellular AMP:ATP ratio. In oxygen consumption experiments on isolated rat liver mitochondria, compound 1 and compound 2 inhibited complex II of the electron transport chain and in freeze-thawed mitochondria succinate dehydrogenase was inhibited. In incubated rat skeletal muscles, both compounds activated AMPK and stimulated glucose uptake. Moreover, these effects were lost in muscles pre-incubated with AMPK inhibitor SBI-0206965, suggesting AMPK dependency. Incubation of mouse hepatocytes with compound 1 or compound 2 led to AMPK activation, but glucose production was decreased in hepatocytes from both wild-type and AMPKβ1-/- mice, suggesting that this effect was not AMPK-dependent. However, when administered intraperitoneally to high-fat diet-induced insulin-resistant mice, compound 1 and compound 2 had blood glucose-lowering effects. In addition, compound 1 and compound 2 reduced the viability of several human cancer cells in culture. The flavonoids we have identified could be a starting point for the development of new drugs to treat type 2 diabetes.
Collapse
|
38
|
Cordycepin for Health and Wellbeing: A Potent Bioactive Metabolite of an Entomopathogenic Cordyceps Medicinal Fungus and Its Nutraceutical and Therapeutic Potential. Molecules 2020; 25:molecules25122735. [PMID: 32545666 PMCID: PMC7356751 DOI: 10.3390/molecules25122735] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Cordyceps is a rare naturally occurring entomopathogenic fungus usually found at high altitudes on the Himalayan plateau and a well-known medicinal mushroom in traditional Chinese medicine. Cordyceps contains various bioactive components, out of which, cordycepin is considered most vital, due to its utmost therapeutic as well as nutraceutical potential. Moreover, the structure similarity of cordycepin with adenosine makes it an important bioactive component, with difference of only hydroxyl group, lacking in the 3′ position of its ribose moiety. Cordycepin is known for various nutraceutical and therapeutic potential, such as anti-diabetic, anti-hyperlipidemia, anti-fungal, anti-inflammatory, immunomodulatory, antioxidant, anti-aging, anticancer, antiviral, hepato-protective, hypo-sexuality, cardiovascular diseases, antimalarial, anti-osteoporotic, anti-arthritic, cosmeceutical etc. which makes it a most valuable medicinal mushroom for helping in maintaining good health. In this review, effort has been made to bring altogether the possible wide range of cordycepin’s nutraceutical potential along with its pharmacological actions and possible mechanism. Additionally, it also summarizes the details of cordycepin based nutraceuticals predominantly available in the market with expected global value. Moreover, this review will attract the attention of food scientists, nutritionists, pharmaceutical and food industries to improve the use of bioactive molecule cordycepin for nutraceutical purposes with commercialization to aid and promote healthy lifestyle, wellness and wellbeing.
Collapse
|
39
|
Metformin: A Possible Option in Cancer Chemotherapy. Anal Cell Pathol (Amst) 2020; 2020:7180923. [PMID: 32399389 PMCID: PMC7201450 DOI: 10.1155/2020/7180923] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/20/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023] Open
Abstract
Metformin has been used for a long time as an antidiabetic medication for type 2 diabetes. It is used either as a monotherapy or in combination with other antidiabetic medications. The drug came into prominence in diabetes and other conditions with cardiovascular risk after the landmark study of 1995 by the United Kingdom Prospective Diabetes Study which emphasized its importance. However, the drug has been used in experimental trials in various aspects of medicine and pharmacology such as in reproductive medicine, cancer chemotherapy, metabolic diseases, and neurodegenerative diseases. It has been in use in the treatment of polycystic ovarian disease and obesity and is being considered in type 1 diabetes. This study seeks to evaluate the relevance of metformin in cancer management. Different mechanisms have been proposed for its antitumor action which involves the following: (a) the activation of adenosine monophosphate kinase, (b) modulation of adenosine A1 receptor (ADORA), (c) reduction in insulin/insulin growth factors, and (d) the role of metformin in the inhibition of endogenous reactive oxygen species (ROS); and its resultant damage to deoxyribonucleic acid (DNA) molecule is another paramount antitumor mechanism.
Collapse
|
40
|
Alhowail A, Aldubayan M, Alqasomi A, Alharbi I, Alharbi H. Effects of Metformin on the Survival Rate of CMF (Cyclophosphamide, Methotrexate and 5-fluorouracil)-treated Rats. INT J PHARMACOL 2020. [DOI: 10.3923/ijp.2020.201.204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
41
|
Analysis of urinary metabolic alteration in type 2 diabetic rats treated with metformin using the metabolomics of quantitative spectral deconvolution 1H NMR spectroscopy. Microchem J 2020. [DOI: 10.1016/j.microc.2019.104513] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
42
|
Kim HS, Kim JH, Jang HJ, Lee J. The addition of metformin to systemic anticancer therapy in advanced or metastatic cancers: a meta-analysis of randomized controlled trials. Int J Med Sci 2020; 17:2551-2560. [PMID: 33029097 PMCID: PMC7532491 DOI: 10.7150/ijms.50338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
Preclinical studies have demonstrated that metformin has anticancer properties and act in additive or synergistic way when combined with anticancer agents. We conducted this meta-analysis of randomized clinical trials to evaluate the effect of metformin added to systemic anticancer therapy in patients with advanced or metastatic cancer. A computerized systematic electronic search was performed using PubMed, PMC, EMBASE, Cochrane Library, and Web of Science databases (up to June 2020). From nine randomized clinical trials, 821 patients were included in the pooled analyses of odds ratios (ORs) with 95% confidence intervals (CIs) for overall response rate (ORR) and hazard ratios (HRs) with 95% CIs for progression-free survival (PFS) and overall survival (OS). The concomitant use of metformin with systemic anticancer therapy did not increase tumor response (the pooled OR of ORR = 1.23, 95% CI: 0.89-1.71, p = 0.21), compared with anticancer therapy alone. In terms of survival, metformin added to anticancer agents failed to prolong PFS (HR = 0.95, 95% CI: 0.75-1.21, p = 0.68) and OS (HR = 0.97, 95% CI: 0.80-1.16, p = 0.71). In conclusion, this meta-analysis of randomized clinical trials indicates that the addition of metformin to systemic anticancer therapy has no clinical benefits in patients with advanced or metastatic cancer.
Collapse
Affiliation(s)
- Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Hwasung 18450, Gyeonggi-Do, Republic of Korea
| | - Jin Lee
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Hwasung 18450, Gyeonggi-Do, Republic of Korea
| |
Collapse
|
43
|
Old wine in new bottles: Drug repurposing in oncology. Eur J Pharmacol 2020; 866:172784. [DOI: 10.1016/j.ejphar.2019.172784] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023]
|
44
|
Participation of the adenosine salvage pathway and cyclic AMP modulation in oocyte energy metabolism. Sci Rep 2019; 9:18395. [PMID: 31804531 PMCID: PMC6895058 DOI: 10.1038/s41598-019-54693-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/18/2019] [Indexed: 12/27/2022] Open
Abstract
A follicular spike in cyclic AMP (cAMP) and its subsequent degradation to AMP promotes oocyte maturation and ovulation. In vitro matured (IVM) oocytes do not receive the cAMP increase that occurs in vivo, and artificial elevation of cAMP in IVM cumulus-oocyte complexes improves oocyte developmental potential. This study examined whether mouse oocytes can use the cAMP degradation product AMP to generate ATP via the adenosine salvage pathway, and examined whether pharmacological elevation of cAMP in IVM cumulus-oocyte complexes alters ATP levels. Oocytes cultured with isotopic 13C5-AMP dose-dependently produced 13C5-ATP, however total cellular ATP remained constant. Pharmacological elevation of cAMP using forskolin and IBMX prior to IVM decreased oocyte ATP and ATP:ADP ratio, and promoted activity of the energy regulator AMPK. Conversely, cumulus cells exhibited higher ATP and no change in AMPK. Culture of oocytes without their cumulus cells or inhibition of their gap-junctional communication yielded lower oocyte 13C5-ATP, indicating that cumulus cells facilitate ATP production via the adenosine salvage pathway. In conclusion, this study demonstrates that mouse oocytes can generate ATP from AMP via the adenosine salvage pathway, and cAMP elevation alters adenine nucleotide metabolism and may provide AMP for energy production via the adenosine salvage pathway during the energetically demanding process of meiotic maturation.
Collapse
|
45
|
Qu YY, Zhao R, Zhang HL, Zhou Q, Xu FJ, Zhang X, Xu WH, Shao N, Zhou SX, Dai B, Zhu Y, Shi GH, Shen YJ, Zhu YP, Han CT, Chang K, Lin Y, Zang WD, Xu W, Ye DW, Zhao SM, Zhao JY. Inactivation of the AMPK-GATA3-ECHS1 Pathway Induces Fatty Acid Synthesis That Promotes Clear Cell Renal Cell Carcinoma Growth. Cancer Res 2019; 80:319-333. [PMID: 31690668 DOI: 10.1158/0008-5472.can-19-1023] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/28/2019] [Accepted: 11/01/2019] [Indexed: 02/05/2023]
Abstract
The tumorigenic role and underlying mechanisms of lipid accumulation, commonly observed in many cancers, remain insufficiently understood. In this study, we identified an AMP-activated protein kinase (AMPK)-GATA-binding protein 3 (GATA3)-enoyl-CoA hydratase short-chain 1 (ECHS1) pathway that induces lipid accumulation and promotes cell proliferation in clear cell renal cell carcinoma (ccRCC). Decreased expression of ECHS1, which is responsible for inactivation of fatty acid (FA) oxidation and activation of de novo FA synthesis, positively associated with ccRCC progression and predicted poor patient survival. Mechanistically, ECHS1 downregulation induced FA and branched-chain amino acid (BCAA) accumulation, which inhibited AMPK-promoted expression of GATA3, a transcriptional activator of ECHS1. BCAA accumulation induced activation of mTORC1 and de novo FA synthesis, and promoted cell proliferation. Furthermore, GATA3 expression phenocopied ECHS1 in predicting ccRCC progression and patient survival. The AMPK-GATA3-ECHS1 pathway may offer new therapeutic approaches and prognostic assessment for ccRCC in the clinic. SIGNIFICANCE: These findings uncover molecular mechanisms underlying lipid accumulation in ccRCC, suggesting the AMPK-GATA3-ECHS1 pathway as a potential therapeutic target and prognostic biomarker.
Collapse
Affiliation(s)
- Yuan-Yuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Rui Zhao
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Hai-Liang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Qian Zhou
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Fu-Jiang Xu
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Xuan Zhang
- Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, P.R. China
| | - Wen-Hao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ning Shao
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Shu-Xian Zhou
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, P.R. China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Guo-Hai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yi-Jun Shen
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yi-Ping Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Cheng-Tao Han
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Kun Chang
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yan Lin
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, P.R. China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Wei-Dong Zang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P.R. China
| | - Wei Xu
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China.,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, P.R. China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Ding-Wei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Shi-Min Zhao
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China. .,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, P.R. China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jian-Yuan Zhao
- Department of Urology, Fudan University Shanghai Cancer Center, the Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, P.R. China. .,Key Laboratory of Reproduction Regulation of NPFPC, Institutes of Biomedical Sciences and Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, P.R. China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
46
|
Madhavi Y, Gaikwad N, Yerra VG, Kalvala AK, Nanduri S, Kumar A. Targeting AMPK in Diabetes and Diabetic Complications: Energy Homeostasis, Autophagy and Mitochondrial Health. Curr Med Chem 2019; 26:5207-5229. [DOI: 10.2174/0929867325666180406120051] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/16/2018] [Accepted: 03/27/2018] [Indexed: 02/06/2023]
Abstract
Adenosine 5′-monophosphate activated protein kinase (AMPK) is a key enzymatic protein involved
in linking the energy sensing to the metabolic manipulation. It is a serine/threonine kinase activated
by several upstream kinases. AMPK is a heterotrimeric protein complex regulated by AMP, ADP, and
ATP allosterically. AMPK is ubiquitously expressed in various tissues of the living system such as heart,
kidney, liver, brain and skeletal muscles. Thus malfunctioning of AMPK is expected to harbor several
human pathologies especially diseases associated with metabolic and mitochondrial dysfunction. AMPK
activators including synthetic derivatives and several natural products that have been found to show therapeutic
relief in several animal models of disease. AMP, 5-Aminoimidazole-4-carboxamide riboside (AICA
riboside) and A769662 are important activators of AMPK which have potential therapeutic importance
in diabetes and diabetic complications. AMPK modulation has shown beneficial effects against
diabetes, cardiovascular complications and diabetic neuropathy. The major impact of AMPK modulation
ensures healthy functioning of mitochondria and energy homeostasis in addition to maintaining a strict
check on inflammatory processes, autophagy and apoptosis. Structural studies on AMP and AICAR suggest
that the free amino group is imperative for AMPK stimulation. A769662, a non-nucleoside
thienopyridone compound which resulted from the lead optimization studies on A-592107 and several
other related compound is reported to exhibit a promising effect on diabetes and its complications through
activation of AMPK. Subsequent to the discovery of A769662, several thienopyridones,
hydroxybiphenyls pyrrolopyridones have been reported as AMPK modulators. The review will explore
the structure-function relationships of these analogues and the prospect of targeting AMPK in diabetes
and diabetic complications.
Collapse
Affiliation(s)
- Y.V. Madhavi
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Telangana, India
| | - Nikhil Gaikwad
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Telangana, India
| | - Veera Ganesh Yerra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Telangana, India
| | - Anil Kumar Kalvala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Telangana, India
| | - Srinivas Nanduri
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Telangana, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Telangana, India
| |
Collapse
|
47
|
Lyu Y, Zhang Y, Yang M, Lin L, Yang X, Cheung SCK, Shaw PC, Chan PKS, Kong APS, Zuo Z. Pharmacokinetic interactions between metformin and berberine in rats: Role of oral administration sequences and microbiota. Life Sci 2019; 235:116818. [DOI: 10.1016/j.lfs.2019.116818] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/14/2019] [Accepted: 08/28/2019] [Indexed: 11/15/2022]
|
48
|
Eicosapentaenoic Acid (EPA) Modulates Glucose Metabolism by Targeting AMP-Activated Protein Kinase (AMPK) Pathway. Int J Mol Sci 2019; 20:ijms20194751. [PMID: 31557807 PMCID: PMC6801536 DOI: 10.3390/ijms20194751] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 11/17/2022] Open
Abstract
EPA, an omega-3 polyunsaturated fatty acid, exerts beneficial effects on human health. However, the molecular mechanisms underlying EPA function are poorly understood. The object was to illuminate molecular mechanism underlying EPA's role. Here, 1H-NMR-based metabolic analysis showed enhanced branched-chain amino acids (BCAAs) and lactate following EPA treatment in skeletal muscle cells. EPA regulated mitochondrial oxygen consumption rate. Furthermore, EPA induced calcium/calmodulin-dependent protein kinase kinase (CaMKK) through the generation of intracellular calcium. This induced the phosphorylation of AMP-activated protein kinase (AMPK) and p38 mitogen-activated protein kinase (p38 MAPK) that led to glucose uptake, and the translocation of glucose transporter type 4 (GLUT4) in muscles. In conclusion, EPA exerts benign effects on glucose through the activation of AMPK-p38 MAPK signaling pathways in skeletal muscles.
Collapse
|
49
|
Tang Z, Ye W, Chen H, Kuang X, Guo J, Xiang M, Peng C, Chen X, Liu H. Role of purines in regulation of metabolic reprogramming. Purinergic Signal 2019; 15:423-438. [PMID: 31493132 DOI: 10.1007/s11302-019-09676-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
Purines, among most influential molecules, are reported to have essential biological function by regulating various cell types. A large number of studies have led to the discovery of many biological functions of the purine nucleotides such as ATP, ADP, and adenosine, as signaling molecules that engage G protein-coupled or ligand-gated ion channel receptors. The role of purines in the regulation of cellular functions at the gene or protein level has been well documented. With the advances in multiomics, including those from metabolomic and bioinformatic analyses, metabolic reprogramming was identified as a key mechanism involved in the regulation of cellular function under physiological or pathological conditions. Recent studies suggest that purines or purine-derived products contribute to important regulatory functions in many fundamental biological and pathological processes related to metabolic reprogramming. Therefore, this review summarizes the role and potential mechanism of purines in the regulation of metabolic reprogramming. In particular, the molecular mechanisms of extracellular purine- and intracellular purine-mediated metabolic regulation in various cells during disease development are discussed. In summary, our review provides an extensive resource for studying the regulatory role of purines in metabolic reprogramming and sheds light on the utilization of the corresponding peptides or proteins for disease diagnosis and therapy.
Collapse
Affiliation(s)
- Zhenwei Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Wenrui Ye
- Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Haotian Chen
- Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Xinwei Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minmin Xiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
50
|
J B, Das A. An edible fungi Pleurotus ostreatus inhibits adipogenesis via suppressing expression of PPAR γ and C/EBP α in 3T3-L1 cells: In vitro validation of gene knock out of RNAs in PPAR γ using CRISPR spcas9. Biomed Pharmacother 2019; 116:109030. [PMID: 31152927 DOI: 10.1016/j.biopha.2019.109030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE Obesity is now well recognized as a disorder, one that is essentially preventable through changes in lifestyle. Obesity is also a main concern associated with expanded morbidity and mortality from many noncommunicable illnesses (NCDs). The study aimed to determine the antiobesity effect of Pleurotus ostreatus (PO) and its bioactive anthraquinone (AQ). The overall promoter genes CEBPα (CCAAT enhancer binding protein α) and PPARγ (Peroxisome proliferator activated receptor γ) in controlling the homeostasis of glucose was analysed using 3T3-L1 cell line. Finally, an insilico study was carried out using CRISPR software to identify the RNA's involved in adipogenesis especially of the control gene PPARγ. MATERIALS AND METHODS Preliminary screening of the edible fungi and their bio actives led to the marvellous discovery of side effect free agonists for treating obesity (adipogenesis). An edible fungi Pleurotus ostreatus (PO) were analysed in a screening platform with different series of tests for adipocyte differentiation, triglyceride analysis, lipolysis determination, glucose uptake assay, cytotoxicity assay and lipase activity followed by specific gene expression analysis. The gene knockout mechanism was also elucidated by CRISPR spcas 9 tool. RESULTS The antiadipogenic (antiobesity) activity of DMSO extract of PO were found to stimulate the insulin dependent uptake of glucose. The extract also decreased the levels of triglycerides and glycerol accumulation in differentiated adipocyte cells. The binding FABP4 (Fatty acid binding protein) and transport protein FATP1 (Fatty acid transport protein) along with the fat breaking LPL (lipoprotein lipase) was found to be inhibited after the PO treatment at varying concentration (0-300 μg/ml). CRISPR spcas9 genome editing software was used as an insilico approach in validating the efficiency of mouse embryonic and human adipogenic cell line (3T3-L1). These tool analysed and found 4 RNAs gene knock out possibilities in PPARγ and their efficiency for further treating obesity. CONCLUSION These novel finding contribute to the confirmation that edible fungi PO and it's bioactive AQ is an adequate supplement for constraining the lipid and triglycerides in differentiated mature adipocytes by reversing the fat deposition. Thereby, forbidding the enzymes linked with fat absorption. Besides, the CRISPR tool identified gene knock out possibilities of control gene PPARγ, will pave a way in further research for treating obesity.
Collapse
Affiliation(s)
- Bindhu J
- Molecular Diagnostics and Bacterial Pathogenomics Research Laboratory, Department of Biotechnology, Bannari Amman Institute of Technology, Sathyamangalam, 638401, India
| | - Arunava Das
- Molecular Diagnostics and Bacterial Pathogenomics Research Laboratory, Department of Biotechnology, Bannari Amman Institute of Technology, Sathyamangalam, 638401, India.
| |
Collapse
|