101
|
Donnell RA, Carré JE, Affourtit C. Acute bioenergetic insulin sensitivity of skeletal muscle cells: ATP-demand-provoked glycolysis contributes to stimulation of ATP supply. Biochem Biophys Rep 2022; 30:101274. [PMID: 35592612 PMCID: PMC9112030 DOI: 10.1016/j.bbrep.2022.101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/04/2022] [Indexed: 11/08/2022] Open
Abstract
Skeletal muscle takes up glucose in an insulin-sensitive manner and is thus important for the maintenance of blood glucose homeostasis. Insulin resistance during development of type 2 diabetes is associated with decreased ATP synthesis, but the causality of this association is controversial. In this paper, we report real-time oxygen uptake and medium acidification data that we use to quantify acute insulin effects on intracellular ATP supply and ATP demand in rat and human skeletal muscle cells. We demonstrate that insulin increases overall cellular ATP supply by stimulating the rate of glycolytic ATP synthesis. Stimulation is immediate and achieved directly by increased glycolytic capacity, and indirectly by elevated ATP demand from protein synthesis. Raised glycolytic capacity does not result from augmented glucose uptake. Notably, insulin-sensitive glucose uptake is increased synergistically by nitrite. While nitrite has a similar stimulatory effect on glycolytic ATP supply as insulin, it does not amplify insulin stimulation. These data highlight the multifarious nature of acute bioenergetic insulin sensitivity of skeletal muscle cells, and are thus important for the interpretation of changes in energy metabolism that are seen in insulin-resistant muscle. Insulin acutely stimulates glycolytic ATP supply in cultured skeletal muscle cells. Insulin affects muscle glycolysis directly and indirectly by increasing ATP demand. Nitrite synergistically increases insulin-sensitive glucose uptake by muscle cells.
Collapse
|
102
|
Hydroxycitric Acid Inhibits Chronic Myelogenous Leukemia Growth through Activation of AMPK and mTOR Pathway. Nutrients 2022; 14:nu14132669. [PMID: 35807850 PMCID: PMC9268148 DOI: 10.3390/nu14132669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic regulation of cancer cell growth via AMP-activated protein kinase (AMPK) activation is a widely studied strategy for cancer treatment, including leukemias. Recent notions that naturally occurring compounds might have AMPK activity led to the search for nutraceuticals with potential AMPK-stimulating activity. We found that hydroxycitric acid (HCA), a natural, safe bioactive from the plant Garcinia gummi-gutta (cambogia), has potent AMPK activity in chronic myelogenous leukemia (CML) cell line K562. HCA is a known competitive inhibitor of ATP citrate lyase (ACLY) and is widely used as a weight loss inducer. We found that HCA was able to inhibit the growth of K562 cells in in vitro and in vivo xenograft models. At the mechanistic level, we identified a direct interaction between AMPK and ACLY that seems to be sensitive to HCA treatment. Additionally, HCA treatment resulted in the co-activation of AMPK and the mammalian target of rapamycin (mTOR) pathways. Moreover, we found an enhanced unfolded protein response as observed by activation of the eIF2α/ATF4 pathway that could explain the induction of cell cycle arrest at the G2/M phase and DNA fragmentation upon HCA treatment in K562 cells. Overall, these findings suggest HCA as a nutraceutical approach for the treatment of CMLs.
Collapse
|
103
|
Dizaji R, Sharafi A, Pourahmad J, Vatanpour S, Dinmohammadi H, Vatanpour H, Hosseini MJ. Correlation between coenzyme Q 10 content and the nutrient sensors in AKI induced by Hemiscorpius lepturus envenomation. BIOIMPACTS : BI 2022; 12:431-438. [PMID: 36381638 PMCID: PMC9596883 DOI: 10.34172/bi.2022.23422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/21/2021] [Accepted: 04/03/2021] [Indexed: 06/16/2023]
Abstract
Introduction: Acute kidney injury (AKI) may have a negative effect on mitochondrial hemostasis and bioenergetics as well as coenzyme Q10 (CoQ10) content. PGC-1α, AMPK, sirtuin 1 (Sirt1), and Sirt3, as the key metabolic regulators under nutritional stress, stimulate energy production via mitochondrial biogenesis during AKI. However, no report is available on the relationship between CoQ10 level and nutrient sensors in the pathophysiology of AKI caused by Hemiscorpius lepturus scorpion envenomation. Methods: Three doses of venoms (1, 5, and 10 mg/kg) were administered by subcutaneous (SC) injection to male albino mice. The animals were sacrificed 1 day or 7 days after administration of venom and their kidneys were collected to analyze gene expression involved in AKI, nutrient sensors, and apoptosis signaling activation by real-time polymerase chain reaction (PCR) and the measurement of CoQ10 level using the High-performance liquid chromatography (HPLC) method. Results: The data indicated a significant decrease in CoQ10 level after the administration of venom in 5 and 10 mg/kg. In addition, 1 day after the treatment, a significant over-expression of Sirt1 (5 and 10 mg/kg) was observed compared with normal mice. Overexpression of Sirt3 occurred 1 day and 7 days after treatment only at the dose of 5.0 mg/kg of venom. Furthermore, over-expression of AMPK as an important mitochondrial energetic sensor happened 1 day and 7 days after the injection of venom (5 mg/kg) (P < 0.01). The significant increase in the gene expression of caspase-9 and 3 after the injection of venom (5 and 10 mg/kg) confirmed the role of cell death signaling. Conclusion: The venom-induced energy-sensing pathways have a key role in gene expression of PGC-1α, AMPK, Sirt3, and CoQ10 content after venom-induced AKI.
Collapse
Affiliation(s)
- Rana Dizaji
- Department of Food Safety and Hygiene, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran
| | - Ali Sharafi
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jalal Pourahmad
- Departments of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saba Vatanpour
- Department of Biology, University of British Columbia, Vancouver, Canada
| | - Hossein Dinmohammadi
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Vatanpour
- Departments of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
104
|
Metabolic reprograming of MDSCs within tumor microenvironment and targeting for cancer immunotherapy. Acta Pharmacol Sin 2022; 43:1337-1348. [PMID: 34561553 PMCID: PMC9160034 DOI: 10.1038/s41401-021-00776-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
A number of emerging studies in field of immune metabolism have indicated that cellular metabolic reprograming serves as a major administrator in maintaining the viability and functions of both tumor cells and immune cells. As one of the most important immunosuppressive cells in tumor stroma, myeloid-derived suppressor cells (MDSCs) dynamically orchestrate their metabolic pathways in response to the complicated tumor microenvironment (TME), a process that consequently limits the therapeutic effectiveness of anti-cancer treatment modalities. In this context, the metabolic vulnerabilities of MDSCs could be exploited as a novel immune metabolic checkpoint upon which to intervene for promoting the efficacy of immunotherapy. Here, we have discussed about recent studies highlighting the important roles of the metabolic reprograming and the core molecular pathways involved in tumor-infiltrating MDSCs. In addition, we have also summarized the state-of-the-art strategies that are currently being employed to target MDSC metabolism and improve the efficacy of antineoplastic immunotherapy.
Collapse
|
105
|
Chen J, Zou L, Lu G, Grinchuk O, Fang L, Ong DST, Taneja R, Ong CN, Shen HM. PFKP alleviates glucose starvation-induced metabolic stress in lung cancer cells via AMPK-ACC2 dependent fatty acid oxidation. Cell Discov 2022; 8:52. [PMID: 35641476 PMCID: PMC9156709 DOI: 10.1038/s41421-022-00406-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/10/2022] [Indexed: 11/08/2022] Open
Abstract
Cancer cells adopt metabolic reprogramming to promote cell survival under metabolic stress. A key regulator of cell metabolism is AMP-activated protein kinase (AMPK) which promotes catabolism while suppresses anabolism. However, the underlying mechanism of AMPK in handling metabolic stress in cancer remains to be fully understood. In this study, by performing a proteomics screening of AMPK-interacting proteins in non-small-cell lung cancer (NSCLC) cells, we discovered the platelet isoform of phosphofructokinase 1 (PFKP), a rate-limiting enzyme in glycolysis. Moreover, PFKP was found to be highly expressed in NSCLC patients associated with poor survival. We demonstrated that the interaction of PFKP and AMPK was greatly enhanced upon glucose starvation, a process regulated by PFKP-associated metabolites. Notably, the PFKP-AMPK interaction promoted mitochondrial recruitment of AMPK which subsequently phosphorylated acetyl-CoA carboxylase 2 (ACC2) to enhance long-chain fatty acid oxidation, a process helping maintenance of the energy and redox homeostasis and eventually promoting cancer cell survival under glucose starvation. Collectively, we revealed a critical non-glycolysis-related function of PFKP in regulating long-chain fatty acid oxidation via AMPK to alleviate glucose starvation-induced metabolic stress in NSCLC cells.
Collapse
Affiliation(s)
- Jiaqing Chen
- NUS Graduate School Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Zou
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Guang Lu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Oleg Grinchuk
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lei Fang
- Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Derrick Sek Tong Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reshma Taneja
- NUS Graduate School Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Choon-Nam Ong
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Han-Ming Shen
- NUS Graduate School Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China.
| |
Collapse
|
106
|
Aiyasiding X, Liao HH, Feng H, Zhang N, Lin Z, Ding W, Yan H, Zhou ZY, Tang QZ. Liquiritin Attenuates Pathological Cardiac Hypertrophy by Activating the PKA/LKB1/AMPK Pathway. Front Pharmacol 2022; 13:870699. [PMID: 35592411 PMCID: PMC9110825 DOI: 10.3389/fphar.2022.870699] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/05/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Liquiritin (LQ) is one of the main flavonoids extracted from the roots of Glycyrrhiza spp., which are widely used in traditional Chinese medicine. Studies in both cellular and animal disease models have shown that LQ attenuates or prevents oxidative stress, inflammation, and apoptosis. However, the potential therapeutic effects of LQ on pressure overload-induced cardiac hypertrophy have not been so far explored. Therefore, we investigated the cardioprotective role of LQ and its underlying mechanisms in the aortic banding (AB)-induced cardiac hypertrophy mouse model. Methods and Results: Starting 3 days after AB surgery, LQ (80 mg/kg/day) was administered daily over 4 weeks. Echocardiography and pressure-volume loop analysis indicated that LQ treatment markedly improved hypertrophy-related cardiac dysfunction. Moreover, hematoxylin and eosin, picrosirius red, and TUNEL staining showed that LQ significantly inhibited cardiomyocyte hypertrophy, interstitial fibrosis, and apoptosis. Western blot assays further showed that LQ activated LKB1/AMPKα2/ACC signaling and inhibited mTORC1 phosphorylation in cardiomyocytes. Notably, LQ treatment failed to prevent cardiac dysfunction, hypertrophy, and fibrosis in AMPKα2 knockout (AMPKα2−/−) mice. However, LQ still induced LKB1 phosphorylation in AMPKα2−/− mouse hearts. In vitro experiments further demonstrated that LQ inhibited Ang II-induced hypertrophy in neonatal rat cardiomyocytes (NRCMs) by increasing cAMP levels and PKA activity. Supporting the central involvement of the cAMP/PKA/LKB1/AMPKα2 signaling pathway in the cardioprotective effects of LQ, inhibition of Ang II-induced hypertrophy and induction of LKB1 and AMPKα phosphorylation were no longer observed after inhibiting PKA activity. Conclusion: This study revealed that LQ alleviates pressure overload-induced cardiac hypertrophy in vivo and inhibits Ang II-induced cardiomyocyte hypertrophy in vitro via activating cAMP/PKA/LKB1/AMPKα2 signaling. These findings suggest that LQ might be a valuable adjunct to therapeutic approaches for treating pathological cardiac remodeling.
Collapse
Affiliation(s)
- Xiahenazi Aiyasiding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Hai-Han Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Hong Feng
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Zheng Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Han Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Zi-Ying Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| |
Collapse
|
107
|
Cheratta AR, Thayyullathil F, Hawley SA, Ross FA, Atrih A, Lamont DJ, Pallichankandy S, Subburayan K, Alakkal A, Rezgui R, Gray A, Hardie DG, Galadari S. Caspase cleavage and nuclear retention of the energy sensor AMPK-α1 during apoptosis. Cell Rep 2022; 39:110761. [PMID: 35508122 PMCID: PMC9108549 DOI: 10.1016/j.celrep.2022.110761] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/11/2022] [Accepted: 04/07/2022] [Indexed: 02/09/2023] Open
Abstract
AMP-activated protein kinase (AMPK) coordinates energy homeostasis during metabolic and energy stress. We report that the catalytic subunit isoform AMPK-α1 (but not α2) is cleaved by caspase-3 at an early stage during induction of apoptosis. AMPK-α1 cleavage occurs following Asp529, generating an ∼58-kDa N-terminal fragment (cl-AMPK-α1) and leading to the precise excision of the nuclear export sequence (NES) from the C-terminal end. This cleavage does not affect (1) the stability of pre-formed heterotrimeric complexes, (2) the ability of cl-AMPK-α1 to become phosphorylated and activated by the upstream kinases LKB1 or CaMKK2, or (3) allosteric activation by AMP or A-769662. Importantly, cl-AMPK-α1 is only detectable in the nucleus, consistent with removal of the NES, and ectopic expression of cleavage-resistant D529A-mutant AMPK-α1 promotes cell death induced by cytotoxic agents. Thus, we have elucidated a non-canonical mechanism of AMPK activation within the nucleus, which protects cells against death induced by DNA damage.
Collapse
Affiliation(s)
- Anees Rahman Cheratta
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Faisal Thayyullathil
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Simon A. Hawley
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Fiona A. Ross
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Abdelmajdid Atrih
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Douglas J. Lamont
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Siraj Pallichankandy
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Karthikeyan Subburayan
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Ameer Alakkal
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Rachid Rezgui
- Core Technology Platform, Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Alex Gray
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - D. Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK,Corresponding author
| | - Sehamuddin Galadari
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE.
| |
Collapse
|
108
|
Li J, Bollati C, Bartolomei M, Mazzolari A, Arnoldi A, Vistoli G, Lammi C. Hempseed ( Cannabis sativa) Peptide H3 (IGFLIIWV) Exerts Cholesterol-Lowering Effects in Human Hepatic Cell Line. Nutrients 2022; 14:1804. [PMID: 35565772 PMCID: PMC9101684 DOI: 10.3390/nu14091804] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 02/04/2023] Open
Abstract
Hempseed (Cannabis sativa) protein is an important source of bioactive peptides. H3 (IGFLIIWV), a transepithelial transported intestinal peptide obtained from the hydrolysis of hempseed protein with pepsin, carries out antioxidant and anti-inflammatory activities in HepG2 cells. In this study, the main aim was to assess its hypocholesterolemic effects at a cellular level and the mechanisms behind this health-promoting activity. The results showed that peptide H3 inhibited the 3-hydroxy-3-methylglutaryl co-enzyme A reductase (HMGCoAR) activity in vitro in a dose-dependent manner with an IC50 value of 59 μM. Furthermore, the activation of the sterol regulatory element binding proteins (SREBP)-2 transcription factor, followed by the increase of low-density lipoprotein (LDL) receptor (LDLR) protein levels, was observed in human hepatic HepG2 cells treated with peptide H3 at 25 µM. Meanwhile, peptide H3 regulated the intracellular HMGCoAR activity through the increase of its phosphorylation by the activation of AMP-activated protein kinase (AMPK)-pathways. Consequently, the augmentation of the LDLR localized on the cellular membranes led to the improved ability of HepG2 cells to uptake extracellular LDL with a positive effect on cholesterol levels. Unlike the complete hempseed hydrolysate (HP), peptide H3 can reduce the proprotein convertase subtilisin/kexin 9 (PCSK9) protein levels and its secretion in the extracellular environment via the decrease of hepatic nuclear factor 1-α (HNF1-α). Considering all these evidences, H3 may represent a new bioactive peptide to be used for the development of dietary supplements and/or peptidomimetics for cardiovascular disease (CVD) prevention.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Carmen Lammi
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy; (J.L.); (C.B.); (M.B.); (A.M.); (A.A.); (G.V.)
| |
Collapse
|
109
|
Zhang C, Lin Y, Yan CH, Zhang W. Adipokine Signaling Pathways in Osteoarthritis. Front Bioeng Biotechnol 2022; 10:865370. [PMID: 35519618 PMCID: PMC9062110 DOI: 10.3389/fbioe.2022.865370] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disease that affects millions of individuals. The pathogenesis of OA has not been fully elucidated. Obesity is a well-recognized risk factor for OA. Multiple studies have demonstrated adipokines play a key role in obesity-induced OA. Increasing evidence show that various adipokines may significantly affect the development or clinical course of OA by regulating the pro/anti-inflammatory and anabolic/catabolic balance, matrix remodeling, chondrocyte apoptosis and autophagy, and subchondral bone sclerosis. Several signaling pathways are involved but still have not been systematically investigated. In this article, we review the cellular and molecular mechanisms of adipokines in OA, and highlight the possible signaling pathways. The review suggested adipokines play important roles in obesity-induced OA, and exert downstream function via the activation of various signaling pathways. In addition, some pharmaceuticals targeting these pathways have been applied into ongoing clinical trials and showed encouraging results. However, these signaling pathways are complex and converge into a common network with each other. In the future work, more research is warranted to further investigate how this network works. Moreover, more high quality randomised controlled trials are needed in order to investigate the therapeutic effects of pharmaceuticals against these pathways for the treatment of OA. This review may help researchers to better understand the pathogenesis of OA, so as to provide new insight for future clinical practices and translational research.
Collapse
Affiliation(s)
- Chaofan Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yunzhi Lin
- Department of Stomatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chun Hoi Yan
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wenming Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
110
|
Amaral AG, Moretto IA, Zandonadi FDS, Zamora-Obando HR, Rocha I, Sussulini A, Thomaz AAD, Oliveira RV, Santos AMD, Simionato AVC. Comprehending Cardiac Dysfunction by Oxidative Stress: Untargeted Metabolomics of In Vitro Samples. Front Chem 2022; 10:836478. [PMID: 35464220 PMCID: PMC9023746 DOI: 10.3389/fchem.2022.836478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/09/2022] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases (CVDs) are noncommunicable diseases known for their complex etiology and high mortality rate. Oxidative stress (OS), a condition in which the release of free radical exceeds endogenous antioxidant capacity, is pivotal in CVC, such as myocardial infarction, ischemia/reperfusion, and heart failure. Due to the lack of information about the implications of OS on cardiovascular conditions, several methodologies have been applied to investigate the causes and consequences, and to find new ways of diagnosis and treatment as well. In the present study, cardiac dysfunction was evaluated by analyzing cells’ alterations with untargeted metabolomics, after simulation of an oxidative stress condition using hydrogen peroxide (H2O2) in H9c2 myocytes. Optimizations of H2O2 concentration, cell exposure, and cell recovery times were performed through MTT assays. Intracellular metabolites were analyzed right after the oxidative stress (oxidative stress group) and after 48 h of cell recovery (recovery group) by ultra-high-performance liquid chromatography coupled to mass spectrometry (UHPLC-MS) in positive and negative ESI ionization mode. Significant alterations were found in pathways such as “alanine, aspartate and glutamate metabolism”, “glycolysis”, and “glutathione metabolism”, mostly with increased metabolites (upregulated). Furthermore, our results indicated that the LC-MS method is effective for studying metabolism in cardiomyocytes and generated excellent fit (R2Y > 0.987) and predictability (Q2 > 0.84) values.
Collapse
|
111
|
Garg C, khan H, Kaur A, Singh TG, Sharma VK, Singh SK. Therapeutic Implications of Sonic Hedgehog Pathway in Metabolic Disorders: Novel Target for Effective Treatment. Pharmacol Res 2022; 179:106194. [DOI: 10.1016/j.phrs.2022.106194] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022]
|
112
|
Loss of LKB1-NUAK1 signalling enhances NF-κB activity in a spheroid model of high-grade serous ovarian cancer. Sci Rep 2022; 12:3011. [PMID: 35194062 PMCID: PMC8863794 DOI: 10.1038/s41598-022-06796-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/01/2022] [Indexed: 01/31/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is an aggressive malignancy often diagnosed at an advanced stage. Although most HGSOC patients respond initially to debulking surgery combined with cytotoxic chemotherapy, many ultimately relapse with platinum-resistant disease. Thus, improving outcomes requires new ways of limiting metastasis and eradicating residual disease. We identified previously that Liver kinase B1 (LKB1) and its substrate NUAK1 are implicated in EOC spheroid cell viability and are required for efficient metastasis in orthotopic mouse models. Here, we sought to identify additional signalling pathways altered in EOC cells due to LKB1 or NUAK1 loss-of-function. Transcriptome analysis revealed that inflammatory signalling mediated by NF-κB transcription factors is hyperactive due to LKB1-NUAK1 loss in HGSOC cells and spheroids. Upregulated NF-κB signalling due to NUAK1 loss suppresses reactive oxygen species (ROS) production and sustains cell survival in spheroids. NF-κB signalling is also activated in HGSOC precursor fallopian tube secretory epithelial cell spheroids, and is further enhanced by NUAK1 loss. Finally, immunohistochemical analysis of OVCAR8 xenograft tumors lacking NUAK1 displayed increased RelB expression and nuclear staining. Our results support the idea that NUAK1 and NF-κB signalling pathways together regulate ROS and inflammatory signalling, supporting cell survival during each step of HGSOC pathogenesis. We propose that their combined inhibition may be efficacious as a novel therapeutic strategy for advanced HGSOC.
Collapse
|
113
|
Boone-Villa D, Ventura-Sobrevilla J, Aguilera-Méndez A, Jiménez-Villarreal J. The effect of adenosine monophosphate-activated protein kinase on lipolysis in adipose tissue: an historical and comprehensive review. Arch Physiol Biochem 2022; 128:7-23. [PMID: 35143739 DOI: 10.1080/13813455.2019.1661495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
CONTEXT Lipolysis is one of the most important pathways for energy management, its control in the adipose tissue (AT) is a potential therapeutic target for metabolic diseases. Adenosine Mono Phosphate-activated Protein Kinase (AMPK) is a key regulatory enzyme in lipids metabolism and a potential target for diabetes and obesity treatment. OBJECTIVE The aim of this work is to analyse the existing information on the relationship of AMPK and lipolysis in the AT. METHODS A thorough search of bibliography was performed in the databases Scopus and Web of Knowledge using the terms lipolysis, adipose tissue, and AMPK, the unrelated publications were excluded, and the documents were analysed. RESULTS Sixty-three works were found and classified in 3 categories: inhibitory effects, stimulatory effect, and diverse relationships; remarkably, the newest researches support an upregulating relationship of AMPK over lipolysis. CONCLUSION The most probable reality is that the relationship AMPK-lipolysis depends on the experimental conditions.
Collapse
Affiliation(s)
- Daniel Boone-Villa
- School of Medicine Northern Unit, Universidad Autonoma de Coahuila, Piedras Negras, México
| | | | - Asdrúbal Aguilera-Méndez
- Institute of Biological Chemistry Research, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, México
| | | |
Collapse
|
114
|
Heidary Moghaddam R, Samimi Z, Asgary S, Mohammadi P, Hozeifi S, Hoseinzadeh-Chahkandak F, Xu S, Farzaei MH. Natural AMPK Activators in Cardiovascular Disease Prevention. Front Pharmacol 2022; 12:738420. [PMID: 35046800 PMCID: PMC8762275 DOI: 10.3389/fphar.2021.738420] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases (CVD), as a life-threatening global disease, is receiving worldwide attention. Seeking novel therapeutic strategies and agents is of utmost importance to curb CVD. AMP-activated protein kinase (AMPK) activators derived from natural products are promising agents for cardiovascular drug development owning to regulatory effects on physiological processes and diverse cardiometabolic disorders. In the past decade, different therapeutic agents from natural products and herbal medicines have been explored as good templates of AMPK activators. Hereby, we overviewed the role of AMPK signaling in the cardiovascular system, as well as evidence implicating AMPK activators as potential therapeutic tools. In the present review, efforts have been made to compile and update relevant information from both preclinical and clinical studies, which investigated the role of natural products as AMPK activators in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Reza Heidary Moghaddam
- Clinical Research Development Center, Imam Ali and Taleghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Samimi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sedigheh Asgary
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute,.Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soroush Hozeifi
- School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Suowen Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
115
|
Plaza-Diaz J, Izquierdo D, Torres-Martos Á, Baig AT, Aguilera CM, Ruiz-Ojeda FJ. Impact of Physical Activity and Exercise on the Epigenome in Skeletal Muscle and Effects on Systemic Metabolism. Biomedicines 2022; 10:126. [PMID: 35052805 PMCID: PMC8773693 DOI: 10.3390/biomedicines10010126] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 02/05/2023] Open
Abstract
Exercise and physical activity induces physiological responses in organisms, and adaptations in skeletal muscle, which is beneficial for maintaining health and preventing and/or treating most chronic diseases. These adaptations are mainly instigated by transcriptional responses that ensue in reaction to each individual exercise, either resistance or endurance. Consequently, changes in key metabolic, regulatory, and myogenic genes in skeletal muscle occur as both an early and late response to exercise, and these epigenetic modifications, which are influenced by environmental and genetic factors, trigger those alterations in the transcriptional responses. DNA methylation and histone modifications are the most significant epigenetic changes described in gene transcription, linked to the skeletal muscle transcriptional response to exercise, and mediating the exercise adaptations. Nevertheless, other alterations in the epigenetics markers, such as epitranscriptomics, modifications mediated by miRNAs, and lactylation as a novel epigenetic modification, are emerging as key events for gene transcription. Here, we provide an overview and update of the impact of exercise on epigenetic modifications, including the well-described DNA methylations and histone modifications, and the emerging modifications in the skeletal muscle. In addition, we describe the effects of exercise on epigenetic markers in other metabolic tissues; also, we provide information about how systemic metabolism or its metabolites influence epigenetic modifications in the skeletal muscle.
Collapse
Affiliation(s)
- Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain; (D.I.); (C.M.A.)
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain;
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada;
| | - David Izquierdo
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain; (D.I.); (C.M.A.)
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain;
| | - Álvaro Torres-Martos
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain;
| | - Aiman Tariq Baig
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada;
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 85M, Canada
| | - Concepción M. Aguilera
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain; (D.I.); (C.M.A.)
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain;
- Center of Biomedical Research, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Avda. del Conocimiento s/n., 18016 Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain; (D.I.); (C.M.A.)
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain;
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz, Center Munich, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
116
|
Singh SSB, Patil KN. Trans-ferulic acid attenuates hyperglycemia-induced oxidative stress and modulates glucose metabolism by activating AMPK signaling pathway in vitro. J Food Biochem 2022; 46:e14038. [PMID: 34981525 DOI: 10.1111/jfbc.14038] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a potent metabolic regulator and an attractive target for antidiabetic activators. Here we report for the first that, trans-ferulic acid (TFA) is a potent dietary bioactive molecule of hydroxycinnamic acid derivative for the activation of AMPK with a maximum increase in phosphorylation (2.71/2.67 ± 0.10; p < .001 vs. high glucose [HG] control) in hyperglycemia-induced human liver cells (HepG2) and rat skeletal muscle cells (L6), where HG suppresses the AMPK pathway. It was also observed that TFA increased activation of AMPK in a dose- and time-dependent manner and also increased the phosphorylation of acetyl-CoA carboxylase (ACC), suggesting that it may promotes fatty acid oxidation; however, pretreatment with compound C reversed the effect. In addition, TFA reduced the level of intracellular reactive oxygen species (ROS) and nitric oxide (NO) induced by hyperglycemia and subsequently increased the level of glutathione. Interestingly, TFA also upregulated the glucose transporters, GLUT2 and GLUT4, and inhibited c-Jun N-terminal protein kinase (JNK1/2) by decreasing the phosphorylation level in tested cells under HG condition. Our studies provide critical insights into the mechanism of action of TFA as a potential natural activator of AMPK under hyperglycemia. PRACTICAL APPLICATIONS: Hydroxycinnamic acid derivatives possess various pharmacological properties and are found to be one of the most ubiquitous groups of plant metabolites in almost all dietary sources. However, the tissue-specific role and its mechanism under hyperglycemic condition remain largely unknown. The present study showed that TFA is a potent activator of AMPK under HG condition and it could be used as a therapeutic agent against hyperglycemia in type 2 diabetes.
Collapse
Affiliation(s)
- Sangeetha S B Singh
- Department of Protein Chemistry and Technology, Council of Scientific & Industrial Research-Central Food Technological Research Institute (CSIR-CFTRI), Mysore, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - K Neelakanteshwar Patil
- Department of Protein Chemistry and Technology, Council of Scientific & Industrial Research-Central Food Technological Research Institute (CSIR-CFTRI), Mysore, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
117
|
Abstract
Autophagy is an intracellular catabolic degradative process in which damaged cellular organelles, unwanted proteins and different cytoplasmic components get recycled to maintain cellular homeostasis or metabolic balance. During autophagy, a double membrane vesicle is formed to engulf these cytosolic materials and fuse to lysosomes wherein the entire cargo degrades to be used again. Because of this unique recycling ability of cells, autophagy is a universal stress response mechanism. Dysregulation of autophagy leads to several diseases, including cancer, neurodegeneration and microbial infection. Thus, autophagy machineries have become targets for therapeutics. This chapter provides an overview of the paradoxical role of autophagy in tumorigenesis in the perspective of metabolism.
Collapse
Affiliation(s)
- Sweta Sikder
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
118
|
Wang Z, Shen J, Feng E, Jiao Y. AMPK as a Potential Therapeutic Target for Intervertebral Disc Degeneration. Front Mol Biosci 2021; 8:789087. [PMID: 34957218 PMCID: PMC8692877 DOI: 10.3389/fmolb.2021.789087] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022] Open
Abstract
As the principal reason for low back pain, intervertebral disc degeneration (IDD) affects the health of people around the world regardless of race or region. Degenerative discs display a series of characteristic pathological changes, including cell apoptosis, senescence, remodeling of extracellular matrix, oxidative stress and inflammatory local microenvironment. As a serine/threonine-protein kinase in eukaryocytes, AMP-activated protein kinase (AMPK) is involved in various cellular processes through the modulation of cell metabolism and energy balance. Recent studies have shown the abnormal activity of AMPK in degenerative disc cells. Besides, AMPK regulates multiple crucial biological behaviors in IDD. In this review, we summarize the pathophysiologic changes of IDD and activation process of AMPK. We also attempt to generalize the role of AMPK in the pathogenesis of IDD. Moreover, therapies targeting AMPK in alleviating IDD are analyzed, for better insight into the potential of AMPK as a therapeutic target.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Orthopedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jianxiong Shen
- Department of Orthopedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Erwei Feng
- Department of Orthopedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Jiao
- Department of Orthopedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
119
|
Yoshida J, Ohishi T, Abe H, Ohba SI, Inoue H, Usami I, Amemiya M, Oriez R, Sakashita C, Dan S, Sugawara M, Kawaguchi T, Ueno J, Asano Y, Ikeda A, Takamatsu M, Amori G, Kondoh Y, Honda K, Osada H, Noda T, Watanabe T, Shimizu T, Shibasaki M, Kawada M. Mitochondrial complex I inhibitors suppress tumor growth through concomitant acidification of the intra- and extracellular environment. iScience 2021; 24:103497. [PMID: 34934919 DOI: 10.1016/j.isci.2021.103497] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/25/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
The disruption of the tumor microenvironment (TME) is a promising anti-cancer strategy, but its effective targeting for solid tumors remains unknown. Here, we investigated the anti-cancer activity of the mitochondrial complex I inhibitor intervenolin (ITV), which modulates the TME independent of energy depletion. By modulating lactate metabolism, ITV induced the concomitant acidification of the intra- and extracellular environment, which synergistically suppressed S6K1 activity in cancer cells through protein phosphatase-2A-mediated dephosphorylation via G-protein-coupled receptor(s). Other complex I inhibitors including metformin and rotenone were also found to exert the same effect through an energy depletion-independent manner as ITV. In mouse and patient-derived xenograft models, ITV was found to suppress tumor growth and its mode of action was further confirmed. The TME is usually acidic owing to glycolytic cancer cell metabolism, and this condition is more susceptible to complex I inhibitors. Thus, we have demonstrated a potential treatment strategy for solid tumors.
Collapse
Affiliation(s)
- Junjiro Yoshida
- Laboratory of Oncology, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Tomokazu Ohishi
- Numazu Branch and Section of Animal Resources, Institute of Microbial Chemistry (BIKAKEN), Numazu-shi, Shizuoka 410-0301, Japan
| | - Hikaru Abe
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Shun-Ichi Ohba
- Numazu Branch and Section of Animal Resources, Institute of Microbial Chemistry (BIKAKEN), Numazu-shi, Shizuoka 410-0301, Japan
| | - Hiroyuki Inoue
- Numazu Branch and Section of Animal Resources, Institute of Microbial Chemistry (BIKAKEN), Numazu-shi, Shizuoka 410-0301, Japan
| | - Ihomi Usami
- Numazu Branch and Section of Animal Resources, Institute of Microbial Chemistry (BIKAKEN), Numazu-shi, Shizuoka 410-0301, Japan
| | - Masahide Amemiya
- Laboratory of Oncology, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Raphael Oriez
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Chiharu Sakashita
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Minoru Sugawara
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Tokuichi Kawaguchi
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Junko Ueno
- Department of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Yuko Asano
- Department of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Ami Ikeda
- Department of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Manabu Takamatsu
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Gulanbar Amori
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Yasumitsu Kondoh
- Chemical Biology Research Group & Drug Discovery Chemical Bank Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Kaori Honda
- Chemical Biology Research Group & Drug Discovery Chemical Bank Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Hiroyuki Osada
- Chemical Biology Research Group & Drug Discovery Chemical Bank Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Tetsuo Noda
- Director's Room, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Takumi Watanabe
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Takao Shimizu
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Laboratory of Virology, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Masakatsu Shibasaki
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
| | - Manabu Kawada
- Laboratory of Oncology, Institute of Microbial Chemistry (BIKAKEN), Shinagawa-ku, Tokyo 141-0021, Japan
- Numazu Branch and Section of Animal Resources, Institute of Microbial Chemistry (BIKAKEN), Numazu-shi, Shizuoka 410-0301, Japan
| |
Collapse
|
120
|
Liu D, Jin X, Yu G, Wang M, Liu L, Zhang W, Wu J, Wang F, Yang J, Luo Q, Cai L, Yang X, Ke X, Qu Y, Xu Z, Jia L, Chen WL. Oleanolic acid blocks the purine salvage pathway for cancer therapy by inactivating SOD1 and stimulating lysosomal proteolysis. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:107-123. [PMID: 34703880 PMCID: PMC8505360 DOI: 10.1016/j.omto.2021.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/24/2021] [Indexed: 12/28/2022]
Abstract
Metabolic reprogramming is a core hallmark of cancer and is key for tumorigenesis and tumor progression. Investigation of metabolic perturbation by anti-cancer compounds would allow a thorough understanding of the underlying mechanisms of these agents and identification of new anti-cancer targets. Here, we demonstrated that the administration of oleanolic acid (OA) rapidly altered cancer metabolism, particularly suppressing the purine salvage pathway (PSP). PSP restoration significantly opposed OA-induced DNA replication and cell proliferation arrest, underscoring the importance of this pathway for the anti-cancer activity of OA. Hypoxanthine-guanine phosphoribosyltransferase (HGPRT) and 5′-nucleotidase (5′-NT), two metabolic enzymes essential for PSP activity, were promptly degraded by OA via the lysosome pathway. Mechanistically, OA selectively targeted superoxide dismutase 1 (SOD1) and yielded reactive oxygen species (ROS) to activate the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin complex 1 (mTORC1)/macroautophagy pathway, thus eliciting lysosomal degradation of HGPRT and 5′-NT. Furthermore, we found that the PSP was overactivated in human lung and breast cancers, with a negative correlation with patient survival. The results of this study elucidated a new anti-cancer mechanism of OA by restraining the PSP via the SOD1/ROS/AMPK/mTORC1/macroautophagy/lysosomal pathway. We also identified the PSP as a new target for cancer treatment and highlighted OA as a potential therapeutic agent for cancers with high PSP activity.
Collapse
Affiliation(s)
- Dan Liu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xing Jin
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guanzhen Yu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Mingsong Wang
- Department of Thoracic Cardiovascular Surgery, Xinhua Hospital of Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Lei Liu
- Department of Thoracic Surgery, the Affiliated Tumor Hospital of Nantong University, Nantong 226361, China
| | - Wenjuan Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jia Wu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Fengying Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jing Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Qin Luo
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lili Cai
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xi Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xisong Ke
- Center for Chemical Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi Qu
- Center for Chemical Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenye Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wen-Lian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
121
|
Aledavood E, Gheeraert A, Forte A, Vuillon L, Rivalta I, Luque FJ, Estarellas C. Elucidating the Activation Mechanism of AMPK by Direct Pan-Activator PF-739. Front Mol Biosci 2021; 8:760026. [PMID: 34805275 PMCID: PMC8602109 DOI: 10.3389/fmolb.2021.760026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Adenosine monophosphate-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 heterotrimeric complexes, which exhibit notable differences in the sensitivity to direct activators. To comprehend the molecular factors of the activation mechanism of AMPK, we have assessed the changes in the structural and dynamical properties of β1- and β2-containing AMPK complexes formed upon binding to the pan-activator PF-739. The analysis revealed the molecular basis of the PF-739-mediated activation of AMPK and enabled us to identify distinctive features that may justify the slightly higher affinity towards the β1−isoform, such as the β1−Asn111 to β2−Asp111 substitution, which seems to be critical for modulating the dynamical sensitivity of β1- and β2 isoforms. The results are valuable in the design of selective activators to improve the tissue specificity of therapeutic treatment.
Collapse
Affiliation(s)
- Elnaz Aledavood
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Barcelona, Spain
| | - Aria Gheeraert
- Dipartimento di Chimica Industriale "Toso Montanari" Università di Bologna, Bologna, Italy.,LAMA, University of Savoie Mont Blanc, CNRS, LAMA, Le Bourget du Lac, France
| | - Alessia Forte
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Barcelona, Spain
| | - Laurent Vuillon
- LAMA, University of Savoie Mont Blanc, CNRS, LAMA, Le Bourget du Lac, France
| | - Ivan Rivalta
- Dipartimento di Chimica Industriale "Toso Montanari" Università di Bologna, Bologna, Italy.,Université de Lyon, École Normale Supérieure de Lyon, CNRS UMR 5182, Laboratoire de Chimie, Lyon, France
| | - F Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Barcelona, Spain.,Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Carolina Estarellas
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
122
|
Natural and chemical compounds as protective agents against cardiac lipotoxicity. Biomed Pharmacother 2021; 145:112413. [PMID: 34781144 DOI: 10.1016/j.biopha.2021.112413] [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] [Received: 09/21/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022] Open
Abstract
Cardiac lipotoxicity results from the deleterious effects of excess lipid deposition in cardiomyocytes. Lipotoxic cardiomyopathy involves cardiac lipid overload leading to changes in myocardial structure and function. Cardiac dysfunction has been associated with cardiac lipotoxicity through abnormal lipid metabolism. Lipid accumulation, especially saturated free fatty acids (SFFAs), in cardiac cells can cause cardiomyocyte distress and subsequent myocardial contractile dysfunction. Reducing the excess FAs supply or promoting FA storage is beneficial for cardiac function, especially under a lipotoxic condition. The protective effects of several compounds against lipotoxicity progression in the heart have been investigated. A variety of mechanisms has been suggested to prevent or treat cardiac lipotoxicity, including improvement of calcium homeostasis, lipid metabolism, and mitochondrial dysfunction. Known targets and signaling pathways involving a select group of chemicals that interfere with cardiac lipotoxicity pathogenesis are reviewed.
Collapse
|
123
|
Tokarska-Schlattner M, Kay L, Perret P, Isola R, Attia S, Lamarche F, Tellier C, Cottet-Rousselle C, Uneisi A, Hininger-Favier I, Foretz M, Dubouchaud H, Ghezzi C, Zuppinger C, Viollet B, Schlattner U. Role of Cardiac AMP-Activated Protein Kinase in a Non-pathological Setting: Evidence From Cardiomyocyte-Specific, Inducible AMP-Activated Protein Kinase α1α2-Knockout Mice. Front Cell Dev Biol 2021; 9:731015. [PMID: 34733845 PMCID: PMC8558539 DOI: 10.3389/fcell.2021.731015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/24/2021] [Indexed: 12/25/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a key regulator of energy homeostasis under conditions of energy stress. Though heart is one of the most energy requiring organs and depends on a perfect match of energy supply with high and fluctuating energy demand to maintain its contractile performance, the role of AMPK in this organ is still not entirely clear, in particular in a non-pathological setting. In this work, we characterized cardiomyocyte-specific, inducible AMPKα1 and α2 knockout mice (KO), where KO was induced at the age of 8 weeks, and assessed their phenotype under physiological conditions. In the heart of KO mice, both AMPKα isoforms were strongly reduced and thus deleted in a large part of cardiomyocytes already 2 weeks after tamoxifen administration, persisting during the entire study period. AMPK KO had no effect on heart function at baseline, but alterations were observed under increased workload induced by dobutamine stress, consistent with lower endurance exercise capacity observed in AMPK KO mice. AMPKα deletion also induced a decrease in basal metabolic rate (oxygen uptake, energy expenditure) together with a trend to lower locomotor activity of AMPK KO mice 12 months after tamoxifen administration. Loss of AMPK resulted in multiple alterations of cardiac mitochondria: reduced respiration with complex I substrates as measured in isolated mitochondria, reduced activity of complexes I and IV, and a shift in mitochondrial cristae morphology from lamellar to mixed lamellar-tubular. A strong tendency to diminished ATP and glycogen level was observed in older animals, 1 year after tamoxifen administration. Our study suggests important roles of cardiac AMPK at increased cardiac workload, potentially limiting exercise performance. This is at least partially due to impaired mitochondrial function and bioenergetics which degrades with age.
Collapse
Affiliation(s)
- Malgorzata Tokarska-Schlattner
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Laurence Kay
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Pascale Perret
- Inserm U1039, Radiopharmaceutiques Biocliniques, Faculté de Médecine, University of Grenoble Alpes, Grenoble, France
| | - Raffaella Isola
- Department of Biomedical Sciences, Division of Cytomorphology, University of Cagliari, Cagliari, Italy
| | - Stéphane Attia
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Frédéric Lamarche
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Cindy Tellier
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Cécile Cottet-Rousselle
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Amjad Uneisi
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Isabelle Hininger-Favier
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Marc Foretz
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Hervé Dubouchaud
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France
| | - Catherine Ghezzi
- Inserm U1039, Radiopharmaceutiques Biocliniques, Faculté de Médecine, University of Grenoble Alpes, Grenoble, France
| | - Christian Zuppinger
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Benoit Viollet
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Uwe Schlattner
- Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Grenoble, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
124
|
Yuan M, Yan R, Zhang Y, Qiu Y, Jiang Z, Liu H, Wang Y, Sun L, Zhang H, Gao P. CARS senses cysteine deprivation to activate AMPK for cell survival. EMBO J 2021; 40:e108028. [PMID: 34472622 DOI: 10.15252/embj.2021108028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/19/2022] Open
Abstract
Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) is an important cellular metabolite-sensing enzyme that can directly sense changes not only in ATP but also in metabolites associated with carbohydrates and fatty acids. However, less is known about whether and how AMPK senses variations in cellular amino acids. Here, we show that cysteine deficiency significantly triggers calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-mediated activation of AMPK. In addition, we found that CaMKK2 directly associates with cysteinyl-tRNA synthetase (CARS), which then binds to AMPKγ2 under cysteine deficiency to activate AMPK. Interestingly, we discovered that cysteine inhibits the binding of CARS to AMPKγ2, and thus, under cysteine deficiency conditions wherein the inhibitory effect of cysteine is abrogated, CARS mediates the binding of AMPK to CaMKK2, resulting in the phosphorylation and activation of AMPK by CaMKK2. Importantly, we demonstrate that blocking AMPK activation leads to cell death under cysteine-deficient conditions. In summary, our study is the first to show that CARS senses the absence of cysteine and activates AMPK through the cysteine-CARS-CaMKK2-AMPKγ2 axis, a novel adaptation strategy for cell survival under nutrient deprivation conditions.
Collapse
Affiliation(s)
- Mengqiu Yuan
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Signaling Network, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Ronghui Yan
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Signaling Network, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Yi Zhang
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Yue Qiu
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Zetan Jiang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Signaling Network, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Haiying Liu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Signaling Network, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Ying Wang
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Linchong Sun
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| | - Huafeng Zhang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Signaling Network, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Ping Gao
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Signaling Network, School of Life Science, University of Science and Technology of China, Hefei, China.,Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, China
| |
Collapse
|
125
|
Qiu Q, Zou H, Zou H, Jing T, Li X, Yan G, Geng N, Zhang B, Zhang Z, Zhang S, Yao B, Zhang G, Zou C. 3-Bromopyruvate-induced glycolysis inhibition impacts larval growth and development and carbohydrate homeostasis in fall webworm, Hyphantria cunea Drury. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 179:104961. [PMID: 34802511 DOI: 10.1016/j.pestbp.2021.104961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 06/13/2023]
Abstract
As a typical glycolytic inhibitor, 3-bromopyruvate (3-BrPA) has been extensively studied in cancer therapy in recent decades. However, few studies focused on 3-BrPA in regulating the growth and development of insects, and the relationship and regulatory mechanism between glycolysis and chitin biosynthesis remain largely unknown. The Hyphantria cunea, named fall webworm, is a notorious defoliator, which caused a huge economic loss to agriculture and forestry. Here, we investigated the effects of 3-BrPA on the growth and development, glycolysis, carbohydrate homeostasis, as well as chitin synthesis in H. cunea larvae. To elucidate the action mechanism of 3-BrPA on H. cunea will provide a new insight for the control of this pest. The results showed that 3-BrPA dramatically restrained the growth and development of H. cunea larvae and resulted in larval lethality. Meanwhile, we confirmed that 3-BrPA caused a significant decrease in carbohydrate, adenosine triphosphate (ATP), pyruvic acid (PA), and triglyceride (TG) levels by inhibiting glycolysis in H. cunea larvae. Further studies indicated that 3-BrPA significantly affected the activities of hexokinase (HK), phosphofructokinase (PFK), pyruvate kinase (PK), glucose 6-phosphate dehydrogenase (G6PDH) and trehalase, as well as expressions of the genes related to glycolysis, resulting in carbohydrate homeostasis disorder. Moreover, it was found that 3-BrPA enhanced 20-hydroxyecdysone (20E) signaling by upregulating HcCYP306A1 and HcCYP314A1, two critical genes in 20E synthesis pathway, and accelerated chitin synthesis by upregulating transcriptional levels of genes in the chitin synthesis pathway in H. cunea larvae. Taken together, our findings provide a novel insight into the mechanism of glycolytic inhibitor in regulating the growth and development of insects, and lay a foundation for the potential application of glycolytic inhibitors in pest control as well.
Collapse
Affiliation(s)
- Qian Qiu
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Haifeng Zou
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Hang Zou
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Tianzhong Jing
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - XingPeng Li
- School of Forestry, Beihua University, Jilin 132013, PR China
| | - Gaige Yan
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Nannan Geng
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Bihan Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Zhidong Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Shengyu Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Bin Yao
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Guocai Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China.
| | - Chuanshan Zou
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China.
| |
Collapse
|
126
|
Chen SP, Lin SR, Chen TH, Ng HS, Yim HS, Leong MK, Weng CF. Mangosteen xanthone γ-mangostin exerts lowering blood glucose effect with potentiating insulin sensitivity through the mediation of AMPK/PPARγ. Biomed Pharmacother 2021; 144:112333. [PMID: 34678724 DOI: 10.1016/j.biopha.2021.112333] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/24/2021] [Accepted: 10/10/2021] [Indexed: 01/03/2023] Open
Abstract
Diabetes mellitus (DM) is concomitant with significant morbidity and mortality and its prevalence is accumulative in worldwide. The conventional antidiabetic agents are known to mitigate the symptoms of diabetes; however, they may also cause side and adverse effects. There is an imperative necessity to conduct preclinical and clinical trials for the discovery of alternative therapeutic agents that can overcome the drawbacks of current synthetic antidiabetic drugs. This study aimed to investigate the efficacy of lowering blood glucose and underlined mechanism of γ-mangostin, mangosteen (Garcinia mangostana) xanthones. The results showed γ-Mangostin had a antihyperglycemic ability in short (2 h)- and long-term (28 days) administrations to diet-induced diabetic mice. The long-term administration of γ-mangostin attenuated fasting blood glucose of diabetic mice and exhibited no hepatotoxicity and nephrotoxicity. Moreover, AMPK, PPARγ, α-amylase, and α-glucosidase were found to be the potential targets for simulating binds with γ-mangostin after molecular docking. To validate the docking results, the inhibitory potency of γ-mangostin againstα-amylase/α-glucosidase was higher than Acarbose via enzymatic assay. Interestingly, an allosteric relationship between γ-mangostin and insulin was also found in the glucose uptake of VSMC, FL83B, C2C12, and 3T3-L1 cells. Taken together, the results showed that γ-mangostin exerts anti-hyperglycemic activity through promoting glucose uptake and reducing saccharide digestion by inhibition of α-amylase/α-glucosidase with insulin sensitization, suggesting that γ-mangostin could be a new clue for drug discovery and development to treat diabetes.
Collapse
Affiliation(s)
- Sih-Pei Chen
- Institute of Respiratory Disease, Department of Physiology, School of Basic Medical Science, Xiamen Medical College, Xiamen 361023, Fujian, China; Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 974301, Taiwan
| | - Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 974301, Taiwan
| | - Ting-Hsu Chen
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 974301, Taiwan
| | - Hui-Suan Ng
- Faculty of Applied Science, UCSI University, UCSI Height, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Hip-Seng Yim
- Faculty of Applied Science, UCSI University, UCSI Height, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Max K Leong
- Department of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan.
| | - Ching-Feng Weng
- Institute of Respiratory Disease, Department of Physiology, School of Basic Medical Science, Xiamen Medical College, Xiamen 361023, Fujian, China; Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 974301, Taiwan; Department of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan.
| |
Collapse
|
127
|
Functionalized Gadofullerene Ameliorates Impaired Glycolipid Metabolism in Type 2 Diabetic Mice. J Genet Genomics 2021; 49:364-376. [PMID: 34687945 DOI: 10.1016/j.jgg.2021.09.004] [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: 06/17/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022]
Abstract
The soaring global prevalence of diabetes makes it urgent to explore new drugs with high efficacy and safety. Nanomaterial-derived bioactive agents are emerging as one of the most promising candidates for biomedical application. In the present study, we investigated the anti-diabetic effects of a functionalized gadofullerene (GF) using obese db/db and non-obese MKR mouse T2DM models. In both mouse models, the diabetic phenotypes including hyperglycemia, impaired glucose tolerance and insulin sensitivity were ameliorated following 2 or 4 weeks of i.p. administration of GF. GF lowered blood glucose levels in a dose-dependent manner. Importantly, the restored blood glucose levels could persist 10 days after withdrawal of GF treatment. The hepatic AKT/GSK3β/FoxO1 pathway is shown to be the main target of GF for re-balancing gluconeogenesis and glycogen synthesis in vivo and in vitro. In addition, GF treatment significantly reduced weight gain of db/db mice with reduced hepatic fat storage by the inhibition of de novo lipogenesis through mTOR/S6K/SREBP1 pathway. Our data provide compelling evidence to support the promising application of GF for the treatment of T2DM.
Collapse
|
128
|
Guo L, Liang Y, Wang S, Li L, Cai L, Heng Y, Yang J, Jin X, Zhang J, Yuan S, Xu T, Jia L. Jujuboside B Inhibits the Proliferation of Breast Cancer Cell Lines by Inducing Apoptosis and Autophagy. Front Pharmacol 2021; 12:668887. [PMID: 34630073 PMCID: PMC8497973 DOI: 10.3389/fphar.2021.668887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022] Open
Abstract
Jujuboside B (JB) is one of the main biologically active ingredients extracted from Zizyphi Spinosi Semen (ZSS), a widely used traditional Chinese medicine for treating insomnia and anxiety. Breast cancer is the most common cancer and the second leading cause of cancer-related death in women worldwide. The purpose of this study was to examine whether JB could prevent breast cancer and its underlying mechanism. First, we reported that JB induced apoptosis and autophagy in MDA-MB-231 and MCF-7 human breast cancer cell lines. Further mechanistic studies have revealed that JB-induced apoptosis was mediated by NOXA in both two cell lines. Moreover, the AMPK signaling pathway plays an important role in JB-induced autophagy in MCF-7. To confirm the anti-breast cancer effect of JB, the interaction of JB-induced apoptosis and autophagy was investigated by both pharmacological and genetic approaches. Results indicated that autophagy played a pro-survival role in attenuating apoptosis. Further in vivo study showed that JB significantly suppressed the growth of MDA-MB-231 and MCF-7 xenografts. In conclusion, our findings indicate that JB exerts its anti-breast cancer effect in association with the induction of apoptosis and autophagy.
Collapse
Affiliation(s)
- Lin Guo
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yupei Liang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shiwen Wang
- Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Lihui Li
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Cai
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongqing Heng
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xing Jin
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junqian Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuying Yuan
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Xu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lijun Jia
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
129
|
Huang W, Liu Y, Luz A, Berrong M, Meyer JN, Zou Y, Swann E, Sundaramoorthy P, Kang Y, Jauhari S, Lento W, Chao N, Racioppi L. Calcium/Calmodulin Dependent Protein Kinase Kinase 2 Regulates the Expansion of Tumor-Induced Myeloid-Derived Suppressor Cells. Front Immunol 2021; 12:754083. [PMID: 34712241 PMCID: PMC8546266 DOI: 10.3389/fimmu.2021.754083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a hetero geneous group of cells, which can suppress the immune response, promote tumor progression and impair the efficacy of immunotherapies. Consequently, the pharmacological targeting of MDSC is emerging as a new immunotherapeutic strategy to stimulate the natural anti-tumor immune response and potentiate the efficacy of immunotherapies. Herein, we leveraged genetically modified models and a small molecule inhibitor to validate Calcium-Calmodulin Kinase Kinase 2 (CaMKK2) as a druggable target to control MDSC accumulation in tumor-bearing mice. The results indicated that deletion of CaMKK2 in the host attenuated the growth of engrafted tumor cells, and this phenomenon was associated with increased antitumor T cell response and decreased accumulation of MDSC. The adoptive transfer of MDSC was sufficient to restore the ability of the tumor to grow in Camkk2-/- mice, confirming the key role of MDSC in the mechanism of tumor rejection. In vitro studies indicated that blocking of CaMKK2 is sufficient to impair the yield of MDSC. Surprisingly, MDSC generated from Camkk2-/- bone marrow cells also showed a higher ability to terminally differentiate toward more immunogenic cell types (e.g inflammatory macrophages and dendritic cells) compared to wild type (WT). Higher intracellular levels of reactive oxygen species (ROS) accumulated in Camkk2-/- MDSC, increasing their susceptibility to apoptosis and promoting their terminal differentiation toward more mature myeloid cells. Mechanistic studies indicated that AMP-activated protein kinase (AMPK), which is a known CaMKK2 proximal target controlling the oxidative stress response, fine-tunes ROS accumulation in MDSC. Accordingly, failure to activate the CaMKK2-AMPK axis can account for the elevated ROS levels in Camkk2-/- MDSC. These results highlight CaMKK2 as an important regulator of the MDSC lifecycle, identifying this kinase as a new druggable target to restrain MDSC expansion and enhance the efficacy of anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Wei Huang
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Yaping Liu
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Anthony Luz
- Duke University Nicholas School of the Environment, Durham, NC, United States
| | - Mark Berrong
- Duke Human Vaccine Institute, Durham, NC, United States
| | - Joel N. Meyer
- Duke University Nicholas School of the Environment, Durham, NC, United States
| | - Yujing Zou
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Excel Swann
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Pasupathi Sundaramoorthy
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Yubin Kang
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Shekeab Jauhari
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - William Lento
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Nelson Chao
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Luigi Racioppi
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
130
|
Zhou B, Zhang Y, Li S, Wu L, Fejes-Toth G, Naray-Fejes-Toth A, Soukas AA. Serum- and glucocorticoid-induced kinase drives hepatic insulin resistance by directly inhibiting AMP-activated protein kinase. Cell Rep 2021; 37:109785. [PMID: 34610303 PMCID: PMC8576737 DOI: 10.1016/j.celrep.2021.109785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/05/2021] [Accepted: 09/10/2021] [Indexed: 12/01/2022] Open
Abstract
A hallmark of type 2 diabetes (T2D) is hepatic resistance to insulin's glucose-lowering effects. The serum- and glucocorticoid-regulated family of protein kinases (SGK) is activated downstream of mechanistic target of rapamycin complex 2 (mTORC2) in response to insulin in parallel to AKT. Surprisingly, despite an identical substrate recognition motif to AKT, which drives insulin sensitivity, pathological accumulation of SGK1 drives insulin resistance. Liver-specific Sgk1-knockout (Sgk1Lko) mice display improved glucose tolerance and insulin sensitivity and are protected from hepatic steatosis when fed a high-fat diet. Sgk1 promotes insulin resistance by inactivating AMP-activated protein kinase (AMPK) via phosphorylation on inhibitory site AMPKαSer485/491. We demonstrate that SGK1 is dominant among SGK family kinases in regulation of insulin sensitivity, as Sgk1, Sgk2, and Sgk3 triple-knockout mice have similar increases in hepatic insulin sensitivity. In aggregate, these data suggest that targeting hepatic SGK1 may have therapeutic potential in T2D.
Collapse
Affiliation(s)
- Ben Zhou
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yuyao Zhang
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Sainan Li
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Lianfeng Wu
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, Hangzhou, 310024, China
| | - Geza Fejes-Toth
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Aniko Naray-Fejes-Toth
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Alexander A Soukas
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
131
|
Wang X, Zhou X, Uberseder B, Lee J, Latimer CS, Furdui CM, Keene CD, Montine TJ, Register TC, Craft S, Shively CA, Ma T. Isoform-specific dysregulation of AMP-activated protein kinase signaling in a non-human primate model of Alzheimer's disease. Neurobiol Dis 2021; 158:105463. [PMID: 34363967 PMCID: PMC8440492 DOI: 10.1016/j.nbd.2021.105463] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/22/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a molecular sensor that is critical for the maintenance of cellular energy homeostasis, disruption of which has been indicated in multiple neurodegenerative diseases including Alzheimer's disease (AD). Mammalian AMPK is a heterotrimeric complex and its enzymatic α subunit exists in two isoforms: AMPKα1 and AMPKα2. Here we took advantage of a recently characterized non-human primate (NHP) model with sporadic AD-like neuropathology to explore potential relationships between AMPK signaling and AD-like neuropathology. Subjects were nine female vervet monkeys aged 19.5 to 23.4 years old. Subjects were classified into three groups, control lacking AD pathology (n = 3), moderate AD pathology (n = 3), and more severe AD Pathology (n = 3). We found increased activity (assessed by phosphorylation) of AMPKα2 in hippocampi of NHP with AD-like neuropathology, compared to the subjects without AD pathology, with no alterations of AMPKα1 activity. Across all subjects, CSF Abeta42 was inversely associated with cerebral amyloid plaque density. Further, Aβ plaque burden is correlated with levels of either soluble or insoluble brain Aβ measurement. Unbiased mass spectrometry based proteomics studies combined with bioinformatics analysis revealed that many of the dysregulated proteins characteristic of AD neuropathology are associated with AMPK signaling. Our findings on the AMPK molecular signaling cascades provide further support for use of the NHP model to investigate new therapeutic strategies and development of novel biomarkers for Alzheimer's disease.
Collapse
Affiliation(s)
- Xin Wang
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Xueyan Zhou
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Beth Uberseder
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jingyun Lee
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Caitlin S Latimer
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Cristina M Furdui
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Thomas C Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Suzanne Craft
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carol A Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tao Ma
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
132
|
Liu Y, Cui S, Sun J, Yan X, Han D. Identification of Potential Biomarkers for Psoriasis by DNA Methylation and Gene Expression Datasets. Front Genet 2021; 12:722803. [PMID: 34512732 PMCID: PMC8427602 DOI: 10.3389/fgene.2021.722803] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/22/2021] [Indexed: 12/30/2022] Open
Abstract
DNA methylation (DNAm) plays an important role in the pathogenesis of psoriasis through regulating mRNA expressions. This study aimed to identify hub genes regulated by DNAm as biomarkers of psoriasis. Psoriatic skin tissues gene expression and methylation datasets were downloaded from Gene Expression Omnibus (GEO) database. Subsequently, multiple computational approaches, including immune infiltration analysis, enrichment analysis, protein-protein interaction (PPI) network establishment, and machine learning algorithm analysis (lasso, random forest, and SVM-RFE), were performed to analyze the regulatory networks, to recognize hub genes, and to clarify the pathogenesis of psoriasis. Finally, the hypermethylated genes were used to immune cell infiltration analysis, which revealed that psoriasis skin tissues were mainly composed of activated dendritic cells, resting mast cells, T follicular helper cells (cTfh), etc. Differentially expressed-methylated genes (DEMGs) were identified and partitioned into four subgroups and the 97 significantly hypermethylated and downregulated (hyper-down) genes accounted for the highest proportion (47%). Hyper-down genes were mainly enriched in glucose homeostasis, AMP-activated protein kinase (AMPK) signaling pathway, lipid storage disease, partial lipodystrophy, and insulin resistance. Furthermore, insulin receptor substrate 1 (IRS1), Rho guanine nucleotide exchange factor 10 (ARHGEF10) and retinoic acid induced 14 (RAI14) were identified as potential targets. These findings provided new ideas for future studies of psoriasis on the occurrence and the molecular mechanisms.
Collapse
Affiliation(s)
- Yong Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Department of Dermatology, Shaanxi Hospital of Chinese Medicine, Xi'an, China
| | - Shengnan Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiayi Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoning Yan
- Department of Dermatology, Shaanxi Hospital of Chinese Medicine, Xi'an, China
| | - Dongran Han
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
133
|
AMPK-mTOR Signaling and Cellular Adaptations in Hypoxia. Int J Mol Sci 2021; 22:ijms22189765. [PMID: 34575924 PMCID: PMC8465282 DOI: 10.3390/ijms22189765] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022] Open
Abstract
Cellular energy is primarily provided by the oxidative degradation of nutrients coupled with mitochondrial respiration, in which oxygen participates in the mitochondrial electron transport chain to enable electron flow through the chain complex (I-IV), leading to ATP production. Therefore, oxygen supply is an indispensable chapter in intracellular bioenergetics. In mammals, oxygen is delivered by the bloodstream. Accordingly, the decrease in cellular oxygen level (hypoxia) is accompanied by nutrient starvation, thereby integrating hypoxic signaling and nutrient signaling at the cellular level. Importantly, hypoxia profoundly affects cellular metabolism and many relevant physiological reactions induce cellular adaptations of hypoxia-inducible gene expression, metabolism, reactive oxygen species, and autophagy. Here, we introduce the current knowledge of hypoxia signaling with two-well known cellular energy and nutrient sensing pathways, AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin complex 1 (mTORC1). Additionally, the molecular crosstalk between hypoxic signaling and AMPK/mTOR pathways in various hypoxic cellular adaptions is discussed.
Collapse
|
134
|
Li W, Wang D, Li M, Li B. Emodin inhibits the proliferation of papillary thyroid carcinoma by activating AMPK. Exp Ther Med 2021; 22:1075. [PMID: 34447468 PMCID: PMC8355685 DOI: 10.3892/etm.2021.10509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 03/25/2021] [Indexed: 01/01/2023] Open
Abstract
Emodin has been demonstrated to serve antitumor roles in a variety of tumor types, but the effect of emodin on papillary thyroid carcinoma and its molecular mechanisms remain unclear. In the current study, the role of emodin on papillary thyroid carcinoma was analyzed in vitro and in vivo. TPC-1 cells were treated with emodin (0, 10, 25 or 50 µM), and cell viability and apoptosis were detected using Cell Counting Kit-8 and flow cytometry, respectively. The expression levels of AMPK-associated proteins were examined using western blot analysis. To study the effect of emodin on the AMPK pathway, AMPK activator, AICAR and an AMPK inhibitor, Dorsomorphin, were used in TPC-1 cells. In vivo, mice were used to confirm the mechanism of emodin on papillary thyroid carcinoma. The results of the current study indicated that emodin treatment induced cell apoptosis and cell cycle arrest in TPC-1 cells. Furthermore, the inhibitory effect increased in a dose dependent manner. Following emodin treatment, the cell viability of TPC-1 cells was significantly decreased, and apoptosis rate increased (P<0.05). Furthermore, the expression levels of AMPK were increased in the emodin group compared with the control group (P<0.05). Similar effects were observed following AMPK activator treatment in TPC-1 cells. Following AMPK activator treatment, cell proliferation and the cell cycle were inhibited. Also, the AMPK inhibitor was demonstrated to mediate the therapeutic effect of emodin. In addition, the results of the present study demonstrated that emodin inhibited the MEK/ERK pathway. Additionally, the in vivo results of the current study were consistent with those in vitro. In conclusion, the current study demonstrated that the administration of Emodin inhibited the proliferation of papillary thyroid cancer cells via activating AMPK pathway activity.
Collapse
Affiliation(s)
- Weilong Li
- Department of Nuclear Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Dong Wang
- Department of Thyroid, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Meijing Li
- Department of Hepatobiliary, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Baoyuan Li
- Department of Thyroid, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
135
|
Sun WX, Lou K, Chen LJ, Liu SD, Pang SG. Lipocalin-2: a role in hepatic gluconeogenesis via AMP-activated protein kinase (AMPK). J Endocrinol Invest 2021; 44:1753-1765. [PMID: 33423221 DOI: 10.1007/s40618-020-01494-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/23/2020] [Indexed: 01/19/2023]
Abstract
PURPOSE Evidence is accumulating that lipocalin2 (LCN2) is implicated in insulin resistance and glucose homeostasis, but the underlying possible mechanisms remain unclear. This study is to investigate the possible linkage between LCN2 and AMP-activated protein kinase (AMPK) or forkhead transcription factor O1 (FoxO1), which influences insulin sensitivity and gluconeogenesis in liver. METHODS LCN2 knockout (LCN2KO) mice and wild-type littermates were used to evaluate the effect of LCN2 on insulin sensitivity and hepatic gluconeogenesis through pyruvate tolerance test (PTT), glucose tolerance test (ipGTT), insulin tolerance test (ITT), and hyperinsulinemic-euglycemic clamps, respectively. LCN2KO mice and WT mice in vivo, and in vitro HepG2 cells were co-transfected with adenoviral FoxO1-siRNA (Ad-FoxO1-siRNA) or adenovirus expressing constitutively active form of AMPK (Ad-CA-AMPK), or dominant negative adenovirus AMPK (Ad-DN-AMPK), the relative mRNA and protein levels of two key gluconeogenic enzymes phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6P) were measured. RESULTS Improved insulin sensitivity and inhibited gluconeogenesis in the LCN2KO mice were confirmed by pyruvate tolerance tests and hyperinsulinemic-euglycemic clamps. Nuclear FoxO1 and its downstream genes PEECK and G6P were decreased in the livers of the LCN2KO mice, and AMPK activity was stimulated and directly phosphorylated FoxO1. In vitro, AMPK activity was inhibited in HepG2 cells overexpressing LCN2 leading to a decrease in phosphorylated FoxO1 and an increase in nuclear FoxO1. CONCLUSION The present study demonstrates that LCN2 regulates insulin sensitivity and glucose metabolism through inhibiting AMPK activity, and regulating FoxO1 and its downstream genes PEPCK/G6P, which regulate hepatic gluconeogenesis.
Collapse
Affiliation(s)
- W-X Sun
- Department of Pharmacy, Taishan Vocational College of Nursing, Taian, 271000, China
| | - K Lou
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, 105 Jiefang Road, Jinan, 250013, Shandong Province, China
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - L-J Chen
- Department of Endocrinology, Shandong Rongjun General Hospital, 23 Jiefang Road, Jinan, 250013, Shandong Province, China
| | - S-D Liu
- Department of Endocrinology, Shandong Rongjun General Hospital, 23 Jiefang Road, Jinan, 250013, Shandong Province, China.
| | - S-G Pang
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, 105 Jiefang Road, Jinan, 250013, Shandong Province, China.
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
| |
Collapse
|
136
|
Monlish DA, Beezhold KJ, Chiaranunt P, Paz K, Moore NJ, Dobbs AK, Brown RA, Ozolek JA, Blazar BR, Byersdorfer CA. Deletion of AMPK minimizes graft-versus-host disease through an early impact on effector donor T cells. JCI Insight 2021; 6:e143811. [PMID: 34291733 PMCID: PMC8410053 DOI: 10.1172/jci.insight.143811] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 06/10/2021] [Indexed: 12/25/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is a viable treatment for multiple hematologic diseases, but its application is often limited by graft-versus-host disease (GVHD), where donor T cells attack host tissues in the skin, liver, and gastrointestinal tract. Here, we examined the role of the cellular energy sensor AMP kinase (AMPK) in alloreactive T cells during GVHD development. Early posttransplant, AMPK activity increased more than 15-fold in allogeneic T cells, and transplantation of T cells deficient in both AMPKα1 and AMPKα2 decreased GVHD severity in multiple disease models. Importantly, a lack of AMPK lessened GVHD without compromising antileukemia responses or impairing lymphopenia-driven immune reconstitution. Mechanistically, absence of AMPK decreased both CD4+ and CD8+ effector T cell numbers as early as day 3 posttransplant, while simultaneously increasing regulatory T cell (Treg) percentages. Improvements in GVHD resulted from cell-intrinsic perturbations in conventional effector T cells as depletion of donor Tregs had minimal impact on AMPK-related improvements. Together, these results highlight a specific role for AMPK in allogeneic effector T cells early posttransplant and suggest that AMPK inhibition may be an innovative approach to mitigate GVHD while preserving graft-versus-leukemia responses and maintaining robust immune reconstitution.
Collapse
Affiliation(s)
- Darlene A Monlish
- Division of Blood and Marrow Transplantation and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kevin J Beezhold
- Division of Blood and Marrow Transplantation and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Pailin Chiaranunt
- Division of Blood and Marrow Transplantation and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Katelyn Paz
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nathan J Moore
- Division of Blood and Marrow Transplantation and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrea K Dobbs
- Division of Blood and Marrow Transplantation and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rebecca A Brown
- Division of Blood and Marrow Transplantation and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John A Ozolek
- Department of Pathology, Anatomy and Laboratory Medicine, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Bruce R Blazar
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Craig A Byersdorfer
- Division of Blood and Marrow Transplantation and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
137
|
Zhang Z, Yang D, Xiang J, Zhou J, Cao H, Che Q, Bai Y, Guo J, Su Z. Non-shivering Thermogenesis Signalling Regulation and Potential Therapeutic Applications of Brown Adipose Tissue. Int J Biol Sci 2021; 17:2853-2870. [PMID: 34345212 PMCID: PMC8326120 DOI: 10.7150/ijbs.60354] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/23/2021] [Indexed: 12/25/2022] Open
Abstract
In mammals, thermogenic organs exist in the body that increase heat production and enhance energy regulation. Because brown adipose tissue (BAT) consumes energy and generates heat, increasing energy expenditure via BAT might be a potential strategy for new treatments for obesity and obesity-related diseases. Thermogenic differentiation affects normal adipose tissue generation, emphasizing the critical role that common transcriptional regulation factors might play in common characteristics and sources. An understanding of thermogenic differentiation and related factors could help in developing ways to improve obesity indirectly or directly through targeting of specific signalling pathways. Many studies have shown that the active components of various natural products promote thermogenesis through various signalling pathways. This article reviews recent major advances in this field, including those in the cyclic adenosine monophosphate-protein kinase A (cAMP-PKA), cyclic guanosine monophosphate-GMP-dependent protein kinase G (cGMP-AKT), AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), transforming growth factor-β/bone morphogenic protein (TGF-β/BMP), transient receptor potential (TRP), Wnt, nuclear factor-κ-light-chain-enhancer of activated B cells (NF-κΒ), Notch and Hedgehog (Hh) signalling pathways in brown and brown-like adipose tissue. To provide effective information for future research on weight-loss nutraceuticals or drugs, this review also highlights the natural products and their active ingredients that have been reported in recent years to affect thermogenesis and thus contribute to weight loss via the above signalling pathways.
Collapse
Affiliation(s)
- Zhengyan Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Di Yang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Junwei Xiang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingwen Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hua Cao
- Guangdong Cosmetics Engineering & Technology Research Center, School of Chemistry and Chemical Engneering, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd., Guangzhou 510663, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
138
|
PPAR γ Plays an Important Role in Acute Hepatic Ischemia-Reperfusion Injury via AMPK/mTOR Pathway. PPAR Res 2021; 2021:6626295. [PMID: 34285690 PMCID: PMC8275421 DOI: 10.1155/2021/6626295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Background Hepatic ischemia-reperfusion (IR) injury is one of the severe complications associated with liver surgery and leads to liver dysfunction. PPARγ is always linked with various physiologic pathways, and it can alleviate liver damage in IR injury. Aim In this study, we explored the potential mechanism of PPARγ in the pathogenesis of hepatic IR injury by mice model. Methods After treated with si-PPARγ or rosiglitazone, mice were subjected to hepatic ischemia-reperfusion. Liver tissue and blood samples were collected to evaluate liver injury and detected relative mRNA and protein expressions. Results The expression of PPARγ was increased after reperfusion. And the alleviation of PPARγ aggravated the liver damage in IR; at the same time, upregulation of the expression of PPARγ released the liver damage. And these effects of PPARγ in IR were related to the AMPK/mTOR/autophagy signaling pathway. Conclusion PPARγ plays an important role in hepatic IR injury at least partly via the AMPK/mTOR/autophagy pathway.
Collapse
|
139
|
Juban G. Transcriptional control of macrophage inflammatory shift during skeletal muscle regeneration. Semin Cell Dev Biol 2021; 119:82-88. [PMID: 34183241 DOI: 10.1016/j.semcdb.2021.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/13/2021] [Accepted: 06/18/2021] [Indexed: 01/05/2023]
Abstract
Skeletal muscle is a tissue able to fully regenerate after an acute injury. Macrophages play an essential role during skeletal muscle regeneration. Resolution of inflammation is a crucial step during the regeneration process, allowing to contain the inflammatory response to avoid damage of the healthy surrounding muscle and triggers the recovery phase during which the muscle regenerates. Resolution of inflammation is mainly mediated by macrophage phenotypic shift that is the transition from a pro-inflammatory damage associated profile towards an anti-inflammatory restorative phenotype, which is characterized by a major transcriptional rewiring. Failure of the resolution of inflammation is observed in chronic diseases such as degenerative myopathies where permanent asynchronous muscle injuries trigger contradictory inflammatory cues, leading to fibrosis and alteration of muscle function. This review will focus on the described molecular pathways that control macrophage inflammatory shift during skeletal muscle regeneration. First, we will highlight the transcriptional changes that characterize macrophage inflammatory shift during skeletal muscle regeneration. Then, we will describe how the signaling pathways and the metabolic changes associated with this shift are controlled. Finally, we will emphasize the transcription factors involved.
Collapse
Affiliation(s)
- Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, Lyon, France.
| |
Collapse
|
140
|
Vahidi Ferdowsi P, Ahuja KDK, Beckett JM, Myers S. TRPV1 Activation by Capsaicin Mediates Glucose Oxidation and ATP Production Independent of Insulin Signalling in Mouse Skeletal Muscle Cells. Cells 2021; 10:cells10061560. [PMID: 34205555 PMCID: PMC8234135 DOI: 10.3390/cells10061560] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Insulin resistance (IR), a key characteristic of type 2 diabetes (T2DM), is manifested by decreased insulin-stimulated glucose transport in target tissues. Emerging research has highlighted transient receptor potential cation channel subfamily V member (TRPV1) activation by capsaicin as a potential therapeutic target for these conditions. However, there are limited data on the effects of capsaicin on cell signalling molecules involved in glucose uptake. METHODS C2C12 cells were cultured and differentiated to acquire the myotube phenotype. The activation status of signalling molecules involved in glucose metabolism, including 5' adenosine monophosphate-activated protein kinase (AMPK), calcium/calmodulin-dependent protein kinase 2 (CAMKK2), extracellular signal-regulated protein kinases 1 and 2 (ERK1/2), protein kinase B (AKT), and src homology phosphatase 2 (SHP2), was examined. Finally, activation of CAMKK2 and AMPK, and glucose oxidation and ATP levels were measured in capsaicin-treated cells in the presence or absence of TRPV1 antagonist (SB-452533). RESULTS Capsaicin activated cell signalling molecules including CAMKK2 and AMPK leading to increased glucose oxidation and ATP generation independent of insulin in the differentiated C2C12 cells. Pharmacological inhibition of TRPV1 diminished the activation of CAMKK2 and AMPK as well as glucose oxidation and ATP production. Moreover, we observed an inhibitory effect of capsaicin in the phosphorylation of ERK1/2 in the mouse myotubes. CONCLUSION Our data show that capsaicin-mediated stimulation of TRPV1 in differentiated C2C12 cells leads to activation of CAMKK2 and AMPK, and increased glucose oxidation which is concomitant with an elevation in intracellular ATP level. Further studies of the effect of TRPV1 channel activation by capsaicin on glucose metabolism could provide novel therapeutic utility for the management of IR and T2DM.
Collapse
|
141
|
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
|
142
|
Bheemanapally K, Ibrahim MMH, Alshamrani A, Briski KP. Ventromedial hypothalamic nucleus glycogen regulation of metabolic-sensory neuron AMPK and neurotransmitter expression: role of lactate. Am J Physiol Regul Integr Comp Physiol 2021; 320:R791-R799. [PMID: 33825506 PMCID: PMC8285616 DOI: 10.1152/ajpregu.00292.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/18/2021] [Accepted: 03/31/2021] [Indexed: 12/23/2022]
Abstract
Astrocyte glycogen is dynamically remodeled during metabolic stability and provides oxidizable l-lactate equivalents during neuroglucopenia. Current research investigated the hypothesis that ventromedial hypothalamic nucleus (VMN) glycogen metabolism controls glucostimulatory nitric oxide (NO) and/or glucoinhibitory gamma-aminobutyric acid (GABA) neuron 5'-AMP-activated protein kinase (AMPK) and transmitter marker, e.g., neuronal nitric oxide synthase (nNOS), and glutamate decarboxylase65/67 (GAD) protein expression. Adult ovariectomized estradiol-implanted female rats were injected into the VMN with the glycogen phosphorylase inhibitor 1,4-dideoxy-1,4-imino-d-arabinitol (DAB) before vehicle or l-lactate infusion. Western blot analysis of laser-catapult-microdissected nitrergic and GABAergic neurons showed that DAB caused lactate-reversible upregulation of nNOS and GAD proteins. DAB suppressed or increased total AMPK content of NO and GABA neurons, respectively, by lactate-independent mechanisms, but lactate prevented drug enhancement of pAMPK expression in nitrergic neurons. Inhibition of VMN glycogen disassembly caused divergent changes in counter-regulatory hormone, e.g. corticosterone (increased) and glucagon (decreased) secretion. Outcomes show that VMN glycogen metabolism controls local glucoregulatory transmission by means of lactate signal volume. Results implicate glycogen-derived lactate deficiency as a physiological stimulus of corticosterone release. Concurrent normalization of nitrergic neuron nNOS and pAMPK protein and corticosterone secretory response to DAB by lactate infers that the hypothalamic-pituitary-adrenal axis may be activated by VMN NO-mediated signals of cellular energy imbalance.
Collapse
Affiliation(s)
- Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Mostafa M H Ibrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Ayed Alshamrani
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| |
Collapse
|
143
|
Yang W, Zhou X, Zimmermann HR, Ma T. Brain-specific suppression of AMPKα2 isoform impairs cognition and hippocampal LTP by PERK-mediated eIF2α phosphorylation. Mol Psychiatry 2021; 26:1880-1897. [PMID: 32366952 PMCID: PMC8054310 DOI: 10.1038/s41380-020-0739-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 04/07/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
The AMP-activated protein kinase (AMPK) is a molecular sensor to maintain energy homeostasis. The two isoforms of the AMPK catalytic subunit (AMPKα1 and α2) are both expressed in brains, but their roles in cognition are unknown. We generated conditional knockout mice in which brain AMPKα isoforms are selectively suppressed (AMPKα1/α2 cKO), and determined the isoform-specific effects in mice of either sex on cognition and synaptic plasticity. AMPKα2 cKO but not AMPKα1 cKO displayed impaired cognition and hippocampal late long-term potentiation (L-LTP). Further, AMPKα2 cKO mice exhibited decreased dendritic spine density and abnormal spine morphology in hippocampus. Electron microscope imaging demonstrated reduced postsynaptic density formation and fewer dendritic polyribosomes in hippocampi of AMPKα2 cKO mice. Biochemical studies revealed unexpected findings that repression of AMPKα2 resulted in increased phosphorylation of mRNA translational factor eIF2α and its kinase PERK. Importantly, L-LTP failure and cognitive impairments displayed in AMPKα2 cKO mice were alleviated by suppressing PERK activity pharmacologically or genetically. In summary, we demonstrate here that brain-specific suppression of AMPKα2 isoform impairs cognition and hippocampal LTP by PERK-mediated eIF2α phosphorylation, providing molecular mechanisms linking metabolism, protein synthesis, and cognition.
Collapse
Affiliation(s)
- Wenzhong Yang
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Xueyan Zhou
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Helena R. Zimmermann
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Tao Ma
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA. .,Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA. .,Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
144
|
Lin Y, Ji H, Cao X, Cen Y, Chen Y, Ji S, Zheng S. Knockdown of AMP-activated protein kinase increases the insecticidal efficiency of pymetrozine to Nilaparvata lugens. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 175:104856. [PMID: 33993974 DOI: 10.1016/j.pestbp.2021.104856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/07/2021] [Accepted: 04/11/2021] [Indexed: 06/12/2023]
Abstract
Insecticides are the main tools used to control Nilaparvata lugens (Stål), a serious pest of rice in Asia. However, repeated application of insecticides has caused many negative effects. Reducing the amount of insecticide used, while maintaining good pest population control, would be valuable. AMP-activated protein kinase (AMPK), a sensor of cellular energy status, helps to maintain insect energy balance at the cellular and whole-body level. The role of AMPK in insect response to insecticide stimulation is unknown. We studied the functions of AMPK catalytic subunit alpha (NlAMPKα) in the development of N. lugens and in response to pymetrozine, an insecticide used to control insect pests with piercing-sucking mouthparts. A phylogenetic analysis of protein sequences from 12 species in six orders showed that insects have only the AMPKα 2 subtype. RNA interference against NlAMPKα demonstrated that blocking the AMPK pathway led to a decrease in the systemic ATP level and an increase in N. lugens mortality. NlAMPKα responded to the energy stress caused by pymetrozine treatment, which activated downstream energy metabolic pathways to compensate for the energy imbalance. However, the ATP level in pymetrozine- treated nymphs was not increased, suggesting that ATP is consumed more than synthesized. When NlAMPKα expression was reduced in pymetrozine-treated nymphs by RNAi, the ATP level was decreased and the mortality was significantly increased. At day eight post 0.5 g/3 L of pymetrozine and dsNlAMPKα treatment, nymph survival was 29.33%, which was similar to the 27.33% survival of 1 g/3 L pymetrozine-treated nymphs. Addition of dsNlAMPKα can reduce the concentration of pymetrozine used by 50% while providing comparable efficacy. These results indicate that AMPK helps maintain the energy metabolism of N. lugens in response to pymetrozine treatment. Knockdown of NlAMPKα increases the insecticidal efficiency of pymetrozine to N. lugens.
Collapse
Affiliation(s)
- Yiguang Lin
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Huijun Ji
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaocong Cao
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yongjie Cen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yumei Chen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Shuangshun Ji
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Sichun Zheng
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
145
|
Therapeutic potential of AMPK signaling targeting in lung cancer: Advances, challenges and future prospects. Life Sci 2021; 278:119649. [PMID: 34043989 DOI: 10.1016/j.lfs.2021.119649] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Lung cancer (LC) is a leading cause of death worldwide with high mortality and morbidity. A wide variety of risk factors are considered for LC development such as smoking, air pollution and family history. It appears that genetic and epigenetic factors are also potential players in LC development and progression. AMP-activated protein kinase (AMPK) is a signaling pathway with vital function in inducing energy balance and homeostasis. An increase in AMP:ATP and ADP:ATP ratio leads to activation of AMPK signaling by upstream mediators such as LKB1 and CamKK. Dysregulation of AMPK signaling is a common finding in different cancers, particularly LC. AMPK activation can significantly enhance LC metastasis via EMT induction. Upstream mediators such as PLAG1, IMPAD1, and TUFM can regulate AMPK-mediated metastasis. AMPK activation can promote proliferation and survival of LC cells via glycolysis induction. In suppressing LC progression, anti-tumor compounds including metformin, ginsenosides, casticin and duloxetine dually induce/inhibit AMPK signaling. This is due to double-edged sword role of AMPK signaling in LC cells. Furthermore, AMPK signaling can regulate response of LC cells to chemotherapy and radiotherapy that are discussed in the current review.
Collapse
|
146
|
Deneka AY, Kopp MC, Nikonova AS, Gaponova AV, Kiseleva AA, Hensley HH, Flieder DB, Serebriiskii IG, Golemis EA. Nedd9 Restrains Autophagy to Limit Growth of Early Stage Non-Small Cell Lung Cancer. Cancer Res 2021; 81:3717-3726. [PMID: 34006524 DOI: 10.1158/0008-5472.can-20-3626] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/16/2021] [Accepted: 04/26/2021] [Indexed: 01/22/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common cancer worldwide. With overall 5-year survival estimated at <17%, it is critical to identify factors that regulate NSCLC disease prognosis. NSCLC is commonly driven by mutations in KRAS and TP53, with activation of additional kinases such as SRC promoting tumor invasion. In this study, we investigated the role of NEDD9, a SRC activator and scaffolding protein, in NSCLC tumorigenesis. In an inducible model of NSCLC dependent on Kras mutation and Trp53 loss (KP mice), deletion of Nedd9 (KPN mice) led to the emergence of larger tumors characterized by accelerated rates of tumor growth and elevated proliferation. Orthotopic injection of KP and KPN tumors into the lungs of Nedd9-wild-type and -null mice indicated the effect of Nedd9 loss was cell-autonomous. Tumors in KPN mice displayed reduced activation of SRC and AKT, indicating that activation of these pathways did not mediate enhanced growth of KPN tumors. NSCLC tumor growth has been shown to require active autophagy, a process dependent on activation of the kinases LKB1 and AMPK. KPN tumors contained high levels of active LKB1 and AMPK and increased autophagy compared with KP tumors. Treatment with the autophagy inhibitor chloroquine completely eliminated the growth advantage of KPN tumors. These data for the first time identify NEDD9 as a negative regulator of LKB1/AMPK-dependent autophagy during early NSCLC tumor growth. SIGNIFICANCE: This study demonstrates a novel role for the scaffolding protein NEDD9 in regulating LKB1-AMPK signaling in early stage non-small cell lung cancer, suppressing autophagy and tumor growth.
Collapse
Affiliation(s)
- Alexander Y Deneka
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.,Kazan Federal University, Kazan, Russian Federation, Kazan, Tatarstan, Russia
| | - Meghan C Kopp
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.,Cancer Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Anna S Nikonova
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Anna V Gaponova
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Anna A Kiseleva
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Harvey H Hensley
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA
| | - Douglas B Flieder
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.,Department of Pathology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | - Erica A Golemis
- Program in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA.
| |
Collapse
|
147
|
Arazi H, Eghbali E. Possible Effects of Beetroot Supplementation on Physical Performance Through Metabolic, Neuroendocrine, and Antioxidant Mechanisms: A Narrative Review of the Literature. Front Nutr 2021; 8:660150. [PMID: 34055855 PMCID: PMC8155490 DOI: 10.3389/fnut.2021.660150] [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: 01/28/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Athletes often seek to use dietary supplements to increase performance during exercise. Among various supplements, much attention has been paid to beetroot in recent years. Beetroot is a source of carbohydrates, fiber, protein, minerals, and vitamins; also, it is a natural source of nitrate and associated with improved sports performance. Nitrates can the modification of skeletal muscle contractile proteins or calcium handling after translation. The time to reach the peak plasma nitrate is between 1 and 3 h after consumption of a single dose of nitrate. Nitrate is metabolized by conversion to nitrite and subsequently nitric oxide. Beetroot can have various effects on athletic performance through nitric oxide. Nitric oxide is an intracellular and extracellular messenger for regulating certain cellular functions and causes vasodilation of blood vessels and increases blood flow. Nitric oxide seems to be effective in improving athletic performance by increasing oxygen, glucose, and other nutrients for better muscle fueling. Nitric oxide plays the main role in anabolic hormones, modulates the release of several neurotransmitters and the major mediators of stress involved in the acute hypothalamic-pituitary-adrenal response to exercise. Beetroot is an important source of compounds such as ascorbic acid, carotenoids, phenolic acids, flavonoids, betaline, and highly active phenolics and has high antioxidant properties. Beetroot supplement provides an important source of dietary polyphenols and due to the many health benefits. Phytochemicals of Beetroot through signaling pathways inhibit inflammatory diseases. In this study, the mechanisms responsible for these effects were examined and the research in this regard was reviewed.
Collapse
Affiliation(s)
- Hamid Arazi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Rasht, Iran
| | - Ehsan Eghbali
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Rasht, Iran
| |
Collapse
|
148
|
Hao Y, Xing M, Gu X. Research Progress on Oxidative Stress and Its Nutritional Regulation Strategies in Pigs. Animals (Basel) 2021; 11:1384. [PMID: 34068057 PMCID: PMC8152462 DOI: 10.3390/ani11051384] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress refers to the dramatic increase in the production of free radicals in human and animal bodies or the decrease in the ability to scavenging free radicals, thus breaking the antioxidation-oxidation balance. Various factors can induce oxidative stress in pig production. Oxidative stress has an important effect on pig performance and healthy growth, and has become one of the important factors restricting pig production. Based on the overview of the generation of oxidative stress, its effects on pigs, and signal transduction pathways, this paper discussed the nutritional measures to alleviate oxidative stress in pigs, in order to provide ideas for the nutritional research of anti-oxidative stress in pigs.
Collapse
Affiliation(s)
| | | | - Xianhong Gu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.H.); (M.X.)
| |
Collapse
|
149
|
Wang X, Zimmermann HR, Lockhart SN, Craft S, Ma T. Decreased Levels of Blood AMPKα1 but not AMPKα2 Isoform in Patients with Mild Cognitive Impairment and Alzheimer's Disease: A Pilot Study. J Alzheimers Dis 2021; 76:217-224. [PMID: 32444538 DOI: 10.3233/jad-191189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is an urgent need to develop feasible biomarkers for diagnosis and prognosis of Alzheimer's disease (AD). Mounting evidence implicates that dysregulation of energy metabolism is a key and early event in AD pathogenesis. AMP-activated protein kinase (AMPK) is a central molecular sensor that plays a critical role in maintaining cellular energy homeostasis, and aberrant brain AMPK activities are linked to AD pathophysiology. OBJECTIVE We aimed to investigated protein levels of AMPKα isoforms, AMPKα1 and AMPKα2, in plasma samples from patients clinically diagnosed with mild cognitive impairment (MCI) or AD, along with age-matched healthy controls. METHODS 30 participants (10 MCI, 10 AD, and 10 controls) were included in our pilot study. Plasma levels of AMPKα1 and AMPKα2 were determined by ELISA. Receiver operating characteristic (ROC) analysis was used to assess sensitivity and specificity. Linear regression was used to assess the correlation between levels of AMPKα isoforms and other biomarkers. RESULTS Plasma levels of AMPKα1 were decreased in MCI and AD patients, while levels of AMPKα2 were unaltered as compared to controls. ROC analysis showed relatively high sensitivity and specificity for AMPKα1 to distinguish MCI and AD from controls. Linear regression analysis showed that plasma levels of AMPKα1 were correlated with a brain imaging biomarker (AD signature cortical thicknesses). CONCLUSION Plasma levels of AMPKα1 were decreased in MCI and AD patients. Future endeavor to explore whether blood AMPKα1 protein expression has the value as a potential biomarker for AD and MCI diagnosis shall be encouraged.
Collapse
Affiliation(s)
- Xin Wang
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Helena R Zimmermann
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Samuel N Lockhart
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Suzanne Craft
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tao Ma
- Department of Internal Medicine, Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Physiology and Pharmacology, and Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
150
|
van der Vaart JI, Boon MR, Houtkooper RH. The Role of AMPK Signaling in Brown Adipose Tissue Activation. Cells 2021; 10:cells10051122. [PMID: 34066631 PMCID: PMC8148517 DOI: 10.3390/cells10051122] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is becoming a pandemic, and its prevalence is still increasing. Considering that obesity increases the risk of developing cardiometabolic diseases, research efforts are focusing on new ways to combat obesity. Brown adipose tissue (BAT) has emerged as a possible target to achieve this for its functional role in energy expenditure by means of increasing thermogenesis. An important metabolic sensor and regulator of whole-body energy balance is AMP-activated protein kinase (AMPK), and its role in energy metabolism is evident. This review highlights the mechanisms of BAT activation and investigates how AMPK can be used as a target for BAT activation. We review compounds and other factors that are able to activate AMPK and further discuss the therapeutic use of AMPK in BAT activation. Extensive research shows that AMPK can be activated by a number of different kinases, such as LKB1, CaMKK, but also small molecules, hormones, and metabolic stresses. AMPK is able to activate BAT by inducing adipogenesis, maintaining mitochondrial homeostasis and inducing browning in white adipose tissue. We conclude that, despite encouraging results, many uncertainties should be clarified before AMPK can be posed as a target for anti-obesity treatment via BAT activation.
Collapse
Affiliation(s)
- Jamie I. van der Vaart
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Mariëtte R. Boon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Leiden University Medical Center, Einthoven Laboratory for Experimental Vascular Medicine, 2333 ZA Leiden, The Netherlands
- Correspondence: (M.R.B.); (R.H.H.)
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Correspondence: (M.R.B.); (R.H.H.)
| |
Collapse
|