1
|
Biswas D, Chakraborty A, Mukherjee S, Ghosh B. Hairy root culture: a potent method for improved secondary metabolite production of Solanaceous plants. FRONTIERS IN PLANT SCIENCE 2023; 14:1197555. [PMID: 37731987 PMCID: PMC10507345 DOI: 10.3389/fpls.2023.1197555] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/13/2023] [Indexed: 09/22/2023]
Abstract
Secondary metabolites synthesized by the Solanaceous plants are of major therapeutic and pharmaceutical importance, many of which are commonly obtained from the roots of these plants. 'Hairy roots', mirroring the same phytochemical pattern of the corresponding root of the parent plant with higher growth rate and productivity, are therefore extensively studied as an effective alternative for the in vitro production of these metabolites. Hairy roots are the transformed roots, generated from the infection site of the wounded plants with Agrobacterium rhizogenes. With their fast growth, being free from pathogen and herbicide contamination, genetic stability, and autotrophic nature for plant hormones, hairy roots are considered as useful bioproduction systems for specialized metabolites. Lately, several elicitation methods have been employed to enhance the accumulation of these compounds in the hairy root cultures for both small and large-scale production. Nevertheless, in the latter case, the cultivation of hairy roots in bioreactors should still be optimized. Hairy roots can also be utilized for metabolic engineering of the regulatory genes in the metabolic pathways leading to enhanced production of metabolites. The present study summarizes the updated and modern biotechnological aspects for enhanced production of secondary metabolites in the hairy root cultures of the plants of Solanaceae and their respective importance.
Collapse
Affiliation(s)
- Diptesh Biswas
- Plant Biotechnology Laboratory, Post Graduate Department of Botany, Ramakrishna Mission Vivekananda Centenary College, Kolkata, India
| | - Avijit Chakraborty
- Plant Biotechnology Laboratory, Post Graduate Department of Botany, Ramakrishna Mission Vivekananda Centenary College, Kolkata, India
| | - Swapna Mukherjee
- Department of Microbiology, Dinabandhu Andrews College, Kolkata, India
| | - Biswajit Ghosh
- Plant Biotechnology Laboratory, Post Graduate Department of Botany, Ramakrishna Mission Vivekananda Centenary College, Kolkata, India
| |
Collapse
|
2
|
Hela F, Aguayo-Mazzucato C. Interaction between Autophagy and Senescence in Pancreatic Beta Cells. BIOLOGY 2023; 12:1205. [PMID: 37759604 PMCID: PMC10525299 DOI: 10.3390/biology12091205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023]
Abstract
Aging leads to an increase in cellular stress due to the fragility of the organism and the inability to cope with it. In this setting, there is a higher chance of developing different cardiometabolic diseases like diabetes. Cellular senescence and autophagy, both hallmarks of aging and stress-coping mechanisms, have gained increased attention for their role in the pathophysiology of diabetes. Studies show that impairing senescence dampens and even prevents diabetes while the role of autophagy is more contradictory, implying a context- and disease-stage-dependent effect. Reports show conflicting data about the effect of autophagy on senescence while the knowledge about this interaction in beta cells remains scarce. Elucidating this interaction between autophagy and senescence in pancreatic beta cells will lead to an identification of their respective roles and the extent of the effect each mechanism has on beta cells and open new horizons for developing novel therapeutic agents. To help illuminate this relationship we will review the latest findings of cellular senescence and autophagy with a special emphasis on pancreatic beta cells and diabetes.
Collapse
Affiliation(s)
| | - Cristina Aguayo-Mazzucato
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
3
|
Gašparini D, Kavazović I, Barković I, Maričić V, Ivaniš V, Samsa DT, Peršić V, Polić B, Turk Wensveen T, Wensveen FM. Extreme anaerobic exercise causes reduced cytotoxicity and increased cytokine production by peripheral blood lymphocytes. Immunol Lett 2022; 248:45-55. [PMID: 35709930 DOI: 10.1016/j.imlet.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/05/2022]
Abstract
Exercise has many beneficial effects for our body, but can become detrimental at high intensity, especially for our immune system. Little is known about the underlying mechanism of impaired immune functionality under conditions of intense physical strain. Freedivers, people who dive to high depths on a single breath, perform extreme exercise under anaerobic conditions. In this study, we investigated the impact of freediving on the cytotoxic arm of the immune system. At rest, elite freedivers did not display changes in their immunological profile compared to non-diving controls. In contrast, after a freedive, granzyme B and IL-2 production were reduced, whereas IFNγ and TNF secretion were increased by cytotoxic immune cells. Using in vitro models mimicking freedive conditions, we could show that hypoxia in combination with stress hyperglycemia had a negative impact on Granzyme B secretion, whereas IL-2 production was inhibited by stress hormones. Our findings suggest that in response to extreme exercise, cytotoxic immune cells transiently change their functional profile to limit tissue damage.
Collapse
Affiliation(s)
- Dora Gašparini
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, Rijeka 51000, Croatia; Center for Diabetes, Endocrinology and Cardiometabolism, Special Hospital for Medical Rehabilitation of Heart, Lung and Rheumatic Diseases Thalassotherapia Opatija, Opatija, Croatia
| | - Inga Kavazović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, Rijeka 51000, Croatia
| | - Igor Barković
- Center for Research and Education in Underwater, Hyperbaric and Maritime Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vitomir Maričić
- International Association for the Development of Apnea, Croatia
| | - Viktor Ivaniš
- Clinic for Heart and Blood Vessels, Special Hospital for Medical Rehabilitation of Heart, Lung and Rheumatic Diseases Thalassotherapia Opatija, Opatija, Croatia
| | - Dijana Travica Samsa
- Clinic for Heart and Blood Vessels, Special Hospital for Medical Rehabilitation of Heart, Lung and Rheumatic Diseases Thalassotherapia Opatija, Opatija, Croatia; Department of Rehabilitation and Sports Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Viktor Peršić
- Clinic for Heart and Blood Vessels, Special Hospital for Medical Rehabilitation of Heart, Lung and Rheumatic Diseases Thalassotherapia Opatija, Opatija, Croatia; Department of Rehabilitation and Sports Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, Rijeka 51000, Croatia
| | - Tamara Turk Wensveen
- Center for Diabetes, Endocrinology and Cardiometabolism, Special Hospital for Medical Rehabilitation of Heart, Lung and Rheumatic Diseases Thalassotherapia Opatija, Opatija, Croatia; Department of Internal Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Department of Endocrinology, Diabetology and Metabolic Diseases, Clinic for Internal Medicine, Clinical Hospital Centre Rijeka, Rijeka, Croatia
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, Rijeka 51000, Croatia.
| |
Collapse
|
4
|
Karsai M, Zuellig RA, Lehmann R, Cuozzo F, Nasteska D, Luca E, Hantel C, Hodson DJ, Spinas GA, Rutter GA, Gerber PA. Lack of ZnT8 protects pancreatic islets from hypoxia- and cytokine-induced cell death. J Endocrinol 2022; 253:1-11. [PMID: 35017316 PMCID: PMC8859919 DOI: 10.1530/joe-21-0271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/11/2022] [Indexed: 11/10/2022]
Abstract
Pancreatic β-cells depend on the well-balanced regulation of cytosolic zinc concentrations, providing sufficient zinc ions for the processing and storage of insulin, but avoiding toxic effects. The zinc transporter ZnT8, encoded by SLC30A8,is a key player regarding islet cell zinc homeostasis, and polymorphisms in this gene are associated with altered type 2 diabetes susceptibility in man. The objective of this study was to investigate the role of ZnT8 and zinc in situations of cellular stress as hypoxia or inflammation. Isolated islets of WT and global ZnT8-/- mice were exposed to hypoxia or cytokines and cell death was measured. To explore the role of changing intracellular Zn2+ concentrations, WT islets were exposed to different zinc concentrations using zinc chloride or the zinc chelator N,N,N',N'-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine (TPEN). Hypoxia or cytokine (TNF-α, IFN-γ, IL1-β) treatment induced islet cell death, but to a lesser extent in islets from ZnT8-/- mice, which were shown to have a reduced zinc content. Similarly, chelation of zinc with TPEN reduced cell death in WT islets treated with hypoxia or cytokines, whereas increased zinc concentrations aggravated the effects of these stressors. This study demonstrates a reduced rate of cell death in islets from ZnT8-/- mice as compared to WT islets when exposed to two distinct cellular stressors, hypoxia or cytotoxic cytokines. This protection from cell death is, in part, mediated by a reduced zinc content in islet cells of ZnT8-/- mice. These findings may be relevant for altered diabetes burden in carriers of risk SLC30A8 alleles in man.
Collapse
Affiliation(s)
- Maria Karsai
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Richard A Zuellig
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Roger Lehmann
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Federica Cuozzo
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Edlira Luca
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Giatgen A Spinas
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- CR-CHUM, University of Montreal, Montreal, QC, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Philipp A Gerber
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
5
|
Eguchi N, Vaziri ND, Dafoe DC, Ichii H. The Role of Oxidative Stress in Pancreatic β Cell Dysfunction in Diabetes. Int J Mol Sci 2021; 22:ijms22041509. [PMID: 33546200 PMCID: PMC7913369 DOI: 10.3390/ijms22041509] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetes is a chronic metabolic disorder characterized by inappropriately elevated glucose levels as a result of impaired pancreatic β cell function and insulin resistance. Extensive studies have been conducted to elucidate the mechanism involved in the development of β cell failure and death under diabetic conditions such as hyperglycemia, hyperlipidemia, and inflammation. Of the plethora of proposed mechanisms, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, and oxidative stress have been shown to play a central role in promoting β cell dysfunction. It has become more evident in recent years that these 3 factors are closely interrelated and importantly aggravate each other. Oxidative stress in particular is of great interest to β cell health and survival as it has been shown that β cells exhibit lower antioxidative capacity. Therefore, this review will focus on discussing factors that contribute to the development of oxidative stress in pancreatic β cells and explore the downstream effects of oxidative stress on β cell function and health. Furthermore, antioxidative capacity of β cells to counteract these effects will be discussed along with new approaches focused on preserving β cells under oxidative conditions.
Collapse
Affiliation(s)
- Natsuki Eguchi
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (D.C.D.)
| | | | - Donald C. Dafoe
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (D.C.D.)
| | - Hirohito Ichii
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (D.C.D.)
- Correspondence: ; Tel.: +1-714-456-8590
| |
Collapse
|
6
|
Behl T, Sharma A, Sharma L, Sehgal A, Zengin G, Brata R, Fratila O, Bungau S. Exploring the Multifaceted Therapeutic Potential of Withaferin A and Its Derivatives. Biomedicines 2020; 8:E571. [PMID: 33291236 PMCID: PMC7762146 DOI: 10.3390/biomedicines8120571] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Withaferin A (WA), a manifold studied, C28-steroidal lactone withanolide found in Withania somnifera. Given its unique beneficial effects, it has gathered attention in the era of modern science. Cancer, being considered a "hopeless case and the leading cause of death worldwide, and the available conventional therapies have many lacunae in the form of side effects. The poly pharmaceutical natural compound, WA treatment, displayed attenuation of various cancer hallmarks by altering oxidative stress, promoting apoptosis, and autophagy, inhibiting cell proliferation, reducing angiogenesis, and metastasis progression. The cellular proteins associated with antitumor pathways were also discussed. WA structural modifications attack multiple signal transduction pathways and enhance the therapeutic outcomes in various diseases. Moreover, it has shown validated pharmacological effects against multiple neurodegenerative diseases by inhibiting acetylcholesterinases and butyrylcholinesterases enzyme activity, antidiabetic activity by upregulating adiponectin and preventing the phosphorylation of peroxisome proliferator-activated receptors (PPARγ), cardioprotective activity by AMP-activated protein kinase (AMPK) activation and suppressing mitochondrial apoptosis. The current review is an extensive survey of various WA associated disease targets, its pharmacokinetics, synergistic combination, modifications, and biological activities.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya 42250, Turkey;
| | - Roxana Brata
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
7
|
Mroueh FM, Noureldein M, Zeidan YH, Boutary S, Irani SAM, Eid S, Haddad M, Barakat R, Harb F, Costantine J, Kanj R, Sauleau EA, Ouhtit A, Azar ST, Eid AH, Eid AA. Unmasking the interplay between mTOR and Nox4: novel insights into the mechanism connecting diabetes and cancer. FASEB J 2019; 33:14051-14066. [DOI: 10.1096/fj.201900396rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Fatima Mohsen Mroueh
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Mohamed Noureldein
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Youssef H. Zeidan
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
- Department of Radiation Oncology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Suzan Boutary
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Sara Abou Merhi Irani
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Stéphanie Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Mary Haddad
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Rasha Barakat
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Frederic Harb
- Department of Life and Earth Sciences, Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | - Joseph Costantine
- Department of Electrical and Computer Engineering, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut, Lebanon
| | - Rouwaida Kanj
- Department of Electrical and Computer Engineering, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut, Lebanon
| | - Erik-André Sauleau
- Department of Biostatistics, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7357 ICube, University of Strasbourg, Strasbourg, France
| | - Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Sami T. Azar
- Department of Internal Medicine, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
- American University of Beirut (AUB) Diabetes, Faculty of Medicine and Medical Center American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
- American University of Beirut (AUB) Diabetes, Faculty of Medicine and Medical Center American University of Beirut, Beirut, Lebanon
| |
Collapse
|
8
|
Zhang Y, Li Y, Ma P, Chen J, Xie W. Ficus carica leaves extract inhibited pancreatic β-cell apoptosis by inhibiting AMPK/JNK/caspase-3 signaling pathway and antioxidation. Biomed Pharmacother 2019; 122:109689. [PMID: 31786467 DOI: 10.1016/j.biopha.2019.109689] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022] Open
Abstract
The aim of this study was to explore the inhibitory effects of Ficus carica leaves (FCL) extract on AMPK/JNK/caspase3 signaling pathway and antioxidation in pancreatic β-cells. H&E staining, insulin immunohistochemistry, and TUNEL methods were used to investigate the effects of FCL on pancreatic histopathology in type 1 diabetic mice. The expression levels of caspase-3, AMPK, and JNK protein in the pancreatic tissue and MIN6 cells [induced by palmitic acid (PA) and hydrogen peroxide] were determined. Flow cytometry was used to detect the effects of FCL on apoptosis and ROS production of MIN6 cells. FCL (2 g/kg, continuous gavage for 6 weeks) significantly improved the pancreatic tissue injury in type 1 diabetic mice and reduced the expression levels of apoptosis-related proteins such as FasL, caspase8, Bax/Bcl-2, Cyt-C, caspase-3, p-AMPK, and p-JNK. FCL inhibited cell apoptosis induced by PA and the protein expression levels of caspase-3, p-AMPK, and p-JNK. The AMPK agonist AICAR could reverse the protective effects of FCL on MIN6 cells. The AMPK inhibitor compound C had a similar effect on MIN6 cells as that of FCL. FCL could inhibit cell apoptosis induced by hydrogen peroxide and reduced the production of ROS. In conclusion, FCL could inhibit pancreatic β-cell apoptosis by inhibiting the AMPK/JNK/caspase-3 signaling pathway and by antioxidation properties.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.
| | - Yingying Li
- Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.
| | - Ping Ma
- Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.
| | - Jincheng Chen
- Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.
| | - Weiping Xie
- Department of Physical and Chemical Analysis, Quanzhou Center for Disease Control and Prevention, Quanzhou, 362000, People's Republic of China.
| |
Collapse
|
9
|
Tekula S, Khurana A, Anchi P, Godugu C. Withaferin-A attenuates multiple low doses of Streptozotocin (MLD-STZ) induced type 1 diabetes. Biomed Pharmacother 2018; 106:1428-1440. [DOI: 10.1016/j.biopha.2018.07.090] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/14/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022] Open
|
10
|
Abstract
PURPOSE OF REVIEW To discuss advances in our understanding of beta-cell heterogeneity and the ramifications of this for type 1 diabetes (T1D) and its therapy. RECENT FINDINGS A number of studies have challenged the long-standing dogma that the majority of beta cells are eliminated in T1D. As many as 80% are present in some T1D subjects. Why don't these cells function properly to release insulin in response to high glucose? Other findings deploying single-cell "omics" to study both healthy and diseased cells-from patients with both T1D and type 2 diabetes (T2D)-have revealed cell subpopulations and heterogeneity at the transcriptomic/protein level between individual cells. Finally, our own and others' findings have demonstrated the importance of functional beta-cell subpopulations for insulin secretion. Heterogeneity may endow beta cells with molecular features that predispose them to failure/death during T1D.
Collapse
Affiliation(s)
- Richard K. P. Benninger
- 0000 0001 0703 675Xgrid.430503.1Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
- 0000 0001 0703 675Xgrid.430503.1Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Craig Dorrell
- 0000 0000 9758 5690grid.5288.7Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239 USA
| | - David J. Hodson
- 0000 0004 1936 7486grid.6572.6Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, B15 2TT UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, B15 2TH UK
- COMPARE, University of Birmingham and University of Nottingham Midlands, Nottingham, UK
| | - Guy A. Rutter
- 0000 0001 2113 8111grid.7445.2Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, W12 0NN UK
| |
Collapse
|
11
|
Wang H, Zhang H, Sun L, Guo W. Roles of hyperuricemia in metabolic syndrome and cardiac-kidney-vascular system diseases. Am J Transl Res 2018; 10:2749-2763. [PMID: 30323864 PMCID: PMC6176241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/14/2018] [Indexed: 06/08/2023]
Abstract
Uric acid is the final product of purine metabolism. Hyperuricemia is defined as a condition where the level of uric acid exceeds the normal range. The most well-known disease induced by hyperuricemia is gout. However, many studies have reported that hyperuricemia also plays important roles in cardiac-kidney-vascular system diseases and metabolic syndrome. Although hyperuricemia has been known for a long time, its pathophysiology remains poorly understood. In this review, we highlight studies on advanced pathological mechanisms for injuries induced by hyperuricemia, summarize epidemiological studies on hyperuricemia and its associated diseases, and take a brief look at hyperuricemia prevention.
Collapse
Affiliation(s)
- Hongsha Wang
- Department of Endocrinology, The First Hospital of Jilin UniversityChangchun 130021, China
| | - Haifeng Zhang
- Department of Interventional, The First Hospital of Jilin UniversityChangchun 130021, China
| | - Lin Sun
- Department of Endocrinology, The First Hospital of Jilin UniversityChangchun 130021, China
| | - Weiying Guo
- Department of Endocrinology, The First Hospital of Jilin UniversityChangchun 130021, China
| |
Collapse
|
12
|
Klangnurak W, Fukuyo T, Rezanujjaman MD, Seki M, Sugano S, Suzuki Y, Tokumoto T. Candidate gene identification of ovulation-inducing genes by RNA sequencing with an in vivo assay in zebrafish. PLoS One 2018; 13:e0196544. [PMID: 29715317 PMCID: PMC5929532 DOI: 10.1371/journal.pone.0196544] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/14/2018] [Indexed: 01/24/2023] Open
Abstract
We previously reported the microarray-based selection of three ovulation-related genes in zebrafish. We used a different selection method in this study, RNA sequencing analysis. An additional eight up-regulated candidates were found as specifically up-regulated genes in ovulation-induced samples. Changes in gene expression were confirmed by qPCR analysis. Furthermore, up-regulation prior to ovulation during natural spawning was verified in samples from natural pairing. Gene knock-out zebrafish strains of one of the candidates, the starmaker gene (stm), were established by CRISPR genome editing techniques. Unexpectedly, homozygous mutants were fertile and could spawn eggs. However, a high percentage of unfertilized eggs and abnormal embryos were produced from these homozygous females. The results suggest that the stm gene is necessary for fertilization. In this study, we selected additional ovulation-inducing candidate genes, and a novel function of the stm gene was investigated.
Collapse
Affiliation(s)
- Wanlada Klangnurak
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, Japan
| | - Taketo Fukuyo
- Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Shizuoka, Japan
| | - M. D. Rezanujjaman
- Biological Science Course, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Oya 836, Suruga-ku, Shizuoka, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Sumio Sugano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Toshinobu Tokumoto
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, Japan
- Department of Biological Science, Faculty of Science, National University Corporation Shizuoka University, Shizuoka, Japan
- Biological Science Course, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Oya 836, Suruga-ku, Shizuoka, Japan
- * E-mail:
| |
Collapse
|
13
|
Zhang B, Lakshmanan J, Du Y, Smith JW, Harbrecht BG. Cell-specific regulation of iNOS by AMP-activated protein kinase in primary rat hepatocytes. J Surg Res 2017; 221:104-112. [PMID: 29229115 DOI: 10.1016/j.jss.2017.08.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/21/2017] [Accepted: 08/14/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) regulates several metabolic pathways in hepatocytes that are critical to the hepatic response to sepsis and shock. Induction of nitric oxide synthesis is an important response to sepsis, inflammation and shock and many of the stimuli that upregulate inducible nitric oxide synthase (iNOS) also activate AMPK. AMPK inhibits nitric oxide (NO) production in skeletal and cardiac muscle cells, but the role of AMPK in regulating iNOS expression in hepatocytes has not been determined. MATERIALS AND METHODS Primary cultured rat hepatocytes were preincubated with an AMPK inhibitor, AMPK activators, or transfected with AMPK siRNA before being treated with the proinflammatory cytokines interleukin-1β (IL-1β) and interferon-γ (IFNγ). The hepatocyte cell lysate and culture supernatants were collected for Western blot analysis and Griess assay. RESULTS IL-1β and IFNγ markedly upregulated iNOS expression and AMPK phosphorylation. IL-1β + IFNγ-induced NO production and iNOS expression were significantly decreased in hepatocytes treated with the AMPK inhibitor compound C and AMPK knockdown by AMPK siRNA. Cytokine-induced iNOS expression was increased by AMPK activators 1-oxo-2-(2H-pyrrolium-1-yl)-1H-inden-3-olate, AMPK signaling activator III and AICA-riboside. Compound C upregulated Akt and c-Jun N-terminal kinase phosphorylation but decreased IκBα phosphorylation. AICA-riboside exerted opposite effects on these signaling pathways in hepatocytes. CONCLUSIONS In contrast to other cell types, AMPK increased IL-1β + IFNγ-induced NO production and iNOS expression through the Akt, c-Jun N-terminal kinase, and NF-κΒ signaling pathways in primary hepatocytes. These data suggest that AMPK-altering medications used clinically may have subsequent effects on iNOS expression and proinflammatory signaling pathways.
Collapse
Affiliation(s)
- Baochun Zhang
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky.
| | - Jaganathan Lakshmanan
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| | - Yibo Du
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| | - Jason W Smith
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| | - Brian G Harbrecht
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| |
Collapse
|
14
|
Ouyang Z, Wang X, Meng Q, Feng L, Sun Y, Wu X, Xu Q. Suppression of adenosine monophosphate-activated protein kinase selectively triggers apoptosis in activated T cells and ameliorates immune diseases. Biochem Biophys Res Commun 2017; 487:223-229. [PMID: 28412370 DOI: 10.1016/j.bbrc.2017.04.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/10/2017] [Indexed: 01/10/2023]
Abstract
Deficient apoptosis of activated T cells can result in immunological disorders. Molecules associated with energy and metabolisms are suggested to be involved in pathogenesis of immune diseases, but remain uninvestigated. In the present study we reported that glibenclamide exerted a new pharmacological effect on inflammatory responses by selectively triggering apoptosis of activated T cells. Glibenclamide demonstrated an inhibition on activated T lymphocytes, whereas showed no toxicity in the naive cells. This effect was mainly related with its ability to facilitate apoptosis in activated T cells with an up-regulation of cleaved-caspases and cleaved-PARP. Glibenclamide enhanced Fas expression and suppressed the expression of antiapoptotic cellular FLICE-inhibitory protein. The underlying mechanism of glibenclamide was not associated with its classical inhibitory effect on ATP-sensitive potassium channels, but due to a unique suppression on the phosphorylation of 5' adenosine monophosphate-activated protein kinase, which was augmented during T cell activation. An in vivo experiment further demonstrated that glibenclamide ameliorated T-cell-mediated contact hypersensitivity in mice. Altogether, these results suggest that AMPK inhibition by glibenclamide can regulate the survival and death of T lymphocytes and be beneficial for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Zijun Ouyang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Xingqi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Qianqian Meng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Lili Feng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China.
| |
Collapse
|
15
|
Marmugi A, Parnis J, Chen X, Carmichael L, Hardy J, Mannan N, Marchetti P, Piemonti L, Bosco D, Johnson P, Shapiro JAM, Cruciani-Guglielmacci C, Magnan C, Ibberson M, Thorens B, Valdivia HH, Rutter GA, Leclerc I. Sorcin Links Pancreatic β-Cell Lipotoxicity to ER Ca2+ Stores. Diabetes 2016; 65:1009-21. [PMID: 26822088 PMCID: PMC4806657 DOI: 10.2337/db15-1334] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/18/2016] [Indexed: 01/02/2023]
Abstract
Preserving β-cell function during the development of obesity and insulin resistance would limit the worldwide epidemic of type 2 diabetes. Endoplasmic reticulum (ER) calcium (Ca(2+)) depletion induced by saturated free fatty acids and cytokines causes β-cell ER stress and apoptosis, but the molecular mechanisms behind these phenomena are still poorly understood. Here, we demonstrate that palmitate-induced sorcin downregulation and subsequent increases in glucose-6-phosphatase catalytic subunit-2 (G6PC2) levels contribute to lipotoxicity. Sorcin is a calcium sensor protein involved in maintaining ER Ca(2+) by inhibiting ryanodine receptor activity and playing a role in terminating Ca(2+)-induced Ca(2+) release. G6PC2, a genome-wide association study gene associated with fasting blood glucose, is a negative regulator of glucose-stimulated insulin secretion (GSIS). High-fat feeding in mice and chronic exposure of human islets to palmitate decreases endogenous sorcin expression while levels of G6PC2 mRNA increase. Sorcin-null mice are glucose intolerant, with markedly impaired GSIS and increased expression of G6pc2 Under high-fat diet, mice overexpressing sorcin in the β-cell display improved glucose tolerance, fasting blood glucose, and GSIS, whereas G6PC2 levels are decreased and cytosolic and ER Ca(2+) are increased in transgenic islets. Sorcin may thus provide a target for intervention in type 2 diabetes.
Collapse
Affiliation(s)
- Alice Marmugi
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Imperial College London, London, U.K
| | - Julia Parnis
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Imperial College London, London, U.K
| | - Xi Chen
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI
| | - LeAnne Carmichael
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Imperial College London, London, U.K
| | - Julie Hardy
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Imperial College London, London, U.K
| | - Naila Mannan
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Imperial College London, London, U.K
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Paul Johnson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, U.K
| | - James A M Shapiro
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | | | - Christophe Magnan
- Unit of Functional and Adaptive Biology, Paris Diderot University-Paris 7, Paris, France
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Héctor H Valdivia
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Imperial College London, London, U.K.
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Imperial College London, London, U.K.
| |
Collapse
|
16
|
Chang TJ, Tseng HC, Liu MW, Chang YC, Hsieh ML, Chuang LM. Glucagon-like peptide-1 prevents methylglyoxal-induced apoptosis of beta cells through improving mitochondrial function and suppressing prolonged AMPK activation. Sci Rep 2016; 6:23403. [PMID: 26997114 PMCID: PMC4800673 DOI: 10.1038/srep23403] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/02/2016] [Indexed: 11/09/2022] Open
Abstract
Accumulation of methylglyoxal (MG) contributes to glucotoxicity and mediates beta cell apoptosis. The molecular mechanism by which GLP-1 protects MG-induced beta cell apoptosis remains unclear. Metformin is a first-line drug for treating type 2 diabetes associated with AMPK activation. However, whether metformin prevents MG-induced beta cell apoptosis is controversial. Here, we explored the signaling pathway involved in the anti-apoptotic effect of GLP-1, and investigated whether metformin had an anti-apoptotic effect on beta cells. MG treatment induced apoptosis of beta cells, impaired mitochondrial function, and prolonged activation of AMP-dependent protein kinase (AMPK). The MG-induced pro-apoptotic effects were abolished by an AMPK inhibitor. Pretreatment of GLP-1 reversed MG-induced apoptosis, and mitochondrial dysfunction, and suppressed prolonged AMPK activation. Pretreatment of GLP-1 reversed AMPK activator 5-aminoimidazole-4-carboxamide riboside (AICAR)-induced apoptosis, and suppressed prolonged AMPK activation. However, metformin neither leads to beta cell apoptosis nor ameliorates MG-induced beta cell apoptosis. In parallel, GLP-1 also prevents MG-induced beta cell apoptosis through PKA and PI3K-dependent pathway. In conclusion, these data indicates GLP-1 but not metformin protects MG-induced beta cell apoptosis through improving mitochondrial function, and alleviating the prolonged AMPK activation. Whether adding GLP-1 to metformin provides better beta cell survival and delays disease progression remains to be validated.
Collapse
Affiliation(s)
- Tien-Jyun Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Hsing-Chi Tseng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan.,Institute of Molecular Medicine, National Taiwan University Medical College, Taipei 10002, Taiwan
| | - Meng-Wei Liu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Yi-Cheng Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, National Taiwan University Medical College, Taipei 10002, Taiwan.,Institute of Biomedical Science, Academia Sinica, Taipei, 11500, Taiwan
| | - Meng-Lun Hsieh
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University Medical College, Taipei 10002, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan.,Institute of Molecular Medicine, National Taiwan University Medical College, Taipei 10002, Taiwan
| |
Collapse
|
17
|
Fred RG, Kappe C, Ameur A, Cen J, Bergsten P, Ravassard P, Scharfmann R, Welsh N. Role of the AMP kinase in cytokine-induced human EndoC-βH1 cell death. Mol Cell Endocrinol 2015. [PMID: 26213325 DOI: 10.1016/j.mce.2015.07.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The aim of the present investigation was to delineate cytokine-induced signaling and death using the EndoC-βH1 cells as a model for primary human beta-cells. The cytokines IL-1β and IFN-γ induced a rapid and transient activation of NF-κB, STAT-1, ERK, JNK and eIF-2α signaling. The EndoC-βH1 cells died rapidly when exposed to IL-1β + IFN-γ, and this occurred also in the presence of the actinomycin D. Inhibition of NF-κB and STAT-1 did not protect against cell death, nor did the cytokines activate iNOS expression. Instead, cytokines promoted a rapid decrease in EndoC-βH1 cell respiration and ATP levels, and we observed protection by the AMPK activator AICAR against cytokine-induced cell death. It is concluded that EndoC-βH1 cell death can be prevented by AMPK activation, which suggests a role for ATP depletion in cytokine-induced human beta-cell death.
Collapse
Affiliation(s)
- Rikard G Fred
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Camilla Kappe
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Adam Ameur
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Jing Cen
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Peter Bergsten
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Phillippe Ravassard
- Biotechnology and Biotherapy Laboratory, Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, CHU Pitié-Salpêtrière, Paris, France
| | - Raphael Scharfmann
- INSERM, U1016, Institut Cochin, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Nils Welsh
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
18
|
Matsuda T, Takahashi H, Mieda Y, Shimizu S, Kawamoto T, Matsuura Y, Takai T, Suzuki E, Kanno A, Koyanagi-Kimura M, Asahara SI, Bartolome A, Yokoi N, Inoue H, Ogawa W, Seino S, Kido Y. Regulation of Pancreatic β Cell Mass by Cross-Interaction between CCAAT Enhancer Binding Protein β Induced by Endoplasmic Reticulum Stress and AMP-Activated Protein Kinase Activity. PLoS One 2015; 10:e0130757. [PMID: 26091000 PMCID: PMC4474801 DOI: 10.1371/journal.pone.0130757] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/22/2015] [Indexed: 01/05/2023] Open
Abstract
During the development of type 2 diabetes, endoplasmic reticulum (ER) stress leads to not only insulin resistance but also to pancreatic beta cell failure. Conversely, cell function under various stressed conditions can be restored by reducing ER stress by activating AMP-activated protein kinase (AMPK). However, the details of this mechanism are still obscure. Therefore, the current study aims to elucidate the role of AMPK activity during ER stress-associated pancreatic beta cell failure. MIN6 cells were loaded with 5-amino-1-β-D-ribofuranosyl-imidazole-4-carboxamide (AICAR) and metformin to assess the relationship between AMPK activity and CCAAT enhancer binding protein β (C/EBPβ) expression levels. The effect of C/EBPβ phosphorylation on expression levels was also investigated. Vildagliptin and metformin were administered to pancreatic beta cell-specific C/EBPβ transgenic mice to investigate the relationship between C/EBPβ expression levels and AMPK activity in the pancreatic islets. When pancreatic beta cells are exposed to ER stress, the accumulation of the transcription factor C/EBPβ lowers the AMP/ATP ratio, thereby decreasing AMPK activity. In an opposite manner, incubation of MIN6 cells with AICAR or metformin activated AMPK, which suppressed C/EBPβ expression. In addition, administration of the dipeptidyl peptidase-4 inhibitor vildagliptin and metformin to pancreatic beta cell-specific C/EBPβ transgenic mice decreased C/EBPβ expression levels and enhanced pancreatic beta cell mass in proportion to the recovery of AMPK activity. Enhanced C/EBPβ expression and decreased AMPK activity act synergistically to induce ER stress-associated pancreatic beta cell failure.
Collapse
Affiliation(s)
- Tomokazu Matsuda
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroaki Takahashi
- Division of Medical Chemistry, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Yusuke Mieda
- Division of Medical Chemistry, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Shinobu Shimizu
- Division of Medical Chemistry, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Takeshi Kawamoto
- Division of Medical Chemistry, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Yuki Matsuura
- Division of Medical Chemistry, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Tomoko Takai
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Emi Suzuki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ayumi Kanno
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Maki Koyanagi-Kimura
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shun-ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Alberto Bartolome
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, United States of America
| | - Norihide Yokoi
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Inoue
- Department of Physiology and Metabolism, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Susumu Seino
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Medical Chemistry, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
- * E-mail:
| |
Collapse
|
19
|
Tong X, Kono T, Evans-Molina C. Nitric oxide stress and activation of AMP-activated protein kinase impair β-cell sarcoendoplasmic reticulum calcium ATPase 2b activity and protein stability. Cell Death Dis 2015; 6:e1790. [PMID: 26086963 PMCID: PMC4669835 DOI: 10.1038/cddis.2015.154] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/13/2015] [Accepted: 04/20/2015] [Indexed: 11/15/2022]
Abstract
The sarcoendoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) pump maintains a steep Ca2+ concentration gradient between the cytosol and ER lumen in the pancreatic β-cell, and the integrity of this gradient has a central role in regulated insulin production and secretion, maintenance of ER function and β-cell survival. We have previously demonstrated loss of β-cell SERCA2b expression under diabetic conditions. To define the mechanisms underlying this, INS-1 cells and rat islets were treated with the proinflammatory cytokine interleukin-1β (IL-1β) combined with or without cycloheximide or actinomycin D. IL-1β treatment led to increased inducible nitric oxide synthase (iNOS) gene and protein expression, which occurred concurrently with the activation of AMP-activated protein kinase (AMPK). IL-1β led to decreased SERCA2b mRNA and protein expression, whereas time-course experiments revealed a reduction in protein half-life with no change in mRNA stability. Moreover, SERCA2b protein but not mRNA levels were rescued by treatment with the NOS inhibitor l-NMMA (NG-monomethyl l-arginine), whereas the NO donor SNAP (S-nitroso-N-acetyl-d,l-penicillamine) and the AMPK activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide) recapitulated the effects of IL-1β on SERCA2b protein stability. Similarly, IL-1β-induced reductions in SERCA2b expression were rescued by pharmacological inhibition of AMPK with compound C or by transduction of a dominant-negative form of AMPK, whereas β-cell death was prevented in parallel. Finally, to determine a functional relationship between NO and AMPK signaling and SERCA2b activity, fura-2/AM (fura-2-acetoxymethylester) Ca2+ imaging experiments were performed in INS-1 cells. Consistent with observed changes in SERCA2b expression, IL-1β, SNAP and AICAR increased cytosolic Ca2+ and decreased ER Ca2+ levels, suggesting congruent modulation of SERCA activity under these conditions. In aggregate, these results show that SERCA2b protein stability is decreased under inflammatory conditions through NO- and AMPK-dependent pathways and provide novel insight into pathways leading to altered β-cell calcium homeostasis and reduced β-cell survival in diabetes.
Collapse
Affiliation(s)
- X Tong
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - T Kono
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - C Evans-Molina
- 1] Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA [2] Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA [3] Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA [4] The Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA [5] The Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
20
|
Lee MS. Role of islet β cell autophagy in the pathogenesis of diabetes. Trends Endocrinol Metab 2014; 25:620-7. [PMID: 25242548 DOI: 10.1016/j.tem.2014.08.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/17/2014] [Accepted: 08/24/2014] [Indexed: 01/03/2023]
Abstract
While the role of autophagy in the physiology of endocrine organs and the development of metabolic disorders or diabetes has been investigated, the relationship between the pancreatic islet and autophagy has not been explored extensively. In this review, studies on the possible involvement of dysregulated autophagy in the pathogenesis of metabolic syndrome and diabetes will be summarized with an emphasis on the autophagic process in pancreatic islet β cells. Novel findings regarding the role of autophagy in human β cell pathology and the development of type 2 diabetes (T2D) characterized by islet amyloid deposition will be discussed. Careful design of agents enhancing autophagic activity in β cells might serve as a novel approach towards therapeutics for the treatment of diabetes.
Collapse
Affiliation(s)
- Myung-Shik Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea.
| |
Collapse
|
21
|
Guan FY, Gu J, Li W, Zhang M, Ji Y, Li J, Chen L, Hatch GM. Compound K protects pancreatic islet cells against apoptosis through inhibition of the AMPK/JNK pathway in type 2 diabetic mice and in MIN6 β-cells. Life Sci 2014; 107:42-9. [PMID: 24802125 DOI: 10.1016/j.lfs.2014.04.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/08/2014] [Accepted: 04/24/2014] [Indexed: 01/27/2023]
Abstract
AIMS Compound K (CK) is known to possess anti-diabetic activities but the mechanism for this action is unknown. The present study observed the protective effect of CK on islet cell apoptosis through the AMP-activated protein kinase (AMPK) mediated C-Jun N-terminal kinase (JNK) pathway. MAIN METHODS Treatment effect of CK on type 2 diabetic (T2D) mice and palmitate-induced MIN6 β-cells injury was observed. Fasting plasma glucose, triacylglycerol, total cholesterol, insulin levels and glucose tolerance test were evaluated. The expression of AMPK and JNK was detected in islet and MIN6 cells. KEY FINDINGS CK treatment (30 mg/kg) decreased fasting plasma glucose, triacylglycerol, total cholesterol, elevated plasma insulin levels and improved glucose tolerance in T2D mice. CK treatment attenuated islet cell apoptosis and caspase-3 activity accompanied by a decrease in AMPK and JNK activation. Meanwhile, CK treatment attenuated the palmitate-induced reduction in MIN6 β-cell viability, apoptosis and caspase-3 activity and activation of AMPK and JNK. The AMPK activator AICAR attenuated the CK-mediated inhibition of palmitate-induced apoptosis. SIGNIFICANCE These data suggest that CK treatment provides a beneficial anti-diabetic effect in mice with T2D and this protective effect may be mediated through prevention of β-cell apoptosis via inhibition of the AMPK-JNK pathway.
Collapse
Affiliation(s)
- Feng Ying Guan
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Jian Gu
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130018, China
| | - Ming Zhang
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Yingshi Ji
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Jing Li
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Li Chen
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China.
| | - Grant M Hatch
- Department of Pharmacology & Therapeutics, University of Manitoba, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| |
Collapse
|
22
|
Allagnat F, Klee P, Cardozo AK, Meda P, Haefliger JA. Connexin36 contributes to INS-1E cells survival through modulation of cytokine-induced oxidative stress, ER stress and AMPK activity. Cell Death Differ 2013; 20:1742-52. [PMID: 24096873 PMCID: PMC3824597 DOI: 10.1038/cdd.2013.134] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 08/21/2013] [Accepted: 08/25/2013] [Indexed: 12/22/2022] Open
Abstract
Cell-to-cell communication mediated by gap junctions made of Connexin36 (Cx36) contributes to pancreatic β-cell function. We have recently demonstrated that Cx36 also supports β-cell survival by a still unclear mechanism. Using specific Cx36 siRNAs or adenoviral vectors, we now show that Cx36 downregulation promotes apoptosis in INS-1E cells exposed to the pro-inflammatory cytokines (IL-1β, TNF-α and IFN-γ) involved at the onset of type 1 diabetes, whereas Cx36 overexpression protects against this effect. Cx36 overexpression also protects INS-1E cells against endoplasmic reticulum (ER) stress-mediated apoptosis, and alleviates the cytokine-induced production of reactive oxygen species, the depletion of the ER Ca(2+) stores, the CHOP overexpression and the degradation of the anti-apoptotic protein Bcl-2 and Mcl-1. We further show that cytokines activate the AMP-dependent protein kinase (AMPK) in a NO-dependent and ER-stress-dependent manner and that AMPK inhibits Cx36 expression. Altogether, the data suggest that Cx36 is involved in Ca(2+) homeostasis within the ER and that Cx36 expression is downregulated following ER stress and subsequent AMPK activation. As a result, cytokine-induced Cx36 downregulation elicits a positive feedback loop that amplifies ER stress and AMPK activation, leading to further Cx36 downregulation. The data reveal that Cx36 plays a central role in the oxidative stress and ER stress induced by cytokines and the subsequent regulation of AMPK activity, which in turn controls Cx36 expression and mitochondria-dependent apoptosis of insulin-producing cells.
Collapse
Affiliation(s)
- F Allagnat
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne. Switzerland
| | - P Klee
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - A K Cardozo
- Laboratoire de Médecine Expérimentale, Université Libre de Bruxelles, Brussels, Belgium
| | - P Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - J-A Haefliger
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne. Switzerland
| |
Collapse
|
23
|
Noh JR, Hwang JH, Kim YH, Kim KS, Gang GT, Kim SW, Kim DK, Shong M, Lee IK, Choi HS, Lee CH. The orphan nuclear receptor small heterodimer partner negatively regulates pancreatic beta cell survival and hyperglycemia in multiple low-dose streptozotocin-induced type 1 diabetic mice. Int J Biochem Cell Biol 2013; 45:1538-45. [DOI: 10.1016/j.biocel.2013.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/02/2013] [Accepted: 05/06/2013] [Indexed: 12/19/2022]
|
24
|
Yeo SH, Noh JR, Kim YH, Gang GT, Kim SW, Kim KS, Hwang JH, Shong M, Lee CH. Increased vulnerability to β-cell destruction and diabetes in mice lacking NAD(P)H:quinone oxidoreductase 1. Toxicol Lett 2013; 219:35-41. [PMID: 23458895 DOI: 10.1016/j.toxlet.2013.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 02/19/2013] [Accepted: 02/19/2013] [Indexed: 10/27/2022]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) has been known to protect cells against stressors, including the diabetogenic reagent streptozotocin (STZ). The present study demonstrated that NQO1 deficiency resulted in increased pancreatic β-cell death induced by multiple low dose of STZ (MLDS) injections. NQO1 knockout (KO) mice showed hyperglycemia, body weight loss, impaired glucose clearance rate and a lower plasma insulin level after MLDS treatment. Moreover, β-cell mass and pancreatic insulin content were significantly lower in KO mice than in wild-type (WT) mice after MLDS treatment. Five days after the first STZ treatment, the islets of KO mice had substantially more TUNEL-positive β-cells than those of WT mice, but there was no difference in the regeneration of β-cells between KO mice and WT mice. At the same time, MLDS-treated KO mice showed significantly increased apoptotic markers in β-cells, including cleaved caspase 3, Smac/DIABLO and AIF (apoptosis inducing factor) in the cytoplasm. These results suggest that mice deficient in NQO1 are vulnerable to MLDS-induced β-cell destruction and diabetes, caused by increase of β-cell apoptosis in pancreas.
Collapse
Affiliation(s)
- Seung-Hoon Yeo
- Laboratory Animal Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejeon 305-806, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Barbagallo D, Piro S, Condorelli AG, Mascali LG, Urbano F, Parrinello N, Monello A, Statello L, Ragusa M, Rabuazzo AM, Di Pietro C, Purrello F, Purrello M. miR-296-3p, miR-298-5p and their downstream networks are causally involved in the higher resistance of mammalian pancreatic α cells to cytokine-induced apoptosis as compared to β cells. BMC Genomics 2013; 14:62. [PMID: 23360399 PMCID: PMC3571888 DOI: 10.1186/1471-2164-14-62] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/26/2013] [Indexed: 01/03/2023] Open
Abstract
Background The molecular bases of mammalian pancreatic α cells higher resistance than β to proinflammatory cytokines are very poorly defined. MicroRNAs are master regulators of cell networks, but only scanty data are available on their transcriptome in these cells and its alterations in diabetes mellitus. Results Through high-throughput real-time PCR, we analyzed the steady state microRNA transcriptome of murine pancreatic α (αTC1-6) and β (βTC1) cells: their comparison demonstrated significant differences. We also characterized the alterations of αTC1-6 cells microRNA transcriptome after treatment with proinflammatory cytokines. We focused our study on two microRNAs, miR-296-3p and miR-298-5p, which were: (1) specifically expressed at steady state in αTC1-6, but not in βTC1 or INS-1 cells; (2) significantly downregulated in αTC1-6 cells after treatment with cytokines in comparison to untreated controls. These microRNAs share more targets than expected by chance and were co-expressed in αTC1-6 during a 6–48 h time course treatment with cytokines. The genes encoding them are physically clustered in the murine and human genome. By exploiting specific microRNA mimics, we demonstrated that experimental upregulation of miR-296-3p and miR-298-5p raised the propensity to apoptosis of transfected and cytokine-treated αTC1-6 cells with respect to αTC1-6 cells, treated with cytokines after transfection with scramble molecules. Both microRNAs control the expression of IGF1Rβ, its downstream targets phospho-IRS-1 and phospho-ERK, and TNFα. Our computational analysis suggests that MAFB (a transcription factor exclusively expressed in pancreatic α cells within adult rodent islets of Langerhans) controls the expression of miR-296-3p and miR-298-5p. Conclusions Altogether, high-throughput microRNA profiling, functional analysis with synthetic mimics and molecular characterization of modulated pathways strongly suggest that specific downregulation of miR-296-3p and miR-298-5p, coupled to upregulation of their targets as IGF1Rβ and TNFα, is a major determinant of mammalian pancreatic α cells resistance to apoptosis induction by cytokines.
Collapse
Affiliation(s)
- Davide Barbagallo
- Dipartimento Gian Filippo Ingrassia, Unità di BioMedicina Molecolare Genomica e dei Sistemi Complessi, Genetica, Biologia Computazionale, Università di Catania, Catania, EU 95123, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Miao XY, Gu ZY, Liu P, Hu Y, Li L, Gong YP, Shu H, Liu Y, Li CL. The human glucagon-like peptide-1 analogue liraglutide regulates pancreatic beta-cell proliferation and apoptosis via an AMPK/mTOR/P70S6K signaling pathway. Peptides 2013; 39:71-9. [PMID: 23116613 DOI: 10.1016/j.peptides.2012.10.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 10/15/2012] [Accepted: 10/22/2012] [Indexed: 02/07/2023]
Abstract
Glucagon-like peptide-1 (GLP-1), an effective therapeutic agent for the treatment of diabetes, has been proven to protect pancreatic beta cells through many pathways. Recent evidence demonstrates that AMP-activated protein kinase (AMPK), as a metabolic regulator, coordinates beta-cell protein synthesis through regulation of the mammalian target of rapamycin (mTOR) signaling pathway. The purpose of the present study was to explore whether liraglutide, a human GLP-1 analogue, protects beta cells via AMPK/mTOR signaling. We evaluated INS-1 beta-cell line proliferation using the Cell Counting Kit-8, and examined the effect of GLP-1 on cellular ATP levels using an ATP assay kit. mTOR pathway protein expression levels were tested by Western blotting and glucolipotoxicity-induced cell apoptosis was evaluated by flow cytometry. Liraglutide increased beta-cell viability at an optimum concentration of 100 nmol/L in the presence of 11.1 or 30 mmol/L glucose. Liraglutide (100 nmol/L) activated mTOR and its downstream effectors, 70-kDa ribosomal protein S6 kinase and eIF4E-binding protein-1, in INS-1 cells. This effect was abated by pathway blockers: the AMPK activator AICAR and the mTOR inhibitor rapamycin. Furthermore, the effect of liraglutide on beta-cell proliferation was inhibited by AICAR and rapamycin. Liraglutide increased cellular ATP levels. In addition, liraglutide protected beta cells from glucolipotoxicity-induced apoptosis. This response was also prevented by rapamycin treatment. These results suggest that the enhancement of beta-cell proliferation by that GLP-1 receptor agonist liraglutide is mediated, at least in part, by AMPK/mTOR signaling. Liraglutide also prevents beta-cell glucolipotoxicity by activating mTOR.
Collapse
Affiliation(s)
- Xin-Yu Miao
- Department of Geriatric Endocrinology, General Hospital of PLA, 100853 Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Salt IP, Palmer TM. Exploiting the anti-inflammatory effects of AMP-activated protein kinase activation. Expert Opin Investig Drugs 2012; 21:1155-67. [PMID: 22694351 DOI: 10.1517/13543784.2012.696609] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION AMP-activated protein kinase (AMPK) is the downstream component of a serine/threonine protein kinase cascade involved in the regulation of metabolism. Many studies have also revealed that AMPK activation can exert significant anti-inflammatory and immunosuppressive effects in a variety of cell types and models of inflammatory/autoimmune disease. Because metformin, an AMPK activator that is a favored first-line therapeutic option for type 2 diabetes, may confer benefits in chronic inflammatory diseases and cancers independent of its ability to normalize blood glucose, there is now considerable interest in identifying and exploiting AMPK's anti-inflammatory effects. AREAS COVERED The authors provide a background to AMPK signaling and describe the pro-inflammatory signaling pathways and processes shown to be regulated by AMPK activation. EXPERT OPINION Identification of AMPK subunits responsible for specific anti-inflammatory effects, and a molecular understanding of the mechanisms involved, will be necessary to exploit AMPK pathway activation in acute and chronic inflammatory disease settings while minimizing adverse reactions due to deregulation of AMPK's wide-ranging effects on metabolism.
Collapse
Affiliation(s)
- Ian P Salt
- University of Glasgow, Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, Glasgow G12 8QQ, Scotland, UK
| | | |
Collapse
|
28
|
Dias T, Liu B, Jones P, Houghton PJ, Mota-Filipe H, Paulo A. Cytoprotective effect of Coreopsis tinctoria extracts and flavonoids on tBHP and cytokine-induced cell injury in pancreatic MIN6 cells. JOURNAL OF ETHNOPHARMACOLOGY 2012; 139:485-92. [PMID: 22143153 DOI: 10.1016/j.jep.2011.11.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 11/02/2011] [Accepted: 11/19/2011] [Indexed: 05/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE [corrected] Coreopsis tinctoria flowering tops infusion is traditionally used in Portugal for treating the symptoms of diabetes. Recent studies have revealed its antihyperglycemic activity when administered for 3 weeks to a STZ-induced glucose intolerance model in the rat and glucose tolerance regain was even clearer and pancreatic function recovery was achieved when administering Coreopsis tinctoria flavonoid-rich AcOEt fraction. In this study we aimed to evaluate the protective effect of Coreopsis tinctoria flowering tops aqueous extract, AcOEt fraction and the pure compounds marein and flavanomarein, against beta-cell injury, in a mouse insulinoma cell line (MIN6) challenged with pro-oxidant tert-butyl-hydroperoxide (tBHP) or cytokines. MATERIALS AND METHODS The protective effects of Coreopsis tinctoria flowering tops extracts and pure compounds were evaluated through pre-incubating MIN6 cells with samples followed by treatment with tBHP (400 μM for 2 h) after which viability was determined through ATP measurements. In order to assess whether plant extracts were involved in decreasing reactive oxygen species, superoxide anion production was determined through a lucigenin-enhanced chemiluminescent method. Lastly, the direct influence of Coreopsis tinctoria extracts and main compounds on cell survival/apoptosis was determined measuring caspase 3 and 7 cleavage induced by cytokines. RESULTS Coreopsis tinctoria flowering tops extracts (25-100 μg/mL) and pure compounds (200-400 μM), when pre-incubated with MIN6 cells did not present any cytotoxicity, instead they increased cell viability in a dose dependent manner when challenged with tBHP. Treatment with this pro-oxidant also showed a rise in superoxide radical anion formation in MIN6 cells. This increase was significantly reduced by treatment with superoxide dismutase enzyme (SOD) but not by pre-treatment with Coreopsis tinctoria flowering tops extracts. Caspase 3/7 activation measurements show that Coreopsis tinctoria flowering tops extracts, as well as marein and flavanomarein, significantly inhibit apoptosis. CONCLUSIONS Coreopsis tinctoria extracts and pure compounds show cytoprotection that seems to be due to inhibition of the apoptotic pathway, and not through a decrease on superoxide radical production.
Collapse
Affiliation(s)
- Teresa Dias
- i.Med-UL-Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | | | | | | | | |
Collapse
|
29
|
Shaked M, Ketzinel-Gilad M, Cerasi E, Kaiser N, Leibowitz G. AMP-activated protein kinase (AMPK) mediates nutrient regulation of thioredoxin-interacting protein (TXNIP) in pancreatic beta-cells. PLoS One 2011; 6:e28804. [PMID: 22194917 PMCID: PMC3237554 DOI: 10.1371/journal.pone.0028804] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 11/15/2011] [Indexed: 02/07/2023] Open
Abstract
Thioredoxin-interacting protein (TXNIP) regulates critical biological processes including inflammation, stress and apoptosis. TXNIP is upregulated by glucose and is a critical mediator of hyperglycemia-induced beta-cell apoptosis in diabetes. In contrast, the saturated long-chain fatty acid palmitate, although toxic to the beta-cell, inhibits TXNIP expression. The mechanisms involved in the opposing effects of glucose and fatty acids on TXNIP expression are unknown. We found that both palmitate and oleate inhibited TXNIP in a rat beta-cell line and islets. Palmitate inhibition of TXNIP was independent of fatty acid beta-oxidation or esterification. AMP-activated protein kinase (AMPK) has an important role in cellular energy sensing and control of metabolic homeostasis; therefore we investigated its involvement in nutrient regulation of TXNIP. As expected, glucose inhibited whereas palmitate stimulated AMPK. Pharmacologic activators of AMPK mimicked fatty acids by inhibiting TXNIP. AMPK knockdown increased TXNIP expression in presence of high glucose with and without palmitate, indicating that nutrient (glucose and fatty acids) effects on TXNIP are mediated in part via modulation of AMPK activity. TXNIP is transcriptionally regulated by carbohydrate response element-binding protein (ChREBP). Palmitate inhibited glucose-stimulated ChREBP nuclear entry and recruitment to the Txnip promoter, thereby inhibiting Txnip transcription. We conclude that AMPK is an important regulator of Txnip transcription via modulation of ChREBP activity. The divergent effects of glucose and fatty acids on TXNIP expression result in part from their opposing effects on AMPK activity. In light of the important role of TXNIP in beta-cell apoptosis, its inhibition by fatty acids can be regarded as an adaptive/protective response to glucolipotoxicity. The finding that AMPK mediates nutrient regulation of TXNIP may have important implications for the pathophysiology and treatment of diabetes.
Collapse
Affiliation(s)
- Maayan Shaked
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Mali Ketzinel-Gilad
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Erol Cerasi
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nurit Kaiser
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Gil Leibowitz
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
30
|
Jahansouz C, Jahansouz C, Kumer SC, Brayman KL. Evolution of β-Cell Replacement Therapy in Diabetes Mellitus: Islet Cell Transplantation. J Transplant 2011; 2011:247959. [PMID: 22013505 PMCID: PMC3195999 DOI: 10.1155/2011/247959] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 08/08/2011] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus remains one of the leading causes of morbidity and mortality worldwide. According to the Centers for Disease Control and Prevention, approximately 23.6 million people in the United States are affected. Of these individuals, 5 to 10% have been diagnosed with Type 1 diabetes mellitus (T1DM), an autoimmune disease. Although it often appears in childhood, T1DM may manifest at any age, leading to significant morbidity and decreased quality of life. Since the 1960s, the surgical treatment for diabetes mellitus has evolved to become a viable alternative to insulin administration, beginning with pancreatic transplantation. While islet cell transplantation has emerged as another potential alternative, its role in the treatment of T1DM remains to be solidified as research continues to establish it as a truly viable alternative for achieving insulin independence. In this paper, the historical evolution, procurement, current status, benefits, risks, and ongoing research of islet cell transplantation are explored.
Collapse
Affiliation(s)
- Cyrus Jahansouz
- School of Medicine, University of Virginia, Charlottesville, VA 22102, USA
| | | | | | | |
Collapse
|
31
|
Santos GJ, Oliveira CA, Boschero AC, Rezende LF. CNTF protects MIN6 cells against apoptosis induced by Alloxan and IL-1β through downregulation of the AMPK pathway. Cell Signal 2011; 23:1669-76. [DOI: 10.1016/j.cellsig.2011.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Revised: 06/01/2011] [Accepted: 06/06/2011] [Indexed: 01/27/2023]
|
32
|
Targeted therapies of the LKB1/AMPK pathway for the treatment of insulin resistance. Future Med Chem 2011; 2:1785-96. [PMID: 21428801 DOI: 10.4155/fmc.10.264] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type II diabetes is characterized by elevated serum glucose levels and altered lipid metabolism due to peripheral insulin resistance and defects of insulin secretion in the pancreatic β-cells. While some cases of obesity and Type II diabetes result from genetic dysfunction, the increased worldwide incidence of these two disorders strongly suggest that the contribution of environmental factors such as sedentary lifestyles and high-calorie intake may disrupt energy balance. AMP-activated protein kinase and its upstream kinase liver kinase B1 are conserved serine/threonine kinases regulating anabolic and catabolic metabolic processes, therefore representing attractive therapeutic targets for the treatment of obesity and Type II diabetes. In this review, we will discuss the advantages of targeting the liver kinase B1/AMP-activated protein kinase pathway for the treatment of metabolic diseases.
Collapse
|
33
|
Abstract
AMP-activated protein kinase AMP-activated protein kinase (AMPK AMPK ), a phylogenetically conserved serine/threonine protein kinase, is a major regulator of cellular and whole-body energy homeostasis that coordinates metabolic pathways in order to balance nutrient supply with energy demand. It is now recognized that pharmacological activation of AMPK improves blood glucose homeostasis, lipid profile, and blood pressure in insulin-resistant rodents. Indeed, AMPK activation mimics the beneficial effects of physical activity or those of calorie restriction calorie restriction by acting on multiple cellular targets. In addition, it is now demonstrated that AMPK is one of the probable (albeit indirect) targets of major antidiabetic drugs drugs including the biguanides (metformin metformin ) and thiazolidinedione thiazolidinedione s, as well as of insulin-sensitizing adipokines (e.g., adiponectin adiponectin ). Taken together, such findings highlight the logic underlying the concept of targeting the AMPK pathway for the treatment of metabolic syndrome and type 2 diabetes.
Collapse
|
34
|
Yun H, Ha J. AMP-activated protein kinase modulators: a patent review (2006 - 2010). Expert Opin Ther Pat 2011; 21:983-1005. [PMID: 21548715 DOI: 10.1517/13543776.2011.577069] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION AMPK is a key player in the regulation of energy balance at both the cellular and whole-body levels, placing it at the center stage in studies of metabolic disorders. Recently, AMPK has also been identified as a potential target for either therapy or prevention of some types of cancer. Thus, identification of AMPK modulators for possible use as novel therapeutic drugs, both for treatment of metabolic disorders and cancer, will have a high commercial potential. AREAS COVERED This review covers the structures and activities of AMPK modulators described in the patent literature since 2006. The patents reviewed include those for direct and/or indirect activators of AMPK, and novel pharmaceutical compounds with potential for use in the prevention and/or treatment of metabolic disorders, and cancer targeting AMPK. EXPERT OPINION Targeting of AMPK appears to be an attractive strategy in the treatment of metabolic disorders. However, some detrimental effects of AMPK have also been reported, including a possible tumor-promoting effect in some settings and a heart disease-causing effect. Moreover, activation of AMPK in the hypothalamus may cause undesired consequences, such as an increase in feeding and body weight gain. These effects, therefore, must be carefully assessed for the development of therapeutic drugs targeting AMPK.
Collapse
Affiliation(s)
- Hee Yun
- Kyung Hee University, Medical Research Center and Biomedical Science Institute, School of Medicine, Department of Biochemistry and Molecular Biology, Seoul, Republic of Korea
| | | |
Collapse
|
35
|
Bitter melon (Momordica charantia) extract suppresses cytokineinduced activation of MAPK and NF-κB in pancreatic β-Cells. Food Sci Biotechnol 2011. [DOI: 10.1007/s10068-011-0074-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
36
|
Diraison F, Hayward K, Sanders KL, Brozzi F, Lajus S, Hancock J, Francis JE, Ainscow E, Bommer UA, Molnar E, Avent ND, Varadi A. Translationally controlled tumour protein (TCTP) is a novel glucose-regulated protein that is important for survival of pancreatic beta cells. Diabetologia 2011; 54:368-79. [PMID: 21063673 DOI: 10.1007/s00125-010-1958-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 09/27/2010] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS This study used proteomics and biochemical approaches to identify novel glucose-regulated proteins and to unveil their role in pancreatic beta cell function. Translationally controlled tumour protein (TCTP) was identified to be one such protein, and further investigations into its function and regulation were carried out. METHODS Global protein profiling of beta cell homogenates following glucose stimulation was performed using two-dimensional gel electrophoresis. Proteins were identified by mass spectroscopy analysis. Immunoblotting was used to investigate alterations in TCTP protein levels in response to glucose stimulation or cell stress induced by palmitate. To investigate the biological function of TCTP, immunolocalisation, gene knockdown and overexpression of Tctp (also known as Tpt1) were performed. Apoptosis was measured in Tctp knockdown or Tctp-overexpressing cells. Glucose-stimulated insulin secretion was carried out in Tctp knockdown cells. RESULTS TCTP was identified as a novel glucose-regulated protein, the level of which is increased at stimulatory glucose concentration. Glucose also induced TCTP dephosphorylation and its partial translocation to the mitochondria and the nucleus. TCTP protein levels were downregulated in response to cell stress induced by palmitate or thapsigargin treatments. Gene knockdown by small interfering RNA led to increased apoptosis, whereas overproduction of TCTP prevented palmitate-induced cell death. CONCLUSIONS/INTERPRETATION Regulation of TCTP protein levels by glucose is likely to be an important cyto-protective mechanism for pancreatic beta cells against damage caused by hyperglycaemia. In contrast, high concentration of palmitate causes cell stress, reduction in TCTP levels and consequently reduced cell viability. Our results imply that TCTP levels influence the sensitivity of beta cells to apoptosis.
Collapse
Affiliation(s)
- F Diraison
- Centre for Research in Biomedicine, Faculty of Health and Life Sciences, University of the West of England, Bristol BS16 1QY, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Exercise, together with a low-energy diet, is the first-line treatment for type 2 diabetes type 2 diabetes . Exercise improves insulin sensitivity insulin sensitivity by increasing the number or function of muscle mitochondria mitochondria and the capacity for aerobic metabolism, all of which are low in many insulin-resistant subjects. Cannabinoid 1-receptor antagonists and β-adrenoceptor agonists improve insulin sensitivity in humans and promote fat oxidation in rodents independently of reduced food intake. Current drugs for the treatment of diabetes are not, however, noted for their ability to increase fat oxidation, although the thiazolidinediones increase the capacity for fat oxidation in skeletal muscle, whilst paradoxically increasing weight gain.There are a number of targets for anti-diabetic drugs that may improve insulin sensitivity insulin sensitivity by increasing the capacity for fat oxidation. Their mechanisms of action are linked, notably through AMP-activated protein kinase, adiponectin, and the sympathetic nervous system. If ligands for these targets have obvious acute thermogenic activity, it is often because they increase sympathetic activity. This promotes fuel mobilisation, as well as fuel oxidation. When thermogenesis thermogenesis is not obvious, researchers often argue that it has occurred by using the inappropriate device of treating animals for days or weeks until there is weight (mainly fat) loss and then expressing energy expenditure energy expenditure relative to body weight. In reality, thermogenesis may have occurred, but it is too small to detect, and this device distracts us from really appreciating why insulin sensitivity has improved. This is that by increasing fatty acid oxidation fatty acid oxidation more than fatty acid supply, drugs lower the concentrations of fatty acid metabolites that cause insulin resistance. Insulin sensitivity improves long before any anti-obesity effect can be detected.
Collapse
Affiliation(s)
- Jonathan R S Arch
- Clore Laboratory, University of Buckingham, Buckingham, MK18 1EG, UK
| |
Collapse
|
38
|
Abstract
The chapters throughout this volume illustrate the many contributions of mitochondria to the maintenance of normal cell and tissue function, experienced as the health of the individual. Mitochondria are essential for maintaining aspects of physiology as fundamental as cellular energy balance, the modulation of calcium signalling, in defining cellular redox balance, and they house significant biosynthetic pathways. Mitochondrial numbers and volume within cells are regulated and have an impact on their functional roles, while, especially in the CNS (central nervous system), mitochondrial trafficking is critical to ensure the cellular distribution and strategic localization of mitochondria, presumably driven by local energy demand. Maintenance of a healthy mitochondrial population involves a complex system of quality control, involving degrading misfolded proteins, while damaged mitochondria are renewed by fusion or removed by autophagy. It seems evident that mechanisms that impair any of these processes will impair mitochondrial function and cell signalling pathways, leading to disordered cell function which manifests as disease. As gatekeepers of cell life and cell death, mitochondria regulate both apoptotic and necrotic cell death, and so at its most extreme, disturbances involving these pathways may trigger untimely cell death. Conversely, the lack of appropriate cell death can lead to inappropriate tissue growth and development of tumours, which are also characterized by altered mitochondrial metabolism. The centrality of mitochondrial dysfunction to a surprisingly wide range of major human diseases is slowly becoming recognized, bringing with it the prospect of novel therapeutic approaches to treat a multitude of unpleasant and pervasive diseases.
Collapse
|
39
|
Irwin N, McKinney JM, Bailey CJ, Flatt PR, McClenaghan NH. Effects of metformin on BRIN-BD11 beta-cell insulin secretory desensitization induced by prolonged exposure to sulphonylureas. Diabetes Obes Metab 2010; 12:1066-71. [PMID: 20977577 DOI: 10.1111/j.1463-1326.2010.01294.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS Prolonged exposure of pancreatic beta-cells in vitro to the sulphonylureas tolbutamide and glibenclamide induces subsequent desensitization of insulinotropic pathways. Clinically, the insulin-sensitizing biguanide drug metformin is often administered alongside sulphonylurea as antidiabetic therapy. The present study examines the functional effects of metformin (200 µM) on tolbutamide- and glibenclamide-induced desensitisation. METHODS Acute and prolonged (18 h) effects of exposure to tolbutamide and glibenclamide alone, or in the presence of metformin, were examined in insulin-secreting BRIN-BD11 cells. RESULTS In acute 20 min incubations at 1.1 mM glucose, metformin increased (1.2-1.7-fold; p < 0.001) the insulin-releasing actions of tolbutamide and glibenclamide. At 16.7 mM glucose, metformin significantly enhanced glibenclamide-induced insulin release at all concentrations (50-400 µM) examined, but tolbutamide-stimulated insulin secretion was only augmented at higher concentrations (300-400 µM). Exposure for 18 h to 100 µM tolbutamide or glibenclamide significantly impaired insulin release in response to glucose and a broad range of insulin secretagogues. Concomitant culture with metformin (200 µM) prevented or partially reversed many of the adverse effects on K(ATP) channel dependent and independent insulinotropic pathways. Beneficial effects of metformin were also observed in cells exposed to glibenclamide for 18 h with significant improvements in the insulin secretory responsiveness to alanine, GLP-1 and sulphonylureas. The decrease of viable cell numbers observed with glibenclamide was reversed by co-culture with metformin, but cellular insulin content was depressed. CONCLUSIONS The results suggest that metformin can prevent the aspects of sulphonylurea-induced beta-cell desensitization.
Collapse
Affiliation(s)
- N Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK.
| | | | | | | | | |
Collapse
|
40
|
McKiney JM, Irwin N, Flatt PR, Bailey CJ, McClenaghan NH. Acute and long-term effects of metformin on the function and insulin secretory responsiveness of clonal β-cells. Biol Chem 2010; 391:1451-9. [DOI: 10.1515/bc.2010.139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Functional effects of acute and prolonged (48 h) exposure to the biguanide drug metformin were examined in the clonal pancreatic β-cell line, BRIN-BD11. Effects of metformin on prolonged exposure to excessive increased concentrations of glucose and palmitic acid were also assessed. In acute 20-min incubations, 12.5–50 μm metformin did not alter basal (1.1 mm glucose) or glucose-stimulated (16.7 mm glucose) insulin secretion. However, higher concentrations of metformin (100–1000 μm) increased (1.3–1.5-fold; p<0.001) insulin release at basal glucose concentrations, but had no effect on glucose-stimulated insulin secretion. There were no apparent acute effects of metformin on intracellular Ca2+ concentrations, but metformin enhanced (p<0.05 to p<0.01) the acute insulinotropic actions of GIP and GLP-1. Exposure for 48 h to 200 μm metformin improved aspects of β-cell insulin secretory function, whereas these benefits were lost at 1 mm metformin. Prolonged glucotoxic and lipotoxic conditions impaired β-cell viability and insulin release in response to glucose and to a broad range of insulin secretagogues. Concomitant culture with 200 μm metformin partially reversed many of the adverse effects of prolonged glucotoxic conditions. However, there were no beneficial effects of metformin under prolonged culture with elevated concentrations of palmitic acid. The results suggest that metformin exerts direct effects on β-cell viability, function and survival that could contribute to the use of this agent in the treatment of type 2 diabetes.
Collapse
|
41
|
Ding Y, Yamada S, Wang KY, Shimajiri S, Guo X, Tanimoto A, Murata Y, Kitajima S, Watanabe T, Izumi H, Kohno K, Sasaguri Y. Overexpression of peroxiredoxin 4 protects against high-dose streptozotocin-induced diabetes by suppressing oxidative stress and cytokines in transgenic mice. Antioxid Redox Signal 2010; 13:1477-90. [PMID: 20446767 DOI: 10.1089/ars.2010.3137] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Peroxiredoxin 4 (PRDX4) is one of a newly discovered family of antioxidative proteins. We generated human PRDX4 (hPRDX4) transgenic (Tg) mice, displaying a high level of hPRDX4 expression in the pancreatic islets, and then focused on the functions of PRDX4 in a type 1 diabetes mellitus (T1DM) model using a single high dose of streptozotocin (SHDS). After SHDS-injection, Tg mice showed significantly less hyperglycemia and hypoinsulinemia and a much faster response on glucose tolerance test than wild-type (WT) mice. Morphologic and immunohistochemical observation revealed that the pancreatic islet areas of Tg mice were larger along with less CD3-positive lymphocyte infiltration compared with WT mice. Upon comparison between these two mouse models, β-cell apoptosis was also repressed, and reversely, β-cell proliferation was enhanced in Tg mice. Real-time RT-PCR demonstrated that the expression of many inflammatory-related molecules and their receptors and transcription factors were significantly downregulated in Tg mice. These data indicate that PRDX4 can protect pancreatic islet β-cells against injury caused by SHDS-induced insulitis, which strongly suggests that oxidative stress plays an essential role in SHDS-induced diabetes. This study, for the first time, implicates that PRDX4 has a pivotal protective function against diabetes progression in this T1DM model.
Collapse
Affiliation(s)
- Yan Ding
- Department of Pathology and Cell Biology, School of Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kilbride SM, Farrelly AM, Bonner C, Ward MW, Nyhan KC, Concannon CG, Wollheim CB, Byrne MM, Prehn JHM. AMP-activated protein kinase mediates apoptosis in response to bioenergetic stress through activation of the pro-apoptotic Bcl-2 homology domain-3-only protein BMF. J Biol Chem 2010; 285:36199-206. [PMID: 20841353 PMCID: PMC2975242 DOI: 10.1074/jbc.m110.138107] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 08/26/2010] [Indexed: 11/06/2022] Open
Abstract
Heterozygous loss-of-function mutations in the hepatocyte nuclear factor 1A (HNF1A) gene result in the pathogenesis of maturity-onset diabetes-of-the-young type 3, (HNF1A-MODY). This disorder is characterized by a primary defect in metabolism-secretion coupling and decreased beta cell mass, attributed to excessive beta cell apoptosis. Here, we investigated the link between energy stress and apoptosis activation following HNF1A inactivation. This study employed single cell fluorescent microscopy, flow cytometry, gene expression analysis, and gene silencing to study the effects of overexpression of dominant-negative (DN)-HNF1A expression on cellular bioenergetics and apoptosis in INS-1 cells. Induction of DN-HNF1A expression led to reduced ATP levels and diminished the bioenergetic response to glucose. This was coupled with activation of the bioenergetic stress sensor AMP-activated protein kinase (AMPK), which preceded the onset of apoptosis. Pharmacological activation of AMPK using aminoimidazole carboxamide ribonucleotide (AICAR) was sufficient to induce apoptosis in naive cells. Conversely, inhibition of AMPK with compound C or AMPKα gene silencing protected against DN-HNF1A-induced apoptosis. Interestingly, AMPK mediated the induction of the pro-apoptotic Bcl-2 homology domain-3-only protein Bmf (Bcl-2-modifying factor). Bmf expression was also elevated in islets of DN-HNF1A transgenic mice. Furthermore, knockdown of Bmf expression in INS-1 cells using siRNA was sufficient to protect against DN-HNF1A-induced apoptosis. Our study suggests that overexpression of DN-HNF1A induces bioenergetic stress and activation of AMPK. This in turn mediates the transcriptional activation of the pro-apoptotic Bcl-2-homology protein BMF, coupling prolonged energy stress to apoptosis activation.
Collapse
Affiliation(s)
- Seán M. Kilbride
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | | | - Caroline Bonner
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Manus W. Ward
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Kristine C. Nyhan
- the Mater Misericordiae Hospital, Eccles Street, Dublin 7, Ireland, and
| | - Caoimhín G. Concannon
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Claes B. Wollheim
- the Department of Cell Physiology and Metabolism, University Medical Center, CH-1211 Geneva, Switzerland
| | - Maria M. Byrne
- the Mater Misericordiae Hospital, Eccles Street, Dublin 7, Ireland, and
| | - Jochen H. M. Prehn
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
43
|
Eid AA, Ford BM, Block K, Kasinath BS, Gorin Y, Ghosh-Choudhury G, Barnes JL, Abboud HE. AMP-activated protein kinase (AMPK) negatively regulates Nox4-dependent activation of p53 and epithelial cell apoptosis in diabetes. J Biol Chem 2010; 285:37503-12. [PMID: 20861022 DOI: 10.1074/jbc.m110.136796] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Diabetes and high glucose (HG) increase the generation of NADPH oxidase-derived reactive oxygen species and induce apoptosis of glomerular epithelial cells (podocytes). Loss of podocytes contributes to albuminuria, a major risk factor for progression of kidney disease. Here, we show that HG inactivates AMP-activated protein kinase (AMPK), up-regulates Nox4, enhances NADPH oxidase activity, and induces podocyte apoptosis. Activation of AMPK blocked HG-induced expression of Nox4, NADPH oxidase activity, and apoptosis. We also identified the tumor suppressor protein p53 as a mediator of podocyte apoptosis in cells exposed to HG. Inactivation of AMPK by HG up-regulated the expression and phosphorylation of p53, and p53 acted downstream of Nox4. To investigate the mechanism of podocyte apoptosis in vivo, we used OVE26 mice, a model of type 1 diabetes. Glomeruli isolated from these mice showed decreased phosphorylation of AMPK and enhanced expression of Nox4 and p53. Pharmacologic activation of AMPK by 5-aminoimidazole-4-carboxamide-1-riboside in OVE26 mice attenuated Nox4 and p53 expression. Administration of 5-aminoimidazole-4-carboxamide-1-riboside also prevented renal hypertrophy, glomerular basement thickening, foot process effacement, and podocyte loss, resulting in marked reduction in albuminuria. Our results uncover a novel function of AMPK that integrates metabolic input to Nox4 and provide new insight for activation of p53 to induce podocyte apoptosis. The data indicate the potential therapeutic utility of AMPK activators to block Nox4 and reactive oxygen species generation and to reduce urinary albumin excretion in type 1 diabetes.
Collapse
Affiliation(s)
- Assaad A Eid
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Sun G, Tarasov AI, McGinty JA, French PM, McDonald A, Leclerc I, Rutter GA. LKB1 deletion with the RIP2.Cre transgene modifies pancreatic beta-cell morphology and enhances insulin secretion in vivo. Am J Physiol Endocrinol Metab 2010; 298:E1261-73. [PMID: 20354156 PMCID: PMC2886523 DOI: 10.1152/ajpendo.00100.2010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The tumor suppressor liver kinase B1 (LKB1), also called STK11, is a protein kinase mutated in Peutz-Jeghers syndrome. LKB1 phosphorylates AMP-activated protein kinase (AMPK) and several related protein kinases. Whereas deletion of both catalytic isoforms of AMPK from the pancreatic beta-cell and hypothalamic neurons using the rat insulin promoter (RIP2).Cre transgene (betaAMPKdKO) diminishes insulin secretion in vivo, deletion of LKB1 in the beta-cell with an inducible Pdx-1.CreER transgene enhances insulin secretion in mice. To determine whether the differences between these models reflect genuinely distinct roles for the two kinases in the beta-cell or simply differences in the timing and site(s) of deletion, we have therefore created mice deleted for LKB1 with the RIP2.Cre transgene. In marked contrast to betaAMPKdKO mice, betaLKB1KO mice showed diminished food intake and weight gain, enhanced insulin secretion, unchanged insulin sensitivity, and improved glucose tolerance. In line with the phenotype of Pdx1-CreER mice, total beta-cell mass and the size of individual islets and beta-cells were increased and islet architecture was markedly altered in betaLKB1KO islets. Signaling by mammalian target of rapamycin (mTOR) to eIF4-binding protein-1 and ribosomal S6 kinase was also enhanced. In contrast to Pdx1-CreER-mediated deletion, the expression of Glut2, glucose-induced changes in membrane potential and intracellular Ca(2+) were sharply reduced in betaLKB1KO mouse islets and the stimulation of insulin secretion was modestly inhibited. We conclude that LKB1 and AMPK play distinct roles in the control of insulin secretion and that the timing of LKB1 deletion, and/or its loss from extrapancreatic sites, influences the final impact on beta-cell function.
Collapse
Affiliation(s)
- Gao Sun
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, UK
| | | | | | | | | | | | | |
Collapse
|
45
|
Sun G, Tarasov A, McGinty J, McDonald A, da Silva Xavier G, Gorman T, Marley A, French PM, Parker H, Gribble F, Reimann F, Prendiville O, Carzaniga R, Viollet B, Leclerc I, Rutter G. Ablation of AMP-activated protein kinase alpha1 and alpha2 from mouse pancreatic beta cells and RIP2.Cre neurons suppresses insulin release in vivo. Diabetologia 2010; 53:924-36. [PMID: 20221584 PMCID: PMC4306708 DOI: 10.1007/s00125-010-1692-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 02/01/2010] [Indexed: 10/19/2022]
Abstract
AIMS/HYPOTHESIS AMP-activated protein kinase (AMPK) is an evolutionarily conserved enzyme and a target of glucose-lowering agents, including metformin. However, the precise role or roles of the enzyme in controlling insulin secretion remain uncertain. METHODS The catalytic alpha1 and alpha2 subunits of AMPK were ablated selectively in mouse pancreatic beta cells and hypothalamic neurons by breeding Ampkalpha1 [also known as Prkaa1]-knockout mice, bearing floxed Ampkalpha2 [also known as Prkaa2] alleles (Ampkalpha1 ( -/- ),alpha2( fl/fl ),), with mice expressing Cre recombinase under the rat insulin promoter (RIP2). RIP2 was used to express constitutively activated AMPK selectively in beta cells in transgenic mice. Food intake, body weight and urinary catecholamines were measured using metabolic cages. Glucose and insulin tolerance were determined after intraperitoneal injection. Beta cell mass and morphology were analysed by optical projection tomography and confocal immunofluorescence microscopy, respectively. Granule docking, insulin secretion, membrane potential and intracellular free Ca(2+) were measured with standard techniques. RESULTS Trigenic Ampkalpha1 ( -/- ),alpha2( fl/fl ) expressing Cre recombinase and lacking both AMPKalpha subunits in the beta cell, displayed normal body weight and increased insulin sensitivity, but were profoundly insulin-deficient. Secreted catecholamine levels were unchanged. Total beta cell mass was unaltered, while mean islet and beta cell volume were reduced. AMPK-deficient beta cells displayed normal glucose-induced changes in membrane potential and intracellular free Ca(2+), while granule docking and insulin secretion were enhanced. Conversely, betaAMPK transgenic mice were glucose-intolerant and displayed defective insulin secretion. CONCLUSIONS/INTERPRETATION Inhibition of AMPK activity within the beta cell is necessary, but not sufficient for stimulation of insulin secretion by glucose to occur. AMPK activation in extrapancreatic RIP2.Cre-expressing cells might also influence insulin secretion in vivo.
Collapse
Affiliation(s)
- G. Sun
- Section of Cell Biology, Division of Endocrinology and Metabolism, Department of Medicine, Imperial College London
| | - A.I. Tarasov
- Section of Cell Biology, Division of Endocrinology and Metabolism, Department of Medicine, Imperial College London
| | - J. McGinty
- Photonics Group, Department of Physics, Imperial College London
| | - A. McDonald
- Section of Cell Biology, Division of Endocrinology and Metabolism, Department of Medicine, Imperial College London
| | - G. da Silva Xavier
- Section of Cell Biology, Division of Endocrinology and Metabolism, Department of Medicine, Imperial College London
| | - T. Gorman
- AstraZeneca, Alderley Edge, Cheshire
| | - A. Marley
- AstraZeneca, Alderley Edge, Cheshire
| | - P. M. French
- Photonics Group, Department of Physics, Imperial College London
| | - H. Parker
- Cambridge Institute of Medical Research, Cambridge, U.K
| | - F. Gribble
- Cambridge Institute of Medical Research, Cambridge, U.K
| | - F. Reimann
- Cambridge Institute of Medical Research, Cambridge, U.K
| | | | - R. Carzaniga
- Electron Microscopy Centre, Imperial College London
| | - B. Viollet
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104)
- Inserm, U1067, Paris, France
| | - I. Leclerc
- Section of Endocrinology and Metabolic Medicine, Division of Endocrinology and Metabolism, Department of Medicine, Imperial College London
| | - G.A. Rutter
- Section of Cell Biology, Division of Endocrinology and Metabolism, Department of Medicine, Imperial College London
| |
Collapse
|
46
|
Activation of autophagy through modulation of 5'-AMP-activated protein kinase protects pancreatic beta-cells from high glucose. Biochem J 2010; 425:541-51. [PMID: 19903169 DOI: 10.1042/bj20090429] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Chronic hyperglycaemia is detrimental to pancreatic beta-cells by causing impaired insulin secretion and diminished beta-cell function through glucotoxicity. Understanding the mechanisms underlying beta-cell survival is crucial for the prevention of beta-cell failure associated with glucotoxicity. Autophagy is a dynamic lysosomal degradation process that protects organisms against metabolic stress. To date, little is known about the physiological function of autophagy in the pathogenesis of diabetes. In the present study, we explored the roles of autophagy in the survival of pancreatic beta-cells exposed to high glucose using pharmacological and genetic manipulation of autophagy. We demonstrated that chronic high glucose increases autophagy in rat INS-1 (832/13) cells and pancreatic islets, and that this increase is enhanced by inhibition of 5'-AMP-activated protein kinase. Our results also indicate that stimulation of autophagy rescues pancreatic beta-cells from high-glucose-induced cell death and inhibition of autophagy augments caspase-3 activation, suggesting that autophagy plays a protective role in the survival of pancreatic beta-cells. Greater knowledge of the molecular mechanisms linking autophagy and beta-cell survival may unveil novel therapeutic targets needed to preserve beta-cell function.
Collapse
|
47
|
Abstract
Mitochondrial dysfunction plays a role in the pathogenesis of a wide range of diseases that involve disordered cellular fuel metabolism and survival/death pathways, including neurodegenerative diseases, cancer and diabetes. Cytokine, virus recognition and cellular stress pathways converging on mitochondria cause apoptotic and/or necrotic cell death of beta-cells in type-1 diabetes. Moreover, since mitochondria generate crucial metabolic signals for glucose stimulated insulin secretion (GSIS), mitochondrial dysfunction underlies both the functional derangement of GSIS and (over-nutrition) stress-induced apoptotic/necrotic beta-cell death, hallmarks of type-2 diabetes. The apparently distinct mechanisms governing beta-cell life/death decisions during the development of diabetes provide a remarkable example where remote metabolic, immune and stress signalling meet with mitochondria mediated apoptotic/necrotic death pathways to determine the fate of the beta-cell. We summarize the main findings supporting such a pivotal role of mitochondria in beta-cell death in the context of current trends in diabetes research.
Collapse
Affiliation(s)
- Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Mitochondrial Biology Group, University College London, Gower Street, WC1E 6BT London, UK.
| | | |
Collapse
|
48
|
Isolation and culture of mouse pancreatic islets for ex vivo imaging studies with trappable or recombinant fluorescent probes. Methods Mol Biol 2010; 633:171-84. [PMID: 20204627 DOI: 10.1007/978-1-59745-019-5_12] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The endocrine pancreas contains small clusters of 1,000-2,000 neuroendocrine cells termed islets of Langerhans. By secreting insulin, glucagon, or other hormones as circumstances dictate, islets play a central role in the control of glucose homeostasis in mammals. Islets are dispersed throughout the exocrine tissue and comprise only 1-2% of the volume of the whole organ; human pancreas contains about 10(6) islets whereas rodents have approximately 2 x 10(3) islets. The isolation of islets from the exocrine tissue usually begins with digestion of the pancreas with collagenase. Collagenase-containing medium is either injected into the pancreatic duct, and the organ left to digest in situ, or added after isolation of the pancreas and its dissection into small pieces ex vivo. Islets can then be separated from the exocrine tissue by gradient density or by handpicking. The islets obtained can either be used intact, for example, to measure insulin or glucagon secretion or be dispersed into single cells with a Ca(2+)-free medium or with trypsin/dispase. The latter facilitates the introduction of recombinant or trappable probes and microimaging studies of, for example, changes in cytosolic-free Ca(2+) concentration or the dynamics of individual organelles or proteins.
Collapse
|
49
|
Meares GP, Hughes KJ, Jaimes KF, Salvatori AS, Rhodes CJ, Corbett JA. AMP-activated protein kinase attenuates nitric oxide-induced beta-cell death. J Biol Chem 2009; 285:3191-200. [PMID: 19933272 DOI: 10.1074/jbc.m109.047365] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
During the initial autoimmune response in type 1 diabetes, islets are exposed to a damaging mix of pro-inflammatory molecules that stimulate the production of nitric oxide by beta-cells. Nitric oxide causes extensive but reversible cellular damage. In response to nitric oxide, the cell activates pathways for functional recovery and adaptation as well as pathways that direct beta-cell death. The molecular events that dictate cellular fate following nitric oxide-induced damage are currently unknown. In this study, we provide evidence that AMPK plays a primary role controlling the response of beta-cells to nitric oxide-induced damage. AMPK is transiently activated by nitric oxide in insulinoma cells and rat islets following IL-1 treatment or by the exogenous addition of nitric oxide. Active AMPK promotes the functional recovery of beta-cell oxidative metabolism and abrogates the induction of pathways that mediate cell death such as caspase-3 activation following exposure to nitric oxide. Overall, these data show that nitric oxide activates AMPK and that active AMPK suppresses apoptotic signaling allowing the beta-cell to recover from nitric oxide-mediated cellular stress.
Collapse
Affiliation(s)
- Gordon P Meares
- Department of Medicine, Division of Endocrinology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
50
|
Fogarty S, Hardie DG. Development of protein kinase activators: AMPK as a target in metabolic disorders and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2009; 1804:581-91. [PMID: 19778642 DOI: 10.1016/j.bbapap.2009.09.012] [Citation(s) in RCA: 273] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 09/10/2009] [Accepted: 09/15/2009] [Indexed: 10/20/2022]
Abstract
AMP-activated protein kinase (AMPK) is a cellular energy sensor activated by metabolic stresses that either inhibit ATP synthesis or accelerate ATP consumption. Activation of AMPK in response to an increase in the cellular AMP:ATP ratio results in inhibition of ATP-consuming processes such as gluconeogenesis and fatty acid synthesis, while stimulating ATP-generating processes, including fatty acid oxidation. These alterations in lipid and glucose metabolism would be expected to ameliorate the pathogenesis of obesity, type 2 diabetes and other metabolic disorders. Recently, AMPK has also been identified as a potential target for cancer prevention and/or treatment. Cell growth and proliferation are energetically demanding, and AMPK may act as an "energy checkpoint" that permits growth and proliferation only when energy reserves are sufficient. Thus, activators of AMPK could have potential as novel therapeutics both for metabolic disorders and for cancer, which together constitute two of the most prevalent groups of diseases worldwide.
Collapse
Affiliation(s)
- S Fogarty
- Division of Molecular Physiology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | | |
Collapse
|