1
|
Song X, Wang X, Gao Y, Xu G, Yan X, Chen Z, Song G. Exploring the Therapeutic Potential of Glycyrrhiza Compounds in Alzheimer's Disease: A Comprehensive Review. Curr Top Med Chem 2025; 25:286-310. [PMID: 39323338 DOI: 10.2174/0115680266322320240911194626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder. Research shows that the development of AD is linked to neuroinflammation, endoplasmic reticulum stress, mitochondrial dysfunction, cell death, and abnormal cholinergic signaling. Glycyrrhiza compounds contain active ingredients and extracts that offer multiple benefits, including targeting various pathways, high efficacy with low toxicity, and long-lasting therapeutic effects. These benefits highlight the significant potential of Glycyrrhiza compounds for preventing and treating AD. This review summarizes recent advancements in Glycyrrhiza compounds for preventing and treating AD. It focuses on their inhibitory effects on key signaling pathways, such as Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), and cholinergic signaling. This study aims to establish a scientific framework for using Glycyrrhiza compounds in the clinical prevention and treatment of AD and to support the development of new therapeutic interventions.
Collapse
Affiliation(s)
- Xiaona Song
- Department of Basic Medical Sciences, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
- Laboratory Animal Center, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
| | - Xiaotang Wang
- Department of Basic Medical Sciences, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
- Laboratory Animal Center, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
| | - Yao Gao
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, No. 85 Jiefang South Rd, Taiyuan, 030001, China
| | - Guoqiang Xu
- Department of Basic Medical Sciences, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
- Laboratory Animal Center, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
| | - Xiaoru Yan
- Department of Basic Medical Sciences, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
- Laboratory Animal Center, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
| | - Zhaoyang Chen
- Department of Basic Medical Sciences, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
- Laboratory Animal Center, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
| | - Guohua Song
- Department of Basic Medical Sciences, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
- Laboratory Animal Center, Shanxi Medical University, No. 56, Xinjian South Rd, Taiyuan, 030001, China
| |
Collapse
|
2
|
Zhou L, Chen S, Wei Y, Sun Y, Yang Y, Lin B, Li Y, Wang C. Glycyrrhizic acid restores the downregulated hepatic ACE2 signaling in the attenuation of mouse steatohepatitis. Eur J Pharmacol 2024; 967:176365. [PMID: 38316247 DOI: 10.1016/j.ejphar.2024.176365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Glycyrrhizic acid (GA), one of the major active components derived from licorice root, exerts liver-protecting activity. Its molecular mechanisms of action, however, remain not completely understood. The angiotensin (Ang) converting enzyme (ACE) 2/Ang-(1-7)/Mas axis, regulated by ACE2 through converting Ang II into Ang-(1-7) to activate Mas receptor, counteracts the pro-inflammatory and pro-steatotic effects of the ACE/Ang II/Ang II receptor type 1 (AT1) axis. Here, it was found that pretreatment with GA suppressed LPS/D-galactosamine-induced serum hyperactivities of alanine aminotransferase and aspartate aminotransferase, hepatomegaly, pathological changes, and over-accumulation of triglycerides and fatty droplets in the liver of mice. GA also diminished LPS/free fatty acid-induced inflammation and steatosis in cultured hepatocytes. Mechanistically, GA restored hepatic protein hypoexpression of ACE2 and Mas receptor, and the decrease in hepatic Ang-(1-7) content. Hepatic overexpression of angiotensin II and AT1 was also suppressed. However, GA did not alter hepatic protein expression of renin and ACE. In addition, GA inhibited hepatic protein over-phosphorylation of the p38 mitogen-activated protein kinase, c-Jun N-terminal kinase, extracellular signal-regulated kinase, and nuclear factor κB at Ser536. Hepatic overexpression of tumor necrosis factor α, interleukin 6, interleukin 1β, sterol regulatory element-binding protein 1c, and fatty acid synthase was also inhibited. GA-elicited recovery of ACE2 and Mas protein hypoexpression was further confirmed in the hepatocyte. Thus, the present results demonstrate that GA restores the downregulated hepatic ACE2-mediated anti-inflammatory and anti-steatotic signaling in the amelioration of steatohepatitis. We suggest that GA may protect the liver from injury by regulating the hepatic ACE2-mediated signaling.
Collapse
Affiliation(s)
- Longyue Zhou
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Shankang Chen
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuanyi Wei
- Department of Pharmacy, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Yihui Sun
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Yifan Yang
- Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, NSW, 2000, Australia.
| | - Bingqi Lin
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuhao Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, NSW, 2000, Australia.
| | - Chunxia Wang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Olloquequi J, Ettcheto M, Cano A, Fortuna A, Bicker J, Sánchez-Lopez E, Paz C, Ureña J, Verdaguer E, Auladell C, Camins A. Licochalcone A: A Potential Multitarget Drug for Alzheimer's Disease Treatment. Int J Mol Sci 2023; 24:14177. [PMID: 37762479 PMCID: PMC10531537 DOI: 10.3390/ijms241814177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Licochalcone A (Lico-A) is a flavonoid compound derived from the root of the Glycyrrhiza species, a plant commonly used in traditional Chinese medicine. While the Glycyrrhiza species has shown promise in treating various diseases such as cancer, obesity, and skin diseases due to its active compounds, the investigation of Licochalcone A's effects on the central nervous system and its potential application in Alzheimer's disease (AD) treatment have garnered significant interest. Studies have reported the neuroprotective effects of Lico-A, suggesting its potential as a multitarget compound. Lico-A acts as a PTP1B inhibitor, enhancing cognitive activity through the BDNF-TrkB pathway and exhibiting inhibitory effects on microglia activation, which enables mitigation of neuroinflammation. Moreover, Lico-A inhibits c-Jun N-terminal kinase 1, a key enzyme involved in tau phosphorylation, and modulates the brain insulin receptor, which plays a role in cognitive processes. Lico-A also acts as an acetylcholinesterase inhibitor, leading to increased levels of the neurotransmitter acetylcholine (Ach) in the brain. This mechanism enhances cognitive capacity in individuals with AD. Finally, Lico-A has shown the ability to reduce amyloid plaques, a hallmark of AD, and exhibits antioxidant properties by activating the nuclear factor erythroid 2-related factor 2 (Nrf2), a key regulator of antioxidant defense mechanisms. In the present review, we discuss the available findings analyzing the potential of Lico-A as a neuroprotective agent. Continued research on Lico-A holds promise for the development of novel treatments for cognitive disorders and neurodegenerative diseases, including AD. Further investigations into its multitarget action and elucidation of underlying mechanisms will contribute to our understanding of its therapeutic potential.
Collapse
Affiliation(s)
- Jordi Olloquequi
- Departament of Biochemistry and Physiology, Physiology Section, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Av. Joan XXIII 27/31, 08028 Barcelona, Spain
- Laboratory of Cellular and Molecular Pathology, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| | - Miren Ettcheto
- Departament of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain; (M.E.); (A.C.)
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (A.C.); (E.S.-L.); (J.U.); (E.V.); (C.A.)
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Reus, Spain
| | - Amanda Cano
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (A.C.); (E.S.-L.); (J.U.); (E.V.); (C.A.)
- Ace Alzheimer Center Barcelona, International University of Catalunya (UIC), 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), 08028 Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Ana Fortuna
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (J.B.)
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), 3000-548 Coimbra, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (J.B.)
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), 3000-548 Coimbra, Portugal
| | - Elena Sánchez-Lopez
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (A.C.); (E.S.-L.); (J.U.); (E.V.); (C.A.)
- Institute of Nanoscience and Nanotechnology (IN2UB), 08028 Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
- Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034 Barcelona, Spain
| | - Cristian Paz
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Jesús Ureña
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (A.C.); (E.S.-L.); (J.U.); (E.V.); (C.A.)
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (A.C.); (E.S.-L.); (J.U.); (E.V.); (C.A.)
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (A.C.); (E.S.-L.); (J.U.); (E.V.); (C.A.)
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Antoni Camins
- Departament of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain; (M.E.); (A.C.)
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (A.C.); (E.S.-L.); (J.U.); (E.V.); (C.A.)
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Reus, Spain
| |
Collapse
|
4
|
Matveevskaya VV, Pavlov DI, Kovrizhina AR, Sukhikh TS, Sadykov EH, Dorovatovskii PV, Lazarenko VA, Khlebnikov AI, Potapov AS. Experimental and Computational Investigation of the Oxime Bond Stereochemistry in c-Jun N-terminal Kinase 3 Inhibitors 11 H-Indeno[1,2- b]quinoxalin-11-one Oxime and Tryptanthrin-6-oxime. Pharmaceutics 2023; 15:1802. [PMID: 37513989 PMCID: PMC10383563 DOI: 10.3390/pharmaceutics15071802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
11H-Indeno[1,2-b]quinoxalin-11-one oxime (IQ-1) and tryptanthrin-6-oxime are potent c-Jun N-terminal kinase 3 (JNK-3) inhibitors demonstrating neuroprotective, anti-inflammatory and anti-arthritic activity. However, the stereochemical configuration of the oxime carbon-nitrogen double bond (E- or Z-) in these compounds was so far unknown. In this contribution, we report the results of the determination of the double bond configuration in the solid state by single crystal X-ray diffraction and in solution by 1D and 2D NMR techniques and DFT calculations. It was found that both in the solid state and in solution, IQ-1 adopts the E-configuration stabilized by intermolecular hydrogen bonds, in contrast to previously assumed Z-configuration that could be stabilized only by an intramolecular hydrogen bond.
Collapse
Affiliation(s)
- Vladislava V Matveevskaya
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch of the Russian Academy of Sciences, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Dmitry I Pavlov
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch of the Russian Academy of Sciences, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Anastasia R Kovrizhina
- Kizhner Research Center, National Research Tomsk Polytechnic University, 30 Lenin Ave., 634050 Tomsk, Russia
| | - Taisiya S Sukhikh
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch of the Russian Academy of Sciences, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Evgeniy H Sadykov
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch of the Russian Academy of Sciences, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Pavel V Dorovatovskii
- National Research Centre "Kurchatov Institute", Kurchatov Square 1, 123182 Moscow, Russia
| | - Vladimir A Lazarenko
- National Research Centre "Kurchatov Institute", Kurchatov Square 1, 123182 Moscow, Russia
| | - Andrei I Khlebnikov
- Kizhner Research Center, National Research Tomsk Polytechnic University, 30 Lenin Ave., 634050 Tomsk, Russia
| | - Andrei S Potapov
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch of the Russian Academy of Sciences, 3 Lavrentiev Ave., 630090 Novosibirsk, Russia
| |
Collapse
|
5
|
El-Bana MA, Ashour MN, Rasheed WI, Diab YM, Medhat D. Bombax ceiba Linn. leaf extract rich in phenolic compounds to mitigate non-alcoholic fatty liver-related complications in experimental model. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2023; 20:343-352. [PMID: 36935561 DOI: 10.1515/jcim-2021-0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 02/24/2023] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Obesity, diabetes mellitus, insulin resistance (IR), and hypertriglyceridemia are common features observed in non-alcoholic fatty liver diseases (NAFLD). There is a critical medical necessity to find novel therapeutics that can halt the development of NAFLD. METHODS Bombax ceiba Linn. leaf extract was prepared and its phytochemical profile was determined. Standard and high carbohydrate high-fat diets (HCHF) were prepared. Rats were fed HCHF for 18 weeks to induce a non-alcoholic fatty liver (NAFL) model. Forty male rats were divided into control, B. ceiba Linn. leaf extract, NAFL, prophylactic, and treated groups. Serum fasting blood sugar (FBS), insulin, insulin resistance (HOMA-IR), cholesterol, high-density lipoprotein (HDL), triglycerides (TG), low density lipoprotein (LDL), alanine aminotransferase (ALT), aspartate aminotransferase (AST), intelectin-1 (ITLN1), p38 MAP kinase (MAPK), peroxisome proliferator-activated receptor alpha (PPAR-α), and interleukin-6 (IL-6) were evaluated. RESULTS Data obtained showed that HCHF-induced NAFL resulting in a significant increase in FBS, serum insulin, HOMA-IR, cholesterol, LDL, TG, ALT, AST, and IL-6 and a significant decrease in serum levels of HDL, ITLN1, p38 MAP kinase, and PPAR-α compared to the control group. The analysis of B. ceiba Linn. leaf extract showed high content of phenol compounds which may cause a significant decrease in the levels of FBS, insulin, HOMA-IR values, lipid profile, and levels of IL-6 while a significant increase in serum levels of LDL, ITLN1, p38 MAP kinase, and PPAR-α compared to the NAFL group. CONCLUSIONS B. ceiba Linn. Leaf extract is a highly protective and promising therapeutic agent against inflammation and oxidative stress in the NAFLD model induced by HCHF.
Collapse
Affiliation(s)
- Mona A El-Bana
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Magdi N Ashour
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Wafaa I Rasheed
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Yasser M Diab
- Department of Biochemistry, Faculty of Agriculture, Fayoum University, Fayoum, Egypt
| | - Dalia Medhat
- Department of Medical Biochemistry, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
6
|
Kabeer SW, Pant R, Sharma S, Tikoo K. Laccaic acid restores epigenetic alterations responsible for high fat diet induced insulin resistance in C57BL/6J mice. Chem Biol Interact 2023; 374:110401. [PMID: 36828244 DOI: 10.1016/j.cbi.2023.110401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/10/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Laccaic acid, the major constituent of the food colouring agent-lac dye, possesses antioxidant and anti-inflammatory properties. Here we have evaluated the effects of laccaic acid on the high-fat diet induced insulin resistance in C57BL/6J mice. Insulin resistance was developed in mice by feeding high-fat diet for 12 weeks. 6 week treatment with laccaic acid showed significant improvement in the morphometric, biochemical parameters and liver function. Western blotting experiments showed, laccaic acid increased phosphorylation of IRS1/2/AKT/GSK3β which is suppressed under insulin-resistant conditions in liver. Furthermore, it also attenuated the inflammatory ERK/NFκB signalling, thereby reducing the expression of inflammatory cytokines- TNFα, IL-1β and IL-6. Concomitantly, laccaic acid increased AMPK/AKT-mediated phosphorylation of FOXO1, preventing its nuclear translocation and transcriptional activation of gluconeogenic genes (G6PC and PCK1). Interestingly, treatment with laccaic acid also prevented high-fat diet induced alterations of histone methylation (H3K27me3 and H3K36me2) at global level. Our chromatin-immunoprecipitation data shows high-fat diet induced loss of inactivation mark H3K27me3 at FOXO1 promoter was regained upon laccaic acid treatment. Additionally, the expression of the H3K27 methylating enzyme EZH2 was also upregulated by laccaic acid. Together it all results in the downregulation of FOXO1 gene expression. To the best of our knowledge, we provide first evidence that laccaic acid either directly or indirectly modulates the epigenetic landscape of genes responsible for high-fat diet induced insulin resistance.
Collapse
Affiliation(s)
- Shaheen Wasil Kabeer
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India
| | - Rajat Pant
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India
| | - Shivam Sharma
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India
| | - Kulbhushan Tikoo
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India.
| |
Collapse
|
7
|
Lahmi A, Oryan S, Eidi A, Rohani AH. Comparative effects of thymol and vitamin E on nonalcoholic fatty liver disease in male Wistar rats. BRAZ J BIOL 2023; 84:e268781. [PMID: 36629640 DOI: 10.1590/1519-6984.268781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 01/10/2023] Open
Abstract
Following the obesity epidemics, nonalcoholic fatty liver disease (NAFLD) has grown in prevalence and become a main cause of morbidity and death, intimately linked to cardiovascular disease, cancer, and cirrhosis. The key factor in the evolution of NAFLD is thought to be oxidative stress. Because most patients cannot change their lifestyle or dietary habits, a pharmaceutical strategy is now required to treat NAFLD. Nonalcoholic fatty liver disease (NAFLD), including nonalcoholic steatohepatitis, is treated with vitamin E. (NASH). Vitamin E is also a powerful antioxidant that has been demonstrated to lower oxidative stress in people with NAFLD. Thymol is a monoterpene phenol with a variety of pharmacological effects, however its anti-fatty liver properties have yet to be investigated. Despite the fact that oxidative stress is thought to have a role in the etiology of nonalcoholic steatohepatitis, antioxidant therapies have not been well studied in the treatment of nonalcoholic steatohepatitis. The goal was to learn more about vitamin E and thymol's biological activities, with a particular emphasis on their therapeutic effectiveness in NAFLD. Four groups of thirty-two adult male rats were formed (healthy control, thymol, Vit E, and fatty liver). For 28 days, rats were given either oral vitamin E (200 mg/kg) or thymol (50 mg/kg) randomly. The levels of ALT, AST, TNF- α, Ferritin, CK-MB enzymes, and MAPK gene expression were then determined in the serum. Based on a random effect model analysis, at the end of 28 days of therapy, ALT (41.43 U/L), AST (47.91 U/L), Ferritin (1.13 pg/dl), CK-MB (251.22 IU/L), TNF-α (95.39 pg/mL) (p≤0.001), and MAPK gene expression levels (p≤0.05) significantly reduced in both experimental groups compared with the fatty liver group. Vitamin E and thymol therapy is a safe, affordable, and effective therapeutic option in the fatty liver group. Patients with fatty liver disease should be encouraged to take vitamin E and Thymol supplements, which are both safe and affordable, because more effective new therapeutic options are lacking.
Collapse
Affiliation(s)
- A Lahmi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - S Oryan
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Biological Science Faculty, Animal Physiology Department, Kharazmi University, Tehran,Iran
| | - A Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - A H Rohani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
8
|
Wu T, Xie Y, Wu Z, Li Y, Jiang M, Yu H, Li X, Wang J, Zhou E, Yang Z. β-Carotene Protects Mice against Lipopolysaccharide and D-Galactosamine Induced Acute Liver Injury via Regulation of NF-κB, MAPK, and Nrf2 Signaling. J Oleo Sci 2023; 72:1027-1035. [PMID: 37914264 DOI: 10.5650/jos.ess23100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
Acute liver injury (ALI), posing a serious threaten to our life, has emerged as a public health issue around the world. β-carotene has plenty of pharmacologic effects, such as anti-inflammatory, antioxidant, and antitumor activities. In this study, we focused on studying the protective role and potential molecular mechanisms of β-carotene against D-galactosamine (D-GalN) and lipopolysaccharide (LPS) induced ALI. Our results indicated that β-carotene pretreatment effectively hindered abnormal changes induced by LPS/D-GalN in liver histopathology. Meanwhile, serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were downgraded with β-carotene pretreatment. β-carotene pretreatment also decreased malondialdehyde content and myeloperoxidase activity, increased glutathione peroxidase and superoxide dismutase levels, and reduced the levels of tumor necrosis factor-a (TNF-α) and interleukin 6 (IL-6) in liver tissues. Further investigations found that β-carotene mediated multiple signaling pathways in LPS/D-GalN-induced ALI, inhibiting NF-κB and MAPK signaling and upregulating the expression of Nrf2 and HO-1 proteins. All findings indicate that β-carotene appears to protect mice against LPS/D-GalN induced ALI by reducing oxidative stress and inflammation, possibly via regulating NF-κB, MAPK, and Nrf2 signaling.
Collapse
Affiliation(s)
- Ting Wu
- College of Life Sciences and Engineering, Foshan University
| | - Yueqing Xie
- College of Life Sciences and Engineering, Foshan University
| | - Zhikai Wu
- College of Life Sciences and Engineering, Foshan University
| | - Yifei Li
- College of Life Sciences and Engineering, Foshan University
| | - Mingzhen Jiang
- College of Life Sciences and Engineering, Foshan University
| | - Hongsen Yu
- College of Life Sciences and Engineering, Foshan University
| | - Xuhai Li
- College of Life Sciences and Engineering, Foshan University
| | - Jingjing Wang
- College of Life Sciences and Engineering, Foshan University
| | - Ershun Zhou
- College of Life Sciences and Engineering, Foshan University
| | - Zhengtao Yang
- College of Life Sciences and Engineering, Foshan University
| |
Collapse
|
9
|
Wang J, Wu S, Zhan H, Bi W, Xu Y, Liang Y, Ge Y, Peng L, Jin X, Lu K, Zhao J, Gao L, He Z. p38α in the preoptic area inhibits brown adipose tissue thermogenesis. Obesity (Silver Spring) 2022; 30:2242-2255. [PMID: 36321273 DOI: 10.1002/oby.23552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/06/2022] [Accepted: 07/21/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Elevation of energy expenditure through an increase of brown adipose tissue (BAT) thermogenesis is regarded as one of the most promising ways to prevent obesity development. The preoptic area (POA) of the hypothalamus is a critical area for control of BAT thermogenesis. However, the intracellular signaling cascades in the POA for regulation of BAT thermogenesis are poorly understood. METHODS Phosphorylation proteomics (phosphoproteomics) and bioinformatics approaches were used to disclose numerous hypothalamic signaling pathways involved in the regulation of BAT thermogenesis. Conditional manipulation of the p38α gene in mouse POA was performed by stereotaxic injection of adeno-associated virus 9 vector to explore the role of p38α in BAT thermogenesis. RESULTS Multiple hypothalamic signaling pathways were triggered by cold exposure, especially the mitogen-activated protein kinase (MAPK) signaling pathway. The p38α activation, but not extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun NH2-terminal kinase (JNK), in the hypothalamus was significantly decreased during cold exposure. p38α deficiency in the POA dramatically elevated energy expenditure owing to a marked increase in BAT thermogenesis, resulting in significantly decreased body weight gain and fat mass. Overexpression of p38α in the POA led to a dramatic increase in weight gain. CONCLUSIONS These results demonstrate that p38α in the POA exacerbates obesity development, at least in part owing to a decrease in BAT thermogenesis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Shanshan Wu
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Huidong Zhan
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Wenkai Bi
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Yang Xu
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Yixiao Liang
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Yueping Ge
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Li Peng
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Xinchen Jin
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
| | - Keke Lu
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiajun Zhao
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ling Gao
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhao He
- Department of Endocrinology, Medical Integration and Practice Center & Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
10
|
Shao Y, Tian J, Yang Y, Hu Y, Zhu Y, Shu Q. Identification of key genes and pathways revealing the central regulatory mechanism of brain-derived glucagon-like peptide-1 on obesity using bioinformatics analysis. Front Neurosci 2022; 16:931161. [PMID: 35992905 PMCID: PMC9389235 DOI: 10.3389/fnins.2022.931161] [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: 04/28/2022] [Accepted: 07/07/2022] [Indexed: 12/01/2022] Open
Abstract
Objective Central glucagon-like peptide-1 (GLP-1) is a target in treating obesity due to its effect on suppressing appetite, but the possible downstream key genes that GLP-1 regulated have not been studied in depth. This study intends to screen out the downstream feeding regulation genes of central GLP-1 neurons through bioinformatics analysis and verify them by chemical genetics, which may provide insights for future research. Materials and methods GSE135862 genetic expression profiles were extracted from the Gene Expression Omnibus (GEO) database. The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) enrichment analyses were carried out. STRING database and Cytoscape software were used to map the protein-protein interaction (PPI) network of the differentially expressed genes (DEGs). After bioinformatics analysis, we applied chemogenetic methods to modulate the activities of GLP-1 neurons in the nucleus tractus solitarius (NTS) and observed the alterations of screened differential genes and their protein expressions in the hypothalamus under different excitatory conditions of GLP-1 neurons. Results A total of 49 DEGs were discovered, including 38 downregulated genes and 11 upregulated genes. The two genes with the highest expression scores were biglycan (Bgn) and mitogen-activated protein kinase activated protein kinase 3 (Mapkapk3). The results of GO analysis showed that there were 10 molecular functions of differential genes. Differential genes were mainly localized in seven regions around the cells, and enriched in 10 biology processes. The results of the KEGG signaling pathway enrichment analysis showed that differential genes played an important role in seven pathways. The top 15 genes selected according to the Cytoscape software included Bgn and Mapkapk3. Chemogenetic activation of GLP-1 in NTS induced a decrease in food intake and body mass, while chemogenetic inhibition induced the opposite effect. The gene and protein expression of GLP-1 were upregulated in NTS when activated by chemogenetics. In addition, the expression of Bgn was upregulated and that of Mapkapk3 was downregulated in the hypothalamus. Conclusion Our data showed that GLP-1 could modulate the protein expression of Bgn and Mapkapk3. Our findings elucidated the regulatory network in GLP-1 to obesity and might provide a novel diagnostic and therapeutic target for obesity.
Collapse
Affiliation(s)
- Yuwei Shao
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Tian
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanan Yang
- Department of Traditional Chinese Medicine, China Resources Wugang General Hospital, Wuhan, China
| | - Yan Hu
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ye Zhu
- College of Health Sciences, Wuhan Sports University, Wuhan, China
| | - Qing Shu
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Qing Shu,
| |
Collapse
|
11
|
Zhao Y, Dai J, Jiang Y, Wu H, Cui Y, Li X, Mao H, Wang B, Ju S, Peng XG. Reducing White Adipose Tissue Browning Using p38α MAPK Inhibitors Ameliorates Cancer-Associated Cachexia as Assessed by Magnetic Resonance Imaging. Nutrients 2022; 14:nu14153013. [PMID: 35893867 PMCID: PMC9331061 DOI: 10.3390/nu14153013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Up to 80% of pancreatic cancer patients suffer from cachexia. White adipose tissue (WAT) browning caused by the tumorigenicity and progression aggravates the cancer-associated cachexia (CAC). Cancer-initiated changes in the protein-38 mitogen-activated protein kinases (p38 MAPK) pathway are likely involved in the development of CAC. Methods: p38 MAPK inhibitors, VCP979 or SB203580, were used in the in vitro and in vivo models of pancreatic cancer cachexia. Expression of uncoupling protein 1 (UCP1) in the p38 MARK pathway and the properties and level of white adipocytes were analyzed and correlated to browning, followed by immunohistochemistry and Western blotting validations. Changes in the volume and fat fraction of WAT in animals were monitored by magnetic resonance imaging (MRI). Results: The size of white adipocytes was increased after being treated with the p38 MAPK inhibitors, along with increase in the MRI-measured volume and fat fraction of WAT. Comparing two p38 MAPK inhibitors, the p38α subunit-specific inhibitor VCP979 had a better therapeutic effect than SB203580, which targets both p38α and β subunits. Conclusions: Blockade of p38 MAPK reduced the WAT browning that contributes to CAC. Thus, p38 MARK inhibitors can potentially be used as a therapy for treating CAC. Non-invasive MRI can also be applied to assess the progression and treatment responses of CAC.
Collapse
Affiliation(s)
- Yufei Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
| | - Jingyue Dai
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
| | - Yang Jiang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
| | - Honghong Wu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
| | - Ying Cui
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
| | - Xinxiang Li
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30329, USA;
| | - Binghui Wang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia;
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
| | - Xin-Gui Peng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; (Y.Z.); (J.D.); (Y.J.); (H.W.); (Y.C.); (X.L.); (S.J.)
- People’s Hospital of Lishui District, 86 Chongwen Road, Yongyang Town, Lishui District, Nanjing 211299, China
- Correspondence: ; Tel.: +86-025-83272115
| |
Collapse
|
12
|
Metabolic Impact of MKP-2 Upregulation in Obesity Promotes Insulin Resistance and Fatty Liver Disease. Nutrients 2022; 14:nu14122475. [PMID: 35745205 PMCID: PMC9228271 DOI: 10.3390/nu14122475] [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: 04/13/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
The mechanisms connecting obesity with type 2 diabetes, insulin resistance, nonalcoholic fatty liver disease, and cardiovascular diseases remain incompletely understood. The function of MAPK phosphatase-2 (MKP-2), a type 1 dual-specific phosphatase (DUSP) in whole-body metabolism, and how this contributes to the development of diet-induced obesity, type 2 diabetes (T2D), and insulin resistance is largely unknown. We investigated the physiological contribution of MKP-2 in whole-body metabolism and whether MKP-2 is altered in obesity and human fatty liver disease using MKP-2 knockout mice models and human liver tissue derived from fatty liver disease patients. We demonstrate that, for the first time, MKP-2 expression was upregulated in liver tissue in humans with obesity and fatty liver disease and in insulin-responsive tissues in mice with obesity. MKP-2-deficient mice have enhanced p38 MAPK, JNK, and ERK activities in insulin-responsive tissues compared with wild-type mice. MKP-2 deficiency in mice protects against diet-induced obesity and hepatic steatosis and was accompanied by improved glucose homeostasis and insulin sensitivity. Mkp-2−/− mice are resistant to diet-induced obesity owing to reduced food intake and associated lower respiratory exchange ratio. This was associated with enhanced circulating insulin-like growth factor-1 (IGF-1) and stromal cell-derived factor 1 (SDF-1) levels in Mkp-2−/− mice. PTEN, a negative regulator of Akt, was downregulated in livers of Mkp-2−/− mice, resulting in enhanced Akt activity consistent with increased insulin sensitivity. These studies identify a novel role for MKP-2 in the regulation of systemic metabolism and pathophysiology of obesity-induced insulin resistance and fatty liver disease.
Collapse
|
13
|
Ficus hirta Vahl. Ameliorates Nonalcoholic Fatty Liver Disease through Regulating Lipid Metabolism and Gut Microbiota. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3474723. [PMID: 35592528 PMCID: PMC9113867 DOI: 10.1155/2022/3474723] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has gradually become one of the most serious liver diseases threatening human health in the world. Currently, Chinese herbal medicine is a potentially important treatment option for NAFLD, and the development of effective Chinese herbal medicine has a good prospect. Previous studies have suggested that Ficus hirta Vahl. (FV) has various protective effects on the liver. In this study, we investigated the therapeutic outcomes of FV treatment for the liver disease and its underlying mechanism using HepG2 cell lines induced by palmitate (PA) and mouse model fed with high-fat diet (HFD). FV mainly exerts pharmacological effects by mediating lipid metabolism and inflammation. During the lipid metabolism regulation process, CD36, SREBP-1, SCD1, PPAR γ, ACOX1, and CPT1α are the key factors related to the healing effects of FV on NAFLD. During the inflammation process, the downregulation of IL-6, IL-1β, and TNF-α is involved in alleviation of NAFLD. Furthermore, CD36 overexpression promotes lipid abnormal metabolism and inflammation in PA-induced HepG2 cells, while CD36 knockdown and FV supplementation reverse these responses. In addition, FV also modulates gut microbiota composition, such as Allobaculum, Faecalibaculum, and Butyricicoccus in HFD-fed mice. In summary, our findings demonstrated that FV exerted a beneficial preventive and therapeutic effect on NAFLD by improving lipid metabolism and inflammation as well as regulating the structure of gut microbiota, and therefore, FV may be a candidate for the treatment of NAFLD.
Collapse
|
14
|
Díaz-Chamorro S, Garrido-Jiménez S, Barrera-López JF, Mateos-Quirós CM, Cumplido-Laso G, Lorenzo MJ, Román ÁC, Bernardo E, Sabio G, Carvajal-González JM, Centeno F. Title: p38δ Regulates IL6 Expression Modulating ERK Phosphorylation in Preadipocytes. Front Cell Dev Biol 2022; 9:708844. [PMID: 35111744 PMCID: PMC8802314 DOI: 10.3389/fcell.2021.708844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
IL6 is an essential cytokine in metabolism regulation and for intercommunication among different organs and tissues. IL6 produced by different tissues has different functions and therefore it is very important to understand the mechanism of its expression in adipose tissue. In this work we demonstrated that IL6 expression in mouse preadipocytes, like in human, is partially dependent on Wnt5a and JNK. Using mouse preadipocytes lacking each one of the p38 SAPK family members, we have shown that IL6 expression is also p38γ and p38δ dependent. In fact, the lack of some of these two kinases increases IL6 expression without altering that of Wnt5a. Moreover, we show that the absence of p38δ promotes greater ERK1/2 phosphorylation in a MEK1/2 independent manner, and that this increased ERK1/2 phosphorylation state is contributing to the higher IL6 expression in p38δ−/- preadipocytes. These results suggest a new crosstalk between two MAPK signaling pathway, p38δ and ERK1/2, where p38δ modulates the phosphorylation state of ERK1/2.
Collapse
Affiliation(s)
- Selene Díaz-Chamorro
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Sergio Garrido-Jiménez
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Juan Francisco Barrera-López
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Clara María Mateos-Quirós
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Guadalupe Cumplido-Laso
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - María Jesús Lorenzo
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Cáceres, Spain
| | - Ángel Carlos Román
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Edgar Bernardo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - José María Carvajal-González
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Francisco Centeno
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| |
Collapse
|
15
|
Nellaiappan K, Preeti K, Khatri DK, Singh SB. Diabetic Complications: An Update on Pathobiology and Therapeutic Strategies. Curr Diabetes Rev 2022; 18:e030821192146. [PMID: 33745424 DOI: 10.2174/1573399817666210309104203] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/28/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022]
Abstract
Despite the advent of novel therapies which manage and control diabetes well, the increased risk of morbidity and mortality in diabetic subjects is associated with the devastating secondary complications it produces. Long-standing diabetes majorly drives cellular and molecular alterations, which eventually damage both small and large blood vessels. The complications are prevalent both in type I and type II diabetic subjects. The microvascular complications include diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, while the macrovascular complications include diabetic heart disease and stroke. The current therapeutic strategy alleviates the complications to some extent but does not cure or prevent them. Also, the recent clinical trial outcomes in this field are disappointing. Success in the drug discovery of diabetic complications may be achieved by a better understanding of the underlying pathophysiology and by recognising the crucial factors contributing to the development and progression of the disease. In this review, we discuss the well-studied cellular mechanisms leading to the development and progression of diabetic complications. In addition, we also highlight the various therapeutic paradigms currently in clinical practice.
Collapse
Affiliation(s)
- Karthika Nellaiappan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| | - Kumari Preeti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| | - Shashi Bala Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana-500037,India
| |
Collapse
|
16
|
Dark and bright side of targeting fibroblast growth factor receptor 4 in the liver. J Hepatol 2021; 75:1440-1451. [PMID: 34364916 DOI: 10.1016/j.jhep.2021.07.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factor (FGF) receptor 4 (FGFR4) and its cognate ligand, FGF19, are implicated in a range of cellular processes, including differentiation, metabolism and proliferation. Indeed, their aberrant activation has been associated with the development of hepatic tumours. Despite great advances in early diagnosis and the development of new therapies, liver cancer is still associated with a high mortality rate, owing primarily to high molecular heterogeneity and unclear molecular targeting. The development of FGFR4 inhibitors is a promising tool in patients with concomitant supraphysiological levels of FGF19 and several clinical trials are testing these treatments for patients with advanced hepatocellular carcinoma (HCC). Conversely, using FGF19 analogues to activate FGFR4-KLOTHO β represents a novel therapeutic strategy in patients presenting with cholestatic liver disorders and non-alcoholic steatohepatitis, which could potentially prevent the development of metabolic HCC. Herein, we provide an overview of the currently available therapeutic options for targeting FGFR4 in HCC and other liver diseases, highlighting the need to carefully stratify patients and personalise therapeutic strategies.
Collapse
|
17
|
Santamans AM, Montalvo-Romeral V, Mora A, Lopez JA, González-Romero F, Jimenez-Blasco D, Rodríguez E, Pintor-Chocano A, Casanueva-Benítez C, Acín-Pérez R, Leiva-Vega L, Duran J, Guinovart JJ, Jiménez-Borreguero J, Enríquez JA, Villlalba-Orero M, Bolaños JP, Aspichueta P, Vázquez J, González-Terán B, Sabio G. p38γ and p38δ regulate postnatal cardiac metabolism through glycogen synthase 1. PLoS Biol 2021; 19:e3001447. [PMID: 34758018 PMCID: PMC8612745 DOI: 10.1371/journal.pbio.3001447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/24/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
During the first weeks of postnatal heart development, cardiomyocytes undergo a major adaptive metabolic shift from glycolytic energy production to fatty acid oxidation. This metabolic change is contemporaneous to the up-regulation and activation of the p38γ and p38δ stress-activated protein kinases in the heart. We demonstrate that p38γ/δ contribute to the early postnatal cardiac metabolic switch through inhibitory phosphorylation of glycogen synthase 1 (GYS1) and glycogen metabolism inactivation. Premature induction of p38γ/δ activation in cardiomyocytes of newborn mice results in an early GYS1 phosphorylation and inhibition of cardiac glycogen production, triggering an early metabolic shift that induces a deficit in cardiomyocyte fuel supply, leading to whole-body metabolic deregulation and maladaptive cardiac pathogenesis. Notably, the adverse effects of forced premature cardiac p38γ/δ activation in neonate mice are prevented by maternal diet supplementation of fatty acids during pregnancy and lactation. These results suggest that diet interventions have a potential for treating human cardiac genetic diseases that affect heart metabolism.
Collapse
Affiliation(s)
| | | | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Juan Antonio Lopez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco González-Romero
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Daniel Jimenez-Blasco
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, CSIC, Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | | | - Rebeca Acín-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Joan J. Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | | | - José Antonio Enríquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - María Villlalba-Orero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan P. Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, CSIC, Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
- BioCruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
18
|
Nordgaard C, Tollenaere MAX, Val AMD, Bekker-Jensen DB, Blasius M, Olsen JV, Bekker-Jensen S. Regulation of the Golgi Apparatus by p38 and JNK Kinases during Cellular Stress Responses. Int J Mol Sci 2021; 22:9595. [PMID: 34502507 PMCID: PMC8431686 DOI: 10.3390/ijms22179595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/20/2021] [Accepted: 08/31/2021] [Indexed: 12/15/2022] Open
Abstract
p38 and c-Jun N-terninal kinase (JNK) are activated in response to acute stress and inflammatory signals. Through modification of a plethora of substrates, these kinases profoundly re-shape cellular physiology for the optimal response to a harmful environment and/or an inflammatory state. Here, we utilized phospho-proteomics to identify several hundred substrates for both kinases. Our results indicate that the scale of signaling from p38 and JNK are of a similar magnitude. Among the many new targets, we highlight the regulation of the transcriptional regulators grb10-interacting GYF protein 1 and 2 (GIGYF1/2) by p38-dependent MAP kinase-activated protein kinase 2 (MK2) phosphorylation and 14-3-3 binding. We also show that the Golgi apparatus contains numerous substrates, and is a major target for regulation by p38 and JNK. When activated, these kinases mediate structural rearrangement of the Golgi apparatus, which positively affects protein flux through the secretory system. Our work expands on our knowledge about p38 and JNK signaling with important biological ramifications.
Collapse
Affiliation(s)
- Cathrine Nordgaard
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (C.N.); (M.A.X.T.); (M.B.)
| | - Maxim A. X. Tollenaere
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (C.N.); (M.A.X.T.); (M.B.)
- LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Ana Martinez Del Val
- Mass Spectrometry for Quantitative Proteomics, Proteomics Program, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (A.M.D.V.); (D.B.B.-J.); (J.V.O.)
| | - Dorte B. Bekker-Jensen
- Mass Spectrometry for Quantitative Proteomics, Proteomics Program, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (A.M.D.V.); (D.B.B.-J.); (J.V.O.)
| | - Melanie Blasius
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (C.N.); (M.A.X.T.); (M.B.)
| | - Jesper V. Olsen
- Mass Spectrometry for Quantitative Proteomics, Proteomics Program, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (A.M.D.V.); (D.B.B.-J.); (J.V.O.)
| | - Simon Bekker-Jensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (C.N.); (M.A.X.T.); (M.B.)
| |
Collapse
|
19
|
Burillo J, Marqués P, Jiménez B, González-Blanco C, Benito M, Guillén C. Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease. Cells 2021; 10:1236. [PMID: 34069890 PMCID: PMC8157600 DOI: 10.3390/cells10051236] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus is a progressive disease that is characterized by the appearance of insulin resistance. The term insulin resistance is very wide and could affect different proteins involved in insulin signaling, as well as other mechanisms. In this review, we have analyzed the main molecular mechanisms that could be involved in the connection between type 2 diabetes and neurodegeneration, in general, and more specifically with the appearance of Alzheimer's disease. We have studied, in more detail, the different processes involved, such as inflammation, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jesús Burillo
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Patricia Marqués
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Beatriz Jiménez
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos González-Blanco
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| |
Collapse
|
20
|
Kang S, Lee M. Beiging Modulates Inflammatory Adipogenesis in Salt-Treated and MEK6-Transfected Adipocytes. Cells 2021; 10:cells10051106. [PMID: 34064531 PMCID: PMC8147997 DOI: 10.3390/cells10051106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/21/2022] Open
Abstract
To investigate whether the beiging process changes the interactive effects of salt and MEK6 gene on inflammatory adipogenesis, the salt treatment (NaCl 50 mM) and MEK6 transfection of Tg(+/+) cells were performed with white adipocytes (WAT) and beige-like-adipocytes (BLA). BLA induced by T3 were confirmed by UCP-1 expression and the MEK6 protein was 3.5 times higher in MEK6 transfected WAT than the control. The adipogenic genes, PPAR-γ and C/EBP-α, were 1.5 times more highly expressed in the salt-treated groups than the non-salt-treated groups, and adipogenesis was greatly increased in Tg(+/+) WAT compared to non-transfected Tg(−/−). The adipogenesis induced by salt treatment and MEK6 transfection was significantly reduced in BLA. The inflammatory adipocytokines, TNF-α, IL-1β, and IL-6, were increased in the salt-treated Tg(+/+) WAT, but an anti-inflammation biomarker, the adiponectin/leptin ratio, was reduced in Tg(+/+), to tenth of that in Tg(−/−). However, the production of adipocytokines in WAT was strongly weakened in BLA, although a combination of salt and MEK6 transfection had the most significant effects on inflammation in both WAT and BLA. Oxygen consumption in mitochondria was maximized in salt-treated and MEK6 transfected WAT, but it was decreased by 50% in BLA. In conclusion, beiging controls the synergistic effects of salt and MEK6 on adipogenesis, inflammation, and energy expenditure.
Collapse
Affiliation(s)
- Songjoo Kang
- Department of Food & Nutrition, Sungshin Women’s University, Seoul 01133, Korea;
| | - Myoungsook Lee
- Department of Food & Nutrition, Sungshin Women’s University, Seoul 01133, Korea;
- Research Institute of Obesity Sciences, Sungshin Women’s University, Seoul 01133, Korea
- Correspondence: ; Tel.: +082-02-920-7211
| |
Collapse
|
21
|
Garg R, Kumariya S, Katekar R, Verma S, Goand UK, Gayen JR. JNK signaling pathway in metabolic disorders: An emerging therapeutic target. Eur J Pharmacol 2021; 901:174079. [PMID: 33812885 DOI: 10.1016/j.ejphar.2021.174079] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023]
Abstract
Metabolic Syndrome is a multifactorial disease associated with increased risk of cardiovascular disorders, type 2 diabetes mellitus, fatty liver disease, etc. Various stress stimuli such as reactive oxygen species, endoplasmic reticulum stress, mitochondrial dysfunction, increased cytokines, or free fatty acids are known to aggravate progressive development of hyperglycemia and hyperlipidemia. Although the exact mechanism contributing to altered metabolism is unclear. Evidence suggests stress kinase role to be a crucial one in metabolic syndrome. Stress kinase, c-jun N-terminal kinase activation (JNK) is involved in various metabolic manifestations including obesity, insulin resistance, fatty liver disease as well as cardiometabolic disorders. It emerged as a foremost mediator in regulating metabolism in the liver, skeletal muscle, adipose tissue as well as pancreatic β cells. It has three isoforms each having a unique and tissue-specific role in altered metabolism. Current findings based on genetic manipulation or chemical inhibition studies identified JNK isoforms to play a central role in the regulation of whole-body metabolism, suggesting it to be a novel therapeutic target. Hence, it is imperative to elucidate its role in metabolic syndrome onset and progression. The purpose of this review is to elucidate in vitro and in vivo implications of JNK signaling along with the therapeutic strategy to inhibit specific isoform. Since metabolic syndrome is an array of diseases and complex pathway, carefully examining each tissue will be important for specific treatment strategies.
Collapse
Affiliation(s)
- Richa Garg
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sanjana Kumariya
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
| | - Roshan Katekar
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Saurabh Verma
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Umesh K Goand
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
22
|
Yousefnia S. Mechanistic effects of arsenic trioxide on acute promyelocytic leukemia and other types of leukemias. Cell Biol Int 2021; 45:1148-1157. [PMID: 33527587 DOI: 10.1002/cbin.11563] [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: 08/09/2020] [Revised: 11/25/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022]
Abstract
Acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia characterized with a translocation between promyelocytic leukemia gene (PML) on chromosome 15 and retinoic acid receptor alpha gene (RARα) on chromosome 17. Transcription of this fusion gene results in PML/RARα fusion protein blocking expression of critical genes involved in differentiation of myeloid cells through interaction with RAR element. PML/RARα fusion protein prevents normal function of PML and RARα as well as inhibiting apoptosis. Arsenic trioxide (ATO) is an important agent for the treatment of relapsed and newly diagnosed APL. ATO induces apoptosis, autophagy, and partial cellular differentiation as well as inhibiting cell growth and angiogenesis. Recognition of signaling pathways and molecular mechanisms induced by ATO can be effective for discovering novel treatment strategies to target leukemia cells. Also, it can be developed for the treatment of a variety of cancer cells. This review provides a perspective on anticancerous effects of ATO on APL and leukemia cells.
Collapse
Affiliation(s)
- Saghar Yousefnia
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
23
|
Abstract
Obesity, which has long since reached epidemic proportions worldwide, is associated with long-term stress to a variety of organs and results in diseases including type 2 diabetes. In the brain, overnutrition induces hypothalamic stress associated with the activation of several signalling pathways, together with central insulin and leptin resistance. This central action of nutrient overload appears very rapidly, suggesting that nutrition-induced hypothalamic stress is a major upstream initiator of obesity and associated diseases. The cellular response to nutrient overload includes the activation of the stress-activated c-Jun N-terminal kinases (JNKs) JNK1, JNK2 and JNK3, which are widely expressed in the brain. Here, we review recent findings on the regulation and effects of these kinases, with particular focus on the hypothalamus, a key brain region in the control of energy and glucose homeostasis. JNK1 blocks the hypothalamic-pituitary-thyroid axis, reducing energy expenditure and promoting obesity. Recently, opposing roles have been identified for JNK1 and JNK3 in hypothalamic agouti gene-related protein (AgRP) neurons: while JNK1 activation in AgRP neurons induces feeding and weight gain and impairs insulin and leptin signalling, JNK3 (also known as MAPK10) deletion in the same neuronal population produces very similar effects. The opposing roles of these kinases, and the unknown role of hypothalamic JNK2, reflect the complexity of JNK biology. Future studies should address the specific function of each kinase, not only in different neuronal subsets, but also in non-neuronal cells in the central nervous system. Decoding the puzzle of brain stress kinases will help to define the central stimuli and mechanisms implicated in the control of energy balance. Graphical abstract.
Collapse
Affiliation(s)
- Rubén Nogueiras
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Guadalupe Sabio
- Department of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
24
|
Herrera-Melle L, Crespo M, Leiva M, Sabio G. Stress-activated kinases signaling pathways in cancer development. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2020.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
25
|
Canovas B, Nebreda AR. Diversity and versatility of p38 kinase signalling in health and disease. Nat Rev Mol Cell Biol 2021; 22:346-366. [PMID: 33504982 PMCID: PMC7838852 DOI: 10.1038/s41580-020-00322-w] [Citation(s) in RCA: 349] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
The ability of cells to deal with different types of stressful situations in a precise and coordinated manner is key for survival and involves various signalling networks. Over the past 25 years, p38 kinases — in particular, p38α — have been implicated in the cellular response to stress at many levels. These span from environmental and intracellular stresses, such as hyperosmolarity, oxidative stress or DNA damage, to physiological situations that involve important cellular changes such as differentiation. Given that p38α controls a plethora of functions, dysregulation of this pathway has been linked to diseases such as inflammation, immune disorders or cancer, suggesting the possibility that targeting p38α could be of therapeutic interest. In this Review, we discuss the organization of this signalling pathway focusing on the diversity of p38α substrates, their mechanisms and their links to particular cellular functions. We then address how the different cellular responses can be generated depending on the signal received and the cell type, and highlight the roles of this kinase in human physiology and in pathological contexts. p38α — the best-characterized member of the p38 kinase family — is a key mediator of cellular stress responses. p38α is activated by a plethora of signals and functions through a multitude of substrates to regulate different cellular behaviours. Understanding context-dependent p38α signalling provides important insights into p38α roles in physiology and pathology.
Collapse
Affiliation(s)
- Begoña Canovas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
26
|
Westenberger G, Sellers J, Fernando S, Junkins S, Han SM, Min K, Lawan A. Function of Mitogen-Activated Protein Kinases in Hepatic Inflammation. JOURNAL OF CELLULAR SIGNALING 2021; 2:172-180. [PMID: 34557866 PMCID: PMC8457364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The western diet and overuse of anti-inflammatory medication have caused a great deal of stress on the liver. Obesity and the associated inflammatory state in insulin-responsive tissues result in the release of pro-inflammatory cytokine that activates the stress-responsive MAPKs, p38 MAPK, and JNK. These MAPKs have figured prominently as critical effectors in physiological and pathophysiological hepatic inflammation. In contrast, evidence for a role for ERK1/2 in hepatic inflammation has been less well developed. In this review article, we describe recent insights into the physiology and pathophysiology of the role of stress-responsive MAPKs in hepatic inflammation during obesity and liver injury with a focus on macrophages, hepatocytes and hepatic stellate cells. In response to metabolic stress and liver injury, JNK activation in macrophages and hepatocytes promotes the secretion of inflammatory cytokines and macrophage and neutrophil infiltration. p38 MAPK plays an important role in contributing to the progression of hepatic inflammation in response to various hepatic cellular stresses, although the precise substrates mediating these effects in hepatocytes and hepatic stellate cells remain to be identified. Both JNK and p38 MAPK promotes profibrotic behavior in hepatic stellate cells.
Collapse
Affiliation(s)
- Gabrielle Westenberger
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, Alabama 35899, USA
| | - Jacob Sellers
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, Alabama 35899, USA
| | - Savanie Fernando
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, Alabama 35899, USA
| | - Sadie Junkins
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, Alabama 35899, USA
| | - Sung Min Han
- Department of Aging and Geriatric Research, Institute of Aging, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Kisuk Min
- Division of Kinesiology, University of Texas at El Paso, El Paso, Texas 79968, USA
| | - Ahmed Lawan
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, Alabama 35899, USA,Correspondence should be addressed to Ahmed Lawan;
| |
Collapse
|
27
|
Crespo M, Gonzalez-Teran B, Nikolic I, Mora A, Folgueira C, Rodríguez E, Leiva-Vega L, Pintor-Chocano A, Fernández-Chacón M, Ruiz-Garrido I, Cicuéndez B, Tomás-Loba A, A-Gonzalez N, Caballero-Molano A, Beiroa D, Hernández-Cosido L, Torres JL, Kennedy NJ, Davis RJ, Benedito R, Marcos M, Nogueiras R, Hidalgo A, Matesanz N, Leiva M, Sabio G. Neutrophil infiltration regulates clock-gene expression to organize daily hepatic metabolism. eLife 2020; 9:59258. [PMID: 33287957 PMCID: PMC7723411 DOI: 10.7554/elife.59258] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022] Open
Abstract
Liver metabolism follows diurnal fluctuations through the modulation of molecular clock genes. Disruption of this molecular clock can result in metabolic disease but its potential regulation by immune cells remains unexplored. Here, we demonstrated that in steady state, neutrophils infiltrated the mouse liver following a circadian pattern and regulated hepatocyte clock-genes by neutrophil elastase (NE) secretion. NE signals through c-Jun NH2-terminal kinase (JNK) inhibiting fibroblast growth factor 21 (FGF21) and activating Bmal1 expression in the hepatocyte. Interestingly, mice with neutropenia, defective neutrophil infiltration or lacking elastase were protected against steatosis correlating with lower JNK activation, reduced Bmal1 and increased FGF21 expression, together with decreased lipogenesis in the liver. Lastly, using a cohort of human samples we found a direct correlation between JNK activation, NE levels and Bmal1 expression in the liver. This study demonstrates that neutrophils contribute to the maintenance of daily hepatic homeostasis through the regulation of the NE/JNK/Bmal1 axis. Every day, the body's biological processes work to an internal clock known as the circadian rhythm. This rhythm is controlled by ‘clock genes’ that are switched on or off by daily physical and environmental cues, such as changes in light levels. These daily rhythms are very finely tuned, and disturbances can lead to serious health problems, such as diabetes or high blood pressure. The ability of the body to cycle through the circadian rhythm each day is heavily influenced by the clock of one key organ: the liver. This organ plays a critical role in converting food and drink into energy. There is evidence that neutrophils – white blood cells that protect the body by being the first response to inflammation – can influence how the liver performs its role in obese people, by for example, releasing a protein called elastase. Additionally, the levels of neutrophils circulating in the blood change following a daily pattern. Crespo, González-Terán et al. wondered whether neutrophils enter the liver at specific times of the day to control liver’s daily rhythm. Crespo, González-Terán et al. revealed that neutrophils visit the liver in a pattern that peaks when it gets light and dips when it gets dark by counting the number of neutrophils in the livers of mice at different times of the day. During these visits, neutrophils secreted elastase, which activated a protein called JNK in the cells of the mice’s liver. This subsequently blocked the activity of another protein, FGF21, which led to the activation of the genes that allow cells to make fat molecules for storage. JNK activation also switched on the clock gene, Bmal1, ultimately causing fat to build up in the mice’s liver. Crespo, González-Terán et al. also found that, in samples from human livers, the levels of elastase, the activity of JNK, and whether the Bmal1 gene was switched on were tightly linked. This suggests that neutrophils may be controlling the liver’s rhythm in humans the same way they do in mice. Overall, this research shows that neutrophils can control and reset the liver's daily rhythm using a precisely co-ordinated series of molecular changes. These insights into the liver's molecular clock suggest that elastase, JNK and BmaI1 may represent new therapeutic targets for drugs or smart medicines to treat metabolic diseases such as diabetes or high blood pressure.
Collapse
Affiliation(s)
- María Crespo
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | | | - Ivana Nikolic
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | | | | | - Irene Ruiz-Garrido
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Beatriz Cicuéndez
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Antonia Tomás-Loba
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Noelia A-Gonzalez
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | | | - Daniel Beiroa
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.,CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Lourdes Hernández-Cosido
- Department of General Surgery, University Hospital of Salamanca-IBSAL, Department of Surgery, University of Salamanca, Salamanca, Spain
| | - Jorge L Torres
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Norman J Kennedy
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Roger J Davis
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Rui Benedito
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Ruben Nogueiras
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.,CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Andrés Hidalgo
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Nuria Matesanz
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Magdalena Leiva
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| |
Collapse
|
28
|
Abstract
Obesity is a health condition that has reached pandemic levels and is implicated in the development and progression of type 2 diabetes mellitus, cancer and heart failure. A key characteristic of obesity is the activation of stress-activated protein kinases (SAPKs), such as the p38 and JNK stress kinases, in several organs, including adipose tissue, liver, skeletal muscle, immune organs and the central nervous system. The correct timing, intensity and duration of SAPK activation contributes to cellular metabolic adaptation. By contrast, uncontrolled SAPK activation has been proposed to contribute to the complications of obesity. The stress kinase signalling pathways have therefore been identified as potential targets for the development of novel therapeutic approaches for metabolic syndrome. The past few decades have seen intense research efforts to determine how these kinases are regulated in a cell-specific manner and to define their contribution to the development of obesity and insulin resistance. Several studies have uncovered new and unexpected functions of the non-classical members of both pathways. Here, we provide an overview of the role of SAPKs in metabolic control and highlight important discoveries in the field.
Collapse
Affiliation(s)
- Ivana Nikolic
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Magdalena Leiva
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
29
|
Kassouf T, Sumara G. Impact of Conventional and Atypical MAPKs on the Development of Metabolic Diseases. Biomolecules 2020; 10:biom10091256. [PMID: 32872540 PMCID: PMC7563211 DOI: 10.3390/biom10091256] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
The family of mitogen-activated protein kinases (MAPKs) consists of fourteen members and has been implicated in regulation of virtually all cellular processes. MAPKs are divided into two groups, conventional and atypical MAPKs. Conventional MAPKs are further classified into four sub-families: extracellular signal-regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK1, 2 and 3), p38 (α, β, γ, δ), and extracellular signal-regulated kinase 5 (ERK5). Four kinases, extracellular signal-regulated kinase 3, 4, and 7 (ERK3, 4 and 7) as well as Nemo-like kinase (NLK) build a group of atypical MAPKs, which are activated by different upstream mechanisms than conventional MAPKs. Early studies identified JNK1/2 and ERK1/2 as well as p38α as a central mediators of inflammation-evoked insulin resistance. These kinases have been also implicated in the development of obesity and diabetes. Recently, other members of conventional MAPKs emerged as important mediators of liver, skeletal muscle, adipose tissue, and pancreatic β-cell metabolism. Moreover, latest studies indicate that atypical members of MAPK family play a central role in the regulation of adipose tissue function. In this review, we summarize early studies on conventional MAPKs as well as recent findings implicating previously ignored members of the MAPK family. Finally, we discuss the therapeutic potential of drugs targeting specific members of the MAPK family.
Collapse
|
30
|
Manieri E, Folgueira C, Rodríguez ME, Leiva-Vega L, Esteban-Lafuente L, Chen C, Cubero FJ, Barrett T, Cavanagh-Kyros J, Seruggia D, Rosell A, Sanchez-Cabo F, Gómez MJ, Monte MJ, G Marin JJ, Davis RJ, Mora A, Sabio G. JNK-mediated disruption of bile acid homeostasis promotes intrahepatic cholangiocarcinoma. Proc Natl Acad Sci U S A 2020; 117:16492-16499. [PMID: 32601222 PMCID: PMC7368313 DOI: 10.1073/pnas.2002672117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Metabolic stress causes activation of the cJun NH2-terminal kinase (JNK) signal transduction pathway. It is established that one consequence of JNK activation is the development of insulin resistance and hepatic steatosis through inhibition of the transcription factor PPARα. Indeed, JNK1/2 deficiency in hepatocytes protects against the development of steatosis, suggesting that JNK inhibition represents a possible treatment for this disease. However, the long-term consequences of JNK inhibition have not been evaluated. Here we demonstrate that hepatic JNK controls bile acid production. We found that hepatic JNK deficiency alters cholesterol metabolism and bile acid synthesis, conjugation, and transport, resulting in cholestasis, increased cholangiocyte proliferation, and intrahepatic cholangiocarcinoma. Gene ablation studies confirmed that PPARα mediated these effects of JNK in hepatocytes. This analysis highlights potential consequences of long-term use of JNK inhibitors for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Elisa Manieri
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - María Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Laura Esteban-Lafuente
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Chaobo Chen
- Department of Immunology, Ophthalmology, and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology, and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Tamera Barrett
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Julie Cavanagh-Kyros
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Davide Seruggia
- Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215
| | - Alejandro Rosell
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Fátima Sanchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Manuel Jose Gómez
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain
| | - Maria J Monte
- Laboratory of Experimental Hepatology and Drug Targeting, National Institute for Study of Liver and Gastrointestinal Diseases (CIBERehd), University of Salamanca, 37007 Salamanca, Spain
| | - Jose J G Marin
- Laboratory of Experimental Hepatology and Drug Targeting, National Institute for Study of Liver and Gastrointestinal Diseases (CIBERehd), University of Salamanca, 37007 Salamanca, Spain
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605;
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain;
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares, Myocardial Pathophysiology Area, 28029 Madrid, Spain;
| |
Collapse
|
31
|
Gusev EY, Zotova NV. Cellular Stress and General Pathological Processes. Curr Pharm Des 2020; 25:251-297. [PMID: 31198111 DOI: 10.2174/1381612825666190319114641] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 02/06/2023]
Abstract
From the viewpoint of the general pathology, most of the human diseases are associated with a limited number of pathogenic processes such as inflammation, tumor growth, thrombosis, necrosis, fibrosis, atrophy, pathological hypertrophy, dysplasia and metaplasia. The phenomenon of chronic low-grade inflammation could be attributed to non-classical forms of inflammation, which include many neurodegenerative processes, pathological variants of insulin resistance, atherosclerosis, and other manifestations of the endothelial dysfunction. Individual and universal manifestations of cellular stress could be considered as a basic element of all these pathologies, which has both physiological and pathophysiological significance. The review examines the causes, main phenomena, developmental directions and outcomes of cellular stress using a phylogenetically conservative set of genes and their activation pathways, as well as tissue stress and its role in inflammatory and para-inflammatory processes. The main ways towards the realization of cellular stress and its functional blocks were outlined. The main stages of tissue stress and the classification of its typical manifestations, as well as its participation in the development of the classical and non-classical variants of the inflammatory process, were also described. The mechanisms of cellular and tissue stress are structured into the complex systems, which include networks that enable the exchange of information with multidirectional signaling pathways which together make these systems internally contradictory, and the result of their effects is often unpredictable. However, the possible solutions require new theoretical and methodological approaches, one of which includes the transition to integral criteria, which plausibly reflect the holistic image of these processes.
Collapse
Affiliation(s)
- Eugeny Yu Gusev
- Laboratory of the Immunology of Inflammation, Institute of Immunology and Physiology, Yekaterinburg, Russian Federation
| | - Natalia V Zotova
- Laboratory of the Immunology of Inflammation, Institute of Immunology and Physiology, Yekaterinburg, Russian Federation.,Department of Medical Biochemistry and Biophysics, Ural Federal University named after B.N.Yeltsin, Yekaterinburg, Russian Federation
| |
Collapse
|
32
|
Bennett AM, Lawan A. Improving Obesity and Insulin Resistance by Targeting Skeletal Muscle MKP-1. JOURNAL OF CELLULAR SIGNALING 2020; 1:160-168. [PMID: 33179019 PMCID: PMC7654974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Obesity has reached a global epidemic and it predisposes to the development of insulin resistance, type 2 diabetes and related metabolic diseases. Current interventions against obesity and/or type 2 diabetes such as calorie restriction, exercise, genetic manipulations or established pharmacological treatments have not been successful for many patients with obesity and/or type 2 diabetes. There is an urgent need for new strategies to treat insulin resistance, T2D and obesity. Increased activity of stress-responsive pathways has been linked to the pathogenesis of insulin resistance in obesity. In this commentary, we argue that chronic upregulation of MKP-1 in skeletal muscle is part of a stress response that contributes to the development of insulin resistance, T2D and obesity. Therefore, inhibition of MKP-1 in skeletal muscle is a potential strategy for the treatment of T2D and obesity. We highlight therapeutic strategies for potential targeting of MKP-1 in skeletal muscle for the treatment of metabolic diseases as well as other diseases of skeletal muscle.
Collapse
Affiliation(s)
- Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA,Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, USA,Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Ahmed Lawan
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, Alabama 35899, USA,Correspondence should be addressed to Ahmed Lawan;
| |
Collapse
|
33
|
Leiva M, Matesanz N, Pulgarín-Alfaro M, Nikolic I, Sabio G. Uncovering the Role of p38 Family Members in Adipose Tissue Physiology. Front Endocrinol (Lausanne) 2020; 11:572089. [PMID: 33424765 PMCID: PMC7786386 DOI: 10.3389/fendo.2020.572089] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
The complex functions of adipose tissue have been a focus of research interest over the past twenty years. Adipose tissue is not only the main energy storage depot, but also one of the largest endocrine organs in the body and carries out crucial metabolic functions. Moreover, brown and beige adipose depots are major sites of energy expenditure through the activation of adaptive, non-shivering thermogenesis. In recent years, numerous signaling molecules and pathways have emerged as critical regulators of adipose tissue, in both homeostasis and obesity-related disease. Among the best characterized are members of the p38 kinase family. The activity of these kinases has emerged as a key contributor to the biology of the white and brown adipose tissues, and their modulation could provide new therapeutic approaches against obesity. Here, we give an overview of the roles of the distinct p38 family members in adipose tissue, focusing on their actions in adipogenesis, thermogenic activity, and secretory function.
Collapse
|
34
|
Yu JX, Craig AJ, Duffy ME, Villacorta-Martin C, Miguela V, Ruiz de Galarreta M, Scopton AP, Silber L, Maldonado AY, Rialdi A, Guccione E, Lujambio A, Villanueva A, Dar AC. Phenotype-Based Screens with Conformation-Specific Inhibitors Reveal p38 Gamma and Delta as Targets for HCC Polypharmacology. Mol Cancer Ther 2019; 18:1506-1519. [PMID: 31213506 PMCID: PMC7017390 DOI: 10.1158/1535-7163.mct-18-0571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 03/14/2019] [Accepted: 06/12/2019] [Indexed: 12/24/2022]
Abstract
The approved kinase inhibitors for hepatocellular carcinoma (HCC) are not matched to specific mutations within tumors. This has presented a daunting challenge; without a clear target or mechanism, no straightforward path has existed to guide the development of improved therapies for HCC. Here, we combine phenotypic screens with a class of conformation-specific kinase inhibitors termed type II to identify a multikinase inhibitor, AD80, with antitumoral activity across a variety of HCC preclinical models, including mouse xenografts. Mass spectrometry profiling found a number of kinases as putative targets for AD80, including several receptor and cytoplasmic protein kinases. Among these, we found p38 gamma and delta as direct targets of AD80. Notably, a closely related analog of AD80 lacking p38δ/γ activity, but retaining several other off-target kinases, lost significant activity in several HCC models. Moreover, forced and sustained MKK6 → p38→ATF2 signaling led to a significant reduction of AD80 activity within HCC cell lines. Together with HCC survival data in The Cancer Genome Atlas and RNA-seq analysis, we suggest p38 delta and gamma as therapeutic targets in HCC and an "AD80 inhibition signature" as identifying those patients with best clinical outcomes.
Collapse
Affiliation(s)
- Jia Xin Yu
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amanda J Craig
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mary E Duffy
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carlos Villacorta-Martin
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Verónica Miguela
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Precision Immunology Institute at Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marina Ruiz de Galarreta
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Precision Immunology Institute at Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexander P Scopton
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lisa Silber
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andres Y Maldonado
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexander Rialdi
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ernesto Guccione
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amaia Lujambio
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York.
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Precision Immunology Institute at Icahn School of Medicine at Mount Sinai, New York, New York
| | - Augusto Villanueva
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York.
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Arvin C Dar
- Department of Oncological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York.
- Department of Pharmacological Sciences, The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, New York
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
35
|
Su C, Sun Q, Liu S, Wang H, Feng L, Cao Y. Targeting p38γ to inhibit human colorectal cancer cell progression. Biochem Biophys Res Commun 2019; 517:172-179. [PMID: 31349971 DOI: 10.1016/j.bbrc.2019.07.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 01/02/2023]
Abstract
Colorectal cancer (CRC) is a common malignancy globally causing significant cancer-related mortality. Recent studies have proposed p38gamma (p38γ) as a novel cyclin-dependent kinase (CDK)-like kinase, promoting tumorigenesis and cancer progression. The current study evaluates p38γ expression and potential role in CRC. In HT-29 cells and primary human colon cancer cells, shRNA-induced p38γ silencing or CRISPR/Cas9-mediated p38γ knockout inhibited cell growth, proliferation, and migration, and induced significant apoptosis. Conversely, ectopic overexpression of p38γ further promoted the growth, proliferation, and migration of HT-29 cells and primary colon cancer cells. Retinoblastoma (Rb) phosphorylation and cyclins (E1/A) expression were decreased by p38γ silencing or KO, but increased with p38γ overexpression. p38γ mRNA and protein levels are significantly upregulated in human colon cancer tissues, when compared to levels in surrounding colon epithelial tissues. These results demonstrate that overexpression of p38γ can promote human CRC cell progression, and identify p38γ as a novel therapeutic target.
Collapse
Affiliation(s)
- Chang Su
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Qi Sun
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Shaoqun Liu
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Huayin Wang
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yiou Cao
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
p38gamma overexpression promotes renal cell carcinoma cell growth, proliferation and migration. Biochem Biophys Res Commun 2019; 516:466-473. [PMID: 31229268 DOI: 10.1016/j.bbrc.2019.06.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/08/2019] [Indexed: 01/08/2023]
Abstract
Recent studies have proposed that p38gamma (p38γ) might be critically involved in tumorigenesis and cancer progression. Its expression and potential functions in human renal cell carcinoma (RCC) are studied here. We show that p38γ mRNA and protein levels are upregulated in human RCC tissues, as compared to its levels in the surrounding normal renal tissues. p38γ upregulation was also detected in established (786-O line) and primary human RCC cells. Functional studies in 786-O cells and primary human RCC cells demonstrated that p38γ silencing (by targeted shRNAs) or CRISPR/Cas9-mediated p38γ knockout (KO) potently inhibited cell growth, viability, proliferation and migration. Furthermore, p38γ shRNA or KO in RCC cells decreased retinoblastoma (Rb) phosphorylation and downregulated cyclin E1/A expression. Additionally, significant apoptosis activation was detected in p38γ-silenced and p38γ-KO RCC cells. Contrarily, ectopic overexpression of p38γ facilitated cell growth, viability, proliferation and migration in RCC cells. Taken together, we show that p38γ overexpression promotes RCC cell growth, proliferation and migration. p38γ could be a novel therapeutic target for human RCC.
Collapse
|
37
|
Lin YC, Chen YC, Hsiao HP, Kuo CH, Chen BH, Chen YT, Wang SL, Tsai ML, Hung CH. The effects of acarbose on chemokine and cytokine production in human monocytic THP-1 cells. Hormones (Athens) 2019; 18:179-187. [PMID: 30827017 DOI: 10.1007/s42000-019-00101-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/15/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Chronic inflammation induced by proinflammatory cytokines and chemokines is postulated to be involved in insulin resistance and β-cell dysfunction in type 2 diabetes mellitus (T2DM). Acarbose, the α-glucosidase inhibitor, is an oral antidiabetic drug for T2DM. Acarbose suppresses inflammatory cytokine production in patients with T2DM, though the underlying mechanisms are unclear. In the present study, we aimed to investigate the anti-inflammatory effects and the exact mechanisms of acarbose in human monocytic THP-1 cells. METHODS THP-1 cells were pretreated with acarbose and then stimulated with lipopolysaccharide (LPS). The levels of Th1-related chemokines, including interferon-γ-inducible protein-10 (IP-10), monocyte chemoattractant protein-1 (MCP-1), Th2-related chemokine macrophage-derived chemokine (MDC), and proinflammatory cytokine tumor necrosis factor-α (TNF-α), were determined by enzyme-linked immunosorbent assay. Intracellular signaling pathways were explored by Western blot analysis and using a chromatin immunoprecipitation assay. RESULTS Acarbose suppressed the levels of IP-10, MCP-1, MDC, and TNF-α and downregulated phosphorylation of p38, c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and nuclear factor-kappa B-p65 (NF-κB-p65) in LPS-stimulated THP-1 cells. Acarbose suppressed LPS-induced acetylation of histones H3 (H3) and H4 in the IP-10 and MCP-1 promoter regions. These findings revealed the suppressive effects of acarbose on IP-10, MCP-1, MDC, and TNF-α production in THP-1 cells via, at least partially, the p38, JNK, ERK, and NF-κB-p65 pathways, as well as through epigenetic regulation via histone H3 and H4 acetylation. CONCLUSION Our study points to the therapeutic anti-inflammatory potential of acarbose.
Collapse
Affiliation(s)
- Yi-Ching Lin
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Laboratory Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Research Center for Environmental Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Yen-Chun Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, No.482, Shanming Road, Siaogang District, Kaohsiung City, 812, Taiwan, Republic of China
| | - Hui-Pin Hsiao
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Chang-Hung Kuo
- Ta-Kuo Clinic, No.69, Ziqiang 2nd Road, Cianjin District, Kaohsiung City, 144, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, No.68, Jhonghua 3rd Road, Cianjin District, Kaohsiung City, 145, Taiwan, Republic of China
| | - Bai-Hsiun Chen
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Research Center for Environmental Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Yi-Ting Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Shih-Ling Wang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Mei-Lan Tsai
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Chih-Hsing Hung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China.
- Research Center for Environmental Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China.
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, No.482, Shanming Road, Siaogang District, Kaohsiung City, 812, Taiwan, Republic of China.
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China.
| |
Collapse
|
38
|
Tangudu NK, Buth N, Strnad P, Cirstea IC, Spasić MV. Deregulation of Hepatic Mek1/2⁻Erk1/2 Signaling Module in Iron Overload Conditions. Pharmaceuticals (Basel) 2019; 12:ph12020070. [PMID: 31067696 PMCID: PMC6631327 DOI: 10.3390/ph12020070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/30/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023] Open
Abstract
The liver, through the production of iron hormone hepcidin, controls body iron levels. High liver iron levels and deregulated hepcidin expression are commonly observed in many liver diseases including highly prevalent genetic iron overload disorders. In spite of a number of breakthrough investigations into the signals that control hepcidin expression, little progress has been made towards investigations into intracellular signaling in the liver under excess of iron. This study examined hepatic signaling pathways underlying acquired and genetic iron overload conditions. Our data demonstrate that hepatic iron overload associates with a decline in the activation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (Erk) kinase (Mek1/2) pathway by selectively affecting the phosphorylation of Erk1/2. We propose that Mek1/2-Erk1/2 signaling is uncoupled from iron-Bmp-Smad-mediated hepcidin induction and that it may contribute to a number of liver pathologies in addition to toxic effects of iron. We believe that our findings will advance the understanding of cellular signaling events in the liver during iron overload of different etiologies.
Collapse
Affiliation(s)
- Naveen Kumar Tangudu
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm 89081, Germany; (N.K.T.); (N.B.); (I.C.C.)
| | - Nils Buth
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm 89081, Germany; (N.K.T.); (N.B.); (I.C.C.)
| | - Pavel Strnad
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen 52074, Germany;
| | - Ion C. Cirstea
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm 89081, Germany; (N.K.T.); (N.B.); (I.C.C.)
| | - Maja Vujić Spasić
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm 89081, Germany; (N.K.T.); (N.B.); (I.C.C.)
- Correspondence: ; Tel.: +49-731-50-32635
| |
Collapse
|
39
|
Abstract
Men are more likely to develop hepatocellular carcinoma (HCC) than women, but it is not clear why. In this issue of JEM, Manieri et al. (https://doi.org/10.1084/jem.20181288) identify reduced adiponectin levels as responsible for the increased incidence of HCC in males.
Collapse
Affiliation(s)
- Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch and National Cancer Institute Center for Cancer Research Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
40
|
Park J, Joe Y, Ryter SW, Surh YJ, Chung HT. Similarities and Distinctions in the Effects of Metformin and Carbon Monoxide in Immunometabolism. Mol Cells 2019; 42:292-300. [PMID: 31091555 PMCID: PMC6530647 DOI: 10.14348/molcells.2019.0016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/14/2019] [Accepted: 04/18/2019] [Indexed: 12/31/2022] Open
Abstract
Immunometabolism, defined as the interaction of metabolic pathways with the immune system, influences the pathogenesis of metabolic diseases. Metformin and carbon monoxide (CO) are two pharmacological agents known to ameliorate metabolic disorders. There are notable similarities and differences in the reported effects of metformin and CO on immunometabolism. Metformin, an anti-diabetes drug, has positive effects on metabolism and can exert anti-inflammatory and anti-cancer effects via adenosine monophosphate-activated protein kinase (AMPK)-dependent and AMPK-independent mechanisms. CO, an endogenous product of heme oxygenase-1 (HO-1), can exert anti-inflammatory and antioxidant effects at low concentration. CO can confer cytoprotection in metabolic disorders and cancer via selective activation of the protein kinase R-like endoplasmic reticulum (ER) kinase (PERK) pathway. Both metformin and CO can induce mitochondrial stress to produce a mild elevation of mitochondrial ROS (mtROS) by distinct mechanisms. Metformin inhibits complex I of the mitochondrial electron transport chain (ETC), while CO inhibits ETC complex IV. Both metformin and CO can differentially induce several protein factors, including fibroblast growth factor 21 (FGF21) and sestrin2 (SESN2), which maintain metabolic homeostasis; nuclear factor erythroid 2-related factor 2 (Nrf2), a master regulator of the antioxidant response; and REDD1, which exhibits an anticancer effect. However, metformin and CO regulate these effects via different pathways. Metformin stimulates p53- and AMPK-dependent pathways whereas CO can selectively trigger the PERK-dependent signaling pathway. Although further studies are needed to identify the mechanistic differences between metformin and CO, pharmacological application of these agents may represent useful strategies to ameliorate metabolic diseases associated with altered immunometabolism.
Collapse
Affiliation(s)
- Jeongmin Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Yeonsoo Joe
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Stefan W. Ryter
- Joan and Sanford I. Weill Department of Medicine, and Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical Center, NY 10065,
USA
| | - Young-Joon Surh
- Tumor microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08733,
Korea
| | - Hun Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| |
Collapse
|
41
|
p38γ is essential for cell cycle progression and liver tumorigenesis. Nature 2019; 568:557-560. [PMID: 30971822 DOI: 10.1038/s41586-019-1112-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/07/2019] [Indexed: 11/08/2022]
Abstract
The cell cycle is a tightly regulated process that is controlled by the conserved cyclin-dependent kinase (CDK)-cyclin protein complex1. However, control of the G0-to-G1 transition is not completely understood. Here we demonstrate that p38 MAPK gamma (p38γ) acts as a CDK-like kinase and thus cooperates with CDKs, regulating entry into the cell cycle. p38γ shares high sequence homology, inhibition sensitivity and substrate specificity with CDK family members. In mouse hepatocytes, p38γ induces proliferation after partial hepatectomy by promoting the phosphorylation of retinoblastoma tumour suppressor protein at known CDK target residues. Lack of p38γ or treatment with the p38γ inhibitor pirfenidone protects against the chemically induced formation of liver tumours. Furthermore, biopsies of human hepatocellular carcinoma show high expression of p38γ, suggesting that p38γ could be a therapeutic target in the treatment of this disease.
Collapse
|
42
|
Keiran N, Ceperuelo-Mallafré V, Calvo E, Hernández-Alvarez MI, Ejarque M, Núñez-Roa C, Horrillo D, Maymó-Masip E, Rodríguez MM, Fradera R, de la Rosa JV, Jorba R, Megia A, Zorzano A, Medina-Gómez G, Serena C, Castrillo A, Vendrell J, Fernández-Veledo S. SUCNR1 controls an anti-inflammatory program in macrophages to regulate the metabolic response to obesity. Nat Immunol 2019; 20:581-592. [PMID: 30962591 DOI: 10.1038/s41590-019-0372-7] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/05/2019] [Indexed: 12/14/2022]
Abstract
Succinate is a signaling metabolite sensed extracellularly by succinate receptor 1 (SUNCR1). The accumulation of succinate in macrophages is known to activate a pro-inflammatory program; however, the contribution of SUCNR1 to macrophage phenotype and function has remained unclear. Here we found that activation of SUCNR1 had a critical role in the anti-inflammatory responses in macrophages. Myeloid-specific deficiency in SUCNR1 promoted a local pro-inflammatory phenotype, disrupted glucose homeostasis in mice fed a normal chow diet, exacerbated the metabolic consequences of diet-induced obesity and impaired adipose-tissue browning in response to cold exposure. Activation of SUCNR1 promoted an anti-inflammatory phenotype in macrophages and boosted the response of these cells to type 2 cytokines, including interleukin-4. Succinate decreased the expression of inflammatory markers in adipose tissue from lean human subjects but not that from obese subjects, who had lower expression of SUCNR1 in adipose-tissue-resident macrophages. Our findings highlight the importance of succinate-SUCNR1 signaling in determining macrophage polarization and assign a role to succinate in limiting inflammation.
Collapse
Affiliation(s)
- Noelia Keiran
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Victoria Ceperuelo-Mallafré
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Calvo
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Isabel Hernández-Alvarez
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Miriam Ejarque
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Catalina Núñez-Roa
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Horrillo
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Biología Molecular, Universidad Rey Juan Carlos, Madrid, Spain
| | - Elsa Maymó-Masip
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - M Mar Rodríguez
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Fradera
- General and Digestive Surgery Service, Hospital St. Pau i Sta Tecla, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain
| | - Juan Vladimir de la Rosa
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitaria (IUBIS), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Rosa Jorba
- General and Digestive Surgery Service, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain
| | - Ana Megia
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Departament de Bioquímica i Biomedicina Molecular, Facultat de Biología, Barcelona, Spain
| | - Gema Medina-Gómez
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Biología Molecular, Universidad Rey Juan Carlos, Madrid, Spain
| | - Carolina Serena
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitaria (IUBIS), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Joan Vendrell
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain. .,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain. .,Universitat Rovira i Virgili, Tarragona, Spain.
| | - Sonia Fernández-Veledo
- Unitat de Recerca, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain. .,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
43
|
Manieri E, Herrera-Melle L, Mora A, Tomás-Loba A, Leiva-Vega L, Fernández DI, Rodríguez E, Morán L, Hernández-Cosido L, Torres JL, Seoane LM, Cubero FJ, Marcos M, Sabio G. Adiponectin accounts for gender differences in hepatocellular carcinoma incidence. J Exp Med 2019; 216:1108-1119. [PMID: 30944152 PMCID: PMC6504215 DOI: 10.1084/jem.20181288] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/11/2019] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Gender disparity in liver cancer incidence is a relevant feature of this malignancy. Manieri et al. show that testosterone-induced JNK1 activation in adipose tissue results in decreased levels of circulating adiponectin, which is responsible for higher incidence of hepatocellular carcinoma in males. Hepatocellular carcinoma (HCC) is the sixth most common cancer type and the fourth leading cause of cancer-related death. This cancer appears with higher incidence in men and during obesity; however, the specific mechanisms underlying this correlation are unknown. Adipose tissue, a key organ in metabolic syndrome, shows evident gender disparities in the production of adipokines. Levels of the important adipokine adiponectin decrease in men during puberty, as well as in the obese state. Here, we show that this decrease in adiponectin levels is responsible for the increased liver cancer risk in males. We found that testosterone activates the protein JNK in mouse and human adipocytes. JNK-mediated inhibition of adiponectin secretion increases liver cancer cell proliferation, since adiponectin protects against liver cancer development through the activation of AMP-activated protein kinase (AMPK) and p38α. This study provides insight into adipose tissue to liver crosstalk and its gender relation during cancer development, having the potential to guide strategies for new cancer therapeutics.
Collapse
Affiliation(s)
- Elisa Manieri
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro Nacional de Biotecnología, Madrid, Spain
| | | | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Antonia Tomás-Loba
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Delia I Fernández
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Laura Morán
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Complutense University School of Medicine, Madrid, Spain.,12 de Octubre Health Research Institute, Madrid, Spain
| | - Lourdes Hernández-Cosido
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Jorge L Torres
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Luisa M Seoane
- Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago, Hospital Clínico Universitario de Santiago de Compostela Servicio Gallego de Salud, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red (CIBER), Fisiopatología Obesidad y Nutrición, Instituto Salud Carlos III, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Complutense University School of Medicine, Madrid, Spain.,12 de Octubre Health Research Institute, Madrid, Spain
| | - Miguel Marcos
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
44
|
Sun Y, Zhang D, Li C, Huang J, Li W, Qiu Y, Mao A, Zhou M, Xue L. Lic regulates JNK-mediated cell death in Drosophila. Cell Prolif 2019; 52:e12593. [PMID: 30847993 PMCID: PMC6536442 DOI: 10.1111/cpr.12593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 01/01/2023] Open
Abstract
Objectives The evolutionary conserved JNK pathway plays crucial role in cell death, yet factors that modulate this signalling have not been fully disclosed. In this study, we aim to identify additional factors that regulate JNK signalling in cell death, and characterize the underlying mechanisms. Materials and Methods Drosophila were raised on standard media, and cross was carried out at 25°C. The Gal4/UAS system was used to express proteins or RNAi in a specific temporal and spatial pattern. Gene expression was revealed by GFP fluorescence, X‐gal staining or immunostaining of 3rd instar larval eye and wing discs. Cell death was visualized by acridine orange (AO) staining. Images of fly eyes and wings were taken by OLYMPUS microscopes. Results We found that licorne (lic) encoding the Drosophila MKK3 is an essential regulator of JNK‐mediated cell death. Firstly, loss of lic suppressed ectopic Egr‐triggered JNK activation and cell death in eye and wing development. Secondary, lic is necessary for loss‐of‐cell polarity‐induced, physiological JNK‐dependent cell death in wing development. Thirdly, Lic overexpression is sufficient to initiate JNK‐mediated cell death in developing eyes and wings. Furthermore, ectopic Lic activates JNK signalling by promoting JNK phosphorylation. Finally, genetic epistatic analysis confirmed that Lic acts in parallel with Hep in the Egr‐JNK pathway. Conclusions This study not only identified Lic as a novel component of the JNK signalling, but also disclosed the crucial roles and mechanism of Lic in cell death.
Collapse
Affiliation(s)
- Yihao Sun
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Di Zhang
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Chenglin Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Jiuhong Huang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing, China
| | - Wenzhe Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yu Qiu
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Aiwu Mao
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingcheng Zhou
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lei Xue
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| |
Collapse
|
45
|
Jiang M, Li C, Liu Q, Wang A, Lei M. Inhibiting Ceramide Synthesis Attenuates Hepatic Steatosis and Fibrosis in Rats With Non-alcoholic Fatty Liver Disease. Front Endocrinol (Lausanne) 2019; 10:665. [PMID: 31616384 PMCID: PMC6775186 DOI: 10.3389/fendo.2019.00665] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common metabolic disorder diseases, which include a histological spectrum of conditions ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). Dysregulated metabolism of sphingomyelin in the liver plays a critical role in the pathogenesis of NAFLD. Ceramides are central molecules of sphingolipid biosynthesis and catabolism and play an important role in insulin resistance, apoptosis, and inflammation. In addition, apoptosis is a main contributor to the development of NAFLD. This study detected whether the inhibition of ceramide synthesis ameliorated hepatic steatosis and fibrosis in rats with NAFLD. Sprague-Dawley rats were used to establish the NAFLD model. Here, we showed that hepatic ceramide, steatosis, and fibrosis increased in liver tissue from rats with NAFLD. Chronic treatment with myriocin inhibited ceramide and lipid accumulation and improved fibrosis in liver tissue samples of high fat diet (HFD)-fed rats. In addition, hepatic inflammation and apoptosis were markedly ameliorated in HFD-fed rats treated with myriocin. Furthermore, myriocin treatment regulated the expression of pro-apoptosis and anti-apoptosis proteins by inactivating the c-Jun N-terminal kinase (JNK) signaling pathway in the liver of HFD-fed rats. Collectively, ceramide plays an important role in the pathogenesis of NASH and may represent a potential therapeutic strategy to prevent NAFLD.
Collapse
|
46
|
Matesanz N, Nikolic I, Leiva M, Pulgarín-Alfaro M, Santamans AM, Bernardo E, Mora A, Herrera-Melle L, Rodríguez E, Beiroa D, Caballero A, Martín-García E, Acín-Pérez R, Hernández-Cosido L, Leiva-Vega L, Torres JL, Centeno F, Nebreda AR, Enríquez JA, Nogueiras R, Marcos M, Sabio G. p38α blocks brown adipose tissue thermogenesis through p38δ inhibition. PLoS Biol 2018; 16:e2004455. [PMID: 29979672 PMCID: PMC6051667 DOI: 10.1371/journal.pbio.2004455] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 07/18/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023] Open
Abstract
Adipose tissue has emerged as an important regulator of whole-body metabolism, and its capacity to dissipate energy in the form of heat has acquired a special relevance in recent years as potential treatment for obesity. In this context, the p38MAPK pathway has arisen as a key player in the thermogenic program because it is required for the activation of brown adipose tissue (BAT) thermogenesis and participates also in the transformation of white adipose tissue (WAT) into BAT-like depot called beige/brite tissue. Here, using mice that are deficient in p38α specifically in adipose tissue (p38αFab-KO), we unexpectedly found that lack of p38α protected against high-fat diet (HFD)-induced obesity. We also showed that p38αFab-KO mice presented higher energy expenditure due to increased BAT thermogenesis. Mechanistically, we found that lack of p38α resulted in the activation of the related protein kinase family member p38δ. Our results showed that p38δ is activated in BAT by cold exposure, and lack of this kinase specifically in adipose tissue (p38δ Fab-KO) resulted in overweight together with reduced energy expenditure and lower body and skin surface temperature in the BAT region. These observations indicate that p38α probably blocks BAT thermogenesis through p38δ inhibition. Consistent with the results obtained in animals, p38α was reduced in visceral and subcutaneous adipose tissue of subjects with obesity and was inversely correlated with body mass index (BMI). Altogether, we have elucidated a mechanism implicated in physiological BAT activation that has potential clinical implications for the treatment of obesity and related diseases such as diabetes.
Collapse
Affiliation(s)
- Nuria Matesanz
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Ivana Nikolic
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Magdalena Leiva
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Marta Pulgarín-Alfaro
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Ayelén M. Santamans
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Edgar Bernardo
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Alfonso Mora
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Leticia Herrera-Melle
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Elena Rodríguez
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Daniel Beiroa
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Ainoa Caballero
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Elena Martín-García
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Rebeca Acín-Pérez
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Lourdes Hernández-Cosido
- Bariatric Surgery Unit, Department of General Surgery, University Hospital of Salamanca, Salamanca, Spain
| | - Luis Leiva-Vega
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Jorge L. Torres
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Francisco Centeno
- Facultad de Ciencias, University of Extremadura, Grupo GIEN (Grupo de Investigación en Enfermedades Neurodegenerativas), Badajoz, Spain
| | - Angel R. Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- ICREA, Barcelona, Spain
| | - José Antonio Enríquez
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Guadalupe Sabio
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
47
|
Lawan A, Min K, Zhang L, Canfran-Duque A, Jurczak MJ, Camporez JPG, Nie Y, Gavin TP, Shulman GI, Fernandez-Hernando C, Bennett AM. Skeletal Muscle-Specific Deletion of MKP-1 Reveals a p38 MAPK/JNK/Akt Signaling Node That Regulates Obesity-Induced Insulin Resistance. Diabetes 2018; 67:624-635. [PMID: 29317435 PMCID: PMC5860856 DOI: 10.2337/db17-0826] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 01/03/2018] [Indexed: 12/16/2022]
Abstract
Stress responses promote obesity and insulin resistance, in part, by activating the stress-responsive mitogen-activated protein kinases (MAPKs), p38 MAPK, and c-Jun NH2-terminal kinase (JNK). Stress also induces expression of MAPK phosphatase-1 (MKP-1), which inactivates both JNK and p38 MAPK. However, the equilibrium between JNK/p38 MAPK and MKP-1 signaling in the development of obesity and insulin resistance is unclear. Skeletal muscle is a major tissue involved in energy expenditure and glucose metabolism. In skeletal muscle, MKP-1 is upregulated in high-fat diet-fed mice and in skeletal muscle of obese humans. Mice lacking skeletal muscle expression of MKP-1 (MKP1-MKO) showed increased skeletal muscle p38 MAPK and JNK activities and were resistant to the development of diet-induced obesity. MKP1-MKO mice exhibited increased whole-body energy expenditure that was associated with elevated levels of myofiber-associated mitochondrial oxygen consumption. miR-21, a negative regulator of PTEN expression, was upregulated in skeletal muscle of MKP1-MKO mice, resulting in increased Akt activity consistent with enhanced insulin sensitivity. Our results demonstrate that skeletal muscle MKP-1 represents a critical signaling node through which inactivation of the p38 MAPK/JNK module promotes obesity and insulin resistance.
Collapse
Affiliation(s)
- Ahmed Lawan
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Kisuk Min
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Lei Zhang
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Alberto Canfran-Duque
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT
| | - Michael J Jurczak
- Cellular & Molecular Physiology and Department of Internal Medicine, Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT
| | - Joao Paulo G Camporez
- Cellular & Molecular Physiology and Department of Internal Medicine, Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT
| | - Yaohui Nie
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN
| | - Timothy P Gavin
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN
| | - Gerald I Shulman
- Cellular & Molecular Physiology and Department of Internal Medicine, Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT
| | - Carlos Fernandez-Hernando
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
48
|
Abstract
The proto-oncogene Myc is well known for its roles in promoting cell growth, proliferation and apoptosis. However, in this study, we found from a genetic screen that Myc inhibits, rather than promotes, cell death triggered by c-Jun N-terminal kinase (JNK) signaling in Drosophila. Firstly, expression of Drosophila Myc (dMyc) suppresses, whereas loss of dMyc enhances, ectopically activated JNK signaling-induced cell death. Secondly, dMyc impedes physiologically activated JNK pathway-mediated cell death. Thirdly, loss of dMyc triggers JNK pathway activation and JNK-dependent cell death. Finally, the mammalian cMyc gene, when expressed in Drosophila, impedes activated JNK signaling-induced cell death. Thus, besides its well-studied apoptosis promoting function, Myc also antagonizes JNK-mediated cell death in Drosophila, and this function is likely conserved from fly to human.
Collapse
Affiliation(s)
- Jiuhong Huang
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Feng
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xinhong Chen
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Wenzhe Li
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| | - Lei Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
49
|
Iglesias C, Floridia E, Sartages M, Porteiro B, Fraile M, Guerrero A, Santos D, Cuñarro J, Tovar S, Nogueiras R, Pombo CM, Zalvide J. The MST3/STK24 kinase mediates impaired fasting blood glucose after a high-fat diet. Diabetologia 2017; 60:2453-2462. [PMID: 28956081 DOI: 10.1007/s00125-017-4433-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/28/2017] [Indexed: 01/15/2023]
Abstract
AIMS/HYPOTHESIS The identification of mediators in the pathogenesis of type 2 diabetes mellitus is essential for the full understanding of this disease. Protein kinases are especially important because of their potential as pharmacological targets. The goal of this study was to investigate whether mammalian sterile-20 3 (MST3/STK24), a stress-regulated kinase, is involved in metabolic alterations in obesity. METHODS Glucose regulation of Mst3 (also known as Stk24)-knockout mice was analysed both in 129;C57 mixed background mice and in C57/BL6J mice fed normally or with a high-fat diet (HFD). This work was complemented with an analysis of the insulin signalling pathway in cultured human liver cells made deficient in MST3 using RNA interference. RESULTS MST3 is phosphorylated in the livers of mice subject to an obesity-promoting HFD, and its deficiency lowers the hyperglycaemia, hyperinsulinaemia and insulin resistance that the animals develop with this diet, an effect that is seen even without complete inactivation of the kinase. Lack of MST3 results in activation of the insulin signalling pathway downstream of IRS1, in both cultured liver cells and the liver of animals after HFD. This effect increases the inhibition of forkhead box (FOX)O1, with subsequent downregulation of the expression of gluconeogenic enzymes. CONCLUSIONS/INTERPRETATION MST3 inhibits the insulin signalling pathway and is important in the development of insulin resistance and impaired blood glucose levels after an HFD.
Collapse
Affiliation(s)
- Cristina Iglesias
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Ebel Floridia
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Miriam Sartages
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Begoña Porteiro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - María Fraile
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Ana Guerrero
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, London, UK
| | - Diana Santos
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Juan Cuñarro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Sulay Tovar
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Rubén Nogueiras
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Celia M Pombo
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain.
| | - Juan Zalvide
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain.
| |
Collapse
|
50
|
Lawan A, Bennett AM. Mitogen-Activated Protein Kinase Regulation in Hepatic Metabolism. Trends Endocrinol Metab 2017; 28:868-878. [PMID: 29128158 PMCID: PMC5774993 DOI: 10.1016/j.tem.2017.10.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/20/2017] [Accepted: 10/20/2017] [Indexed: 01/11/2023]
Abstract
The mitogen-activated protein kinases (MAPKs) participate in a multitude of processes that control hepatic metabolism. The liver regulates glucose and lipid metabolism, and under pathophysiological conditions such as obesity, type 2 diabetes mellitus (T2DM), and non-alcoholic fatty liver disease (NAFLD) these processes become dysfunctional. Stress responses activate the hepatic MAPKs, and this is thought to impair insulin action and lipid metabolism. The MAPKs also activate the MAPK phosphatases (MKPs) which oppose their actions. How the MAPK/MKP balance is controlled in liver metabolism and how perturbations in these activities contribute to metabolic disease remains unclear. Discussion of recent insights into the MAPK/MKP signaling role in hepatic metabolic function and disease will be the focus of this review.
Collapse
Affiliation(s)
- Ahmed Lawan
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA.
| | - Anton M Bennett
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University, New Haven, CT 06520, USA
| |
Collapse
|