1
|
Oh E, Lee J, Cho S, Kim SW, Won K, Shin WS, Gwak SH, Ha J, Jeon SY, Park JH, Song IS, Thoudam T, Lee IK, Kim S, Choi SY, Kim KT. Gossypetin Prevents the Progression of Nonalcoholic Steatohepatitis by Regulating Oxidative Stress and AMP-Activated Protein Kinase. Mol Pharmacol 2023; 104:214-229. [PMID: 37595967 DOI: 10.1124/molpharm.123.000675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe liver metabolic disorder, however, there are still no effective and safe drugs for its treatment. Previous clinical trials used various therapeutic approaches to target individual pathologic mechanisms, but these approaches were unsuccessful because of the complex pathologic causes of NASH. Combinatory therapy in which two or more drugs are administered simultaneously to patients with NASH, however, carries the risk of side effects associated with each individual drug. To solve this problem, we identified gossypetin as an effective dual-targeting agent that activates AMP-activated protein kinase (AMPK) and decreases oxidative stress. Administration of gossypetin decreased hepatic steatosis, lobular inflammation and liver fibrosis in the liver tissue of mice with choline-deficient high-fat diet and methionine-choline deficient diet (MCD) diet-induced NASH. Gossypetin functioned directly as an antioxidant agent, decreasing hydrogen peroxide and palmitate-induced oxidative stress in the AML12 cells and liver tissue of MCD diet-fed mice without regulating the antioxidant response factors. In addition, gossypetin acted as a novel AMPK activator by binding to the allosteric drug and metabolite site, which stabilizes the activated structure of AMPK. Our findings demonstrate that gossypetin has the potential to serve as a novel therapeutic agent for nonalcoholic fatty liver disease /NASH. SIGNIFICANCE STATEMENT: This study demonstrates that gossypetin has preventive effect to progression of nonalcoholic steatohepatitis (NASH) as a novel AMP-activated protein kinase (AMPK) activator and antioxidants. Our findings indicate that simultaneous activation of AMPK and oxidative stress using gossypetin has the potential to serve as a novel therapeutic approach for nonalcoholic fatty liver disease /NASH patients.
Collapse
Affiliation(s)
- Eunji Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Jae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Sungji Cho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Sung Wook Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Kyung Won
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Won Sik Shin
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Seung Hee Gwak
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Joohun Ha
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - So Yeon Jeon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Jin-Hyang Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Im-Sook Song
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Themis Thoudam
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - In-Kyu Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Seonyong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Se-Young Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Kyong-Tai Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| |
Collapse
|
2
|
Deliktas O, Gedik ME, Koc I, Gunaydin G, Kiratli H. Modulation of AMPK Significantly Alters Uveal Melanoma Tumor Cell Viability. Ophthalmic Res 2023; 66:1230-1244. [PMID: 37647867 PMCID: PMC10614466 DOI: 10.1159/000533806] [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: 11/25/2022] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
INTRODUCTION Uveal melanoma (UM) responds poorly to targeted therapies or immune checkpoint inhibitors. Adenosine monophosphate-activated protein kinase (AMPK) is a pivotal serine/threonine protein kinase that coordinates vital processes such as cell growth. Targeting AMPK pathway, which represents a critical mechanism mediating the survival of UM cells, may prove to be a novel treatment strategy for UM. We aimed to demonstrate the effects of AMPK modulation on UM cells. METHODS In silico analyses were performed to compare UM and normal melanocyte cells via Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA). The effects of AMPK modulation on cell viability and proliferation in UM cell lines with different molecular profiles (i.e., 92-1, MP46, OMM2.5, and Mel270) were investigated via XTT cell viability and proliferation assays after treating the cells with varying concentrations of A-769662 (AMPK activator) or dorsomorphin (AMPK inhibitor). RESULTS KEGG/GSEA studies demonstrated that genes implicated in the AMPK signaling pathway were differentially regulated in UM. Gene sets comprising genes involved in AMPK signaling and genes involved in energy-dependent regulation of mammalian target of rapamycin by liver kinase B1-AMPK were downregulated in UM. We observed gradual decreases in the numbers of viable UM cells as the concentration of A-769662 treatment increased. All UM cells demonstrated statistically significant decreases in cell viability when treated with 200 µm A-769662. Moreover, the effects of AMPK inhibition on UM cells were potent, since low doses of dorsomorphin treatment resulted in significant decreases in viabilities of UM cells. The half maximal inhibitory concentration (IC50) values confirmed the potency of dorsomorphin treatment against UM in vitro. CONCLUSION AMPK may act like a friend or a foe in cancer depending on the context. As such, the current study contributes to the literature in determining the effects of therapeutic strategies targeting AMPK in several UM cells. We propose a new perspective in the treatment of UM. Targeting AMPK pathway may open up new avenues in developing novel therapeutic approaches to improve overall survival in UM.
Collapse
Affiliation(s)
- Ozge Deliktas
- Department of Ophthalmology, Hacettepe University Medical School, Ankara, Turkey
- Department of Ophthalmology, Bursa City Hospital, Nilufer, Turkey
| | - M. Emre Gedik
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Irem Koc
- Department of Ophthalmology, Hacettepe University Medical School, Ankara, Turkey
| | - Gurcan Gunaydin
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hayyam Kiratli
- Department of Ophthalmology, Hacettepe University Medical School, Ankara, Turkey
| |
Collapse
|
3
|
Moldakozhayev A, Gladyshev VN. Metabolism, homeostasis, and aging. Trends Endocrinol Metab 2023; 34:158-169. [PMID: 36681595 PMCID: PMC11096277 DOI: 10.1016/j.tem.2023.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/21/2023]
Abstract
We propose a two-mode (pursuit/maintenance) model of metabolism defined by usable resource availability. Pursuit, consisting of anabolism and catabolism, dominates when usable resources are plentiful and leads to the generation of metabolic waste. In turn, maintenance of a system is activated by elevated metabolic waste during resource depletion. Interaction with the environment results in pendulum-like swings between these metabolic states in thriveless attempts to maintain the least deleterious organismal state - ephemeral homeostasis. Imperfectness of biological processes during these attempts supports the accumulation of the deleteriome, driving organismal aging. We discuss how metabolic adjustment by the environment and resource stabilization may modulate healthspan and lifespan.
Collapse
Affiliation(s)
- Alibek Moldakozhayev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, QC H3A 2B4, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, QC H4A 3J1, Canada
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Tarasiuk O, Miceli M, Di Domizio A, Nicolini G. AMPK and Diseases: State of the Art Regulation by AMPK-Targeting Molecules. BIOLOGY 2022; 11:biology11071041. [PMID: 36101419 PMCID: PMC9312068 DOI: 10.3390/biology11071041] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022]
Abstract
5′-adenosine monophosphate (AMP)-activated protein kinase (AMPK) is an enzyme that regulates cellular energy homeostasis, glucose, fatty acid uptake, and oxidation at low cellular ATP levels. AMPK plays an important role in several molecular mechanisms and physiological conditions. It has been shown that AMPK can be dysregulated in different chronic diseases, such as inflammation, diabetes, obesity, and cancer. Due to its fundamental role in physiological and pathological cellular processes, AMPK is considered one of the most important targets for treating different diseases. Over decades, different AMPK targeting compounds have been discovered, starting from those that activate AMPK indirectly by altering intracellular AMP:ATP ratio to compounds that activate AMPK directly by binding to its activation sites. However, indirect altering of intracellular AMP:ATP ratio influences different cellular processes and induces side effects. Direct AMPK activators showed more promising results in eliminating side effects as well as the possibility to engineer drugs for specific AMPK isoforms activation. In this review, we discuss AMPK targeting drugs, especially concentrating on those compounds that activate AMPK by mimicking AMP. These compounds are poorly described in the literature and still, a lot of questions remain unanswered about the exact mechanism of AMP regulation. Future investigation of the mechanism of AMP binding will make it possible to develop new compounds that, in combination with others, can activate AMPK in a synergistic manner.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
- Correspondence:
| | - Matteo Miceli
- SPILLOproject—Innovative In Silico Solutions for Drug R&D and Pharmacology, 20037 Paderno Dugnano, Italy; (M.M.); (A.D.D.)
| | - Alessandro Di Domizio
- SPILLOproject—Innovative In Silico Solutions for Drug R&D and Pharmacology, 20037 Paderno Dugnano, Italy; (M.M.); (A.D.D.)
| | - Gabriella Nicolini
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
5
|
Tseng H, Zeng Y, Lin YJ, Huang J, Lin C, Lee M, Yang F, Fang T, Mar A, Su J. A novel AMPK activator shows therapeutic potential in hepatocellular carcinoma by suppressing HIF1α-mediated aerobic glycolysis. Mol Oncol 2022; 16:2274-2294. [PMID: 35298869 PMCID: PMC9168760 DOI: 10.1002/1878-0261.13211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/15/2022] [Accepted: 03/15/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by rapid growth, early vascular invasion, and high metastasis. Currently available US Food and Drug Administration (FDA)-approved drugs show low therapeutic efficacy, limiting HCC treatment to chemotherapy. We designed and synthesized a novel small molecule, SCT-1015, that allosterically activated adenosine monophosphate-activated protein kinase (AMPK) to suppress the aerobic glycolysis in HCC. SCT-1015 was shown to bind the AMPK α and β-subunit interface, thereby exposing the kinase α domain to the upstream kinases, resulting in the increased AMPK activity. SCT-1015 dramatically reduced HCC cell growth in vitro and tumor growth in vivo. We further found that AMPK formed protein complexes with hypoxia-inducible factor 1-alpha (HIF1α) and that SCT-1015-activated AMPK promoted hydroxylation of HIF1α (402P and 564P), resulting in HIF1α degradation by the ubiquitin-proteasome system. With declined HIF1α abundance, many glycolysis-related enzymes were downregulated, suppressing aerobic glycolysis, and promoting oxidative phosphorylation. These results indicated that SCT-1015 channeled HCC cells into an unfavorable metabolic status. Overall, we reported SCT-1015 as a direct activator of AMPK signaling that held therapeutic potential in HCC.
Collapse
Affiliation(s)
- Hsing‐I Tseng
- Department of PharmacyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Institute of Biopharmaceutical SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yi‐Siang Zeng
- Department of PharmacyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department & Institute of PhysiologyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Ying‐Chung Jimmy Lin
- Department of Life Science and Institute of Plant BiologyNational Taiwan UniversityTaipeiTaiwan
- Genome and Systems Biology Degree ProgramNational Taiwan University and Academia SinicaTaipeiTaiwan
| | - Jui‐Wen Huang
- Biomedical Technology and Device Research LabsIndustrial Technology Research InstituteHsinchuTaiwan
| | - Chih‐Lung Lin
- Biomedical Technology and Device Research LabsIndustrial Technology Research InstituteHsinchuTaiwan
| | - Meng‐Hsuan Lee
- Department of PharmacyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Fan‐Wei Yang
- Department of PharmacyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Te‐Ping Fang
- Department of PharmacyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Ai‐Chung Mar
- Taiwan International Graduate Program in Molecular MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipeiTaiwan
| | - Jung‐Chen Su
- Department of PharmacyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
6
|
Abstract
In 2011, CAMKK2, the gene encoding calcium/calmodulin-dependent kinase kinase 2 (CAMKK2), was demonstrated to be a direct target of the androgen receptor and a driver of prostate cancer progression. Results from multiple independent studies have confirmed these findings and demonstrated the potential role of CAMKK2 as a clinical biomarker and therapeutic target in advanced prostate cancer using a variety of preclinical models. Drug development efforts targeting CAMKK2 have begun accordingly. CAMKK2 regulation can vary across disease stages, which might have important implications in the use of CAMKK2 as a biomarker. Moreover, new non-cell-autonomous roles for CAMKK2 that could affect tumorigenesis, metastasis and possible comorbidities linked to disease and treatment have emerged and could present novel treatment opportunities for prostate cancer.
Collapse
|
7
|
Nguyen YTK, To NB, Truong VNP, Kim HY, Ediriweera MK, Lim Y, Cho SK. Impairment of Glucose Metabolism and Suppression of Stemness in MCF-7/SC Human Breast Cancer Stem Cells by Nootkatone. Pharmaceutics 2022; 14:pharmaceutics14050906. [PMID: 35631492 PMCID: PMC9145028 DOI: 10.3390/pharmaceutics14050906] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
Targeting cancer stem cell metabolism has emerged as a promising therapeutic strategy for cancer treatment. Breast cancer stem cells (BCSCs) exert distinct metabolism machinery, which plays a major role in radiation and multidrug resistance. Therefore, exploring the mechanisms involved in energy utilization of BCSCs could improve the effectiveness of therapeutic strategies aimed at their elimination. This study was conducted to clarify the glucose metabolism machinery and the function of nootkatone, a bioactive component of grapefruit, in regulating glucose metabolism and stemness characteristics in human breast carcinoma MCF-7 stem cells (MCF-7SCs). In vivo experiments, transcriptomic analysis, seahorse XF analysis, MTT assay, Western blotting, mammosphere formation, wound healing, invasion assay, flow cytometric analysis, reverse transcription-quantitative polymerase chain reaction, and in silico docking experiments were performed. MCF-7SCs showed a greater tumorigenic capacity and distinct gene profile with enrichment of the genes involved in stemness and glycolysis signaling pathways compared to parental MCF-7 cells, indicating that MCF-7SCs use glycolysis rather than oxidative phosphorylation (OXPHOS) for their energy supply. Nootkatone impaired glucose metabolism through AMPK activation and reduced the stemness characteristics of MCF-7SCs. In silico docking analysis demonstrated that nootkatone efficiently bound to the active site of AMPK. Therefore, this study indicates that regulation of glucose metabolism through AMPK activation could be an attractive target for BCSCs.
Collapse
Affiliation(s)
- Yen Thi-Kim Nguyen
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (Y.T.-K.N.); (N.B.T.); (V.N.-P.T.); (H.Y.K.)
| | - Ngoc Bao To
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (Y.T.-K.N.); (N.B.T.); (V.N.-P.T.); (H.Y.K.)
| | - Vi Nguyen-Phuong Truong
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (Y.T.-K.N.); (N.B.T.); (V.N.-P.T.); (H.Y.K.)
| | - Hee Young Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (Y.T.-K.N.); (N.B.T.); (V.N.-P.T.); (H.Y.K.)
| | - Meran Keshawa Ediriweera
- Subtropical—Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea;
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Colombo, Colombo 00300, Sri Lanka
| | - Yoongho Lim
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea;
| | - Somi Kim Cho
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (Y.T.-K.N.); (N.B.T.); (V.N.-P.T.); (H.Y.K.)
- Subtropical—Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea;
- Correspondence: ; Tel.: +82-10-8660-1842
| |
Collapse
|
8
|
Hyun DH, Lee J. A New Insight into an Alternative Therapeutic Approach to Restore Redox Homeostasis and Functional Mitochondria in Neurodegenerative Diseases. Antioxidants (Basel) 2021; 11:antiox11010007. [PMID: 35052511 PMCID: PMC8772965 DOI: 10.3390/antiox11010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative diseases are accompanied by oxidative stress and mitochondrial dysfunction, leading to a progressive loss of neuronal cells, formation of protein aggregates, and a decrease in cognitive or motor functions. Mitochondrial dysfunction occurs at the early stage of neurodegenerative diseases. Protein aggregates containing oxidatively damaged biomolecules and other misfolded proteins and neuroinflammation have been identified in animal models and patients with neurodegenerative diseases. A variety of neurodegenerative diseases commonly exhibits decreased activity of antioxidant enzymes, lower amounts of antioxidants, and altered cellular signalling. Although several molecules have been approved clinically, there is no known cure for neurodegenerative diseases, though some drugs are focused on improving mitochondrial function. Mitochondrial dysfunction is caused by oxidative damage and impaired cellular signalling, including that of peroxisome proliferator-activated receptor gamma coactivator 1α. Mitochondrial function can also be modulated by mitochondrial biogenesis and the mitochondrial fusion/fission cycle. Mitochondrial biogenesis is regulated mainly by sirtuin 1, NAD+, AMP-activated protein kinase, mammalian target of rapamycin, and peroxisome proliferator-activated receptor γ. Altered mitochondrial dynamics, such as increased fission proteins and decreased fusion products, are shown in neurodegenerative diseases. Due to the restrictions of a target-based approach, a phenotype-based approach has been performed to find novel proteins or pathways. Alternatively, plasma membrane redox enzymes improve mitochondrial function without the further production of reactive oxygen species. In addition, inducers of antioxidant response elements can be useful to induce a series of detoxifying enzymes. Thus, redox homeostasis and metabolic regulation can be important therapeutic targets for delaying the progression of neurodegenerative diseases.
Collapse
|
9
|
Chen J, Ou Q, Wang Z, Liu Y, Hu S, Liu Y, Tian H, Xu J, Gao F, Lu L, Jin C, Xu GT, Cui HP. Small-Molecule Induction Promotes Corneal Endothelial Cell Differentiation From Human iPS Cells. Front Bioeng Biotechnol 2021; 9:788987. [PMID: 34976977 PMCID: PMC8714889 DOI: 10.3389/fbioe.2021.788987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose: Corneal endothelial cells (CECs) serve as a barrier and foothold for the corneal stroma to maintain the function and transparency of the cornea. Loss of CECs during aging or disease states leads to blindness, and cell replacement therapy using either donated or artificially differentiated CECs remains the only curative approach. Methods: Human induced pluripotent stem cells (hiPSCs) that were cultured in chemically defined medium were induced with dual-SMAD inhibition to differentiate into neural crest cells (NCCs). A small-molecule library was screened to differentiate the NCCs into corneal endothelial-like cells. The characteristics of these cells were identified with real-time PCR and immunofluorescence. Western blotting was applied to detect the signaling pathways and key factors regulated by the small molecules. Results: We developed an effective protocol to differentiate hiPSCs into CECs with defined small molecules. The hiPSC-CECs were characterized by ZO-1, AQP1, Vimentin and Na+/K+-ATPase. Based on our small-molecule screen, we identified a small-molecule combination, A769662 and AT13148, that enabled the most efficient production of CECs. The combination of A769662 and AT13148 upregulated the PKA/AKT signaling pathway, FOXO1 and PITX2 to promote the conversion of NCCs to CECs. Conclusion: We established an efficient small molecule-based method to differentiate hiPSCs into corneal endothelial-like cells, which might facilitate drug discovery and the development of cell-based therapies for corneal diseases.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qingjian Ou
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhe Wang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yifan Liu
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuqin Hu
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yumeilan Liu
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haibin Tian
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingying Xu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Furong Gao
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lixia Lu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Caixia Jin
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Guo-Tong Xu
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Hong-Ping Cui
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Dai X, Bu X, Gao Y, Guo J, Hu J, Jiang C, Zhang Z, Xu K, Duan J, He S, Zhang J, Wan L, Liu T, Zhou X, Hung MC, Freeman GJ, Wei W. Energy status dictates PD-L1 protein abundance and anti-tumor immunity to enable checkpoint blockade. Mol Cell 2021; 81:2317-2331.e6. [PMID: 33909988 PMCID: PMC8178223 DOI: 10.1016/j.molcel.2021.03.037] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/26/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Aberrant energy status contributes to multiple metabolic diseases, including obesity, diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4 immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on Ser283, thereby disrupting its interaction with CMTM4 and subsequently triggering PD-L1 degradation. In addition, AMPK phosphorylates EZH2, which disrupts PRC2 function, leading to enhanced IFNs and antigen presentation gene expression. Through these mechanisms, AMPK agonists or ketogenic diets enhance the efficacy of anti-CTLA-4 immunotherapy and improve the overall survival rate in syngeneic mouse tumor models. Our findings reveal a pivotal role for AMPK in regulating the immune response to immune-checkpoint blockade and advocate for combining ketogenic diets or AMPK agonists with anti-CTLA4 immunotherapy to combat cancer.
Collapse
Affiliation(s)
- Xiaoming Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Xia Bu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yang Gao
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jia Hu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Cong Jiang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Zhao Zhang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kexin Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jinzhi Duan
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shaohui He
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jinfang Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Lixin Wan
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Alghamdi F, Alshuweishi Y, Salt IP. Regulation of nutrient uptake by AMP-activated protein kinase. Cell Signal 2020; 76:109807. [DOI: 10.1016/j.cellsig.2020.109807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
|
12
|
Gupta P, Taiyab A, Hassan MI. Emerging role of protein kinases in diabetes mellitus: From mechanism to therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 124:47-85. [PMID: 33632470 DOI: 10.1016/bs.apcsb.2020.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Diabetes mellitus has emerged as a severe burden on the medical health system across the globe. Presently, around 422 million people are suffering from diabetes which is speculated to be expanded to about 600 million by 2035. Patients with type 2 diabetes are at increased risk of developing detrimental metabolic and cardiovascular complications. The scientific understanding of this chronic disease and its underlying root cause is not yet fully unraveled. Protein kinases are well known to regulate almost every cellular process through phosphorylation of target protein in diverse signaling pathways. The important role of several protein kinases including AMP-activated protein kinase, IκB kinase and protein kinase C have been well demonstrated in various animal models. They modulate glucose tolerance, inflammation and insulin resistance in the cells via acting on diverse downstream targets and signaling pathways. Thus, modulating the activity of potential human kinases which are significantly involved in diabetes by targeting with small molecule inhibitors could be an attractive therapeutic strategy to tackle diabetes. In this chapter, we have discussed the potential role of protein kinases in glucose metabolism and insulin sensitivity, and in the pathogenesis of diabetes mellitus. Furthermore, the small molecules reported in the literature that can be potentially used for the treatment of diabetes have been discussed in detail.
Collapse
Affiliation(s)
- Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
13
|
Lospinoso Severini L, Ghirga F, Bufalieri F, Quaglio D, Infante P, Di Marcotullio L. The SHH/GLI signaling pathway: a therapeutic target for medulloblastoma. Expert Opin Ther Targets 2020; 24:1159-1181. [PMID: 32990091 DOI: 10.1080/14728222.2020.1823967] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Medulloblastoma (MB) is a heterogeneous tumor of the cerebellum that is divided into four main subgroups with distinct molecular and clinical features. Sonic Hedgehog MB (SHH-MB) is the most genetically understood and occurs predominantly in childhood. Current therapies consist of aggressive and non-targeted multimodal approaches that are often ineffective and cause long-term complications. These problems intensify the need to develop molecularly targeted therapies to improve outcome and reduce treatment-related morbidities. In this scenario, Hedgehog (HH) signaling, a developmental pathway whose deregulation is involved in the pathogenesis of several malignancies, has emerged as an attractive druggable pathway for SHH-MB therapy. AREAS COVERED This review provides an overview of the advancements in the HH antagonist research field. We place an emphasis on Smoothened (SMO) and glioma-associated oncogene homolog (GLI) inhibitors and immunotherapy approaches that are validated in preclinical SHH-MB models and that have therapeutic potential for MB patients. Literature from Pubmed and data reported on ClinicalTrial.gov up to August 2020 were considered. EXPERT OPINION Extensive-omics analysis has enhanced our knowledge and has transformed the way that MB is studied and managed. The clinical use of SMO antagonists has yet to be determined, however, future GLI inhibitors and multitargeting approaches are promising.
Collapse
Affiliation(s)
| | - Francesca Ghirga
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia , 00161, Rome, Italy
| | - Francesca Bufalieri
- Department of Molecular Medicine, University of Rome La Sapienza , 00161, Rome, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, University of Rome La Sapienza, 00185 , Rome, Italy
| | - Paola Infante
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia , 00161, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University of Rome La Sapienza , 00161, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza , 00161, Rome, Italy
| |
Collapse
|
14
|
Johansson KS, Sonne DP, Knop FK, Christensen MB. What is on the horizon for type 2 diabetes pharmacotherapy? – An overview of the antidiabetic drug development pipeline. Expert Opin Drug Discov 2020; 15:1253-1265. [DOI: 10.1080/17460441.2020.1791078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Karl Sebastian Johansson
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - David Peick Sonne
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Filip Krag Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Bring Christensen
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
The influence of hypoxia and energy depletion on the response of endothelial cells to the vascular disrupting agent combretastatin A-4-phosphate. Sci Rep 2020; 10:9926. [PMID: 32555222 PMCID: PMC7303175 DOI: 10.1038/s41598-020-66568-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/15/2020] [Indexed: 12/25/2022] Open
Abstract
Combretastatin A-4 phosphate (CA4P) is a microtubule-disrupting tumour-selective vascular disrupting agent (VDA). CA4P activates the actin-regulating RhoA-GTPase/ ROCK pathway, which is required for full vascular disruption. While hypoxia renders tumours resistant to many conventional therapies, little is known about its influence on VDA activity. Here, we found that active RhoA and ROCK effector phospho-myosin light chain (pMLC) were downregulated in endothelial cells by severe hypoxia. CA4P failed to activate RhoA/ROCK/pMLC but its activity was restored upon reoxygenation. Hypoxia also inhibited CA4P-mediated actinomyosin contractility, VE-cadherin junction disruption and permeability rise. Glucose withdrawal downregulated pMLC, and coupled with hypoxia, reduced pMLC faster and more profoundly than hypoxia alone. Concurrent inhibition of glycolysis (2-deoxy-D-glucose, 2DG) and mitochondrial respiration (rotenone) caused profound actin filament loss, blocked RhoA/ROCK signalling and rendered microtubules CA4P-resistant. Withdrawal of the metabolism inhibitors restored the cytoskeleton and CA4P activity. The AMP-activated kinase AMPK was investigated as a potential mediator of pMLC downregulation. Pharmacological AMPK activators that generate AMP, unlike allosteric activators, downregulated pMLC but only when combined with 2DG and/or rotenone. Altogether, our results suggest that Rho/ROCK and actinomyosin contractility are regulated by AMP/ATP levels independently of AMPK, and point to hypoxia/energy depletion as potential modifiers of CA4P response.
Collapse
|
16
|
Lorca RA, Matarazzo CJ, Bales ES, Houck JA, Orlicky DJ, Euser AG, Julian CG, Moore LG. AMPK activation in pregnant human myometrial arteries from high-altitude and intrauterine growth-restricted pregnancies. Am J Physiol Heart Circ Physiol 2020; 319:H203-H212. [PMID: 32502374 DOI: 10.1152/ajpheart.00644.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High-altitude (>2,500 m) residence increases the incidence of intrauterine growth restriction (IUGR) due, in part, to reduced uterine artery blood flow and impaired myometrial artery (MA) vasodilator response. A role for the AMP-activated protein kinase (AMPK) pathway in protecting against hypoxia-associated IUGR is suggested by genomic and transcriptomic studies in humans and functional studies in mice. AMPK is a hypoxia-sensitive metabolic sensor with vasodilatory properties. Here we hypothesized that AMPK-dependent vasodilation was increased in MAs from high versus low-altitude (<1,700 m) Colorado women with appropriate for gestational age (AGA) pregnancies and reduced in IUGR pregnancies regardless of altitude. Vasoreactivity studies showed that, in AGA pregnancies, MAs from high-altitude women were more sensitive to vasodilation by activation of AMPK with A769662 due chiefly to increased endothelial nitric oxide production, whereas MA responses to AMPK activation in the low-altitude women were endothelium independent. MAs from IUGR compared with AGA pregnancies had blunted vasodilator responses to acetylcholine at high altitude. We concluded that 1) blunted vasodilator responses in IUGR pregnancies confirm the importance of MA vasodilation for normal fetal growth and 2) the increased sensitivity to AMPK activation in AGA pregnancies at high altitude suggests that AMPK activation helped maintain MA vasodilation and fetal growth. These results highlight a novel mechanism for vasodilation of MAs under conditions of chronic hypoxia and suggest that AMPK activation could provide a therapy for increasing uteroplacental blood flow and improving fetal growth in IUGR pregnancies.NEW & NOTEWORTHY Intrauterine growth restriction (IUGR) impairs infant well- being and increases susceptibility to later-in-life diseases for mother and child. Our study reveals a novel role for AMPK in vasodilating the myometrial artery (MA) from women residing at high altitude (>2,500 m) with appropriate for gestational age pregnancies but not in IUGR pregnancies at any altitude.
Collapse
Affiliation(s)
- Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Christopher J Matarazzo
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Elise S Bales
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Julie A Houck
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - David J Orlicky
- Department of Pathology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Anna G Euser
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Colleen G Julian
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
17
|
Collodet C, Foretz M, Deak M, Bultot L, Metairon S, Viollet B, Lefebvre G, Raymond F, Parisi A, Civiletto G, Gut P, Descombes P, Sakamoto K. AMPK promotes induction of the tumor suppressor FLCN through activation of TFEB independently of mTOR. FASEB J 2019; 33:12374-12391. [PMID: 31404503 PMCID: PMC6902666 DOI: 10.1096/fj.201900841r] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AMPK is a central regulator of energy homeostasis. AMPK not only elicits acute metabolic responses but also promotes metabolic reprogramming and adaptations in the long-term through regulation of specific transcription factors and coactivators. We performed a whole-genome transcriptome profiling in wild-type (WT) and AMPK-deficient mouse embryonic fibroblasts (MEFs) and primary hepatocytes that had been treated with 2 distinct classes of small-molecule AMPK activators. We identified unique compound-dependent gene expression signatures and several AMPK-regulated genes, including folliculin (Flcn), which encodes the tumor suppressor FLCN. Bioinformatics analysis highlighted the lysosomal pathway and the associated transcription factor EB (TFEB) as a key transcriptional mediator responsible for AMPK responses. AMPK-induced Flcn expression was abolished in MEFs lacking TFEB and transcription factor E3, 2 transcription factors with partially redundant function; additionally, the promoter activity of Flcn was profoundly reduced when its putative TFEB-binding site was mutated. The AMPK-TFEB-FLCN axis is conserved across species; swimming exercise in WT zebrafish induced Flcn expression in muscle, which was significantly reduced in AMPK-deficient zebrafish. Mechanistically, we have found that AMPK promotes dephosphorylation and nuclear localization of TFEB independently of mammalian target of rapamycin activity. Collectively, we identified the novel AMPK-TFEB-FLCN axis, which may function as a key cascade for cellular and metabolic adaptations.—Collodet, C., Foretz, M., Deak, M., Bultot, L., Metairon, S., Viollet, B., Lefebvre, G., Raymond, F., Parisi, A., Civiletto, G., Gut, P., Descombes, P., Sakamoto, K. AMPK promotes induction of the tumor suppressor FLCN through activation of TFEB independently of mTOR.
Collapse
Affiliation(s)
- Caterina Collodet
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland.,School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Marc Foretz
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria Deak
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Laurent Bultot
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Sylviane Metairon
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Benoit Viollet
- INSERM Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Gregory Lefebvre
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Frederic Raymond
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Alice Parisi
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Gabriele Civiletto
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Philipp Gut
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Patrick Descombes
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland.,School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| | - Kei Sakamoto
- Nestlé Research, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland.,School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, Lausanne, Switzerland
| |
Collapse
|
18
|
Nguyen TMD. Main signaling pathways involved in the control of fowl sperm motility. Poult Sci 2019; 98:1528-1538. [DOI: 10.3382/ps/pey465] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/09/2018] [Indexed: 12/28/2022] Open
|
19
|
Zhang X, Yang S, Chen J, Su Z. Unraveling the Regulation of Hepatic Gluconeogenesis. Front Endocrinol (Lausanne) 2019; 9:802. [PMID: 30733709 PMCID: PMC6353800 DOI: 10.3389/fendo.2018.00802] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023] Open
Abstract
Hepatic gluconeogenesis, de novo glucose synthesis from available precursors, plays a crucial role in maintaining glucose homeostasis to meet energy demands during prolonged starvation in animals. The abnormally increased rate of hepatic gluconeogenesis contributes to hyperglycemia in diabetes. Gluconeogenesis is regulated on multiple levels, such as hormonal secretion, gene transcription, and posttranslational modification. We review here the molecular mechanisms underlying the transcriptional regulation of gluconeogenesis in response to nutritional and hormonal changes. The nutrient state determines the hormone release, which instigates the signaling cascades in the liver to modulate the activities of various transcriptional factors through various post-translational modifications like phosphorylation, methylation, and acetylation. AMP-activated protein kinase (AMPK) can mediate the activities of some transcription factors, however its role in the regulation of gluconeogenesis remains uncertain. Metformin, a primary hypoglycemic agent of type 2 diabetes, ameliorates hyperglycemia predominantly through suppression of hepatic gluconeogenesis. Several molecular mechanisms have been proposed to be metformin's mode of action.
Collapse
Affiliation(s)
| | | | | | - Zhiguang Su
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Role of AMPK in mammals reproduction: Specific controls and whole-body energy sensing. C R Biol 2018; 342:1-6. [PMID: 30580936 DOI: 10.1016/j.crvi.2018.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 10/10/2018] [Accepted: 10/31/2018] [Indexed: 01/02/2023]
Abstract
AMP-activated protein kinase (AMPK) is a key enzyme involved in linking the energy sensing to metabolic pathways. As such, it plays a central role at the whole-body level to translate endocrine communications into adapted responses aimed either at saving energy when food is scarce or at allocating it to various functions, particularly reproduction, when food is available. AMPK also plays major roles in the energy individual cells use in order to realize their specific functions. This is of course especially true for all cells involved in the reproductive function (gonads, gametes) or in its control (hypothalamus, pituitary). In the present review, I report a survey of the various roles of AMPK functions in reproduction, either directly in reproductive organs, or indirectly in organs controlling reproduction, particularly at hypothalamus level.
Collapse
|
21
|
A769662 Inhibits Insulin-Stimulated Akt Activation in Human Macrovascular Endothelial Cells Independent of AMP-Activated Protein Kinase. Int J Mol Sci 2018; 19:ijms19123886. [PMID: 30563079 PMCID: PMC6321332 DOI: 10.3390/ijms19123886] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/03/2018] [Indexed: 01/15/2023] Open
Abstract
Protein kinase B (Akt) is a key enzyme in the insulin signalling cascade, required for insulin-stimulated NO production in endothelial cells (ECs). Previous studies have suggested that AMP-activated protein kinase (AMPK) activation stimulates NO synthesis and enhances insulin-stimulated Akt activation, yet these studies have largely used indirect activators of AMPK. The effects of the allosteric AMPK activator A769662 on insulin signalling and endothelial function was therefore examined in cultured human macrovascular ECs. Surprisingly, A769662 inhibited insulin-stimulated NO synthesis and Akt phosphorylation in human ECs from umbilical veins (HUVECs) and aorta (HAECs). In contrast, the AMPK activators compound 991 and AICAR had no substantial inhibitory effect on insulin-stimulated Akt phosphorylation in ECs. Inhibition of AMPK with SBI-0206965 had no effect on the inhibition of insulin-stimulated Akt phosphorylation by A769662, suggesting the inhibitory action of A769662 is AMPK-independent. A769662 decreased IGF1-stimulated Akt phosphorylation yet had no effect on VEGF-stimulated Akt signalling in HUVECs, suggesting that A769662 attenuates early insulin/IGF1 signalling. The effects of A769662 on insulin-stimulated Akt phosphorylation were specific to human ECs, as no effect was observed in the human cancer cell lines HepG2 or HeLa, as well as in mouse embryonic fibroblasts (MEFs). A769662 inhibited insulin-stimulated Erk1/2 phosphorylation in HAECs and MEFs, an effect that was independent of AMPK in MEFs. Therefore, despite being a potent AMPK activator, A769662 has effects unlikely to be mediated by AMPK in human macrovascular ECs that reduce insulin sensitivity and eNOS activation.
Collapse
|
22
|
Abstract
AMP-activated protein kinase (AMPK) is the main cellular energy sensor. Activated following a depletion of cellular energy stores, AMPK will restore the energy homoeostasis by increasing energy production and limiting energy waste. At a central level, the AMPK pathway will integrate peripheral signals (mostly hormones and metabolites) through neuronal networks. Hypothalamic AMPK is directly implicated in feeding behaviour, brown adipose tissue (BAT) thermogenesis and browning of white adipose tissue (WAT). It also participates in other metabolic functions: glucose and muscle metabolisms, as well as hepatic function. Numerous anti-obesity and/or antidiabetic agents, such as nicotine, metformin and liraglutide, are known to induce their effects through a modulation of AMPK pathway, either at central or at peripheral levels. Moreover, the weight-gaining side effects of antipsychotic drugs, such as olanzapine, are also mediated by hypothalamic AMPK. Therefore, considering hypothalamic AMPK as a therapeutic target in metabolic diseases appears as an interesting strategy due to its implication in feeding and energy expenditure, the two sides of the energy balance equation.
Collapse
Affiliation(s)
- Miguel López
- NeurObesity Group, 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
| |
Collapse
|
23
|
Olivier S, Foretz M, Viollet B. Promise and challenges for direct small molecule AMPK activators. Biochem Pharmacol 2018; 153:147-158. [PMID: 29408352 DOI: 10.1016/j.bcp.2018.01.049] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
Abstract
AMP-activated protein kinase (AMPK) is an evolutionary conserved and ubiquitously expressed serine/threonine kinase playing a central role in the coordination of energy homeostasis. Based on the beneficial outcomes of its activation on metabolism, AMPK has emerged as an attractive target for the treatment of metabolic diseases. Identification of novel downstream targets of AMPK beyond the regulation of energy metabolism has renewed considerable attention in exploiting AMPK signaling for novel therapeutic targeting strategies including treatment of cancer and inflammatory diseases. The complexity of AMPK system with tissue- and species-specific expression of multiple isoform combination regulated by various inputs, post-traductional modifications and subcellular locations presents unique challenges for drug discovery. Here, we review the most recent advances in the understanding of the mechanism(s) of action of direct small molecule AMPK activators and the potential therapeutic opportunities.
Collapse
Affiliation(s)
- Séverine Olivier
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Marc Foretz
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France.
| |
Collapse
|
24
|
Gélinas R, Mailleux F, Dontaine J, Bultot L, Demeulder B, Ginion A, Daskalopoulos EP, Esfahani H, Dubois-Deruy E, Lauzier B, Gauthier C, Olson AK, Bouchard B, Des Rosiers C, Viollet B, Sakamoto K, Balligand JL, Vanoverschelde JL, Beauloye C, Horman S, Bertrand L. AMPK activation counteracts cardiac hypertrophy by reducing O-GlcNAcylation. Nat Commun 2018; 9:374. [PMID: 29371602 PMCID: PMC5785516 DOI: 10.1038/s41467-017-02795-4] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 12/28/2017] [Indexed: 12/11/2022] Open
Abstract
AMP-activated protein kinase (AMPK) has been shown to inhibit cardiac hypertrophy. Here, we show that submaximal AMPK activation blocks cardiomyocyte hypertrophy without affecting downstream targets previously suggested to be involved, such as p70 ribosomal S6 protein kinase, calcineurin/nuclear factor of activated T cells (NFAT) and extracellular signal-regulated kinases. Instead, cardiomyocyte hypertrophy is accompanied by increased protein O-GlcNAcylation, which is reversed by AMPK activation. Decreasing O-GlcNAcylation by inhibitors of the glutamine:fructose-6-phosphate aminotransferase (GFAT), blocks cardiomyocyte hypertrophy, mimicking AMPK activation. Conversely, O-GlcNAcylation-inducing agents counteract the anti-hypertrophic effect of AMPK. In vivo, AMPK activation prevents myocardial hypertrophy and the concomitant rise of O-GlcNAcylation in wild-type but not in AMPKα2-deficient mice. Treatment of wild-type mice with O-GlcNAcylation-inducing agents reverses AMPK action. Finally, we demonstrate that AMPK inhibits O-GlcNAcylation by mainly controlling GFAT phosphorylation, thereby reducing O-GlcNAcylation of proteins such as troponin T. We conclude that AMPK activation prevents cardiac hypertrophy predominantly by inhibiting O-GlcNAcylation.
Collapse
Affiliation(s)
- Roselle Gélinas
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Florence Mailleux
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Justine Dontaine
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Laurent Bultot
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Bénédicte Demeulder
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Audrey Ginion
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Evangelos P Daskalopoulos
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Hrag Esfahani
- Pole of Pharmacotherapy, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Emilie Dubois-Deruy
- Pole of Pharmacotherapy, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Benjamin Lauzier
- l'institut du thorax, INSERM, CNRS, Univ. Nantes, Nantes, 44007, France
| | - Chantal Gauthier
- l'institut du thorax, INSERM, CNRS, Univ. Nantes, Nantes, 44007, France
| | - Aaron K Olson
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, Seattle, 98105-0371, WA, USA.,Montreal Heart Institute, Montreal, H1T 1C8, Canada
| | | | - Christine Des Rosiers
- Montreal Heart Institute, Montreal, H1T 1C8, Canada.,Department of Nutrition, Université de Montréal, Montreal, H3T 1A8, Canada
| | - Benoit Viollet
- Institut Cochin, INSERM U1016, 75014, Paris, France.,CNRS UMR8104, 75014, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014, France
| | - Kei Sakamoto
- Nestlé Institute of Health Sciences SA, Lausanne, 1015, Switzerland
| | - Jean-Luc Balligand
- Pole of Pharmacotherapy, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Jean-Louis Vanoverschelde
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium.,Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, 1200, Belgium
| | - Christophe Beauloye
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium.,Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, 1200, Belgium
| | - Sandrine Horman
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, 1200, Belgium.
| |
Collapse
|
25
|
Boudaba N, Marion A, Huet C, Pierre R, Viollet B, Foretz M. AMPK Re-Activation Suppresses Hepatic Steatosis but its Downregulation Does Not Promote Fatty Liver Development. EBioMedicine 2018; 28:194-209. [PMID: 29343420 PMCID: PMC5835560 DOI: 10.1016/j.ebiom.2018.01.008] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease is a highly prevalent component of disorders associated with disrupted energy homeostasis. Although dysregulation of the energy sensor AMP-activated protein kinase (AMPK) is viewed as a pathogenic factor in the development of fatty liver its role has not been directly demonstrated. Unexpectedly, we show here that liver-specific AMPK KO mice display normal hepatic lipid homeostasis and are not prone to fatty liver development, indicating that the decreases in AMPK activity associated with hepatic steatosis may be a consequence, rather than a cause, of changes in hepatic metabolism. In contrast, we found that pharmacological re-activation of downregulated AMPK in fatty liver is sufficient to normalize hepatic lipid content. Mechanistically, AMPK activation reduces hepatic triglyceride content both by inhibiting lipid synthesis and by stimulating fatty acid oxidation in an LKB1-dependent manner, through a transcription-independent mechanism. Furthermore, the effect of the antidiabetic drug metformin on lipogenesis inhibition and fatty acid oxidation stimulation was enhanced by combination treatment with small-molecule AMPK activators in primary hepatocytes from mice and humans. Overall, these results demonstrate that AMPK downregulation is not a triggering factor in fatty liver development but in contrast, establish the therapeutic impact of pharmacological AMPK re-activation in the treatment of fatty liver disease. Hepatic AMPK deficiency is not sufficient to trigger fatty liver development Re-activation of downregulated AMPK in fatty liver normalizes hepatic lipid content Hepatic AMPK activation both inhibits lipogenesis and stimulates fatty acid oxidation AMPK activation modulates lipid metabolism via a transcription-independent mechanism Small-molecule AMPK activators enhance metformin effects on hepatic lipid metabolism
Nonalcoholic fatty liver disease is a highly prevalent component of metabolic syndrome, for which treatment options are limited. Downregulation of the energy sensor AMPK is viewed as a pathogenic factor in the development of fatty liver. However, we show here hepatic AMPK suppression is not sufficient to promote hepatic lipid accumulation, indicating that the decreases in AMPK activity associated with hepatic steatosis may be a consequence, rather than a cause, of changes in hepatic metabolism. In contrast, we found that pharmacological re-activation of downregulated AMPK in fatty liver is sufficient to normalize hepatic lipid content. Thus, these results establish the therapeutic impact of pharmacological AMPK re-activation in the treatment of fatty liver disease.
Collapse
Affiliation(s)
- Nadia Boudaba
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Allison Marion
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Camille Huet
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Rémi Pierre
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Marc Foretz
- INSERM, U1016, Institut Cochin, Paris 75014, France; CNRS, UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France.
| |
Collapse
|
26
|
Lopez-Mejia IC, Lagarrigue S, Giralt A, Martinez-Carreres L, Zanou N, Denechaud PD, Castillo-Armengol J, Chavey C, Orpinell M, Delacuisine B, Nasrallah A, Collodet C, Zhang L, Viollet B, Hardie DG, Fajas L. CDK4 Phosphorylates AMPKα2 to Inhibit Its Activity and Repress Fatty Acid Oxidation. Mol Cell 2017; 68:336-349.e6. [PMID: 29053957 DOI: 10.1016/j.molcel.2017.09.034] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 07/17/2017] [Accepted: 09/22/2017] [Indexed: 01/18/2023]
Abstract
The roles of CDK4 in the cell cycle have been extensively studied, but less is known about the mechanisms underlying the metabolic regulation by CDK4. Here, we report that CDK4 promotes anaerobic glycolysis and represses fatty acid oxidation in mouse embryonic fibroblasts (MEFs) by targeting the AMP-activated protein kinase (AMPK). We also show that fatty acid oxidation (FAO) is specifically induced by AMPK complexes containing the α2 subunit. Moreover, we report that CDK4 represses FAO through direct phosphorylation and inhibition of AMPKα2. The expression of non-phosphorylatable AMPKα2 mutants, or the use of a CDK4 inhibitor, increased FAO rates in MEFs and myotubes. In addition, Cdk4-/- mice have increased oxidative metabolism and exercise capacity. Inhibition of CDK4 mimicked these alterations in normal mice, but not when skeletal muscle was AMPK deficient. This novel mechanism explains how CDK4 promotes anabolism by blocking catabolic processes (FAO) that are activated by AMPK.
Collapse
Affiliation(s)
- Isabel C Lopez-Mejia
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Sylviane Lagarrigue
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Albert Giralt
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | | | - Nadège Zanou
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland; Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland
| | - Pierre-Damien Denechaud
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | | | - Carine Chavey
- IGMM, Université de Montpellier, UMR 5535 CNRS, 34293 Montpellier, France
| | - Meritxell Orpinell
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Brigitte Delacuisine
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Anita Nasrallah
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Caterina Collodet
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, 1015 Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, School of Life Sciences, 1015 Lausanne, Switzerland
| | - Lianjun Zhang
- Ludwig Center for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland
| | - Benoît Viollet
- Institut Cochin, INSERM U1016, Paris, France; CNRS, UMR 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - D Grahame Hardie
- School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
27
|
Speirs C, Williams JJL, Riches K, Salt IP, Palmer TM. Linking energy sensing to suppression of JAK-STAT signalling: A potential route for repurposing AMPK activators? Pharmacol Res 2017; 128:88-100. [PMID: 29037480 DOI: 10.1016/j.phrs.2017.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/12/2017] [Accepted: 10/12/2017] [Indexed: 02/07/2023]
Abstract
Exaggerated Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling is key to the pathogenesis of pro-inflammatory disorders, such as rheumatoid arthritis and cardiovascular diseases. Mutational activation of JAKs is also responsible for several haematological malignancies, including myeloproliferative neoplasms and acute lymphoblastic leukaemia. Accumulating evidence links adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), an energy sensor and regulator of organismal and cellular metabolism, with the suppression of immune and inflammatory processes. Recent studies have shown that activation of AMPK can limit JAK-STAT-dependent signalling pathways via several mechanisms. These novel findings support AMPK activation as a strategy for management of an array of disorders characterised by hyper-activation of the JAK-STAT pathway. This review discusses the pivotal role of JAK-STAT signalling in a range of disorders and how both established clinically used and novel AMPK activators might be used to treat these conditions.
Collapse
Affiliation(s)
- Claire Speirs
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Jamie J L Williams
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Kirsten Riches
- School of Chemistry and Biosciences, University of Bradford, Bradford BD7 1DP, UK
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Timothy M Palmer
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford BD7 1DP, UK.
| |
Collapse
|
28
|
Calle-Guisado V, Hurtado de Llera A, González-Fernández L, Bragado MJ, Garcia-Marin LJ. Human sperm motility is downregulated by the AMPK activator A769662. Andrology 2017; 5:1131-1140. [DOI: 10.1111/andr.12423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 07/18/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022]
Affiliation(s)
- V. Calle-Guisado
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP); Institute of Biotechnology in Agriculture and Livestock (INBIO G+C); University of Extremadura; Caceres Spain
| | - A. Hurtado de Llera
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP); Institute of Biotechnology in Agriculture and Livestock (INBIO G+C); University of Extremadura; Caceres Spain
| | - L. González-Fernández
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP); Institute of Biotechnology in Agriculture and Livestock (INBIO G+C); University of Extremadura; Caceres Spain
- CECA/ICETA-Animal Science Centre; ICBAS-University of Porto; Vairão Portugal
| | - M. J. Bragado
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP); Institute of Biotechnology in Agriculture and Livestock (INBIO G+C); University of Extremadura; Caceres Spain
| | - L. J. Garcia-Marin
- Research Group of Intracellular Signalling and Technology of Reproduction (SINTREP); Institute of Biotechnology in Agriculture and Livestock (INBIO G+C); University of Extremadura; Caceres Spain
| |
Collapse
|
29
|
Guigas B, Viollet B. Targeting AMPK: From Ancient Drugs to New Small-Molecule Activators. ACTA ACUST UNITED AC 2017; 107:327-350. [PMID: 27812986 DOI: 10.1007/978-3-319-43589-3_13] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The AMP-activated protein kinase (AMPK) is an evolutionary conserved and ubiquitously expressed serine/threonine kinase mainly acting as a key regulator of cellular energy homeostasis. AMPK is a heterotrimeric protein complex, consisting of a catalytic α subunit and two regulatory β and γ subunits, whose activity is tightly regulated by changes in adenine nucleotides and several posttranslational modifications. Once activated in response to energy deficit, AMPK concomitantly inhibits ATP-consuming anabolic processes and promotes ATP-generating catabolic pathways via direct phosphorylation of multiple downstream effectors, leading to restoration of cellular energy balance. A growing number of energy/nutrient-independent functions of AMPK are also regularly reported, progressively expanding its role to regulation of non-metabolic cellular processes. Historically, AMPK as a therapeutic target has attracted much of interest due to its potential impact on metabolic disorders, such as obesity and type 2 diabetes, but has also recently received considerable renewed attention in the framework of cancer studies, highlighting the persistent need for selective, reversible, potent, and tissue-specific activators. In this chapter, we review the most recent advances in the understanding of the mechanism(s) of action of the current portfolio of AMPK activators, including plant-derived natural compounds and newly discovered small-molecule agonists directly targeting various AMPK subunits.
Collapse
Affiliation(s)
- Bruno Guigas
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Parasitology, Leiden University Medical Center, 9600, Postzone L40-Q, 2300 RC, Leiden, The Netherlands.
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
| |
Collapse
|
30
|
Chen L, Chen Q, Rong P, Wang HY, Chen S. The energy sensing LKB1-AMPKα1 pathway regulates IGF1 secretion and consequent activation of the IGF1R-PKB pathway in primary hepatocytes. FEBS J 2017; 284:2096-2109. [PMID: 28500773 DOI: 10.1111/febs.14106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 05/05/2017] [Accepted: 05/10/2017] [Indexed: 12/22/2022]
Abstract
The insulin-like growth factor 1 (IGF1) pathway has been linked with various diseases including diabetes, cancer and aging. In contrast to the well-established regulatory mechanisms controlling IGF1 expression, molecular mechanisms regulating its secretion are not fully understood. The AMP-activated protein kinase (AMPK) is a key energy sensor, and cumulative evidence shows that it is an attractive therapeutic target for treatment of diabetes, cancer and aging. Here we found that deficiency of AMPK promoted IGF1 secretion in mouse primary hepatocytes. Furthermore, we found that AMPKα1 but not AMPKα2 was involved in regulation of IGF1 secretion in mouse primary hepatocytes. Knockout of AMPK caused activation of the IGF1 receptor (IGF1R)-protein kinase B (PKB; also known as Akt) pathway in hepatocytes, which was mediated by hypersecretion of IGF1. Upstream of AMPK, liver kinase B1 (LKB1) was responsible for AMPK-dependent suppression of IGF1 secretion in hepatocytes. Collectively, these findings demonstrate that the energy-sensing LKB1-AMPK pathway regulates IGF1 secretion in mouse primary hepatocytes, which in turn regulates activation of the IGF1R-PKB pathway.
Collapse
Affiliation(s)
- Liang Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, China
| | - Qiaoli Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, China
| | - Ping Rong
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, China
| | - Hong Yu Wang
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, China
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, China
| |
Collapse
|
31
|
López M. EJE PRIZE 2017: Hypothalamic AMPK: a golden target against obesity? Eur J Endocrinol 2017; 176:R235-R246. [PMID: 28232370 PMCID: PMC5425938 DOI: 10.1530/eje-16-0927] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/26/2017] [Accepted: 02/22/2017] [Indexed: 12/16/2022]
Abstract
AMP-activated protein kinase (AMPK) is a cellular gauge that is activated under conditions, such as low energy, increasing energy production and reducing energy waste. Centrally, the AMPK pathway is a canonical route regulating energy homeostasis, by integrating peripheral signals, such as hormones and metabolites, with neuronal networks. Current evidence links hypothalamic AMPK with feeding, brown adipose tissue (BAT) thermogenesis and browning of white adipose tissue (WAT), as well as muscle metabolism, hepatic function and glucose homeostasis. The relevance of these data is interesting from a therapeutic point of view as several agents with potential anti-obesity and/or antidiabetic effects, some currently in clinical use, such as nicotine, metformin and liraglutide are known to act through AMPK, either peripherally or centrally. Furthermore, the orexigenic and weight-gaining effects of the worldwide use of antipsychotic drugs (APDs), such as olanzapine, are also mediated by hypothalamic AMPK. Overall, this evidence makes hypothalamic AMPK signaling an interesting target for the drug development, with its potential for controlling both sides of the energy balance equation, namely feeding and energy expenditure through defined metabolic pathways.
Collapse
Affiliation(s)
- Miguel López
- Department of PhysiologyNeurObesity Group, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Correspondence should be addressed to M López;
| |
Collapse
|
32
|
López M, Tena-Sempere M. Estradiol effects on hypothalamic AMPK and BAT thermogenesis: A gateway for obesity treatment? Pharmacol Ther 2017; 178:109-122. [PMID: 28351720 DOI: 10.1016/j.pharmthera.2017.03.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/21/2017] [Indexed: 12/24/2022]
Abstract
In addition to their prominent roles in the control of reproduction, estrogens are important modulators of energy balance, as evident in conditions of deficiency of estrogens, which are characterized by increased feeding and decreased energy expenditure, leading to obesity. AMP-activated protein kinase (AMPK) is a ubiquitous cellular energy gauge that is activated under conditions of low energy, increasing energy production and reducing energy wasting. Centrally, the AMPK pathway is a canonical route regulating energy homeostasis, by integrating peripheral signals, such as hormones and metabolites, with neuronal networks. As a result of those actions, hypothalamic AMPK modulates feeding, as well as brown adipose tissue (BAT) thermogenesis and browning of white adipose tissue (WAT). Here, we will review the central actions of estrogens on energy balance, with particular focus on hypothalamic AMPK. The relevance of this interaction is noteworthy, because some agents with known actions on metabolic homeostasis, such as nicotine, metformin, liraglutide, olanzapine and also natural molecules, such as resveratrol and flavonoids, exert their actions by modulating AMPK. This evidence highlights the possibility that hypothalamic AMPK might be a potential target for the treatment of obesity.
Collapse
Affiliation(s)
- Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos II, Spain.
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos II, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Reina Sofía, 14004 Córdoba, Spain; FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland.
| |
Collapse
|
33
|
Nguyen TMD. Impact of 5'-amp-activated Protein Kinase on Male Gonad and Spermatozoa Functions. Front Cell Dev Biol 2017; 5:25. [PMID: 28386541 PMCID: PMC5362614 DOI: 10.3389/fcell.2017.00025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022] Open
Abstract
As we already know, the male reproductive system requires less energetic investment than the female one. Nevertheless, energy balance is an important feature for spermatozoa production in the testis and for spermatozoa properties after ejaculation. The 5'-AMP-activated protein kinase, AMPK, is a sensor of cell energy, that regulates many metabolic pathways and that has been recently shown to control spermatozoa quality and functions. It is indeed involved in the regulation of spermatozoa quality through its action on the proliferation of testicular somatic cells (Sertoli and Leydig), on spermatozoa motility and acrosome reaction. It also favors spermatozoa quality through the management of lipid peroxidation and antioxidant enzymes. I review here the most recent data available on the roles of AMPK in vertebrate spermatozoa functions.
Collapse
Affiliation(s)
- Thi Mong Diep Nguyen
- Physiologie de la Reproduction et des Comportements, INRANouzilly, France; Quy Nhon UniversityQuy Nhon, Vietnam
| |
Collapse
|
34
|
Guo X, Huang X, Chen MJ. Reversible phosphorylation of the 26S proteasome. Protein Cell 2017; 8:255-272. [PMID: 28258412 PMCID: PMC5359188 DOI: 10.1007/s13238-017-0382-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/26/2017] [Indexed: 01/09/2023] Open
Abstract
The 26S proteasome at the center of the ubiquitin-proteasome system (UPS) is essential for virtually all cellular processes of eukaryotes. A common misconception about the proteasome is that, once made, it remains as a static and uniform complex with spontaneous and constitutive activity for protein degradation. Recent discoveries have provided compelling evidence to support the exact opposite insomuch as the 26S proteasome undergoes dynamic and reversible phosphorylation under a variety of physiopathological conditions. In this review, we summarize the history and current understanding of proteasome phosphorylation, and advocate the idea of targeting proteasome kinases/phosphatases as a new strategy for clinical interventions of several human diseases.
Collapse
Affiliation(s)
- Xing Guo
- The Life Sciences Institute of Zhejiang University, Hangzhou, 310058, China.
| | - Xiuliang Huang
- Ministry of Education Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Mark J Chen
- Department of Bioinformatics and Computational Biology, Genentech Inc., South San Francisco, CA, 94080, USA
| |
Collapse
|
35
|
Khan AS, Frigo DE. A spatiotemporal hypothesis for the regulation, role, and targeting of AMPK in prostate cancer. Nat Rev Urol 2017; 14:164-180. [PMID: 28169991 PMCID: PMC5672799 DOI: 10.1038/nrurol.2016.272] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The 5'-AMP-activated protein kinase (AMPK) is a master regulator of cellular homeostasis. Despite AMPK's known function in physiology, its role in pathological processes such as prostate cancer is enigmatic. However, emerging evidence is now beginning to decode the paradoxical role of AMPK in cancer and, therefore, inform clinicians if - and how - AMPK could be therapeutically targeted. Spatiotemporal regulation of AMPK complexes could be one of the mechanisms that governs this kinase's role in cancer. We hypothesize that different upstream stimuli will activate select subcellular AMPK complexes. This hypothesis is supported by the distinct subcellular locations of the various AMPK subunits. Each of these unique AMPK complexes regulates discrete downstream processes that can be tumour suppressive or oncogenic. AMPK's final biological output is then determined by the weighted net function of these downstream signalling events, influenced by additional prostate-specific signalling.
Collapse
Affiliation(s)
- Ayesha S. Khan
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX USA
| | - Daniel E. Frigo
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX USA
- Genomic Medicine Program, The Houston Methodist Research Institute, Houston, TX USA
| |
Collapse
|
36
|
Mancini SJ, White AD, Bijland S, Rutherford C, Graham D, Richter EA, Viollet B, Touyz RM, Palmer TM, Salt IP. Activation of AMP-activated protein kinase rapidly suppresses multiple pro-inflammatory pathways in adipocytes including IL-1 receptor-associated kinase-4 phosphorylation. Mol Cell Endocrinol 2017; 440:44-56. [PMID: 27840174 PMCID: PMC5228585 DOI: 10.1016/j.mce.2016.11.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/29/2022]
Abstract
Inflammation of adipose tissue in obesity is associated with increased IL-1β, IL-6 and TNF-α secretion and proposed to contribute to insulin resistance. AMP-activated protein kinase (AMPK) regulates nutrient metabolism and is reported to have anti-inflammatory actions in adipose tissue, yet the mechanisms underlying this remain poorly characterised. The effect of AMPK activation on cytokine-stimulated proinflammatory signalling was therefore assessed in cultured adipocytes. AMPK activation inhibited IL-1β-stimulated CXCL10 secretion, associated with reduced interleukin-1 receptor associated kinase-4 (IRAK4) phosphorylation and downregulated MKK4/JNK and IKK/IκB/NFκB signalling. AMPK activation inhibited TNF-α-stimulated IKK/IκB/NFκB signalling but had no effect on JNK phosphorylation. The JAK/STAT3 pathway was also suppressed by AMPK after IL-6 stimulation and during adipogenesis. Adipose tissue from AMPKα1-/- mice exhibited increased JNK and STAT3 phosphorylation, supporting suppression of these distinct proinflammatory pathways by AMPK in vivo. The inhibition of multiple pro-inflammatory signalling pathways by AMPK may underlie the reported beneficial effects of AMPK activation in adipose tissue.
Collapse
Affiliation(s)
- Sarah J Mancini
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Anna D White
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Silvia Bijland
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Claire Rutherford
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Timothy M Palmer
- School of Pharmacy, University of Bradford, Bradford, West Yorkshire, BD7 1DP, United Kingdom
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom.
| |
Collapse
|
37
|
Ruiz S, Carrera C, Villuendas P, Urriolabeitia EP. Ru-Catalysed synthesis of fused heterocycle-pyridinones and -pyrones. Org Biomol Chem 2017; 15:8904-8913. [DOI: 10.1039/c7ob01497j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Heterocycle-pyridinones and heterocycle-pyranones have been prepared by Ru-catalysed oxidative coupling of N-unprotected primary heterocycle-amides and heterocycle-carboxylic acids with internal alkynes.
Collapse
Affiliation(s)
- S. Ruiz
- Instituto de Síntesis Química y Catálisis Homogénea
- CSIC-Universidad de Zaragoza
- E-50009 Zaragoza
- Spain
| | - C. Carrera
- Instituto de Síntesis Química y Catálisis Homogénea
- CSIC-Universidad de Zaragoza
- E-50009 Zaragoza
- Spain
| | - P. Villuendas
- Instituto de Síntesis Química y Catálisis Homogénea
- CSIC-Universidad de Zaragoza
- E-50009 Zaragoza
- Spain
| | - E. P. Urriolabeitia
- Instituto de Síntesis Química y Catálisis Homogénea
- CSIC-Universidad de Zaragoza
- E-50009 Zaragoza
- Spain
| |
Collapse
|
38
|
Nandipati KC, Subramanian S, Agrawal DK. Protein kinases: mechanisms and downstream targets in inflammation-mediated obesity and insulin resistance. Mol Cell Biochem 2016; 426:27-45. [PMID: 27868170 DOI: 10.1007/s11010-016-2878-8] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/07/2016] [Indexed: 12/23/2022]
Abstract
Obesity-induced low-grade inflammation (metaflammation) impairs insulin receptor signaling. This has been implicated in the development of insulin resistance. Insulin signaling in the target tissues is mediated by stress kinases such as p38 mitogen-activated protein kinase, c-Jun NH2-terminal kinase, inhibitor of NF-kB kinase complex β (IKKβ), AMP-activated protein kinase, protein kinase C, Rho-associated coiled-coil containing protein kinase, and RNA-activated protein kinase. Most of these kinases phosphorylate several key regulators in glucose homeostasis. The phosphorylation of serine residues in the insulin receptor and IRS-1 molecule results in diminished enzymatic activity in the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. This has been one of the key mechanisms observed in the tissues that are implicated in insulin resistance especially in type 2 diabetes mellitus (T2-DM). Identifying the specific protein kinases involved in obesity-induced chronic inflammation may help in developing the targeted drug therapies to minimize the insulin resistance. This review is focused on the protein kinases involved in the inflammatory cascade and molecular mechanisms and their downstream targets with special reference to obesity-induced T2-DM.
Collapse
Affiliation(s)
- Kalyana C Nandipati
- Department of Surgery, Creighton University School of Medicine, 601 N. 30th Street, Suite # 3700, Omaha, NE, 68131, USA.
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA.
| | - Saravanan Subramanian
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA
| |
Collapse
|
39
|
Cameron KO, Kurumbail RG. Recent progress in the identification of adenosine monophosphate-activated protein kinase (AMPK) activators. Bioorg Med Chem Lett 2016; 26:5139-5148. [PMID: 27727125 DOI: 10.1016/j.bmcl.2016.09.065] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/20/2016] [Accepted: 09/26/2016] [Indexed: 12/31/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK), a serine/threonine heterotrimeric protein kinase, is a critical regulator of cellular and whole body energy homeostasis. There are twelve known AMPK isoforms that are differentially expressed in tissues and species. Dysregulation of AMPK signaling is associated with a multitude of human pathologies. Hence isoform-selective activators of AMPK are actively being sought for the treatment of cardiovascular and metabolic diseases. The present review summarizes the status of direct AMPK activators from the patent and published literature.
Collapse
Affiliation(s)
- Kimberly O Cameron
- Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Chemistry, 610 Main Street, Cambridge, MA 02139, USA.
| | - Ravi G Kurumbail
- Pfizer Global Research and Development, Worldwide Medicinal Chemistry, Eastern Point Road, Groton, CT 06340, USA
| |
Collapse
|
40
|
Kim J, Yang G, Kim Y, Kim J, Ha J. AMPK activators: mechanisms of action and physiological activities. Exp Mol Med 2016; 48:e224. [PMID: 27034026 PMCID: PMC4855276 DOI: 10.1038/emm.2016.16] [Citation(s) in RCA: 546] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 12/28/2015] [Accepted: 12/29/2015] [Indexed: 02/07/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is a central regulator of energy homeostasis, which coordinates metabolic pathways and thus balances nutrient supply with energy demand. Because of the favorable physiological outcomes of AMPK activation on metabolism, AMPK has been considered to be an important therapeutic target for controlling human diseases including metabolic syndrome and cancer. Thus, activators of AMPK may have potential as novel therapeutics for these diseases. In this review, we provide a comprehensive summary of both indirect and direct AMPK activators and their modes of action in relation to the structure of AMPK. We discuss the functional differences among isoform-specific AMPK complexes and their significance regarding the development of novel AMPK activators and the potential for combining different AMPK activators in the treatment of human disease.
Collapse
Affiliation(s)
- Joungmok Kim
- Depatment of Oral Biochemistry and Molecular Biology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Goowon Yang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Yeji Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Jin Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, Graduate School, Kyung Hee University, Seoul, Korea
| |
Collapse
|
41
|
Ross E, Ata R, Thavarajah T, Medvedev S, Bowden P, Marshall JG, Antonescu CN. AMP-Activated Protein Kinase Regulates the Cell Surface Proteome and Integrin Membrane Traffic. PLoS One 2015; 10:e0128013. [PMID: 26010094 PMCID: PMC4444004 DOI: 10.1371/journal.pone.0128013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/21/2015] [Indexed: 12/11/2022] Open
Abstract
The cell surface proteome controls numerous cellular functions including cell migration and adhesion, intercellular communication and nutrient uptake. Cell surface proteins are controlled by acute changes in protein abundance at the plasma membrane through regulation of endocytosis and recycling (endomembrane traffic). Many cellular signals regulate endomembrane traffic, including metabolic signaling; however, the extent to which the cell surface proteome is controlled by acute regulation of endomembrane traffic under various conditions remains incompletely understood. AMP-activated protein kinase (AMPK) is a key metabolic sensor that is activated upon reduced cellular energy availability. AMPK activation alters the endomembrane traffic of a few specific proteins, as part of an adaptive response to increase energy intake and reduce energy expenditure. How increased AMPK activity during energy stress may globally regulate the cell surface proteome is not well understood. To study how AMPK may regulate the cell surface proteome, we used cell-impermeable biotinylation to selectively purify cell surface proteins under various conditions. Using ESI-MS/MS, we found that acute (90 min) treatment with the AMPK activator A-769662 elicits broad control of the cell surface abundance of diverse proteins. In particular, A-769662 treatment depleted from the cell surface proteins with functions in cell migration and adhesion. To complement our mass spectrometry results, we used other methods to show that A-769662 treatment results in impaired cell migration. Further, A-769662 treatment reduced the cell surface abundance of β1-integrin, a key cell migration protein, and AMPK gene silencing prevented this effect. While the control of the cell surface abundance of various proteins by A-769662 treatment was broad, it was also selective, as this treatment did not change the cell surface abundance of the transferrin receptor. Hence, the cell surface proteome is subject to acute regulation by treatment with A-769662, at least some of which is mediated by the metabolic sensor AMPK.
Collapse
Affiliation(s)
- Eden Ross
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Rehman Ata
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Thanusi Thavarajah
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Sergei Medvedev
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Peter Bowden
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - John G Marshall
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| |
Collapse
|
42
|
Mo JS, Meng Z, Kim YC, Park HW, Hansen CG, Kim S, Lim DS, Guan KL. Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat Cell Biol 2015; 17:500-10. [PMID: 25751140 PMCID: PMC4380774 DOI: 10.1038/ncb3111] [Citation(s) in RCA: 435] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/14/2015] [Indexed: 12/15/2022]
Abstract
YAP (Yes-associated protein) is a transcription co-activator in the Hippo tumor suppressor pathway and controls cell growth, tissue homeostasis, and organ size. YAP is inhibited by the kinase Lats, which phosphorylates YAP to induce its cytoplasmic localization and proteasomal degradation. YAP induces gene expression by binding to the TEAD family transcription factors. Dysregulation of the Hippo-YAP pathway is frequently observed in human cancers. Here we show that cellular energy stress induces YAP phosphorylation, in part due to AMPK-dependent Lats activation, thereby inhibiting YAP activity. Moreover, AMPK directly phosphorylates YAP S94, a residue essential for the interaction with TEAD, thus disrupting the YAP-TEAD interaction. AMPK-induced YAP inhibition can suppress oncogenic transformation of Lats-null cells with high YAP activity. Our study establishes a molecular mechanism and functional significance of AMPK in linking cellular energy status to the Hippo-YAP pathway.
Collapse
Affiliation(s)
- Jung-Soon Mo
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Young Chul Kim
- Department of Cardiology, Veterans Medical Research Foundation, 3350 La Jolla Village Dr., San Diego, California 92161, USA
| | - Hyun Woo Park
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Carsten Gram Hansen
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Soohyun Kim
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Dae-Sik Lim
- National Creative Research Initiatives Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Republic of Korea
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
43
|
Weston AH, Egner I, Dong Y, Porter EL, Heagerty AM, Edwards G. Stimulated release of a hyperpolarizing factor (ADHF) from mesenteric artery perivascular adipose tissue: involvement of myocyte BKCa channels and adiponectin. Br J Pharmacol 2014; 169:1500-9. [PMID: 23488724 DOI: 10.1111/bph.12157] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 12/31/2012] [Accepted: 03/03/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Perivascular adipose tissue (PVAT) releases adipocyte-derived hyperpolarizing factors (ADHFs) that may partly act by opening myocyte K(+) channels. The present study in rat and mouse mesenteric arteries aimed to identify the myocyte K(+) channel activated by PVAT and to determine whether adiponectin contributed to the hyperpolarizing effects of PVAT. EXPERIMENTAL APPROACH Myocyte membrane potential was recorded from de-endothelialized, non-contracted rat and mouse mesenteric arteries in the presence and absence of PVAT. KEY RESULTS The β3 -adrenoceptor agonist, CL-316,243 (10 μM), generated PVAT-dependent, iberiotoxin-sensitive myocyte hyperpolarizations resulting from BKCa channel opening and which were partially blocked by L-NMMA (100 μM). Adiponectin (5 μg·mL(-1) ) also produced iberiotoxin-sensitive hyperpolarizations in PVAT-denuded arterioles. Activation of myocyte AMP-activated protein kinase (AMPK) using 5 μM A-769662 also induced BKCa -mediated hyperpolarizations. Dorsomorphin abolished hyperpolarizations to CL-316,243, adiponectin and A-769662. In vessels from Adipo(-/-) mice, hyperpolarizations to CL-316,243 were absent whereas those to A-769662 and adiponectin were normal. In rat vessels, adipocyte-dependent hyperpolarizations were blocked by glibenclamide and clotrimazole but those to NS1619 (33 μM) were unaltered. CONCLUSIONS AND IMPLICATIONS Under basal, non-contracted conditions, β3 -adrenoceptor stimulation of PVAT releases an ADHF, which is probably adiponectin. This activates AMPK to open myocyte BKCa channels indirectly and additionally liberates NO, which also contributes to the observed PVAT-dependent myocyte hyperpolarizations. Clotrimazole and glibenclamide each reversed hyperpolarizations to adiponectin and A-769662, suggesting the involvement of myocyte TRPM4 channels in the ADHF-induced myocyte electrical changes mediated via the opening of BKCa channels.
Collapse
Affiliation(s)
- A H Weston
- The University of Manchester, Manchester, UK
| | | | | | | | | | | |
Collapse
|
44
|
Hindupur SK, Balaji SA, Saxena M, Pandey S, Sravan GS, Heda N, Kumar MV, Mukherjee G, Dey D, Rangarajan A. Identification of a novel AMPK-PEA15 axis in the anoikis-resistant growth of mammary cells. Breast Cancer Res 2014; 16:420. [PMID: 25096718 PMCID: PMC4303232 DOI: 10.1186/s13058-014-0420-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 07/25/2014] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Matrix detachment triggers anoikis, a form of apoptosis, in most normal epithelial cells, while acquisition of anoikis resistance is a prime requisite for solid tumor growth. Of note, recent studies have revealed that a small population of normal human mammary epithelial cells (HMECs) survive in suspension and generate multicellular spheroids termed 'mammospheres'. Therefore, understanding how normal HMECs overcome anoikis may provide insights into breast cancer initiation and progression. METHODS Primary breast tissue-derived normal HMECs were grown as adherent monolayers or mammospheres. The status of AMP-activated protein kinase (AMPK) and PEA15 signaling was investigated by immunoblotting. Pharmacological agents and an RNA interference (RNAi) approach were employed to gauge their roles in mammosphere formation. Immunoprecipitation and in vitro kinase assays were undertaken to evaluate interactions between AMPK and PEA15. In vitro sphere formation and tumor xenograft assays were performed to understand their roles in tumorigenicity. RESULTS In this study, we show that mammosphere formation by normal HMECs is accompanied with an increase in AMPK activity. Inhibition or knockdown of AMPK impaired mammosphere formation. Concomitant with AMPK activation, we detected increased Ser116 phosphorylation of PEA15, which promotes its anti-apoptotic functions. Inhibition or knockdown of AMPK impaired PEA15 Ser116 phosphorylation and increased apoptosis. Knockdown of PEA15, or overexpression of the nonphosphorylatable S116A mutant of PEA15, also abrogated mammosphere formation. We further demonstrate that AMPK directly interacts with and phosphorylates PEA15 at Ser116 residue, thus identifying PEA15 as a novel AMPK substrate. Together, these data revealed that AMPK activation facilitates mammosphere formation by inhibition of apoptosis, at least in part, through Ser116 phosphorylation of PEA15. Since anoikis resistance plays a critical role in solid tumor growth, we investigated the relevance of these findings in the context of breast cancer. Significantly, we show that the AMPK-PEA15 axis plays an important role in the anchorage-independent growth of breast cancer cells both in vitro and in vivo. CONCLUSIONS Our study identifies a novel AMPK-PEA15 signaling axis in the anchorage-independent growth of both normal and cancerous mammary epithelial cells, suggesting that breast cancer cells may employ mechanisms of anoikis resistance already inherent within a subset of normal HMECs. Thus, targeting the AMPK-PEA15 axis might prevent breast cancer dissemination and metastasis.
Collapse
|
45
|
Coughlan KA, Valentine RJ, Ruderman NB, Saha AK. AMPK activation: a therapeutic target for type 2 diabetes? Diabetes Metab Syndr Obes 2014; 7:241-53. [PMID: 25018645 PMCID: PMC4075959 DOI: 10.2147/dmso.s43731] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disease characterized by insulin resistance, β-cell dysfunction, and elevated hepatic glucose output. Over 350 million people worldwide have T2D, and the International Diabetes Federation projects that this number will increase to nearly 600 million by 2035. There is a great need for more effective treatments for maintaining glucose homeostasis and improving insulin sensitivity. AMP-activated protein kinase (AMPK) is an evolutionarily conserved serine/threonine kinase whose activation elicits insulin-sensitizing effects, making it an ideal therapeutic target for T2D. AMPK is an energy-sensing enzyme that is activated when cellular energy levels are low, and it signals to stimulate glucose uptake in skeletal muscles, fatty acid oxidation in adipose (and other) tissues, and reduces hepatic glucose production. There is substantial evidence suggesting that AMPK is dysregulated in animals and humans with metabolic syndrome or T2D, and that AMPK activation (physiological or pharmacological) can improve insulin sensitivity and metabolic health. Numerous pharmacological agents, natural compounds, and hormones are known to activate AMPK, either directly or indirectly - some of which (for example, metformin and thiazolidinediones) are currently used to treat T2D. This paper will review the regulation of the AMPK pathway and its role in T2D, some of the known AMPK activators and their mechanisms of action, and the potential for future improvements in targeting AMPK for the treatment of T2D.
Collapse
Affiliation(s)
- Kimberly A Coughlan
- Endocrinology and Diabetes, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Rudy J Valentine
- Endocrinology and Diabetes, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Neil B Ruderman
- Endocrinology and Diabetes, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Asish K Saha
- Endocrinology and Diabetes, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| |
Collapse
|
46
|
Zadra G, Photopoulos C, Tyekucheva S, Heidari P, Weng QP, Fedele G, Liu H, Scaglia N, Priolo C, Sicinska E, Mahmood U, Signoretti S, Birnberg N, Loda M. A novel direct activator of AMPK inhibits prostate cancer growth by blocking lipogenesis. EMBO Mol Med 2014; 6:519-38. [PMID: 24497570 PMCID: PMC3992078 DOI: 10.1002/emmm.201302734] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
5′AMP-activated kinase (AMPK) constitutes a hub for cellular metabolic and growth control, thus representing an ideal therapeutic target for prostate cancers (PCas) characterized by increased lipogenesis and activation of mTORC1 pathway. However, whether AMPK activation itself is sufficient to block cancer cell growth remains to be determined. A small molecule screening was performed and identified MT 63–78, a specific and potent direct AMPK activator. Here, we show that direct activation of AMPK inhibits PCa cell growth in androgen sensitive and castration resistant PCa (CRPC) models, induces mitotic arrest, and apoptosis. In vivo, AMPK activation is sufficient to reduce PCa growth, whereas the allelic loss of its catalytic subunits fosters PCa development. Importantly, despite mTORC1 blockade, the suppression of de novo lipogenesis is the underpinning mechanism responsible for AMPK-mediated PCa growth inhibition, suggesting AMPK as a therapeutic target especially for lipogenesis-driven PCas. Finally, we demonstrate that MT 63–78 enhances the growth inhibitory effect of AR signaling inhibitors MDV3100 and abiraterone. This study thus provides a rationale for their combined use in CRPC treatment.
Collapse
Affiliation(s)
- Giorgia Zadra
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Taylor A, Westveld AH, Szkudlinska M, Guruguri P, Annabi E, Patwardhan A, Price TJ, Yassine HN. The use of metformin is associated with decreased lumbar radiculopathy pain. J Pain Res 2013; 6:755-63. [PMID: 24357937 PMCID: PMC3862700 DOI: 10.2147/jpr.s52205] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Lumbar radiculopathy pain represents a major public health problem, with few effective long-term treatments. Preclinical neuropathic and postsurgical pain studies implicate the kinase adenosine monophosphate activated kinase (AMPK) as a potential pharmacological target for the treatment of chronic pain conditions. Metformin, which acts via AMPK, is a safe and clinically available drug used in the treatment of diabetes. Despite the strong preclinical rationale, the utility of metformin as a potential pain therapeutic has not yet been studied in humans. Our objective was to assess whether metformin is associated with decreased lumbar radiculopathy pain, in a retrospective chart review. We completed a retrospective chart review of patients who sought care from a university pain specialist for lumbar radiculopathy between 2008 and 2011. Patients on metformin at the time of visit to a university pain specialist were compared with patients who were not on metformin. We compared the pain outcomes in 46 patients on metformin and 94 patients not taking metformin therapy. The major finding was that metformin use was associated with a decrease in the mean of "pain now," by -1.85 (confidence interval: -3.6 to -0.08) on a 0-10 visual analog scale, using a matched propensity scoring analysis and confirmed using a Bayesian analysis, with a significant mean decrease of -1.36 (credible interval: -2.6 to -0.03). Additionally, patients on metformin showed a non-statistically significant trend toward decreased pain on a variety of other pain descriptors. Our proof-of-concept findings suggest that metformin use is associated with a decrease in lumbar radiculopathy pain, providing a rational for larger retrospective trials in different pain populations and for prospective trials, to test the effectiveness of metformin in reducing neuropathic pain.
Collapse
Affiliation(s)
- Amber Taylor
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Anton H Westveld
- Statistics Laboratory, Bio5 Institute, Statistics GIDP, University of Arizona, Tucson, AZ, USA ; Faculty of ESTeM, University of Canberra, Canberra, ACT, Australia
| | | | | | - Emil Annabi
- Department of Anesthesia, University of Arizona, Tucson, AZ, USA
| | - Amol Patwardhan
- Department of Anesthesia, University of Arizona, Tucson, AZ, USA
| | - Theodore J Price
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Hussein N Yassine
- Department of Medicine, University of Southern California, LA, CA, USA
| |
Collapse
|
48
|
Abstract
Mitochondrial dysfunction is not only a hallmark of rare inherited mitochondrial disorders but also implicated in age-related diseases, including those that affect the metabolic and nervous system, such as type 2 diabetes and Parkinson's disease. Numerous pathways maintain and/or restore proper mitochondrial function, including mitochondrial biogenesis, mitochondrial dynamics, mitophagy and the mitochondrial unfolded protein response. New and powerful phenotypic assays in cell-based models as well as multicellular organisms have been developed to explore these different aspects of mitochondrial function. Modulating mitochondrial function has therefore emerged as an attractive therapeutic strategy for several diseases, which has spurred active drug discovery efforts in this area.
Collapse
|
49
|
Zadra G, Photopoulos C, Loda M. The fat side of prostate cancer. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1518-32. [PMID: 23562839 DOI: 10.1016/j.bbalip.2013.03.010] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 03/23/2013] [Accepted: 03/24/2013] [Indexed: 12/28/2022]
Abstract
Prostate cancer (PCa) metabolism appears to be unique in comparison with other types of solid cancers. Normal prostate cells mainly rely on glucose oxidation to provide precursors for the synthesis and secretion of citrate, resulting in an incomplete Krebs cycle and minimal oxidative phosphorylation for energy production. In contrast, during transformation, PCa cells no longer secrete citrate and they reactivate the Krebs cycle as energy source. Moreover, primary PCas do not show increased aerobic glycolysis and therefore they are not efficiently detectable with (18)F-FDG-PET. However, increased de novo lipid synthesis, strictly intertwined with deregulation in classical oncogenes and oncosuppressors, is an early event of the disease. Up-regulation and increased activity of lipogenic enzymes (including fatty acid synthase and choline kinase) occurs throughout PCa carcinogenesis and correlates with worse prognosis and poor survival. Thus, lipid precursors such as acetate and choline have been successfully used as alternative tracers for PET imaging. Lipid synthesis intermediates and FA catabolism also emerged as important players in PCa maintenance. Finally, epidemiologic studies suggested that systemic metabolic disorders including obesity, metabolic syndrome, and diabetes as well as hypercaloric and fat-rich diets might increase the risk of PCa. However, how metabolic disorders contribute to PCa development and whether dietary lipids and de novo lipids synthesized intra-tumor are differentially metabolized still remains unclear. In this review, we examine the switch in lipid metabolism supporting the development and progression of PCa and we discuss how we can exploit its lipogenic nature for therapeutic and diagnostic purposes. This article is part of a Special Issue entitled Lipid Metabolism in Cancer.
Collapse
Affiliation(s)
- Giorgia Zadra
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
50
|
Diabetes Mellitus: New Challenges and Innovative Therapies. NEW STRATEGIES TO ADVANCE PRE/DIABETES CARE: INTEGRATIVE APPROACH BY PPPM 2013; 3. [PMCID: PMC7120768 DOI: 10.1007/978-94-007-5971-8_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diabetes is a common chronic disease affecting an estimated 285 million adults worldwide. The rising incidence of diabetes, metabolic syndrome, and subsequent vascular diseases is a major public health problem in industrialized countries. This chapter summarizes current pharmacological approaches to treat diabetes mellitus and focuses on novel therapies for diabetes mellitus that are under development. There is great potential for developing a new generation of therapeutics that offer better control of diabetes, its co-morbidities and its complications. Preclinical results are discussed for new approaches including AMPK activation, the FGF21 target, cell therapy approaches, adiponectin mimetics and novel insulin formulations. Gene-based therapies are among the most promising emerging alternatives to conventional treatments. Therapies based on gene silencing using vector systems to deliver interference RNA to cells (i.e. against VEGF in diabetic retinopathy) are also a promising therapeutic option for the treatment of several diabetic complications. In conclusion, treatment of diabetes faces now a new era that is characterized by a variety of innovative therapeutic approaches that will improve quality of life in the near future.
Collapse
|